51
|
Scherbakov D, Duscha S, Juskeviciene R, Restelli L, Frank S, Laczko E, Boettger EC. Mitochondrial misreading in skeletal muscle accelerates metabolic aging and confers lipid accumulation and increased inflammation. RNA (NEW YORK, N.Y.) 2020; 27:rna.077347.120. [PMID: 33262249 PMCID: PMC7901843 DOI: 10.1261/rna.077347.120] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/23/2020] [Indexed: 06/12/2023]
Abstract
We have recently reported on an experimental model of mitochondrial mistranslation conferred by amino acid exchange V338Y in the mitochondrial ribosomal protein MrpS5. Here we used a combination of RNA-Seq and metabolic profiling of homozygous transgenic MrpS5V338Y/V338Y mice to analyze the changes associated with the V338Y mutation in post-mitotic skeletal muscle. Metabolic profiling demonstrated age-dependent metabolic changes in the mutant V338Y animals, which included enhanced levels of age-associated metabolites and which were accompanied by increased glycolysis, lipid desaturation and eicosanoid biosynthesis, and alterations of the pentose phosphate pathway. In addition, transcriptome signatures of aged V338Y mutant muscle pointed to elevated inflammation, likely reflecting the increased levels of bioactive lipids. Our findings indicate that mistranslation-mediated chronic impairment of mitochondrial function affects specific bioenergetic processes in muscle in an age-dependent manner.
Collapse
|
52
|
The dialogue between the ubiquitin-proteasome system and autophagy: Implications in ageing. Ageing Res Rev 2020; 64:101203. [PMID: 33130248 DOI: 10.1016/j.arr.2020.101203] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/09/2020] [Accepted: 10/25/2020] [Indexed: 02/06/2023]
Abstract
Dysregulated proteostasis is one of the hallmarks of ageing. Damaged proteins may impair cellular function and their accumulation may lead to tissue dysfunction and disease. This is why protective mechanisms to safeguard the cell proteome have evolved. These mechanisms consist of cellular machineries involved in protein quality control, including regulators of protein translation, folding, trafficking and degradation. In eukaryotic cells, protein degradation occurs via two main pathways: the ubiquitin-proteasome system (UPS) and the autophagy-lysosome pathway. Although distinct pathways, they are not isolated systems and have a complementary nature, as evidenced by recent studies. These findings raise the question of how autophagy and the proteasome crosstalk. In this review we address how the two degradation pathways impact each other, thereby adding a new layer of regulation to protein degradation. We also analyze the implications of the UPS and autophagy in ageing.
Collapse
|
53
|
Zhou M, Zhang L, Yang Q, Yan C, Jiang P, Lan Y, Wang J, Tang R, He M, Lei G, Sun P, Su N, Price M, Li J, Lin F, Yue B, Fan Z. Age-related gene expression and DNA methylation changes in rhesus macaque. Genomics 2020; 112:5147-5156. [DOI: 10.1016/j.ygeno.2020.09.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/23/2020] [Accepted: 09/08/2020] [Indexed: 02/06/2023]
|
54
|
Abstract
Mitochondria contain about 1,000-1,500 proteins that fulfil multiple functions. Mitochondrial proteins originate from two genomes: mitochondrial and nuclear. Hence, proper mitochondrial function requires synchronization of gene expression in the nucleus and in mitochondria and necessitates efficient import of mitochondrial proteins into the organelle from the cytosol. Furthermore, the mitochondrial proteome displays high plasticity to allow the adaptation of mitochondrial function to cellular requirements. Maintenance of this complex and adaptable mitochondrial proteome is challenging, but is of crucial importance to cell function. Defects in mitochondrial proteostasis lead to proteotoxic insults and eventually cell death. Different quality control systems monitor the mitochondrial proteome. The cytosolic ubiquitin-proteasome system controls protein transport across the mitochondrial outer membrane and removes damaged or mislocalized proteins. Concomitantly, a number of mitochondrial chaperones and proteases govern protein folding and degrade damaged proteins inside mitochondria. The quality control factors also regulate processing and turnover of native proteins to control protein import, mitochondrial metabolism, signalling cascades, mitochondrial dynamics and lipid biogenesis, further ensuring proper function of mitochondria. Thus, mitochondrial protein quality control mechanisms are of pivotal importance to integrate mitochondria into the cellular environment.
Collapse
|
55
|
Gamage R, Wagnon I, Rossetti I, Childs R, Niedermayer G, Chesworth R, Gyengesi E. Cholinergic Modulation of Glial Function During Aging and Chronic Neuroinflammation. Front Cell Neurosci 2020; 14:577912. [PMID: 33192323 PMCID: PMC7594524 DOI: 10.3389/fncel.2020.577912] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/07/2020] [Indexed: 12/12/2022] Open
Abstract
Aging is a complex biological process that increases the risk of age-related cognitive degenerative diseases such as dementia, including Alzheimer’s disease (AD), Lewy Body Dementia (LBD), and mild cognitive impairment (MCI). Even non-pathological aging of the brain can involve chronic oxidative and inflammatory stress, which disrupts the communication and balance between the brain and the immune system. There has been an increasingly strong connection found between chronic neuroinflammation and impaired memory, especially in AD. While microglia and astrocytes, the resident immune cells of the central nervous system (CNS), exerting beneficial effects during the acute inflammatory phase, during chronic neuroinflammation they can become more detrimental. Central cholinergic circuits are involved in maintaining normal cognitive function and regulating signaling within the entire cerebral cortex. While neuronal-glial cholinergic signaling is anti-inflammatory and anti-oxidative, central cholinergic neuronal degeneration is implicated in impaired learning, memory sleep regulation, and attention. Although there is evidence of cholinergic involvement in memory, fewer studies have linked the cholinergic anti-inflammatory and anti-oxidant pathways to memory processes during development, normal aging, and disease states. This review will summarize the current knowledge of cholinergic effects on microglia and astroglia, and their role in both anti-inflammatory and anti-oxidant mechanisms, concerning normal aging and chronic neuroinflammation. We provided details on how stimulation of α7 nicotinic acetylcholine (α7nACh) receptors can be neuroprotective by increasing amyloid-β phagocytosis, decreasing inflammation and reducing oxidative stress by promoting the nuclear factor erythroid 2-related factor 2 (Nrf2) pathways and decreasing the release of pro-inflammatory cytokines. There is also evidence for astroglial α7nACh receptor stimulation mediating anti-inflammatory and antioxidant effects by inhibiting the nuclear factor-κB (NF-κB) pathway and activating the Nrf2 pathway respectively. We conclude that targeting cholinergic glial interactions between neurons and glial cells via α7nACh receptors could regulate neuroinflammation and oxidative stress, relevant to the treatment of several neurodegenerative diseases.
Collapse
Affiliation(s)
- Rashmi Gamage
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Ingrid Wagnon
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Ilaria Rossetti
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Ryan Childs
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Garry Niedermayer
- School of Science, Western Sydney University, Penrith, NSW, Australia
| | - Rose Chesworth
- School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Erika Gyengesi
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| |
Collapse
|
56
|
Webb M, Sideris DP. Intimate Relations-Mitochondria and Ageing. Int J Mol Sci 2020; 21:ijms21207580. [PMID: 33066461 PMCID: PMC7589147 DOI: 10.3390/ijms21207580] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/14/2022] Open
Abstract
Mitochondrial dysfunction is associated with ageing, but the detailed causal relationship between the two is still unclear. We review the major phenomenological manifestations of mitochondrial age-related dysfunction including biochemical, regulatory and energetic features. We conclude that the complexity of these processes and their inter-relationships are still not fully understood and at this point it seems unlikely that a single linear cause and effect relationship between any specific aspect of mitochondrial biology and ageing can be established in either direction.
Collapse
Affiliation(s)
- Michael Webb
- Mitobridge Inc., an Astellas Company, 1030 Massachusetts Ave, Cambridge, MA 02138, USA
| | - Dionisia P Sideris
- Mitobridge Inc., an Astellas Company, 1030 Massachusetts Ave, Cambridge, MA 02138, USA
| |
Collapse
|
57
|
Xie K, Kapetanou M, Sidiropoulou K, Bano D, Gonos ES, Djordjevic AM, Ehninger D. Signaling pathways of dietary energy restriction and metabolism on brain physiology and in age-related neurodegenerative diseases. Mech Ageing Dev 2020; 192:111364. [PMID: 32991920 DOI: 10.1016/j.mad.2020.111364] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 08/17/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023]
Abstract
Several laboratory animal models have shown that dietary energy restriction (ER) can promote longevity and improve various health aspects in old age. However, whether the entire spectrum of ER-induced short- and long-term physiological and metabolic adaptions is translatable to humans remains to be determined. In this review article, we present recent evidence towards the elucidation of the impact of ER on brain physiology and in age-related neurodegenerative diseases. We also discuss modulatory influences of ER on metabolism and overall on human health, limitations of current experimental designs as well as future perspectives for ER trials in humans. Finally, we summarize signaling pathways and processes known to be affected by both aging and ER with a special emphasis on the link between ER and cellular proteostasis.
Collapse
Affiliation(s)
- Kan Xie
- Molecular and Cellular Cognition Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany
| | - Marianna Kapetanou
- Institute of Chemical Biology, National Hellenic Research Foundation, 48 Vassileos Constantinou Ave., Athens, 11635, Greece
| | | | - Daniele Bano
- Aging and Neurodegeneration Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany
| | - Efstathios S Gonos
- Institute of Chemical Biology, National Hellenic Research Foundation, 48 Vassileos Constantinou Ave., Athens, 11635, Greece
| | - Aleksandra Mladenovic Djordjevic
- Department of Neurobiology, Institute for Biological Research 'Sinisa Stankovic', University of Belgrade, National Institute of Republic of Serbia, Boulevard Despota Stefana 142, 11000 Belgrade, Serbia
| | - Dan Ehninger
- Molecular and Cellular Cognition Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127 Bonn, Germany.
| |
Collapse
|
58
|
Adornetto A, Morrone LA, Satriano A, Laganà ML, Licastro E, Nucci C, Corasaniti MT, Tonin P, Bagetta G, Russo R. Effects of caloric restriction on retinal aging and neurodegeneration. PROGRESS IN BRAIN RESEARCH 2020; 256:189-207. [PMID: 32958212 DOI: 10.1016/bs.pbr.2020.07.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Glaucoma is the most common neurodegenerative cause of irreversible blindness worldwide. Restricted caloric regimens are an attractive approach for delaying the progression of neurodegenerative diseases. Here we review the current literature on the effects of caloric restriction on retinal neurons, under physiological and pathological conditions. We focused on autophagy as one of the mechanisms modulated by restricted caloric regimens and involved in the death of retinal ganglion cells (RGCs) over the course of glaucoma.
