501
|
Kaeberlein M, Creevy KE, Promislow DEL. The dog aging project: translational geroscience in companion animals. Mamm Genome 2016; 27:279-88. [PMID: 27143112 DOI: 10.1007/s00335-016-9638-7] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 04/15/2016] [Indexed: 12/16/2022]
Abstract
Studies of the basic biology of aging have identified several genetic and pharmacological interventions that appear to modulate the rate of aging in laboratory model organisms, but a barrier to further progress has been the challenge of moving beyond these laboratory discoveries to impact health and quality of life for people. The domestic dog, Canis familiaris, offers a unique opportunity for surmounting this barrier in the near future. In particular, companion dogs share our environment and play an important role in improving the quality of life for millions of people. Here, we present a rationale for increasing the role of companion dogs as an animal model for both basic and clinical geroscience and describe complementary approaches and ongoing projects aimed at achieving this goal.
Collapse
Affiliation(s)
- Matt Kaeberlein
- Department of Pathology, University of Washington, Seattle, WA, USA.
| | - Kate E Creevy
- Department of Small Animal Medicine and Surgery, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | | |
Collapse
|
502
|
Abstract
For decades, researchers in the biology of aging have focused on defining mechanisms that modulate aging by primarily studying a single metric, sometimes described as the "gold standard" lifespan. Increasingly, geroscience research is turning towards defining functional domains of aging such as the cardiovascular system, skeletal integrity, and metabolic health as being a more direct route to understand why tissues decline in function with age. Each model used in aging research has strengths and weaknesses, yet we know surprisingly little about how critical tissues decline in health with increasing age. Here I discuss popular model systems used in geroscience research and their utility as possible tools in preclinical studies in aging.
Collapse
Affiliation(s)
- Simon Melov
- Buck Institute for Research on Aging, Novato, CA, USA
| |
Collapse
|
503
|
Abstract
Aging is characterized by the progressive accumulation of degenerative changes, culminating in impaired function and increased probability of death. It is the major risk factor for many human pathologies - including cancer, type 2 diabetes, and cardiovascular and neurodegenerative diseases - and consequently exerts an enormous social and economic toll. The major goal of aging research is to develop interventions that can delay the onset of multiple age-related diseases and prolong healthy lifespan (healthspan). The observation that enhanced longevity and health can be achieved in model organisms by dietary restriction or simple genetic manipulations has prompted the hunt for chemical compounds that can increase lifespan. Most of the pathways that modulate the rate of aging in mammals have homologs in yeast, flies, and worms, suggesting that initial screening to identify such pharmacological interventions may be possible using invertebrate models. In recent years, several compounds have been identified that can extend lifespan in invertebrates, and even in rodents. Here, we summarize the strategies employed, and the progress made, in identifying compounds capable of extending lifespan in organisms ranging from invertebrates to mice and discuss the formidable challenges in translating this work to human therapies.
Collapse
Affiliation(s)
- Surinder Kumar
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - David B Lombard
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA; Institute of Gerontology, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
504
|
Treuting PM, Snyder JM, Ikeno Y, Schofield PN, Ward JM, Sundberg JP. The Vital Role of Pathology in Improving Reproducibility and Translational Relevance of Aging Studies in Rodents. Vet Pathol 2016; 53:244-9. [PMID: 26792843 PMCID: PMC4835687 DOI: 10.1177/0300985815620629] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Pathology is a discipline of medicine that adds great benefit to aging studies of rodents by integrating in vivo, biochemical, and molecular data. It is not possible to diagnose systemic illness, comorbidities, and proximate causes of death in aging studies without the morphologic context provided by histopathology. To date, many rodent aging studies do not utilize end points supported by systematic necropsy and histopathology, which leaves studies incomplete, contradictory, and difficult to interpret. As in traditional toxicity studies, if the effect of a drug, dietary treatment, or altered gene expression on aging is to be studied, systematic pathology analysis must be included to determine the causes of age-related illness, moribundity, and death. In this Commentary, the authors discuss the factors that should be considered in the design of aging studies in mice, with the inclusion of robust pathology practices modified after those developed by toxicologic and discovery research pathologists. Investigators in the field of aging must consider the use of histopathology in their rodent aging studies in this era of integrative and preclinical geriatric science (geroscience).
Collapse
Affiliation(s)
- P M Treuting
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, WA, USA
| | - J M Snyder
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, WA, USA
| | - Y Ikeno
- Barshop Institute and Department of Pathology, University of Texas Health Science Center at San Antonio; Research Service and Geriatric Research and Education Clinical Center, Audie L. Murphy VA Hospital, South Texas Veterans Health Care System, San Antonio, TX, USA
| | - P N Schofield
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK The Jackson Laboratory, Bar Harbor, ME, USA
| | - J M Ward
- Global VetPathology, Montgomery Village, MD, USA
| | | |
Collapse
|
505
|
The role of autophagy in axonal degeneration of the optic nerve. Exp Eye Res 2016; 144:81-9. [DOI: 10.1016/j.exer.2015.08.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 08/06/2015] [Accepted: 08/18/2015] [Indexed: 11/21/2022]
|
506
|
Kirkland JL. Translating the Science of Aging into Therapeutic Interventions. Cold Spring Harb Perspect Med 2016; 6:a025908. [PMID: 26931808 DOI: 10.1101/cshperspect.a025908] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Life and health span have been extended in experimental animals using drugs that are potentially translatable into humans. Considerable effort is needed beyond the usual steps in drug development to devise the models, and realistic preclinical and clinical trial strategies are required to advance these agents into clinical application. It will be important to focus on subjects who already have symptoms or are at imminent risk of developing disorders related to fundamental aging processes, to use short-term, clinically relevant outcomes, as opposed to long-term outcomes, such as health span or life span, and to validate endpoint measures so they are acceptable to regulatory agencies. Funding is a roadblock, as is shortage of investigators with combined expertise in the basic biology of aging, clinical geriatrics, and investigational new drug clinical trials. Strategies for developing a path from the bench to the bedside are reviewed for interventions that target fundamental aging mechanisms.
