101
|
Role of recombination and faithfulness to partner in sex chromosome degeneration. Sci Rep 2018; 8:8978. [PMID: 29895905 PMCID: PMC5997740 DOI: 10.1038/s41598-018-27219-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 05/24/2018] [Indexed: 11/08/2022] Open
Abstract
Sex determination in mammals is strongly linked to sex chromosomes. In most cases, females possess two copies of X chromosome while males have one X and one Y chromosome. It is assumed that these chromosomes originated from a pair of homologous autosomes, which diverged when recombination between them was suppressed. However, it is still debated why the sex chromosomes stopped recombining and how this process spread out over most part of the chromosomes. To study this problem, we developed a simulation model, in which the recombination rate between the sex chromosomes can freely evolve. We found that the suppression of recombination between the X and Y is spontaneous and proceeds very quickly during the evolution of population, which leads to the degeneration of the Y in males. Interestingly, the degeneration happens only when mating pairs are unfaithful. This evolutionary strategy purifies the X chromosome from defective alleles and leads to the larger number of females than males in the population. In consequence, the reproductive potential of the whole population increases. Our results imply that both the suppression of recombination and the degeneration of Y chromosome may be associated with reproductive strategy and favoured in polygamous populations with faithless mating partners.
Collapse
|
102
|
Loss of Malat1 does not modify age- or diet-induced adipose tissue accretion and insulin resistance in mice. PLoS One 2018; 13:e0196603. [PMID: 29746487 PMCID: PMC5944987 DOI: 10.1371/journal.pone.0196603] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 04/16/2018] [Indexed: 12/18/2022] Open
Abstract
Several studies have suggested that signals emerging from white adipose tissue can contribute to the control of longevity. In turn, aging is associated with perturbed regulation and partitioning of fat depots and insulin resistance. However, the exact mechanisms involved in these relationships remain undetermined. Using RAP-PCR on adipose tissue of young and old male mice coupled with qPCR validation, we have uncovered the long non-coding RNA Malat1 as a gene robustly downregulated in visceral white adipose tissue (vWAT) during normal aging in male mice and men. Reductions in Malat1 expression in subcutaneous WAT (scWAT) were also observed in genetic (ob and db) as well as diet-induced models of obesity. Based on these findings, Malat1+/+ and Malat1-/- mouse littermates were thus probed to detect whether loss of Malat1 would impact age or diet-induced gain in fat mass and development of glucose intolerance. Contrary to this hypothesis, male and female Malat1-deficient mice gained as much weight, and developed insulin resistance to a similar extent as their Malat1+/+ littermates when studied up to eight months old on regular chow or a high-fat, high-sucrose diet. Moreover, we observed no marked difference in oxygen consumption, food intake, or lipid profiles between Malat1+/+ and Malat1-/- mice. Therefore, we conclude that the overall metabolic impact of the absence of Malat1 on adipose tissue accretion and glucose intolerance is either physiologically not relevant upon aging and obesity, or that it is masked by as yet unknown compensatory mechanisms.
Collapse
|
103
|
Mishra R, Upadhyay A, Prajapati VK, Mishra A. Proteasome-mediated proteostasis: Novel medicinal and pharmacological strategies for diseases. Med Res Rev 2018; 38:1916-1973. [DOI: 10.1002/med.21502] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 03/13/2018] [Accepted: 04/04/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Ribhav Mishra
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan India
| | - Arun Upadhyay
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan India
| | - Vijay Kumar Prajapati
- Department of Biochemistry; School of Life Sciences; Central University of Rajasthan; Rajasthan India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan India
| |
Collapse
|
104
|
Zullo A, Simone E, Grimaldi M, Musto V, Mancini FP. Sirtuins as Mediator of the Anti-Ageing Effects of Calorie Restriction in Skeletal and Cardiac Muscle. Int J Mol Sci 2018; 19:E928. [PMID: 29561771 PMCID: PMC5979282 DOI: 10.3390/ijms19040928] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 03/14/2018] [Accepted: 03/20/2018] [Indexed: 12/17/2022] Open
Abstract
Fighting diseases and controlling the signs of ageing are the major goals of biomedicine. Sirtuins, enzymes with mainly deacetylating activity, could be pivotal targets of novel preventive and therapeutic strategies to reach such aims. Scientific proofs are accumulating in experimental models, but, to a minor extent, also in humans, that the ancient practice of calorie restriction could prove an effective way to prevent several degenerative diseases and to postpone the detrimental signs of ageing. In the present review, we summarize the evidence about the central role of sirtuins in mediating the beneficial effects of calorie restriction in skeletal and cardiac muscle since these tissues are greatly damaged by diseases and advancing years. Moreover, we entertain the possibility that the identification of sirtuin activators that mimic calorie restriction could provide the benefits without the inconvenience of this dietary style.
Collapse
Affiliation(s)
- Alberto Zullo
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy.
- CEINGE Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy.
| | - Emanuela Simone
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy.
| | - Maddalena Grimaldi
- Department of Pediatric Oncology and Hematology, Charité University Hospital, 13353 Berlin, Germany.
| | - Vincenzina Musto
- Department of Sciences and Technologies, University of Sannio, 82100 Benevento, Italy.
| | | |
Collapse
|
105
|
Spurrier J, Shukla AK, McLinden K, Johnson K, Giniger E. Altered expression of the Cdk5 activator-like protein, Cdk5α, causes neurodegeneration, in part by accelerating the rate of aging. Dis Model Mech 2018; 11:dmm031161. [PMID: 29469033 PMCID: PMC5897722 DOI: 10.1242/dmm.031161] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 02/02/2018] [Indexed: 12/11/2022] Open
Abstract
Aging is the greatest risk factor for neurodegeneration, but the connection between the two processes remains opaque. This is in part for want of a rigorous way to define physiological age, as opposed to chronological age. Here, we develop a comprehensive metric for physiological age in Drosophila, based on genome-wide expression profiling. We applied this metric to a model of adult-onset neurodegeneration, increased or decreased expression of the activating subunit of the Cdk5 protein kinase, encoded by the gene Cdk5α, the ortholog of mammalian p35. Cdk5α-mediated degeneration was associated with a 27-150% acceleration of the intrinsic rate of aging, depending on the tissue and genetic manipulation. Gene ontology analysis and direct experimental tests revealed that affected age-associated processes included numerous core phenotypes of neurodegeneration, including enhanced oxidative stress and impaired proteostasis. Taken together, our results suggest that Cdk5α-mediated neurodegeneration results from accelerated aging, in combination with cell-autonomous neuronal insults. These data fundamentally recast our picture of the relationship between neurodegeneration and its most prominent risk factor, natural aging.
Collapse
Affiliation(s)
- Joshua Spurrier
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 02892, USA
- The Johns Hopkins University/National Institutes of Health Graduate Partnership Program, National Institutes of Health, Bethesda, MD 02892, USA
| | - Arvind Kumar Shukla
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 02892, USA
| | - Kristina McLinden
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 02892, USA
| | - Kory Johnson
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 02892, USA
| | - Edward Giniger
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 02892, USA
| |
Collapse
|
106
|
Abstract
INTRODUCTION Pearson syndrome (PS) is a sporadic and very rare syndrome classically associated with single large-scale deletions of mitochondrial DNA and characterized by refractory sideroblastic anemia during infancy. Areas covered: This review presents an analysis and interpretation of the published data that forms the basis for our understanding of PS. PubMed, Google Scholarand Thompson ISI Web of Knowledge were searched for relevant data. Expert commentary: PS is a very rare mitochodrial disease that involves different organs and systems. Clinical phenotype is extremely variable and may change over the course of disease itself with the possibility both of worsenings and improvements. Outcome is invariably lethal and at the moment no cure is available. Accurate supportive treatment and follow up program in centres with experience in mitochondrial diseases and marrow failure may positively influence quality and duration of life.
Collapse
Affiliation(s)
- Piero Farruggia
- a Pediatric Hematology and Oncology Unit, Oncology Department , A.R.N.A.S. Ospedali Civico, Di Cristina e Benfratelli , Palermo , Italy
| | - Floriana Di Marco
- a Pediatric Hematology and Oncology Unit, Oncology Department , A.R.N.A.S. Ospedali Civico, Di Cristina e Benfratelli , Palermo , Italy
| | - Carlo Dufour
- b Clinical and Experimental Hematology Unit, G. Gaslini Children's Hospital , Genova , Italy
| |
Collapse
|
107
|
Aging-Associated Decline of Epidermal PSMD8 Contributes to Impaired Skin Function. J Invest Dermatol 2017; 138:976-978. [PMID: 29183730 DOI: 10.1016/j.jid.2017.10.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 10/06/2017] [Accepted: 10/25/2017] [Indexed: 01/11/2023]
|
108
|
Soundararajan R, Stearns TM, Czachor A, Fukumoto J, Turn C, Westermann-Clark E, Breitzig M, Tan L, Lockey RF, King BL, Kolliputi N. Global gene profiling of aging lungs in Atp8b1 mutant mice. Aging (Albany NY) 2017; 8:2232-2252. [PMID: 27689529 PMCID: PMC5076460 DOI: 10.18632/aging.101056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/10/2016] [Indexed: 12/18/2022]
Abstract
Objective Recent studies implicate cardiolipin oxidation in several age-related diseases. Atp8b1 encoding Type 4 P-type ATPases is a cardiolipin transporter. Mutation in Atp8b1 gene or inflammation of the lungs impairs the capacity of Atp8b1 to clear cardiolipin from lung fluid. However, the link between Atp8b1 mutation and age-related gene alteration is unknown. Therefore, we investigated how Atp8b1 mutation alters age-related genes. Methods We performed Affymetrix gene profiling of lungs isolated from young (7-9 wks, n=6) and aged (14 months, 14 M, n=6) C57BL/6 and Atp8b1 mutant mice. In addition, Ingenuity Pathway Analysis (IPA) was performed. Differentially expressed genes were validated by quantitative real-time PCR (qRT-PCR). Results Global transcriptome analysis revealed 532 differentially expressed genes in Atp8b1 lungs, 157 differentially expressed genes in C57BL/6 lungs, and 37 overlapping genes. IPA of age-related genes in Atp8b1 lungs showed enrichment of Xenobiotic metabolism and Nrf2-mediated signaling pathways. The increase in Adamts2 and Mmp13 transcripts in aged Atp8b1 lungs was validated by qRT-PCR. Similarly, the decrease in Col1a1 and increase in Cxcr6 transcripts was confirmed in both Atp8b1 mutant and C57BL/6 lungs. Conclusion Based on transcriptome profiling, our study indicates that Atp8b1 mutant mice may be susceptible to age-related lung diseases.
