1
|
Shu Z, Li X, Zhang W, Huyan Z, Cheng D, Xie S, Cheng H, Wang J, Du B. MG-132 activates sodium palmitate-induced autophagy in human vascular smooth muscle cells and inhibits senescence via the PI3K/AKT/mTOR axis. Lipids Health Dis 2024; 23:282. [PMID: 39232759 PMCID: PMC11373134 DOI: 10.1186/s12944-024-02268-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 08/19/2024] [Indexed: 09/06/2024] Open
Abstract
OBJECTIVE This study aimed to reveal the role and mechanism of MG-132 in delaying hyperlipidemia-induced senescence of vascular smooth muscle cells (VSMCs). METHODS Immunohistochemistry and hematoxylin-eosin staining confirmed the therapeutic effect of MG-132 on arterial senescence in vivo and its possible mechanism. Subsequently, VSMCs were treated with sodium palmitate (PA), an activator (Recilisib) or an inhibitor (Pictilisib) to activate or inhibit PI3K, and CCK-8 and EdU staining, wound healing assays, Transwell cell migration assays, autophagy staining assays, reactive oxygen species assays, senescence-associated β-galactosidase staining, and Western blotting were performed to determine the molecular mechanism by which MG-132 inhibits VSMC senescence. Validation of the interaction between MG-132 and PI3K using molecular docking. RESULTS Increased expression of p-PI3K, a key protein of the autophagy regulatory system, and decreased expression of the autophagy-associated proteins Beclin 1 and ULK1 were observed in the aortas of C57BL/6J mice fed a high-fat diet (HFD), and autophagy was inhibited in aortic smooth muscle. MG-132 inhibits atherosclerosis by activating autophagy in VSMCs to counteract PA-induced cell proliferation, migration, oxidative stress, and senescence, thereby inhibiting VSMC senescence in the aorta. This process is achieved through the PI3K/AKT/mTOR signaling pathway. CONCLUSION MG-132 activates autophagy by inhibiting the PI3K/AKT/mTOR pathway, thereby inhibiting palmitate-induced proliferation, migration, and oxidative stress in vascular smooth muscle cells and suppressing their senescence.
Collapse
MESH Headings
- Autophagy/drug effects
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/cytology
- TOR Serine-Threonine Kinases/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- Animals
- Cellular Senescence/drug effects
- Humans
- Phosphatidylinositol 3-Kinases/metabolism
- Mice
- Signal Transduction/drug effects
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Leupeptins/pharmacology
- Male
- Mice, Inbred C57BL
- Palmitic Acid/pharmacology
- Cell Proliferation/drug effects
- Cell Movement/drug effects
- Diet, High-Fat/adverse effects
Collapse
Affiliation(s)
- Zhiyun Shu
- Department of Cardiology, First Hospital of Jilin University, Changchun, Jilin, 130000, China
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Rd, Changchun, Jilin, 130000, China
| | - Xiangjun Li
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Rd, Changchun, Jilin, 130000, China
| | - Wenqing Zhang
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Rd, Changchun, Jilin, 130000, China
| | - Zixu Huyan
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Rd, Changchun, Jilin, 130000, China
| | - Dong Cheng
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Shishun Xie
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Rd, Changchun, Jilin, 130000, China
| | - Hongyuan Cheng
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Rd, Changchun, Jilin, 130000, China
| | - Jiajia Wang
- Department of Cardiology, First Hospital of Jilin University, Changchun, Jilin, 130000, China
| | - Bing Du
- Department of Cardiology, First Hospital of Jilin University, Changchun, Jilin, 130000, China.
| |
Collapse
|
2
|
Mafra D, Borges NA, Baptista BG, Martins LF, Borland G, Shiels PG, Stenvinkel P. What Can the Gut Microbiota of Animals Teach Us about the Relationship between Nutrition and Burden of Lifestyle Diseases? Nutrients 2024; 16:1789. [PMID: 38892721 PMCID: PMC11174762 DOI: 10.3390/nu16111789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
The gut microbiota performs several crucial roles in a holobiont with its host, including immune regulation, nutrient absorption, synthesis, and defense against external pathogens, significantly influencing host physiology. Disruption of the gut microbiota has been linked to various chronic conditions, including cardiovascular, kidney, liver, respiratory, and intestinal diseases. Studying how animals adapt their gut microbiota across their life course at different life stages and under the dynamics of extreme environmental conditions can provide valuable insights from the natural world into how the microbiota modulates host biology, with a view to translating these into treatments or preventative measures for human diseases. By modulating the gut microbiota, opportunities to address many complications associated with chronic diseases appear. Such a biomimetic approach holds promise for exploring new strategies in healthcare and disease management.
Collapse
Affiliation(s)
- Denise Mafra
- Graduate Program in Medical Sciences and Graduate Program in Nutrition Sciences, Federal Fluminense University (UFF), Niterói 24020-141, Brazil;
- Graduate Program in Biological Sciences—Physiology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-909, Brazil
| | - Natália A. Borges
- Graduate Program in Food, Nutrition, and Health, Institute of Nutrition, State University of Rio de Janeiro (UERJ), Rio de Janeiro 21941-909, Brazil;
| | - Beatriz G. Baptista
- Graduate Program in Medical Sciences and Graduate Program in Nutrition Sciences, Federal Fluminense University (UFF), Niterói 24020-141, Brazil;
| | - Layla F. Martins
- Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), São Paulo 05508-220, Brazil;
| | - Gillian Borland
- School of Molecular Biosciences, University of Glasgow, Glasgow G12 8QQ, UK; (G.B.); (P.G.S.)
| | - Paul G. Shiels
- School of Molecular Biosciences, University of Glasgow, Glasgow G12 8QQ, UK; (G.B.); (P.G.S.)
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science, Technology and Intervention, Karolinska Institutet, 17165 Stockholm, Sweden;
| |
Collapse
|
3
|
Sun F, Sutovsky P, Patterson AL, Balboula AZ. Mechanisms of DNA Damage Response in Mammalian Oocytes. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2024; 238:47-68. [PMID: 39030354 DOI: 10.1007/978-3-031-55163-5_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/21/2024]
Abstract
DNA damage poses a significant challenge to all eukaryotic cells, leading to mutagenesis, genome instability and senescence. In somatic cells, the failure to repair damaged DNA can lead to cancer development, whereas, in oocytes, it can lead to ovarian dysfunction and infertility. The response of the cell to DNA damage entails a series of sequential and orchestrated events including sensing the DNA damage, activating DNA damage checkpoint, chromatin-related conformational changes, activating the DNA damage repair machinery and/or initiating the apoptotic cascade. This chapter focuses on how somatic cells and mammalian oocytes respond to DNA damage. Specifically, we will discuss how and why fully grown mammalian oocytes differ drastically from somatic cells and growing oocytes in their response to DNA damage.
Collapse
Affiliation(s)
- Fei Sun
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Peter Sutovsky
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, USA
| | - Amanda L Patterson
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, USA
| | - Ahmed Z Balboula
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
4
|
Wlaschek M, Singh K, Maity P, Scharffetter-Kochanek K. The skin of the naked mole-rat and its resilience against aging and cancer. Mech Ageing Dev 2023; 216:111887. [PMID: 37993056 DOI: 10.1016/j.mad.2023.111887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 10/10/2023] [Accepted: 11/14/2023] [Indexed: 11/24/2023]
Abstract
The naked mole-rat (NMR) Heterocephalus glaber (from the Greek/latin words ἕτερος, heteros = divergent, κεφαλή, kephalē = head and glabra = hairless) was first described by Rüppell (Fig. 1) and belongs to the Hystricognath (from the Greek words ὕστριξ, hystrix = porcupine and γνάθος, gnathos = jaw) as a suborder of rodents. NMR are characterized by the highest longevity among rodents and reveal a profound cancer resistance. Details of its skin-specific protective and resistance mechanisms against aging and carcinogenesis have so far not been adequately characterized. Recently, our knowledge of NMR skin biology was complemented and expanded by published data using state-of-the art histological and molecular techniques. Here we review and integrate novel published data regarding skin morphology and histology of the aging NMR and the underlying mechanisms at the cellular and molecular level. We relate this data to the longevity of the NMR and its resistance to neoplastic transformation and discuss further open questions to understand its extraordinary longevity. In addition, we will address the exposome, defined as "the total of all non-genetic, endogenous and exogenous environmental influences" on the skin, respiratory tract, stomach, and intestine. Finally, we will discuss in perspective further intriguing possibilities arising from the interaction of skin with other organs.
Collapse
Affiliation(s)
- Meinhard Wlaschek
- Department of Dermatology and Allergic Diseases, Ulm University, Ulm, Germany.
| | - Karmveer Singh
- Department of Dermatology and Allergic Diseases, Ulm University, Ulm, Germany.
| | - Pallab Maity
- Department of Dermatology and Allergic Diseases, Ulm University, Ulm, Germany.
| | | |
Collapse
|
5
|
Gressler AE, Leng H, Zinecker H, Simon AK. Proteostasis in T cell aging. Semin Immunol 2023; 70:101838. [PMID: 37708826 PMCID: PMC10804938 DOI: 10.1016/j.smim.2023.101838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 09/16/2023]
Abstract
Aging leads to a decline in immune cell function, which leaves the organism vulnerable to infections and age-related multimorbidities. One major player of the adaptive immune response are T cells, and recent studies argue for a major role of disturbed proteostasis contributing to reduced function of these cells upon aging. Proteostasis refers to the state of a healthy, balanced proteome in the cell and is influenced by synthesis (translation), maintenance and quality control of proteins, as well as degradation of damaged or unwanted proteins by the proteasome, autophagy, lysosome and cytoplasmic enzymes. This review focuses on molecular processes impacting on proteostasis in T cells, and specifically functional or quantitative changes of each of these upon aging. Importantly, we describe the biological consequences of compromised proteostasis in T cells, which range from impaired T cell activation and function to enhancement of inflamm-aging by aged T cells. Finally, approaches to improve proteostasis and thus rejuvenate aged T cells through pharmacological or physical interventions are discussed.
Collapse
Affiliation(s)
- A Elisabeth Gressler
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Houfu Leng
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX3 7FY, United Kingdom; Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Heidi Zinecker
- Ascenion GmbH, Am Zirkus 1, Bertold-Brecht-Platz 3, 10117 Berlin, Germany
| | - Anna Katharina Simon
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany; Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX3 7FY, United Kingdom.
| |
Collapse
|
6
|
Dysregulation of sphingosine-1-phosphate (S1P) and S1P receptor 1 signaling in the 5xFAD mouse model of Alzheimer's disease. Brain Res 2023; 1799:148171. [PMID: 36410428 DOI: 10.1016/j.brainres.2022.148171] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/22/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022]
Abstract
Sphingolipid-1-phosphate (S1P) signaling through the activation S1P receptors (S1PRs) plays critical roles in cellular events in the brain. Aberrant S1P metabolism has been identified in the brains of Alzheimer's disease (AD) patients. Our recent studies have shown that treatment with fingolimod, an analog of sphingosine, provides neuroprotective effects in five familiar Alzheimer disease (5xFAD) transgenic mice, resulting in the reduction of amyloid-β (Aβ) neurotoxicity, inhibition of activation of microglia and astrocytes, increased hippocampal neurogenesis, and improved learning and memory. However, the pathways by which dysfunctional S1P and S1PR signaling may associate with the development of AD-like pathology remain unknown. In this study, we investigated the alteration of signaling of S1P/S1P receptor 1 (S1PR1), the most abundant S1PR subtype in the brain, in the cortex of 5xFAD transgenic mice at 3, 8, and 14 months of age. Compared to non-transgenic wildtype (WT) littermates, we found significant decreased levels of sphingosine kinases (SphKs), increased S1P lyase (S1PL), and increased S1PR1 in 8- and 14-month-old, but not in 3-month-old 5xFAD mice. Furthermore, we detected increased activation of the S1PR1 downstream pathway of Akt/mTor/Tau signaling in aging 5xFAD mice. Treatment with fingolimod from 1 to 8 months of age reversed the levels of SphKs, S1PL, and furthermore, those of S1PR1 and its downstream pathway of Akt/mTor/Tau signaling. Together the data reveal that dysregulation of S1P and S1PR signaling may associate with the development of AD-like pathology through Akt/mTor/Tau signaling.