Collapse
Affiliation(s)
- James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota 55905
| |
Collapse
|
507
|
Billings H, Crowe SM, Palmer CS. Does immunometabolism provide new targets to treat HIV-mediated inflammatory diseases? Future Virol 2016. [DOI: 10.2217/fvl.16.4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Hugh Billings
- Centre for Biomedical Research, Burnet Institute, Melbourne 3001, Australia
| | - Suzanne M Crowe
- Centre for Biomedical Research, Burnet Institute, Melbourne 3001, Australia
- Department of Infectious Diseases, Monash University, Melbourne 3800, Australia
- Infectious Diseases Department, The Alfred Hospital, Melbourne 3004, Australia
| | - Clovis S Palmer
- Centre for Biomedical Research, Burnet Institute, Melbourne 3001, Australia
- Department of Infectious Diseases, Monash University, Melbourne 3800, Australia
| |
Collapse
|
508
|
McElhaney JE, Kuchel GA, Zhou X, Swain SL, Haynes L. T-Cell Immunity to Influenza in Older Adults: A Pathophysiological Framework for Development of More Effective Vaccines. Front Immunol 2016; 7:41. [PMID: 26941738 PMCID: PMC4766518 DOI: 10.3389/fimmu.2016.00041] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 01/28/2016] [Indexed: 02/03/2023] Open
Abstract
One of the most profound public health consequences of immune senescence is reflected in an increased susceptibility to influenza and other acute respiratory illnesses, as well as a loss of influenza vaccine effectiveness in older people. Common medical conditions and mental and psychosocial health issues as well as degree of frailty and functional dependence accelerate changes associated with immune senescence. All contribute to the increased risk for complications of influenza infection, including pneumonias, heart diseases, and strokes that lead to hospitalization, disability, and death in the over 65 population. Changes in mucosal barrier mechanisms and both innate and adaptive immune functions converge in the reduced response to influenza infection, and lead to a loss of antibody-mediated protection against influenza with age. The interactions of immune senescence and reduced adaptive immune responses, persistent cytomegalovirus infection, inflammaging (chronic elevation of inflammatory cytokines), and dysregulated cytokine production, pose major challenges to the development of vaccines designed to improve T-cell-mediated immunity. In older adults, the goal of vaccination is more realistically targeted to providing clinical protection against disease rather than to inducing sterilizing immunity to infection. Standard assays of antibody titers correlate with protection against influenza illness but do not detect important changes in cellular immune mechanisms that correlate with vaccine-mediated protection against influenza in older people. This article will discuss: (i) the burden of influenza in older adults and how this relates to changes in T-cell function, (ii) age-related changes in different T-cell subsets and immunologic targets for improved influenza vaccine efficacy in older, and (iii) the development of correlates of clinical protection against influenza disease to expedite the process of new vaccine development for the 65 and older population. Ultimately, these efforts will address the public health need for improved protection against influenza in older adults and “vaccine preventable disability.”
Collapse
Affiliation(s)
- Janet E McElhaney
- Health Sciences North Research Institute, Sudbury, ON, Canada; UConn Center on Aging, University of Connecticut School of Medicine, Farmington, CT, USA
| | - George A Kuchel
- UConn Center on Aging, University of Connecticut School of Medicine , Farmington, CT , USA
| | - Xin Zhou
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, CT, USA; Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Susan L Swain
- Department of Pathology, University of Massachusetts Medical School , North Worcester, MA , USA
| | - Laura Haynes
- UConn Center on Aging, University of Connecticut School of Medicine, Farmington, CT, USA; Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, USA
| |
Collapse
|
509
|
Beyond adjuvants: Antagonizing inflammation to enhance vaccine immunity. Vaccine 2016; 33 Suppl 2:B55-9. [PMID: 26022570 DOI: 10.1016/j.vaccine.2015.03.058] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 03/19/2015] [Indexed: 11/20/2022]
Abstract
Since the development of the first vaccine over 200 years ago, vaccines have saved millions of lives and have become the most cost-effective modern medical intervention. However, over 70 years ago, Freund recognized that the effectiveness of the vaccine-induced immune responses could be vastly improved via the co-delivery of inflammation-induced agents, giving birth to the adjuvant field. Since the first description of adjuvants, revolutionary discoveries, including the discovery of dendritic cells and pattern recognition receptors, that drive remarkably different biological profiles, have opened the landscape of opportunities for the development of novel adjuvants able to trigger a remarkably diverse inflammatory profiles, thereby qualitatively and quantitatively skewing adaptive immunity in a tailored manner against target pathogens. However, mounting data point to a critical role for pre-existing inflammation as a predictor of vaccine responsiveness. Thus, in this review we will discuss novel opportunities by which pre-existing inflammation may be modulated, skewed, or tuned via next-generation vaccine approaches to enhanced vaccine-induced immunity in the elderly, immunocompromised, or subjects with chronic diseases.
Collapse
|
510
|
Kaeberlein M, Rabinovitch PS, Martin GM. Healthy aging: The ultimate preventative medicine. Science 2016; 350:1191-3. [PMID: 26785476 DOI: 10.1126/science.aad3267] [Citation(s) in RCA: 221] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Age is the greatest risk factor for nearly every major cause of mortality in developed nations. Despite this, most biomedical research focuses on individual disease processes without much consideration for the relationships between aging and disease. Recent discoveries in the field of geroscience, which aims to explain biological mechanisms of aging, have provided insights into molecular processes that underlie biological aging and, perhaps more importantly, potential interventions to delay aging and promote healthy longevity. Here we describe some of these advances, along with efforts to move geroscience from the bench to the clinic. We also propose that greater emphasis should be placed on research into basic aging processes, because interventions that slow aging will have a greater effect on quality of life compared with disease-specific approaches.
Collapse
Affiliation(s)
- Matt Kaeberlein
- Department of Pathology, University of Washington, Seattle, WA 98195, USA.
| | | | - George M Martin
- Department of Pathology, University of Washington, Seattle, WA 98195, USA. Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
511
|
Arriola Apelo SI, Neuman JC, Baar EL, Syed FA, Cummings NE, Brar HK, Pumper CP, Kimple ME, Lamming DW. Alternative rapamycin treatment regimens mitigate the impact of rapamycin on glucose homeostasis and the immune system. Aging Cell 2016; 15:28-38. [PMID: 26463117 PMCID: PMC4717280 DOI: 10.1111/acel.12405] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2015] [Indexed: 12/23/2022] Open
Abstract
Inhibition of the mechanistic target of rapamycin (mTOR) signaling pathway by the FDA-approved drug rapamycin has been shown to promote lifespan and delay age-related diseases in model organisms including mice. Unfortunately, rapamycin has potentially serious side effects in humans, including glucose intolerance and immunosuppression, which may preclude the long-term prophylactic use of rapamycin as a therapy for age-related diseases. While the beneficial effects of rapamycin are largely mediated by the inhibition of mTOR complex 1 (mTORC1), which is acutely sensitive to rapamycin, many of the negative side effects are mediated by the inhibition of a second mTOR-containing complex, mTORC2, which is much less sensitive to rapamycin. We hypothesized that different rapamycin dosing schedules or the use of FDA-approved rapamycin analogs with different pharmacokinetics might expand the therapeutic window of rapamycin by more specifically targeting mTORC1. Here, we identified an intermittent rapamycin dosing schedule with minimal effects on glucose tolerance, and we find that this schedule has a reduced impact on pyruvate tolerance, fasting glucose and insulin levels, beta cell function, and the immune system compared to daily rapamycin treatment. Further, we find that the FDA-approved rapamycin analogs everolimus and temsirolimus efficiently inhibit mTORC1 while having a reduced impact on glucose and pyruvate tolerance. Our results suggest that many of the negative side effects of rapamycin treatment can be mitigated through intermittent dosing or the use of rapamycin analogs.