Collapse
Affiliation(s)
- Ramani Soundararajan
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | | | - Alexander Czachor
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Jutaro Fukumoto
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Christina Turn
- University of Florida College of Medicine, Gainesville, FL 32608, USA
| | - Emma Westermann-Clark
- Division of Allergy and Immunology, Department of Internal Medicine, James A Haley Veterans Hospital, Tampa, FL 33612, USA
| | - Mason Breitzig
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Lee Tan
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Richard F Lockey
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | | | - Narasaiah Kolliputi
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
109
|
Bonasera SJ, Arikkath J, Boska MD, Chaudoin TR, DeKorver NW, Goulding EH, Hoke TA, Mojtahedzedah V, Reyelts CD, Sajja B, Schenk AK, Tecott LH, Volden TA. Age-related changes in cerebellar and hypothalamic function accompany non-microglial immune gene expression, altered synapse organization, and excitatory amino acid neurotransmission deficits. Aging (Albany NY) 2017; 8:2153-2181. [PMID: 27689748 PMCID: PMC5076456 DOI: 10.18632/aging.101040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 09/07/2016] [Indexed: 11/25/2022]
Abstract
We describe age-related molecular and neuronal changes that disrupt mobility or energy balance based on brain region and genetic background. Compared to young mice, aged C57BL/6 mice exhibit marked locomotor (but not energy balance) impairments. In contrast, aged BALB mice exhibit marked energy balance (but not locomotor) impairments. Age-related changes in cerebellar or hypothalamic gene expression accompany these phenotypes. Aging evokes upregulation of immune pattern recognition receptors and cell adhesion molecules. However, these changes do not localize to microglia, the major CNS immunocyte. Consistent with a neuronal role, there is a marked age-related increase in excitatory synapses over the cerebellum and hypothalamus. Functional imaging of these regions is consistent with age-related synaptic impairments. These studies suggest that aging reactivates a developmental program employed during embryogenesis where immune molecules guide synapse formation and pruning. Renewed activity in this program may disrupt excitatory neurotransmission, causing significant behavioral deficits.
Collapse
Affiliation(s)
- Stephen J Bonasera
- Division of Geriatrics, University of Nebraska Medical Center, Durham Research Center II, Omaha, NE 68198, USA
| | - Jyothi Arikkath
- Monroe-Meyer Institute, University of Nebraska Medical Center, Durham Research Center II, Omaha, NE 68198, USA
| | - Michael D Boska
- Department of Radiology, University of Nebraska Medical Center, College of Medicine, Omaha, NE 68198, USA
| | - Tammy R Chaudoin
- Division of Geriatrics, University of Nebraska Medical Center, Durham Research Center II, Omaha, NE 68198, USA
| | - Nicholas W DeKorver
- Division of Geriatrics, University of Nebraska Medical Center, Durham Research Center II, Omaha, NE 68198, USA
| | - Evan H Goulding
- Department of Psychiatry and Behavioral Sciences, Northwestern University, Chicago, IL 60611, USA
| | - Traci A Hoke
- Division of Geriatrics, University of Nebraska Medical Center, Durham Research Center II, Omaha, NE 68198, USA
| | | | - Crystal D Reyelts
- Division of Geriatrics, University of Nebraska Medical Center, Durham Research Center II, Omaha, NE 68198, USA
| | - Balasrinivasa Sajja
- Department of Radiology, University of Nebraska Medical Center, College of Medicine, Omaha, NE 68198, USA
| | - A Katrin Schenk
- Department of Physics, Randolph College, Lynchburg, VA 24503, USA
| | - Laurence H Tecott
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Tiffany A Volden
- Division of Geriatrics, University of Nebraska Medical Center, Durham Research Center II, Omaha, NE 68198, USA
| |
Collapse
|
110
|
Das SK, Balasubramanian P, Weerasekara YK. Nutrition modulation of human aging: The calorie restriction paradigm. Mol Cell Endocrinol 2017; 455:148-157. [PMID: 28412520 PMCID: PMC7153268 DOI: 10.1016/j.mce.2017.04.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 04/08/2017] [Accepted: 04/09/2017] [Indexed: 12/20/2022]
Abstract
Globally, the aging population is growing rapidly, creating an urgent need to attenuate age-related health conditions, including metabolic disease and disability. A promising strategy for healthy aging based on consistently positive results from studies with a variety of species, including non-human primates (NHP), is calorie restriction (CR), or the restriction of energy intake while maintaining intake of essential nutrients. The burgeoning evidence for this approach in humans is reviewed and the major study to date to address this question, CALERIE (Comprehensive Assessment of the Long-term Effects of Reducing Intake of Energy), is described. CALERIE findings indicate the feasibility of CR in non-obese humans, confirm observations in NHP, and are consistent with improvements in disease risk reduction and potential anti-aging effects. Finally, the mechanisms of CR in humans are reviewed which sums up the fact that evolutionarily conserved mechanisms mediate the anti-aging effects of CR. Overall, the prospect for further research in both NHP and humans is highly encouraging.
Collapse
Affiliation(s)
- Sai Krupa Das
- Jean Mayer, US Department of Agriculture, Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA.
| | - Priya Balasubramanian
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison WI, USA.
| | - Yasoma K Weerasekara
- Jean Mayer, US Department of Agriculture, Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA.
| |
Collapse
|
111
|
Cummings NE, Lamming DW. Regulation of metabolic health and aging by nutrient-sensitive signaling pathways. Mol Cell Endocrinol 2017; 455:13-22. [PMID: 27884780 PMCID: PMC5440210 DOI: 10.1016/j.mce.2016.11.014] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 10/07/2016] [Accepted: 11/19/2016] [Indexed: 01/20/2023]
Abstract
All organisms need to be capable of adapting to changes in the availability and composition of nutrients. Over 75 years ago, researchers discovered that a calorie restricted (CR) diet could significantly extend the lifespan of rats, and since then a CR diet has been shown to increase lifespan and healthspan in model organisms ranging from yeast to non-human primates. In this review, we discuss the effects of a CR diet on metabolism and healthspan, and highlight emerging evidence that suggests that dietary composition - the precise macronutrients that compose the diet - may be just as important as caloric content. In particular, we discuss recent evidence that suggests protein quality may influence metabolic health. Finally, we discuss key metabolic pathways which may influence the response to CR diets and altered macronutrient composition. Understanding the molecular mechanisms responsible for the effects of CR and dietary composition on health and longevity may allow the design of novel therapeutic approaches to age-related diseases.
Collapse
Affiliation(s)
- Nicole E Cummings
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI, USA; Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI, USA; Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
112
|
Moreno CL, Mobbs CV. Epigenetic mechanisms underlying lifespan and age-related effects of dietary restriction and the ketogenic diet. Mol Cell Endocrinol 2017; 455:33-40. [PMID: 27884781 DOI: 10.1016/j.mce.2016.11.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 11/18/2016] [Accepted: 11/19/2016] [Indexed: 02/08/2023]
Abstract
Aging constitutes the central risk factor for major diseases including many forms of cancer, neurodegeneration, and cardiovascular diseases. The aging process is characterized by both global and tissue-specific changes in gene expression across taxonomically diverse species. While aging has historically been thought to entail cell-autonomous, even stochastic changes, recent evidence suggests that modulation of this process can be hierarchal, wherein manipulations of nutrient-sensing neurons (e.g., in the hypothalamus) produce peripheral effects that may modulate the aging process itself. The most robust intervention extending lifespan, plausibly impinging on the aging process, involves different modalities of dietary restriction (DR). Lifespan extension by DR is associated with broad protection against diseases (natural and engineered). Here we review potential epigenetic processes that may link lifespan to age-related diseases, particularly in the context of DR and (other) ketogenic diets, focusing on brain and hypothalamic mechanisms.
Collapse
Affiliation(s)
- Cesar L Moreno
- Department of Neurology, 1470 Madison Ave., 9-119, New York, NY 10029, USA
| | - Charles V Mobbs
- Departments of Neuroscience, Endocrinology, and Geriatrics, Icahn School of Medicine at Mount Sinai, 1470 Madison Ave., 9-119, New York, NY 10029, USA.
| |
Collapse
|
113
|
|
114
|
Sato S, Solanas G, Peixoto FO, Bee L, Symeonidi A, Schmidt MS, Brenner C, Masri S, Benitah SA, Sassone-Corsi P. Circadian Reprogramming in the Liver Identifies Metabolic Pathways of Aging. Cell 2017; 170:664-677.e11. [PMID: 28802039 PMCID: PMC7792549 DOI: 10.1016/j.cell.2017.07.042] [Citation(s) in RCA: 260] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/30/2017] [Accepted: 07/25/2017] [Indexed: 12/14/2022]
Abstract
The process of aging and circadian rhythms are intimately intertwined, but how peripheral clocks involved in metabolic homeostasis contribute to aging remains unknown. Importantly, caloric restriction (CR) extends lifespan in several organisms and rewires circadian metabolism. Using young versus old mice, fed ad libitum or under CR, we reveal reprogramming of the circadian transcriptome in the liver. These age-dependent changes occur in a highly tissue-specific manner, as demonstrated by comparing circadian gene expression in the liver versus epidermal and skeletal muscle stem cells. Moreover, de novo oscillating genes under CR show an enrichment in SIRT1 targets in the liver. This is accompanied by distinct circadian hepatic signatures in NAD+-related metabolites and cyclic global protein acetylation. Strikingly, this oscillation in acetylation is absent in old mice while CR robustly rescues global protein acetylation. Our findings indicate that the clock operates at the crossroad between protein acetylation, liver metabolism, and aging.
Collapse
Affiliation(s)
- Shogo Sato
- Center for Epigenetics and Metabolism, U1233 INSERM, University of California, Irvine, Irvine, CA 92607, USA
| | - Guiomar Solanas
- Institute for Research in Biomedicine, The Barcelona Institute of Science and Technology, 08028, Barcelona, Spain
| | - Francisca Oliveira Peixoto
- Institute for Research in Biomedicine, The Barcelona Institute of Science and Technology, 08028, Barcelona, Spain
| | - Leonardo Bee
- Center for Epigenetics and Metabolism, U1233 INSERM, University of California, Irvine, Irvine, CA 92607, USA
| | - Aikaterini Symeonidi
- Institute for Research in Biomedicine, The Barcelona Institute of Science and Technology, 08028, Barcelona, Spain
| | - Mark S Schmidt
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Charles Brenner
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Selma Masri
- Center for Epigenetics and Metabolism, U1233 INSERM, University of California, Irvine, Irvine, CA 92607, USA
| | - Salvador Aznar Benitah
- Institute for Research in Biomedicine, The Barcelona Institute of Science and Technology, 08028, Barcelona, Spain; ICREA, Catalan Institution for Research and Advanced Studies, Barcelona, Spain.
| | - Paolo Sassone-Corsi
- Center for Epigenetics and Metabolism, U1233 INSERM, University of California, Irvine, Irvine, CA 92607, USA.
| |
Collapse
|
115
|
Proteostasis of Huntingtin in Health and Disease. Int J Mol Sci 2017; 18:ijms18071568. [PMID: 28753941 PMCID: PMC5536056 DOI: 10.3390/ijms18071568] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 07/15/2017] [Accepted: 07/18/2017] [Indexed: 12/28/2022] Open
Abstract
Huntington's disease (HD) is a fatal neurodegenerative disorder characterized by motor dysfunction, cognitive deficits and psychosis. HD is caused by mutations in the Huntingtin (HTT) gene, resulting in the expansion of polyglutamine (polyQ) repeats in the HTT protein. Mutant HTT is prone to aggregation, and the accumulation of polyQ-expanded fibrils as well as intermediate oligomers formed during the aggregation process contribute to neurodegeneration. Distinct protein homeostasis (proteostasis) nodes such as chaperone-mediated folding and proteolytic systems regulate the aggregation and degradation of HTT. Moreover, polyQ-expanded HTT fibrils and oligomers can lead to a global collapse in neuronal proteostasis, a process that contributes to neurodegeneration. The ability to maintain proteostasis of HTT declines during the aging process. Conversely, mechanisms that preserve proteostasis delay the onset of HD. Here we will review the link between proteostasis, aging and HD-related changes.
