1
|
Bernardová N, Novák J, Horák P, Fan CK, Kolářová L. Neurobehavioral Disorders and Pathological Changes in the Brain of Mice Are Caused by Chronic Toxocara canis Larval Invasion with Low to Moderate Inoculum. Acta Parasitol 2024; 69:1736-1748. [PMID: 39240446 DOI: 10.1007/s11686-024-00869-0] [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: 06/05/2024] [Accepted: 07/23/2024] [Indexed: 09/07/2024]
Abstract
Toxocara canis larvae are one of the most overlooked agents of nervous system infection in paratenic hosts. Previous studies in mouse models have shown that infection with various (mainly high) numbers of larvae leads to neurobehavioral disturbances and pathological changes. Our study investigated whether the infection with low and moderate numbers of larvae could affect the physical condition, motor skills, and pathogenesis in the brains of experimentally infected mice.Two groups of BALB/c mice were orally infected with 10 and 100 T. canis larvae per animal and examined regularly until the 97th week after infection. General appearance, specific antibody responses, and motor/balance skills were assessed. The number and viability of larvae in the liver, spleen, lungs, and brain were assessed by quantitative compressed biopsy technique, while the pathological changes of the brain infection were studied histologically.As a result, changes were observed in overall appearance, activity, as well as motor and balance ability. The infections were associated with an increased IgG antibody response to the specific anti-T. canis excretory/secretory antigen and tissue damage in the brain characterized by necrosis, cell infiltrations, including foamy cells, and hemorrhages.The study demonstrated the effects of low and moderate T. canis infection in a paratenic host during the chronic phase of infection, which lasted up to 97 weeks for the first time.
Collapse
Affiliation(s)
- Nicol Bernardová
- Institute of Immunology and Microbiology, 1st Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia.
| | - Jan Novák
- Institute of Immunology and Microbiology, 1st Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Petr Horák
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czechia
| | - Chia-Kwung Fan
- Department of Molecular Parasitology and Tropical Diseases, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Libuše Kolářová
- Institute of Immunology and Microbiology, 1st Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
- National Reference Laboratory for Tissue Helminthosis, General University Hospital, Prague, Czechia
| |
Collapse
|
2
|
Mansfield L, Ramponi V, Gupta K, Stevenson T, Mathew AB, Barinda AJ, Herbstein F, Morsli S. Emerging insights in senescence: pathways from preclinical models to therapeutic innovations. NPJ AGING 2024; 10:53. [PMID: 39578455 PMCID: PMC11584693 DOI: 10.1038/s41514-024-00181-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/25/2024] [Indexed: 11/24/2024]
Abstract
Senescence is a crucial hallmark of ageing and a significant contributor to the pathology of age-related disorders. As committee members of the young International Cell Senescence Association (yICSA), we aim to synthesise recent advancements in the identification, characterisation, and therapeutic targeting of senescence for clinical translation. We explore novel molecular techniques that have enhanced our understanding of senescent cell heterogeneity and their roles in tissue regeneration and pathology. Additionally, we delve into in vivo models of senescence, both non-mammalian and mammalian, to highlight tools available for advancing the contextual understanding of in vivo senescence. Furthermore, we discuss innovative diagnostic tools and senotherapeutic approaches, emphasising their potential for clinical application. Future directions of senescence research are explored, underscoring the need for precise, context-specific senescence classification and the integration of advanced technologies such as machine learning, long-read sequencing, and multifunctional senoprobes and senolytics. The dual role of senescence in promoting tissue homoeostasis and contributing to chronic diseases highlights the complexity of targeting these cells for improved clinical outcomes.
Collapse
Affiliation(s)
- Luke Mansfield
- The Bateson Centre, School of Medicine and Population Health, The University of Sheffield, Western Bank, Sheffield, UK
| | - Valentina Ramponi
- Cellular Plasticity and Disease Group, Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Kavya Gupta
- Department of Cellular and Molecular Biology and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | | | - Abraham Binoy Mathew
- Department of Developmental Biology and Genetics, Biological Sciences, Indian Institute of Science, Bangalore, India
| | - Agian Jeffilano Barinda
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Metabolic, Cardiovascular, and Aging Cluster, Indonesia Medical Education and Research Institute (IMERI), Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Florencia Herbstein
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires, Argentina.
| | - Samir Morsli
- Karolinska Institutet, Department of Cell and Molecular Biology, Biomedicum Q6A, Stockholm, Sweden.
