1
|
Slade L, Etheridge T, Szewczyk NJ. Consolidating multiple evolutionary theories of ageing suggests a need for new approaches to study genetic contributions to ageing decline. Ageing Res Rev 2024; 100:102456. [PMID: 39153601 DOI: 10.1016/j.arr.2024.102456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 08/05/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Understanding mechanisms of ageing remains a complex challenge for biogerontologists, but recent adaptations of evolutionary ageing theories offer a compelling lens in which to view both age-related molecular and physiological deterioration. Ageing is commonly associated with progressive declines in biochemical and molecular processes resulting from damage accumulation, yet the role of continued developmental gene activation is less appreciated. Natural selection pressures are at their highest in youthful periods to modify gene expression towards maximising reproductive capacity. After sexual maturation, selective pressure diminishes, subjecting individuals to maladaptive pleiotropic gene functions that were once beneficial for developmental growth but become pathogenic later in life. Due to this selective 'shadowing' in ageing, mechanisms to counter such hyper/hypofunctional genes are unlikely to evolve. Interventions aimed at targeting gene hyper/hypofunction during ageing might, therefore, represent an attractive therapeutic strategy. The nematode Caenorhabditis elegans offers a strong model for post-reproductive mechanistic and therapeutic investigations, yet studies examining the mechanisms of, and countermeasures against, ageing decline largely intervene from larval stages onwards. Importantly, however, lifespan extending conditions frequently impair early-life fitness and fail to correspondingly increase healthspan. Here, we consolidate multiple evolutionary theories of ageing and discuss data supporting hyper/hypofunctional changes at a global molecular and functional level in C. elegans, and how classical lifespan-extension mutations alter these dynamics. The relevance of such mutant models for exploring mechanisms of ageing are discussed, highlighting that post-reproductive gene optimisation represents a more translatable approach for C. elegans research that is not constrained by evolutionary trade-offs. Where some genetic mutations in C. elegans that promote late-life health map accordingly with healthy ageing in humans, other widely used genetic mutations that extend worm lifespan are associated with life-limiting pathologies in people. Lifespan has also become the gold standard for quantifying 'ageing', but we argue that gerospan compression (i.e., 'healthier' ageing) is an appropriate goal for anti-ageing research, the mechanisms of which appear distinct from those regulating lifespan alone. There is, therefore, an evident need to re-evaluate experimental approaches to study the role of hyper/hypofunctional genes in ageing in C. elegans.
Collapse
Affiliation(s)
- Luke Slade
- University of Exeter Medical School, Exeter, UK.
| | - Timothy Etheridge
- Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Nathaniel J Szewczyk
- Ohio Musculoskeletal and Neurological Institute, Heritage College of Osteopathic Medicine, Athens, OH 45701, United States.
| |
Collapse
|
2
|
Coler-Reilly A, Pincus Z, Scheller EL, Civitelli R. Six drivers of aging identified among genes differentially expressed with age. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.02.606402. [PMID: 39149379 PMCID: PMC11326176 DOI: 10.1101/2024.08.02.606402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Many studies have compared gene expression in young and old samples to gain insights on aging, the primary risk factor for most major chronic diseases. However, these studies only describe associations, failing to distinguish drivers of aging from compensatory geroprotective responses and incidental downstream effects. Here, we introduce a workflow to characterize the causal effects of differentially expressed genes on lifespan. First, we performed a meta-analysis of 25 gene expression datasets comprising samples of various tissues from healthy, untreated adult mammals (humans, dogs, and rodents) at two distinct ages. We ranked each gene according to the number of distinct datasets in which the gene was differentially expressed with age in a consistent direction. The top age-upregulated genes were TMEM176A, EFEMP1, CP, and HLA-A; the top age-downregulated genes were CA4, SIAH, SPARC, and UQCR10. Second, the effects of the top ranked genes on lifespan were measured by applying post-developmental RNA interference of the corresponding ortholog in the nematode C. elegans (two trials, with roughly 100 animals per genotype per trial). Out of 10 age-upregulated and 9 age-downregulated genes that were tested, two age-upregulated genes (csp-3/CASP1 and spch-2/RSRC1) and four age-downregulated genes (C42C1.8/DIRC2, ost-1/SPARC, fzy-1/CDC20, and cah-3/CA4) produced significant and reproducible lifespan extension. Notably, the data do not suggest that the direction of differential expression with age is predictive of the effect on lifespan. Our study provides novel insight into the relationship between differential gene expression and aging phenotypes, pilots an unbiased workflow that can be easily repeated and expanded, and pinpoints six genes with evolutionarily conserved, causal roles in the aging process for further study.
Collapse
Affiliation(s)
- Ariella Coler-Reilly
- Division of Bone and Mineral Diseases, Musculoskeletal Research Center
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO, USA
| | - Zachary Pincus
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Erica L. Scheller
- Division of Bone and Mineral Diseases, Musculoskeletal Research Center
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Cell Biology and Physiology; Washington University School of Medicine, St. Louis, MO, USA
| | - Roberto Civitelli
- Division of Bone and Mineral Diseases, Musculoskeletal Research Center
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Cell Biology and Physiology; Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
3
|
Kumar A, Saha MK, Kumar V, Bhattacharya A, Barge S, Mukherjee AK, Kalita MC, Khan MR. Heat-killed probiotic Levilactobacillus brevis MKAK9 and its exopolysaccharide promote longevity by modulating aging hallmarks and enhancing immune responses in Caenorhabditis elegans. Immun Ageing 2024; 21:52. [PMID: 39095841 PMCID: PMC11295351 DOI: 10.1186/s12979-024-00457-w] [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: 05/15/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND Proteostasis is a critical aging hallmark responsible for removing damaged or misfolded proteins and their aggregates by improving proteasomal degradation through the autophagy-lysosome pathway (ALP) and the ubiquitin-proteasome system (UPS). Research on the impact of heat-killed probiotic bacteria and their structural components on aging hallmarks and innate immune responses is scarce, yet enhancing these effects could potentially delay age-related diseases. RESULTS This study introduces a novel heat-killed Levilactobacillus brevis strain MKAK9 (HK MKAK9), along with its exopolysaccharide (EPS), demonstrating their ability to extend longevity by improving proteostasis and immune responses in wild-type Caenorhabditis elegans. We elucidate the underlying mechanisms through a comprehensive approach involving mRNA- and small RNA sequencing, proteomic analysis, lifespan assays on loss-of-function mutants, and quantitative RT-PCR. Mechanistically, HK MKAK9 and its EPS resulted in downregulation of the insulin-like signaling pathway in a DAF-16-dependent manner, enhancing protein ubiquitination and subsequent proteasomal degradation through activation of the ALP pathway, which is partially mediated by microRNA mir-243. Importantly, autophagosomes engulf ubiquitinylated proteins, as evidenced by increased expression of the autophagy receptor sqst-3, and subsequently fuse with lysosomes, facilitated by increased levels of the lysosome-associated membrane protein (LAMP) lmp-1, suggesting the formation of autolysosomes for degradation of the selected cargo. Moreover, HK MKAK9 and its EPS activated the p38 MAPK pathway and its downstream SKN-1 transcription factor, which are known to regulate genes involved in innate immune response (thn-1, ilys-1, cnc-2, spp-9, spp-21, clec-47, and clec-266) and antioxidation (sod-3 and gst-44), thereby reducing the accumulation of reactive oxygen species (ROS) at both cellular and mitochondrial levels. Notably, SOD-3 emerged as a transcriptional target of both DAF-16 and SKN-1 transcription factors. CONCLUSION Our research sets a benchmark for future investigations by demonstrating that heat-killed probiotic and its specific cellular component, EPS, can downregulate the insulin-signaling pathway, potentially improving the autophagy-lysosome pathway (ALP) for degrading ubiquitinylated proteins and promoting organismal longevity. Additionally, we discovered that increased expression of microRNA mir-243 regulates insulin-like signaling and its downstream ALP pathway. Our findings also indicate that postbiotic treatment may bolster antioxidative and innate immune responses, offering a promising avenue for interventions in aging-related diseases.
Collapse
Affiliation(s)
- Arun Kumar
- Molecular Biology and Microbial Biotechnology Laboratory, Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Assam, Guwahati-781035, India
| | | | - Vipin Kumar
- Application Specialist, Research Business Cytiva, Gurugram, Haryana, India
| | - Anupam Bhattacharya
- Molecular Biology and Microbial Biotechnology Laboratory, Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Assam, Guwahati-781035, India
| | - Sagar Barge
- Molecular Biology and Microbial Biotechnology Laboratory, Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Assam, Guwahati-781035, India
| | - Ashis K Mukherjee
- Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Assam, Guwahati-781035, India
- Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Tezpur, Assam, 784028, India
| | - Mohan C Kalita
- Department of Biotechnology, Gauhati University, Guwahati, Assam, 781014, India
| | - Mojibur R Khan
- Molecular Biology and Microbial Biotechnology Laboratory, Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Assam, Guwahati-781035, India.
| |
Collapse
|
4
|
Rothi MH, Haddad JA, Sarkar GC, Mitchell W, Ying K, Pohl N, Sotomayor R, Natale J, Dellacono S, Gladyshev VN, Greer EL. The 18S rRNA Methyltransferase DIMT-1 Regulates Lifespan in the Germline Later in Life. RESEARCH SQUARE 2024:rs.3.rs-4421268. [PMID: 38946979 PMCID: PMC11213213 DOI: 10.21203/rs.3.rs-4421268/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Ribosome heterogeneity has emerged as an important regulatory control feature for determining which proteins are synthesized, however, the influence of age on ribosome heterogeneity is not fully understood. Whether mRNA transcripts are selectively translated in young versus old cells and whether dysregulation of this process drives organismal aging is unknown. Here we examined the role of ribosomal RNA (rRNA) methylation in maintaining appropriate translation as organisms age. In a directed RNAi screen, we identified the 18S rRNA N6'-dimethyl adenosine (m6,2A) methyltransferase, dimt-1, as a regulator of C. elegans lifespan and stress resistance. Lifespan extension induced by dimt-1 deficiency required a functional germline and was dependent on the known regulator of protein translation, the Rag GTPase, raga-1, which links amino acid sensing to the mechanistic target of rapamycin complex (mTORC)1. Using an auxin-inducible degron tagged version of dimt-1, we demonstrate that DIMT-1 functions in the germline after mid-life to regulate lifespan. We further found that knock-down of dimt-1 leads to selective translation of transcripts important for stress resistance and lifespan regulation in the C. elegans germline in mid-life including the cytochrome P450 daf-9, which synthesizes a steroid that signals from the germline to the soma to regulate lifespan. We found that dimt-1 induced lifespan extension was dependent on the daf-9 signaling pathway. This finding reveals a new layer of proteome dysfunction, beyond protein synthesis and degradation, as an important regulator of aging. Our findings highlight a new role for ribosome heterogeneity, and specific rRNA modifications, in maintaining appropriate translation later in life to promote healthy aging.
Collapse
Affiliation(s)
- M. Hafiz Rothi
- Department of Pediatrics, HMS Initiative for RNA Medicine, Harvard Medical School, Boston MA, USA
- Division of Newborn Medicine, Boston Children’s Hospital, Boston MA, USA
| | - Joseph Al Haddad
- Division of Newborn Medicine, Boston Children’s Hospital, Boston MA, USA
| | - Gautam Chandra Sarkar
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Wayne Mitchell
- Division of Genetics, Department of Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston MA 02115, USA
| | - Kejun Ying
- Division of Newborn Medicine, Boston Children’s Hospital, Boston MA, USA
| | - Nancy Pohl
- Division of Newborn Medicine, Boston Children’s Hospital, Boston MA, USA
| | - Roberto Sotomayor
- Division of Newborn Medicine, Boston Children’s Hospital, Boston MA, USA
| | - Julia Natale
- Division of Newborn Medicine, Boston Children’s Hospital, Boston MA, USA
| | - Scarlett Dellacono
- Department of Pediatrics, HMS Initiative for RNA Medicine, Harvard Medical School, Boston MA, USA
- Division of Newborn Medicine, Boston Children’s Hospital, Boston MA, USA
| | - Vadim N. Gladyshev
- Division of Genetics, Department of Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston MA 02115, USA
| | - Eric Lieberman Greer
- Department of Pediatrics, HMS Initiative for RNA Medicine, Harvard Medical School, Boston MA, USA
- Division of Newborn Medicine, Boston Children’s Hospital, Boston MA, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
5
|
Hafiz Rothi M, Sarkar GC, Haddad JA, Mitchell W, Ying K, Pohl N, Sotomayor-Mena RG, Natale J, Dellacono S, Gladyshev VN, Lieberman Greer E. The 18S rRNA Methyltransferase DIMT-1 Regulates Lifespan in the Germline Later in Life. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.594211. [PMID: 38798397 PMCID: PMC11118296 DOI: 10.1101/2024.05.14.594211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Ribosome heterogeneity has emerged as an important regulatory control feature for determining which proteins are synthesized, however, the influence of age on ribosome heterogeneity is not fully understood. Whether mRNA transcripts are selectively translated in young versus old cells and whether dysregulation of this process drives organismal aging is unknown. Here we examined the role of ribosomal RNA (rRNA) methylation in maintaining appropriate translation as organisms age. In a directed RNAi screen, we identified the 18S rRNA N6'-dimethyl adenosine (m6,2A) methyltransferase, dimt-1, as a regulator of C. elegans lifespan and stress resistance. Lifespan extension induced by dimt-1 deficiency required a functional germline and was dependent on the known regulator of protein translation, the Rag GTPase, raga-1, which links amino acid sensing to the mechanistic target of rapamycin complex (mTORC)1. Using an auxin-inducible degron tagged version of dimt-1, we demonstrate that DIMT-1 functions in the germline after mid-life to regulate lifespan. We further found that knock-down of dimt-1 leads to selective translation of transcripts important for stress resistance and lifespan regulation in the C. elegans germline in mid-life including the cytochrome P450 daf-9, which synthesizes a steroid that signals from the germline to the soma to regulate lifespan. We found that dimt-1 induced lifespan extension was dependent on the daf-9 signaling pathway. This finding reveals a new layer of proteome dysfunction, beyond protein synthesis and degradation, as an important regulator of aging. Our findings highlight a new role for ribosome heterogeneity, and specific rRNA modifications, in maintaining appropriate translation later in life to promote healthy aging.