Collapse
|
116
|
Stegeman R, Weake VM. Transcriptional Signatures of Aging. J Mol Biol 2017; 429:2427-2437. [PMID: 28684248 DOI: 10.1016/j.jmb.2017.06.019] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 06/02/2017] [Accepted: 06/28/2017] [Indexed: 12/31/2022]
Abstract
Genome-wide studies of aging have identified subsets of genes that show age-related changes in expression. Although the types of genes that are age regulated vary among different tissues and organisms, some patterns emerge from these large data sets. First, aging is associated with a broad induction of stress response pathways, although the specific genes and pathways involved differ depending on cell type and species. In contrast, a wide variety of functional classes of genes are downregulated with age, often including tissue-specific genes. Although the upregulation of age-regulated genes is likely to be governed by stress-responsive transcription factors, questions remain as to why particular genes are susceptible to age-related transcriptional decline. Here, we discuss recent findings showing that splicing is misregulated with age. While defects in splicing could lead to changes in protein isoform levels, they could also impact gene expression through nonsense-mediated decay of intron-retained transcripts. The discovery that splicing is misregulated with age suggests that other aspects of gene expression, such as transcription elongation, termination, and polyadenylation, must also be considered as potential mechanisms for age-related changes in transcript levels. Moreover, the considerable variation between genome-wide aging expression studies indicates that there is a critical need to analyze the transcriptional signatures of aging in single-cell types rather than whole tissues. Since age-associated decreases in gene expression could contribute to a progressive decline in cellular function, understanding the mechanisms that determine the aging transcriptome provides a potential target to extend healthy cellular lifespan.
Collapse
Affiliation(s)
- R Stegeman
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907, USA
| | - V M Weake
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907, USA; Purdue University Center for Cancer Research, West Lafayette, IN 47907, USA.
| |
Collapse
|
117
|
Ahanger MA, Akram NA, Ashraf M, Alyemeni MN, Wijaya L, Ahmad P. Plant responses to environmental stresses-from gene to biotechnology. AOB PLANTS 2017; 9:plx025. [PMID: 28775828 PMCID: PMC5534019 DOI: 10.1093/aobpla/plx025] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 06/25/2017] [Indexed: 05/21/2023]
Abstract
Increasing global population, urbanization and industrialization are increasing the rate of conversion of arable land into wasteland. Supplying food to an ever-increasing population is one of the biggest challenges that agriculturalists and plant scientists are currently confronting. Environmental stresses make this situation even graver. Despite the induction of several tolerance mechanisms, sensitive plants often fail to survive under environmental extremes. New technological approaches are imperative. Conventional breeding methods have a limited potential to improve plant genomes against environmental stress. Recently, genetic engineering has contributed enormously to the development of genetically modified varieties of different crops such as cotton, maize, rice, canola and soybean. The identification of stress-responsive genes and their subsequent introgression or overexpression within sensitive crop species are now being widely carried out by plant scientists. Engineering of important tolerance pathways, like antioxidant enzymes, osmolyte accumulation, membrane-localized transporters for efficient compartmentation of deleterious ions and accumulation of essential elements and resistance against pests or pathogens is also an area that has been intensively researched. In this review, the role of biotechnology and its successes, prospects and challenges in developing stress-tolerant crop cultivars are discussed.
Collapse
Affiliation(s)
| | - Nudrat Aisha Akram
- Department of Botany, Government College University, Faisalabad 38000, Pakistan
| | - Muhammad Ashraf
- Pakistan Science Foundation, Islamabad, Pakistan
- Department of Botany & Microbiology, King Saud University, Riyadh, Saudi Arabia
| | | | - Leonard Wijaya
- Department of Botany & Microbiology, King Saud University, Riyadh, Saudi Arabia
| | - Parvaiz Ahmad
- Department of Botany & Microbiology, King Saud University, Riyadh, Saudi Arabia
- Department of Botany, S.P. College, Srinagar, Jammu and Kashmir 190001, India
| |
Collapse
|
118
|
Trial J, Heredia CP, Taffet GE, Entman ML, Cieslik KA. Dissecting the role of myeloid and mesenchymal fibroblasts in age-dependent cardiac fibrosis. Basic Res Cardiol 2017; 112:34. [PMID: 28478479 DOI: 10.1007/s00395-017-0623-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 04/27/2017] [Indexed: 12/24/2022]
Abstract
Aging is associated with increased cardiac interstitial fibrosis and diastolic dysfunction. Our previous study has shown that mesenchymal fibroblasts in the C57BL/6J (B6J) aging mouse heart acquire an inflammatory phenotype and produce higher levels of chemokines. Monocyte chemoattractant protein-1 (MCP-1) secreted by these aged fibroblasts promotes leukocyte uptake into the heart. Some of the monocytes that migrate into the heart polarize into M2a macrophages/myeloid fibroblasts. The number of activated mesenchymal fibroblasts also increases with age, and consequently, both sources of fibroblasts contribute to fibrosis. Here, we further investigate mechanisms by which inflammation influences activation of myeloid and mesenchymal fibroblasts and their collagen synthesis. We examined cardiac fibrosis and heart function in three aged mouse strains; we compared C57BL/6J (B6J) with two other strains that have reduced inflammation via different mechanisms. Aged C57BL/6N (B6N) hearts are protected from oxidative stress and fibroblasts derived from them do not develop an inflammatory phenotype. Likewise, these mice have preserved diastolic function. Aged MCP-1 null mice on the B6J background (MCP-1KO) are protected from elevated leukocyte infiltration; they develop moderate but reduced fibrosis and diastolic dysfunction. Based on these studies, we further delineated the role of resident versus monocyte-derived M2a macrophages in myeloid-dependent fibrosis and found that the number of monocyte-derived M2a (but not resident) macrophages correlates with age-related fibrosis and diastolic dysfunction. In conclusion, we have found that ROS and inflammatory mediators are necessary for activation of fibroblasts of both developmental origins, and prevention of either led to better functional outcomes.
Collapse
Affiliation(s)
- JoAnn Trial
- Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, M.S. BCM620, Houston, TX, 77030, USA
| | - Celia Pena Heredia
- Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, M.S. BCM620, Houston, TX, 77030, USA
| | - George E Taffet
- Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, M.S. BCM620, Houston, TX, 77030, USA
| | - Mark L Entman
- Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, M.S. BCM620, Houston, TX, 77030, USA.,Houston Methodist, Houston, TX, USA
| | - Katarzyna A Cieslik
- Division of Cardiovascular Sciences and the DeBakey Heart Center, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, M.S. BCM620, Houston, TX, 77030, USA.
| |
Collapse
|
119
|
Role of DNA methylation in the dietary restriction mediated cellular memory. GeroScience 2017; 39:331-345. [PMID: 28477138 PMCID: PMC5505897 DOI: 10.1007/s11357-017-9976-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Accepted: 04/11/2017] [Indexed: 01/13/2023] Open
Abstract
An important facet of dietary restriction (DR) that has been largely overlooked is that DR can have early effects that create a cellular memory, which persists even when DR is discontinued. The goal of this study was to determine if DNA methylation played a role in the cellular memory of DR by examining the effect of short-term DR on gene expression and DNA methylation and determining if the changes in expression and DNA methylation persist when DR is discontinued and mice returned to ad libitum (AL) feeding. We show that DR can induce substantial changes in gene expression within 1 month of its implementation in various tissues, and more interestingly, ~19–50% of these changes in gene expression persist across the tissues even when DR is discontinued. We then determined whether DR induced changes in DNA methylation in the promoter of three candidate genes identified from our gene expression analysis (Pomc, Hsph1, and Nts1) that correlated with the changes in the expression of these genes. Decreased methylation at three specific CG sites in the promoter of the Nts1 gene encompassing the distal consensus AP-1 site was correlated with increased Nts1 expression. Both the promoter hypomethylation and increased Nts1 expression persisted even after DR was discontinued and mice fed AL, supporting our hypothesis that DNA methylation could play a role in the memory effect of DR. The changes in DNA methylation in the Nts1 gene are likely to occur in intestinal stem cells and could play a role in preserving the intestinal stem cell pool in DR mice.
Collapse
|
120
|
Feng Q, Chai GS, Wang ZH, Hu Y, Sun DS, Li XG, Ma RH, Li YR, Ke D, Wang JZ, Liu GP. Knockdown of pp32 Increases Histone Acetylation and Ameliorates Cognitive Deficits. Front Aging Neurosci 2017; 9:104. [PMID: 28473768 PMCID: PMC5397422 DOI: 10.3389/fnagi.2017.00104] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 03/31/2017] [Indexed: 01/10/2023] Open
Abstract
Aging is a cause of cognitive decline in the elderly and the major risk factor for Alzheimer's disease, however, aging people are not all destined to develop into cognitive deficits, the molecular mechanisms underlying this difference in cognition of aging people are obscure. Epigenetic modifications, particularly histone acetylation in the nervous system, play a critical role in regulation of gene expression for learning and memory. An inhibitor of acetyltransferases (INHAT) is reported to suppress histone acetylation via a histone-masking mechanism, and pp32 is a key component of INHAT complex. In the present study, we divided ~18 m-old aged mice into the cognitive-normal and the cognitive-impaired group by Morris water maze, and found that pp32 level was significantly increased in the hippocampus of cognitive-impaired aged mice. The mRNA and protein levels of synaptic-associated proteins decreased with reduced dendrite complexity and histone acetylation. Knockdown of pp32 rescued cognitive decline in cognitive-impaired aged mice with restoration of synaptic-associated proteins, the increase of spine density and elevation of histone acetylation. Our study reveals a novel mechanism underlying the aging-associated cognitive disturbance, indicating that suppression of pp32 might represent a promising therapeutic approach for learning and memory impairments.
Collapse
Affiliation(s)
- Qiong Feng
- Key Laboratory of Ministry of Education of China for Neurological Disorders, Department of Pathophysiology, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Gao-Shang Chai
- Key Laboratory of Ministry of Education of China for Neurological Disorders, Department of Pathophysiology, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China.,Department of Basic Medicine, Wuxi Medical School, Jiangnan UniversityWuxi, China
| | - Zhi-Hao Wang
- Key Laboratory of Ministry of Education of China for Neurological Disorders, Department of Pathophysiology, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Yu Hu
- Key Laboratory of Ministry of Education of China for Neurological Disorders, Department of Pathophysiology, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Dong-Sheng Sun
- Key Laboratory of Ministry of Education of China for Neurological Disorders, Department of Pathophysiology, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Xiao-Guang Li
- Key Laboratory of Ministry of Education of China for Neurological Disorders, Department of Pathophysiology, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Rong-Hong Ma
- Department of Laboratory Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and TechnologyWuhan, China
| | - Yi-Rong Li
- Department of Laboratory Medicine, Zhongnan Hospital, Wuhan UniversityWuhan, China
| | - Dan Ke
- Key Laboratory of Ministry of Education of China for Neurological Disorders, Department of Pathophysiology, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Jian-Zhi Wang
- Key Laboratory of Ministry of Education of China for Neurological Disorders, Department of Pathophysiology, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China.,Co-Innovation Center of Neuroregeneration, Nantong UniversityNantong, China
| | - Gong-Ping Liu
- Key Laboratory of Ministry of Education of China for Neurological Disorders, Department of Pathophysiology, Collaborative Innovation Center for Brain Science, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China.,Co-Innovation Center of Neuroregeneration, Nantong UniversityNantong, China
| |
Collapse
|
121
|
Lima GC, Vieira VCC, Cazarin CBB, Ribeiro RDR, Junior SB, de Albuquerque CL, Vidal RO, Netto CC, Yamada ÁT, Augusto F, Maróstica Junior MR. Fructooligosaccharide intake promotes epigenetic changes in the intestinal mucosa in growing and ageing rats. Eur J Nutr 2017; 57:1499-1510. [PMID: 28324207 DOI: 10.1007/s00394-017-1435-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 03/06/2017] [Indexed: 01/16/2023]
Abstract
PURPOSE The aim of this study was to investigate the relationship between fructooligosaccharide (FOS) intake at different life stages of Wistar rats and its stimulatory effects on intestinal parameters. METHODS Recently weaned and ageing female rats were divided into growing and ageing treatments, which were fed diets that partially replaced sucrose with FOS for 12 weeks. RESULTS Dietary FOS intake induced a significant increase in the numbers of Bifidobacterium and Lactobacillus in growing rats. FOS intake was associated with increased butyric acid levels and a reduced pH of the caecal contents at both ages. Differential gene expression patterns were observed by microarray analysis of growing and ageing animals fed the FOS diet. A total of 133 genes showed detectable changes in expression in the growing rats, while there were only 19 gene expression changes in ageing rats fed with FOS. CONCLUSION These results suggest that dietary FOS intake may be beneficial for some parameters of intestinal health in growing rats.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ramon Oliveira Vidal
- Sainte-Justine University Hospital Center, Université de Montreal, Montreal, Canada
| | - Claudia Cardoso Netto
- Department of Biochemistry, Biological Sciences and Health Center, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Fabio Augusto
- Institute of Chemistry, University of Campinas, Campinas, São Paulo, Brazil
| | - Mário Roberto Maróstica Junior
- School of Food Engineering, University of Campinas, Campinas, São Paulo, Brazil. .,Laboratório de Nutrição e Metabolismo-Departamento de Alimentos e Nutrição, Faculdade de Engenharia de Alimentos, Universidade Estadual de Campinas, R. Monteiro Lobato 80, Campinas, SP, 13083-862, Brazil.