Collapse
|
7
|
Rappe A, McWilliams TG. Mitophagy in the aging nervous system. Front Cell Dev Biol 2022; 10:978142. [PMID: 36303604 PMCID: PMC9593040 DOI: 10.3389/fcell.2022.978142] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 09/07/2022] [Indexed: 02/01/2024] Open
Abstract
Aging is characterised by the progressive accumulation of cellular dysfunction, stress, and inflammation. A large body of evidence implicates mitochondrial dysfunction as a cause or consequence of age-related diseases including metabolic disorders, neuropathies, various forms of cancer and neurodegenerative diseases. Because neurons have high metabolic demands and cannot divide, they are especially vulnerable to mitochondrial dysfunction which promotes cell dysfunction and cytotoxicity. Mitophagy neutralises mitochondrial dysfunction, providing an adaptive quality control strategy that sustains metabolic homeostasis. Mitophagy has been extensively studied as an inducible stress response in cultured cells and short-lived model organisms. In contrast, our understanding of physiological mitophagy in mammalian aging remains extremely limited, particularly in the nervous system. The recent profiling of mitophagy reporter mice has revealed variegated vistas of steady-state mitochondrial destruction across different tissues. The discovery of patients with congenital autophagy deficiency provokes further intrigue into the mechanisms that underpin neural integrity. These dimensions have considerable implications for targeting mitophagy and other degradative pathways in age-related neurological disease.
Collapse
Affiliation(s)
- Anna Rappe
- Translational Stem Cell Biology and Metabolism Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Thomas G. McWilliams
- Translational Stem Cell Biology and Metabolism Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
8
|
Sefiani A, Rusyn I, Geoffroy CG. Novel adult cortical neuron processing and screening method illustrates sex- and age-dependent effects of pharmaceutical compounds. Sci Rep 2022; 12:13125. [PMID: 35908049 PMCID: PMC9338961 DOI: 10.1038/s41598-022-17389-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 07/25/2022] [Indexed: 11/22/2022] Open
Abstract
Neurodegenerative diseases and neurotraumatic injuries are typically age-associated disorders that can reduce neuron survival, neurite outgrowth, and synaptic plasticity leading to loss of cognitive capacity, executive function, and motor control. In pursuit of reducing the loss of said neurological functions, novel compounds are sought that promote neuron viability, neuritogenesis, and/or synaptic plasticity. Current high content in vitro screenings typically use cells that are iPSC-derived, embryonic, or originate from post-natal tissues; however, most patients suffering from neurodegenerative diseases and neurotrauma are of middle-age and older. The chasm in maturity between the neurons used in drug screens and those in a target population is a barrier for translational success of in vitro results. It has been historically challenging to culture adult neurons let alone conduct screenings; therefore, age-appropriate drug screenings have previously not been plausible. We have modified Miltenyi's protocol to increase neuronal yield, neuron purity, and neural viability at a reduced cost to expand our capacity to screen compounds directly in primary adult neurons. To our knowledge, we developed the first morphology-based screening system using adult cortical neurons and the first to incorporate age and sex as biological variables in a screen using adult cortical neurons. By using primary adult cortical neurons from mice that were 4 to 48 weeks old for screening pharmaceutical agents, we have demonstrated age- and sex-dependent effects on neuritogenesis and neuron survival in vitro. Utilizing age- and sex-appropriate in vitro models to find novel compounds increasing neuron survival and neurite outgrowth, made possible by our modified adult neuron processing method, will greatly increase the relevance of in vitro screening for finding neuroprotective compounds.
Collapse
Affiliation(s)
- Arthur Sefiani
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University, Bryan, TX, 77807, USA
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Cédric G Geoffroy
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University, Bryan, TX, 77807, USA.
| |
Collapse
|
9
|
Zaman Q, Zhang D, Reddy OS, Wong WT, Lai WF. Roles and Mechanisms of Astragaloside IV in Combating Neuronal Aging. Aging Dis 2022; 13:1845-1861. [DOI: 10.14336/ad.2022.0126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 01/26/2022] [Indexed: 11/18/2022] Open
|
10
|
Kallergi E, Nikoletopoulou V. Macroautophagy and normal aging of the nervous system: Lessons from animal models. Cell Stress 2021; 5:146-166. [PMID: 34708187 PMCID: PMC8490955 DOI: 10.15698/cst2021.10.257] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 01/18/2023] Open
Abstract
Aging represents a cumulative form of cellular stress, which is thought to challenge many aspects of proteostasis. The non-dividing, long-lived neurons are particularly vulnerable to stress, and, not surprisingly, even normal aging is highly associated with a decline in brain function in humans, as well as in other animals. Macroautophagy is a fundamental arm of the proteostasis network, safeguarding proper protein turnover during different cellular states and against diverse cellular stressors. An intricate interplay between macroautophagy and aging is beginning to unravel, with the emergence of new tools, including those for monitoring autophagy in cultured neurons and in the nervous system of different organisms in vivo. Here, we review recent findings on the impact of aging on neuronal integrity and on neuronal macroautophagy, as they emerge from studies in invertebrate and mammalian models.
Collapse
Affiliation(s)
- Emmanouela Kallergi
- University of Lausanne, Department of Fundamental Neurosciences, Lausanne, Switzerland
| | | |
Collapse
|
11
|
Narayan V, McMahon M, O'Brien JJ, McAllister F, Buffenstein R. Insights into the Molecular Basis of Genome Stability and Pristine Proteostasis in Naked Mole-Rats. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1319:287-314. [PMID: 34424521 DOI: 10.1007/978-3-030-65943-1_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The naked mole-rat (Heterocephalus glaber) is the longest-lived rodent, with a maximal reported lifespan of 37 years. In addition to its long lifespan - which is much greater than predicted based on its small body size (longevity quotient of ~4.2) - naked mole-rats are also remarkably healthy well into old age. This is reflected in a striking resistance to tumorigenesis and minimal declines in cardiovascular, neurological and reproductive function in older animals. Over the past two decades, researchers have been investigating the molecular mechanisms regulating the extended life- and health- span of this animal, and since the sequencing and assembly of the naked mole-rat genome in 2011, progress has been rapid. Here, we summarize findings from published studies exploring the unique molecular biology of the naked mole-rat, with a focus on mechanisms and pathways contributing to genome stability and maintenance of proteostasis during aging. We also present new data from our laboratory relevant to the topic and discuss our findings in the context of the published literature.
Collapse
Affiliation(s)
| | - Mary McMahon
- Calico Life Sciences, LLC, South San Francisco, CA, USA
| | | | | | - Rochelle Buffenstein
- Calico Life Sciences, LLC, South San Francisco, CA, USA. .,Department of Pharmacology, University of Texas Health at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
12
|
Pergande MR, Amoroso VG, Nguyen TTA, Li W, Vice E, Park TJ, Cologna SM. PPARα and PPARγ Signaling Is Enhanced in the Brain of the Naked Mole-Rat, a Mammal that Shows Intrinsic Neuroprotection from Oxygen Deprivation. J Proteome Res 2021; 20:4258-4271. [PMID: 34351155 DOI: 10.1021/acs.jproteome.1c00131] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Naked mole-rats (NMRs) are a long-lived animal that do not develop age-related diseases including neurodegeneration and cancer. Additionally, NMRs have a profound ability to consume reactive oxygen species (ROS) and survive long periods of oxygen deprivation. Here, we evaluated the unique proteome across selected brain regions of NMRs at different ages. Compared to mice, we observed numerous differentially expressed proteins related to altered mitochondrial function in all brain regions, suggesting that the mitochondria in NMRs may have adapted to compensate for energy demands associated with living in a harsh, underground environment. Keeping in mind that ROS can induce polyunsaturated fatty acid peroxidation under periods of neuronal stress, we investigated docosahexaenoic acid (DHA) and arachidonic acid (AA) peroxidation under oxygen-deprived conditions and observed that NMRs undergo DHA and AA peroxidation to a far less extent compared to mice. Further, our proteomic analysis also suggested enhanced peroxisome proliferator-activated receptor (PPAR)-retinoid X receptor (RXR) activation in NMRs via the PPARα-RXR and PPARγ-RXR complexes. Correspondingly, we present several lines of evidence supporting PPAR activation, including increased eicosapetenoic and omega-3 docosapentaenoic acid, as well as an upregulation of fatty acid-binding protein 3 and 4, known transporters of omega-3 fatty acids and PPAR activators. These results suggest enhanced PPARα and PPARγ signaling as a potential, innate neuroprotective mechanism in NMRs.
Collapse
Affiliation(s)
- Melissa R Pergande
- Department of Chemistry, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| | - Vince G Amoroso
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| | - Thu T A Nguyen
- Department of Chemistry, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| | - Wenping Li
- Department of Chemistry, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| | - Emily Vice
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| | - Thomas J Park
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois 60607, United States.,Laboratory for Integrative Neuroscience, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| | - Stephanie M Cologna
- Department of Chemistry, University of Illinois at Chicago, Chicago, Illinois 60607, United States.,Laboratory for Integrative Neuroscience, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| |
Collapse
|
13
|
Aman Y, Schmauck-Medina T, Hansen M, Morimoto RI, Simon AK, Bjedov I, Palikaras K, Simonsen A, Johansen T, Tavernarakis N, Rubinsztein DC, Partridge L, Kroemer G, Labbadia J, Fang EF. Autophagy in healthy aging and disease. NATURE AGING 2021; 1:634-650. [PMID: 34901876 PMCID: PMC8659158 DOI: 10.1038/s43587-021-00098-4] [Citation(s) in RCA: 541] [Impact Index Per Article: 180.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 07/07/2021] [Indexed: 12/12/2022]
Abstract
Autophagy is a fundamental cellular process that eliminates molecules and subcellular elements, including nucleic acids, proteins, lipids and organelles, via lysosome-mediated degradation to promote homeostasis, differentiation, development and survival. While autophagy is intimately linked to health, the intricate relationship among autophagy, aging and disease remains unclear. This Review examines several emerging features of autophagy and postulates how they may be linked to aging as well as to the development and progression of disease. In addition, we discuss current preclinical evidence arguing for the use of autophagy modulators as suppressors of age-related pathologies such as neurodegenerative diseases. Finally, we highlight key questions and propose novel research avenues that will likely reveal new links between autophagy and the hallmarks of aging. Understanding the precise interplay between autophagy and the risk of age-related pathologies across organisms will eventually facilitate the development of clinical applications that promote long-term health.