Collapse
Affiliation(s)
- Annagrazia Adornetto
- Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Luigi Antonio Morrone
- Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Andrea Satriano
- Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Maria Luisa Laganà
- Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Ester Licastro
- Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Carlo Nucci
- Ophthalmology Unit, Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Maria Tiziana Corasaniti
- School of Hospital Pharmacy, University "Magna Graecia" of Catanzaro and Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Paolo Tonin
- Regional Center for Serious Brain Injuries, S. Anna Institute, Crotone, Italy
| | - Giacinto Bagetta
- Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Rossella Russo
- Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy.
| |
Collapse
|
59
|
Zou K, Rouskin S, Dervishi K, McCormick MA, Sasikumar A, Deng C, Chen Z, Kaeberlein M, Brem RB, Polymenis M, Kennedy BK, Weissman JS, Zheng J, Ouyang Q, Li H. Life span extension by glucose restriction is abrogated by methionine supplementation: Cross-talk between glucose and methionine and implication of methionine as a key regulator of life span. SCIENCE ADVANCES 2020; 6:eaba1306. [PMID: 32821821 PMCID: PMC7406366 DOI: 10.1126/sciadv.aba1306] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 06/22/2020] [Indexed: 05/03/2023]
Abstract
Caloric restriction (CR) is known to extend life span across species; however, the molecular mechanisms are not well understood. We investigate the mechanism by which glucose restriction (GR) extends yeast replicative life span, by combining ribosome profiling and RNA-seq with microfluidic-based single-cell analysis. We discovered a cross-talk between glucose sensing and the regulation of intracellular methionine: GR down-regulated the transcription and translation of methionine biosynthetic enzymes and transporters, leading to a decreased intracellular methionine concentration; external supplementation of methionine cancels the life span extension by GR. Furthermore, genetic perturbations that decrease methionine synthesis/uptake extend life span. These observations suggest that intracellular methionine mediates the life span effects of various nutrient and genetic perturbations, and that the glucose-methionine cross-talk is a general mechanism for coordinating the nutrient status and the translation/growth of a cell. Our work also implicates proteasome as a downstream effector of the life span extension by GR.
Collapse
Affiliation(s)
- Ke Zou
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
- The State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences at Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Silvia Rouskin
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California Institute for Quantitative Biomedical Research and Howard Hughes Medical Institute, San Francisco, CA 94158, USA
| | | | - Mark A. McCormick
- Department of Biochemistry and Molecular Biology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
- Autophagy Inflammation and Metabolism Center of Biomedical Research Excellence, Albuquerque, NM 87131, USA
| | | | - Changhui Deng
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Zhibing Chen
- Department of Physical Therapy and Rehabilitation Science, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Matt Kaeberlein
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Rachel B. Brem
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Michael Polymenis
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Brian K. Kennedy
- Buck Institute for Research on Aging, Novato, CA 94945, USA
- Departments of Biochemistry and Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Centre for Healthy Longevity, National University Health System, Singapore
- Singapore Institute for Clinical Sciences, A*STAR, Singapore
| | - Jonathan S. Weissman
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California Institute for Quantitative Biomedical Research and Howard Hughes Medical Institute, San Francisco, CA 94158, USA
| | - Jiashun Zheng
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Qi Ouyang
- The State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences at Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Hao Li
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
60
|
Fonseca Costa SS, Robinson-Rechavi M, Ripperger JA. Single-cell transcriptomics allows novel insights into aging and circadian processes. Brief Funct Genomics 2020; 19:343-349. [PMID: 32633783 PMCID: PMC7716582 DOI: 10.1093/bfgp/elaa014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/18/2020] [Accepted: 06/04/2020] [Indexed: 12/14/2022] Open
Abstract
Aging and circadian rhythms are two biological processes that affect an organism, although at different time scales. Nevertheless, due to the overlap of their actions, it was speculated that both interfere or interact with each other. However, to address this question, a much deeper insight into these processes is necessary, especially at the cellular level. New methods such as single-cell RNA-sequencing (scRNA-Seq) have the potential to close this gap in our knowledge. In this review, we analyze applications of scRNA-Seq from the aging and circadian rhythm fields and highlight new findings emerging from the analysis of single cells, especially in humans or rodents. Furthermore, we judge the potential of scRNA-Seq to identify common traits of both processes. Overall, this method offers several advantages over more traditional methods analyzing gene expression and will become an important tool to unravel the link between these biological processes.
Collapse
Affiliation(s)
- Sara S Fonseca Costa
- Department of Ecology and Evolution, University of Lausanne, 1015 Lausanne, Switzerland.,SIB Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Marc Robinson-Rechavi
- Department of Ecology and Evolution, University of Lausanne, 1015 Lausanne, Switzerland.,SIB Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | | |
Collapse
|
61
|
Abstract
IMPACT STATEMENT NAD is a central metabolite connecting energy balance and organismal growth with genomic integrity and function. It is involved in the development of malignancy and has a regulatory role in the aging process. These processes are mediated by a diverse series of enzymes whose common focus is either NAD's biosynthesis or its utilization as a redox cofactor or enzyme substrate. These enzymes include dehydrogenases, cyclic ADP-ribose hydrolases, mono(ADP-ribosyl)transferases, poly(ADP-ribose) polymerases, and sirtuin deacetylases. This article describes the manifold pathways that comprise NAD metabolism and promotes an increased awareness of how perturbations in these systems may be important in disease prevention and/or progression.
Collapse
Affiliation(s)
- John Wr Kincaid
- Department of Nutrition, 12304Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.,151230Case Comprehensive Cancer Center, 12304Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Nathan A Berger
- 151230Case Comprehensive Cancer Center, 12304Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.,Department of Biochemistry, 12304Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.,Department of Genetics and Genome Sciences, 12304Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.,Department of Medicine, 12304Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.,Center for Science, Health and Society, 12304Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
62
|
Mastaloudis A, Sheth C, Hester SN, Wood SM, Prescot A, McGlade E, Renshaw PF, Yurgelun-Todd DA. Supplementation with a putative calorie restriction mimetic micronutrient blend increases glutathione concentrations and improves neuroenergetics in brain of healthy middle-aged men and women. Free Radic Biol Med 2020; 153:112-121. [PMID: 32335159 DOI: 10.1016/j.freeradbiomed.2020.04.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/17/2020] [Accepted: 04/19/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND Caloric restriction (CR) without micronutrient deficiency has been shown to increase both lifespan and healthspan. In animals, CR has been demonstrated to increase glutathione (GSH), a neuroprotective antioxidant, in the brain and preserve brain mitochondrial function by altering neuroenergetics. In humans it has been associated with improvements in mood states and cognitive function. However, most CR studies have employed a 30-60% reduction in calories which is likely too stringent for most people to adhere to long-term. Thus, there is an unmet need for nutritional supplements which can mimic the biological effects of CR, without the need for calorie limitations. AIM The purpose of the present randomized, placebo-controlled clinical trial was to use Proton (1H) Magnetic Resonance Spectroscopic (MRS) measurements to determine non-invasively whether a blend of micronutrients, a putative CR mimetic, positively modulates metabolites related to neuroprotection and neuroenergetics in the brain. METHODS Healthy middle-aged men and women (N = 63 [33 women]; age: 40-60 years) were randomized in a double-blind manner to 6 weeks supplementation with either the putative CR mimetic or placebo. At baseline and 6 weeks, subjects underwent MRS at 3 T to investigate changes in brain chemistry, including the neurometabolites: GSH, Glutamate (Glu), Glutamine (Gln) and N-Acetylaspartate (NAA). RESULTS GSH, a marker of antioxidant and cellular redox status, increased in the brain of participants in the supplement group. The supplement group also showed an increase in the Glu/Gln ratio, a marker of excitatory neurotransmission and bioenergetics. A trend for an increase in NAA/H2O, a marker of neuronal integrity, was observed in females in the supplement group. CONCLUSIONS The present study reveals that 6-weeks daily supplementation with a micronutrient blend elicits positive changes in brain neurochemistry. This is the first study to demonstrate that a putative CR mimetic increases brain GSH concentrations and improves neuroprotection and neuroenergetics in the brain of healthy humans. This study was registered at www.clinicaltrials.gov as NCT02439983.
Collapse
Affiliation(s)
| | - Chandni Sheth
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT, USA; Diagnostic Neuroimaging, University of Utah, Salt Lake City, UT, USA.
| | | | - Steven M Wood
- Pharmanex Research, NSE Products, Inc., Provo, UT, USA
| | - Andrew Prescot
- Department of Radiology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Erin McGlade
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT, USA; Diagnostic Neuroimaging, University of Utah, Salt Lake City, UT, USA; George E. Wahlen Department of Veterans Affairs Medical Center, VA VISN 19 Mental Illness Research, Education and Clinical Center (MIRREC), Salt Lake City, UT, USA
| | - Perry F Renshaw
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT, USA; Diagnostic Neuroimaging, University of Utah, Salt Lake City, UT, USA; George E. Wahlen Department of Veterans Affairs Medical Center, VA VISN 19 Mental Illness Research, Education and Clinical Center (MIRREC), Salt Lake City, UT, USA
| | - Deborah A Yurgelun-Todd
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT, USA; Diagnostic Neuroimaging, University of Utah, Salt Lake City, UT, USA; George E. Wahlen Department of Veterans Affairs Medical Center, VA VISN 19 Mental Illness Research, Education and Clinical Center (MIRREC), Salt Lake City, UT, USA
| |
Collapse
|
63
|
Buffenstein R, Lewis KN, Gibney PA, Narayan V, Grimes KM, Smith M, Lin TD, Brown-Borg HM. Probing Pedomorphy and Prolonged Lifespan in Naked Mole-Rats and Dwarf Mice. Physiology (Bethesda) 2020; 35:96-111. [DOI: 10.1152/physiol.00032.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Pedomorphy, maintenance of juvenile traits throughout life, is most pronounced in extraordinarily long-lived naked mole-rats. Many of these traits (e.g., slow growth rates, low hormone levels, and delayed sexual maturity) are shared with spontaneously mutated, long-lived dwarf mice. Although some youthful traits likely evolved as adaptations to subterranean habitats (e.g., thermolability), the nature of these intrinsic pedomorphic features may also contribute to their prolonged youthfulness, longevity, and healthspan.
Collapse
Affiliation(s)
| | | | - Patrick A. Gibney
- Calico Life Sciences LLC, South San Francisco, California
- Department of Food Science, College of Agriculture and Life Sciences, Stocking Hall, Cornell University, Ithaca, New York
| | - Vikram Narayan
- Calico Life Sciences LLC, South San Francisco, California
| | - Kelly M. Grimes
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Megan Smith
- Calico Life Sciences LLC, South San Francisco, California
| | - Tzuhua D. Lin
- Calico Life Sciences LLC, South San Francisco, California
| | - Holly M. Brown-Borg
- Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota
| |
Collapse
|
64
|
Deshwal S, Fiedler KU, Langer T. Mitochondrial Proteases: Multifaceted Regulators of Mitochondrial Plasticity. Annu Rev Biochem 2020; 89:501-528. [PMID: 32075415 DOI: 10.1146/annurev-biochem-062917-012739] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mitochondria are essential metabolic hubs that dynamically adapt to physiological demands. More than 40 proteases residing in different compartments of mitochondria, termed mitoproteases, preserve mitochondrial proteostasis and are emerging as central regulators of mitochondrial plasticity. These multifaceted enzymes limit the accumulation of short-lived, regulatory proteins within mitochondria, modulate the activity of mitochondrial proteins by protein processing, and mediate the degradation of damaged proteins. Various signaling cascades coordinate the activity of mitoproteases to preserve mitochondrial homeostasis and ensure cell survival. Loss of mitoproteases severely impairs the functional integrity of mitochondria, is associated with aging, and causes pleiotropic diseases. Understanding the dual function of mitoproteases as regulatory and quality control enzymes will help unravel the role of mitochondrial plasticity in aging and disease.