| |
Collapse
|
122
|
The impact of oxidative stress and inflammation on RPE degeneration in non-neovascular AMD. Prog Retin Eye Res 2017; 60:201-218. [PMID: 28336424 DOI: 10.1016/j.preteyeres.2017.03.002] [Citation(s) in RCA: 551] [Impact Index Per Article: 68.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/13/2017] [Accepted: 03/14/2017] [Indexed: 02/07/2023]
Abstract
The retinal pigment epithelium (RPE) is a highly specialized, unique epithelial cell that interacts with photoreceptors on its apical side and with Bruch's membrane and the choriocapillaris on its basal side. Due to vital functions that keep photoreceptors healthy, the RPE is essential for maintaining vision. With aging and the accumulated effects of environmental stresses, the RPE can become dysfunctional and die. This degeneration plays a central role in age-related macular degeneration (AMD) pathobiology, the leading cause of blindness among the elderly in western societies. Oxidative stress and inflammation have both physiological and potentially pathological roles in RPE degeneration. Given the central role of the RPE, this review will focus on the impact of oxidative stress and inflammation on the RPE with AMD pathobiology. Physiological sources of oxidative stress as well as unique sources from photo-oxidative stress, the phagocytosis of photoreceptor outer segments, and modifiable factors such as cigarette smoking and high fat diet ingestion that can convert oxidative stress into a pathological role, and the negative impact of impairing the cytoprotective roles of mitochondrial dynamics and the Nrf2 signaling system on RPE health in AMD will be discussed. Likewise, the response by the innate immune system to an inciting trigger, and the potential role of local RPE production of inflammation, as well as a potential role for damage by inflammation with chronicity if the inciting trigger is not neutralized, will be debated.
Collapse
|
123
|
Uno Y, Takata R, Kito G, Yamazaki H, Nakagawa K, Nakamura Y, Kamataki T, Katagiri T. Sex- and age-dependent gene expression in human liver: An implication for drug-metabolizing enzymes. Drug Metab Pharmacokinet 2017; 32:100-107. [DOI: 10.1016/j.dmpk.2016.10.409] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 10/17/2016] [Accepted: 10/17/2016] [Indexed: 01/08/2023]
|
124
|
Keenan KP, Hoe CM, Mixson L, McCoy CL, Coleman JB, Mattson BA, Ballam GA, Gumprecht LA, Soper KA. Diabesity: A Polygenic Model of Dietary-Induced Obesity from Ad Libitum Overfeeding of Sprague–Dawley Rats and Its Modulation by Moderate and Marked Dietary Restriction. Toxicol Pathol 2017; 33:650-74. [PMID: 16207639 DOI: 10.1080/01926230500311222] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
This study compared the effects of ad libitum (AL) overfeeding and moderate or marked dietary restriction (DR) on the pathogenesis of a metabolic syndrome of diabesity comprised of age-related degenerative diseases and obesity in a outbred stock of Sprague–Dawley (SD) rats [Crl:CD (SD) IGS BR]. SD rats were fed Purina Certified Rodent Diet AL (group 1), DR at 72–79% of AL (group 2), DR at 68–72% of AL (group 3) or DR at 47–48% of AL (group 4) for 106 weeks. Interim necropsies were performed at 13, 26, and 53 weeks, after a 7-day 5-bromo-2-deoxyuridine (BrdU)-filled minipump implantation. Body weights, organ weights, carcass analysis, in-life data including estrous cyclicity, and histopathology were determined. At 6–7 weeks of age SD rats had 6% body fat. AL-feeding resulted in hypertriglyceridemia, hypercholesterolemia, and dietary-induced obesity (DIO) by study week 14, with 25% body fat that progressed to 36–42% body fat by 106 weeks. As early as 14 weeks, key biomarkers developed for spontaneous nephropathy, cardiomyopathy, and degenerative changes in multiple organ systems. Early endocrine disruption was indicated by changes in metabolic and endocrine profiles and the early development and progression of lesions in the pituitary, pancreatic islets, adrenals, thyroids, parathyroids, liver, kidneys, and other tissues. Reproductive senescence was seen by 9 months with declines in estrous cyclicity and pathological changes in the reproductive organs of both sexes fed AL or moderate DR, but not marked DR. The diabesity syndrome in AL-fed, DIO SD rats was readily modulated or prevented by moderate to marked DR. Moderate DR of balanced diets resulted in a better toxicology model by significantly improving survival, controlling adult body weight and obesity, reducing the onset, severity, and morbidity of age-related renal, endocrine, metabolic, and cardiac diseases. Moderate DR feeding reduces study-to-study variability, increases treatment exposure time, and increases the ability to distinguish true treatment effects from spontaneous aging. The structural and metabolic differences between the phenotypes of DIO and DR SD rats indicated changes of polygenic expression over time in this outbred stock. AL-overfeeding of SD rats produces a needed model of DIO and diabesity that needs further study of its patterns of polygenic expression and phenotype.
Collapse
Affiliation(s)
- Kevin P Keenan
- Merck Research Laboratories, Department of Biometrics, West Point, Pennsylvania 19486, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
125
|
Chung HJ, Lee HK, Kim HJ, Baek SH, Hong ST. Gene expression profiles and physiological data from mice fed resveratrol-enriched rice DJ526. Sci Data 2016; 3:160114. [PMID: 27996975 PMCID: PMC5170592 DOI: 10.1038/sdata.2016.114] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 11/01/2016] [Indexed: 01/07/2023] Open
Abstract
The molecular mechanism underlying lifespan extension by resveratrol remains widely discussed. To help study this mechanism, we previously created resveratrol-enriched rice, DJ526, by transferring the resveratrol biosynthesis gene into Dongjin rice. DJ526 accumulates 1.4-1.9 μg g-1 of resveratrol in its grain and can ameliorates age-related deterioration in mice, as compared to control animals, based on assessments of motor coordination, physical strength and cutaneous tissue aging. Here, we present raw data sets, deposited in public repositories, from microarray analysis and physiological data of mice fed with DJ526 and Dongjin rice and treated with resveratrol. We also provide a method to analyze blood serum at micron levels. These data sets may help other researchers find new clues regarding the etiology of the anti-aging process and signaling pathways induced by resveratrol, rice, or DJ526.
Collapse
Affiliation(s)
- Hea-Jong Chung
- Department of Biomedical Sciences and Institute for Medical Science, Chonbuk National University Medical School, Jeonju, Chonbuk 54907, South Korea
| | - Heui-Kwan Lee
- Deparment of Radiation oncology, Presbyterian Medical Center, Seonam University Medical School, Jeonju, Chonbuk 54987, South Korea
| | - Hyeon-Jin Kim
- JINIS BDRD institute, JINIS Biopharmaceuticals Co., 948-9 Dunsan, Bongdong, Wanju, Chonbuk 55321, South Korea
| | - So-Hyeon Baek
- Department of Well-being Resources, Sunchon National University, Suncheon, Jeonnam 57922, South Korea
| | - Seong-Tshool Hong
- Department of Biomedical Sciences and Institute for Medical Science, Chonbuk National University Medical School, Jeonju, Chonbuk 54907, South Korea
| |
Collapse
|
126
|
Bota DA, Davies KJA. Mitochondrial Lon protease in human disease and aging: Including an etiologic classification of Lon-related diseases and disorders. Free Radic Biol Med 2016; 100:188-198. [PMID: 27387767 PMCID: PMC5183306 DOI: 10.1016/j.freeradbiomed.2016.06.031] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 06/21/2016] [Accepted: 06/29/2016] [Indexed: 12/20/2022]
Abstract
The Mitochondrial Lon protease, also called LonP1 is a product of the nuclear gene LONP1. Lon is a major regulator of mitochondrial metabolism and response to free radical damage, as well as an essential factor for the maintenance and repair of mitochondrial DNA. Lon is an ATP-stimulated protease that cycles between being bound (at the inner surface of the inner mitochondrial membrane) to the mitochondrial genome, and being released into the mitochondrial matrix where it can degrade matrix proteins. At least three different roles or functions have been ascribed to Lon: 1) Proteolytic digestion of oxidized proteins and the turnover of specific essential mitochondrial enzymes such as aconitase, TFAM, and StAR; 2) Mitochondrial (mt)DNA-binding protein, involved in mtDNA replication and mitogenesis; and 3) Protein chaperone, interacting with the Hsp60-mtHsp70 complex. LONP1 orthologs have been studied in bacteria, yeast, flies, worms, and mammals, evincing the widespread importance of the gene, as well as its remarkable evolutionary conservation. In recent years, we have witnessed a significant increase in knowledge regarding Lon's involvement in physiological functions, as well as in an expanding array of human disorders, including cancer, neurodegeneration, heart disease, and stroke. In addition, Lon appears to have a significant role in the aging process. A number of mitochondrial diseases have now been identified whose mechanisms involve various degrees of Lon dysfunction. In this paper we review current knowledge of Lon's function, under normal conditions, and we propose a new classification of human diseases characterized by a either over-expression or decline or loss of function of Lon. Lon has also been implicated in human aging, and we review the data currently available as well as speculating about possible interactions of aging and disease. Finally, we also discuss Lon as potential therapeutic target in human disease.