Collapse
Affiliation(s)
- Yahyah Aman
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog, Norway
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK
- These authors contributed equally: Yahyah Aman, Tomas Schmauck-Medina
| | - Tomas Schmauck-Medina
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog, Norway
- These authors contributed equally: Yahyah Aman, Tomas Schmauck-Medina
| | - Malene Hansen
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Richard I. Morimoto
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, IL, USA
| | | | - Ivana Bjedov
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK
- UCL Cancer Institute, University College London, London, UK
| | - Konstantinos Palikaras
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Anne Simonsen
- Department of Molecular Medicine, Institute of Basic Medical Sciences and Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, The University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, Oslo, Norway
| | - Terje Johansen
- Molecular Cancer Research Group, Institute of Medical Biology, University of Tromsø–The Arctic University of Norway, Tromsø, Norway
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology–Hellas, Heraklion, Greece
- Department of Basic Sciences, School of Medicine, University of Crete, Heraklion, Greece
| | - David C. Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge, UK
| | - Linda Partridge
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK
- Department of Biological Mechanisms of Ageing, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue contre le Cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China
- Karolinska Institute, Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
| | - John Labbadia
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Evandro F. Fang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog, Norway
- The Norwegian Centre on Healthy Ageing (NO-Age), Oslo, Norway
| |
Collapse
|
14
|
Zia A, Pourbagher-Shahri AM, Farkhondeh T, Samarghandian S. Molecular and cellular pathways contributing to brain aging. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2021; 17:6. [PMID: 34118939 PMCID: PMC8199306 DOI: 10.1186/s12993-021-00179-9] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/27/2021] [Indexed: 12/12/2022]
Abstract
Aging is the leading risk factor for several age-associated diseases such as neurodegenerative diseases. Understanding the biology of aging mechanisms is essential to the pursuit of brain health. In this regard, brain aging is defined by a gradual decrease in neurophysiological functions, impaired adaptive neuroplasticity, dysregulation of neuronal Ca2+ homeostasis, neuroinflammation, and oxidatively modified molecules and organelles. Numerous pathways lead to brain aging, including increased oxidative stress, inflammation, disturbances in energy metabolism such as deregulated autophagy, mitochondrial dysfunction, and IGF-1, mTOR, ROS, AMPK, SIRTs, and p53 as central modulators of the metabolic control, connecting aging to the pathways, which lead to neurodegenerative disorders. Also, calorie restriction (CR), physical exercise, and mental activities can extend lifespan and increase nervous system resistance to age-associated neurodegenerative diseases. The neuroprotective effect of CR involves increased protection against ROS generation, maintenance of cellular Ca2+ homeostasis, and inhibition of apoptosis. The recent evidence about the modem molecular and cellular methods in neurobiology to brain aging is exhibiting a significant potential in brain cells for adaptation to aging and resistance to neurodegenerative disorders.
Collapse
Affiliation(s)
- Aliabbas Zia
- Department of Biochemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Ali Mohammad Pourbagher-Shahri
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences (BUMS), 9717853577 Birjand, Iran
| | - Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
- Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Samarghandian
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
15
|
Boughey H, Jurga M, El-Khamisy SF. DNA Homeostasis and Senescence: Lessons from the Naked Mole Rat. Int J Mol Sci 2021; 22:ijms22116011. [PMID: 34199458 PMCID: PMC8199619 DOI: 10.3390/ijms22116011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/28/2021] [Accepted: 05/31/2021] [Indexed: 12/13/2022] Open
Abstract
As we age, our bodies accrue damage in the form of DNA mutations. These mutations lead to the generation of sub-optimal proteins, resulting in inadequate cellular homeostasis and senescence. The build-up of senescent cells negatively affects the local cellular micro-environment and drives ageing associated disease, including neurodegeneration. Therefore, limiting the accumulation of DNA damage is essential for healthy neuronal populations. The naked mole rats (NMR) are from eastern Africa and can live for over three decades in chronically hypoxic environments. Despite their long lifespan, NMRs show little to no biological decline, neurodegeneration, or senescence. Here, we discuss molecular pathways and adaptations that NMRs employ to maintain genome integrity and combat the physiological and pathological decline in organismal function.
Collapse
Affiliation(s)
- Harvey Boughey
- The Healthy Lifespan Institute and the Institute of Neuroscience, University of Sheffield, Sheffield S10 2TN, UK;
| | - Mateusz Jurga
- The Institute of Cancer Therapeutics, University of Bradford, Bradford BD7 1DP, UK;
| | - Sherif F. El-Khamisy
- The Healthy Lifespan Institute and the Institute of Neuroscience, University of Sheffield, Sheffield S10 2TN, UK;
- The Institute of Cancer Therapeutics, University of Bradford, Bradford BD7 1DP, UK;
- Correspondence: ; Tel.: +44-(0)-114-2222-791; Fax: +44-(0)-114-222-2850
| |
Collapse
|
16
|
Pras A, Nollen EAA. Regulation of Age-Related Protein Toxicity. Front Cell Dev Biol 2021; 9:637084. [PMID: 33748125 PMCID: PMC7973223 DOI: 10.3389/fcell.2021.637084] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 02/10/2021] [Indexed: 12/23/2022] Open
Abstract
Proteome damage plays a major role in aging and age-related neurodegenerative diseases. Under healthy conditions, molecular quality control mechanisms prevent toxic protein misfolding and aggregation. These mechanisms include molecular chaperones for protein folding, spatial compartmentalization for sequestration, and degradation pathways for the removal of harmful proteins. These mechanisms decline with age, resulting in the accumulation of aggregation-prone proteins that are harmful to cells. In the past decades, a variety of fast- and slow-aging model organisms have been used to investigate the biological mechanisms that accelerate or prevent such protein toxicity. In this review, we describe the most important mechanisms that are required for maintaining a healthy proteome. We describe how these mechanisms decline during aging and lead to toxic protein misassembly, aggregation, and amyloid formation. In addition, we discuss how optimized protein homeostasis mechanisms in long-living animals contribute to prolonging their lifespan. This knowledge might help us to develop interventions in the protein homeostasis network that delay aging and age-related pathologies.
Collapse
Affiliation(s)
| | - Ellen A. A. Nollen
- Laboratory of Molecular Neurobiology of Ageing, European Research Institute for the Biology of Ageing, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
17
|
Kapetanou M, Nespital T, Tain LS, Pahl A, Partridge L, Gonos ES. FoxO1 Is a Novel Regulator of 20S Proteasome Subunits Expression and Activity. Front Cell Dev Biol 2021; 9:625715. [PMID: 33634126 PMCID: PMC7901890 DOI: 10.3389/fcell.2021.625715] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
Proteostasis collapses during aging resulting, among other things, in the accumulation of damaged and aggregated proteins. The proteasome is the main cellular proteolytic system and plays a fundamental role in the maintenance of protein homeostasis. Our previous work has demonstrated that senescence and aging are related to a decline in proteasome content and activities, while its activation extends lifespan in vitro and in vivo in various species. However, the mechanisms underlying this age-related decline of proteasome function and the down-regulation in expression of its subunits remain largely unclear. Here, we demonstrate that the Forkhead box-O1 (FoxO1) transcription factor directly regulates the expression of a 20S proteasome catalytic subunit and, hence, proteasome activity. Specifically, we demonstrate that knockout of FoxO1, but not of FoxO3, in mice severely impairs proteasome activity in several tissues, while depletion of IRS1 enhances proteasome function. Importantly, we show that FoxO1 directly binds on the promoter region of the rate-limiting catalytic β5 proteasome subunit to regulate its expression. In summary, this study reveals the direct role of FoxO factors in the regulation of proteasome function and provides new insight into how FoxOs affect proteostasis and, in turn, longevity.
Collapse
Affiliation(s)
- Marianna Kapetanou
- Laboratory of Molecular and Cellular Aging, Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Tobias Nespital
- Department of Biological Mechanisms of Ageing, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Luke S Tain
- Department of Biological Mechanisms of Ageing, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Andre Pahl
- Department of Biological Mechanisms of Ageing, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Linda Partridge
- Department of Biological Mechanisms of Ageing, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Efstathios S Gonos
- Laboratory of Molecular and Cellular Aging, Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| |
Collapse
|
18
|
Adult Neural Plasticity in Naked Mole-Rats: Implications of Fossoriality, Longevity and Sociality on the Brain's Capacity for Change. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1319:105-135. [PMID: 34424514 DOI: 10.1007/978-3-030-65943-1_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Naked mole-rats (Heterocephalus glaber) are small African rodents that have many unique behavioral and physiological adaptations well-suited for testing hypotheses about mammalian neural plasticity. In this chapter, we focus on three features of naked mole-rat biology and how they impact neural plasticity in this species: (1) their fossorial lifestyle, (2) their extreme longevity with a lack of demonstrable senescence, and (3) their unusual social structure. Critically, each of these features requires some degree of biological flexibility. First, their fossorial habitat situates them in an environment with characteristics to which the central nervous system is particularly sensitive (e.g., oxygen content, photoperiod, spatial complexity). Second, their long lifespan requires adaptations to combat senescence and declines in neural functioning. Finally, their extreme reproductive skew and sustained ability for release from reproductive suppression indicates remarkable neural sensitivity to the sociosexual environment that is distinct from chronological age. These three features of naked mole-rat life are not mutually exclusive, but they do each offer unique considerations for the possibilities, constraints, and mechanisms associated with adult neural plasticity.
Collapse
|
19
|
Ward JM, Cartoceti AN, Delaney MA. Brain Lesions in Aging Zoo-Housed Naked Mole-Rats ( Heterocephalus glaber). Vet Pathol 2020; 58:142-146. [PMID: 33205701 DOI: 10.1177/0300985820969982] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Naked mole-rats (NMRs) are common in the managed care of zoos and valuable models for aging research. Limited information on NMR neuropathology is available despite many studies regarding their aging physiology. Histologic sections of brain from 27 adult (5-27 years old) NMRs from 2 zoos were reviewed to determine presence or absence of lesions associated with advanced age in humans and other mammals. A majority (23/27; 85%) of NMR brains had cerebral cortical neuronal changes with rounded or angular neurons, cytoplasmic vacuoles containing pale yellow pigment, periodic acid-Schiff (PAS)-positive granules and green autofluorescence, compatible with lipofuscinosis. Less severe lesions were present in cerebellar Purkinje cells, medulla, and hippocampal neurons. The hypothalamic neuropil of all NMRs had scattered variably sized PAS-positive granules and 10 (37%) had larger round bodies consistent with corpora amylacea. The youngest NMRs, 5 to 7 years old, generally had minimal or no cerebrocortical lesions. Further studies will help understand brain aging in this long-lived species.