Collapse
Affiliation(s)
- Soni Deshwal
- Max Planck Institute for Biology of Ageing, 50931 Cologne, Germany;
| | - Kai Uwe Fiedler
- Max Planck Institute for Biology of Ageing, 50931 Cologne, Germany;
| | - Thomas Langer
- Max Planck Institute for Biology of Ageing, 50931 Cologne, Germany; .,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
65
|
Serna E, Mastaloudis A, Martorell P, Wood SM, Hester SN, Bartlett M, Prolla TA, Viña J. A Novel Micronutrient Blend Mimics Calorie Restriction Transcriptomics in Multiple Tissues of Mice and Increases Lifespan and Mobility in C. elegans. Nutrients 2020; 12:nu12020486. [PMID: 32075050 PMCID: PMC7071149 DOI: 10.3390/nu12020486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/07/2020] [Accepted: 02/10/2020] [Indexed: 11/17/2022] Open
Abstract
Background: We previously described a novel micronutrient blend that behaves like a putative calorie restriction mimetic. The aim of this paper was to analyze the beneficial effects of our micronutrient blend in mice and C. elegans, and compare them with calorie restriction. Methods: Whole transcriptomic analysis was performed in the brain cortex, skeletal muscle and heart in three groups of mice: old controls (30 months), old + calorie restriction and old + novel micronutrient blend. Longevity and vitality were tested in C. elegans. Results: The micronutrient blend elicited transcriptomic changes in a manner similar to those in the calorie-restricted group and different from those in the control group. Subgroup analysis revealed that nuclear hormone receptor, proteasome complex and angiotensinogen genes, all of which are known to be directly related to aging, were the most affected. Furthermore, a functional analysis in C. elegans was used. We found that feeding C. elegans the micronutrient blend increased longevity as well as vitality. Conclusions: We describe a micronutrient supplement that causes similar changes (transcriptomic and promoting longevity and vitality) as a calorie restriction in mice and C. elegans, respectively, but further studies are required to confirm these effects in humans.
Collapse
Affiliation(s)
- Eva Serna
- Freshage Research Group-Dept. Physiology-University of Valencia, CIBERFES, INCLIVA, 46010 Valencia, Spain;
| | - Angela Mastaloudis
- Pharmanex Research, NSE Products, Inc., Provo, UT 84601, USA; (A.M.); (S.M.W.); (S.N.H.); (M.B.)
| | - Patricia Martorell
- Cell Biology Laboratory/ADM Nutrition/Biopolis SL/Archer Daniels Midland, 46980 Paterna, Valencia, Spain;
| | - Steven M. Wood
- Pharmanex Research, NSE Products, Inc., Provo, UT 84601, USA; (A.M.); (S.M.W.); (S.N.H.); (M.B.)
| | - Shelly N. Hester
- Pharmanex Research, NSE Products, Inc., Provo, UT 84601, USA; (A.M.); (S.M.W.); (S.N.H.); (M.B.)
| | - Mark Bartlett
- Pharmanex Research, NSE Products, Inc., Provo, UT 84601, USA; (A.M.); (S.M.W.); (S.N.H.); (M.B.)
| | - Tomas A. Prolla
- LifeGen Technologies LLC, Madison, WI 53719, USA;
- Departments of Genetics and Medical Genetics; University of Wisconsin; Madison, WI 53706, USA
| | - Jose Viña
- Freshage Research Group-Dept. Physiology-University of Valencia, CIBERFES, INCLIVA, 46010 Valencia, Spain;
- Correspondence: ; Tel.: +34-963864650
| |
Collapse
|
66
|
Huminiecki L, Atanasov AG, Horbańczuk J. Etiology of atherosclerosis informs choice of animal models and tissues for initial functional genomic studies of resveratrol. Pharmacol Res 2020; 156:104598. [PMID: 32067842 DOI: 10.1016/j.phrs.2019.104598] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 12/09/2019] [Accepted: 12/11/2019] [Indexed: 01/03/2023]
Abstract
Resveratrol, a phytoalexin, is a natural polyphenol synthesized exclusively by plants in response to environmental stresses. However, the molecule has also many exogenous bioactivities in animal cells. These bioactivities may lead to anti-cancer and cardio-protective health benefits. Because cellular responses to the treatment with resveratrol include the changes of expression patterns, functional genomics is an attractive tool to study them. In recent and today's experimental practice, this mostly means microarray profiling of gene expression (using RNAs isolated from bulk tissues). Herein, we review such published studies undertaken in the context of cardiovascular diseases (CVDs). CVDs are a number one public health problem in developed countries, outweighing in magnitude even cancer. In particular, we review the studies of resveratrol in several animal models relevant to CVDs. These models included: normal and pre-mature aging in mice, as well as atherogenic diet in mice / pigs / non-human primates. Additionally, there were few clinical studies published in the context of the comorbidities of atherosclerosis in humans (e.g. obesity, diabetes, hypertension). For the purposes of these studies, three types of samples were most commonly profiled with microarrays: the liver, the skeletal muscle, and peripheral blood mononuclear cells. Resveratrol-induced changes of gene expression typically mimicked those associated with calorie restriction and lifespan extension. They also opposed changes induced by the atherogenic diet. We conclude by discussing few experimental factors that were relatively neglected thus far, but which could be interesting to investigate in the future. These factors include sex and the exact formulation of resveratrol (plant extract, or synthetic chemical).
Collapse
Affiliation(s)
- Lukasz Huminiecki
- The Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Postępu 36A, Jastrzębiec, 05-552 Magdalenka, Poland.
| | - Atanas G Atanasov
- The Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Postępu 36A, Jastrzębiec, 05-552 Magdalenka, Poland; Department of Pharmacognosy, Faculty of Life Sciences, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria
| | - Jarosław Horbańczuk
- The Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Postępu 36A, Jastrzębiec, 05-552 Magdalenka, Poland
| |
Collapse
|
67
|
The Mitochondrial Lon Protease: Novel Functions off the Beaten Track? Biomolecules 2020; 10:biom10020253. [PMID: 32046155 PMCID: PMC7072132 DOI: 10.3390/biom10020253] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 12/11/2022] Open
Abstract
To maintain organellar function, mitochondria contain an elaborate endogenous protein quality control system. As one of the two soluble energy-dependent proteolytic enzymes in the matrix compartment, the protease Lon is a major component of this system, responsible for the degradation of misfolded proteins, in particular under oxidative stress conditions. Lon defects have been shown to negatively affect energy production by oxidative phosphorylation but also mitochondrial gene expression. In this review, recent studies on the role of Lon in mammalian cells, in particular on its protective action under diverse stress conditions and its relationship to important human diseases are summarized and commented.
Collapse
|
68
|
McCracken AW, Adams G, Hartshorne L, Tatar M, Simons MJP. The hidden costs of dietary restriction: Implications for its evolutionary and mechanistic origins. SCIENCE ADVANCES 2020; 6:eaay3047. [PMID: 32128403 PMCID: PMC7034997 DOI: 10.1126/sciadv.aay3047] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 12/04/2019] [Indexed: 05/14/2023]
Abstract
Dietary restriction (DR) extends life span across taxa. Despite considerable research, universal mechanisms of DR have not been identified, limiting its translational potential. Guided by the conviction that DR evolved as an adaptive, pro-longevity physiological response to food scarcity, biomedical science has interpreted DR as an activator of pro-longevity molecular pathways. Current evolutionary theory predicts that organisms invest in their soma during DR, and thus when resource availability improves, should outcompete rich-fed controls in survival and/or reproduction. Testing this prediction in Drosophila melanogaster (N > 66,000 across 11 genotypes), our experiments revealed substantial, unexpected mortality costs when flies returned to a rich diet following DR. The physiological effects of DR should therefore not be interpreted as intrinsically pro-longevity, acting via somatic maintenance. We suggest DR could alternatively be considered an escape from costs incurred under nutrient-rich conditions, in addition to costs associated with DR.
Collapse
Affiliation(s)
- Andrew W. McCracken
- Department of Animal and Plant Sciences and Bateson Centre, The University of Sheffield, Sheffield S10 2TN, UK
| | - Gracie Adams
- Department of Animal and Plant Sciences and Bateson Centre, The University of Sheffield, Sheffield S10 2TN, UK
| | - Laura Hartshorne
- Department of Animal and Plant Sciences and Bateson Centre, The University of Sheffield, Sheffield S10 2TN, UK
| | - Marc Tatar
- Department of Ecology and Evolutionary Biology, Brown University, Providence, RI, USA
| | - Mirre J. P. Simons
- Department of Animal and Plant Sciences and Bateson Centre, The University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
69
|
Shukla R, Oh H, Sibille E. Molecular and Cellular Evidence for Age by Disease Interactions: Updates and Path Forward. Am J Geriatr Psychiatry 2020; 28:237-247. [PMID: 31285153 DOI: 10.1016/j.jagp.2019.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/14/2019] [Accepted: 06/01/2019] [Indexed: 12/31/2022]
Abstract
Characterization of age-associated gene expression changes shows that the brain engages a specific set of genes and biologic pathways along a continuous life-long trajectory and that these genes and pathways overlap with those associated with brain-related disorders. Based on this correlative observation, we have suggested a model of age-by-disease interaction by which brain ageing promotes biologic changes associated with diseases and where deviations from expected age-related trajectories, due to biologic and environmental factors, contribute to defining disease risk or resiliency. In this review, we first evaluate various biomarkers that can be used to study age-by-disease interactions and then focus on transcriptome analysis (i.e., the set of all expressed genes) as a useful tool to explore this interaction. Using the specific example of brain-derived neurotrophic factor and brain-derived neurotrophic factor-associated genes, we then describe molecular events and mechanisms potentially contributing to age-by-disease interactions. Finally, we suggest that long-term biologic adaptations within distinct cellular components of cortical microcircuits, as determined by transcriptome analysis, may integrate and mediate the effects of ageing and diseases. Moving forward, we suggest that analysis of transcriptome similarities between ageing and small molecule-induced system perturbations may lead to novel therapeutics discovery.
Collapse
Affiliation(s)
- Rammohan Shukla
- Campbell Family Mental Health Research Institute of CAMH, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Hyunjung Oh
- Campbell Family Mental Health Research Institute of CAMH, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute of CAMH, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
70
|
Katsimpardi L, Kuperwasser N, Camus C, Moigneu C, Chiche A, Tolle V, Li H, Kokovay E, Lledo P. Systemic GDF11 stimulates the secretion of adiponectin and induces a calorie restriction-like phenotype in aged mice. Aging Cell 2020; 19:e13038. [PMID: 31637864 PMCID: PMC6974718 DOI: 10.1111/acel.13038] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 07/06/2019] [Accepted: 08/19/2019] [Indexed: 01/11/2023] Open
Abstract
Aging is a negative regulator of general homeostasis, tissue function, and regeneration. Changes in organismal energy levels and physiology, through systemic manipulations such as calorie restriction and young blood infusion, can regenerate tissue activity and increase lifespan in aged mice. However, whether these two systemic manipulations could be linked has never been investigated. Here, we report that systemic GDF11 triggers a calorie restriction-like phenotype without affecting appetite or GDF15 levels in the blood, restores the insulin/IGF-1 signaling pathway, and stimulates adiponectin secretion from white adipose tissue by direct action on adipocytes, while repairing neurogenesis in the aged brain. These findings suggest that GDF11 has a pleiotropic effect on an organismal level and that it could be a linking mechanism of rejuvenation between heterochronic parabiosis and calorie restriction. As such, GDF11 could be considered as an important therapeutic candidate for age-related neurodegenerative and metabolic disorders.