Collapse
Affiliation(s)
- Sebastian I. Arriola Apelo
- Department of Medicine University of Wisconsin‐Madison Madison WI USA
- William S. Middleton Memorial Veterans Hospital Madison WI USA
| | - Joshua C. Neuman
- William S. Middleton Memorial Veterans Hospital Madison WI USA
- Interdisciplinary Graduate Program in Nutritional Sciences University of Wisconsin‐Madison Madison WI USA
| | - Emma L. Baar
- Department of Medicine University of Wisconsin‐Madison Madison WI USA
- William S. Middleton Memorial Veterans Hospital Madison WI USA
| | - Faizan A. Syed
- Department of Medicine University of Wisconsin‐Madison Madison WI USA
- William S. Middleton Memorial Veterans Hospital Madison WI USA
| | - 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
| | - Harpreet K. Brar
- Department of Medicine University of Wisconsin‐Madison Madison WI USA
- William S. Middleton Memorial Veterans Hospital Madison WI USA
| | - Cassidy P. Pumper
- Department of Medicine University of Wisconsin‐Madison Madison WI USA
- William S. Middleton Memorial Veterans Hospital Madison WI USA
| | - Michelle E. Kimple
- Department of Medicine University of Wisconsin‐Madison Madison WI USA
- William S. Middleton Memorial Veterans Hospital Madison WI USA
- Interdisciplinary Graduate Program in Nutritional Sciences University of Wisconsin‐Madison 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
- Interdisciplinary Graduate Program in Nutritional Sciences University of Wisconsin‐Madison Madison WI USA
- Endocrinology and Reproductive Physiology Graduate Training Program University of Wisconsin‐Madison Madison WI USA
- University of Wisconsin Carbone Cancer Center Madison WI USA
| |
Collapse
|
512
|
Walters HE, Deneka-Hannemann S, Cox LS. Reversal of phenotypes of cellular senescence by pan-mTOR inhibition. Aging (Albany NY) 2016; 8:231-44. [PMID: 26851731 PMCID: PMC4789579 DOI: 10.18632/aging.100872] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cellular senescence, a state of essentially irreversible proliferation arrest, serves as a potent tumour suppressor mechanism. However, accumulation of senescent cells with chronological age is likely to contribute to loss of tissue and organ function and organismal aging. A crucial biochemical modulator of aging is mTOR; here, we have addressed the question of whether acute mTORC inhibition in near-senescent cells can modify phenotypes of senescence. We show that acute short term treatment of human skin fibroblasts with low dose ATP mimetic pan-mTORC inhibitor AZD8055 leads to reversal of many phenotypes that develop as cells near replicative senescence, including reduction in cell size and granularity, loss of SA-β-gal staining and reacquisition of fibroblastic spindle morphology. AZD8055 treatment also induced rearrangement of the actin cytoskeleton, providing a possible mechanism of action for the observed rejuvenation. Importantly, short-term drug exposure had no detrimental effects on cell proliferation control across the life-course of the fibroblasts. Our findings suggest that combined inhibition of both mTORC1 and mTORC2 may provide a promising strategy to reverse the development of senescence-associated features in near-senescent cells.
Collapse
Affiliation(s)
- Hannah E Walters
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, United Kingdom
| | - Sylwia Deneka-Hannemann
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, United Kingdom.,Current address: Oxford BioMedica Plc, Oxford, OX4 6LT, United Kingdom
| | - Lynne S Cox
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, United Kingdom
| |
Collapse
|
513
|
Pre-vaccination inflammation and B-cell signalling predict age-related hyporesponse to hepatitis B vaccination. Nat Commun 2016; 7:10369. [PMID: 26742691 PMCID: PMC4729923 DOI: 10.1038/ncomms10369] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 12/04/2015] [Indexed: 01/10/2023] Open
Abstract
Aging is associated with hyporesponse to vaccination, whose mechanisms remain unclear. In this study hepatitis B virus (HBV)-naive older adults received three vaccines, including one against HBV. Here we show, using transcriptional and cytometric profiling of whole blood collected before vaccination, that heightened expression of genes that augment B-cell responses and higher memory B-cell frequencies correlate with stronger responses to HBV vaccine. In contrast, higher levels of inflammatory response transcripts and increased frequencies of pro-inflammatory innate cells correlate with weaker responses to this vaccine. Increased numbers of erythrocytes and the haem-induced response also correlate with poor response to the HBV vaccine. A transcriptomics-based pre-vaccination predictor of response to HBV vaccine is built and validated in distinct sets of older adults. This moderately accurate (area under the curve≈65%) but robust signature is supported by flow cytometry and cytokine profiling. This study is the first that identifies baseline predictors and mechanisms of response to the HBV vaccine. Ageing is associated with poor responses to vaccines but the underlying mechanism remains unclear. Here the authors use a systems-based approach to define molecular signatures present before vaccination that correlate with non-responsiveness to hepatitis B vaccination in healthy, elderly adults.
Collapse
|
514
|
Schreiber KH, O’Leary MN, Kennedy BK. The mTOR Pathway and Aging. HANDBOOK OF THE BIOLOGY OF AGING 2016:55-81. [DOI: 10.1016/b978-0-12-411596-5.00002-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
515
|
Frasca D, Blomberg BB. B Cell-Specific Biomarkers for Optimal Antibody Responses to Influenza Vaccination and Molecular Pathways That Reduce B Cell Function with Aging. Crit Rev Immunol 2016; 36:523-537. [PMID: 28845758 DOI: 10.1615/critrevimmunol.2017020113] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
This review highlights recent findings on the effects of aging on influenza vaccine responses, with major emphasis on T and B cells, which are significantly impaired by aging. We discuss changes in T cell production and thymic output; T cell subsets; and TCR repertoire, function, and response to latent persistent infection. We also discuss changes in B cell subsets, repertoire, and function, and how function is impaired by increased intrinsic B cell inflammation and reduced signal transduction. This review presents age-related effects on antigen-presenting cells, summarizes recent studies, including our own, aimed at the identification of biomarkers of protective vaccine responses, and provides examples of recent technical advances and insights into human vaccine responses that are helping to define the features associated with successful vaccination and that may enable a more predictive vaccinology in the future.