Collapse
Affiliation(s)
- M. Hafiz Rothi
- Department of Pediatrics, HMS Initiative for RNA Medicine, Harvard Medical School, Boston MA, USA
- Division of Newborn Medicine, Boston Children’s Hospital, Boston MA, USA
| | - Gautam Chandra Sarkar
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Joseph Al Haddad
- Division of Newborn Medicine, Boston Children’s Hospital, Boston MA, USA
| | - Wayne Mitchell
- Division of Genetics, Department of Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston MA 02115, USA
| | - Kejun Ying
- Department of Pediatrics, HMS Initiative for RNA Medicine, Harvard Medical School, Boston MA, USA
- Division of Newborn Medicine, Boston Children’s Hospital, Boston MA, USA
| | - Nancy Pohl
- Department of Pediatrics, HMS Initiative for RNA Medicine, Harvard Medical School, Boston MA, USA
- Division of Newborn Medicine, Boston Children’s Hospital, Boston MA, USA
| | - Roberto G. Sotomayor-Mena
- Department of Pediatrics, HMS Initiative for RNA Medicine, Harvard Medical School, Boston MA, USA
- Division of Newborn Medicine, Boston Children’s Hospital, Boston MA, USA
| | - Julia Natale
- Division of Newborn Medicine, Boston Children’s Hospital, Boston MA, USA
| | - Scarlett Dellacono
- Department of Pediatrics, HMS Initiative for RNA Medicine, Harvard Medical School, Boston MA, USA
- Division of Newborn Medicine, Boston Children’s Hospital, Boston MA, USA
| | - Vadim N. Gladyshev
- Division of Genetics, Department of Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston MA 02115, USA
| | - Eric Lieberman Greer
- Department of Pediatrics, HMS Initiative for RNA Medicine, Harvard Medical School, Boston MA, USA
- Division of Newborn Medicine, Boston Children’s Hospital, Boston MA, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
6
|
Zang X, Wang Q, Zhang H, Zhang Y, Wang Z, Wu Z, Chen D. Knockdown of neuronal DAF-15/Raptor promotes healthy aging in C. elegans. J Genet Genomics 2024; 51:507-516. [PMID: 37951302 DOI: 10.1016/j.jgg.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 11/06/2023] [Accepted: 11/06/2023] [Indexed: 11/13/2023]
Abstract
The highly conserved target of rapamycin (TOR) pathway plays an important role in aging across species. Previous studies have established that inhibition of the TOR complex 1 (TORC1) significantly extends lifespan in Caenorhabditiselegans. However, it has not been clear whether TORC1 perturbation affects aging in a spatiotemporal manner. Here, we apply the auxin-inducible degradation tool to knock down endogenous DAF-15, the C. elegans ortholog of regulatory associated protein of TOR (Raptor), to characterize its roles in aging. Global or tissue-specific inhibition of DAF-15 during development results in various growth defects, whereas neuron-specific knockdown of DAF-15 during adulthood significantly extends lifespan and healthspan. The neuronal DAF-15 deficiency-induced longevity requires the intestinal activities of DAF-16/FOXO and PHA-4/FOXA transcription factors, as well as the AAK-2/AMP-activated protein kinase α catalytic subunit. Transcriptome profiling reveals that the neuronal DAF-15 knockdown promotes the expression of genes involved in protection. These findings define the tissue-specific roles of TORC1 in healthy aging and highlight the importance of neuronal modulation of aging.
Collapse
Affiliation(s)
- Xiao Zang
- Model Animal Research Center of Medical School, Nanjing University, Nanjing, Jiangsu 210061, China; Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Haining, Zhejiang 314400, China
| | - Qi Wang
- Model Animal Research Center of Medical School, Nanjing University, Nanjing, Jiangsu 210061, China; Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Haining, Zhejiang 314400, China
| | - Hanxin Zhang
- Model Animal Research Center of Medical School, Nanjing University, Nanjing, Jiangsu 210061, China
| | - Yiyan Zhang
- Model Animal Research Center of Medical School, Nanjing University, Nanjing, Jiangsu 210061, China; Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Haining, Zhejiang 314400, China
| | - Zi Wang
- Model Animal Research Center of Medical School, Nanjing University, Nanjing, Jiangsu 210061, China
| | - Zixing Wu
- Model Animal Research Center of Medical School, Nanjing University, Nanjing, Jiangsu 210061, China
| | - Di Chen
- Model Animal Research Center of Medical School, Nanjing University, Nanjing, Jiangsu 210061, China; Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Haining, Zhejiang 314400, China; Department of Colorectal Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
7
|
Rathor L, Curry S, Park Y, McElroy T, Robles B, Sheng Y, Chen WW, Min K, Xiao R, Lee MH, Han SM. Mitochondrial stress in GABAergic neurons non-cell autonomously regulates organismal health and aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.20.585932. [PMID: 38585797 PMCID: PMC10996468 DOI: 10.1101/2024.03.20.585932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Mitochondrial stress within the nervous system can trigger non-cell autonomous responses in peripheral tissues. However, the specific neurons involved and their impact on organismal aging and health have remained incompletely understood. Here, we demonstrate that mitochondrial stress in γ-aminobutyric acid-producing (GABAergic) neurons in Caenorhabditis elegans ( C. elegans ) is sufficient to significantly alter organismal lifespan, stress tolerance, and reproductive capabilities. This mitochondrial stress also leads to significant changes in mitochondrial mass, energy production, and levels of reactive oxygen species (ROS). DAF-16/FoxO activity is enhanced by GABAergic neuronal mitochondrial stress and mediates the induction of these non-cell-autonomous effects. Moreover, our findings indicate that GABA signaling operates within the same pathway as mitochondrial stress in GABAergic neurons, resulting in non-cell-autonomous alterations in organismal stress tolerance and longevity. In summary, these data suggest the crucial role of GABAergic neurons in detecting mitochondrial stress and orchestrating non-cell-autonomous changes throughout the organism.
Collapse
|
8
|
Sokolowski DJ, Vasquez OE, Wilson MD, Sokolowski MB, Anreiter I. Transcriptomic effects of the foraging gene shed light on pathways of pleiotropy and plasticity. Ann N Y Acad Sci 2023; 1526:99-113. [PMID: 37350250 DOI: 10.1111/nyas.15015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/24/2023]
Abstract
Genes are often pleiotropic and plastic in their expression, features which increase and diversify the functionality of the genome. The foraging (for) gene in Drosophila melanogaster is highly pleiotropic and a long-standing model for studying individual differences in behavior and plasticity from ethological, evolutionary, and genetic perspectives. Its pleiotropy is known to be linked to its complex molecular structure; however, the downstream pathways and interactors remain mostly elusive. To uncover these pathways and interactors and gain a better understanding of how pleiotropy and plasticity are achieved at the molecular level, we explore the effects of different for alleles on gene expression at baseline and in response to 4 h of food deprivation, using RNA sequencing analysis in different Drosophila larval tissues. The results show tissue-specific transcriptomic dynamics influenced by for allelic variation and food deprivation, as well as genotype by treatment interactions. Differentially expressed genes yielded pathways linked to previously described for phenotypes and several potentially novel phenotypes. Together, these findings provide putative genes and pathways through which for might regulate its varied phenotypes in a pleiotropic, plastic, and gene-structure-dependent manner.
Collapse
Affiliation(s)
- Dustin J Sokolowski
- Genetics and Genome Biology, SickKids Research Institute, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Oscar E Vasquez
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, Ontario, Canada
| | - Michael D Wilson
- Genetics and Genome Biology, SickKids Research Institute, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Marla B Sokolowski
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, Ontario, Canada
- Program in Child and Brain Development, Canadian Institute for Advanced Research, Toronto, Ontario, Canada
| | - Ina Anreiter
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
| |
Collapse
|
9
|
Vijay Kumar MJ, Morales R, Tsvetkov AS. G-quadruplexes and associated proteins in aging and Alzheimer's disease. FRONTIERS IN AGING 2023; 4:1164057. [PMID: 37323535 PMCID: PMC10267416 DOI: 10.3389/fragi.2023.1164057] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 05/17/2023] [Indexed: 06/17/2023]
Abstract
Aging is a prominent risk factor for many neurodegenerative disorders, such as Alzheimer's disease (AD). Alzheimer's disease is characterized by progressive cognitive decline, memory loss, and neuropsychiatric and behavioral symptoms, accounting for most of the reported dementia cases. This disease is now becoming a major challenge and burden on modern society, especially with the aging population. Over the last few decades, a significant understanding of the pathophysiology of AD has been gained by studying amyloid deposition, hyperphosphorylated tau, synaptic dysfunction, oxidative stress, calcium dysregulation, and neuroinflammation. This review focuses on the role of non-canonical secondary structures of DNA/RNA G-quadruplexes (G4s, G4-DNA, and G4-RNA), G4-binding proteins (G4BPs), and helicases, and their roles in aging and AD. Being critically important for cellular function, G4s are involved in the regulation of DNA and RNA processes, such as replication, transcription, translation, RNA localization, and degradation. Recent studies have also highlighted G4-DNA's roles in inducing DNA double-strand breaks that cause genomic instability and G4-RNA's participation in regulating stress granule formation. This review emphasizes the significance of G4s in aging processes and how their homeostatic imbalance may contribute to the pathophysiology of AD.
Collapse
Affiliation(s)
- M. J. Vijay Kumar
- The Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, TX, United States
| | - Rodrigo Morales
- The Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, TX, United States
- Centro Integrativo de Biologia y Quimica Aplicada (CIBQA), Universidad Bernardo O’Higgins, Santiago, Chile
| | - Andrey S. Tsvetkov
- The Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, TX, United States
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX, United States
- UTHealth Consortium on Aging, The University of Texas McGovern Medical School, Houston, TX, United States
| |
Collapse
|
10
|
Kim HS, Pickering AM. Protein translation paradox: Implications in translational regulation of aging. Front Cell Dev Biol 2023; 11:1129281. [PMID: 36711035 PMCID: PMC9880214 DOI: 10.3389/fcell.2023.1129281] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/02/2023] [Indexed: 01/15/2023] Open
Abstract
Protein translation is an essential cellular process playing key roles in growth and development. Protein translation declines over the course of age in multiple animal species, including nematodes, fruit flies, mice, rats, and even humans. In all these species, protein translation transiently peaks in early adulthood with a subsequent drop over the course of age. Conversely, lifelong reductions in protein translation have been found to extend lifespan and healthspan in multiple animal models. These findings raise the protein synthesis paradox: age-related declines in protein synthesis should be detrimental, but life-long reductions in protein translation paradoxically slow down aging and prolong lifespan. This article discusses the nature of this paradox and complies an extensive body of work demonstrating protein translation as a modulator of lifespan and healthspan.
Collapse
Affiliation(s)
- Harper S. Kim
- Center for Neurodegeneration and Experimental Therapeutics (CNET), Department of Neurology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Medical Scientist Training Program, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Andrew M. Pickering
- Center for Neurodegeneration and Experimental Therapeutics (CNET), Department of Neurology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
11
|
Silencing of the mitochondrial ribosomal protein L-24 gene activates the oxidative stress response in Caenorhabditis elegans. Biochim Biophys Acta Gen Subj 2023; 1867:130255. [PMID: 36265765 DOI: 10.1016/j.bbagen.2022.130255] [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: 05/20/2022] [Revised: 10/09/2022] [Accepted: 10/12/2022] [Indexed: 11/13/2022]
Abstract
The mitochondrial translation machinery allows the synthesis of the mitochondrial-encoded subunits of the electron transport chain. Defects in this process lead to mitochondrial physiology failure; in humans, they are associated with early-onset, extremely variable and often fatal disorder. The use of a simple model to study the mitoribosomal defects is mandatory to overcome the difficulty to analyze the impact of pathological mutations in humans. In this paper we study in nematode Caenorhabditis elegans the silencing effect of the mrpl-24 gene, coding for the mitochondrial ribosomal protein L-24 (MRPL-24). This is a structural protein of the large subunit 39S of the mitoribosome and its effective physiological function is not completely elucidated. We have evaluated the nematode's fitness fault and investigated the mitochondrial defects associated with MRPL-24 depletion. The oxidative stress response activation due to the mitochondrial alteration has been also investigated as a compensatory physiological mechanism. For the first time, we demonstrated that MRPL-24 reduction increases the expression of detoxifying enzymes such as SOD-3 and GST-4 through the involvement of transcription factor SKN-1. BACKGROUND In humans, mutations in genes encoding mitochondrial ribosomal proteins (MRPs) often cause early-onset, severe, fatal and extremely variable clinical defects. Mitochondrial ribosomal protein L-24 (MRPL24) is a structural protein of the large subunit 39S of the mitoribosome. It is highly conserved in different species and its effective physiological function is not completely elucidated. METHODS We characterized the MRPL24 functionality using the animal model Caenorhabditis elegans. We performed the RNA mediated interference (RNAi) by exposing the nematodes' embryos to double-stranded RNA (dsRNA) specific for the MRPL-24 coding sequence. We investigated for the first time in C. elegans, the involvement of the MRPL-24 on the nematode's fitness and its mitochondrial physiology. RESULTS Mrpl-24 silencing in C. elegans negatively affected the larval development, progeny production and body bending. The analysis of mitochondrial functionality revealed loss of mitochondrial network and impairment of mitochondrial functionality, as the decrease of oxygen consumption rate and the ROS production, as well as reduction of mitochondrial protein synthesis. Finally, the MRPL-24 depletion activated the oxidative stress response, increasing the expression levels of two detoxifying enzymes, SOD-3 and GST-4. CONCLUSIONS In C. elegans the MRPL-24 depletion activated the oxidative stress response. This appears as a compensatory mechanism to the alteration of the mitochondrial functionality and requires the involvement of transcription factor SKN-1. GENERAL SIGNIFICANCE C. elegans resulted in a good model for the study of mitochondrial disorders and its use as a simple and pluricellular organism could open interesting perspectives to better investigate the pathologic mechanisms underlying these devastating diseases.