| |
Collapse
|
3
|
Fernandes S, Revanna J, Pratt J, Hayes N, Marchetto MC, Gage FH. Modeling Alzheimer's disease using human cell derived brain organoids and 3D models. Front Neurosci 2024; 18:1434945. [PMID: 39156632 PMCID: PMC11328153 DOI: 10.3389/fnins.2024.1434945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 07/10/2024] [Indexed: 08/20/2024] Open
Abstract
Age-related neurodegenerative diseases, like Alzheimer's disease (AD), are challenging diseases for those affected with no cure and limited treatment options. Functional, human derived brain tissues that represent the diverse genetic background and cellular subtypes contributing to sporadic AD (sAD) are limited. Human stem cell derived brain organoids recapitulate some features of human brain cytoarchitecture and AD-like pathology, providing a tool for illuminating the relationship between AD pathology and neural cell dysregulation leading to cognitive decline. In this review, we explore current strategies for implementing brain organoids in the study of AD as well as the challenges associated with investigating age-related brain diseases using organoid models.
Collapse
Affiliation(s)
- Sarah Fernandes
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, United States
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Jasmin Revanna
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, United States
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Joshua Pratt
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, United States
- Department of Biology, San Diego State University, San Diego, CA, United States
| | - Nicholas Hayes
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, United States
- Department of Biological Sciences, California State University, San Marcos, CA, United States
| | - Maria C. Marchetto
- Department of Anthropology, Center for Academic Research and Training in Anthropogeny (CARTA), University of California, San Diego, La Jolla, CA, United States
| | - Fred H. Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA, United States
| |
Collapse
|
4
|
Gille B, Müller-Eigner A, Gottschalk S, Wytrwat E, Langhammer M, Peleg S. Titan mice as a model to test interventions that attenuate frailty and increase longevity. GeroScience 2024; 46:3599-3606. [PMID: 38177967 PMCID: PMC11226689 DOI: 10.1007/s11357-023-01045-4] [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: 09/20/2023] [Accepted: 12/11/2023] [Indexed: 01/06/2024] Open
Abstract
Wild-type murine models for aging research have lifespans of several years, which results in long experimental duration and late output. Here we explore the short-lived non-inbred Titan mouse (DU6) as a mouse model to test longevity interventions. We show that Titan mice exhibit increased frailty and senescence-associated beta-galactosidase activity at an early age. Dietary intervention attenuates the frailty progression of Titan mice. Additionally, cyclic administration of the senolytic drug Navitoclax at an early age increases the lifespan and reduces senescence-associated beta-galactosidase activity. Our data suggests that Titan mice can serve as a cost-effective and timely model for longevity interventions in mammals.
Collapse
Affiliation(s)
- Benedikt Gille
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Annika Müller-Eigner
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Shari Gottschalk
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Erika Wytrwat
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Martina Langhammer
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - Shahaf Peleg
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany.
| |
Collapse
|
5
|
Hilmer SN, Johnell K, Mach J. Pre-clinical Models for Geriatric Pharmacotherapy. Drugs Aging 2024; 41:633-640. [PMID: 38982010 PMCID: PMC11322264 DOI: 10.1007/s40266-024-01129-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2024] [Indexed: 07/11/2024]
Abstract
With ageing of the population worldwide and discovery of new medications for prevention and management of age-related conditions, there is increasing use of medications by older adults. There are international efforts to increase the representativeness of participants in clinical trials to match the intended real-world users of the medications across a range of characteristics including age, multimorbidity, polypharmacy and frailty. Currently, much of the data on medication-related harm in older adults are from pharmacovigilance studies. New methods in pre-clinical models have allowed for measurement of exposures (such as chronic exposure, polypharmacy and deprescribing) and outcomes (such as health span functional measures and frailty) that are highly relevant to geriatric pharmacotherapy. Here we describe opportunities for design and implementation of pre-clinical models that can better predict drug effects in geriatric patients. This could improve the translation of new drugs from bench to bedside and improve outcomes of pharmacotherapy in older adults.