Collapse
Affiliation(s)
- Daniela A Bota
- Department of Neurology and Chao Family Comprehensive Cancer Center, UC Irvine School of Medicine, 200 S. Manchester Ave., Suite 206, Orange, CA 92868, USA.
| | - Kelvin J A Davies
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, Los Angeles, CA 90089-0191, USA; Division of Molecular & Computational Biology, Department of Biological Sciences, Dornsife College of Letters, Arts, & Sciences, The University of Southern California, Los Angeles, CA 90089-0191, USA
| |
Collapse
|
127
|
Hibshman JD, Hung A, Baugh LR. Maternal Diet and Insulin-Like Signaling Control Intergenerational Plasticity of Progeny Size and Starvation Resistance. PLoS Genet 2016; 12:e1006396. [PMID: 27783623 PMCID: PMC5081166 DOI: 10.1371/journal.pgen.1006396] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 09/29/2016] [Indexed: 12/12/2022] Open
Abstract
Maternal effects of environmental conditions produce intergenerational phenotypic plasticity. Adaptive value of these effects depends on appropriate anticipation of environmental conditions in the next generation, and mismatch between conditions may contribute to disease. However, regulation of intergenerational plasticity is poorly understood. Dietary restriction (DR) delays aging but maternal effects have not been investigated. We demonstrate maternal effects of DR in the roundworm C. elegans. Worms cultured in DR produce fewer but larger progeny. Nutrient availability is assessed in late larvae and young adults, rather than affecting a set point in young larvae, and maternal age independently affects progeny size. Reduced signaling through the insulin-like receptor daf-2/InsR in the maternal soma causes constitutively large progeny, and its effector daf-16/FoxO is required for this effect. nhr-49/Hnf4, pha-4/FoxA, and skn-1/Nrf also regulate progeny-size plasticity. Genetic analysis suggests that insulin-like signaling controls progeny size in part through regulation of nhr-49/Hnf4, and that pha-4/FoxA and skn-1/Nrf function in parallel to insulin-like signaling and nhr-49/Hnf4. Furthermore, progeny of DR worms are buffered from adverse consequences of early-larval starvation, growing faster and producing more offspring than progeny of worms fed ad libitum. These results suggest a fitness advantage when mothers and their progeny experience nutrient stress, compared to an environmental mismatch where only progeny are stressed. This work reveals maternal provisioning as an organismal response to DR, demonstrates potentially adaptive intergenerational phenotypic plasticity, and identifies conserved pathways mediating these effects. Information from a mother’s environment can be transmitted to her offspring. In theory, the way mothers provision offspring can be beneficial or pathological depending on whether the environments of the mother and her offspring match. We find that roundworms fed a restricted diet produce fewer but larger offspring. These offspring recover better from starvation, growing faster and having increased fertility. Thus, we find that worms are more likely to thrive after early-life starvation if their mothers have been preconditioned with limited nutrient availability. We describe a genetic network that mediates effects of a mother’s diet on the size and starvation resistance of her offspring. The same genes required to extend the lifespan of worms fed a restricted diet are also required for the differential maternal provisioning we describe. In particular, insulin-like signaling, pha-4/FoxA, skn-1/Nrf, and nhr-49/Hnf4 function in the mother to transmit information about her diet to her offspring. Our work underscores the impact of maternal diet on reproductive health, with consequences for offspring physiology. The conserved genetic network controlling such effects of diet across generations is likely relevant to human diseases related to nutrient sensing and storage.
Collapse
Affiliation(s)
- Jonathan D. Hibshman
- Department of Biology, Duke University, Durham, North Carolina, United States of America
- University Program in Genetics and Genomics, Duke University, Durham, North Carolina, United States of America
| | - Anthony Hung
- Department of Biology, Duke University, Durham, North Carolina, United States of America
| | - L. Ryan Baugh
- Department of Biology, Duke University, Durham, North Carolina, United States of America
- University Program in Genetics and Genomics, Duke University, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
128
|
Ntsapi C, Loos B. Caloric restriction and the precision-control of autophagy: A strategy for delaying neurodegenerative disease progression. Exp Gerontol 2016; 83:97-111. [DOI: 10.1016/j.exger.2016.07.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Revised: 07/18/2016] [Accepted: 07/25/2016] [Indexed: 01/07/2023]
|
129
|
Liu Y, Wang TT, Zhang R, Fu WY, Wang X, Wang F, Gao P, Ding YN, Xie Y, Hao DL, Chen HZ, Liu DP. Calorie restriction protects against experimental abdominal aortic aneurysms in mice. J Exp Med 2016; 213:2473-2488. [PMID: 27670594 PMCID: PMC5068228 DOI: 10.1084/jem.20151794] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 08/26/2016] [Indexed: 12/29/2022] Open
Abstract
Abdominal aortic aneurysm (AAA), characterized by a localized dilation of the abdominal aorta, is a life-threatening vascular pathology. Because of the current lack of effective treatment for AAA rupture, prevention is of prime importance for AAA management. Calorie restriction (CR) is a nonpharmacological intervention that delays the aging process and provides various health benefits. However, whether CR prevents AAA formation remains untested. In this study, we subjected Apoe-/- mice to 12 wk of CR and then examined the incidence of angiotensin II (AngII)-induced AAA formation. We found that CR markedly reduced the incidence of AAA formation and attenuated aortic elastin degradation in Apoe-/- mice. The expression and activity of Sirtuin 1 (SIRT1), a key metabolism/energy sensor, were up-regulated in vascular smooth muscle cells (VSMCs) upon CR. Importantly, the specific ablation of SIRT1 in smooth muscle cells abolished the preventive effect of CR on AAA formation in Apoe-/- mice. Mechanistically, VSMC-SIRT1-dependent deacetylation of histone H3 lysine 9 on the matrix metallopeptidase 2 (Mmp2) promoter was required for CR-mediated suppression of AngII-induced MMP2 expression. Together, our findings suggest that CR may be an effective intervention that protects against AAA formation.
Collapse
Affiliation(s)
- Yue Liu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Ting-Ting Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Ran Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Wen-Yan Fu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Xu Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Fang Wang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Peng Gao
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yang-Nan Ding
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yan Xie
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - De-Long Hao
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Hou-Zao Chen
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - De-Pei Liu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
130
|
Hoeijmakers L, Heinen Y, van Dam AM, Lucassen PJ, Korosi A. Microglial Priming and Alzheimer's Disease: A Possible Role for (Early) Immune Challenges and Epigenetics? Front Hum Neurosci 2016; 10:398. [PMID: 27555812 PMCID: PMC4977314 DOI: 10.3389/fnhum.2016.00398] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 07/26/2016] [Indexed: 12/11/2022] Open
Abstract
Neuroinflammation is thought to contribute to Alzheimer's disease (AD) pathogenesis that is, to a large extent, mediated by microglia. Given the tight interaction between the immune system and the brain, peripheral immune challenges can profoundly affect brain function. Indeed, both preclinical and clinical studies have indicated that an aberrant inflammatory response can elicit behavioral impairments and cognitive deficits, especially when the brain is in a vulnerable state, e.g., during early development, as a result of aging, or under disease conditions like AD. However, how exactly peripheral immune challenges affect brain function and whether this is mediated by aberrant microglial functioning remains largely elusive. In this review, we hypothesize that: (1) systemic immune challenges occurring during vulnerable periods of life can increase the propensity to induce later cognitive dysfunction and accelerate AD pathology; and (2) that "priming" of microglial cells is instrumental in mediating this vulnerability. We highlight how microglia can be primed by both neonatal infections as well as by aging, two periods of life during which microglial activity is known to be specifically upregulated. Lasting changes in (the ratios of) specific microglial phenotypes can result in an exaggerated pro-inflammatory cytokine response to subsequent inflammatory challenges. While the resulting changes in brain function are initially transient, a continued and/or excess release of such pro-inflammatory cytokines can activate various downstream cellular cascades known to be relevant for AD. Finally, we discuss microglial priming and the aberrant microglial response as potential target for treatment strategies for AD.
Collapse
Affiliation(s)
- Lianne Hoeijmakers
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam Amsterdam, Netherlands
| | - Yvonne Heinen
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam Amsterdam, Netherlands
| | - Anne-Marie van Dam
- Department of Anatomy and Neurosciences, Neuroscience Campus Amsterdam, VU University Medical Center Amsterdam, Netherlands
| | - Paul J Lucassen
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam Amsterdam, Netherlands
| | - Aniko Korosi
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam Amsterdam, Netherlands
| |
Collapse
|
131
|
Affiliation(s)
- S Michal Jazwinski
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1901 Perdido Street, Box P7-2, New Orleans, Louisiana 70112 USA
| |
Collapse
|
132
|
Gadbury GL, Page GP, Edwards J, Kayo T, Prolla TA, Weindruch R, Permana PA, Mountz JD, Allison DB. Power and sample size estimation in high dimensional biology. Stat Methods Med Res 2016. [DOI: 10.1191/0962280204sm369ra] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Genomic scientists often test thousands of hypotheses in a single experiment. One example is a microarray experiment that seeks to determine differential gene expression among experimental groups. Planning such experiments involves a determination of sample size that will allow meaningful interpretations. Traditional power analysis methods may not be well suited to this task when thousands of hypotheses are tested in a discovery oriented basic research. We introduce the concept of expected discovery rate (EDR) and an approach that combines parametric mixture modelling with parametric bootstrapping to estimate the sample size needed for a desired accuracy of results. While the examples included are derived from microarray studies, the methods, herein, are ‘extraparadigmatic’ in the approach to study design and are applicable to most high dimensional biological situations. Pilot data from three different microarray experiments are used to extrapolate EDR as well as the related false discovery rate at different sample sizes and thresholds.
Collapse
Affiliation(s)
- Gary L Gadbury
- Department of Mathematics and Statistics, University of Missouri - Rolla, MO, USA
| | - Grier P Page
- USDA ARS, Department of Agronomy, Iowa State University, Ames, IA, USA
| | - Jode Edwards
- USDA ARS, Department of Agronomy, Iowa State University, Ames, IA, USA
| | - Tsuyoshi Kayo
- Wisconsin Regional Primate Research Center, Madison, WI, USA
| | - Tomas A Prolla
- Department of Genetics and Medical Genetics, University of Wisconsin, Madison, WI, USA
| | - Richard Weindruch
- Department of Medicine, University of Wisconsin and The Geriatric Research, Education, and Clinical Center, William S Middleton VA Hospital, Madison, WI, USA
| | - Paska A Permana
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, USA
| | - John D Mountz
- The Birmingham Veterans Administration Medical Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David B Allison
- Department of Biostatistics, Section on Statistical Genetics, and Clinical Nutrition Research Center, University of Alabama at Birmingham, Birmingham, AL, USA,
| |
Collapse
|
133
|
Tepp K, Timohhina N, Puurand M, Klepinin A, Chekulayev V, Shevchuk I, Kaambre T. Bioenergetics of the aging heart and skeletal muscles: Modern concepts and controversies. Ageing Res Rev 2016; 28:1-14. [PMID: 27063513 DOI: 10.1016/j.arr.2016.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 04/01/2016] [Accepted: 04/04/2016] [Indexed: 01/03/2023]
Abstract
Age-related alterations in the bioenergetics of the heart and oxidative skeletal muscle tissues are of crucial influence on their performance. Until now the prevailing concept of aging was the mitochondrial theory, the increased production of reactive oxygen species, mediated by deficiency in the activity of respiratory chain complexes. However, studies with mitochondria in situ have presented results which, to some extent, disagree with previous ones, indicating that the mitochondrial theory of aging may be overestimated. The studies reporting age-related decline in mitochondrial function were performed using mainly isolated mitochondria. Measurements on this level are not able to take into account the system level properties. The relevant information can be obtained only from appropriate studies using cells or tissue fibers. The functional interactions between the components of Intracellular Energetic Unit (ICEU) regulate the energy production and consumption in oxidative muscle cells. The alterations of these interactions in ICEU should be studied in order to find a more effective protocol to decelerate the age-related changes taking place in the energy metabolism. In this article, an overview is given of the present theories and controversies of causes of age-related alterations in bioenergetics. Also, branches of study, which need more emphasis, are indicated.