Collapse
|
12
|
Zheng T, Luo Q, Han C, Zhou J, Gong J, Chun L, Xu XZS, Liu J. Cytoplasmic and mitochondrial aminoacyl-tRNA synthetases differentially regulate lifespan in Caenorhabditis elegans. iScience 2022; 25:105266. [PMID: 36304099 PMCID: PMC9593246 DOI: 10.1016/j.isci.2022.105266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/21/2022] [Accepted: 09/29/2022] [Indexed: 11/12/2022] Open
Abstract
Reducing the rate of translation promotes longevity in multiple organisms, representing a conserved mechanism for lifespan extension. Aminoacyl-tRNA synthetases (ARSs) catalyze the loading of amino acids to their cognate tRNAs, thereby playing an essential role in translation. Mutations in ARS genes are associated with various human diseases. However, little is known about the role of ARSs in aging, particularly whether and how these genes regulate lifespan. Here, using Caenorhabditis elegans as a model, we systematically characterized the role of all three types of ARS genes in lifespan regulation, including mitochondrial, cytoplasmic, and cyto-mito bifunctional ARS genes. We found that, as expected, RNAi knockdown of mitochondrial ARS genes extended lifespan. Surprisingly, knocking down cytoplasmic or cyto-mito bifunctional ARS genes shortened lifespan, though such treatment reduced the rate of translation. These results reveal opposing roles of mitochondrial and cytoplasmic ARSs in lifespan regulation, demonstrating that inhibiting translation may not always extend lifespan. RNAi knockdown of mitochondrial ARS genes extends lifespan via UPRmt RNAi knockdown of cytoplasmic or cyto-mito bifunctional ARS genes shortens lifespan Inhibiting translation may not always extend lifespan
Collapse
Affiliation(s)
- Tianlin Zheng
- College of Life Science and Technology, Key Laboratory of Molecular Biophysics of MOE, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Qiang Luo
- College of Life Science and Technology, Key Laboratory of Molecular Biophysics of MOE, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Chengxuan Han
- College of Life Science and Technology, Key Laboratory of Molecular Biophysics of MOE, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Jiejun Zhou
- College of Life Science and Technology, Key Laboratory of Molecular Biophysics of MOE, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Jianke Gong
- College of Life Science and Technology, Key Laboratory of Molecular Biophysics of MOE, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China.,Life Sciences Institute and Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lei Chun
- College of Life Science and Technology, Key Laboratory of Molecular Biophysics of MOE, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - X Z Shawn Xu
- Life Sciences Institute and Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jianfeng Liu
- College of Life Science and Technology, Key Laboratory of Molecular Biophysics of MOE, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| |
Collapse
|
13
|
Mukherjee P, Panda P, Kasturi P. A comparative meta-analysis of membraneless organelle-associated proteins with age related proteome of C. elegans. Cell Stress Chaperones 2022; 27:619-631. [PMID: 36169889 PMCID: PMC9672229 DOI: 10.1007/s12192-022-01299-5] [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: 04/18/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 01/25/2023] Open
Abstract
Proteome imbalance can lead to protein misfolding and aggregation which is associated with pathologies. Protein aggregation can also be an active, organized process and can be exploited by cells as a survival strategy. In adverse conditions, it is beneficial to deposit the proteins in a condensate rather degrading and resynthesizing. Membraneless organelles (MLOs) are biological condensates formed through liquid-liquid phase separation (LLPS), involving cellular components such as nucleic acids and proteins. LLPS is a regulated process, which when perturbed, can undergo a transition from a physiological liquid condensate to pathological solid-like protein aggregates. To understand how the MLO-associated proteins (MLO-APs) behave during aging, we performed a comparative meta-analysis with age-related proteome of C. elegans. We found that the MLO-APs are highly abundant throughout the lifespan in wild-type and long-lived daf-2 mutant animals. Interestingly, they are aggregating more in long-lived mutant animals compared to the age matched wild-type and short-lived daf-16 and hsf-1 mutant animals. GO term analysis revealed that the cell cycle and embryonic development are among the top enriched processes in addition to RNP components in aggregated proteome. Considering antagonistic pleotropic nature of these developmental genes and post mitotic status of C. elegans, we assume that these proteins phase transit during post development. As the organism ages, these MLO-APs either mature to become more insoluble or dissolve in uncontrolled manner. However, in the long-lived daf-2 mutant animals, the MLOs may attain protective states due to extended availability and association of molecular chaperones.
Collapse
Affiliation(s)
- Pritam Mukherjee
- BioX Centre, School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Kamand, Himachal Pradesh, 175005, India
| | - Prajnadipta Panda
- BioX Centre, School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Kamand, Himachal Pradesh, 175005, India
| | - Prasad Kasturi
- BioX Centre, School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Kamand, Himachal Pradesh, 175005, India.
| |
Collapse
|
14
|
Javidnia S, Cranwell S, Mueller SH, Selman C, Tullet JM, Kuchenbaecker K, Alic N. Mendelian randomization analyses implicate biogenesis of translation machinery in human aging. Genome Res 2022; 32:258-265. [PMID: 35078808 PMCID: PMC8805714 DOI: 10.1101/gr.275636.121] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 12/15/2021] [Indexed: 11/25/2022]
Abstract
Reduced provision of protein translation machinery promotes healthy aging in a number of animal models. In humans, however, inborn impairments in translation machinery are a known cause of several developmental disorders, collectively termed ribosomopathies. Here, we use casual inference approaches in genetic epidemiology to investigate whether adult, tissue-specific biogenesis of translation machinery drives human aging. We assess naturally occurring variation in the expression of genes encoding subunits specific to the two RNA polymerases (Pols) that transcribe ribosomal and transfer RNAs, namely Pol I and III, and the variation in expression of ribosomal protein (RP) genes, using Mendelian randomization. We find each causally associated with human longevity (β = −0.15 ± 0.047, P = 9.6 × 10−4, q = 0.015; β = −0.13 ± 0.040, P = 1.4 × 10−3, q = 0.023; β = −0.048 ± 0.016, P = 3.5 × 10−3, q = 0.056, respectively), and this does not appear to be mediated by altered susceptibility to a single disease. We find that reduced expression of Pol III, RPs, or Pol I promotes longevity from different organs, namely visceral adipose, liver, and skeletal muscle, echoing the tissue specificity of ribosomopathies. Our study shows the utility of leveraging genetic variation in expression to elucidate how essential cellular processes impact human aging. The findings extend the evolutionary conservation of protein synthesis as a critical process that drives animal aging to include humans.
Collapse
|
15
|
Wilson KA, Chamoli M, Hilsabeck TA, Pandey M, Bansal S, Chawla G, Kapahi P. Evaluating the beneficial effects of dietary restrictions: A framework for precision nutrigeroscience. Cell Metab 2021; 33:2142-2173. [PMID: 34555343 PMCID: PMC8845500 DOI: 10.1016/j.cmet.2021.08.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/17/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022]
Abstract
Dietary restriction (DR) has long been viewed as the most robust nongenetic means to extend lifespan and healthspan. Many aging-associated mechanisms are nutrient responsive, but despite the ubiquitous functions of these pathways, the benefits of DR often vary among individuals and even among tissues within an individual, challenging the aging research field. Furthermore, it is often assumed that lifespan interventions like DR will also extend healthspan, which is thus often ignored in aging studies. In this review, we provide an overview of DR as an intervention and discuss the mechanisms by which it affects lifespan and various healthspan measures. We also review studies that demonstrate exceptions to the standing paradigm of DR being beneficial, thus raising new questions that future studies must address. We detail critical factors for the proposed field of precision nutrigeroscience, which would utilize individualized treatments and predict outcomes using biomarkers based on genotype, sex, tissue, and age.
Collapse
Affiliation(s)
| | - Manish Chamoli
- The Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Tyler A Hilsabeck
- The Buck Institute for Research on Aging, Novato, CA 94945, USA; Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Manish Pandey
- Regional Centre for Biotechnology, Faridabad, Haryana 121001, India
| | - Sakshi Bansal
- Regional Centre for Biotechnology, Faridabad, Haryana 121001, India
| | - Geetanjali Chawla
- Regional Centre for Biotechnology, Faridabad, Haryana 121001, India.
| | - Pankaj Kapahi
- The Buck Institute for Research on Aging, Novato, CA 94945, USA; Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
16
|
Knapp-Wilson A, Pereira GC, Buzzard E, Ford HC, Richardson A, Corey RA, Neal C, Verkade P, Halestrap AP, Gold VAM, Kuwabara PE, Collinson I. Maintenance of complex I and its supercomplexes by NDUF-11 is essential for mitochondrial structure, function and health. J Cell Sci 2021; 134:jcs258399. [PMID: 34106255 PMCID: PMC8277142 DOI: 10.1242/jcs.258399] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 05/28/2021] [Indexed: 12/22/2022] Open
Abstract
Mitochondrial supercomplexes form around a conserved core of monomeric complex I and dimeric complex III; wherein a subunit of the former, NDUFA11, is conspicuously situated at the interface. We identified nduf-11 (B0491.5) as encoding the Caenorhabditis elegans homologue of NDUFA11. Animals homozygous for a CRISPR-Cas9-generated knockout allele of nduf-11 arrested at the second larval (L2) development stage. Reducing (but not eliminating) expression using RNAi allowed development to adulthood, enabling characterisation of the consequences: destabilisation of complex I and its supercomplexes and perturbation of respiratory function. The loss of NADH dehydrogenase activity was compensated by enhanced complex II activity, with the potential for detrimental reactive oxygen species (ROS) production. Cryo-electron tomography highlighted aberrant morphology of cristae and widening of both cristae junctions and the intermembrane space. The requirement of NDUF-11 for balanced respiration, mitochondrial morphology and development presumably arises due to its involvement in complex I and supercomplex maintenance. This highlights the importance of respiratory complex integrity for health and the potential for its perturbation to cause mitochondrial disease. This article has an associated First Person interview with Amber Knapp-Wilson, joint first author of the paper.
Collapse
Affiliation(s)
| | | | - Emma Buzzard
- Living Systems Institute, Stocker Road, University of Exeter, Exeter EX4 4QD, UK
- College of Life and Environmental Sciences,Geoffrey Pope Building, University of Exeter, Stocker Road, Exeter EX4 4QD, UK
| | - Holly C. Ford
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | | | - Robin A. Corey
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| | - Chris Neal
- Wolfson Bioimaging Facility, Faculty of Life Sciences, University of Bristol, Bristol BS8 1TD, UK
| | - Paul Verkade
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | | | - Vicki A. M. Gold
- Living Systems Institute, Stocker Road, University of Exeter, Exeter EX4 4QD, UK
- College of Life and Environmental Sciences,Geoffrey Pope Building, University of Exeter, Stocker Road, Exeter EX4 4QD, UK
| | | | - Ian Collinson
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| |
Collapse
|
17
|
Sokolov AS, Nekrasov PV, Shaposhnikov MV, Moskalev AA. Hydrogen sulfide in longevity and pathologies: Inconsistency is malodorous. Ageing Res Rev 2021; 67:101262. [PMID: 33516916 DOI: 10.1016/j.arr.2021.101262] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/18/2021] [Accepted: 01/24/2021] [Indexed: 02/08/2023]
Abstract
Hydrogen sulfide (H2S) is one of the biologically active gases (gasotransmitters), which plays an important role in various physiological processes and aging. Its production in the course of methionine and cysteine catabolism and its degradation are finely balanced, and impairment of H2S homeostasis is associated with various pathologies. Despite the strong geroprotective action of exogenous H2S in C. elegans, there are controversial effects of hydrogen sulfide and its donors on longevity in other models, as well as on stress resistance, age-related pathologies and aging processes, including regulation of senescence-associated secretory phenotype (SASP) and senescent cell anti-apoptotic pathways (SCAPs). Here we discuss that the translation potential of H2S as a geroprotective compound is influenced by a multiplicity of its molecular targets, pleiotropic biological effects, and the overlapping ranges of toxic and beneficial doses. We also consider the challenges of the targeted delivery of H2S at the required dose. Along with this, the complexity of determining the natural levels of H2S in animal and human organs and their ambiguous correlations with longevity are reviewed.
Collapse
|
18
|
Wu D, Cai W, Zhang X, Lan J, Zou L, Chen SJ, Wu Z, Chen D. Inhibition of PAR-1 delays aging via activating AMPK in C. elegans. Aging (Albany NY) 2020; 12:25700-25717. [PMID: 33232266 PMCID: PMC7803586 DOI: 10.18632/aging.104180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023]
Abstract
The antagonistic pleiotropy theory of aging suggests that genes essential for growth and development are likely to modulate aging later in life. Previous studies in C. elegans demonstrate that inhibition of certain developmentally essential genes during adulthood leads to significant lifespan extension. PAR-1, a highly conserved serine/threonine kinase, functions as a key cellular polarity regulator during the embryonic development. However, the role of PAR-1 during adulthood remains unknown. Here we show that inhibition of par-1 either by a temperature-sensitive mutant or by RNAi knockdown only during adulthood is sufficient to extend lifespan in C. elegans. Inhibition of par-1 also improves healthspan, as indicated by increased stress resistance, enhanced proteotoxicity resistance, as well as reduced muscular function decline over time. Additionally, tissue-enriched RNAi knockdown analysis reveals that PAR-1 mainly functions in the epidermis to regulate lifespan. Further genetic epistatic and molecular studies demonstrate that the effect of par-1 on lifespan requires the AMP-activated protein kinase (AMPK), and RNAi knockdown of par-1 results in age-dependent AMPK activation and reduced lipid accumulation in the metabolic tissue. Taken together, our findings reveal a previously undescribed function of PAR-1 in adulthood, which will help to understand the molecular links between development and aging.