Collapse
Affiliation(s)
- Sarah N Hilmer
- Kolling Institute, The University of Sydney and Northern Sydney Local Health District, St Leonards, NSW, Australia.
| | - Kristina Johnell
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - John Mach
- Kolling Institute, The University of Sydney and Northern Sydney Local Health District, St Leonards, NSW, Australia
| |
Collapse
|
6
|
Carr LM, Mustafa S, Care A, Collins-Praino LE. More than a number: Incorporating the aged phenotype to improve in vitro and in vivo modeling of neurodegenerative disease. Brain Behav Immun 2024; 119:554-571. [PMID: 38663775 DOI: 10.1016/j.bbi.2024.04.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 03/04/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024] Open
Abstract
Age is the number one risk factor for developing a neurodegenerative disease (ND), such as Alzheimer's disease (AD) or Parkinson's disease (PD). With our rapidly ageing world population, there will be an increased burden of ND and need for disease-modifying treatments. Currently, however, translation of research from bench to bedside in NDs is poor. This may be due, at least in part, to the failure to account for the potential effect of ageing in preclinical modelling of NDs. While ageing can impact upon physiological response in multiple ways, only a limited number of preclinical studies of ND have incorporated ageing as a factor of interest. Here, we evaluate the aged phenotype and highlight the critical, but unmet, need to incorporate aspects of this phenotype into both the in vitro and in vivo models used in ND research. Given technological advances in the field over the past several years, we discuss how these could be harnessed to create novel models of ND that more readily incorporate aspects of the aged phenotype. This includes a recently described in vitro panel of ageing markers, which could help lead to more standardised models and improve reproducibility across studies. Importantly, we cannot assume that young cells or animals yield the same responses as seen in the context of ageing; thus, an improved understanding of the biology of ageing, and how to appropriately incorporate this into the modelling of ND, will ensure the best chance for successful translation of new therapies to the aged patient.
Collapse
Affiliation(s)
- Laura M Carr
- School of Biomedicine, University of Adelaide, Adelaide, SA, Australia
| | - Sanam Mustafa
- School of Biomedicine, University of Adelaide, Adelaide, SA, Australia; Australian Research Council Centre of Excellence for Nanoscale Biophotonics, The University of Adelaide, Adelaide, SA, Australia; Davies Livestock Research Centre, The University of Adelaide, Roseworthy, SA, Australia
| | - Andrew Care
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Lyndsey E Collins-Praino
- School of Biomedicine, University of Adelaide, Adelaide, SA, Australia; Australian Research Council Centre of Excellence for Nanoscale Biophotonics, The University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
7
|
Sember E, Chennakesavula R, Beard B, Opoola M, Hwangbo DS. Dietary restriction fails to extend lifespan of Drosophila model of Werner syndrome. G3 (BETHESDA, MD.) 2024; 14:jkae056. [PMID: 38491858 PMCID: PMC11075538 DOI: 10.1093/g3journal/jkae056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/28/2024] [Accepted: 03/04/2024] [Indexed: 03/18/2024]
Abstract
Werner syndrome (WS) is a rare genetic disease in humans, caused by mutations in the WRN gene that encodes a protein containing helicase and exonuclease domains. WS is characterized by symptoms of accelerated aging in multiple tissues and organs, involving increased risk of cancer, heart failure, and metabolic dysfunction. These conditions ultimately lead to the premature mortality of patients with WS. In this study, using the null mutant flies (WRNexoΔ) for the gene WRNexo (CG7670), homologous to the exonuclease domain of WRN in humans, we examined how diets affect the lifespan, stress resistance, and sleep/wake patterns of a Drosophila model of WS. We observed that dietary restriction (DR), one of the most robust nongenetic interventions to extend lifespan in animal models, failed to extend the lifespan of WRNexoΔ mutant flies and even had a detrimental effect in females. Interestingly, the mean lifespan of WRNexoΔ mutant flies was not reduced on a protein-rich diet compared to that of wild-type (WT) flies. Compared to WT control flies, the mutant flies also exhibited altered responses to DR in their resistance to starvation and oxidative stress, as well as changes in sleep/wake patterns. These findings show that the WRN protein is necessary for mediating the effects of DR and suggest that the exonuclease domain of WRN plays an important role in metabolism in addition to its primary role in DNA-repair and genome stability.