Collapse
Affiliation(s)
- Lida Katsimpardi
- Perception and Memory Lab Neuroscience Department Institut Pasteur Paris France
- Centre National de la Recherche Scientifique Unité Mixte de Recherche 3571 Paris France
| | - Nicolas Kuperwasser
- Department of Cell Growth and Signaling Institut National de la Santé et de la Recherche Médicale (INSERM) U1151Institut Necker Enfants Malades (INEM)Université Paris Descartes France
| | - Claire Camus
- Perception and Memory Lab Neuroscience Department Institut Pasteur Paris France
- Centre National de la Recherche Scientifique Unité Mixte de Recherche 3571 Paris France
| | - Carine Moigneu
- Perception and Memory Lab Neuroscience Department Institut Pasteur Paris France
- Centre National de la Recherche Scientifique Unité Mixte de Recherche 3571 Paris France
| | - Aurélie Chiche
- Department of Developmental & Stem Cell Biology Cellular Plasticity & Disease Modelling CNRS UMR 3738Institut Pasteur Paris France
| | - Virginie Tolle
- Centre de Psychiatrie et Neurosciences UMR‐S 894INSERMUniversité Paris Descartes Sorbonne Paris Cité Paris France
| | - Han Li
- Department of Developmental & Stem Cell Biology Cellular Plasticity & Disease Modelling CNRS UMR 3738Institut Pasteur Paris France
| | - Erzsebet Kokovay
- Cell Systems and Anatomy Brashop Institute for Longevity and Aging Studies University of Texas Health Science Center at San Antonio San Antonio TX USA
| | - Pierre‐Marie Lledo
- Perception and Memory Lab Neuroscience Department Institut Pasteur Paris France
- Centre National de la Recherche Scientifique Unité Mixte de Recherche 3571 Paris France
| |
Collapse
|
71
|
Control of Inflammation by Calorie Restriction Mimetics: On the Crossroad of Autophagy and Mitochondria. Cells 2019; 9:cells9010082. [PMID: 31905682 PMCID: PMC7017321 DOI: 10.3390/cells9010082] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 12/17/2019] [Accepted: 12/25/2019] [Indexed: 01/07/2023] Open
Abstract
Mitochondrial metabolism and autophagy are two of the most metabolically active cellular processes, playing a crucial role in regulating organism longevity. In fact, both mitochondrial dysfunction or autophagy decline compromise cellular homeostasis and induce inflammation. Calorie restriction (CR) is the oldest strategy known to promote healthspan, and a plethora of CR mimetics have been used to emulate its beneficial effects. Herein, we discuss how CR and CR mimetics, by modulating mitochondrial metabolism or autophagic flux, prevent inflammatory processes, protect the intestinal barrier function, and dampen both inflammaging and neuroinflammation. We outline the effects of some compounds classically known as modulators of autophagy and mitochondrial function, such as NAD+ precursors, metformin, spermidine, rapamycin, and resveratrol, on the control of the inflammatory cascade and how these anti-inflammatory properties could be involved in their ability to increase resilience to age-associated diseases.
Collapse
|
72
|
Caloric restriction rescues yeast cells from alpha-synuclein toxicity through autophagic control of proteostasis. Aging (Albany NY) 2019; 10:3821-3833. [PMID: 30530923 PMCID: PMC6326672 DOI: 10.18632/aging.101675] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 11/18/2018] [Indexed: 01/31/2023]
Abstract
α-Synuclein (SNCA) is a presynaptic protein that is associated with the pathophysiology of synucleinopathies, including Parkinson's disease. SNCA is a naturally aggregation-prone protein, which may be degraded by the ubiquitin-proteasome system (UPS) and by lysosomal degradation pathways. Besides being a target of the proteolytic systems, SNCA can also alter the function of these pathways further, contributing to the progression of neurodegeneration. Deterioration of UPS and autophagy activities with aging further aggravates this toxic cycle. Caloric restriction (CR) is still the most effective non-genetic intervention promoting lifespan extension. It is known that CR-mediated lifespan extension is linked to the regulation of proteolytic systems, but the mechanisms underlying CR rescue of SNCA toxicity remain poorly understood. This study shows that CR balances UPS and autophagy activities during aging. CR enhances UPS activity, reversing the decline of the UPS activity promoted by SNCA, and keeps autophagy at homeostatic levels. Maintenance of autophagy at homeostatic levels appears to be relevant for UPS activity and for the mechanism underlying rescue of cells from SNCA-mediated toxicity by CR.
Collapse
|
73
|
Candesartan Neuroprotection in Rat Primary Neurons Negatively Correlates with Aging and Senescence: a Transcriptomic Analysis. Mol Neurobiol 2019; 57:1656-1673. [PMID: 31811565 DOI: 10.1007/s12035-019-01800-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 09/22/2019] [Indexed: 12/11/2022]
Abstract
Preclinical experiments and clinical trials demonstrated that angiotensin II AT1 receptor overactivity associates with aging and cellular senescence and that AT1 receptor blockers (ARBs) protect from age-related brain disorders. In a primary neuronal culture submitted to glutamate excitotoxicity, gene set enrichment analysis (GSEA) revealed expression of several hundred genes altered by glutamate and normalized by candesartan correlated with changes in expression in Alzheimer's patient's hippocampus. To further establish whether our data correlated with gene expression alterations associated with aging and senescence, we compared our global transcriptional data with additional published datasets, including alterations in gene expression in the neocortex and cerebellum of old mice, human frontal cortex after age of 40, gene alterations in the Werner syndrome, rodent caloric restriction, Ras and oncogene-induced senescence in fibroblasts, and to tissues besides the brain such as the muscle and kidney. The most significant and enriched pathways associated with aging and senescence were positively correlated with alterations in gene expression in glutamate-injured neurons and, conversely, negatively correlated when the injured neurons were treated with candesartan. Our results involve multiple genes and pathways, including CAV1, CCND1, CDKN1A, CHEK1, ICAM1, IL-1B, IL-6, MAPK14, PTGS2, SERPINE1, and TP53, encoding proteins associated with aging and senescence hallmarks, such as inflammation, oxidative stress, cell cycle and mitochondrial function alterations, insulin resistance, genomic instability including telomere shortening and DNA damage, and the senescent-associated secretory phenotype. Our results demonstrate that AT1 receptor blockade ameliorates central mechanisms of aging and senescence. Using ARBs for prevention and treatment of age-related disorders has important translational value.
Collapse
|
74
|
Ozkurede U, Miller RA. Improved mitochondrial stress response in long-lived Snell dwarf mice. Aging Cell 2019; 18:e13030. [PMID: 31423721 PMCID: PMC6826134 DOI: 10.1111/acel.13030] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 07/31/2019] [Accepted: 08/06/2019] [Indexed: 12/20/2022] Open
Abstract
Prolonged lifespan and improved health in late adulthood can be achieved by partial inhibition of mitochondrial proteins in yeast, worms, fruit flies, and mice. Upregulation of the mitochondrial unfolded protein response (mtUPR) has been proposed as a common pathway in lifespan extension induced by mitochondrial defects. However, it is not known whether mtUPR is elevated in long‐lived mouse models. Here, we report that Snell dwarf mice, which show 30%–40% lifespan extension and prolonged healthspan, exhibit augmented mitochondrial stress responses. Cultured cells from Snell mice show elevated levels of the mitochondrial chaperone HSP60 and mitochondrial protease LONP1, two components of the mtUPR. In response to mitochondrial stress, the increase in Tfam (mitochondrial transcription factor A), a regulator of mitochondrial transcription, is higher in Snell cells, while Pgc‐1α, the main regulator of mitochondrial biogenesis, is upregulated only in Snell cells. Consistent with these differences, Snell cells maintain oxidative respiration rate, ATP content, and expression of mitochondrial‐DNA‐encoded genes after exposure to doxycycline stress. In vivo, compared to normal mice, Snell mice show stronger hepatic mtUPR induction and maintain mitochondrial protein stoichiometry after mitochondrial stress exposure. Overall, our work demonstrates that a long‐lived mouse model exhibits improved mitochondrial stress response, and provides a rationale for future mouse lifespan studies involving compounds that induce mtUPR. Further research on mitochondrial homeostasis in long‐lived mice may facilitate development of interventions that blunt mitochondrial deterioration in neurodegenerative diseases such as Alzheimer's and Parkinson's and postpone diseases of aging in humans.
Collapse
Affiliation(s)
- Ulas Ozkurede
- Department of Molecular and Cellular Pathology, University of Michigan Geriatrics Center University of Michigan School of Medicine Ann Arbor MI USA
| | - Richard A. Miller
- Department of Molecular and Cellular Pathology, University of Michigan Geriatrics Center University of Michigan School of Medicine Ann Arbor MI USA
- Department of Pathology, University of Michigan Geriatrics Center University of Michigan School of Medicine Ann Arbor MI USA
| |
Collapse
|
75
|
Sándor S, Kubinyi E. Genetic Pathways of Aging and Their Relevance in the Dog as a Natural Model of Human Aging. Front Genet 2019; 10:948. [PMID: 31681409 PMCID: PMC6813227 DOI: 10.3389/fgene.2019.00948] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 09/05/2019] [Indexed: 12/15/2022] Open
Abstract
Aging research has experienced a burst of scientific efforts in the last decades as the growing ratio of elderly people has begun to pose an increased burden on the healthcare and pension systems of developed countries. Although many breakthroughs have been reported in understanding the cellular mechanisms of aging, the intrinsic and extrinsic factors that contribute to senescence on higher biological levels are still barely understood. The dog, Canis familiaris, has already served as a valuable model of human physiology and disease. The possible role the dog could play in aging research is still an open question, although utilization of dogs may hold great promises as they naturally develop age-related cognitive decline, with behavioral and histological characteristics very similar to those of humans. In this regard, family dogs may possess unmatched potentials as models for investigations on the complex interactions between environmental, behavioral, and genetic factors that determine the course of aging. In this review, we summarize the known genetic pathways in aging and their relevance in dogs, putting emphasis on the yet barely described nature of certain aging pathways in canines. Reasons for highlighting the dog as a future aging and gerontology model are also discussed, ranging from its unique evolutionary path shared with humans, its social skills, and the fact that family dogs live together with their owners, and are being exposed to the same environmental effects.