Collapse
Affiliation(s)
| | - Andrew N Cartoceti
- National Zoological Park, 53587Smithsonian Institution, Washington, DC, USA
| | - Martha A Delaney
- Zoological Pathology Program, College of Veterinary Medicine, 14589University of Illinois, Brookfield, IL, USA
| |
Collapse
|
20
|
Du Z, Chakrabarti S, Kulaberoglu Y, Smith ESJ, Dobson CM, Itzhaki LS, Kumita JR. Probing the unfolded protein response in long-lived naked mole-rats. Biochem Biophys Res Commun 2020; 529:1151-1157. [PMID: 32819579 PMCID: PMC7453385 DOI: 10.1016/j.bbrc.2020.06.118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 06/23/2020] [Indexed: 11/08/2022]
Abstract
The long-living naked mole-rat (NMR) shows negligible senescence and resistance to age-associated diseases. Recent evidence, based on protein-level assays, suggests that enhanced protein homeostasis machinery contributes to NMR stress-resistance and longevity. Here, we develop NMR-specific, transcriptional assays for measuring the unfolded protein response (UPR), a component of ER proteostasis. By varying doses and response times of pharmacological ER stressors applied to NMR kidney fibroblasts, we probe the NMR UPR in detail, demonstrating that NMR fibroblasts have a higher UPR activation threshold compared to mouse fibroblasts under mild ER-stress induction; whereas temporal analysis reveals that severe ER-stress induction results in no comparative differences. Probing NMR UPR activation with our robust assays may lead to insights into the proteostasis and ageing relationship.
Collapse
Affiliation(s)
- Zhen Du
- Department of Pharmacology, University of Cambridge, Tennis Court Rd., Cambridge, CB2 1PD, UK; Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Lensfield Rd., Cambridge, CB2 1EW, UK
| | - Sampurna Chakrabarti
- Department of Pharmacology, University of Cambridge, Tennis Court Rd., Cambridge, CB2 1PD, UK
| | - Yavuz Kulaberoglu
- Department of Pharmacology, University of Cambridge, Tennis Court Rd., Cambridge, CB2 1PD, UK; UCL Institute of Healthy Ageing, Darwin Building, 104 Gower St, Bloomsbury, London, WC1E 6AD, UK
| | - Ewan St John Smith
- Department of Pharmacology, University of Cambridge, Tennis Court Rd., Cambridge, CB2 1PD, UK
| | - Christopher M Dobson
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Lensfield Rd., Cambridge, CB2 1EW, UK
| | - Laura S Itzhaki
- Department of Pharmacology, University of Cambridge, Tennis Court Rd., Cambridge, CB2 1PD, UK.
| | - Janet R Kumita
- Department of Pharmacology, University of Cambridge, Tennis Court Rd., Cambridge, CB2 1PD, UK; Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Lensfield Rd., Cambridge, CB2 1EW, UK.
| |
Collapse
|
21
|
Sunchu B, Riordan RT, Yu Z, Almog I, Dimas-Munoz J, Drake AC, Perez VI. Aggresome-Like Formation Promotes Resistance to Proteotoxicity in Cells from Long-Lived Species. J Gerontol A Biol Sci Med Sci 2020; 75:1439-1447. [PMID: 32515471 PMCID: PMC7357592 DOI: 10.1093/gerona/glaa069] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Indexed: 11/13/2022] Open
Abstract
The capacity of cells to maintain proteostasis declines with age, causing rapid accumulation of damaged proteins and protein aggregates, which plays an important role in age-related disease etiology. While our group and others have identified that proteostasis is enhanced in long-lived species, there are no data on whether this leads to better resistance to proteotoxicity. We compared the sensitivity of cells from long- (naked mole rat [NMR]) and short- (Mouse) lived species to proteotoxicity, by measuring the survival of fibroblasts under polyglutamine (polyQ) toxicity, a well-established model of protein aggregation. Additionally, to evaluate the contribution of proteostatic mechanisms to proteotoxicity resistance, we down-regulated a key protein of each mechanism (autophagy-ATG5; ubiquitin-proteasome-PSMD14; and chaperones-HSP27) in NMR fibroblasts. Furthermore, we analyzed the formation and subcellular localization of inclusions in long- and short-lived species. Here, we show that fibroblasts from long-lived species are more resistant to proteotoxicity than their short-lived counterparts. Surprisingly, this does not occur because the NMR cells have less polyQ82 protein aggregates, but rather they have an enhanced capacity to handle misfolded proteins and form protective perinuclear and aggresome-like inclusions. All three proteostatic mechanisms contribute to this resistance to polyQ toxicity but autophagy has the greatest effect. Overall, our data suggest that the resistance to proteotoxicity observed in long-lived species is not due to a lower level of protein aggregates but rather to enhanced handling of the protein aggregates through the formation of aggresome-like inclusions, a well-recognized protective mechanism against proteotoxicty.
Collapse
Affiliation(s)
- Bharath Sunchu
- Linus Pauling Institute, Oregon State University, Corvallis
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis
| | - Ruben T Riordan
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis
| | - Zhen Yu
- Linus Pauling Institute, Oregon State University, Corvallis
| | - Ido Almog
- Linus Pauling Institute, Oregon State University, Corvallis
| | - Jovita Dimas-Munoz
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis
| | - Andrew C Drake
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis
| | - Viviana I Perez
- Linus Pauling Institute, Oregon State University, Corvallis
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis
| |
Collapse
|
22
|
Shepard A, Kissil JL. The use of non-traditional models in the study of cancer resistance-the case of the naked mole rat. Oncogene 2020; 39:5083-5097. [PMID: 32535616 DOI: 10.1038/s41388-020-1355-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/15/2020] [Accepted: 06/03/2020] [Indexed: 12/16/2022]
Abstract
Non-traditional model organisms are typically defined as any model the deviates from the typical laboratory animals, such as mouse, rat, and worm. These models are becoming increasingly important in human disease research, such as cancer, as they often display unusual biological features. Naked mole rats (NMRs) are currently one of the most popular non-traditional model, particularly in the longevity and cancer research fields. NMRs display an exceptionally long lifespan (~30 years), yet have been observed to display a low incidence of cancer, making them excellent candidates for understanding endogenous cancer resistance mechanisms. Over the past decade, many potential resistance mechanisms have been characterized. These include unique biological mechanisms involved in genome stability, protein stability, oxidative metabolism, and other cellular mechanisms such as cell cycle regulation and senescence. This review aims to summarize the many identified cancer resistance mechanisms to understand some of the main hypotheses that have thus far been generated. Many of these proposed mechanisms remain to be fully characterized or confirmed in vivo, giving the field a direction to grow and further understand the complex biology displayed by the NMR.
Collapse
Affiliation(s)
- Alyssa Shepard
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, 33458, USA
| | - Joseph L Kissil
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, 33458, USA.
| |
Collapse
|
23
|
Lee BP, Smith M, Buffenstein R, Harries LW. Negligible senescence in naked mole rats may be a consequence of well-maintained splicing regulation. GeroScience 2020; 42:633-651. [PMID: 31927681 PMCID: PMC7205774 DOI: 10.1007/s11357-019-00150-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 12/27/2019] [Indexed: 02/07/2023] Open
Abstract
Naked mole-rats (NMRs) have amongst the longest lifespans relative to body size of any known, non-volant mammalian species. They also display an enhanced stress resistance phenotype, negligible senescence and very rarely are they burdened with chronic age-related diseases. Alternative splicing (AS) dysregulation is emerging as a potential driver of senescence and ageing. We hypothesised that the expression of splicing factors, important regulators of patterns of AS, may differ in NMRs when compared to other species with relatively shorter lifespans. We designed assays specific to NMR splicing regulatory factors and also to a panel of pre-selected brain-expressed genes known to demonstrate senescence-related alterations in AS in other species, and measured age-related changes in the transcript expression levels of these using embryonic and neonatal developmental stages through to extreme old age in NMR brain samples. We also compared splicing factor expression in both young mouse and NMR spleen and brain samples. Both NMR tissues showed approximately double the expression levels observed in tissues from similarly sized mice. Furthermore, contrary to observations in other species, following a brief period of labile expression in early life stages, adult NMR splicing factors and patterns of AS for functionally relevant brain genes remained remarkably stable for at least two decades. These findings are consistent with a model whereby the conservation of splicing regulation and stable patterns of AS may contribute to better molecular stress responses and the avoidance of senescence in NMRs, contributing to their exceptional lifespan and prolonged healthspan.
Collapse
Affiliation(s)
- B P Lee
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Barrack Road, Exeter, EX2 5DW, UK
| | - M Smith
- Calico Life Sciences LLC, 1170 Veterans Blvd., South San Francisco, CA, 94080, USA
| | - R Buffenstein
- Calico Life Sciences LLC, 1170 Veterans Blvd., South San Francisco, CA, 94080, USA.
| | - L W Harries
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Barrack Road, Exeter, EX2 5DW, UK.
| |
Collapse
|
24
|
Buffenstein R, Lewis KN, Gibney PA, Narayan V, Grimes KM, Smith M, Lin TD, Brown-Borg HM. Probing Pedomorphy and Prolonged Lifespan in Naked Mole-Rats and Dwarf Mice. Physiology (Bethesda) 2020; 35:96-111. [DOI: 10.1152/physiol.00032.2019] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Pedomorphy, maintenance of juvenile traits throughout life, is most pronounced in extraordinarily long-lived naked mole-rats. Many of these traits (e.g., slow growth rates, low hormone levels, and delayed sexual maturity) are shared with spontaneously mutated, long-lived dwarf mice. Although some youthful traits likely evolved as adaptations to subterranean habitats (e.g., thermolability), the nature of these intrinsic pedomorphic features may also contribute to their prolonged youthfulness, longevity, and healthspan.
Collapse
Affiliation(s)
| | | | - Patrick A. Gibney
- Calico Life Sciences LLC, South San Francisco, California
- Department of Food Science, College of Agriculture and Life Sciences, Stocking Hall, Cornell University, Ithaca, New York
| | - Vikram Narayan
- Calico Life Sciences LLC, South San Francisco, California
| | - Kelly M. Grimes
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Megan Smith
- Calico Life Sciences LLC, South San Francisco, California
| | - Tzuhua D. Lin
- Calico Life Sciences LLC, South San Francisco, California
| | - Holly M. Brown-Borg
- Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota
| |
Collapse
|
25
|
Popov NA, Skulachev VP. Neotenic Traits in Heterocephalus glaber and Homo sapiens. BIOCHEMISTRY (MOSCOW) 2019; 84:1484-1489. [DOI: 10.1134/s0006297919120071] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
26
|
Di Domenico F, Zuliani I, Tramutola A. Shining a light on defective autophagy by proteomics approaches: implications for neurodegenerative illnesses. Expert Rev Proteomics 2019; 16:951-964. [PMID: 31709850 DOI: 10.1080/14789450.2019.1691919] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Autophagy is one of the most conserved clearance systems through which eukaryotes manage to handle dysfunctional and excess organelles and macromolecules. This catabolic process has not only a role in the maintenance of basal turnover of cellular components, but it is also essential in cells adaptation to stress conditions. In the last decades, defects in autophagic machinery have been identified as a feature in neurodegenerative diseases. In this context, mass spectrometry-based proteomics has become an important tool in the comprehensive analysis of proteins involved in the autophagic flux.Area covered: In this review, we discuss recent contributions of proteomic techniques in the study of defective autophagy related to neurodegenerative illness. Particular emphasis is given to the identification of i) shared autophagic markers between different disorders, which support common pathological mechanisms; ii) unique autophagic signature, which could aid to discriminate among diseases.Expert opinion: Proteomic approaches are valuable in the identification of alterations of components to the autophagic process at different steps of the process. The investigation of autophagic defects associated with neurological disorders is crucial in order to unravel all the potential mechanism leading to neurodegeneration and propose effective therapeutic strategies targeting autophagy.