Collapse
Affiliation(s)
- Di Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center of Medical School, Nanjing University, Pukou, Nanjing 210061, Jiangsu, China
| | - Waijiao Cai
- Institute of Traditional Chinese and Western Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xuan Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center of Medical School, Nanjing University, Pukou, Nanjing 210061, Jiangsu, China
| | - Jianfeng Lan
- Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Lina Zou
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center of Medical School, Nanjing University, Pukou, Nanjing 210061, Jiangsu, China
| | - Samuel J Chen
- Department of Mechanical, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Zixing Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center of Medical School, Nanjing University, Pukou, Nanjing 210061, Jiangsu, China
| | - Di Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center of Medical School, Nanjing University, Pukou, Nanjing 210061, Jiangsu, China
| |
Collapse
|
19
|
Carolina de Souza-Guerreiro T, Meng X, Dacheux E, Firczuk H, McCarthy J. Translational control of gene expression noise and its relationship to ageing in yeast. FEBS J 2020; 288:2278-2293. [PMID: 33090724 DOI: 10.1111/febs.15594] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 09/30/2020] [Accepted: 10/07/2020] [Indexed: 11/29/2022]
Abstract
Gene expression noise influences organism evolution and fitness but is poorly understood. There is increasing evidence that the functional roles of components of the translation machinery influence noise intensity. In addition, modulation of the activities of at least some of these same components affects the replicative lifespan of a broad spectrum of organisms. In a novel comparative approach, we modulate the activities of the translation initiation factors eIFG1 and eIF4G2, both of which are involved in the process of recruiting ribosomal 43S preinitiation complexes to the 5' end of eukaryotic mRNAs. We show that tagging of the cell wall using a fluorescent dye allows us to follow gene expression noise as different yeast strains progress through successive cycles of replicative ageing. This procedure reveals a relationship between global protein synthesis rate and gene expression noise (cell-to-cell heterogeneity), which is accompanied by a parallel correlation between gene expression noise and the replicative age of mother cells. An alternative approach, based on microfluidics, confirms the interdependence between protein synthesis rate, gene expression noise and ageing. We additionally show that it is important to characterize the influence of the design of the microfluidic device on the nutritional state of the cells during such experiments. Analysis of the noise data derived from flow cytometry and fluorescence microscopy measurements indicates that both the intrinsic and the extrinsic noise components increase as a function of ageing.
Collapse
Affiliation(s)
| | - Xiang Meng
- Warwick Integrative Synthetic Biology Centre (WISB) and School of Life Sciences, University of Warwick, Coventry, UK
| | - Estelle Dacheux
- Warwick Integrative Synthetic Biology Centre (WISB) and School of Life Sciences, University of Warwick, Coventry, UK
| | - Helena Firczuk
- Warwick Integrative Synthetic Biology Centre (WISB) and School of Life Sciences, University of Warwick, Coventry, UK
| | - John McCarthy
- Warwick Integrative Synthetic Biology Centre (WISB) and School of Life Sciences, University of Warwick, Coventry, UK
| |
Collapse
|
20
|
Skariah G, Todd PK. Translational control in aging and neurodegeneration. WILEY INTERDISCIPLINARY REVIEWS-RNA 2020; 12:e1628. [PMID: 32954679 DOI: 10.1002/wrna.1628] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/19/2020] [Accepted: 09/07/2020] [Indexed: 12/13/2022]
Abstract
Protein metabolism plays central roles in age-related decline and neurodegeneration. While a large body of research has explored age-related changes in protein degradation, alterations in the efficiency and fidelity of protein synthesis with aging are less well understood. Age-associated changes occur in both the protein synthetic machinery (ribosomal proteins and rRNA) and within regulatory factors controlling translation. At the same time, many of the interventions that prolong lifespan do so in part by pre-emptively decreasing protein synthesis rates to allow better harmonization to age-related declines in protein catabolism. Here we review the roles of translation regulation in aging, with a specific focus on factors implicated in age-related neurodegeneration. We discuss how emerging technologies such as ribosome profiling and superior mass spectrometric approaches are illuminating age-dependent mRNA-specific changes in translation rates across tissues to reveal a critical interplay between catabolic and anabolic pathways that likely contribute to functional decline. These new findings point to nodes in posttranscriptional gene regulation that both contribute to aging and offer targets for therapy. This article is categorized under: Translation > Translation Regulation Translation > Ribosome Biogenesis Translation > Translation Mechanisms.
Collapse
Affiliation(s)
- Geena Skariah
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Peter K Todd
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
- Ann Arbor VA Healthcare System, Department of Veterans Affairs, Ann Arbor, Michigan, USA
| |
Collapse
|
21
|
Bjedov I, Rallis C. The Target of Rapamycin Signalling Pathway in Ageing and Lifespan Regulation. Genes (Basel) 2020; 11:E1043. [PMID: 32899412 PMCID: PMC7565554 DOI: 10.3390/genes11091043] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 08/28/2020] [Accepted: 08/30/2020] [Indexed: 12/11/2022] Open
Abstract
Ageing is a complex trait controlled by genes and the environment. The highly conserved mechanistic target of rapamycin signalling pathway (mTOR) is a major regulator of lifespan in all eukaryotes and is thought to be mediating some of the effects of dietary restriction. mTOR is a rheostat of energy sensing diverse inputs such as amino acids, oxygen, hormones, and stress and regulates lifespan by tuning cellular functions such as gene expression, ribosome biogenesis, proteostasis, and mitochondrial metabolism. Deregulation of the mTOR signalling pathway is implicated in multiple age-related diseases such as cancer, neurodegeneration, and auto-immunity. In this review, we briefly summarise some of the workings of mTOR in lifespan and ageing through the processes of transcription, translation, autophagy, and metabolism. A good understanding of the pathway's outputs and connectivity is paramount towards our ability for genetic and pharmacological interventions for healthy ageing and amelioration of age-related disease.
Collapse
Affiliation(s)
- Ivana Bjedov
- UCL Cancer Institute, Paul O’Gorman Building, 72 Huntley Street, London WC1E 6DD, UK
| | - Charalampos Rallis
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, UK
| |
Collapse
|
22
|
Lan J, Rollins JA, Zang X, Wu D, Zou L, Wang Z, Ye C, Wu Z, Kapahi P, Rogers AN, Chen D. Translational Regulation of Non-autonomous Mitochondrial Stress Response Promotes Longevity. Cell Rep 2020; 28:1050-1062.e6. [PMID: 31340143 PMCID: PMC6684276 DOI: 10.1016/j.celrep.2019.06.078] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 06/04/2019] [Accepted: 06/21/2019] [Indexed: 01/12/2023] Open
Abstract
Reduced mRNA translation delays aging, but the underlying mechanisms remain underexplored. Mutations in both DAF-2 (IGF-1 receptor) and RSKS-1 (ribosomal S6 kinase/S6K) cause synergistic lifespan extension in C. elegans. To understand the roles of translational regulation in this process, we performed polysomal profiling and identified translationally regulated ribosomal and cytochrome c (CYC-2.1) genes as key mediators of longevity. cyc-2.1 knockdown significantly extends lifespan by activating the intestinal mitochondrial unfolded protein response (UPRmt), mitochondrial fission, and AMP-activated kinase (AMPK). The germline serves as the key tissue for cyc-2.1 to regulate lifespan, and germline-specific cyc-2.1 knockdown non-autonomously activates intestinal UPRmt and AMPK. Furthermore, the RNA-binding protein GLD-1-mediated translational repression of cyc-2.1 in the germline is important for the non-autonomous activation of UPRmt and synergistic longevity of the daf-2 rsks-1 mutant. Altogether, these results illustrate a translationally regulated non-autonomous mitochondrial stress response mechanism in the modulation of lifespan by insulin-like signaling and S6K. To understand how reduced translation delays aging, Lan et al. perform translational profiling in C. elegans and propose that, in the significantly long-lived daf-2 rsks-1 mutant, serial translational regulation leads to reduced cytochrome c in the germline, which non-autonomously activates UPRmt and AMPK in the metabolic tissue to ensure longevity.
Collapse
Affiliation(s)
- Jianfeng Lan
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, 12 Xuefu Rd, Pukou, Nanjing, Jiangsu 210061, China
| | - Jarod A Rollins
- MDI Biological Laboratory, 159 Old Bar Harbor Rd., Salisbury Cove, ME 04672, USA
| | - Xiao Zang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, 12 Xuefu Rd, Pukou, Nanjing, Jiangsu 210061, China
| | - Di Wu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, 12 Xuefu Rd, Pukou, Nanjing, Jiangsu 210061, China
| | - Lina Zou
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, 12 Xuefu Rd, Pukou, Nanjing, Jiangsu 210061, China
| | - Zi Wang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, 12 Xuefu Rd, Pukou, Nanjing, Jiangsu 210061, China
| | - Chang Ye
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, 12 Xuefu Rd, Pukou, Nanjing, Jiangsu 210061, China
| | - Zixing Wu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, 12 Xuefu Rd, Pukou, Nanjing, Jiangsu 210061, China
| | - Pankaj Kapahi
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA.
| | - Aric N Rogers
- MDI Biological Laboratory, 159 Old Bar Harbor Rd., Salisbury Cove, ME 04672, USA.
| | - Di Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, 12 Xuefu Rd, Pukou, Nanjing, Jiangsu 210061, China.
| |
Collapse
|
23
|
Heras J, Aguilar A. Comparative Transcriptomics Reveals Patterns of Adaptive Evolution Associated with Depth and Age Within Marine Rockfishes (Sebastes). J Hered 2020; 110:340-350. [PMID: 30602025 DOI: 10.1093/jhered/esy070] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 12/31/2018] [Indexed: 01/21/2023] Open
Abstract
The genetic underpinnings that contribute to ecological adaptation and speciation are not completely understood, especially within marine ecosystems. These evolutionary processes can be elucidated by studying adaptive radiations, because they provide replicates of divergence within a given environment or time-frame. Marine rockfishes (genus Sebastes) are an adaptive radiation and unique model system for studying adaptive evolution in the marine realm. We investigated molecular evolution associated with ecological (depth) and life history (lifespan) divergence in 2 closely related clades of Sebastes. Brain transcriptomes were sequenced via RNA-Seq from 3 species within the subgenus Pteropodus and a pair of related congeners from the subgenus Sebastosomus in order to identify patterns of adaptive evolution. De novo assemblies from these transcriptomes were used to identify 3867 orthologous clusters, and genes subject to positive selection were identified based on all 5 species, depth, and lifespan. Within all our analyses, we identified hemoglobin subunit α to be under strong positive selection and is associated with the depth of occurrence. In our lifespan analysis we identified immune function genes under positive selection in association with maximum lifespan. This study provides insight on the molecular evolution of rockfishes and these candidate genes may provide a better understanding of how these subgenera radiated within the Northeast Pacific.
Collapse
Affiliation(s)
- Joseph Heras
- School of Natural Sciences and Graduate Group in Quantitative and Systems Biology, University of California, Merced, CA
| | - Andres Aguilar
- School of Natural Sciences and Graduate Group in Quantitative and Systems Biology, University of California, Merced, CA
| |
Collapse
|
24
|
Molenaars M, Daniels EG, Meurs A, Janssens GE, Houtkooper RH. Mitochondrial cross-compartmental signalling to maintain proteostasis and longevity. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190414. [PMID: 32362258 DOI: 10.1098/rstb.2019.0414] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Lifespan in eukaryotic species can be prolonged by shifting from cellular states favouring growth to those favouring maintenance and stress resistance. For instance, perturbations in mitochondrial oxidative phosphorylation (OXPHOS) can shift cells into this latter state and extend lifespan. Because mitochondria rely on proteins synthesized from nuclear as well as mitochondrial DNA, they need to constantly send and receive messages from other compartments of the cell in order to function properly and maintain homeostasis, and lifespan extension is often dependent on this cross-compartmental signalling. Here, we describe the mechanisms of bi-directional mitochondrial cross-compartmental signalling resulting in proteostasis and longevity. These proteostasis mechanisms are highly context-dependent, governed by the origin and extent of stress. Furthermore, we discuss the translatability of these mechanisms and explore therapeutic developments, such as the antibiotic studies targeting mitochondria or mitochondria-derived peptides as therapies for age-related diseases such as neurodegeneration and cancer. This article is part of the theme issue 'Retrograde signalling from endosymbiotic organelles'.
Collapse
Affiliation(s)
- Marte Molenaars
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Eileen G Daniels
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Amber Meurs
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Georges E Janssens
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Mechanisms of Lifespan Regulation by Calorie Restriction and Intermittent Fasting in Model Organisms. Nutrients 2020; 12:nu12041194. [PMID: 32344591 PMCID: PMC7230387 DOI: 10.3390/nu12041194] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/19/2020] [Accepted: 04/22/2020] [Indexed: 12/18/2022] Open
Abstract
Genetic and pharmacological interventions have successfully extended healthspan and lifespan in animals, but their genetic interventions are not appropriate options for human applications and pharmacological intervention needs more solid clinical evidence. Consequently, dietary manipulations are the only practical and probable strategies to promote health and longevity in humans. Caloric restriction (CR), reduction of calorie intake to a level that does not compromise overall health, has been considered as being one of the most promising dietary interventions to extend lifespan in humans. Although it is straightforward, continuous reduction of calorie or food intake is not easy to practice in real lives of humans. Recently, fasting-related interventions such as intermittent fasting (IF) and time-restricted feeding (TRF) have emerged as alternatives of CR. Here, we review the history of CR and fasting-related strategies in animal models, discuss the molecular mechanisms underlying these interventions, and propose future directions that can fill the missing gaps in the current understanding of these dietary interventions. CR and fasting appear to extend lifespan by both partially overlapping common mechanisms such as the target of rapamycin (TOR) pathway and circadian clock, and distinct independent mechanisms that remain to be discovered. We propose that a systems approach combining global transcriptomic, metabolomic, and proteomic analyses followed by genetic perturbation studies targeting multiple candidate pathways will allow us to better understand how CR and fasting interact with each other to promote longevity.