Collapse
Affiliation(s)
- Sára Sándor
- Department of Ethology, Eötvös Loránd University, Budapest, Hungary
| | | |
Collapse
|
76
|
Pibiri M. Liver regeneration in aged mice: new insights. Aging (Albany NY) 2019; 10:1801-1824. [PMID: 30157472 PMCID: PMC6128415 DOI: 10.18632/aging.101524] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 08/10/2018] [Indexed: 02/06/2023]
Abstract
The regenerative capacity of the liver after resection is reduced with aging. Recent studies on rodents revealed that both intracellular and extracellular factors are involved in the impairment of liver mass recovery during aging. Among the intracellular factors, age-dependent decrease of BubR1 (budding uninhibited by benzimidazole-related 1), YAP (Yes-associated protein) and SIRT1 (Sirtuin-1) have been associated to dampening of tissue reconstitution and inhibition of cell cycle genes following partial hepatectomy. Extra-cellular factors, such as age-dependent changes in hepatic stellate cells affect liver regeneration through inhibition of progenitor cells and reduction of liver perfusion. Furthermore, chronic release of pro-inflammatory proteins by senescent cells (SASP) affects cell proliferation suggesting that senescent cell clearance might improve tissue regeneration. Accordingly, young plasma restores liver regeneration in aged animals through autophagy re-establishment. This review will discuss how intracellular and extracellular factors cooperate to guarantee a proper liver regeneration and the possible causes of its impairment during aging. The possibility that an improvement of the liver regenerative capacity in elderly might be achieved through elimination of senescent cells via autophagy or by administration of direct mitogenic agents devoid of cytotoxicity will also be entertained.
Collapse
Affiliation(s)
- Monica Pibiri
- Department of Biomedical Sciences, Oncology and Molecular Pathology Unit, University of Cagliari, Cagliari 09124, Italy
| |
Collapse
|
77
|
Barja G. Towards a unified mechanistic theory of aging. Exp Gerontol 2019; 124:110627. [DOI: 10.1016/j.exger.2019.05.016] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/08/2019] [Accepted: 05/30/2019] [Indexed: 12/18/2022]
|
78
|
Salcan S, Bongardt S, Monteiro Barbosa D, Efimov IR, Rassaf T, Krüger M, Kötter S. Elastic titin properties and protein quality control in the aging heart. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1867:118532. [PMID: 31421188 DOI: 10.1016/j.bbamcr.2019.118532] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 07/12/2019] [Accepted: 08/12/2019] [Indexed: 01/09/2023]
Abstract
Cardiac aging affects the heart on the functional, structural, and molecular level and shares characteristic hallmarks with the development of chronic heart failure. Apart from age-dependent left ventricular hypertrophy and fibrosis that impairs diastolic function, diminished activity of cardiac protein-quality-control systems increases the risk of cytotoxic accumulation of defective proteins. Here, we studied the impact of cardiac aging on the sarcomeric protein titin by analyzing titin-based cardiomyocyte passive tension, titin modification and proteasomal titin turnover. We analyzed left ventricular samples from young (6 months) and old (20 months) wild-type mice and healthy human donor patients grouped according to age in young (17-50 years) and aged hearts (51-73 years). We found no age-dependent differences in titin isoform composition of mouse or human hearts. In aged hearts from mice and human we determined altered titin phosphorylation at serine residues S4010 and S4099 in the elastic N2B domain, but no significant changes in phosphorylation of S11878 and S12022 in the elastic PEVK region. Importantly, overall titin-based cardiomyocyte passive tension remained unchanged. In aged hearts, the calcium-activated protease calpain-1, which provides accessibility to ubiquitination by releasing titin from the sarcomere, showed decreased proteolytic activity. In addition, we observed a reduction in the proteasomal activities. Taken together, our data indicate that cardiac aging does not affect titin-based passive properties of the cardiomyocytes, but impairs protein-quality control, including titin, which may result in a diminished adaptive capacity of the aged myocardium.
Collapse
Affiliation(s)
- Senem Salcan
- Department of Cardiovascular Physiology, Medical Faculty, Heinrich Heine-University Düsseldorf, D-40225 Düsseldorf, Germany
| | - Sabine Bongardt
- Department of Cardiovascular Physiology, Medical Faculty, Heinrich Heine-University Düsseldorf, D-40225 Düsseldorf, Germany
| | - David Monteiro Barbosa
- Department of Cardiovascular Physiology, Medical Faculty, Heinrich Heine-University Düsseldorf, D-40225 Düsseldorf, Germany
| | - Igor R Efimov
- George Washington University, Department of Biomedical Engineering, Science and Engineering Hall, Washington DC-20052, USA
| | - Tienush Rassaf
- University Hospital Essen, Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, 45147 Essen, Germany
| | - Martina Krüger
- Department of Cardiovascular Physiology, Medical Faculty, Heinrich Heine-University Düsseldorf, D-40225 Düsseldorf, Germany.
| | - Sebastian Kötter
- Department of Cardiovascular Physiology, Medical Faculty, Heinrich Heine-University Düsseldorf, D-40225 Düsseldorf, Germany.
| |
Collapse
|
79
|
Chaves CF, Mazzotti DR, Cendoroglo MS, Ramos LR, Tufik S, Silva VCD, D'Almeida V. Genes related to maintenance of autophagy and successful aging. ARQUIVOS DE NEURO-PSIQUIATRIA 2019; 76:831-839. [PMID: 30698207 DOI: 10.1590/0004-282x20180142] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/25/2018] [Indexed: 01/11/2023]
Abstract
Considering aging as a phenomenon in which there is a decline in essential processes for cell survival, we investigated the autophagic and proteasome pathways in three different groups: young, older and oldest old male adults. The expression profile of autophagic pathway-related genes was carried out in peripheral blood, and the proteasome quantification was performed in plasma. No significant changes were found in plasma proteasome concentrations or in correlations between proteasome concentrations and ages. However, some autophagy- and/or apoptosis-related genes were differentially expressed. In addition, the network and enrichment analysis showed an interaction between four of the five differentially expressed genes and an association of these genes with the transcriptional process. Considering that the oldest old individuals maintained both the expression of genes linked to the autophagic machinery, and the proteasome levels, when compared with the older group, we concluded that these factors could be considered crucial for successful aging.
Collapse
Affiliation(s)
| | - Diego Robles Mazzotti
- Universidade Federal de São Paulo, Departamento de Psicobiologia, São Paulo SP, Brasil
| | | | - Luiz Roberto Ramos
- Universidade Federal de São Paulo, Departamento de Medicina Preventiva, São Paulo SP, Brasil
| | - Sergio Tufik
- Universidade Federal de São Paulo, Departamento de Psicobiologia, São Paulo SP, Brasil
| | | | - Vânia D'Almeida
- Universidade Federal de São Paulo, Departamento de Psicobiologia, São Paulo SP, Brasil
| |
Collapse
|
80
|
Kwak SE, Shin HE, Zhang DD, Lee J, Yoon KJ, Bae JH, Moon HY, Song W. Potential role of exercise-induced glucose-6-phosphate isomerase in skeletal muscle function. J Exerc Nutrition Biochem 2019; 23:28-33. [PMID: 31337203 PMCID: PMC6651688 DOI: 10.20463/jenb.2019.0014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 06/19/2019] [Indexed: 12/28/2022] Open
Abstract
[Purpose] Recent studies have shown that glucose-6-phosphate isomerase (GPI)—which is a glycolysis interconversion enzyme—reduces oxidative stress. However, these studies are limited to tumors such as fibrosarcoma, and there are no studies that have examined the effects of exercise on GPI expression in mice skeletal muscle. Furthermore, GPI acts in an autocrine manner thorough its receptor, autocrine motility factor receptor (AMFR); therefore, we investigated expression level changes of secreted GPI from skeletal muscle in in vitro study to examine the potential role of GPI on skeletal muscle. [Methods] First, we performed an in vitro study, to identify the condition that upregulates GPI levels in skeletal muscle cells; we treated C2C12 muscle cells with an exercise-mimicking chemical, AICAR. AICAR treatment upregulated GPI expression level in C2C12 cell and its secretomes. To confirm the direct effect of GPI on skeletal muscle cells, we treated C2C12 cells with GPI recombinant protein. [Results] We found that GPI improved the viability of C2C12 cells. In the in vivo study, the exercise-treated mice group showed upregulated GPI expression in skeletal muscle. Based on the in vitro study results, we speculated that expression level of GPI in skeletal muscle might be associated with muscle function. We analyzed the association between GPI expression level and the grip strength of the all mice group. The mice group’s grip strengths were upregulated after 2 weeks of treadmill exercise, and GPI expression level positively correlated with the grip strength. [Conclusion] These results suggested that the exercise-induced GPI expression in skeletal muscle might have a positive effect on skeletal muscle function.
Collapse
|
81
|
Rinschen MM, Ivanisevic J, Giera M, Siuzdak G. Identification of bioactive metabolites using activity metabolomics. Nat Rev Mol Cell Biol 2019; 20:353-367. [PMID: 30814649 PMCID: PMC6613555 DOI: 10.1038/s41580-019-0108-4] [Citation(s) in RCA: 660] [Impact Index Per Article: 110.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The metabolome, the collection of small-molecule chemical entities involved in metabolism, has traditionally been studied with the aim of identifying biomarkers in the diagnosis and prediction of disease. However, the value of metabolome analysis (metabolomics) has been redefined from a simple biomarker identification tool to a technology for the discovery of active drivers of biological processes. It is now clear that the metabolome affects cellular physiology through modulation of other 'omics' levels, including the genome, epigenome, transcriptome and proteome. In this Review, we focus on recent progress in using metabolomics to understand how the metabolome influences other omics and, by extension, to reveal the active role of metabolites in physiology and disease. This concept of utilizing metabolomics to perform activity screens to identify biologically active metabolites - which we term activity metabolomics - is already having a broad impact on biology.
Collapse
Affiliation(s)
- Markus M Rinschen
- The Scripps Research Institute, Center for Metabolomics and Mass Spectrometry, La Jolla, CA, USA
| | - Julijana Ivanisevic
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Martin Giera
- Leiden University Medical Center, Center for Proteomics & Metabolomics, Leiden, Netherlands.
| | - Gary Siuzdak
- The Scripps Research Institute, Center for Metabolomics and Mass Spectrometry, La Jolla, CA, USA.
| |
Collapse
|
82
|
Klein BEK, Lee KE, Maynard JD, Johnson CA, Danforth L, Klein R. Skin Intrinsic Fluorescence and Selected Measures of Visual Function and aging in Older Adults. Ophthalmic Epidemiol 2019; 26:264-269. [PMID: 31030599 DOI: 10.1080/09286586.2019.1604977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Purpose: Functional, structural and metabolic decline in many systems and in combination contribute to biologic aging and may be manifest as increased risk of morbid events such as neuropathy, albuminuria, and coronary artery disease or mortality. A biologic marker of aging may be a useful tool in identifying persons at increased risk of morbidity or mortality. We have measured skin intrinsic fluorescence (SIF) in a group of older adults to determine whether this easily determined measure could serve as such a biomarker. Methods: Survivors of a population based study of older adults in a moderate sized Midwestern town. Of the 1181 persons participating, 939 had measures of skin intrinsic fluorescence (SIF) and at least one functional or diagnostic characteristic at the most recent examination. Characteristics such as blood pressure, forced expiratory volume, vision, time to walk a standard course and medical history and their associations with SIF measures were examined. Mortality after the last examination with respect to SIF was also investigated. There were 118 deaths among those who participated in this phase of the study. All analyses pertinent to these findings were adjusted for age. Results: SIF measures were significantly associated with low contrast sensitivity, more errors on frequency doubling technology testing (loss of peripheral vision), self-reported poor vision, slow gait, poor forced expiratory volume, and self-reported poor health. SIF was also associated with increased risk of death. All of these analyses were adjusted for age. Conclusions: Skin intrinsic fluorescence provides easily obtained markers of age-related functional outcomes, suggesting SIF measurements may be useful to identify persons who may benefit from more frequent medical scrutiny to decrease morbidity and mortality.