Collapse
Affiliation(s)
- Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Ilaria Zuliani
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| | - Antonella Tramutola
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
27
|
Jalali Z, Parvaz N. Molecular evolution of autophagy rate-limiting factor LAMP2 in placental mammals. Gene 2019; 727:144231. [PMID: 31707000 DOI: 10.1016/j.gene.2019.144231] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/08/2019] [Accepted: 10/27/2019] [Indexed: 01/24/2023]
Abstract
Autophagy is the cellular process of removal of misfolded or damaged macromolecules and organelles. Experimental studies have demonstrated autophagy as a major mechanism of lifespan extension in long-lived mammals such as bats and mole rat rodents. Moreover, the role of this biological process has been well documented in protection against age-associated diseases and viral infection. However, studies on the molecular adaptive changes of autophagy factors during evolution are scarce. Here, we conducted a bioinformatics study of the molecular evolution of the Lysosomal Associated Membrane Protein 2 (LAMP2), as a rate-limiting factor in the lysosomal degradation stage of autophagy (the communal step of two of autophagy types: macroautophagy and chaperone-mediated). Analyzing LAMP2 across placental mammals, our phylogenetic-based maximum likelihood analyses indicate that the majority of the coding sites undergo purifying selection. However, around 27% of sites display a relaxation of purifying constraints (average ω = 0.42128), among which, 14 particular sites undergo positive selection (ω > 1). These sites are mostly located in the first luminal domain of LAMP2 (N-domain), with a hotspot region in the 135-144 codons interval. Therefore, the N-domain may account for the functional diversity and regulation of LAMP2. In addition, the identified positive selection sites could act as key regulatory sites in the LAMP2 function. On the other hand, testing the rate of evolution in LAMP2 along different clades of placental mammals revealed a relatively relaxed evolution in LAMP2 along megabats' clade. It is not clear yet whether an expedited evolution of LAMP2 in megabats has contributed to their reported up-regulation of autophagy. Finally, our data indicate positive selection sites along the ancestral branch of the clades of rodents, mouse-related rodents, and mole-rats; and suggest the potentially important regulatory role of these sites in LAMP2. Identifying the residues under positive selection, our findings pave the way for future experimental investigations to define how these selective substitutions have functionally affected autophagy.
Collapse
Affiliation(s)
- Zahra Jalali
- Non-Communicable Diseases Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Clinical Biochemistry, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | - Najmeh Parvaz
- Department of Clinical Biochemistry, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
28
|
Butterfield DA. Phosphoproteomics of Alzheimer disease brain: Insights into altered brain protein regulation of critical neuronal functions and their contributions to subsequent cognitive loss. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2031-2039. [PMID: 31167728 PMCID: PMC6602546 DOI: 10.1016/j.bbadis.2018.08.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 08/22/2018] [Accepted: 08/25/2018] [Indexed: 02/07/2023]
Abstract
Alzheimer disease (AD) is the major locus of dementia worldwide. In the USA there are nearly 6 million persons with this disorder, and estimates of 13-20 million AD cases in the next three decades. The molecular bases for AD remain unknown, though processes involving amyloid beta-peptide as small oligomeric forms are gaining attention as known agents to both lead to oxidative stress and synaptic dysfunction associated with cognitive dysfunction in AD and its earlier forms, including amnestic mild cognitive impairment (MCI) and possibly preclinical Alzheimer disease (PCAD). Altered brain protein phosphorylation is a hallmark of AD, and phosphoproteomics offers an opportunity to identify these altered phosphoproteins in order to gain more insights into molecular mechanisms of neuronal dysfunction and death that lead to cognitive loss. This paper reviews what, to this author, are believed to be the known phosphoproteomics studies related to in vitro and in vivo models of AD as well as phosphoproteomics studies of brains from subjects with AD, and in at least one case in MCI and PCAD as well. The results of this review are discussed with relevance to new insights into AD brain protein dysregulation in critical neuronal functions and to potential therapeutic targets to slow, or in favorable cases, halt progression of this dementing disorder that affects millions of persons and their families worldwide.
Collapse
Affiliation(s)
- D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA.
| |
Collapse
|
29
|
Viltard M, Durand S, Pérez-Lanzón M, Aprahamian F, Lefevre D, Leroy C, Madeo F, Kroemer G, Friedlander G. The metabolomic signature of extreme longevity: naked mole rats versus mice. Aging (Albany NY) 2019; 11:4783-4800. [PMID: 31346149 PMCID: PMC6682510 DOI: 10.18632/aging.102116] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 07/16/2019] [Indexed: 04/11/2023]
Abstract
The naked mole-rat (Heterocephalus glaber) is characterized by a more than tenfold higher life expectancy compared to another rodent species of the same size, namely, the laboratory mouse (Mus musculus). We used mass spectrometric metabolomics to analyze circulating plasma metabolites in both species at different ages. Interspecies differences were much more pronounced than age-associated alterations in the metabolome. Such interspecies divergences affected multiple metabolic pathways involving amino, bile and fatty acids as well as monosaccharides and nucleotides. The most intriguing metabolites were those that had previously been linked to pro-health and antiaging effects in mice and that were significantly increased in the long-lived rodent compared to its short-lived counterpart. This pattern applies to α-tocopherol (also known as vitamin E) and polyamines (in particular cadaverine, N8-acetylspermidine and N1,N8-diacetylspermidine), all of which were more abundant in naked mole-rats than in mice. Moreover, the age-associated decline in spermidine and N1-acetylspermidine levels observed in mice did not occur, or is even reversed (in the case of N1-acetylspermidine) in naked mole-rats. In short, the present metabolomics analysis provides a series of testable hypotheses to explain the exceptional longevity of naked mole-rats.
Collapse
Affiliation(s)
- Mélanie Viltard
- Fondation pour la Recherche en Physiologie, Brussels, Belgium
| | - Sylvère Durand
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Equipe Labellisée par la Ligue contre le Cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
| | - Maria Pérez-Lanzón
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Equipe Labellisée par la Ligue contre le Cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, France
| | - Fanny Aprahamian
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Equipe Labellisée par la Ligue contre le Cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
| | - Deborah Lefevre
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Equipe Labellisée par la Ligue contre le Cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
| | - Christine Leroy
- INSERM UMR_S1151 CNRS UMR8253 Institut Necker-Enfants Malades (INEM), Paris, France
| | - Frank Madeo
- Institute of Molecular Biosciences, University of Graz, NAWI Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Equipe Labellisée par la Ligue contre le Cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Suzhou Institute for Systems Medicine, Chinese Academy of Sciences, Suzhou, China
- Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Gérard Friedlander
- INSERM UMR_S1151 CNRS UMR8253 Institut Necker-Enfants Malades (INEM), Paris, France
- Service de Physiologie et Explorations Fonctionnelles, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France
- Université de Paris - Paris Descartes, Faculté de Médecine, Paris, France
| |
Collapse
|
30
|
Papadopoli D, Boulay K, Kazak L, Pollak M, Mallette FA, Topisirovic I, Hulea L. mTOR as a central regulator of lifespan and aging. F1000Res 2019; 8:F1000 Faculty Rev-998. [PMID: 31316753 PMCID: PMC6611156 DOI: 10.12688/f1000research.17196.1] [Citation(s) in RCA: 220] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/20/2019] [Indexed: 12/17/2022] Open
Abstract
The mammalian/mechanistic target of rapamycin (mTOR) is a key component of cellular metabolism that integrates nutrient sensing with cellular processes that fuel cell growth and proliferation. Although the involvement of the mTOR pathway in regulating life span and aging has been studied extensively in the last decade, the underpinning mechanisms remain elusive. In this review, we highlight the emerging insights that link mTOR to various processes related to aging, such as nutrient sensing, maintenance of proteostasis, autophagy, mitochondrial dysfunction, cellular senescence, and decline in stem cell function.
Collapse
Affiliation(s)
- David Papadopoli
- Gerald Bronfman Department of Oncology, McGill University, 5100 de Maisonneuve Blvd. West, Suite 720, Montréal, QC, H4A 3T2, Canada
- Lady Davis Institute, SMBD JGH, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada
| | - Karine Boulay
- Lady Davis Institute, SMBD JGH, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada
- Maisonneuve-Rosemont Hospital Research Centre, 5415 Assumption Blvd, Montréal, QC, H1T 2M4, Canada
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, CP 6128, Succursale Centre-Ville, Montréal, QC, H3C 3J7, Canada
| | - Lawrence Kazak
- Department of Biochemistry, McGill University, 3655 Promenade Sir William Osler, Montréal, QC, H3G 1Y6, Canada
- Goodman Cancer Research Centre, 1160 Pine Avenue West, Montréal, QC, H3A 1A3, Canada
| | - Michael Pollak
- Gerald Bronfman Department of Oncology, McGill University, 5100 de Maisonneuve Blvd. West, Suite 720, Montréal, QC, H4A 3T2, Canada
- Lady Davis Institute, SMBD JGH, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada
- Goodman Cancer Research Centre, 1160 Pine Avenue West, Montréal, QC, H3A 1A3, Canada
- Department of Experimental Medicine, McGill University, 845 Sherbrooke Street West, Montréal, QC, H3A 0G4, Canada
| | - Frédérick A. Mallette
- Maisonneuve-Rosemont Hospital Research Centre, 5415 Assumption Blvd, Montréal, QC, H1T 2M4, Canada
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, CP 6128, Succursale Centre-Ville, Montréal, QC, H3C 3J7, Canada
- Département de Médecine, Université de Montréal, CP 6128, Succursale Centre-Ville, Montréal, QC, H3C 3J7, Canada
| | - Ivan Topisirovic
- Gerald Bronfman Department of Oncology, McGill University, 5100 de Maisonneuve Blvd. West, Suite 720, Montréal, QC, H4A 3T2, Canada
- Lady Davis Institute, SMBD JGH, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada
- Department of Biochemistry, McGill University, 3655 Promenade Sir William Osler, Montréal, QC, H3G 1Y6, Canada
- Department of Experimental Medicine, McGill University, 845 Sherbrooke Street West, Montréal, QC, H3A 0G4, Canada
| | - Laura Hulea
- Maisonneuve-Rosemont Hospital Research Centre, 5415 Assumption Blvd, Montréal, QC, H1T 2M4, Canada
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, CP 6128, Succursale Centre-Ville, Montréal, QC, H3C 3J7, Canada
- Département de Médecine, Université de Montréal, CP 6128, Succursale Centre-Ville, Montréal, QC, H3C 3J7, Canada
| |
Collapse
|
31
|
Loeffler DA. Influence of Normal Aging on Brain Autophagy: A Complex Scenario. Front Aging Neurosci 2019; 11:49. [PMID: 30914945 PMCID: PMC6421305 DOI: 10.3389/fnagi.2019.00049] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 02/19/2019] [Indexed: 12/12/2022] Open
Abstract
Misfolded proteins are pathological findings in some chronic neurodegenerative disorders including Alzheimer's, Parkinson's, and Huntington's diseases. Aging is a major risk factor for these disorders, suggesting that the mechanisms responsible for clearing misfolded proteins from the brain, the ubiquitin-proteasome system and the autophagy-lysosomal pathway, may decline with age. Although autophagic mechanisms have been found to decrease with age in many experimental models, whether they do so in the brain is unclear. This review examines the literature with regard to age-associated changes in macroautophagy and chaperone-mediated autophagy (CMA) in the central nervous system (CNS). Beclin 1, LC3-II, and the LC3-II/LC3-I ratio have frequently been used to examine changes in macroautophagic activity, while lamp2a and HSPA8 (also known as hsc70) have been used to measure CMA activity. Three gene expression analyses found evidence for an age-related downregulation of macroautophagy in human brain, but no published studies were found of age-related changes in CMA in human brain, although cerebrospinal fluid concentrations of HSPA8 were reported to decrease with age. Most studies of age-related changes in brain autophagy in experimental animals have found age-related declines in macroautophagy, and macroautophagy is necessary for normal lifespan in Caenorhabditis elegans, Drosophila, and mice. However, the few studies of age-related changes in brain CMA in experimental animals have produced conflicting results. Investigations of the influence of aging on macroautophagy in experimental animals in systems other than the CNS have generally found an age-related decrease in Beclin 1, but conflicting results for LC3-II and the LC3-II/LC3-I ratio, while CMA decreases with age in most models. CONCLUSION: while indirect evidence suggests that brain autophagy may decrease with normal aging, this issue has not been investigated sufficiently, particularly in human brain. Measuring autophagic activity in the brain can be challenging because of differences in basal autophagic activity between experimental models, and the inability to include lysosomal inhibitors when measuring the LC3-II/LC3-I ratio in postmortem specimens. If autophagy does decrease in the brain with aging, then pharmacological interventions and/or lifestyle alterations to slow this decline could reduce the risk of developing age-related neurodegenerative disorders.