Collapse
|
26
|
Anisimova AS, Alexandrov AI, Makarova NE, Gladyshev VN, Dmitriev SE. Protein synthesis and quality control in aging. Aging (Albany NY) 2019; 10:4269-4288. [PMID: 30562164 PMCID: PMC6326689 DOI: 10.18632/aging.101721] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 12/10/2018] [Indexed: 12/22/2022]
Abstract
Aging is characterized by the accumulation of damage and other deleterious changes, leading to the loss of functionality and fitness. Age-related changes occur at most levels of organization of a living organism (molecular, organellar, cellular, tissue and organ). However, protein synthesis is a major biological process, and thus understanding how it changes with age is of paramount importance. Here, we discuss the relationships between lifespan, aging, protein synthesis and translational control, and expand this analysis to the various aspects of proteome behavior in organisms with age. Characterizing the consequences of changes in protein synthesis and translation fidelity, and determining whether altered translation is pathological or adaptive is necessary for understanding the aging process, as well as for developing approaches to target dysfunction in translation as a strategy for extending lifespan.
Collapse
Affiliation(s)
- Aleksandra S Anisimova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia.,School of Bioengineering and Bioinformatics Lomonosov Moscow State University, Moscow 119234, Russia
| | - Alexander I Alexandrov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia.,Bach Institute of Biochemistry of the Russian Academy of Sciences, Moscow 119071, Russia
| | - Nadezhda E Makarova
- School of Bioengineering and Bioinformatics Lomonosov Moscow State University, Moscow 119234, Russia
| | - Vadim N Gladyshev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia.,Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Sergey E Dmitriev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia.,School of Bioengineering and Bioinformatics Lomonosov Moscow State University, Moscow 119234, Russia.,Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia
| |
Collapse
|
27
|
Medkour Y, Mohammad K, Arlia-Ciommo A, Svistkova V, Dakik P, Mitrofanova D, Rodriguez MEL, Junio JAB, Taifour T, Escudero P, Goltsios FF, Soodbakhsh S, Maalaoui H, Simard É, Titorenko VI. Mechanisms by which PE21, an extract from the white willow Salix alba, delays chronological aging in budding yeast. Oncotarget 2019; 10:5780-5816. [PMID: 31645900 PMCID: PMC6791382 DOI: 10.18632/oncotarget.27209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 08/27/2019] [Indexed: 01/05/2023] Open
Abstract
We have recently found that PE21, an extract from the white willow Salix alba, slows chronological aging and prolongs longevity of the yeast Saccharomyces cerevisiae more efficiently than any of the previously known pharmacological interventions. Here, we investigated mechanisms through which PE21 delays yeast chronological aging and extends yeast longevity. We show that PE21 causes a remodeling of lipid metabolism in chronologically aging yeast, thereby instigating changes in the concentrations of several lipid classes. We demonstrate that such changes in the cellular lipidome initiate three mechanisms of aging delay and longevity extension. The first mechanism through which PE21 slows aging and prolongs longevity consists in its ability to decrease the intracellular concentration of free fatty acids. This postpones an age-related onset of liponecrotic cell death promoted by excessive concentrations of free fatty acids. The second mechanism of aging delay and longevity extension by PE21 consists in its ability to decrease the concentrations of triacylglycerols and to increase the concentrations of glycerophospholipids within the endoplasmic reticulum membrane. This activates the unfolded protein response system in the endoplasmic reticulum, which then decelerates an age-related decline in protein and lipid homeostasis and slows down an aging-associated deterioration of cell resistance to stress. The third mechanisms underlying aging delay and longevity extension by PE21 consists in its ability to change lipid concentrations in the mitochondrial membranes. This alters certain catabolic and anabolic processes in mitochondria, thus amending the pattern of aging-associated changes in several key aspects of mitochondrial functionality.
Collapse
Affiliation(s)
- Younes Medkour
- Department of Biology, Concordia University, Montreal, Quebec H4B 1R6, Canada
| | - Karamat Mohammad
- Department of Biology, Concordia University, Montreal, Quebec H4B 1R6, Canada
| | | | - Veronika Svistkova
- Department of Biology, Concordia University, Montreal, Quebec H4B 1R6, Canada
| | - Pamela Dakik
- Department of Biology, Concordia University, Montreal, Quebec H4B 1R6, Canada
| | - Darya Mitrofanova
- Department of Biology, Concordia University, Montreal, Quebec H4B 1R6, Canada
| | | | | | - Tarek Taifour
- Department of Biology, Concordia University, Montreal, Quebec H4B 1R6, Canada
| | - Paola Escudero
- Department of Biology, Concordia University, Montreal, Quebec H4B 1R6, Canada
| | - Fani-Fay Goltsios
- Department of Biology, Concordia University, Montreal, Quebec H4B 1R6, Canada
| | - Sahar Soodbakhsh
- Department of Biology, Concordia University, Montreal, Quebec H4B 1R6, Canada
| | - Hana Maalaoui
- Department of Biology, Concordia University, Montreal, Quebec H4B 1R6, Canada
| | - Éric Simard
- Idunn Technologies Inc., Rosemere, Quebec J7A 4A5, Canada
| | | |
Collapse
|
28
|
Blackwell TK, Sewell AK, Wu Z, Han M. TOR Signaling in Caenorhabditis elegans Development, Metabolism, and Aging. Genetics 2019; 213:329-360. [PMID: 31594908 PMCID: PMC6781902 DOI: 10.1534/genetics.119.302504] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 07/18/2019] [Indexed: 12/30/2022] Open
Abstract
The Target of Rapamycin (TOR or mTOR) is a serine/threonine kinase that regulates growth, development, and behaviors by modulating protein synthesis, autophagy, and multiple other cellular processes in response to changes in nutrients and other cues. Over recent years, TOR has been studied intensively in mammalian cell culture and genetic systems because of its importance in growth, metabolism, cancer, and aging. Through its advantages for unbiased, and high-throughput, genetic and in vivo studies, Caenorhabditis elegans has made major contributions to our understanding of TOR biology. Genetic analyses in the worm have revealed unexpected aspects of TOR functions and regulation, and have the potential to further expand our understanding of how growth and metabolic regulation influence development. In the aging field, C. elegans has played a leading role in revealing the promise of TOR inhibition as a strategy for extending life span, and identifying mechanisms that function upstream and downstream of TOR to influence aging. Here, we review the state of the TOR field in C. elegans, and focus on what we have learned about its functions in development, metabolism, and aging. We discuss knowledge gaps, including the potential pitfalls in translating findings back and forth across organisms, but also describe how TOR is important for C. elegans biology, and how C. elegans work has developed paradigms of great importance for the broader TOR field.
Collapse
Affiliation(s)
- T Keith Blackwell
- Research Division, Joslin Diabetes Center, Department of Genetics, Harvard Medical School, Harvard Stem Cell Institute, Boston, Massachusetts
| | - Aileen K Sewell
- Department of MCDB, University of Colorado at Boulder, and
- Howard Hughes Medical Institute, Boulder, Colorado
| | - Ziyun Wu
- Research Division, Joslin Diabetes Center, Department of Genetics, Harvard Medical School, Harvard Stem Cell Institute, Boston, Massachusetts
| | - Min Han
- Department of MCDB, University of Colorado at Boulder, and
- Howard Hughes Medical Institute, Boulder, Colorado
| |
Collapse
|
29
|
Topf U, Uszczynska-Ratajczak B, Chacinska A. Mitochondrial stress-dependent regulation of cellular protein synthesis. J Cell Sci 2019; 132:132/8/jcs226258. [DOI: 10.1242/jcs.226258] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
ABSTRACT
The production of newly synthesized proteins is vital for all cellular functions and is a determinant of cell growth and proliferation. The synthesis of polypeptide chains from mRNA molecules requires sophisticated machineries and mechanisms that need to be tightly regulated, and adjustable to current needs of the cell. Failures in the regulation of translation contribute to the loss of protein homeostasis, which can have deleterious effects on cellular function and organismal health. Unsurprisingly, the regulation of translation appears to be a crucial element in stress response mechanisms. This review provides an overview of mechanisms that modulate cytosolic protein synthesis upon cellular stress, with a focus on the attenuation of translation in response to mitochondrial stress. We then highlight links between mitochondrion-derived reactive oxygen species and the attenuation of reversible cytosolic translation through the oxidation of ribosomal proteins at their cysteine residues. We also discuss emerging concepts of how cellular mechanisms to stress are adapted, including the existence of alternative ribosomes and stress granules, and the regulation of co-translational import upon organelle stress.
Collapse
Affiliation(s)
- Ulrike Topf
- Centre of New Technologies, University of Warsaw, Banacha 2C, Warsaw 02-097, Poland
- Department of Genetics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, Warsaw 02-106, Poland
| | | | - Agnieszka Chacinska
- Centre of New Technologies, University of Warsaw, Banacha 2C, Warsaw 02-097, Poland
- ReMedy International Research Agenda Unit, University of Warsaw, Banacha 2C, Warsaw 02-097, Poland
| |
Collapse
|
30
|
Turi Z, Lacey M, Mistrik M, Moudry P. Impaired ribosome biogenesis: mechanisms and relevance to cancer and aging. Aging (Albany NY) 2019; 11:2512-2540. [PMID: 31026227 PMCID: PMC6520011 DOI: 10.18632/aging.101922] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 04/04/2019] [Indexed: 02/06/2023]
Abstract
The biosynthesis of ribosomes is a complex process that requires the coordinated action of many factors and a huge energy investment from the cell. Ribosomes are essential for protein production, and thus for cellular survival, growth and proliferation. Ribosome biogenesis is initiated in the nucleolus and includes: the synthesis and processing of ribosomal RNAs, assembly of ribosomal proteins, transport to the cytoplasm and association of ribosomal subunits. The disruption of ribosome biogenesis at various steps, with either increased or decreased expression of different ribosomal components, can promote cell cycle arrest, senescence or apoptosis. Additionally, interference with ribosomal biogenesis is often associated with cancer, aging and age-related degenerative diseases. Here, we review current knowledge on impaired ribosome biogenesis, discuss the main factors involved in stress responses under such circumstances and focus on examples with clinical relevance.
Collapse
Affiliation(s)
- Zsofia Turi
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic
| | - Matthew Lacey
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic
| | - Martin Mistrik
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic
| | - Pavel Moudry
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic
| |
Collapse
|
31
|
Austad SN, Hoffman JM. Is antagonistic pleiotropy ubiquitous in aging biology? Evol Med Public Health 2018; 2018:287-294. [PMID: 30524730 PMCID: PMC6276058 DOI: 10.1093/emph/eoy033] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 10/08/2018] [Indexed: 01/22/2023] Open
Abstract
Lay Summary: An evolutionary mechanism of aging was hypothesized 60 years ago to be the genetic trade-off between early life fitness and late life mortality. Genetic evidence supporting this hypothesis was unavailable then, but has accumulated recently. These tradeoffs, known as antagonistic pleiotropy, are common, perhaps ubiquitous. George Williams' 1957 paper developed the antagonistic pleiotropy hypothesis of aging, which had previously been hinted at by Peter Medawar. Antagonistic pleiotropy, as it applies to aging, hypothesizes that animals possess genes that enhance fitness early in life but diminish it in later life and that such genes can be favored by natural selection because selection is stronger early in life even as they cause the aging phenotype to emerge. No genes of the sort hypothesized by Williams were known 60 years ago, but modern molecular biology has now discovered hundreds of genes that, when their activity is enhanced, suppressed, or turned off, lengthen life and enhance health under laboratory conditions. Does this provide strong support for Williams' hypothesis? What are the implications of Williams' hypothesis for the modern goal of medically intervening to enhance and prolong human health? Here we briefly review the current state of knowledge on antagonistic pleiotropy both under wild and laboratory conditions. Overall, whenever antagonistic pleiotropy effects have been seriously investigated, they have been found. However, not all trade-offs are directly between reproduction and longevity as is often assumed. The discovery that antagonistic pleiotropy is common if not ubiquitous implies that a number of molecular mechanisms of aging may be widely shared among organisms and that these mechanisms of aging can be potentially alleviated by targeted interventions.
Collapse
Affiliation(s)
- Steven N Austad
- Department of Biology, CH464, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jessica M Hoffman
- Department of Biology, CH464, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
32
|
Roy D, Kahler DJ, Yun C, Hubbard EJA. Functional Interactions Between rsks-1/S6K, glp-1/Notch, and Regulators of Caenorhabditis elegans Fertility and Germline Stem Cell Maintenance. G3 (BETHESDA, MD.) 2018; 8:3293-3309. [PMID: 30126834 PMCID: PMC6169383 DOI: 10.1534/g3.118.200511] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/06/2018] [Indexed: 12/17/2022]
Abstract
The proper accumulation and maintenance of stem cells is critical for organ development and homeostasis. The Notch signaling pathway maintains stem cells in diverse organisms and organ systems. In Caenorhabditis elegans, GLP-1/Notch activity prevents germline stem cell (GSC) differentiation. Other signaling mechanisms also influence the maintenance of GSCs, including the highly-conserved TOR substrate ribosomal protein S6 kinase (S6K). Although C. elegans bearing either a null mutation in rsks-1/S6K or a reduction-of-function (rf) mutation in glp-1/Notch produce half the normal number of adult germline progenitors, virtually all these single mutant animals are fertile. However, glp-1(rf) rsks-1(null) double mutant animals are all sterile, and in about half of their gonads, all GSCs differentiate, a distinctive phenotype associated with a significant reduction or loss of GLP-1 signaling. How rsks-1/S6K promotes GSC fate is unknown. Here, we determine that rsks-1/S6K acts germline-autonomously to maintain GSCs, and that it does not act through Cyclin-E or MAP kinase in this role. We found that interfering with translation also enhances glp-1(rf), but that regulation through rsks-1 cannot fully account for this effect. In a genome-scale RNAi screen for genes that act similarly to rsks-1/S6K, we identified 56 RNAi enhancers of glp-1(rf) sterility, many of which were previously not known to interact functionally with Notch. Further investigation revealed at least six candidates that, by genetic criteria, act linearly with rsks-1/S6K. These include genes encoding translation-related proteins, cacn-1/Cactin, an RNA exosome component, and a Hedgehog-related ligand. We found that additional Hedgehog-related ligands may share functional relationships with glp-1/Notch and rsks-1/S6K in maintaining germline progenitors.