Collapse
Affiliation(s)
- Barbara E K Klein
- a Department of Ophthalmology and Visual Sciences , University of Wisconsin School of Medicine and Public Health , Madison , Wisconsin
| | - Kristine E Lee
- a Department of Ophthalmology and Visual Sciences , University of Wisconsin School of Medicine and Public Health , Madison , Wisconsin
| | - John D Maynard
- b Medical Device and Diagnostics Consulting , Albuquerque , New Mexico , USA
| | - Chris A Johnson
- c University of Iowa Hospitals and Clinics , Iowa City , Iowa , USA.,d Ophthalmology and Visual Sciences , University of Iowa , Iowa City , Iowa , USA
| | - Lorraine Danforth
- a Department of Ophthalmology and Visual Sciences , University of Wisconsin School of Medicine and Public Health , Madison , Wisconsin
| | - Ronald Klein
- a Department of Ophthalmology and Visual Sciences , University of Wisconsin School of Medicine and Public Health , Madison , Wisconsin
| |
Collapse
|
83
|
The Impact of Caloric Restriction on the Epigenetic Signatures of Aging. Int J Mol Sci 2019; 20:ijms20082022. [PMID: 31022953 PMCID: PMC6515465 DOI: 10.3390/ijms20082022] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/19/2019] [Accepted: 04/23/2019] [Indexed: 12/14/2022] Open
Abstract
Aging is characterized by an extensive remodeling of epigenetic patterns, which has been implicated in the physiopathology of age-related diseases. Nutrition plays a significant role in modulating the epigenome, and a growing amount of data indicate that dietary changes can modify the epigenetic marks associated with aging. In this review, we will assess the current advances in the relationship between caloric restriction, a proven anti-aging intervention, and epigenetic signatures of aging. We will specifically discuss the impact of caloric restriction on epigenetic regulation and how some of the favorable effects of caloric restriction on lifespan and healthspan could be mediated by epigenetic modifications.
Collapse
|
84
|
Erwin AA, Blumenstiel JP. Aging in the Drosophila ovary: contrasting changes in the expression of the piRNA machinery and mitochondria but no global release of transposable elements. BMC Genomics 2019; 20:305. [PMID: 31014230 PMCID: PMC6480902 DOI: 10.1186/s12864-019-5668-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 04/08/2019] [Indexed: 01/06/2023] Open
Abstract
Background Evolutionary theory indicates that the dynamics of aging in the soma and reproductive tissues may be distinct. This difference arises from the fact that only the germline lineage establishes future generations. In the soma, changes in the landscape of heterochromatin have been proposed to have an important role in aging. This is because redistribution of heterochromatin during aging has been linked to the derepression of transposable elements and an overall loss of somatic gene regulation. A role for changes in the chromatin landscape in the aging of reproductive tissues is less well established. Whether or not epigenetic factors, such as heterochromatin marks, are perturbed in aging reproductive tissues is of interest because, in special cases, epigenetic variation may be heritable. Using mRNA sequencing data from late-stage egg chambers in Drosophila melanogaster, we characterized the landscape of altered gene and transposable element expression in aged reproductive tissues. This allowed us to test the hypothesis that reproductive tissues may differ from somatic tissues in their response to aging. Results We show that age-related expression changes in late-stage egg chambers tend to occur in genes residing in heterochromatin, particularly on the largely heterochromatic 4th chromosome. However, these expression differences are seen as both decreases and increases during aging, inconsistent with a general loss of heterochromatic silencing. We also identify an increase in expression of the piRNA machinery, suggesting an age-related increased investment in the maintenance of genome stability. We further identify a strong age-related reduction in the expression of mitochondrial transcripts. However, we find no evidence for global TE derepression in reproductive tissues. Rather, the observed effects of aging on TEs are primarily strain and family specific. Conclusions These results identify unique responses in somatic versus reproductive tissue with regards to aging. As in somatic tissues, female reproductive tissues show reduced expression of mitochondrial genes. In contrast, the piRNA machinery shows increased expression during aging. Overall, these results also indicate that global loss of TE control observed in other studies may be unique to the soma and sensitive to genetic background and TE family. Electronic supplementary material The online version of this article (10.1186/s12864-019-5668-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alexandra A Erwin
- Department of Ecology and Evolutionary Biology, University of Kansas, Lawrence, KS, 66045, USA.
| | - Justin P Blumenstiel
- Department of Ecology and Evolutionary Biology, University of Kansas, Lawrence, KS, 66045, USA.
| |
Collapse
|
85
|
Mitochondrial LonP1 protects cardiomyocytes from ischemia/reperfusion injury in vivo. J Mol Cell Cardiol 2019; 128:38-50. [DOI: 10.1016/j.yjmcc.2018.12.017] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 12/10/2018] [Accepted: 12/29/2018] [Indexed: 11/18/2022]
|
86
|
Kane AE, Sinclair DA. Epigenetic changes during aging and their reprogramming potential. Crit Rev Biochem Mol Biol 2019; 54:61-83. [PMID: 30822165 PMCID: PMC6424622 DOI: 10.1080/10409238.2019.1570075] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 01/09/2019] [Accepted: 01/11/2019] [Indexed: 02/07/2023]
Abstract
The aging process results in significant epigenetic changes at all levels of chromatin and DNA organization. These include reduced global heterochromatin, nucleosome remodeling and loss, changes in histone marks, global DNA hypomethylation with CpG island hypermethylation, and the relocalization of chromatin modifying factors. Exactly how and why these changes occur is not fully understood, but evidence that these epigenetic changes affect longevity and may cause aging, is growing. Excitingly, new studies show that age-related epigenetic changes can be reversed with interventions such as cyclic expression of the Yamanaka reprogramming factors. This review presents a summary of epigenetic changes that occur in aging, highlights studies indicating that epigenetic changes may contribute to the aging process and outlines the current state of research into interventions to reprogram age-related epigenetic changes.
Collapse
Affiliation(s)
- Alice E. Kane
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - David A. Sinclair
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Pharmacology, The University of New South Wales, Sydney, Australia
| |
Collapse
|
87
|
Xie J, Liu Y, Du X, Wu Y. TGF-β1 promotes the invasion and migration of papillary thyroid carcinoma cells by inhibiting the expression of lncRNA-NEF. Oncol Lett 2019; 17:3125-3132. [PMID: 30867742 PMCID: PMC6396259 DOI: 10.3892/ol.2019.9947] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 12/13/2018] [Indexed: 01/21/2023] Open
Abstract
Increased expression levels of transforming growth factor-β1 (TGF-β1) are associated with metastasis in papillary thyroid carcinoma (PTC), although the mechanisms involved remain unknown. The present study aimed to investigate these mechanisms. The results demonstrated that the expression levels of TGF-β1 mRNA were higher and those of lncRNA-NEF were lower in tumor tissues compared with in healthy tissues. High levels of TGF-β1 mRNA and low levels of lncRNA-NEF were also detected in the serum of patients with PTC compared with in healthy controls. Serum levels of TGF-β1 and lncRNA-NEF were significantly associated with lymph node metastasis. Furthermore, TGF-β1 promoted cell migration and invasion of PTC cell lines, but not the normal cell line, whereas lncRNA-NEF overexpression inhibited these phenomena. TGF-β1 also inhibited lncRNA-NEF expression in PTC cell lines, but not in the normal cell line. Conversely, lncRNA-NEF overexpression exhibited no effects on TGF-β1. In conclusion, the present study suggested that TGF-β1 may promote the invasion and migration of PTC cells by inhibiting the expression of lncRNA-NEF.
Collapse
Affiliation(s)
- Jiaqi Xie
- Department of Thyroid and Breast Surgery, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning 116000, P.R. China
| | - Yi Liu
- Department of Thyroid and Breast Surgery, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning 116000, P.R. China
| | - Xiaolong Du
- Department of Thyroid and Breast Surgery, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning 116000, P.R. China
| | - Ying Wu
- Intensive Care Unit, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning 116000, P.R. China
| |
Collapse
|
88
|
Larsson L, Degens H, Li M, Salviati L, Lee YI, Thompson W, Kirkland JL, Sandri M. Sarcopenia: Aging-Related Loss of Muscle Mass and Function. Physiol Rev 2019; 99:427-511. [PMID: 30427277 PMCID: PMC6442923 DOI: 10.1152/physrev.00061.2017] [Citation(s) in RCA: 962] [Impact Index Per Article: 160.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 05/14/2018] [Accepted: 06/15/2018] [Indexed: 12/11/2022] Open
Abstract
Sarcopenia is a loss of muscle mass and function in the elderly that reduces mobility, diminishes quality of life, and can lead to fall-related injuries, which require costly hospitalization and extended rehabilitation. This review focuses on the aging-related structural changes and mechanisms at cellular and subcellular levels underlying changes in the individual motor unit: specifically, the perikaryon of the α-motoneuron, its neuromuscular junction(s), and the muscle fibers that it innervates. Loss of muscle mass with aging, which is largely due to the progressive loss of motoneurons, is associated with reduced muscle fiber number and size. Muscle function progressively declines because motoneuron loss is not adequately compensated by reinnervation of muscle fibers by the remaining motoneurons. At the intracellular level, key factors are qualitative changes in posttranslational modifications of muscle proteins and the loss of coordinated control between contractile, mitochondrial, and sarcoplasmic reticulum protein expression. Quantitative and qualitative changes in skeletal muscle during the process of aging also have been implicated in the pathogenesis of acquired and hereditary neuromuscular disorders. In experimental models, specific intervention strategies have shown encouraging results on limiting deterioration of motor unit structure and function under conditions of impaired innervation. Translated to the clinic, if these or similar interventions, by saving muscle and improving mobility, could help alleviate sarcopenia in the elderly, there would be both great humanitarian benefits and large cost savings for health care systems.