Collapse
Affiliation(s)
- David A Loeffler
- Beaumont Research Institute, Department of Neurology, Beaumont Health, Royal Oak, MI, United States
| |
Collapse
|
32
|
Ivanova ME, Trubilin VN, Atarshchikov DS, Demchinsky AM, Strelnikov VV, Tanas AS, Orlova OM, Machalov AS, Overchenko KV, Markova TV, Golenkova DM, Anoshkin KI, Volodin IV, Zaletaev DV, Pulin AA, Nadelyaeva II, Kalinkin AI, Barh D. Genetic screening of Russian Usher syndrome patients toward selection for gene therapy. Ophthalmic Genet 2018; 39:706-713. [PMID: 30358468 DOI: 10.1080/13816810.2018.1532527] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Usher syndrome (USH) is heterogeneous in nature and requires genetic test for diagnosis and management. Mutations in USH associated genes are reported in some populations except Russians. Here, we first time represented the mutation spectrum of a Russian USH cohort. METHODS Twenty-eight patients with USH were selected from 3214 patients from Deaf-Blind Support Foundation "Con-nection" during 2014-2016 following the observational study NCT03319524. Complete ophthalmologic, ENT, and vestibular medical tests were done for clinical characterization. NGS, MLPA, and Sanger sequencing were considered for genetic analysis. RESULTS Around 53.57% and 39.28% patients had USH1 and USH2, respectively; 17.85% cases (n = 5/28) had no known mutation. Eleven (73.33%) subjects showed variations in USH1 associated genes MYO7A (72.72%), CDH23 (9.09%), PCDH15 (9.09%), and USH1C (9.09%). Eleven mutations are detected in MYO7A where 54.54% are novel. MYO7A: p.Q18* was most frequent (27.27%) mutation and is associated with early manifestation and most severe clinical picture. Two novel mutations (p.E1301* and c.158-?_318+?del) are detected in PCDH15 gene. Around 90.90% patients suspected to be USH2 are confirmed by genetic testing. Eleven mutations detected in the USH2A gene, where 27.27% were novel. Most common USH2A mutation is p.W3955* (50%) followed by p.E767fs, p.R1653*, and c.8682-9A> G (20% each). CONCLUSION The Russian USH cohort shows both novel and known USH mutations. Clinically the prevalence of USH2 is low (39.28%) and the frequency of MYO7A mutations responsible for USH1B is very high (63.63%, N = 7/11) compared to other cohorts. These seven patients carrying MYO7A mutations are preliminarily eligible for the UshStat® gene therapy.
Collapse
Affiliation(s)
| | - Vladimir N Trubilin
- b Center of Ophthalmology , Federal Medical-Biological Agency State Research Center , Burnasyan, Moscow , Russia
| | | | | | - Vladimir V Strelnikov
- e Federal State Budgetary Institution , Research Centre for Medical Genetics , Moscow , Russia
| | - Alexander S Tanas
- e Federal State Budgetary Institution , Research Centre for Medical Genetics , Moscow , Russia
| | - Olga M Orlova
- b Center of Ophthalmology , Federal Medical-Biological Agency State Research Center , Burnasyan, Moscow , Russia
| | - Anton S Machalov
- f Surdology and Otoneurology Departments , Scientific and Clinical Center for Otorhinolaryngology of FMBA of Russia , Moscow , Russia
| | - Kira V Overchenko
- f Surdology and Otoneurology Departments , Scientific and Clinical Center for Otorhinolaryngology of FMBA of Russia , Moscow , Russia
| | - Tatiana V Markova
- e Federal State Budgetary Institution , Research Centre for Medical Genetics , Moscow , Russia
| | - Daria M Golenkova
- b Center of Ophthalmology , Federal Medical-Biological Agency State Research Center , Burnasyan, Moscow , Russia
| | - Kirill I Anoshkin
- e Federal State Budgetary Institution , Research Centre for Medical Genetics , Moscow , Russia
| | - Ilya V Volodin
- e Federal State Budgetary Institution , Research Centre for Medical Genetics , Moscow , Russia
| | - Dmitry V Zaletaev
- g Laboratory of Medical Genetics, Institute of Molecular Medicine , I. M. Sechenov First Moscow State Medical University , Moscow , Russia
| | - Andrey A Pulin
- h Laboratory of Cell Biology and Developmental Pathology , Federal State Budgetary Scientific Institution "Institute of General Pathology and Pathophysiology" , Moscow , Russia
| | - Irina I Nadelyaeva
- i Federal State Budget Institution of Higher Education , A.I. Yevdokimov Moscow State University of Medicine and Dentistry, The Ministry of Health Care of the Russia
| | - Alexey I Kalinkin
- e Federal State Budgetary Institution , Research Centre for Medical Genetics , Moscow , Russia.,g Laboratory of Medical Genetics, Institute of Molecular Medicine , I. M. Sechenov First Moscow State Medical University , Moscow , Russia
| | - Debmalya Barh
- j Center for Genomics and Applied Gene Technology , Institute of Integrative Omics and Applied Biotechnology (IIOAB) , Nonakuri, Purba Medinipur , West Bengal , India.,k Division of Bioinformatics and Computational Genomics , NITTE University Center for Science Education and Research (NUCSER), NITTE (Deemed to be University) , Mangaluru , Karnataka , India
| |
Collapse
|
33
|
11β-HSD1 Inhibition by RL-118 Promotes Autophagy and Correlates with Reduced Oxidative Stress and Inflammation, Enhancing Cognitive Performance in SAMP8 Mouse Model. Mol Neurobiol 2018; 55:8904-8915. [PMID: 29611102 DOI: 10.1007/s12035-018-1026-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 03/20/2018] [Indexed: 01/14/2023]
Abstract
Elevated glucocorticoid (GC) exposure is widely accepted as a key factor in the age-related cognitive decline in rodents and humans. 11β-HSD1 is a key enzyme in the GCs pathway, catalyzing the conversion of 11β-dehydrocorticosterone to corticosterone in mice, with possible implications in neurodegenerative processes and cognitive impairment. Here, we determined the effect of a new 11β-HSD1 inhibitor, RL-118, administered to 12-month-old senescence-accelerated mouse-prone 8 (SAMP8) mice with neuropathological AD-like hallmarks and widely used as a rodent model of cognitive dysfunction. Behavioral tests (open field and object location) and neurodegeneration molecular markers were studied. After RL-118 treatment, increased locomotor activity and cognitive performance were found. Likewise, we found changes in hippocampal autophagy markers such as Beclin1, LC3B, AMPKα, and mTOR, indicating a progression in the autophagy process. In line with autophagy increase, a diminution in phosphorylated tau species (Ser 396 and Ser 404) jointly with an increase in ADAM10 and sAPPα indicated that an improvement in removing the abnormal proteins by autophagy might be implicated in the neuroprotective role of the 11β-HSD1 inhibitor. In addition, gene expression of oxidative stress (OS) and inflammatory markers, such as Hmox1, Aldh2, Il-1β, and Ccl3, were reduced in old treated mice in comparison to that of the control group. Consistent with this, we further demonstrate a significant correlation with autophagy markers and cognitive improvement and significant inverse correlation with autophagy, OS, and neuroinflammation markers. We concluded that inhibition of 11β-HSD1 by RL-118 prevented neurodegenerative processes and cognitive decline, acting on autophagy process, being an additional neuroprotective mechanism not described previously.
Collapse
|
34
|
Zhao S, Li L, Wang S, Yu C, Xiao B, Lin L, Cong W, Cheng J, Yang W, Sun W, Cui S. H2O2 treatment or serum deprivation induces autophagy and apoptosis in naked mole-rat skin fibroblasts by inhibiting the PI3K/Akt signaling pathway. Oncotarget 2018; 7:84839-84850. [PMID: 27863375 PMCID: PMC5356702 DOI: 10.18632/oncotarget.13321] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 10/28/2016] [Indexed: 01/07/2023] Open
Abstract
Naked mole-rats (NMR; Heterocephalus glaber) display extreme longevity and resistance to cancer. Here, we examined whether autophagy contributes to the longevity of NMRs by assessing the effects of the PI3K/Akt pathway inhibitor LY294002 and the autophagy inhibitor chloroquine (CQ) on autophagy and apoptosis in NMR skin fibroblasts. Serum starvation, H2O2 treatment, and LY294002 treatment all increased the LC3-II/LC3-I ratio and numbers of double-membraned autophagosomes and autophagic vacuoles, and decreased levels of p70S6K, p-AktSer473, and p-AktThr308. By contrast, CQ treatment decreased p70S6K, AktSer473, and AktThr308 levels. The Bax/Bcl-2 ratio increased after 12 h of exposure to LY294002 or CQ. These data show that inhibiting the Akt pathway promotes autophagy and apoptosis in NMR skin fibroblasts. Furthermore, LY294002 or CQ treatment decreased caspase-3, p53, and HIF1-α levels, suggesting that serum starvation or H2O2 treatment increase autophagy and apoptosis in NMR skin fibroblasts by inhibiting the PI3K/Akt pathway. CQ-induced inhibition of late autophagy stages also prevented Akt activation and induced apoptosis. Finally, the HIF-1α and p53 pathways were involved in serum starvation- or H2O2-induced autophagy in NMR skin fibroblasts.