Collapse
Affiliation(s)
- Debasmita Roy
- Skirball Institute of Biomolecular Medicine, Departments of Cell Biology and Pathology, New York University School of Medicine, New York, NY 10016
| | - David J Kahler
- NYU High Throughput Biology Laboratory, NYU Langone Health, New York, NY 10016
| | - Chi Yun
- NYU High Throughput Biology Laboratory, NYU Langone Health, New York, NY 10016
| | - E Jane Albert Hubbard
- Skirball Institute of Biomolecular Medicine, Departments of Cell Biology and Pathology, New York University School of Medicine, New York, NY 10016
| |
Collapse
|
33
|
Hu Z, Xia B, Postnikoff SD, Shen ZJ, Tomoiaga AS, Harkness TA, Seol JH, Li W, Chen K, Tyler JK. Ssd1 and Gcn2 suppress global translation efficiency in replicatively aged yeast while their activation extends lifespan. eLife 2018; 7:35551. [PMID: 30117416 PMCID: PMC6097839 DOI: 10.7554/elife.35551] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 08/03/2018] [Indexed: 12/21/2022] Open
Abstract
Translational efficiency correlates with longevity, yet its role in lifespan determination remains unclear. Using ribosome profiling, translation efficiency is globally reduced during replicative aging in budding yeast by at least two mechanisms: Firstly, Ssd1 is induced during aging, sequestering mRNAs to P-bodies. Furthermore, Ssd1 overexpression in young cells reduced translation and extended lifespan, while loss of Ssd1 reduced the translational deficit of old cells and shortened lifespan. Secondly, phosphorylation of eIF2α, mediated by the stress kinase Gcn2, was elevated in old cells, contributing to the global reduction in translation without detectable induction of the downstream Gcn4 transcriptional activator. tRNA overexpression activated Gcn2 in young cells and extended lifespan in a manner dependent on Gcn4. Moreover, overexpression of Gcn4 sufficed to extend lifespan in an autophagy-dependent manner in the absence of changes in global translation, indicating that Gcn4-mediated autophagy induction is the ultimate downstream target of activated Gcn2, to extend lifespan.
Collapse
Affiliation(s)
- Zheng Hu
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, United States
| | - Bo Xia
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, United States.,Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, United States
| | - Spike Dl Postnikoff
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, United States
| | - Zih-Jie Shen
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, United States
| | - Alin S Tomoiaga
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, United States.,Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, United States.,Manhattan College, Bronx, United States
| | - Troy A Harkness
- Department of Anatomy and Cell Biology, University of Saskatchewan, Saskatoon, Canada
| | - Ja Hwan Seol
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, United States
| | - Wei Li
- Dan L. Duncan Cancer Center and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, United States
| | - Kaifu Chen
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, United States.,Department of Cardiothoracic Surgery, Weill Cornell Medicine, New York, United States
| | - Jessica K Tyler
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, United States
| |
Collapse
|
34
|
Wilhelm T, Byrne J, Medina R, Kolundžić E, Geisinger J, Hajduskova M, Tursun B, Richly H. Neuronal inhibition of the autophagy nucleation complex extends life span in post-reproductive C. elegans. Genes Dev 2017; 31:1561-1572. [PMID: 28882853 PMCID: PMC5630021 DOI: 10.1101/gad.301648.117] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 08/09/2017] [Indexed: 12/19/2022]
Abstract
Here, Wilhelm et al. performed an RNAi screen in C. elegans to identify genes mediating post-reproductive longevity. They found that the inhibition of vesicle nucleation in the post-reproductive animal prevents age-associated neuronal degeneration, which leads to increased health and life span. Autophagy is a ubiquitous catabolic process that causes cellular bulk degradation of cytoplasmic components and is generally associated with positive effects on health and longevity. Inactivation of autophagy has been linked with detrimental effects on cells and organisms. The antagonistic pleiotropy theory postulates that some fitness-promoting genes during youth are harmful during aging. On this basis, we examined genes mediating post-reproductive longevity using an RNAi screen. From this screen, we identified 30 novel regulators of post-reproductive longevity, including pha-4. Through downstream analysis of pha-4, we identified that the inactivation of genes governing the early stages of autophagy up until the stage of vesicle nucleation, such as bec-1, strongly extend both life span and health span. Furthermore, our data demonstrate that the improvements in health and longevity are mediated through the neurons, resulting in reduced neurodegeneration and sarcopenia. We propose that autophagy switches from advantageous to harmful in the context of an age-associated dysfunction.
Collapse
Affiliation(s)
- Thomas Wilhelm
- Laboratory of Molecular Epigenetics, Institute of Molecular Biology (IMB), 55128 Mainz, Germany.,Faculty of Biology, Johannes Gutenberg University, 55099 Mainz, Germany
| | - Jonathan Byrne
- Laboratory of Molecular Epigenetics, Institute of Molecular Biology (IMB), 55128 Mainz, Germany.,Faculty of Biology, Johannes Gutenberg University, 55099 Mainz, Germany
| | - Rebeca Medina
- Laboratory of Molecular Epigenetics, Institute of Molecular Biology (IMB), 55128 Mainz, Germany
| | - Ena Kolundžić
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center (MDC), 13125 Berlin, Germany
| | - Johannes Geisinger
- Laboratory of Molecular Epigenetics, Institute of Molecular Biology (IMB), 55128 Mainz, Germany.,Faculty of Biology, Johannes Gutenberg University, 55099 Mainz, Germany
| | - Martina Hajduskova
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center (MDC), 13125 Berlin, Germany
| | - Baris Tursun
- Berlin Institute for Medical Systems Biology (BIMSB), Max Delbrück Center (MDC), 13125 Berlin, Germany
| | - Holger Richly
- Laboratory of Molecular Epigenetics, Institute of Molecular Biology (IMB), 55128 Mainz, Germany
| |
Collapse
|
35
|
May CM, Zwaan BJ. Relating past and present diet to phenotypic and transcriptomic variation in the fruit fly. BMC Genomics 2017; 18:640. [PMID: 28830340 PMCID: PMC5568309 DOI: 10.1186/s12864-017-3968-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 07/31/2017] [Indexed: 01/22/2023] Open
Abstract
Background Sub-optimal developmental diets often have adverse effects on long-term fitness and health. One hypothesis is that such effects are caused by mismatches between the developmental and adult environment, and may be mediated by persistent changes in gene expression. However, there are few experimental tests of this hypothesis. Here we address this using the fruit fly, Drosophila melanogaster. We vary diet during development and adulthood in a fully factorial design and assess the consequences for both adult life history traits and gene expression at middle and old age. Results We find no evidence that mismatches between developmental and adult diet are detrimental to either lifespan or fecundity. Rather, developmental and adult diet exert largely independent effects on both lifespan and gene expression, with adult diet having considerably more influence on both traits. Furthermore, we find effects of developmental diet on the transcriptome that persist into middle and old-age. Most of the genes affected show no correlation with the observed phenotypic effects of larval diet on lifespan. However, in each sex we identify a cluster of ribosome, transcription, and translation-related genes whose expression is altered across the lifespan and negatively correlated with lifespan. Conclusions As several recent studies have linked decreased expression of ribosomal and transcription related proteins to increased lifespan, these provide promising candidates for mediating the effects of larval diet on lifespan. We place our findings in the context of theories linking developmental conditions to late-life phenotypes and discuss the likelihood that gene expression differences caused by developmental exposure causally relate to adult ageing phenotypes. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3968-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christina M May
- Laboratory of Genetics, Plant Sciences, Wageningen University, Wageningen, 6708, PB, the Netherlands.
| | - Bas J Zwaan
- Laboratory of Genetics, Plant Sciences, Wageningen University, Wageningen, 6708, PB, the Netherlands
| |
Collapse
|
36
|
Sutphin GL, Backer G, Sheehan S, Bean S, Corban C, Liu T, Peters MJ, van Meurs JBJ, Murabito JM, Johnson AD, Korstanje R. Caenorhabditis elegans orthologs of human genes differentially expressed with age are enriched for determinants of longevity. Aging Cell 2017; 16:672-682. [PMID: 28401650 PMCID: PMC5506438 DOI: 10.1111/acel.12595] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2017] [Indexed: 12/21/2022] Open
Abstract
We report a systematic RNAi longevity screen of 82 Caenorhabditis elegans genes selected based on orthology to human genes differentially expressed with age. We find substantial enrichment in genes for which knockdown increased lifespan. This enrichment is markedly higher than published genomewide longevity screens in C. elegans and similar to screens that preselected candidates based on longevity‐correlated metrics (e.g., stress resistance). Of the 50 genes that affected lifespan, 46 were previously unreported. The five genes with the greatest impact on lifespan (>20% extension) encode the enzyme kynureninase (kynu‐1), a neuronal leucine‐rich repeat protein (iglr‐1), a tetraspanin (tsp‐3), a regulator of calcineurin (rcan‐1), and a voltage‐gated calcium channel subunit (unc‐36). Knockdown of each gene extended healthspan without impairing reproduction. kynu‐1(RNAi) alone delayed pathology in C. elegans models of Alzheimer's disease and Huntington's disease. Each gene displayed a distinct pattern of interaction with known aging pathways. In the context of published work, kynu‐1, tsp‐3, and rcan‐1 are of particular interest for immediate follow‐up. kynu‐1 is an understudied member of the kynurenine metabolic pathway with a mechanistically distinct impact on lifespan. Our data suggest that tsp‐3 is a novel modulator of hypoxic signaling and rcan‐1 is a context‐specific calcineurin regulator. Our results validate C. elegans as a comparative tool for prioritizing human candidate aging genes, confirm age‐associated gene expression data as valuable source of novel longevity determinants, and prioritize select genes for mechanistic follow‐up.
Collapse
Affiliation(s)
| | - Grant Backer
- The Jackson Laboratory; 600 Main Street Bar Harbor ME 04609 USA
| | - Susan Sheehan
- The Jackson Laboratory; 600 Main Street Bar Harbor ME 04609 USA
| | - Shannon Bean
- The Jackson Laboratory; 600 Main Street Bar Harbor ME 04609 USA
| | - Caroline Corban
- The Jackson Laboratory; 600 Main Street Bar Harbor ME 04609 USA
| | - Teresa Liu
- The Jackson Laboratory; 600 Main Street Bar Harbor ME 04609 USA
| | - Marjolein J. Peters
- Department of Internal Medicine; Erasmus Medical Center; Postbus 2040 3000 CA Rotterdam The Netherlands
| | - Joyce B. J. van Meurs
- Department of Internal Medicine; Erasmus Medical Center; Postbus 2040 3000 CA Rotterdam The Netherlands
| | - Joanne M. Murabito
- Section of General Internal Medicine; Boston University School of Medicine; 801 Massachusetts Ave, Crosstown Center Boston MA 02118 USA
- The National Heart, Lung, and Blood Institute's Framingham Heart Study; 73 Mt. Wayte Ave, Suite 2 Framingham MA 01702-5827 USA
| | - Andrew D. Johnson
- The National Heart, Lung, and Blood Institute's Framingham Heart Study; 73 Mt. Wayte Ave, Suite 2 Framingham MA 01702-5827 USA
- Population Sciences Branch; National Heart, Lung, and Blood Institute; Building 31, Room 5A52, 31 Center Drive MSC 2486 Bethesda MD 20892 USA
| | - Ron Korstanje
- The Jackson Laboratory; 600 Main Street Bar Harbor ME 04609 USA
| | | |
Collapse
|
37
|
Mason AG, Garza RM, McCormick MA, Patel B, Kennedy BK, Pillus L, La Spada AR. The replicative lifespan-extending deletion of SGF73 results in altered ribosomal gene expression in yeast. Aging Cell 2017; 16:785-796. [PMID: 28568901 PMCID: PMC5506417 DOI: 10.1111/acel.12611] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2017] [Indexed: 12/29/2022] Open
Abstract
Sgf73, a core component of SAGA, is the yeast orthologue of ataxin‐7, which undergoes CAG–polyglutamine repeat expansion leading to the human neurodegenerative disease spinocerebellar ataxia type 7 (SCA7). Deletion of SGF73 dramatically extends replicative lifespan (RLS) in yeast. To further define the basis for Sgf73‐mediated RLS extension, we performed ChIP‐Seq, identified 388 unique genomic regions occupied by Sgf73, and noted enrichment in promoters of ribosomal protein (RP)‐encoding genes. Of 388 Sgf73 binding sites, 33 correspond to 5′ regions of genes implicated in RLS extension, including 20 genes encoding RPs. Furthermore, half of Sgf73‐occupied, RLS‐linked RP genes displayed significantly reduced expression in sgf73Δ mutants, and double null strains lacking SGF73 and a Sgf73‐regulated, RLS‐linked RP gene exhibited no further increase in replicative lifespan. We also found that sgf73Δ mutants display altered acetylation of Ifh1, an important regulator of RP gene transcription. These findings implicate altered ribosomal protein expression in sgf73Δ yeast RLS and highlight altered acetylation as a pathway of relevance for SCA7 neurodegeneration.