Collapse
Affiliation(s)
- Lars Larsson
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Hans Degens
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Meishan Li
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Leonardo Salviati
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Young Il Lee
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Wesley Thompson
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - James L Kirkland
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Marco Sandri
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| |
Collapse
|
89
|
Wang Z, Yang W. Impaired capacity to restore proteostasis in the aged brain after ischemia: Implications for translational brain ischemia research. Neurochem Int 2018; 127:87-93. [PMID: 30599146 DOI: 10.1016/j.neuint.2018.12.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 12/28/2018] [Accepted: 12/28/2018] [Indexed: 12/15/2022]
Abstract
Brain ischemia induced by cardiac arrest or ischemic stroke is a severe form of metabolic stress that substantially disrupts cellular homeostasis, especially protein homeostasis (proteostasis). As proteostasis is fundamental for cellular and organismal health, cells have developed a complex network to restore proteostasis impaired by stress. Many components of this network - including ubiquitination, small ubiquitin-like modifier (SUMO) conjugation, autophagy, and the unfolded protein response (UPR) - are activated in the post-ischemic brain, and play a crucial role in cell survival and recovery of neurologic function. Importantly, recent studies have shown that ischemia-induced activation of these proteostasis-related pathways in the aged brain is impaired, indicating an aging-related decline in the self-healing capacity of the brain. This impaired capacity is a significant factor for consideration in the field of brain ischemia because the vast majority of cardiac arrest and stroke patients are elderly. In this review, we focus on the effects of aging on these critical proteostasis-related pathways in the brain, and discuss their implications in translational brain ischemia research.
Collapse
Affiliation(s)
- Zhuoran Wang
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Wei Yang
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
90
|
Calorie restriction and its impact on gut microbial composition and global metabolism. Front Med 2018; 12:634-644. [PMID: 30446879 DOI: 10.1007/s11684-018-0670-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 09/27/2018] [Indexed: 02/08/2023]
Abstract
Calorie restriction (CR) is a dietary regimen that reduces calorie intake without incurring malnutrition or a reduction in essential nutrients. It has long been recognized as a natural strategy for promoting health, extending longevity, and prevents the development of metabolic and age-related diseases. In the present review, we focus on the general effect of CR on gut microbiota composition and global metabolism. We also propose mechanisms for its beneficial effect. Results showed that probiotic and butyrate-producing microbes increased their relative abundance, whereas proinflammatory strains exhibited suppressed relative abundance following CR. Analyses of the gut microbial and host metabolisms revealed that most host microbial co-metabolites were changed due to CR. Examples of dramatic CR-induced changes in host metabolism included a decrease in the rate of lipid biosynthesis and an increase in the rates of fatty acid catabolism, β-oxidation, glycogenolysis, and gluconeogenesis. The observed phenotypes and the further verification of the direct link between gut microbiota and metabolome may benefit patients that are at risk for developing metabolic disease. Thus, improved gut microbiota composition and metabolome are potential biomarkers for determining the effectiveness of dietary interventions for age-related and metabolic diseases.
Collapse
|
91
|
Castillo-Morales A, Monzón-Sandoval J, Urrutia AO, Gutiérrez H. Postmitotic cell longevity-associated genes: a transcriptional signature of postmitotic maintenance in neural tissues. Neurobiol Aging 2018; 74:147-160. [PMID: 30448614 DOI: 10.1016/j.neurobiolaging.2018.10.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 10/03/2018] [Accepted: 10/11/2018] [Indexed: 12/24/2022]
Abstract
Different cell types have different postmitotic maintenance requirements. Nerve cells, however, are unique in this respect as they need to survive and preserve their functional complexity for the entire lifetime of the organism, and failure at any level of their supporting mechanisms leads to a wide range of neurodegenerative conditions. Whether these differences across tissues arise from the activation of distinct cell type-specific maintenance mechanisms or the differential activation of a common molecular repertoire is not known. To identify the transcriptional signature of postmitotic cellular longevity (PMCL), we compared whole-genome transcriptome data from human tissues ranging in longevity from 120 days to over 70 years and found a set of 81 genes whose expression levels are closely associated with increased cell longevity. Using expression data from 10 independent sources, we found that these genes are more highly coexpressed in longer-living tissues and are enriched in specific biological processes and transcription factor targets compared with randomly selected gene samples. Crucially, we found that PMCL-associated genes are downregulated in the cerebral cortex and substantia nigra of patients with Alzheimer's and Parkinson's disease, respectively, as well as Hutchinson-Gilford progeria-derived fibroblasts, and that this downregulation is specifically linked to their underlying association with cellular longevity. Moreover, we found that sexually dimorphic brain expression of PMCL-associated genes reflects sexual differences in lifespan in humans and macaques. Taken together, our results suggest that PMCL-associated genes are part of a generalized machinery of postmitotic maintenance and functional stability in both neural and non-neural cells and support the notion of a common molecular repertoire differentially engaged in different cell types with different survival requirements.
Collapse
Affiliation(s)
- Atahualpa Castillo-Morales
- School of Life Sciences, University of Lincoln, Lincoln, UK; Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, Bath, UK
| | - Jimena Monzón-Sandoval
- School of Life Sciences, University of Lincoln, Lincoln, UK; Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, Bath, UK
| | - Araxi O Urrutia
- Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, Bath, UK; Instituto de Ecología, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.
| | | |
Collapse
|
92
|
Abstract
SIGNIFICANCE Aging is a complex trait that is influenced by a combination of genetic and environmental factors. Although many cellular and physiological changes have been described to occur with aging, the precise molecular causes of aging remain unknown. Given the biological complexity and heterogeneity of the aging process, understanding the mechanisms that underlie aging requires integration of data about age-dependent changes that occur at the molecular, cellular, tissue, and organismal levels. Recent Advances: The development of high-throughput technologies such as next-generation sequencing, proteomics, metabolomics, and automated imaging techniques provides researchers with new opportunities to understand the mechanisms of aging. Using these methods, millions of biological molecules can be simultaneously monitored during the aging process with high accuracy and specificity. CRITICAL ISSUES Although the ability to produce big data has drastically increased over the years, integration and interpreting of high-throughput data to infer regulatory relationships between biological factors and identify causes of aging remain the major challenges. In this review, we describe recent advances and survey emerging omics approaches in aging research. We then discuss their limitations and emphasize the need for the further development of methods for the integration of different types of data. FUTURE DIRECTIONS Combining omics approaches and novel methods for single-cell analysis with systems biology tools would allow building interaction networks and investigate how these networks are perturbed with aging and disease states. Together, these studies are expected to provide a better understanding of the aging process and could provide insights into the pathophysiology of many age-associated human diseases. Antioxid. Redox Signal. 29, 985-1002.
Collapse
Affiliation(s)
- Jared S Lorusso
- 1 Department of Dermatology, Boston University School of Medicine , Boston, Massachusetts
| | - Oleg A Sviderskiy
- 2 Department of Ecology and Life Safety, Samara National Research University , Samara, Russia
| | - Vyacheslav M Labunskyy
- 1 Department of Dermatology, Boston University School of Medicine , Boston, Massachusetts
| |
Collapse
|
93
|
Song R, Sarnoski EA, Acar M. The Systems Biology of Single-Cell Aging. iScience 2018; 7:154-169. [PMID: 30267677 PMCID: PMC6153419 DOI: 10.1016/j.isci.2018.08.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/30/2018] [Accepted: 08/29/2018] [Indexed: 12/12/2022] Open
Abstract
Aging is a leading cause of human morbidity and mortality, but efforts to slow or reverse its effects are hampered by an incomplete understanding of its multi-faceted origins. Systems biology, the use of quantitative and computational methods to understand complex biological systems, offers a toolkit well suited to elucidating the root cause of aging. We describe the known components of the aging network and outline innovative techniques that open new avenues of investigation to the aging research community. We propose integration of the systems biology and aging fields, identifying areas of complementarity based on existing and impending technological capabilities.
Collapse
Affiliation(s)
- Ruijie Song
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, 300 George Street, Suite 501, New Haven, CT 06511, USA; Systems Biology Institute, Yale University, 850 West Campus Drive, West Haven, CT 06516, USA
| | - Ethan A Sarnoski
- Systems Biology Institute, Yale University, 850 West Campus Drive, West Haven, CT 06516, USA; Department of Molecular Cellular and Developmental Biology, Yale University, 219 Prospect Street, New Haven, CT 06511, USA
| | - Murat Acar
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, 300 George Street, Suite 501, New Haven, CT 06511, USA; Systems Biology Institute, Yale University, 850 West Campus Drive, West Haven, CT 06516, USA; Department of Molecular Cellular and Developmental Biology, Yale University, 219 Prospect Street, New Haven, CT 06511, USA; Department of Physics, Yale University, 217 Prospect Street, New Haven, CT 06511, USA.
| |
Collapse
|
94
|
Leysen H, van Gastel J, Hendrickx JO, Santos-Otte P, Martin B, Maudsley S. G Protein-Coupled Receptor Systems as Crucial Regulators of DNA Damage Response Processes. Int J Mol Sci 2018; 19:E2919. [PMID: 30261591 PMCID: PMC6213947 DOI: 10.3390/ijms19102919] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 09/14/2018] [Accepted: 09/15/2018] [Indexed: 12/11/2022] Open
Abstract
G protein-coupled receptors (GPCRs) and their associated proteins represent one of the most diverse cellular signaling systems involved in both physiological and pathophysiological processes. Aging represents perhaps the most complex biological process in humans and involves a progressive degradation of systemic integrity and physiological resilience. This is in part mediated by age-related aberrations in energy metabolism, mitochondrial function, protein folding and sorting, inflammatory activity and genomic stability. Indeed, an increased rate of unrepaired DNA damage is considered to be one of the 'hallmarks' of aging. Over the last two decades our appreciation of the complexity of GPCR signaling systems has expanded their functional signaling repertoire. One such example of this is the incipient role of GPCRs and GPCR-interacting proteins in DNA damage and repair mechanisms. Emerging data now suggest that GPCRs could function as stress sensors for intracellular damage, e.g., oxidative stress. Given this role of GPCRs in the DNA damage response process, coupled to the effective history of drug targeting of these receptors, this suggests that one important future activity of GPCR therapeutics is the rational control of DNA damage repair systems.
Collapse
Affiliation(s)
- Hanne Leysen
- Department of Biomedical Sciences, University of Antwerp, 2610 Antwerp, Belgium.
| | - Jaana van Gastel
- Department of Biomedical Sciences, University of Antwerp, 2610 Antwerp, Belgium.
- Translational Neurobiology Group, Center of Molecular Neurology, VIB, 2610 Antwerp, Belgium.
| | - Jhana O Hendrickx
- Department of Biomedical Sciences, University of Antwerp, 2610 Antwerp, Belgium.
- Translational Neurobiology Group, Center of Molecular Neurology, VIB, 2610 Antwerp, Belgium.
| | - Paula Santos-Otte
- Institute of Biophysics, Humboldt-Universität zu Berlin, 10115 Berlin, Germany.
| | - Bronwen Martin
- Department of Biomedical Sciences, University of Antwerp, 2610 Antwerp, Belgium.
| | - Stuart Maudsley
- Department of Biomedical Sciences, University of Antwerp, 2610 Antwerp, Belgium.