Collapse
Affiliation(s)
- Shanmin Zhao
- Laboratory Animal Centre, Second Military Medical University, Shanghai, China
| | - Li Li
- Department of Training, Second Military Medical University, Shanghai, China
| | - Shiyong Wang
- Informatization Office, Second Military Medical University, Shanghai, China
| | - Chenlin Yu
- Laboratory Animal Centre, Second Military Medical University, Shanghai, China
| | - Bang Xiao
- Laboratory Animal Centre, Second Military Medical University, Shanghai, China
| | - Lifang Lin
- Laboratory Animal Centre, Second Military Medical University, Shanghai, China
| | - Wei Cong
- Laboratory Animal Centre, Second Military Medical University, Shanghai, China
| | - Jishuai Cheng
- Laboratory Animal Centre, Second Military Medical University, Shanghai, China
| | - Wenjing Yang
- Laboratory Animal Centre, Second Military Medical University, Shanghai, China
| | - Wei Sun
- Laboratory Animal Centre, Second Military Medical University, Shanghai, China
| | - Shufang Cui
- Laboratory Animal Centre, Second Military Medical University, Shanghai, China
| |
Collapse
|
35
|
Novel treatment strategies for chronic kidney disease: insights from the animal kingdom. Nat Rev Nephrol 2018; 14:265-284. [PMID: 29332935 DOI: 10.1038/nrneph.2017.169] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Many of the >2 million animal species that inhabit Earth have developed survival mechanisms that aid in the prevention of obesity, kidney disease, starvation, dehydration and vascular ageing; however, some animals remain susceptible to these complications. Domestic and captive wild felids, for example, show susceptibility to chronic kidney disease (CKD), potentially linked to the high protein intake of these animals. By contrast, naked mole rats are a model of longevity and are protected from extreme environmental conditions through mechanisms that provide resistance to oxidative stress. Biomimetic studies suggest that the transcription factor nuclear factor erythroid 2-related factor 2 (NRF2) offers protection in extreme environmental conditions and promotes longevity in the animal kingdom. Similarly, during months of fasting, immobilization and anuria, hibernating bears are protected from muscle wasting, azotaemia, thrombotic complications, organ damage and osteoporosis - features that are often associated with CKD. Improved understanding of the susceptibility and protective mechanisms of these animals and others could provide insights into novel strategies to prevent and treat several human diseases, such as CKD and ageing-associated complications. An integrated collaboration between nephrologists and experts from other fields, such as veterinarians, zoologists, biologists, anthropologists and ecologists, could introduce a novel approach for improving human health and help nephrologists to find novel treatment strategies for CKD.
Collapse
|
36
|
Tidwell TR, Søreide K, Hagland HR. Aging, Metabolism, and Cancer Development: from Peto's Paradox to the Warburg Effect. Aging Dis 2017; 8:662-676. [PMID: 28966808 PMCID: PMC5614328 DOI: 10.14336/ad.2017.0713] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 06/13/2017] [Indexed: 12/15/2022] Open
Abstract
Medical advances made over the last century have increased our lifespan, but age-related diseases are a fundamental health burden worldwide. Aging is therefore a major risk factor for cardiovascular disease, cancer, diabetes, obesity, and neurodegenerative diseases, all increasing in prevalence. However, huge inter-individual variations in aging and disease risk exist, which cannot be explained by chronological age, but rather physiological age decline initiated even at young age due to lifestyle. At the heart of this lies the metabolic system and how this is regulated in each individual. Metabolic turnover of food to energy leads to accumulation of co-factors, byproducts, and certain proteins, which all influence gene expression through epigenetic regulation. How these epigenetic markers accumulate over time is now being investigated as the possible link between aging and many diseases, such as cancer. The relationship between metabolism and cancer was described as early as the late 1950s by Dr. Otto Warburg, before the identification of DNA and much earlier than our knowledge of epigenetics. However, when the stepwise gene mutation theory of cancer was presented, Warburg's theories garnered little attention. Only in the last decade, with epigenetic discoveries, have Warburg's data on the metabolic shift in cancers been brought back to life. The stepwise gene mutation theory fails to explain why large animals with more cells, do not have a greater cancer incidence than humans, known as Peto's paradox. The resurgence of research into the Warburg effect has given us insight to what may explain Peto's paradox. In this review, we discuss these connections and how age-related changes in metabolism are tightly linked to cancer development, which is further affected by lifestyle choices modulating the risk of aging and cancer through epigenetic control.
Collapse
Affiliation(s)
- Tia R. Tidwell
- Department of Mathematics and Natural Sciences, Centre for Organelle Research, University of Stavanger, Stavanger, Norway
- Gastrointestinal Translational Research Unit, Molecular Laboratory, Hillevaåg, Stavanger University Hospital, Stavanger, Norway
| | - Kjetil Søreide
- Gastrointestinal Translational Research Unit, Molecular Laboratory, Hillevaåg, Stavanger University Hospital, Stavanger, Norway
- Department of Gastrointestinal Surgery, Stavanger University Hospital, Stavanger, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Hanne R. Hagland
- Department of Mathematics and Natural Sciences, Centre for Organelle Research, University of Stavanger, Stavanger, Norway
- Gastrointestinal Translational Research Unit, Molecular Laboratory, Hillevaåg, Stavanger University Hospital, Stavanger, Norway
| |
Collapse
|
37
|
Loos B, Klionsky DJ, Wong E. Augmenting brain metabolism to increase macro- and chaperone-mediated autophagy for decreasing neuronal proteotoxicity and aging. Prog Neurobiol 2017; 156:90-106. [DOI: 10.1016/j.pneurobio.2017.05.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 05/06/2017] [Accepted: 05/08/2017] [Indexed: 12/14/2022]
|
38
|
Martínez G, Duran‐Aniotz C, Cabral‐Miranda F, Vivar JP, Hetz C. Endoplasmic reticulum proteostasis impairment in aging. Aging Cell 2017; 16:615-623. [PMID: 28436203 PMCID: PMC5506418 DOI: 10.1111/acel.12599] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2017] [Indexed: 12/12/2022] Open
Abstract
Perturbed neuronal proteostasis is a salient feature shared by both aging and protein misfolding disorders. The proteostasis network controls the health of the proteome by integrating pathways involved in protein synthesis, folding, trafficking, secretion, and their degradation. A reduction in the buffering capacity of the proteostasis network during aging may increase the risk to undergo neurodegeneration by enhancing the accumulation of misfolded proteins. As almost one-third of the proteome is synthetized at the endoplasmic reticulum (ER), maintenance of its proper function is fundamental to sustain neuronal function. In fact, ER stress is a common feature of most neurodegenerative diseases. The unfolded protein response (UPR) operates as central player to maintain ER homeostasis or the induction of cell death of chronically damaged cells. Here, we discuss recent evidence placing ER stress as a driver of brain aging, and the emerging impact of neuronal UPR in controlling global proteostasis at the whole organismal level. Finally, we discuss possible therapeutic interventions to improve proteostasis and prevent pathological brain aging.
Collapse
Affiliation(s)
- Gabriela Martínez
- Center for Geroscience, Brain Health and MetabolismSantiagoChile
- Biomedical Neuroscience InstituteFaculty of MedicineUniversity of ChileSantiagoChile
- Program of Cellular and Molecular BiologyInstitute of Biomedical SciencesUniversity of ChileSantiagoChile
- Center for Integrative BiologyUniversidad MayorSantiagoChile
| | - Claudia Duran‐Aniotz
- Center for Geroscience, Brain Health and MetabolismSantiagoChile
- Biomedical Neuroscience InstituteFaculty of MedicineUniversity of ChileSantiagoChile
- Program of Cellular and Molecular BiologyInstitute of Biomedical SciencesUniversity of ChileSantiagoChile
| | - Felipe Cabral‐Miranda
- Center for Geroscience, Brain Health and MetabolismSantiagoChile
- Biomedical Neuroscience InstituteFaculty of MedicineUniversity of ChileSantiagoChile
- Program of Cellular and Molecular BiologyInstitute of Biomedical SciencesUniversity of ChileSantiagoChile
- Instituto de Ciências BiomédicasUniversidade Federal do Rio de JaneiroRio de JaneiroBrasil
| | - Juan P. Vivar
- Center for Geroscience, Brain Health and MetabolismSantiagoChile
- Biomedical Neuroscience InstituteFaculty of MedicineUniversity of ChileSantiagoChile
- Program of Cellular and Molecular BiologyInstitute of Biomedical SciencesUniversity of ChileSantiagoChile
| | - Claudio Hetz
- Center for Geroscience, Brain Health and MetabolismSantiagoChile
- Biomedical Neuroscience InstituteFaculty of MedicineUniversity of ChileSantiagoChile
- Program of Cellular and Molecular BiologyInstitute of Biomedical SciencesUniversity of ChileSantiagoChile
- Buck Institute for Research on AgingNovatoCA94945USA
- Department of Immunology and Infectious diseasesHarvard School of Public HealthBostonMA02115USA
| |
Collapse
|
39
|
Dammann P. Slow aging in mammals-Lessons from African mole-rats and bats. Semin Cell Dev Biol 2017; 70:154-163. [PMID: 28698112 DOI: 10.1016/j.semcdb.2017.07.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 07/04/2017] [Accepted: 07/05/2017] [Indexed: 12/30/2022]
Abstract
Traditionally, the main mammalian models used in aging research have been mice and rats, i.e. short-lived species that obviously lack effective maintenance mechanisms to keep their soma in a functional state for prolonged periods of time. It is doubtful that life-extending mechanisms identified only in such short-lived species adequately reflect the diversity of longevity pathways that have naturally evolved in mammals, or that they have much relevance for long-lived species such as humans. Therefore, some complementary, long-lived mammalian models have been introduced to aging research in the past 15-20 years, particularly naked mole-rats (and to a lesser extent also other mole-rats) and bats. Here, I summarize and compare the most important results regarding various aspects of aging - oxidative stress, molecular homeostasis and repair, and endocrinology - that have been obtained from studies using these new mammalian models of high longevity. I argue that the inclusion of these models was an important step forward, because it drew researchers' attention to certain oversimplifications of existing aging theories and to several features that appear to be universal components of enhanced longevity in mammals. However, even among mammals with high longevity, considerable variation exists with respect to other candidate mechanisms that also must be taken into account if inadequate generalizations are to be avoided.
Collapse
Affiliation(s)
- Philip Dammann
- Central Animal Laboratory, Faculty of Medicine, University of Duisburg, Essen, Germany.
| |
Collapse
|
40
|
Yu Y, Feng L, Li J, Lan X, A L, Lv X, Zhang M, Chen L. The alteration of autophagy and apoptosis in the hippocampus of rats with natural aging-dependent cognitive deficits. Behav Brain Res 2017; 334:155-162. [PMID: 28688896 DOI: 10.1016/j.bbr.2017.07.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 06/28/2017] [Accepted: 07/03/2017] [Indexed: 11/19/2022]
Abstract
AIMS The present study was aim to explore aging-dependent changes in hippocampal autophagy and apoptosis in a natural aging rat model from adult to old stages and to discover a suitable age for treating neurodegenerative diseases. METHODS Wistar rats at 5, 18 and 24months of age were used to mimic the adulthood, initial old, and old phases, respectively. The learning and cognitive ability of the rats was detected by the Morris water maze test. Morphological changes in the hippocampus were observed. Expressions of apoptosis and autophagy-related proteins were examined by Western blot. RESULTS The adult group (5months) exhibited high levels of autophagy related p-ULK p-ULK-1/ULK-1 ratio, Beclin-1, LC3II and cell survival, maintaining normal learning and cognitive function and integrated hippocampal morphology. The initial old group (18 months) presented a reduced number of neurons and cognitive deficits, and exhibited high levels of apoptosis related Bax/Bcl-2 ratio, Caspase-3 activation and autophagy related p-ULK p-ULK-1/ULK-1 ratio, Beclin-1, LC3II compared to the adult group. The old group (24 months) exhibited a high level of apoptosis related Bax/Bcl-2 ratio, Caspase-3 activation and a low level of autophagy related p-ULK p-ULK-1/ULK-1 ratio, Beclin-1, LC3II compared to its younger group, as well as significant neuronal death and cognitive deficits. The degree of autophagy was generally consistent with its negative regulator, the PI3K/Akt/mTOR axis, in all groups. CONCLUSION Our data suggest that cognitive deficits are first observed in the initial old stage. The levels of autophagy and apoptosis tend to be opposite in the adult and old phases. High levels of autophagy and apoptosis coexist in the initial old stage. Our study indicates that up-regulation of autophagy in the initial old phase to anti-cognitive deficits must be further evaluated.