Collapse
Affiliation(s)
- Daniela Frasca
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33101, USA
| | - Bonnie B Blomberg
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33101, USA
| |
Collapse
|
516
|
Stenvinkel P, Kooman JP, Shiels PG. Nutrients and ageing: what can we learn about ageing interactions from animal biology? Curr Opin Clin Nutr Metab Care 2016; 19:19-25. [PMID: 26485336 DOI: 10.1097/mco.0000000000000234] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW Many prevalent clinical conditions, such as chronic kidney disease, diabetes mellitus, chronic obstructive pulmonary, and cardiovascular disease associate with features of premature ageing, such as muscle wasting, hypogonadism, osteoporosis, and arteriosclerosis. Studies on various animal models have shown that caloric restriction prolongs lifespan. Studies of animals with unusual long or short life for their body size may also contribute to better understanding of ageing processes. The aim of the present article is to review what we can learn about nutritional modulations and ageing interactions from animal biology. RECENT FINDINGS Caloric restriction is a powerful intervention that increases longevity in animals ranging from short-lived species, such as worms and flies, to primates. As long-term studies on caloric restriction are not feasible to conduct in humans, much interest has focused on the impact of caloric restriction mimetics, such as resveratrol, on ageing processes. Recent data from studies on the long-lived naked mole rat have provided important novel information on metabolic alterations and antioxidative defense mechanisms that characterize longevity. SUMMARY Better understanding of the biology of exceptionally long-lived animals will contribute to better understanding of ageing processes and novel interventions to extend lifespan also in humans.
Collapse
Affiliation(s)
- Peter Stenvinkel
- aDivision of Renal Medicine, Karolinska University Hospital at Huddinge, Karolinska Institutet Stockholm, Sweden bDivision of Nephrology, Department of Internal Medicine, University Hospital Maastricht, the Netherlands cInstitute of Cancer Sciences, Wolfson Wohl Translational Research Center, University of Glasgow, Glasgow, UK
| | | | | |
Collapse
|
517
|
Abstract
The demographics of the world's population are changing, with many adults now surviving into their 80s. With this change comes the need to protect the aging and other underserved populations not only against infectious diseases but also against cancer and other chronic conditions. New technologies derived from recent advances in the fields of immunology, structural biology, synthetic biology, and genomics have brought a revolution in the vaccine field. Among them, vaccine adjuvants have the potential to harness the immune system to provide protection against new types of diseases, improve protection in young children, and expand this protection to adults and the elderly. However, in order to do so we need also to overcome the nontechnical challenges that could limit the implementation of innovative vaccines, including controversies regarding the safety of adjuvants, increasing regulatory complexity, the inadequate methods used to assess the value of novel vaccines, and the resulting industry alienation from future investment. This Perspective summarizes the outcome of a recent multidisciplinary symposium entitled "Enhancing Vaccine Immunity and Value," held in Siena, Italy, in July 2014, that addressed two related questions: how to improve vaccine efficacy by using breakthrough technologies and how to capture the full potential of novel vaccines.
Collapse
Affiliation(s)
- Steven Black
- Center for Global Health, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | | | | |
Collapse
|
518
|
Milman S, Barzilai N. Dissecting the Mechanisms Underlying Unusually Successful Human Health Span and Life Span. Cold Spring Harb Perspect Med 2015; 6:a025098. [PMID: 26637439 DOI: 10.1101/cshperspect.a025098] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Humans age at different rates and families with exceptional survival provide the opportunity to understand why some people age slower than others. Unique features exhibited by centenarians include a family history of longevity, compression of morbidity with resultant extension of health span, and biomarkers such as low-circulating insulin-like growth factor 1 (IGF-1) and elevated high-density lipoprotein (HDL) cholesterol levels. Given the rarity of the centenarian phenotype, it has not been surprising that the use of discovery methods that relied on common population single nucleotide polymorphisms (SNPs) to unlock the genetic determinants of exceptional longevity have not yielded significant results. Conversely, gene sequencing has resulted in discoveries of functional gene variants that support several of the centenarian phenotypes. These discoveries have led to the strategic developments of drugs that may delay aging and prolong health span.
Collapse
Affiliation(s)
- Sofiya Milman
- Department of Medicine, Division of Endocrinology, Albert Einstein College of Medicine, New York, New York 10461 Institute for Aging Research, Albert Einstein College of Medicine, New York, New York 10461
| | - Nir Barzilai
- Department of Medicine, Division of Endocrinology, Albert Einstein College of Medicine, New York, New York 10461 Institute for Aging Research, Albert Einstein College of Medicine, New York, New York 10461 Department of Genetics, Albert Einstein College of Medicine, Bronx, New York 10461
| |
Collapse
|
519
|
Hurez V, Dao V, Liu A, Pandeswara S, Gelfond J, Sun L, Bergman M, Orihuela CJ, Galvan V, Padrón Á, Drerup J, Liu Y, Hasty P, Sharp ZD, Curiel TJ. Chronic mTOR inhibition in mice with rapamycin alters T, B, myeloid, and innate lymphoid cells and gut flora and prolongs life of immune-deficient mice. Aging Cell 2015; 14:945-56. [PMID: 26315673 PMCID: PMC4693453 DOI: 10.1111/acel.12380] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2015] [Indexed: 01/24/2023] Open
Abstract
The mammalian (mechanistic) target of rapamycin (mTOR) regulates critical immune processes that remain incompletely defined. Interest in mTOR inhibitor drugs is heightened by recent demonstrations that the mTOR inhibitor rapamycin extends lifespan and healthspan in mice. Rapamycin or related analogues (rapalogues) also mitigate age-related debilities including increasing antigen-specific immunity, improving vaccine responses in elderly humans, and treating cancers and autoimmunity, suggesting important new clinical applications. Nonetheless, immune toxicity concerns for long-term mTOR inhibition, particularly immunosuppression, persist. Although mTOR is pivotal to fundamental, important immune pathways, little is reported on immune effects of mTOR inhibition in lifespan or healthspan extension, or with chronic mTOR inhibitor use. We comprehensively analyzed immune effects of rapamycin as used in lifespan extension studies. Gene expression profiling found many and novel changes in genes affecting differentiation, function, homeostasis, exhaustion, cell death, and inflammation in distinct T- and B-lymphocyte and myeloid cell subpopulations. Immune functions relevant to aging and inflammation, and to cancer and infections, and innate lymphoid cell effects were validated in vitro and in vivo. Rapamycin markedly prolonged lifespan and healthspan in cancer- and infection-prone mice supporting disease mitigation as a mechanism for mTOR suppression-mediated longevity extension. It modestly altered gut metagenomes, and some metagenomic effects were linked to immune outcomes. Our data show novel mTOR inhibitor immune effects meriting further studies in relation to longevity and healthspan extension.