- Translational Neurobiology Group, Center of Molecular Neurology, VIB, 2610 Antwerp, Belgium.
| |
Collapse
|
95
|
Aim for the core: suitability of the ubiquitin-independent 20S proteasome as a drug target in neurodegeneration. Transl Res 2018; 198:48-57. [PMID: 30244692 PMCID: PMC6154511 DOI: 10.1016/j.trsl.2018.05.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 05/14/2018] [Accepted: 05/15/2018] [Indexed: 02/06/2023]
Abstract
Neurodegenerative diseases are a class of age-associated proteopathies characterized by the accumulation of misfolded and/or aggregation-prone proteins. This imbalance has been attributed, in part, to an age-dependent decay in the capacity of protein turnover. Most proteins are degraded by the ubiquitin-proteasome system (UPS), which is composed of ubiquitin ligases and regulatory particles, such as the 19S, that deliver cargo to the proteolytically active 20S proteasome (20S) core. However, a subset of clients, especially intrinsically disordered proteins (IDPs), are also removed by the action of the ubiquitin-independent proteasome system (UIPS). What are the specific contributions of the UPS and UIPS in the context of neurodegeneration? Here, we explore how age-associated changes in the relative contribution of the UPS and UIPS, combined with the IDP-like structure of many neurodegenerative disease-associated proteins, might contribute. Strikingly, the 20S has been shown to predominate in older neurons and to preferentially act on relevant substrates, such as synuclein and tau. Moreover, pharmacological activation of the 20S has been shown to accelerate removal of aggregation-prone proteins in some models. Together, these recent studies are turning attention to the 20S and the UIPS as potential therapeutic targets in neurodegeneration.
Collapse
|
96
|
Chaudhari SN, Kipreos ET. The Energy Maintenance Theory of Aging: Maintaining Energy Metabolism to Allow Longevity. Bioessays 2018; 40:e1800005. [PMID: 29901833 PMCID: PMC6314662 DOI: 10.1002/bies.201800005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 04/28/2018] [Indexed: 12/17/2022]
Abstract
Fused, elongated mitochondria are more efficient in generating ATP than fragmented mitochondria. In diverse C. elegans longevity pathways, increased levels of fused mitochondria are associated with lifespan extension. Blocking mitochondrial fusion in these animals abolishes their extended longevity. The long-lived C. elegans vhl-1 mutant is an exception that does not have increased fused mitochondria, and is not dependent on fusion for longevity. Loss of mammalian VHL upregulates alternate energy generating pathways. This suggests that mitochondrial fusion facilitates longevity in C. elegans by increasing energy metabolism. In diverse animals, ATP levels broadly decreases with age. Substantial evidence supports the theory that increasing or maintaining energy metabolism promotes the survival of older animals. Increased ATP levels in older animals allow energy-intensive repair and homeostatic mechanisms such as proteostasis that act to prevent cellular aging. These observations support the emerging paradigm that maintaining energy metabolism promotes the survival of older animals.
Collapse
Affiliation(s)
- Snehal N. Chaudhari
- Department of Cellular Biology University of Georgia Athens, GA 30602
- Present address: Department of Biological Chemistry and Molecular Pharmacology Harvard Medical School Boston, MA 02115
| | - Edward T. Kipreos
- Department of Cellular Biology University of Georgia Athens, GA 30602
| |
Collapse
|
97
|
Muller AWJ. Aging is an adaptation that selects in animals against disruption of homeostasis. Med Hypotheses 2018; 119:68-78. [PMID: 30122495 DOI: 10.1016/j.mehy.2018.07.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/17/2018] [Accepted: 07/25/2018] [Indexed: 01/10/2023]
Abstract
During evolution, Muller's ratchet permanently generates deleterious germline mutations that eventually must be defused by selection. It seems widely held that cancer and aging-related diseases (ARDs) cannot contribute to this germline gene selection because they tail reproduction and thus occur too late, at the end of the life cycle. Here we posit however that by lessening the offspring's survival by proxy through diminishing parental care, they can still contribute to the selection. The hypothesis in detail: The widespread occurrence of aging in animals suggests that it is an adaptation. But to what benefit? Aging seems to have only drawbacks. In humans, ARDs cause today almost all mortality; they include heart disease, cerebrovascular disease, Alzheimer's disease, kidney disease and cancer. Compensation seems unthinkable. For cancer, the author proposed in a previous study a benefit to the species: purifying selection against deleterious germline genes that when expressed enhance intracellular energy dissipation. This multicausal energy dissipation, posited as the universal origin of cancer initiation, relates to cellular heat generation, disrupted metabolism, and inflammation. The organism reproduces during cancer's dormancy, and when approaching its end of life, the onset of cancer is accelerated in proportion to the cancer-initiating signal. Through cancer, the organism, now a parent, implements the self-actuated programmed death of Skulachev's phenoptosis. This "first death" enhances by proxy the offspring's chance of "second death" (or "double death") through diminished parental care. Repetition over generations realizes a purifying selection against genes causing energy dissipation. The removal of the deleterious germline gene mutations permanently generated by Muller's ratchet gives a benefit. We generalize, motivated by the parallels between cancer and aging, the purifying selection posited for cancer to aging. An ARD would be initiated in the organ by multicausal disruption of homeostasis, and be followed by dormancy and senescence until its onset near the end of the life cycle. Just as for cancer, the ARD eventually enhances double death, and the realized permanent selection gives a benefit to the species through the selection against germ line genes that disrupt homeostasis. Given their similarities, cancer and aging are combined in the posited Unified Cancer-Aging Adaptation (UCAA) model, which may be confirmed by next-generation sequencing data. Also because of the emerging important role of cellular senescence, the hypothesis may guide the development of therapies against both cancer and aging.
Collapse
Affiliation(s)
- Anthonie W J Muller
- Synthetic Systems Biology - Nuclear Organization Group, Swammerdam Institute for Life Sciences/University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands.
| |
Collapse
|
98
|
Deori NM, Kale A, Maurya PK, Nagotu S. Peroxisomes: role in cellular ageing and age related disorders. Biogerontology 2018; 19:303-324. [PMID: 29968207 DOI: 10.1007/s10522-018-9761-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 06/27/2018] [Indexed: 12/12/2022]
Abstract
Peroxisomes are dynamic organelles essential for optimum functioning of a eukaryotic cell. Biogenesis of these organelles and the diverse functions performed by them have been extensively studied in the past decade. Their ability to perform functions depending on the cell type and growth conditions is unique and remarkable. Oxidation of fatty acids and reactive oxygen species metabolism are the two most important functions of these ubiquitous organelles. They are often referred to as both source and sink of reactive oxygen species in a cell. Recent research connects peroxisome dysfunction to fatal oxidative damage associated with ageing-related diseases/disorders. It is now widely accepted that mitochondria and peroxisomes are required to maintain oxidative balance in a cell. However, our understanding on the inter-dependence of these organelles to maintain cellular homeostasis of reactive oxygen species is still in its infancy. Herein, we summarize findings that highlight the role of peroxisomes in cellular reactive oxygen species metabolism, ageing and age-related disorders.
Collapse
Affiliation(s)
- Nayan M Deori
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Avinash Kale
- UM-DAE, Centre for Excellence in Basic Sciences, Health Centre, University of Mumbai, Mumbai, 400098, India
| | - Pawan K Maurya
- Interdisciplinary Laboratory for Clinical Neuroscience (LiNC), Universidade Federal de Sao Paulo-UNIFESP, Sao Paulo, Brazil
| | - Shirisha Nagotu
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India.
| |
Collapse
|
99
|
Lebeau J, Rainbolt TK, Wiseman RL. Coordinating Mitochondrial Biology Through the Stress-Responsive Regulation of Mitochondrial Proteases. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 340:79-128. [PMID: 30072094 PMCID: PMC6402875 DOI: 10.1016/bs.ircmb.2018.05.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Proteases are localized throughout mitochondria and function as critical regulators of all aspects of mitochondrial biology. As such, the activities of these proteases are sensitively regulated through transcriptional and post-translational mechanisms to adapt mitochondrial function to specific cellular demands. Here, we discuss the stress-responsive mechanisms responsible for regulating mitochondrial protease activity and the implications of this regulation on mitochondrial function. Furthermore, we describe how imbalances in the activity or regulation of mitochondrial proteases induced by genetic, environmental, or aging-related factors influence mitochondria in the context of disease. Understanding the molecular mechanisms by which cells regulate mitochondrial function through alterations in protease activity provide insights into the contributions of these proteases in pathologic mitochondrial dysfunction and reveals new therapeutic opportunities to ameliorate this dysfunction in the context of diverse classes of human disease.
Collapse
Affiliation(s)
- Justine Lebeau
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - T Kelly Rainbolt
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - R Luke Wiseman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| |
Collapse
|
100
|
Pu M, Wang M, Wang W, Velayudhan SS, Lee SS. Unique patterns of trimethylation of histone H3 lysine 4 are prone to changes during aging in Caenorhabditis elegans somatic cells. PLoS Genet 2018; 14:e1007466. [PMID: 29912876 PMCID: PMC6023244 DOI: 10.1371/journal.pgen.1007466] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 06/28/2018] [Accepted: 06/02/2018] [Indexed: 11/18/2022] Open
Abstract
Tri-methylation on histone H3 lysine 4 (H3K4me3) is associated with active gene expression but its regulatory role in transcriptional activation is unclear. Here we used Caenorhabditis elegans to investigate the connection between H3K4me3 and gene expression regulation during aging. We uncovered around 30% of H3K4me3 enriched regions to show significant and reproducible changes with age. We further showed that these age-dynamic H3K4me3 regions largely mark gene-bodies and are acquired during adult stages. We found that these adult-specific age-dynamic H3K4me3 regions are correlated with gene expression changes with age. In contrast, H3K4me3 marking established during developmental stages remained largely stable with age, even when the H3K4me3 associated genes exhibited RNA expression changes during aging. Importantly, the genes associated with changes in H3K4me3 and RNA levels with age are enriched for functional groups commonly implicated in aging biology. Therefore, our findings suggested divergent roles of H3K4me3 in gene expression regulation during aging, with important implications on aging-dependent pathophysiologies. Histone modifications, the specific chemical modifications on histone proteins, are key for regulating the packing of DNA, and thus have important influence on diverse biological processes. An intensely studied function of histone modifications is their contribution to regulating gene expression. Recent studies in diverse model organisms demonstrated that the global alterations of particular histone modifications, for instance H3K4me3, extend the lifespan of the organism. However, the underlying molecular mechanisms remain largely unclear. In this study, we monitored whether and how the genome-wide pattern of the histone modification H3K4me3 changes during aging in the somatic cells of the model organism C. elegans. We identified interesting and non-conventional patterns of H3K4me3, which span gene-bodies and are acquired during adulthood, that are particularly prone to changes with aging. This is contrasted to the well-studied H3K4me3 patterns that span transcriptional start sites and 5’ promoter regions and are established early during development, which remain stable with age. Consistent with the close association between H3K4me3 marking and active transcription, we observed that the age-dynamic H3K4me3 markings are highly correlated with corresponding RNA expression changes. Importantly, the genes that are associated with both H3K4me3 and RNA expression changes with age are over-represented for functional groups commonly implicated in aging biology. In summary, our findings revealed a lesser known pattern of H3K4me3 modification that can have important biological roles in aging.
Collapse
Affiliation(s)
- Mintie Pu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York City, United States of America
- * E-mail: (MP); (SSL)
| | - Minghui Wang
- Computational Biology Service Unit, Cornell University, Ithaca, New York City, United States of America
| | - Wenke Wang
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York City, United States of America
| | - Satheeja Santhi Velayudhan
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York City, United States of America
| | - Siu Sylvia Lee
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York City, United States of America
- * E-mail: (MP); (SSL)
| |
Collapse
|