Collapse
Affiliation(s)
- Kersti Tepp
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia.
| | - Natalja Timohhina
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia
| | - Marju Puurand
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia
| | - Aleksandr Klepinin
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia
| | - Vladimir Chekulayev
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia
| | - Igor Shevchuk
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia
| | - Tuuli Kaambre
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia; Faculty of Science, Tallinn University, Narva mnt. 25, 10120, Estonia
| |
Collapse
|
134
|
Bullard SA, Seo S, Schilling B, Dyle MC, Dierdorff JM, Ebert SM, DeLau AD, Gibson BW, Adams CM. Gadd45a Protein Promotes Skeletal Muscle Atrophy by Forming a Complex with the Protein Kinase MEKK4. J Biol Chem 2016; 291:17496-17509. [PMID: 27358404 PMCID: PMC5016147 DOI: 10.1074/jbc.m116.740308] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Indexed: 12/13/2022] Open
Abstract
Skeletal muscle atrophy is a serious and highly prevalent condition that remains poorly understood at the molecular level. Previous work found that skeletal muscle atrophy involves an increase in skeletal muscle Gadd45a expression, which is necessary and sufficient for skeletal muscle fiber atrophy. However, the direct mechanism by which Gadd45a promotes skeletal muscle atrophy was unknown. To address this question, we biochemically isolated skeletal muscle proteins that associate with Gadd45a as it induces atrophy in mouse skeletal muscle fibers in vivo. We found that Gadd45a interacts with multiple proteins in skeletal muscle fibers, including, most prominently, MEKK4, a mitogen-activated protein kinase kinase kinase that was not previously known to play a role in skeletal muscle atrophy. Furthermore, we found that, by forming a complex with MEKK4 in skeletal muscle fibers, Gadd45a increases MEKK4 protein kinase activity, which is both sufficient to induce skeletal muscle fiber atrophy and required for Gadd45a-mediated skeletal muscle fiber atrophy. Together, these results identify a direct biochemical mechanism by which Gadd45a induces skeletal muscle atrophy and provide new insight into the way that skeletal muscle atrophy occurs at the molecular level.
Collapse
Affiliation(s)
- Steven A Bullard
- From the Department of Internal Medicine.,Fraternal Order of Eagles Diabetes Research Center, and.,Departments of Molecular Physiology and Biophysics and.,the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Seongjin Seo
- Ophthalmology and Visual Sciences, University of Iowa, Iowa City, Iowa 52242
| | - Birgit Schilling
- the Buck Institute for Research on Aging, Novato, California 94945, and
| | - Michael C Dyle
- From the Department of Internal Medicine.,Fraternal Order of Eagles Diabetes Research Center, and.,Departments of Molecular Physiology and Biophysics and.,the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246.,Emmyon, Inc., Coralville, Iowa 52241
| | - Jason M Dierdorff
- From the Department of Internal Medicine.,Fraternal Order of Eagles Diabetes Research Center, and.,Departments of Molecular Physiology and Biophysics and.,the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Scott M Ebert
- From the Department of Internal Medicine.,Fraternal Order of Eagles Diabetes Research Center, and.,Departments of Molecular Physiology and Biophysics and.,the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246.,Emmyon, Inc., Coralville, Iowa 52241
| | - Austin D DeLau
- From the Department of Internal Medicine.,Fraternal Order of Eagles Diabetes Research Center, and.,Departments of Molecular Physiology and Biophysics and.,the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246
| | - Bradford W Gibson
- the Buck Institute for Research on Aging, Novato, California 94945, and.,the Department of Pharmaceutical Chemistry, University of California at San Francisco, San Francisco, California 94143
| | - Christopher M Adams
- From the Department of Internal Medicine, .,Fraternal Order of Eagles Diabetes Research Center, and.,Departments of Molecular Physiology and Biophysics and.,the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246.,Emmyon, Inc., Coralville, Iowa 52241
| |
Collapse
|
135
|
Liebl MP, Hoppe T. It's all about talking: two-way communication between proteasomal and lysosomal degradation pathways via ubiquitin. Am J Physiol Cell Physiol 2016; 311:C166-78. [PMID: 27225656 DOI: 10.1152/ajpcell.00074.2016] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Selective degradation of proteins requires a fine-tuned coordination of the two major proteolytic pathways, the ubiquitin-proteasome system (UPS) and autophagy. Substrate selection and proteolytic activity are defined by a plethora of regulatory cofactors influencing each other. Both proteolytic pathways are initiated by ubiquitylation to mark substrate proteins for degradation, although the size and/or topology of the modification are different. In this context E3 ubiquitin ligases, ensuring the covalent attachment of activated ubiquitin to the substrate, are of special importance. The regulation of E3 ligase activity, competition between different E3 ligases for binding E2 conjugation enzymes and substrates, as well as their interplay with deubiquitylating enzymes (DUBs) represent key events in the cross talk between the UPS and autophagy. The coordination between both degradation routes is further influenced by heat shock factors and ubiquitin-binding proteins (UBPs) such as p97, p62, or optineurin. Mutations in enzymes and ubiquitin-binding proteins or a general decline of both proteolytic systems during aging result in accumulation of damaged and aggregated proteins. Thus further mechanistic understanding of how UPS and autophagy communicate might allow therapeutic intervention especially against age-related diseases.
Collapse
Affiliation(s)
- Martina P Liebl
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Thorsten Hoppe
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| |
Collapse
|
136
|
Boehm A, Arnoldini M, Bergmiller T, Röösli T, Bigosch C, Ackermann M. Genetic Manipulation of Glycogen Allocation Affects Replicative Lifespan in E. coli. PLoS Genet 2016; 12:e1005974. [PMID: 27093302 PMCID: PMC4836754 DOI: 10.1371/journal.pgen.1005974] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 03/13/2016] [Indexed: 11/18/2022] Open
Abstract
In bacteria, replicative aging manifests as a difference in growth or survival between the two cells emerging from division. One cell can be regarded as an aging mother with a decreased potential for future survival and division, the other as a rejuvenated daughter. Here, we aimed at investigating some of the processes involved in aging in the bacterium Escherichia coli, where the two types of cells can be distinguished by the age of their cell poles. We found that certain changes in the regulation of the carbohydrate metabolism can affect aging. A mutation in the carbon storage regulator gene, csrA, leads to a dramatically shorter replicative lifespan; csrA mutants stop dividing once their pole exceeds an age of about five divisions. These old-pole cells accumulate glycogen at their old cell poles; after their last division, they do not contain a chromosome, presumably because of spatial exclusion by the glycogen aggregates. The new-pole daughters produced by these aging mothers are born young; they only express the deleterious phenotype once their pole is old. These results demonstrate how manipulations of nutrient allocation can lead to the exclusion of the chromosome and limit replicative lifespan in E. coli, and illustrate how mutations can have phenotypic effects that are specific for cells with old poles. This raises the question how bacteria can avoid the accumulation of such mutations in their genomes over evolutionary times, and how they can achieve the long replicative lifespans that have recently been reported. Bacteria were often considered to be potentially immortal and free of aging. This expectation was based on the idea that the two cells emerging from bacterial division are identical and thus also equally old. However, a number of recent studies followed individual bacterial cells over consecutive divisions and reported that individuals with old cell poles show reduced survival and growth. This indicates that at least some types of bacteria age. We were interested in how mutations can affect the aging process and replicative lifespan of bacteria. In eukaryotes the aging process is thought to be modulated by mutations with age-specific effects. For example, there are mutations that have no measureable phenotypic effect early in life but are deleterious later in life. Such mutations play a key role in the evolution of eukaryotic aging and are thought to be responsible for lifespan differences between species. Here, we report that mutations can also have age-specific effects in bacteria. We describe a mutation in E. coli with a deleterious effect on growth and division that only manifests in cells whose cell pole is about four to five divisions old. These results illustrate how mutations can act in an age-specific manner in bacteria, and raise questions about how bacterial lifespan is modulated by such mutations.
Collapse
Affiliation(s)
- Alex Boehm
- Biozentrum, University of Basel, Switzerland
- Philipps-Universität Marburg, LOEWE-Zentrum für Synthetische Mikrobiologie, Marburg, Germany
| | - Markus Arnoldini
- Institute of Biogeochemistry and Pollutant Dynamics, Department of Environmental Systems Science, ETH Zurich, Switzerland
- Department of Environmental Microbiology, Eawag, Dübendorf, Switzerland
- Department of Physics, University of California, San Diego, La Jolla, United States of America
| | - Tobias Bergmiller
- Institute of Biogeochemistry and Pollutant Dynamics, Department of Environmental Systems Science, ETH Zurich, Switzerland
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Thomas Röösli
- Institute of Biogeochemistry and Pollutant Dynamics, Department of Environmental Systems Science, ETH Zurich, Switzerland
- Department of Environmental Microbiology, Eawag, Dübendorf, Switzerland
| | - Colette Bigosch
- Institute of Biogeochemistry and Pollutant Dynamics, Department of Environmental Systems Science, ETH Zurich, Switzerland
- Department of Environmental Microbiology, Eawag, Dübendorf, Switzerland
| | - Martin Ackermann
- Institute of Biogeochemistry and Pollutant Dynamics, Department of Environmental Systems Science, ETH Zurich, Switzerland
- Department of Environmental Microbiology, Eawag, Dübendorf, Switzerland
- * E-mail:
| |
Collapse
|
137
|
López-Lluch G, Navas P. Calorie restriction as an intervention in ageing. J Physiol 2016; 594:2043-60. [PMID: 26607973 PMCID: PMC4834802 DOI: 10.1113/jp270543] [Citation(s) in RCA: 189] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 11/21/2015] [Indexed: 12/20/2022] Open
Abstract
Ageing causes loss of function in tissues and organs, is accompanied by a chronic inflammatory process and affects life- and healthspan. Calorie restriction (CR) is a non-genetic intervention that prevents age-associated diseases and extends longevity in most of the animal models studied so far. CR produces a pleiotropic effect and improves multiple metabolic pathways, generating benefits to the whole organism. Among the effects of CR, modulation of mitochondrial activity and a decrease in oxidative damage are two of the hallmarks. Oxidative damage is reduced by the induction of endogenous antioxidant systems and modulation of the peroxidability index in cell membranes. Mitochondrial activity changes are regulated by inhibition of IGF-1 and Target of Rapamycin (TOR)-dependent activities and activation of AMP-dependent kinase (AMPK) and the sirtuin family of proteins. The activity of PGC-1α and FoxO is regulated by these systems and is involved in mitochondria biogenesis, oxidative metabolism activity and mitochondrial turnover. The use of mimetics and the regulation of common factors have demonstrated that these molecular pathways are essential to explain the effect of CR in the organism. Finally, the anti-inflammatory effect of CR is an interesting emerging factor to be taken into consideration. In the present revision we focus on the general effect of CR and other mimetics in longevity, focusing especially on the cardiovascular system and skeletal muscle.