Collapse
Affiliation(s)
- Vincent Hurez
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Vinh Dao
- Graduate School of Biomedical Sciences, University of Texas Health Science Center, San Antonio, TX, USA
| | - Aijie Liu
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Srilakshmi Pandeswara
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Jonathan Gelfond
- Department of Epidemiology and Biostatistics, University of Texas Health Science Center, San Antonio, TX, USA
| | - Lishi Sun
- Graduate School of Biomedical Sciences, University of Texas Health Science Center, San Antonio, TX, USA
| | - Molly Bergman
- Graduate School of Biomedical Sciences, University of Texas Health Science Center, San Antonio, TX, USA
| | - Carlos J Orihuela
- Graduate School of Biomedical Sciences, University of Texas Health Science Center, San Antonio, TX, USA
| | - Veronica Galvan
- Graduate School of Biomedical Sciences, University of Texas Health Science Center, San Antonio, TX, USA
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center, San Antonio, TX, USA
- Department of Physiology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Álvaro Padrón
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Justin Drerup
- Graduate School of Biomedical Sciences, University of Texas Health Science Center, San Antonio, TX, USA
| | - Yang Liu
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
| | - Paul Hasty
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center, San Antonio, TX, USA
- Department of Molecular Medicine, Institute of Biotechnology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Zelton Dave Sharp
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center, San Antonio, TX, USA
| | - Tyler J Curiel
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX, USA
- Graduate School of Biomedical Sciences, University of Texas Health Science Center, San Antonio, TX, USA
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center, San Antonio, TX, USA
- Cancer Therapy & Research Center, University of Texas Health Science Center, San Antonio, TX, USA
| |
Collapse
|
520
|
Lovelace ES, Polyak SJ. Natural Products as Tools for Defining How Cellular Metabolism Influences Cellular Immune and Inflammatory Function during Chronic Infection. Viruses 2015; 7:6218-32. [PMID: 26633463 PMCID: PMC4690857 DOI: 10.3390/v7122933] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 11/13/2015] [Accepted: 11/19/2015] [Indexed: 12/21/2022] Open
Abstract
Chronic viral infections like those caused by hepatitis C virus (HCV) and human immunodeficiency virus (HIV) cause disease that establishes an ongoing state of chronic inflammation. While there have been tremendous improvements towards curing HCV with directly acting antiviral agents (DAA) and keeping HIV viral loads below detection with antiretroviral therapy (ART), there is still a need to control inflammation in these diseases. Recent studies indicate that many natural products like curcumin, resveratrol and silymarin alter cellular metabolism and signal transduction pathways via enzymes such as adenosine monophosphate kinase (AMPK) and mechanistic target of rapamycin (mTOR), and these pathways directly influence cellular inflammatory status (such as NF-κB) and immune function. Natural products represent a vast toolkit to dissect and define how cellular metabolism controls cellular immune and inflammatory function.
Collapse
Affiliation(s)
- Erica S Lovelace
- Department of Laboratory Medicine, University of Washington, Seattle, WA 98195, USA.
| | - Stephen J Polyak
- Department of Laboratory Medicine, University of Washington, Seattle, WA 98195, USA.
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA.
- Department of Global Health, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
521
|
Eiden AM, Zhang S, Gary JM, Simmons JK, Mock BA. Molecular Pathways: Increased Susceptibility to Infection Is a Complication of mTOR Inhibitor Use in Cancer Therapy. Clin Cancer Res 2015; 22:277-83. [PMID: 26607598 DOI: 10.1158/1078-0432.ccr-14-3239] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 11/04/2015] [Indexed: 12/20/2022]
Abstract
As one of the earliest examples of "chemical biology," the M: echanistic T: arget of R: apamycin (mTOR) protein and its chemical inhibitors have been extensively studied across a spectrum of physiologic and pathologic processes at the molecular, organismal, and patient population levels. There are several FDA-approved mTOR inhibitors (sirolimus, everolimus, and temsirolimus) with indications for cancer treatment and for prevention of solid organ rejection. Dozens of mTOR inhibitors are currently being evaluated in hundreds of ongoing clinical trials across a spectrum of diseases, including numerous cancer indications, autoimmune diseases, and a number of congenital disorders. As many of the approved and investigational indications for mTOR inhibitors require long-term treatment, the magnitude and incidence of particular side effects differ from those observed in shorter-term treatments. Here, we focus on the increased risk of infections in patients being treated with mTOR inhibitors. While increased infection rates might be expected from a class of drugs approved as posttransplant immunosuppressants, we review reports from clinical, mechanistic, and genetically engineered mouse model studies detailing a much more nuanced view of mTOR inhibitor drug action and target biology.
Collapse
Affiliation(s)
- Adrian M Eiden
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Shuling Zhang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Joy M Gary
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - John K Simmons
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Beverly A Mock
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
522
|
Recent advances in the molecular design of synthetic vaccines. Nat Chem 2015; 7:952-60. [DOI: 10.1038/nchem.2396] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 10/13/2015] [Indexed: 01/07/2023]
|
523
|
Zhavoronkov A, Bhullar B. Classifying aging as a disease in the context of ICD-11. Front Genet 2015; 6:326. [PMID: 26583032 PMCID: PMC4631811 DOI: 10.3389/fgene.2015.00326] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/20/2015] [Indexed: 01/16/2023] Open
Abstract
Aging is a complex continuous multifactorial process leading to loss of function and crystalizing into the many age-related diseases. Here, we explore the arguments for classifying aging as a disease in the context of the upcoming World Health Organization's 11th International Statistical Classification of Diseases and Related Health Problems (ICD-11), expected to be finalized in 2018. We hypothesize that classifying aging as a disease with a "non-garbage" set of codes will result in new approaches and business models for addressing aging as a treatable condition, which will lead to both economic and healthcare benefits for all stakeholders. Actionable classification of aging as a disease may lead to more efficient allocation of resources by enabling funding bodies and other stakeholders to use quality-adjusted life years (QALYs) and healthy-years equivalent (HYE) as metrics when evaluating both research and clinical programs. We propose forming a Task Force to interface the WHO in order to develop a multidisciplinary framework for classifying aging as a disease with multiple disease codes facilitating for therapeutic interventions and preventative strategies.