Collapse
Affiliation(s)
- Amanda G. Mason
- Department of Pediatrics; University of California, San Diego; La Jolla CA USA
- Division of Biological Sciences; University of California, San Diego; La Jolla CA USA
| | - Renee M. Garza
- Division of Biological Sciences; University of California, San Diego; La Jolla CA USA
- Moores Cancer Center; University of California, San Diego; La Jolla CA USA
| | - Mark A. McCormick
- Buck Institute for Research on Aging; Novato CA USA
- Department of Biochemistry; University of Washington; Seattle WA USA
| | - Bhumil Patel
- Buck Institute for Research on Aging; Novato CA USA
| | - Brian K. Kennedy
- Buck Institute for Research on Aging; Novato CA USA
- Department of Biochemistry; University of Washington; Seattle WA USA
| | - Lorraine Pillus
- Division of Biological Sciences; University of California, San Diego; La Jolla CA USA
- Moores Cancer Center; University of California, San Diego; La Jolla CA USA
| | - Albert R. La Spada
- Department of Pediatrics; University of California, San Diego; La Jolla CA USA
- Division of Biological Sciences; University of California, San Diego; La Jolla CA USA
- Departments of Cellular & Molecular Medicine and Neurosciences; University of California, San Diego; La Jolla CA USA
- Institute for Genomic Medicine; University of California, San Diego; La Jolla CA USA
- Sanford Consortium for Regenerative Medicine; University of California, San Diego; La Jolla CA USA. Rady Children's Hospital; San Diego CA USA
| |
Collapse
|
38
|
Gonskikh Y, Polacek N. Alterations of the translation apparatus during aging and stress response. Mech Ageing Dev 2017; 168:30-36. [PMID: 28414025 DOI: 10.1016/j.mad.2017.04.003] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 03/28/2017] [Accepted: 04/10/2017] [Indexed: 01/03/2023]
Abstract
Aging is a biological process characterized by the irreversible and time-dependent deterioration of cell functions, tissues, and organs. Accumulating studies in a wide range of species from yeast to human revealed changes associated with the aging process to be conserved throughout evolution. The main characteristics of aging are (i) genomic instability, (ii) loss of telomere function, (iii) epigenetic changes,(iv) increased cellular senescence, (v) depletion of the stem cell pool, (vi) altered intercellular communication and (vii) loss of protein homeostasis. Among the multiple molecular mechanisms underlying aging, alterations of the translation machinery affecting the rate and selectivity of protein biosynthesis seem to play a central role. At the very heart of translation is the ribosome, a multifaceted and universally conserved RNA-protein particle responsible for accurate polypeptide synthesis and co-translational protein folding. Here we summarize and discuss recent developments on the contribution of altered translation and age-dependent modifications on the ribosome structure to aging and cellular senescence.
Collapse
Affiliation(s)
- Yulia Gonskikh
- Department of Chemistry and Biochemistry, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Norbert Polacek
- Department of Chemistry and Biochemistry, University of Bern, Freiestrasse 3, 3012 Bern, Switzerland.
| |
Collapse
|
39
|
Jeong DE, Lee D, Hwang SY, Lee Y, Lee JE, Seo M, Hwang W, Seo K, Hwang AB, Artan M, Son HG, Jo JH, Baek H, Oh YM, Ryu Y, Kim HJ, Ha CM, Yoo JY, Lee SJV. Mitochondrial chaperone HSP-60 regulates anti-bacterial immunity via p38 MAP kinase signaling. EMBO J 2017; 36:1046-1065. [PMID: 28283579 DOI: 10.15252/embj.201694781] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 02/08/2017] [Accepted: 02/10/2017] [Indexed: 12/13/2022] Open
Abstract
Mitochondria play key roles in cellular immunity. How mitochondria contribute to organismal immunity remains poorly understood. Here, we show that HSP-60/HSPD1, a major mitochondrial chaperone, boosts anti-bacterial immunity through the up-regulation of p38 MAP kinase signaling. We first identify 16 evolutionarily conserved mitochondrial components that affect the immunity of Caenorhabditis elegans against pathogenic Pseudomonas aeruginosa (PA14). Among them, the mitochondrial chaperone HSP-60 is necessary and sufficient to increase resistance to PA14. We show that HSP-60 in the intestine and neurons is crucial for the resistance to PA14. We then find that p38 MAP kinase signaling, an evolutionarily conserved anti-bacterial immune pathway, is down-regulated by genetic inhibition of hsp-60, and up-regulated by increased expression of hsp-60 Overexpression of HSPD1, the mammalian ortholog of hsp-60, increases p38 MAP kinase activity in human cells, suggesting an evolutionarily conserved mechanism. Further, cytosol-localized HSP-60 physically binds and stabilizes SEK-1/MAP kinase kinase 3, which in turn up-regulates p38 MAP kinase and increases immunity. Our study suggests that mitochondrial chaperones protect host eukaryotes from pathogenic bacteria by up-regulating cytosolic p38 MAPK signaling.
Collapse
Affiliation(s)
- Dae-Eun Jeong
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| | - Dongyeop Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| | - Sun-Young Hwang
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| | - Yujin Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| | - Jee-Eun Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| | - Mihwa Seo
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| | - Wooseon Hwang
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| | - Keunhee Seo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| | - Ara B Hwang
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| | - Murat Artan
- Information Technology Convergence Engineering, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| | - Heehwa G Son
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| | - Jay-Hyun Jo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| | - Haeshim Baek
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| | - Young Min Oh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| | - Youngjae Ryu
- Research Division, Korea Brain Research Institute, Daegu, Korea
| | - Hyung-Jun Kim
- Research Division, Korea Brain Research Institute, Daegu, Korea
| | - Chang Man Ha
- Research Division, Korea Brain Research Institute, Daegu, Korea
| | - Joo-Yeon Yoo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| | - Seung-Jae V Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea .,School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea.,Information Technology Convergence Engineering, Pohang University of Science and Technology, Pohang, Gyeongbuk, Korea
| |
Collapse
|
40
|
Pu YZ, Wan QL, Ding AJ, Luo HR, Wu GS. Quantitative proteomics analysis of Caenorhabditis elegans upon germ cell loss. J Proteomics 2017; 156:85-93. [DOI: 10.1016/j.jprot.2017.01.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 01/14/2017] [Accepted: 01/19/2017] [Indexed: 10/20/2022]
|
41
|
Bacillus subtilis biofilm extends Caenorhabditis elegans longevity through downregulation of the insulin-like signalling pathway. Nat Commun 2017; 8:14332. [PMID: 28134244 PMCID: PMC5290332 DOI: 10.1038/ncomms14332] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 12/19/2016] [Indexed: 12/24/2022] Open
Abstract
Beneficial bacteria have been shown to affect host longevity, but the molecular mechanisms mediating such effects remain largely unclear. Here we show that formation of Bacillus subtilis biofilms increases Caenorhabditis elegans lifespan. Biofilm-proficient B. subtilis colonizes the C. elegans gut and extends worm lifespan more than biofilm-deficient isogenic strains. Two molecules produced by B. subtilis — the quorum-sensing pentapeptide CSF and nitric oxide (NO) — are sufficient to extend C. elegans longevity. When B. subtilis is cultured under biofilm-supporting conditions, the synthesis of NO and CSF is increased in comparison with their production under planktonic growth conditions. We further show that the prolongevity effect of B. subtilis biofilms depends on the DAF-2/DAF-16/HSF-1 signalling axis and the downregulation of the insulin-like signalling (ILS) pathway. Probiotic bacteria can improve host health, but the mechanisms underlying such beneficial effects are often unclear. Here, the authors show that biofilm formation of the probiotic bacterium B. subtilis extends the lifespan of its host, the nematode C. elegans, by reducing insulin-like signalling.
Collapse
|
42
|
Jiang X, Jiang X, Feng Y, Xu R, Wang Q, Deng H. Proteomic Analysis of eIF5B Silencing-Modulated Proteostasis. PLoS One 2016; 11:e0168387. [PMID: 27959964 PMCID: PMC5154608 DOI: 10.1371/journal.pone.0168387] [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: 06/21/2016] [Accepted: 11/29/2016] [Indexed: 11/17/2022] Open
Abstract
Protein translational machinery is an important component of the proteostasis network that maintains cellular proteostasis and regulates aging and other cellular processes. Ample evidence indicates that inhibition of translation initiation factor activities enhances stress resistance in model organisms. Eukaryotic translation initiation factor 5B (eIF5B) acts by joining the pre-40S subunit with the 60S ribosomal unit to form an 80S-like complex during protein translational initiation. Reduced eIF5B expression may disrupt proteostasis and trigger cellular processes associated with stress responses. In this study, the physiological effects of altered eIF5B expression were examined in 293T and HepG2 cells. Cells with eIF5B-knockdown (eIF5B-KN) grew more slowly than control cells, and had a lower level of intracellular reactive oxygen species (ROS), increased resistance to oxidative stress and enhanced autophagy. Proteomic analysis showed that eIF5B knockdown resulted in upregulation of 88 proteins and downregulation of 130 proteins compared with control cells. The differentially expressed proteins were associated with diverse cellular processes including amino acid metabolism, RNA processing and protein metabolism, and DNA synthesis. Autonomous downregulation of the mitogen-activated protein kinase (MAPK) signaling pathway was identified as confirmed by western blotting and qPCR. We proposed that deactivation of MAPK pathway modulated proteostasis and induced prolonged S-phase of the cell-cycle, contributing to the slow growth of eIF5B-KN cells. eIF5B silencing also inactivated the mTOR pathway, downregulated glutamine transporters, enhanced autophagy, and decreased 28S rRNA and 5.8S rRNA expression levels which were reversed by restoration of eIF5B expression. Taken together, these results suggest that eIF5B silencing provides a negative feedback to deactivate MAPK signaling, leading to reduced cell growth. These findings provide a useful resource to further biological exploration of the functions of protein synthesis in regulation of proteostasis and stress responses.
Collapse
Affiliation(s)
- Xu Jiang
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xiaoyong Jiang
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yun Feng
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, China
| | - Renhua Xu
- School of Nursing, Binzhou Medical University, Yantai, China
| | - Qingtao Wang
- Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
43
|
Cattie DJ, Richardson CE, Reddy KC, Ness-Cohn EM, Droste R, Thompson MK, Gilbert WV, Kim DH. Mutations in Nonessential eIF3k and eIF3l Genes Confer Lifespan Extension and Enhanced Resistance to ER Stress in Caenorhabditis elegans. PLoS Genet 2016; 12:e1006326. [PMID: 27690135 PMCID: PMC5045169 DOI: 10.1371/journal.pgen.1006326] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 08/26/2016] [Indexed: 11/18/2022] Open
Abstract
The translation initiation factor eIF3 is a multi-subunit protein complex that coordinates the assembly of the 43S pre-initiation complex in eukaryotes. Prior studies have demonstrated that not all subunits of eIF3 are essential for the initiation of translation, suggesting that some subunits may serve regulatory roles. Here, we show that loss-of-function mutations in the genes encoding the conserved eIF3k and eIF3l subunits of the translation initiation complex eIF3 result in a 40% extension in lifespan and enhanced resistance to endoplasmic reticulum (ER) stress in Caenorhabditis elegans. In contrast to previously described mutations in genes encoding translation initiation components that confer lifespan extension in C. elegans, loss-of-function mutations in eif-3.K or eif-3.L are viable, and mutants show normal rates of growth and development, and have wild-type levels of bulk protein synthesis. Lifespan extension resulting from EIF-3.K or EIF-3.L deficiency is suppressed by a mutation in the Forkhead family transcription factor DAF-16. Mutations in eif-3.K or eif-3.L also confer enhanced resistance to ER stress, independent of IRE-1-XBP-1, ATF-6, and PEK-1, and independent of DAF-16. Our data suggest a pivotal functional role for conserved eIF3k and eIF3l accessory subunits of eIF3 in the regulation of cellular and organismal responses to ER stress and aging.
Collapse
Affiliation(s)
- Douglas J. Cattie
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Claire E. Richardson
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Kirthi C. Reddy
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Elan M. Ness-Cohn
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Rita Droste
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Howard Hughes Medical Institute, McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Mary K. Thompson
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Wendy V. Gilbert
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Dennis H. Kim
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
44
|
Hekimi S, Wang Y, Noë A. Mitochondrial ROS and the Effectors of the Intrinsic Apoptotic Pathway in Aging Cells: The Discerning Killers! Front Genet 2016; 7:161. [PMID: 27683586 PMCID: PMC5021979 DOI: 10.3389/fgene.2016.00161] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 08/30/2016] [Indexed: 01/06/2023] Open
Abstract
It has become clear that mitochondrial reactive oxygen species (mtROS) are not simply villains and mitochondria the hapless targets of their attacks. Rather, it appears that mitochondrial dysfunction itself and the signaling function of mtROS can have positive effects on lifespan, helping to extend longevity. If events in the mitochondria can lead to better cellular homeostasis and better survival of the organism in ways beyond providing ATP and biosynthetic products, we can conjecture that they act on other cellular components through appropriate signaling pathways. We describe recent advances in a variety of species which promoted our understanding of how changes of mtROS generation are part of a system of signaling pathways that emanate from the mitochondria to impact organism lifespan through global changes, including in transcriptional patterns. In unraveling this, many old players in cellular homeostasis were encountered. Among these, maybe most strikingly, is the intrinsic apoptotic signaling pathway, which is the conduit by which at least one class of mtROS exercise their actions in the nematode Caenorhabditis elegans. This is a pathway that normally contributes to organismal homeostasis by killing defective or otherwise unwanted cells, and whose various compounds have also been implicated in other cellular processes. However, it was a surprise that that appropriate activation of a cell killing pathway can in fact prolong the lifespan of the organism. In the soma of adult C. elegans, all cells are post-mitotic, like many of our neurons and possibly some of our immune cells. These cells cannot simply be killed and replaced when showing signs of dysfunction. Thus, we speculate that it is the ability of the apoptotic pathway to pull together information about the functional and structural integrity of different cellular compartments that is the key property for why this pathway is used to decide when to boost defensive and repair processes in irreplaceable cells. When this process is artificially stimulated in mutants with elevated mtROS generation or with drug treatments it leads to lifespan prolongations beyond the normal lifespan of the organism.
Collapse
Affiliation(s)
| | - Ying Wang
- Department of Biology, McGill University Montreal, QC, Canada
| | - Alycia Noë
- Department of Biology, McGill University Montreal, QC, Canada
| |
Collapse
|
45
|
C. elegans screening strategies to identify pro-longevity interventions. Mech Ageing Dev 2016; 157:60-9. [PMID: 27473404 DOI: 10.1016/j.mad.2016.07.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 07/22/2016] [Accepted: 07/25/2016] [Indexed: 02/07/2023]
Abstract
Drugs screenings in search of enhancers or suppressors of selected readout(s) are nowadays mainly carried out in single cells systems. These approaches are however limited when searching for compounds with effects at the organismal level. To overcome this drawback the use of different model organisms to carry out modifier screenings has exponentially grown in the past decade. Unique characteristics such as easy manageability, low cost, fast reproductive cycle, short lifespan, simple anatomy and genetic amenability, make the nematode Caenorhabditis elegans especially suitable for this purpose. Here we briefly review the different high-throughput and high-content screenings which exploited the nematode to identify new compounds extending healthy lifespan. In this context, we describe our recently developed screening strategy to search for pro-longevity interventions taking advantage of the very reproducible phenotypes observed in C. elegans upon different degrees of mitochondrial stress. Indeed, in Mitochondrial mutants, the processes induced to cope with mild mitochondrial alterations during development, and ultimately extending animal lifespan, lead to reduced size and induction of specific stress responses. Instead, upon strong mitochondrial dysfunction, worms arrest their development. Exploiting these automatically quantifiable phenotypic readouts, we developed a new screening approach using the Cellomics ArrayScanVTI-HCS Reader and identified a new pro-longevity drug.