Collapse
Affiliation(s)
- Guillermo López-Lluch
- Universidad Pablo de Olavide, Centro Andaluz de Biología del Desarrollo, CABD-CSIC, CIBERER, Instituto de Salud Carlos III, Carretera de Utrera km. 1, 41013, Sevilla, Spain
| | - Plácido Navas
- Universidad Pablo de Olavide, Centro Andaluz de Biología del Desarrollo, CABD-CSIC, CIBERER, Instituto de Salud Carlos III, Carretera de Utrera km. 1, 41013, Sevilla, Spain
| |
Collapse
|
138
|
Conover CA, Bale LK, Nair KS. Comparative gene expression and phenotype analyses of skeletal muscle from aged wild-type and PAPP-A-deficient mice. Exp Gerontol 2016; 80:36-42. [PMID: 27086066 DOI: 10.1016/j.exger.2016.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 04/05/2016] [Accepted: 04/06/2016] [Indexed: 12/17/2022]
Abstract
Mice deficient in pregnancy-associated plasma protein-A (PAPP-A) have extended lifespan associated with decreased incidence and severity of degenerative diseases of age, such as cardiomyopathy and nephropathy. In this study, the effect of PAPP-A deficiency on aging skeletal muscle was investigated. Whole-genome expression profiling was performed on soleus muscles from 18-month-old wild-type (WT) and PAPP-A knock-out (KO) mice of the same sex and from the same litter ('womb-mates') to identify potential mechanisms of skeletal muscle aging and its retardation in PAPP-A deficiency. Top genes regulated in PAPP-A KO compared to WT muscle were associated with increased muscle function, increased metabolism, in particular lipid metabolism, and decreased stress. Fiber cross-sectional area was significantly increased in solei from PAPP-A KO mice. In vitro contractility experiments indicated increased specific force and decreased fatigue in solei from PAPP-A KO mice. Intrinsic mitochondrial oxidative capacity was significantly increased in skeletal muscle of aged PAPP-A KO compared to WT mice. Moreover, 18-month-old PAPP-A KO mice exhibited significantly enhanced endurance running on a treadmill. Thus, PAPP-A deficiency in mice is associated with indices of healthy skeletal muscle function with age.
Collapse
Affiliation(s)
- Cheryl A Conover
- Division of Endocrinology, Metabolism, and Nutrition, Endocrine Research Unit, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, United States.
| | - Laurie K Bale
- Division of Endocrinology, Metabolism, and Nutrition, Endocrine Research Unit, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, United States.
| | - K Sreekumaran Nair
- Division of Endocrinology, Metabolism, and Nutrition, Endocrine Research Unit, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, United States.
| |
Collapse
|
139
|
The resveratrol-enriched rice DJ526 boosts motor coordination and physical strength. Sci Rep 2016; 6:23958. [PMID: 27044601 PMCID: PMC4820726 DOI: 10.1038/srep23958] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 03/16/2016] [Indexed: 01/02/2023] Open
Abstract
The main objective of current genetic modifications in crops is to boost agricultural production or to develop GM crops with an improved nutrient profile by introducing a new trait to the plants. A GM crop surpassing the ability of the introduced genetic characteristics has not been developed yet. Here, we show that the resveratrol-enriched rice DJ526, a GM crop, has unexpectedly high beneficial health effects surpassing the introduced genetic characteristic of resveratrol synthetic ability. The synergistic effect of its innate and transgenic properties not only ameliorates age-related deterioration but also boosts motor coordination and physical strength during the aging process. The gene expression profiling analyses by DNA chip showed that the gene expression pattern of mice fed resveratrol-enriched rice DJ526 was very different from mice fed either resveratrol or Dongjin rice alone, respectively, modifying expression of genes related to aging regulation, cell differentiation, extracellular matrix, neurogenesis, or secretion.
Collapse
|
140
|
Individual variability in human blood metabolites identifies age-related differences. Proc Natl Acad Sci U S A 2016; 113:4252-9. [PMID: 27036001 DOI: 10.1073/pnas.1603023113] [Citation(s) in RCA: 264] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Metabolites present in human blood document individual physiological states influenced by genetic, epigenetic, and lifestyle factors. Using high-resolution liquid chromatography-mass spectrometry (LC-MS), we performed nontargeted, quantitative metabolomics analysis in blood of 15 young (29 ± 4 y of age) and 15 elderly (81 ± 7 y of age) individuals. Coefficients of variation (CV = SD/mean) were obtained for 126 blood metabolites of all 30 donors. Fifty-five RBC-enriched metabolites, for which metabolomics studies have been scarce, are highlighted here. We found 14 blood compounds that show remarkable age-related increases or decreases; they include 1,5-anhydroglucitol, dimethyl-guanosine, acetyl-carnosine, carnosine, ophthalmic acid, UDP-acetyl-glucosamine,N-acetyl-arginine,N6-acetyl-lysine, pantothenate, citrulline, leucine, isoleucine, NAD(+), and NADP(+) Six of them are RBC-enriched, suggesting that RBC metabolomics is highly valuable for human aging research. Age differences are partly explained by a decrease in antioxidant production or increasing inefficiency of urea metabolism among the elderly. Pearson's coefficients demonstrated that some age-related compounds are correlated, suggesting that aging affects them concomitantly. Although our CV values are mostly consistent with those CVs previously published, we here report previously unidentified CVs of 51 blood compounds. Compounds having moderate to high CV values (0.4-2.5) are often modified. Compounds having low CV values, such as ATP and glutathione, may be related to various diseases because their concentrations are strictly controlled, and changes in them would compromise health. Thus, human blood is a rich source of information about individual metabolic differences.
Collapse
|
141
|
Hou L, Wang D, Chen D, Liu Y, Zhang Y, Cheng H, Xu C, Sun N, McDermott J, Mair WB, Han JDJ. A Systems Approach to Reverse Engineer Lifespan Extension by Dietary Restriction. Cell Metab 2016; 23:529-40. [PMID: 26959186 PMCID: PMC5110149 DOI: 10.1016/j.cmet.2016.02.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 12/14/2015] [Accepted: 02/03/2016] [Indexed: 12/16/2022]
Abstract
Dietary restriction (DR) is the most powerful natural means to extend lifespan. Although several genes can mediate responses to alternate DR regimens, no single genetic intervention has recapitulated the full effects of DR, and no unified system is known for different DR regimens. Here we obtain temporally resolved transcriptomes during calorie restriction and intermittent fasting in Caenorhabditis elegans and find that early and late responses involve metabolism and cell cycle/DNA damage, respectively. We uncover three network modules of DR regulators by their target specificity. By genetic manipulations of nodes representing discrete modules, we induce transcriptomes that progressively resemble DR as multiple nodes are perturbed. Targeting all three nodes simultaneously results in extremely long-lived animals that are refractory to DR. These results and dynamic simulations demonstrate that extensive feedback controls among regulators may be leveraged to drive the regulatory circuitry to a younger steady state, recapitulating the full effect of DR.
Collapse
Affiliation(s)
- Lei Hou
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | - Dan Wang
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China; Graduate University of Chinese Academy of Sciences, Beijing 100049, China
| | - Di Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, Jiangsu 210061, China
| | - Yi Liu
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China; Beijing Key Lab of Traffic Data Analysis and Mining, School of Computer and Information Technology, Beijing Jiaotong University, Beijing 100044, China
| | - Yue Zhang
- Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Hao Cheng
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China; Graduate University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chi Xu
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China; Graduate University of Chinese Academy of Sciences, Beijing 100049, China
| | - Na Sun
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | - Joseph McDermott
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | - William B Mair
- Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Jing-Dong J Han
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China.
| |
Collapse
|
142
|
Dal Monego S, Colitti M, Pallavicini A, D’Andrea M, Pilla F, Graziosi G, Stefanon B. Evaluation of gene expression profiles of pig skeletal muscle in response to energy content of the diets using human microarrays. ITALIAN JOURNAL OF ANIMAL SCIENCE 2016. [DOI: 10.4081/ijas.2007.45] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
143
|
Age-dependent defects of alpha-synuclein oligomer uptake in microglia and monocytes. Acta Neuropathol 2016; 131:379-91. [PMID: 26576561 DOI: 10.1007/s00401-015-1504-2] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 10/27/2015] [Accepted: 10/28/2015] [Indexed: 12/25/2022]
Abstract
Extracellular alpha-synuclein (αsyn) oligomers, associated to exosomes or free, play an important role in the pathogenesis of Parkinson's disease (PD). Increasing evidence suggests that these extracellular moieties activate microglia leading to enhanced neuronal damage. Despite extensive efforts on studying neuroinflammation in PD, little is known about the impact of age on microglial activation and phagocytosis, especially of extracellular αsyn oligomers. Here, we show that microglia isolated from adult mice, in contrast to microglia from young mice, display phagocytosis deficits of free and exosome-associated αsyn oligomers combined with enhanced TNFα secretion. In addition, we describe a dysregulation of monocyte subpopulations with age in mice and humans. Accordingly, human monocytes from elderly donors also show reduced phagocytic activity of extracellular αsyn. These findings suggest that these age-related alterations may contribute to an increased susceptibility to pathogens or abnormally folded proteins with age in neurodegenerative diseases.
Collapse
|
144
|
Pomatto LCD, Raynes R, Davies KJA. The peroxisomal Lon protease LonP2 in aging and disease: functions and comparisons with mitochondrial Lon protease LonP1. Biol Rev Camb Philos Soc 2016; 92:739-753. [PMID: 26852705 DOI: 10.1111/brv.12253] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 12/02/2015] [Accepted: 12/23/2015] [Indexed: 01/24/2023]
Abstract
Peroxisomes are ubiquitous eukaryotic organelles with the primary role of breaking down very long- and branched-chain fatty acids for subsequent β-oxidation in the mitochondrion. Like mitochondria, peroxisomes are major sites for oxygen utilization and potential contributors to cellular oxidative stress. The accumulation of oxidatively damaged proteins, which often develop into inclusion bodies (of oxidized, aggregated, and cross-linked proteins) within both mitochondria and peroxisomes, results in loss of organelle function that may contribute to the aging process. Both organelles possess an isoform of the Lon protease that is responsible for degrading proteins damaged by oxidation. While the importance of mitochondrial Lon (LonP1) in relation to oxidative stress and aging has been established, little is known regarding the role of LonP2 and aging-related changes in the peroxisome. Recently, peroxisome dysfunction has been associated with aging-related diseases indicating that peroxisome maintenance is a critical component of 'healthy aging'. Although mitochondria and peroxisomes are both needed for fatty acid metabolism, little work has focused on understanding the relationship between these two organelles including how age-dependent changes in one organelle may be detrimental for the other. Herein, we summarize findings that establish proteolytic degradation of damaged proteins by the Lon protease as a vital mechanism to maintain protein homeostasis within the peroxisome. Due to the metabolic coordination between peroxisomes and mitochondria, understanding the role of Lon in the aging peroxisome may help to elucidate cellular causes for both peroxisome and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Laura C D Pomatto
- Ethel Percy Andrus Gerontology Center of the Davis School of Gerontology and Division of Molecular & Computational Biology, Department of Biological Sciences of the College of Letters, Arts & Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA, 90089-0191, U.S.A
| | - Rachel Raynes
- Ethel Percy Andrus Gerontology Center of the Davis School of Gerontology and Division of Molecular & Computational Biology, Department of Biological Sciences of the College of Letters, Arts & Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA, 90089-0191, U.S.A
| | - Kelvin J A Davies
- Ethel Percy Andrus Gerontology Center of the Davis School of Gerontology and Division of Molecular & Computational Biology, Department of Biological Sciences of the College of Letters, Arts & Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA, 90089-0191, U.S.A
| |
Collapse
|
145
|
Papaevgeniou N, Chondrogianni N. UPS Activation in the Battle Against Aging and Aggregation-Related Diseases: An Extended Review. Methods Mol Biol 2016; 1449:1-70. [PMID: 27613027 DOI: 10.1007/978-1-4939-3756-1_1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Aging is a biological process accompanied by gradual increase of damage in all cellular macromolecules, i.e., nucleic acids, lipids, and proteins. When the proteostasis network (chaperones and proteolytic systems) cannot reverse the damage load due to its excess as compared to cellular repair/regeneration capacity, failure of homeostasis is established. This failure is a major hallmark of aging and/or aggregation-related diseases. Dysfunction of the major cellular proteolytic machineries, namely the proteasome and the lysosome, has been reported during the progression of aging and aggregation-prone diseases. Therefore, activation of these pathways is considered as a possible preventive or therapeutic approach against the progression of these processes. This chapter focuses on UPS activation studies in cellular and organismal models and the effects of such activation on aging, longevity and disease prevention or reversal.