Collapse
Affiliation(s)
- Alex Zhavoronkov
- The Biogerontology Research Foundation, Oxford, UK
- Insilico Medicine Inc, Baltimore, MD, USA
| | - Bhupinder Bhullar
- Novartis Pharma AG, Department of Developmental and Molecular Pathways, Novartis Institute for Biomedical Research, Basel, Switzerland
| |
Collapse
|
524
|
Bitto A, Wang AM, Bennett CF, Kaeberlein M. Biochemical Genetic Pathways that Modulate Aging in Multiple Species. Cold Spring Harb Perspect Med 2015; 5:5/11/a025114. [PMID: 26525455 DOI: 10.1101/cshperspect.a025114] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The mechanisms underlying biological aging have been extensively studied in the past 20 years with the avail of mainly four model organisms: the budding yeast Saccharomyces cerevisiae, the nematode Caenorhabditis elegans, the fruitfly Drosophila melanogaster, and the domestic mouse Mus musculus. Extensive research in these four model organisms has identified a few conserved genetic pathways that affect longevity as well as metabolism and development. Here, we review how the mechanistic target of rapamycin (mTOR), sirtuins, adenosine monophosphate-activated protein kinase (AMPK), growth hormone/insulin-like growth factor 1 (IGF-1), and mitochondrial stress-signaling pathways influence aging and life span in the aforementioned models and their possible implications for delaying aging in humans. We also draw some connections between these biochemical pathways and comment on what new developments aging research will likely bring in the near future.
Collapse
Affiliation(s)
- Alessandro Bitto
- Department of Pathology, University of Washington, Seattle, Washington 98195
| | - Adrienne M Wang
- Department of Pathology, University of Washington, Seattle, Washington 98195
| | | | - Matt Kaeberlein
- Department of Pathology, University of Washington, Seattle, Washington 98195
| |
Collapse
|
525
|
Blackwell TK, Steinbaugh MJ, Hourihan JM, Ewald CY, Isik M. SKN-1/Nrf, stress responses, and aging in Caenorhabditis elegans. Free Radic Biol Med 2015; 88:290-301. [PMID: 26232625 PMCID: PMC4809198 DOI: 10.1016/j.freeradbiomed.2015.06.008] [Citation(s) in RCA: 429] [Impact Index Per Article: 42.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 06/17/2015] [Accepted: 06/18/2015] [Indexed: 01/06/2023]
Abstract
The mammalian Nrf/CNC proteins (Nrf1, Nrf2, Nrf3, p45 NF-E2) perform a wide range of cellular protective and maintenance functions. The most thoroughly described of these proteins, Nrf2, is best known as a regulator of antioxidant and xenobiotic defense, but more recently has been implicated in additional functions that include proteostasis and metabolic regulation. In the nematode Caenorhabditis elegans, which offers many advantages for genetic analyses, the Nrf/CNC proteins are represented by their ortholog SKN-1. Although SKN-1 has diverged in aspects of how it binds DNA, it exhibits remarkable functional conservation with Nrf/CNC proteins in other species and regulates many of the same target gene families. C. elegans may therefore have considerable predictive value as a discovery model for understanding how mammalian Nrf/CNC proteins function and are regulated in vivo. Work in C. elegans indicates that SKN-1 regulation is surprisingly complex and is influenced by numerous growth, nutrient, and metabolic signals. SKN-1 is also involved in a wide range of homeostatic functions that extend well beyond the canonical Nrf2 function in responses to acute stress. Importantly, SKN-1 plays a central role in diverse genetic and pharmacologic interventions that promote C. elegans longevity, suggesting that mechanisms regulated by SKN-1 may be of conserved importance in aging. These C. elegans studies predict that mammalian Nrf/CNC protein functions and regulation may be similarly complex and that the proteins and processes that they regulate are likely to have a major influence on mammalian life- and healthspan.
Collapse
Affiliation(s)
- T Keith Blackwell
- Research Division, Joslin Diabetes Center, One Joslin Place, Boston, MA 02215, USA; Department of Genetics and Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02215, USA.
| | - Michael J Steinbaugh
- Research Division, Joslin Diabetes Center, One Joslin Place, Boston, MA 02215, USA; Department of Genetics and Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02215, USA
| | - John M Hourihan
- Research Division, Joslin Diabetes Center, One Joslin Place, Boston, MA 02215, USA; Department of Genetics and Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Collin Y Ewald
- Research Division, Joslin Diabetes Center, One Joslin Place, Boston, MA 02215, USA; Department of Genetics and Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Meltem Isik
- Research Division, Joslin Diabetes Center, One Joslin Place, Boston, MA 02215, USA; Department of Genetics and Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
526
|
Natural products as probes in pharmaceutical research. J Ind Microbiol Biotechnol 2015; 43:249-60. [PMID: 26438431 DOI: 10.1007/s10295-015-1691-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/16/2015] [Indexed: 10/23/2022]
Abstract
From the start of the pharmaceutical research natural products played a key role in drug discovery and development. Over time many discoveries of fundamental new biology were triggered by the unique biological activity of natural products. Unprecedented chemical structures, novel chemotypes, often pave the way to investigate new biology and to explore new pathways and targets. This review summarizes the recent results in the area with a focus on research done in the laboratories of Novartis Institutes for BioMedical Research. We aim to put the technological advances in target identification techniques in the context to the current revival of phenotypic screening and the increasingly complex biological questions related to drug discovery.
Collapse
|
527
|
Liko D, Hall MN. mTOR in health and in sickness. J Mol Med (Berl) 2015; 93:1061-73. [DOI: 10.1007/s00109-015-1326-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 07/14/2015] [Accepted: 07/22/2015] [Indexed: 01/12/2023]
|
528
|
|
529
|
Abstract
Aging is accompanied by immune decline leading to increased incidence of infections and malignancies, given the demographic shift of humans towards an older age the identification of strategies for the manipulation of immunity is an important goal. Evidence implicates mammalian target of rapamycin (mTOR) to be a key modulator of aging and the use of mTOR inhibitors has been shown to ameliorate much age-related pathology; however, recent data suggest that senescent CD8(+) T-cells function independently of mTOR. This review article will challenge the perceived dogma that mTOR universally controls CD8(+) T-cell function.
Collapse
|
530
|
Taylor JG, Gribben JG. Microenvironment abnormalities and lymphomagenesis: Immunological aspects. Semin Cancer Biol 2015; 34:36-45. [PMID: 26232774 DOI: 10.1016/j.semcancer.2015.07.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 07/19/2015] [Accepted: 07/21/2015] [Indexed: 01/06/2023]
Abstract
Innate and adaptive immune cells within the microenvironment identify and eliminate cells displaying signs of malignant potential. Immunosurveillance effector Natural Killer (NK) cells and Cytotoxic T Lymphocytes (CTL) identify malignant cells through germline receptors such as NKG2D and in the case of CTLs, presentation of antigen through the T cell receptor. Manipulation of immunosurveillance through altered tumor-identifying ligand expression or secretion, resistance to cytotoxicity, or compromised cytotoxic cell activity through immune tolerance mechanisms all contribute to failure of these systems to prevent cancer development. This review examines the diverse mechanisms by which alterations in the immune microenvironment can promote lymphomagenesis.