Collapse
Affiliation(s)
- Yang Yu
- Department of Pharmacology, College of Basic Medical Sciences, School of Nursing, Jilin University, Changchun, Jilin, China
| | - Linjing Feng
- Department of Pharmacology, College of Basic Medical Sciences, School of Nursing, Jilin University, Changchun, Jilin, China
| | - Junnan Li
- Department of Pharmacology, College of Basic Medical Sciences, School of Nursing, Jilin University, Changchun, Jilin, China
| | - Xiaoxin Lan
- Department of Pharmacology, College of Basic Medical Sciences, School of Nursing, Jilin University, Changchun, Jilin, China
| | - Lixiang A
- Department of Pharmacology, College of Basic Medical Sciences, School of Nursing, Jilin University, Changchun, Jilin, China
| | - Xiaoyan Lv
- Department of Pharmacology, College of Basic Medical Sciences, School of Nursing, Jilin University, Changchun, Jilin, China
| | - Ming Zhang
- Department of Pharmacology, College of Basic Medical Sciences, School of Nursing, Jilin University, Changchun, Jilin, China.
| | - Li Chen
- Department of Pharmacology, College of Basic Medical Sciences, School of Nursing, Jilin University, Changchun, Jilin, China
| |
Collapse
|
41
|
Skulachev VP, Holtze S, Vyssokikh MY, Bakeeva LE, Skulachev MV, Markov AV, Hildebrandt TB, Sadovnichii VA. Neoteny, Prolongation of Youth: From Naked Mole Rats to “Naked Apes” (Humans). Physiol Rev 2017; 97:699-720. [DOI: 10.1152/physrev.00040.2015] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
It has been suggested that highly social mammals, such as naked mole rats and humans, are long-lived due to neoteny (the prolongation of youth). In both species, aging cannot operate as a mechanism facilitating natural selection because the pressure of this selection is strongly reduced due to 1) a specific social structure where only the “queen” and her “husband(s)” are involved in reproduction (naked mole rats) or 2) substituting fast technological progress for slow biological evolution (humans). Lists of numerous traits of youth that do not disappear with age in naked mole rats and humans are presented and discussed. A high resistance of naked mole rats to cancer, diabetes, cardiovascular and brain diseases, and many infections explains why their mortality rate is very low and almost age-independent and why their lifespan is more than 30 years, versus 3 years in mice. In young humans, curves of mortality versus age start at extremely low values. However, in the elderly, human mortality strongly increases. High mortality rates in other primates are observed at much younger ages than in humans. The inhibition of the aging process in humans by specific drugs seems to be a promising approach to prolong our healthspan. This might be a way to retard aging, which is already partially accomplished via the natural physiological phenomenon neoteny.
Collapse
Affiliation(s)
- Vladimir P. Skulachev
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, Russia; Lomonosov Moscow State University, Institute of Mitoengineering, Moscow, Russia; Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany; Lomonosov Moscow State University, Biological Faculty, Moscow, Russia; Lomonosov Moscow State University, Faculty of Mechanics and Mathematics, Moscow, Russia
| | - Susanne Holtze
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, Russia; Lomonosov Moscow State University, Institute of Mitoengineering, Moscow, Russia; Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany; Lomonosov Moscow State University, Biological Faculty, Moscow, Russia; Lomonosov Moscow State University, Faculty of Mechanics and Mathematics, Moscow, Russia
| | - Mikhail Y. Vyssokikh
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, Russia; Lomonosov Moscow State University, Institute of Mitoengineering, Moscow, Russia; Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany; Lomonosov Moscow State University, Biological Faculty, Moscow, Russia; Lomonosov Moscow State University, Faculty of Mechanics and Mathematics, Moscow, Russia
| | - Lora E. Bakeeva
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, Russia; Lomonosov Moscow State University, Institute of Mitoengineering, Moscow, Russia; Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany; Lomonosov Moscow State University, Biological Faculty, Moscow, Russia; Lomonosov Moscow State University, Faculty of Mechanics and Mathematics, Moscow, Russia
| | - Maxim V. Skulachev
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, Russia; Lomonosov Moscow State University, Institute of Mitoengineering, Moscow, Russia; Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany; Lomonosov Moscow State University, Biological Faculty, Moscow, Russia; Lomonosov Moscow State University, Faculty of Mechanics and Mathematics, Moscow, Russia
| | - Alexander V. Markov
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, Russia; Lomonosov Moscow State University, Institute of Mitoengineering, Moscow, Russia; Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany; Lomonosov Moscow State University, Biological Faculty, Moscow, Russia; Lomonosov Moscow State University, Faculty of Mechanics and Mathematics, Moscow, Russia
| | - Thomas B. Hildebrandt
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, Russia; Lomonosov Moscow State University, Institute of Mitoengineering, Moscow, Russia; Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany; Lomonosov Moscow State University, Biological Faculty, Moscow, Russia; Lomonosov Moscow State University, Faculty of Mechanics and Mathematics, Moscow, Russia
| | - Viktor A. Sadovnichii
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, Russia; Lomonosov Moscow State University, Institute of Mitoengineering, Moscow, Russia; Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany; Lomonosov Moscow State University, Biological Faculty, Moscow, Russia; Lomonosov Moscow State University, Faculty of Mechanics and Mathematics, Moscow, Russia
| |
Collapse
|
42
|
Yin Y, Sun G, Li E, Kiselyov K, Sun D. ER stress and impaired autophagy flux in neuronal degeneration and brain injury. Ageing Res Rev 2017; 34:3-14. [PMID: 27594375 DOI: 10.1016/j.arr.2016.08.008] [Citation(s) in RCA: 156] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 08/31/2016] [Indexed: 12/12/2022]
Abstract
Autophagy is a highly controlled lysosome-mediated function in eukaryotic cells to eliminate damaged or aged long-lived proteins and organelles. It is required for restoring cellular homeostasis in cell survival under multiple stresses. Autophagy is known to be a double-edged sword because too much activation or inhibition of autophagy can disrupt homeostatic degradation of protein and organelles within the brain and play a role in neuronal cell death. Many factors affect autophagy flux function in the brain, including endoplasmic reticulum (ER) stress, oxidative stress, and aging. Newly emerged research indicates that altered autophagy flux functionality is involved in neurodegeneration of the aged brain, chronic neurological diseases, and after traumatic and ischemic brain injuries. In search to identify neuroprotective agents that may reduce oxidative stress and stimulate autophagy, one particular neuroprotective agent docosahexaenoic acid (DHA) presents unique functions in reducing ER and oxidative stress and modulating autophagy. This review will summarize the recent findings on changes of autophagy in aging, neurodegenerative diseases, and brain injury after trauma or ischemic strokes. Discussion of DHA functions is focused on modulating ER stress and autophagy in regard to its neuroprotection and anti-tumor functions.
Collapse
Affiliation(s)
- Yan Yin
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian 116023, PR China; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, United States.
| | - George Sun
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - Eric Li
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - Kirill Kiselyov
- Department of Biological Science, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, United States; Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Education and Clinical Center, Pittsburgh, PA 15213, United States.
| |
Collapse
|
43
|
Garza-Lombó C, Gonsebatt ME. Mammalian Target of Rapamycin: Its Role in Early Neural Development and in Adult and Aged Brain Function. Front Cell Neurosci 2016; 10:157. [PMID: 27378854 PMCID: PMC4910040 DOI: 10.3389/fncel.2016.00157] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 05/30/2016] [Indexed: 01/14/2023] Open
Abstract
The kinase mammalian target of rapamycin (mTOR) integrates signals triggered by energy, stress, oxygen levels, and growth factors. It regulates ribosome biogenesis, mRNA translation, nutrient metabolism, and autophagy. mTOR participates in various functions of the brain, such as synaptic plasticity, adult neurogenesis, memory, and learning. mTOR is present during early neural development and participates in axon and dendrite development, neuron differentiation, and gliogenesis, among other processes. Furthermore, mTOR has been shown to modulate lifespan in multiple organisms. This protein is an important energy sensor that is present throughout our lifetime its role must be precisely described in order to develop therapeutic strategies and prevent diseases of the central nervous system. The aim of this review is to present our current understanding of the functions of mTOR in neural development, the adult brain and aging.
Collapse
Affiliation(s)
- Carla Garza-Lombó
- Departamento de Medicina Genómica, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México México
| | - María E Gonsebatt
- Departamento de Medicina Genómica, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México México
| |
Collapse
|
44
|
Lees H, Walters H, Cox LS. Animal and human models to understand ageing. Maturitas 2016; 93:18-27. [PMID: 27372369 DOI: 10.1016/j.maturitas.2016.06.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/07/2016] [Accepted: 06/08/2016] [Indexed: 12/12/2022]
Abstract
Human ageing is the gradual decline in organ and tissue function with increasing chronological time, leading eventually to loss of function and death. To study the processes involved over research-relevant timescales requires the use of accessible model systems that share significant similarities with humans. In this review, we assess the usefulness of various models, including unicellular yeasts, invertebrate worms and flies, mice and primates including humans, and highlight the benefits and possible drawbacks of each model system in its ability to illuminate human ageing mechanisms. We describe the strong evolutionary conservation of molecular pathways that govern cell responses to extracellular and intracellular signals and which are strongly implicated in ageing. Such pathways centre around insulin-like growth factor signalling and integration of stress and nutritional signals through mTOR kinase. The process of cellular senescence is evaluated as a possible underlying cause for many of the frailties and diseases of human ageing. Also considered is ageing arising from systemic changes that cannot be modelled in lower organisms and instead require studies either in small mammals or in primates. We also touch briefly on novel therapeutic options arising from a better understanding of the biology of ageing.
Collapse
Affiliation(s)
- Hayley Lees
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Hannah Walters
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Lynne S Cox
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK.
| |
Collapse
|
45
|
Triplett JC, Swomley AM, Kirk J, Grimes KM, Lewis KN, Orr ME, Rodriguez KA, Cai J, Klein JB, Buffenstein R, Butterfield DA. Reaching Out to Send a Message: Proteins Associated with Neurite Outgrowth and Neurotransmission are Altered with Age in the Long-Lived Naked Mole-Rat. Neurochem Res 2016; 41:1625-34. [DOI: 10.1007/s11064-016-1877-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Revised: 02/06/2016] [Accepted: 02/18/2016] [Indexed: 10/22/2022]
|