Collapse
|
46
|
Calvert S, Tacutu R, Sharifi S, Teixeira R, Ghosh P, de Magalhães JP. A network pharmacology approach reveals new candidate caloric restriction mimetics in C. elegans. Aging Cell 2016; 15:256-66. [PMID: 26676933 PMCID: PMC4783339 DOI: 10.1111/acel.12432] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2015] [Indexed: 01/04/2023] Open
Abstract
Caloric restriction (CR), a reduction in calorie intake without malnutrition, retards aging in several animal models from worms to mammals. Developing CR mimetics, compounds that reproduce the longevity benefits of CR without its side effects, is of widespread interest. Here, we employed the Connectivity Map to identify drugs with overlapping gene expression profiles with CR. Eleven statistically significant compounds were predicted as CR mimetics using this bioinformatics approach. We then tested rapamycin, allantoin, trichostatin A, LY‐294002 and geldanamycin in Caenorhabditis elegans. An increase in lifespan and healthspan was observed for all drugs except geldanamycin when fed to wild‐type worms, but no lifespan effects were observed in eat‐2 mutant worms, a genetic model of CR, suggesting that life‐extending effects may be acting via CR‐related mechanisms. We also treated daf‐16 worms with rapamycin, allantoin or trichostatin A, and a lifespan extension was observed, suggesting that these drugs act via DAF‐16‐independent mechanisms, as would be expected from CR mimetics. Supporting this idea, an analysis of predictive targets of the drugs extending lifespan indicates various genes within CR and longevity networks. We also assessed the transcriptional profile of worms treated with either rapamycin or allantoin and found that both drugs use several specific pathways that do not overlap, indicating different modes of action for each compound. The current work validates the capabilities of this bioinformatic drug repositioning method in the context of longevity and reveals new putative CR mimetics that warrant further studies.
Collapse
Affiliation(s)
- Shaun Calvert
- Integrative Genomics of Ageing Group; Institute of Integrative Biology; University of Liverpool; Liverpool UK
| | - Robi Tacutu
- Integrative Genomics of Ageing Group; Institute of Integrative Biology; University of Liverpool; Liverpool UK
| | - Samim Sharifi
- Integrative Genomics of Ageing Group; Institute of Integrative Biology; University of Liverpool; Liverpool UK
| | - Rute Teixeira
- Integrative Genomics of Ageing Group; Institute of Integrative Biology; University of Liverpool; Liverpool UK
| | - Pratul Ghosh
- Integrative Genomics of Ageing Group; Institute of Integrative Biology; University of Liverpool; Liverpool UK
| | - João Pedro de Magalhães
- Integrative Genomics of Ageing Group; Institute of Integrative Biology; University of Liverpool; Liverpool UK
| |
Collapse
|
47
|
McCormick MA, Delaney JR, Tsuchiya M, Tsuchiyama S, Shemorry A, Sim S, Chou ACZ, Ahmed U, Carr D, Murakami CJ, Schleit J, Sutphin GL, Wasko BM, Bennett CF, Wang AM, Olsen B, Beyer RP, Bammler TK, Prunkard D, Johnson SC, Pennypacker JK, An E, Anies A, Castanza AS, Choi E, Dang N, Enerio S, Fletcher M, Fox L, Goswami S, Higgins SA, Holmberg MA, Hu D, Hui J, Jelic M, Jeong KS, Johnston E, Kerr EO, Kim J, Kim D, Kirkland K, Klum S, Kotireddy S, Liao E, Lim M, Lin MS, Lo WC, Lockshon D, Miller HA, Moller RM, Muller B, Oakes J, Pak DN, Peng ZJ, Pham KM, Pollard TG, Pradeep P, Pruett D, Rai D, Robison B, Rodriguez AA, Ros B, Sage M, Singh MK, Smith ED, Snead K, Solanky A, Spector BL, Steffen KK, Tchao BN, Ting MK, Vander Wende H, Wang D, Welton KL, Westman EA, Brem RB, Liu XG, Suh Y, Zhou Z, Kaeberlein M, Kennedy BK. A Comprehensive Analysis of Replicative Lifespan in 4,698 Single-Gene Deletion Strains Uncovers Conserved Mechanisms of Aging. Cell Metab 2015; 22:895-906. [PMID: 26456335 PMCID: PMC4862740 DOI: 10.1016/j.cmet.2015.09.008] [Citation(s) in RCA: 173] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 07/31/2015] [Accepted: 09/08/2015] [Indexed: 02/05/2023]
Abstract
Many genes that affect replicative lifespan (RLS) in the budding yeast Saccharomyces cerevisiae also affect aging in other organisms such as C. elegans and M. musculus. We performed a systematic analysis of yeast RLS in a set of 4,698 viable single-gene deletion strains. Multiple functional gene clusters were identified, and full genome-to-genome comparison demonstrated a significant conservation in longevity pathways between yeast and C. elegans. Among the mechanisms of aging identified, deletion of tRNA exporter LOS1 robustly extended lifespan. Dietary restriction (DR) and inhibition of mechanistic Target of Rapamycin (mTOR) exclude Los1 from the nucleus in a Rad53-dependent manner. Moreover, lifespan extension from deletion of LOS1 is nonadditive with DR or mTOR inhibition, and results in Gcn4 transcription factor activation. Thus, the DNA damage response and mTOR converge on Los1-mediated nuclear tRNA export to regulate Gcn4 activity and aging.
Collapse
Affiliation(s)
- Mark A McCormick
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Joe R Delaney
- Department of Pathology, University of Washington, Seattle, WA 98195, USA; Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Mitsuhiro Tsuchiya
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Scott Tsuchiyama
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Anna Shemorry
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Sylvia Sim
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | | | - Umema Ahmed
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Daniel Carr
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | | | - Jennifer Schleit
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - George L Sutphin
- Department of Pathology, University of Washington, Seattle, WA 98195, USA; Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Brian M Wasko
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Christopher F Bennett
- Department of Pathology, University of Washington, Seattle, WA 98195, USA; Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Adrienne M Wang
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Brady Olsen
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Richard P Beyer
- Department of Occupational and Environmental Health Sciences, University of Washington, Seattle, WA 98195, USA
| | - Theodor K Bammler
- Department of Occupational and Environmental Health Sciences, University of Washington, Seattle, WA 98195, USA
| | - Donna Prunkard
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Simon C Johnson
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | | | - Elroy An
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Arieanna Anies
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Anthony S Castanza
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Eunice Choi
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Nick Dang
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Shiena Enerio
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Marissa Fletcher
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Lindsay Fox
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Sarani Goswami
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Sean A Higgins
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Molly A Holmberg
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Di Hu
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Jessica Hui
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Monika Jelic
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Ki-Soo Jeong
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Elijah Johnston
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Emily O Kerr
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Jin Kim
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Diana Kim
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Katie Kirkland
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Shannon Klum
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Soumya Kotireddy
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Eric Liao
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Michael Lim
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Michael S Lin
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Winston C Lo
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Dan Lockshon
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Hillary A Miller
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Richard M Moller
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Brian Muller
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Jonathan Oakes
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Diana N Pak
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Zhao Jun Peng
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Kim M Pham
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Tom G Pollard
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Prarthana Pradeep
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Dillon Pruett
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Dilreet Rai
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Brett Robison
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Ariana A Rodriguez
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Bopharoth Ros
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Michael Sage
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Manpreet K Singh
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Erica D Smith
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Katie Snead
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Amrita Solanky
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Benjamin L Spector
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Kristan K Steffen
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Bie Nga Tchao
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Marc K Ting
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Helen Vander Wende
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Dennis Wang
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - K Linnea Welton
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Eric A Westman
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Rachel B Brem
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Xin-Guang Liu
- Aging Research Institute, Guangdong Medical College, Dongguan 523808, Guangdong, P.R. China
| | - Yousin Suh
- Aging Research Institute, Guangdong Medical College, Dongguan 523808, Guangdong, P.R. China; Department of Genetics, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Zhongjun Zhou
- Department of Biochemistry, University of Hong Kong, Hong Kong
| | - Matt Kaeberlein
- Department of Pathology, University of Washington, Seattle, WA 98195, USA.
| | - Brian K Kennedy
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA; Department of Biochemistry, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
48
|
Systematic analysis of asymmetric partitioning of yeast proteome between mother and daughter cells reveals "aging factors" and mechanism of lifespan asymmetry. Proc Natl Acad Sci U S A 2015; 112:11977-82. [PMID: 26351681 DOI: 10.1073/pnas.1506054112] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Budding yeast divides asymmetrically, giving rise to a mother cell that progressively ages and a daughter cell with full lifespan. It is generally assumed that mother cells retain damaged, lifespan limiting materials ("aging factors") through asymmetric division. However, the identity of these aging factors and the mechanisms through which they limit lifespan remain poorly understood. Using a flow cytometry-based, high-throughput approach, we quantified the asymmetric partitioning of the yeast proteome between mother and daughter cells during cell division, discovering 74 mother-enriched and 60 daughter-enriched proteins. While daughter-enriched proteins are biased toward those needed for bud construction and genome maintenance, mother-enriched proteins are biased towards those localized in the plasma membrane and vacuole. Deletion of 23 of the 74 mother-enriched proteins leads to lifespan extension, a fraction that is about six times that of the genes picked randomly from the genome. Among these lifespan-extending genes, three are involved in endosomal sorting/endosome to vacuole transport, and three are nitrogen source transporters. Tracking the dynamic expression of specific mother-enriched proteins revealed that their concentration steadily increases in the mother cells as they age, but is kept relatively low in the daughter cells via asymmetric distribution. Our results suggest that some mother-enriched proteins may increase to a concentration that becomes deleterious and lifespan-limiting in aged cells, possibly by upsetting homeostasis or leading to aberrant signaling. Our study provides a comprehensive resource for analyzing asymmetric cell division and aging in yeast, which should also be valuable for understanding similar phenomena in other organisms.
Collapse
|
49
|
Heras J, McClintock K, Sunagawa S, Aguilar A. Gonadal transcriptomics elucidate patterns of adaptive evolution within marine rockfishes (Sebastes). BMC Genomics 2015; 16:656. [PMID: 26329285 PMCID: PMC4557894 DOI: 10.1186/s12864-015-1870-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 08/20/2015] [Indexed: 12/12/2022] Open
Abstract
Background The genetic mechanisms of speciation and adaptation in the marine environment are not well understood. The rockfish genus Sebastes provides a unique model system for studying adaptive evolution because of the extensive diversity found within this group, which includes morphology, ecology, and a broad range of life spans. Examples of adaptive radiations within marine ecosystems are considered an anomaly due to the absence of geographical barriers and the presence of gene flow. Using marine rockfishes, we identified signatures of natural selection from transcriptomes developed from gonadal tissue of two rockfish species (Sebastes goodei and S. saxicola). We predicted orthologous transcript pairs, and estimated their distributions of nonsynonymous (Ka) and synonymous (Ks) substitution rates. Results We identified 144 genes out of 1079 orthologous pairs under positive selection, of which 11 are functionally annotated to reproduction based on gene ontologies (GOs). One orthologous pair of the zona pellucida gene family, which is known for its role in the selection of sperm by oocytes, out of ten was identified to be evolving under positive selection. In addition to our results in the protein coding-regions of transcripts, we found substitution rates in 3’ and 5’ UTRs to be significantly lower than Ks substitution rates implying negative selection in these regions. Conclusions We were able to identify a series of candidate genes that are useful for the assessment of the critical genes that diverged and are responsible for the radiation within this genus. Genes associated with longevity hold potential for understanding the molecular mechanisms that have contributed to the radiation within this genus. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1870-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Joseph Heras
- Department of Ecology and Evolutionary Biology, University of California Irvine, 321 Steinhaus Hall, Irvine, CA, 92697, USA.
| | - Kelly McClintock
- School of Natural Sciences and Graduate Group in Quantitative and Systems Biology, University of California Merced, 5200 N Lake Rd, Merced, CA, 95344, USA.
| | - Shinichi Sunagawa
- European Molecular Biology Laboratory, Meyerhofstr 1, 69117, Heidelberg, Germany.
| | - Andres Aguilar
- Department of Biological Sciences, California State University Los Angeles, 5151 State University Dr, Los Angeles, CA, 90032, USA.
| |
Collapse
|
50
|
Mitochondrial responsibility in ageing process: innocent, suspect or guilty. Biogerontology 2015; 16:599-620. [PMID: 26105157 DOI: 10.1007/s10522-015-9585-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 06/13/2015] [Indexed: 12/22/2022]
Abstract
Ageing is accompanied by the accumulation of damaged molecules in cells due to the injury produced by external and internal stressors. Among them, reactive oxygen species produced by cell metabolism, inflammation or other enzymatic processes are considered key factors. However, later research has demonstrated that a general mitochondrial dysfunction affecting electron transport chain activity, mitochondrial biogenesis and turnover, apoptosis, etc., seems to be in a central position to explain ageing. This key role is based on several effects from mitochondrial-derived ROS production to the essential maintenance of balanced metabolic activities in old organisms. Several studies have demonstrated caloric restriction, exercise or bioactive compounds mainly found in plants, are able to affect the activity and turnover of mitochondria by increasing biogenesis and mitophagy, especially in postmitotic tissues. Then, it seems that mitochondria are in the centre of metabolic procedures to be modified to lengthen life- or health-span. In this review we show the importance of mitochondria to explain the ageing process in different models or organisms (e.g. yeast, worm, fruitfly and mice). We discuss if the cause of aging is dependent on mitochondrial dysfunction of if the mitochondrial changes observed with age are a consequence of events taking place outside the mitochondrial compartment.
Collapse
|