Collapse
Affiliation(s)
| | - John G Gribben
- Barts Cancer Institute, Queen Mary University of London, UK.
| |
Collapse
|
531
|
Castillo-Quan JI, Kinghorn KJ, Bjedov I. Genetics and pharmacology of longevity: the road to therapeutics for healthy aging. ADVANCES IN GENETICS 2015; 90:1-101. [PMID: 26296933 DOI: 10.1016/bs.adgen.2015.06.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aging can be defined as the progressive decline in tissue and organismal function and the ability to respond to stress that occurs in association with homeostatic failure and the accumulation of molecular damage. Aging is the biggest risk factor for human disease and results in a wide range of aging pathologies. Although we do not completely understand the underlying molecular basis that drives the aging process, we have gained exceptional insights into the plasticity of life span and healthspan from the use of model organisms such as the worm Caenorhabditis elegans and the fruit fly Drosophila melanogaster. Single-gene mutations in key cellular pathways that regulate environmental sensing, and the response to stress, have been identified that prolong life span across evolution from yeast to mammals. These genetic manipulations also correlate with a delay in the onset of tissue and organismal dysfunction. While the molecular genetics of aging will remain a prosperous and attractive area of research in biogerontology, we are moving towards an era defined by the search for therapeutic drugs that promote healthy aging. Translational biogerontology will require incorporation of both therapeutic and pharmacological concepts. The use of model organisms will remain central to the quest for drug discovery, but as we uncover molecular processes regulated by repurposed drugs and polypharmacy, studies of pharmacodynamics and pharmacokinetics, drug-drug interactions, drug toxicity, and therapeutic index will slowly become more prevalent in aging research. As we move from genetics to pharmacology and therapeutics, studies will not only require demonstration of life span extension and an underlying molecular mechanism, but also the translational relevance for human health and disease prevention.
Collapse
Affiliation(s)
- Jorge Iván Castillo-Quan
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK; Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Kerri J Kinghorn
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK; Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Ivana Bjedov
- Cancer Institute, University College London, London, UK
| |
Collapse
|
532
|
Richner JM, Gmyrek GB, Govero J, Tu Y, van der Windt GJW, Metcalf TU, Haddad EK, Textor J, Miller MJ, Diamond MS. Age-Dependent Cell Trafficking Defects in Draining Lymph Nodes Impair Adaptive Immunity and Control of West Nile Virus Infection. PLoS Pathog 2015. [PMID: 26204259 PMCID: PMC4512688 DOI: 10.1371/journal.ppat.1005027] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Impaired immune responses in the elderly lead to reduced vaccine efficacy and increased susceptibility to viral infections. Although several groups have documented age-dependent defects in adaptive immune priming, the deficits that occur prior to antigen encounter remain largely unexplored. Herein, we identify novel mechanisms for compromised adaptive immunity that occurs with aging in the context of infection with West Nile virus (WNV), an encephalitic flavivirus that preferentially causes disease in the elderly. An impaired IgM and IgG response and enhanced vulnerability to WNV infection during aging was linked to delayed germinal center formation in the draining lymph node (DLN). Adoptive transfer studies and two-photon intravital microscopy revealed a decreased trafficking capacity of donor naïve CD4+ T cells from old mice, which manifested as impaired T cell diapedesis at high endothelial venules and reduced cell motility within DLN prior to antigen encounter. Furthermore, leukocyte accumulation in the DLN within the first few days of WNV infection or antigen-adjuvant administration was diminished more generally in old mice and associated with a second aging-related defect in local cytokine and chemokine production. Thus, age-dependent cell-intrinsic and environmental defects in the DLN result in delayed immune cell recruitment and antigen recognition. These deficits compromise priming of early adaptive immune responses and likely contribute to the susceptibility of old animals to acute WNV infection.
Collapse
Affiliation(s)
- Justin M. Richner
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Grzegorz B. Gmyrek
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Jennifer Govero
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Yizheng Tu
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Gerritje J. W. van der Windt
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Talibah U. Metcalf
- Vaccine and Gene Therapy Institute of Florida, Port St. Lucie, Florida, United States of America
| | - Elias K. Haddad
- Vaccine and Gene Therapy Institute of Florida, Port St. Lucie, Florida, United States of America
| | - Johannes Textor
- Department of Theoretical Biology & Bioinformatics, Utrecht University, Utrecht, Netherlands
| | - Mark J. Miller
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Michael S. Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
533
|
Montgomery RR, Shaw AC. Paradoxical changes in innate immunity in aging: recent progress and new directions. J Leukoc Biol 2015; 98:937-43. [PMID: 26188078 DOI: 10.1189/jlb.5mr0315-104r] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 06/23/2015] [Indexed: 12/29/2022] Open
Abstract
Immunosenescence, describing alterations, including decline of immune responses with age, is comprised of inappropriate elevations, decreases, and dysregulated immune responses, leading to more severe consequences of bacterial and viral infections and reduced responses to vaccination. In adaptive immunity, these changes include increased proportions of antigen-experienced B and T cells at the cost of naïve cell populations. Innate immune changes in aging are complex in spanning multiple cell types, activation states, and tissue context. Innate immune responses are dampened in aging, yet there is also a paradoxical increase in certain signaling pathways and cytokine levels. Here, we review recent progress and highlight novel directions for expected advances that can lead the aging field to a new era of discovery that will embrace the complexity of aging in human populations.
Collapse
Affiliation(s)
- Ruth R Montgomery
- Sections of *Rheumatology and Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Albert C Shaw
- Sections of *Rheumatology and Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
534
|
Faragher RGA. Should we treat aging as a disease? The consequences and dangers of miscategorisation. Front Genet 2015; 6:171. [PMID: 26236330 PMCID: PMC4500987 DOI: 10.3389/fgene.2015.00171] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 04/17/2015] [Indexed: 11/24/2022] Open
Abstract
The aging of the population represents one of the largest healthcare challenges facing the world today. The available scientific evidence shows that interventions are available now that can target fundamental "aging" processes or pathways. Sufficient economic evidence is available to argue convincingly that this approach will also save enormous sums of money which could then be deployed to solve other urgent global problems. However, as yet this scenario has barely entered the public consciousness and, far from being a point of vigorous debate, seems to be ignored by policy makers. Understanding why this lethargy exists is important given the urgent need to deal with the challenge represented by population aging. In this paper I hypothesize that one major cause of inaction is a widely held, but flawed, conceptual framework concerning the relationship between aging and disease that categorizes the former as "natural" and the latter as "abnormal." This perspective is sufficient in itself to act as a disincentive to intervention by rendering those who hold it prone to the "naturalistic fallacy" but can give rise to active hostility to biogerontology if coupled with loose and/or blurred understanding of the goals and potential of the field.