Collapse
Affiliation(s)
- Nikoletta Papaevgeniou
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 48 Vassileos Constantinou Ave., Athens, 11635, Greece
| | - Niki Chondrogianni
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 48 Vassileos Constantinou Ave., Athens, 11635, Greece.
| |
Collapse
|
146
|
Pinti M, Gibellini L, Liu Y, Xu S, Lu B, Cossarizza A. Mitochondrial Lon protease at the crossroads of oxidative stress, ageing and cancer. Cell Mol Life Sci 2015; 72:4807-24. [PMID: 26363553 PMCID: PMC11113732 DOI: 10.1007/s00018-015-2039-3] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 09/01/2015] [Accepted: 09/07/2015] [Indexed: 11/26/2022]
Abstract
Lon protease is a nuclear DNA-encoded mitochondrial enzyme highly conserved throughout evolution, involved in the degradation of damaged and oxidized proteins of the mitochondrial matrix, in the correct folding of proteins imported in mitochondria, and in the maintenance of mitochondrial DNA. Lon expression is induced by various stimuli, including hypoxia and reactive oxygen species, and provides protection against cell stress. Lon down-regulation is associated with ageing and with cell senescence, while up-regulation is observed in tumour cells, and is correlated with a more aggressive phenotype of cancer. Lon up-regulation contributes to metabolic reprogramming observed in cancer, favours the switch from a respiratory to a glycolytic metabolism, helping cancer cell survival in the tumour microenvironment, and contributes to epithelial to mesenchymal transition. Silencing of Lon, or pharmacological inhibition of its activity, causes cell death in various cancer cells. Thus, Lon can be included in the growing class of proteins that are not responsible for oncogenic transformation, but that are essential for survival and proliferation of cancer cells, and that can be considered as a new target for development of anticancer drugs.
Collapse
Affiliation(s)
- Marcello Pinti
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Campi, 287, 41125, Modena, Italy.
| | - Lara Gibellini
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Yongzhang Liu
- School of Life Sciences, Institute of Biophysics, Attardi Institute of Mitochondrial Biomedicine and Zhejiang Provincial Key Laboratory of Medical Genetics, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Shan Xu
- School of Life Sciences, Institute of Biophysics, Attardi Institute of Mitochondrial Biomedicine and Zhejiang Provincial Key Laboratory of Medical Genetics, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Bin Lu
- School of Life Sciences, Institute of Biophysics, Attardi Institute of Mitochondrial Biomedicine and Zhejiang Provincial Key Laboratory of Medical Genetics, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Andrea Cossarizza
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
147
|
Hopp L, Willscher E, Löffler-Wirth H, Binder H. Function Shapes Content: DNA-Methylation Marker Genes and their Impact for Molecular Mechanisms of Glioma. ACTA ACUST UNITED AC 2015. [DOI: 10.6000/1929-2279.2015.04.04.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
|
148
|
Lam YY, Ghosh S, Civitarese AE, Ravussin E. Six-month Calorie Restriction in Overweight Individuals Elicits Transcriptomic Response in Subcutaneous Adipose Tissue That is Distinct From Effects of Energy Deficit. J Gerontol A Biol Sci Med Sci 2015; 71:1258-65. [PMID: 26486851 DOI: 10.1093/gerona/glv194] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 10/02/2015] [Indexed: 12/11/2022] Open
Abstract
Calorie restriction confers health benefits distinct from energy deficit by exercise. We characterized the adipose-transcriptome to investigate the molecular basis of the differential phenotypic responses. Abdominal subcutaneous fat was collected from 24 overweight participants randomized in three groups (N = 8/group): weight maintenance (control), 25% energy deficit by calorie restriction alone (CR), and 25% energy deficit by calorie restriction with structured exercise (CREX). Within each group, gene expression was compared between 6 months and baseline with cutoffs at nominal p ≤ .01 and absolute fold-change ≥ 1.5. Gene-set enrichment analysis (false discovery rate < 5%) was used to identify significantly regulated biological pathways. CR and CREX elicited similar overall clinical response to energy deficit and a comparable reduction in gene transcription specific to oxidative phosphorylation and proteasome function. CR vastly outweighed CREX in the number of differentially regulated genes (88 vs 39) and pathways (28 vs 6). CR specifically downregulated the chemokine signaling-related pathways. Among the CR-regulated genes, 27 functioned as transcription/translation regulators (eg, mRNA processing or transcription/translation initiation), whereas CREX regulated only one gene in this category. Our data suggest that CR has a broader effect on the transcriptome compared with CREX which may mediate its specific impact on delaying primary aging.
Collapse
Affiliation(s)
- Yan Y Lam
- Pennington Biomedical Research Center, Baton Rouge, Louisiana. Boden Institute of Obesity, Nutrition, Exercise & Eating Disorders, University of Sydney, Sydney, New South Wales, Australia.
| | - Sujoy Ghosh
- Pennington Biomedical Research Center, Baton Rouge, Louisiana. Centre for Computational Biology & Program in Cardiovascular and Metabolic Disorders, Duke-NUS Graduate Medical School, Singapore
| | - Anthony E Civitarese
- Pennington Biomedical Research Center, Baton Rouge, Louisiana. Novo Nordisk Research Center, Seattle, Washington
| | - Eric Ravussin
- Pennington Biomedical Research Center, Baton Rouge, Louisiana
| |
Collapse
|
149
|
Ebert SM, Dyle MC, Bullard SA, Dierdorff JM, Murry DJ, Fox DK, Bongers KS, Lira VA, Meyerholz DK, Talley JJ, Adams CM. Identification and Small Molecule Inhibition of an Activating Transcription Factor 4 (ATF4)-dependent Pathway to Age-related Skeletal Muscle Weakness and Atrophy. J Biol Chem 2015; 290:25497-511. [PMID: 26338703 PMCID: PMC4646196 DOI: 10.1074/jbc.m115.681445] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 08/30/2015] [Indexed: 12/17/2022] Open
Abstract
Aging reduces skeletal muscle mass and strength, but the underlying molecular mechanisms remain elusive. Here, we used mouse models to investigate molecular mechanisms of age-related skeletal muscle weakness and atrophy as well as new potential interventions for these conditions. We identified two small molecules that significantly reduce age-related deficits in skeletal muscle strength, quality, and mass: ursolic acid (a pentacyclic triterpenoid found in apples) and tomatidine (a steroidal alkaloid derived from green tomatoes). Because small molecule inhibitors can sometimes provide mechanistic insight into disease processes, we used ursolic acid and tomatidine to investigate the pathogenesis of age-related muscle weakness and atrophy. We found that ursolic acid and tomatidine generate hundreds of small positive and negative changes in mRNA levels in aged skeletal muscle, and the mRNA expression signatures of the two compounds are remarkably similar. Interestingly, a subset of the mRNAs repressed by ursolic acid and tomatidine in aged muscle are positively regulated by activating transcription factor 4 (ATF4). Based on this finding, we investigated ATF4 as a potential mediator of age-related muscle weakness and atrophy. We found that a targeted reduction in skeletal muscle ATF4 expression reduces age-related deficits in skeletal muscle strength, quality, and mass, similar to ursolic acid and tomatidine. These results elucidate ATF4 as a critical mediator of age-related muscle weakness and atrophy. In addition, these results identify ursolic acid and tomatidine as potential agents and/or lead compounds for reducing ATF4 activity, weakness, and atrophy in aged skeletal muscle.
Collapse
Affiliation(s)
- Scott M Ebert
- From the Departments of Internal Medicine, the Fraternal Order of Eagles Diabetes Research Center, and the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246, and Emmyon, Inc., Coralville, Iowa 52241
| | - Michael C Dyle
- From the Departments of Internal Medicine, the Fraternal Order of Eagles Diabetes Research Center, and the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246, and Molecular Physiology and Biophysics
| | - Steven A Bullard
- From the Departments of Internal Medicine, the Fraternal Order of Eagles Diabetes Research Center, and the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246, and
| | - Jason M Dierdorff
- From the Departments of Internal Medicine, the Fraternal Order of Eagles Diabetes Research Center, and the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246, and
| | - Daryl J Murry
- the College of Pharmacy, The University of Iowa, Iowa City, Iowa 52242
| | - Daniel K Fox
- From the Departments of Internal Medicine, the Fraternal Order of Eagles Diabetes Research Center, and the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246, and Molecular Physiology and Biophysics
| | - Kale S Bongers
- From the Departments of Internal Medicine, the Fraternal Order of Eagles Diabetes Research Center, and the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246, and Molecular Physiology and Biophysics
| | - Vitor A Lira
- the Fraternal Order of Eagles Diabetes Research Center, and Health and Human Physiology, and
| | | | | | - Christopher M Adams
- From the Departments of Internal Medicine, the Fraternal Order of Eagles Diabetes Research Center, and the Iowa City Veterans Affairs Medical Center, Iowa City, Iowa 52246, and Emmyon, Inc., Coralville, Iowa 52241 Molecular Physiology and Biophysics,
| |
Collapse
|
150
|
Hagiwara K, Gailhouste L, Yasukawa K, Kosaka N, Ochiya T. A robust screening method for dietary agents that activate tumour-suppressor microRNAs. Sci Rep 2015; 5:14697. [PMID: 26423775 PMCID: PMC4589759 DOI: 10.1038/srep14697] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 09/07/2015] [Indexed: 12/17/2022] Open
Abstract
Certain dietary agents, such as natural products, have been reported to show anti-cancer effects. However, the underlying mechanisms of these substances in human cancer remain unclear. We recently found that resveratrol exerts an anti-cancer effect by upregulating tumour-suppressor microRNAs (miRNAs). In the current study, we aimed to identify new dietary products that have the ability to activate tumour-suppressor miRNAs and that therefore may serve as novel tools for the prevention and treatment of human cancers. We describe the generation and use of an original screening system based on a luciferase-based reporter vector for monitoring miR-200c tumour-suppressor activity. By screening a library containing 139 natural substances, three natural compounds — enoxolone, magnolol and palmatine chloride — were identified as being capable of inducing miR-200c expression in breast cancer cells at 10 μM. Moreover, these molecules suppressed the invasiveness of breast cancer cells in vitro. Next, we identified a molecular pathway by which the increased expression of miR-200c induced by natural substances led to ZEB1 inhibition and E-cadherin induction. These results indicate that our method is a valuable tool for a fast identification of natural molecules that exhibit tumour-suppressor activity in human cancer through miRNA activation.
Collapse
Affiliation(s)
- Keitaro Hagiwara
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.,Department of Biological Sciences, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Luc Gailhouste
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Ken Yasukawa
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.,Integrative Bioscience and Biomedical Engineering, Graduate School of Science and Engineering, Waseda University, 2-2 Wakamatsu, Shinjuku, Tokyo 162-8480, Japan
| | - Nobuyoshi Kosaka
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Takahiro Ochiya
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| |
Collapse
|