Collapse
|
535
|
|
536
|
de Mare-Bredemeijer ELD, Shi XL, Mancham S, van Gent R, van der Heide-Mulder M, de Boer R, Heemskerk MHM, de Jonge J, van der Laan LJW, Metselaar HJ, Kwekkeboom J. Cytomegalovirus-Induced Expression of CD244 after Liver Transplantation Is Associated with CD8+ T Cell Hyporesponsiveness to Alloantigen. THE JOURNAL OF IMMUNOLOGY 2015; 195:1838-48. [DOI: 10.4049/jimmunol.1500440] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 06/02/2015] [Indexed: 12/31/2022]
|
537
|
Abstract
The field of aging research has progressed rapidly over the past few decades. Genetic modulators of aging rate that are conserved over a broad evolutionary distance have now been identified. Several physiological and environmental interventions have also been shown to influence the rate of aging in organisms ranging from yeast to mammals. Here we briefly review these conserved pathways and interventions and highlight some key unsolved challenges that remain. Although the molecular mechanisms by which these modifiers of aging act are only partially understood, interventions to slow aging are nearing clinical application, and it is likely that we will begin to reap the benefits of aging research prior to solving all of the mysteries that the biology of aging has to offer. Old age is the single greatest risk factor for the leading causes of death in the developed world. Advances in aging research promise to alleviate the diseases of aging by targeting aging itself.
Collapse
|
538
|
Saffrey J, Brown JE. The interconnectedness of ageing: does the convoy principal apply? Biogerontology 2015; 16:131-2. [DOI: 10.1007/s10522-015-9556-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 01/29/2015] [Indexed: 10/24/2022]
|
539
|
The S6K protein family in health and disease. Life Sci 2015; 131:1-10. [PMID: 25818187 DOI: 10.1016/j.lfs.2015.03.001] [Citation(s) in RCA: 188] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 03/02/2015] [Accepted: 03/05/2015] [Indexed: 02/06/2023]
Abstract
The S6K proteins are mTOR pathway effectors and accumulative evidence suggest that mTOR/S6K signaling contributes to several pathological conditions, such as diabetes, cancer and obesity. The activation of the mTOR/S6K axis stimulates protein synthesis and cell growth. S6K1 has two well-known isoforms, p70-S6K1 and p85-S6K1, generated by alternative translation initiation sites. A third isoform, named p31-S6K1, has been characterized as a truncated type of the protein due to alternative splicing, and reports have shown its important role in cancer. Studies involving S6K2 are scarce. This article aims to review what is new in the literature about these kinases and establish differences regarding their interacting proteins, activation and function, connecting their roles in the homeostasis of the cell and in pathological conditions.
Collapse
|
540
|
Dao V, Pandeswara S, Liu Y, Hurez V, Dodds S, Callaway D, Liu A, Hasty P, Sharp ZD, Curiel TJ. Prevention of carcinogen and inflammation-induced dermal cancer by oral rapamycin includes reducing genetic damage. Cancer Prev Res (Phila) 2015; 8:400-9. [PMID: 25736275 DOI: 10.1158/1940-6207.capr-14-0313-t] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 02/26/2015] [Indexed: 01/22/2023]
Abstract
Cancer prevention is a cost-effective alternative to treatment. In mice, the mTOR inhibitor rapamycin prevents distinct spontaneous, noninflammatory cancers, making it a candidate broad-spectrum cancer prevention agent. We now show that oral microencapsulated rapamycin (eRapa) prevents skin cancer in dimethylbenz(a)anthracene (DMBA)/12-O-tetradecanoylphorbol-13-acetate (TPA) carcinogen-induced, inflammation-driven carcinogenesis. eRapa given before DMBA/TPA exposure significantly increased tumor latency, reduced papilloma prevalence and numbers, and completely inhibited malignant degeneration into squamous cell carcinoma. Rapamycin is primarily an mTORC1-specific inhibitor, but eRapa did not reduce mTORC1 signaling in skin or papillomas, and did not reduce important proinflammatory factors in this model, including p-Stat3, IL17A, IL23, IL12, IL1β, IL6, or TNFα. In support of lack of mTORC1 inhibition, eRapa did not reduce numbers or proliferation of CD45(-)CD34(+)CD49f(mid) skin cancer initiating stem cells in vivo and marginally reduced epidermal hyperplasia. Interestingly, eRapa reduced DMBA/TPA-induced skin DNA damage and the hras codon 61 mutation that specifically drives carcinogenesis in this model, suggesting reduction of DNA damage as a cancer prevention mechanism. In support, cancer prevention and DNA damage reduction effects were lost when eRapa was given after DMBA-induced DNA damage in vivo. eRapa afforded picomolar concentrations of rapamycin in skin of DMBA/TPA-exposed mice, concentrations that also reduced DMBA-induced DNA damage in mouse and human fibroblasts in vitro. Thus, we have identified DNA damage reduction as a novel mechanism by which rapamycin can prevent cancer, which could lay the foundation for its use as a cancer prevention agent in selected human populations.
Collapse
Affiliation(s)
- Vinh Dao
- The Graduate School of Biomedical Sciences, University of Texas Health Science Center, San Antonio, Texas. Department of Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - Srilakshmi Pandeswara
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - Yang Liu
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas. Xiangya School of Medicine, Central South University, Changsha, Hunan, P.R. China
| | - Vincent Hurez
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - Sherry Dodds
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - Danielle Callaway
- The Graduate School of Biomedical Sciences, University of Texas Health Science Center, San Antonio, Texas
| | - Aijie Liu
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - Paul Hasty
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - Zelton D Sharp
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, Texas
| | - Tyler J Curiel
- The Graduate School of Biomedical Sciences, University of Texas Health Science Center, San Antonio, Texas. Department of Medicine, University of Texas Health Science Center, San Antonio, Texas. Cancer Therapy and Research Center, University of Texas Health Science Center, San Antonio, Texas.
| |
Collapse
|
541
|
Warner HR. NIA's Intervention Testing Program at 10 years of age. AGE (DORDRECHT, NETHERLANDS) 2015; 37:22. [PMID: 25726185 PMCID: PMC4344944 DOI: 10.1007/s11357-015-9761-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 02/18/2015] [Indexed: 06/04/2023]
Abstract
The previous 20 years of basic research on aging has identified a large number of genes and gene products whose expression can be manipulated in a variety of ways to increase the healthy life span of animal models such as yeast, nematodes, fruit flies, and mice. In an overt attempt to capitalize on this information, the National Institute on Aging (NIA) began a program in 2003 to identify nutritional and pharmaceutical interventions that could be safely employed to extend the healthy life span of mice. This program is called the Intervention Testing Program (ITP), and this article briefly describes the development of this initiative and some of the early success achieved during its first 10 years (2004-2014) of operation.
Collapse
Affiliation(s)
- Huber R Warner
- College of Biological Sciences, University of Minnesota, St. Paul, MN, 55108, USA,
| |
Collapse
|