Collapse
Affiliation(s)
- Eileen Sember
- Department of Biology, University of Louisville, Louisville, KY 40292, USA
| | | | - Breanna Beard
- Department of Biology, University of Louisville, Louisville, KY 40292, USA
| | - Mubaraq Opoola
- Department of Biology, University of Louisville, Louisville, KY 40292, USA
| | - Dae-Sung Hwangbo
- Department of Biology, University of Louisville, Louisville, KY 40292, USA
| |
Collapse
|
8
|
Park S, Laskow TC, Chen J, Guha P, Dawn B, Kim D. Microphysiological systems for human aging research. Aging Cell 2024; 23:e14070. [PMID: 38180277 PMCID: PMC10928588 DOI: 10.1111/acel.14070] [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: 06/30/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 01/06/2024] Open
Abstract
Recent advances in microphysiological systems (MPS), also known as organs-on-a-chip (OoC), enable the recapitulation of more complex organ and tissue functions on a smaller scale in vitro. MPS therefore provide the potential to better understand human diseases and physiology. To date, numerous MPS platforms have been developed for various tissues and organs, including the heart, liver, kidney, blood vessels, muscle, and adipose tissue. However, only a few studies have explored using MPS platforms to unravel the effects of aging on human physiology and the pathogenesis of age-related diseases. Age is one of the risk factors for many diseases, and enormous interest has been devoted to aging research. As such, a human MPS aging model could provide a more predictive tool to understand the molecular and cellular mechanisms underlying human aging and age-related diseases. These models can also be used to evaluate preclinical drugs for age-related diseases and translate them into clinical settings. Here, we provide a review on the application of MPS in aging research. First, we offer an overview of the molecular, cellular, and physiological changes with age in several tissues or organs. Next, we discuss previous aging models and the current state of MPS for studying human aging and age-related conditions. Lastly, we address the limitations of current MPS and present future directions on the potential of MPS platforms for human aging research.
Collapse
Affiliation(s)
- Seungman Park
- Department of Mechanical EngineeringUniversity of Nevada, Las VegasLas VegasNevadaUSA
| | - Thomas C. Laskow
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Jingchun Chen
- Nevada Institute of Personalized MedicineUniversity of Nevada, Las VegasLas VegasNevadaUSA
| | - Prasun Guha
- Nevada Institute of Personalized MedicineUniversity of Nevada, Las VegasLas VegasNevadaUSA
- School of Life SciencesUniversity of Nevada, Las VegasLas VegasNevadaUSA
| | - Buddhadeb Dawn
- Department of Internal Medicine, Kirk Kerkorian School of MedicineUniversity of Nevada, Las VegasLas VegasNevadaUSA
| | - Deok‐Ho Kim
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Biomedical EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
- Center for Microphysiological SystemsJohns Hopkins UniversityBaltimoreMarylandUSA
| |
Collapse
|
9
|
Polis B, Samson AO. Addressing the Discrepancies Between Animal Models and Human Alzheimer's Disease Pathology: Implications for Translational Research. J Alzheimers Dis 2024; 98:1199-1218. [PMID: 38517793 DOI: 10.3233/jad-240058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2024]
Abstract
Animal models, particularly transgenic mice, are extensively used in Alzheimer's disease (AD) research to emulate key disease hallmarks, such as amyloid plaques and neurofibrillary tangles formation. Although these models have contributed to our understanding of AD pathogenesis and can be helpful in testing potential therapeutic interventions, their reliability is dubious. While preclinical studies have shown promise, clinical trials often yield disappointing results, highlighting a notable gap and disparity between animal models and human AD pathology. Existing models frequently overlook early-stage human pathologies and other key AD characteristics, thereby limiting their application in identifying optimal therapeutic interventions. Enhancing model reliability necessitates rigorous study design, comprehensive behavioral evaluations, and biomarker utilization. Overall, a nuanced understanding of each model's neuropathology, its fidelity to human AD, and its limitations is essential for accurate interpretation and successful translation of findings. This article analyzes the discrepancies between animal models and human AD pathology that complicate the translation of findings from preclinical studies to clinical applications. We also delve into AD pathogenesis and attributes to propose a new perspective on this pathology and deliberate over the primary limitations of key experimental models. Additionally, we discuss several fundamental problems that may explain the translational failures and suggest some possible directions for more effective preclinical studies.
Collapse
Affiliation(s)
- Baruh Polis
- Bar-Ilan University Azrieli Faculty of Medicine, Safed, Israel
| | | |
Collapse
|
10
|
Pu X, Qu Y. A study on the delayed effect of tilapia skin collagen on skin aging for mice and its possible mechanism. J Cosmet Dermatol 2023; 22:3436-3444. [PMID: 37328955 DOI: 10.1111/jocd.15835] [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: 01/07/2023] [Revised: 04/05/2023] [Accepted: 05/14/2023] [Indexed: 06/18/2023]
Abstract
OBJECTIVE Study the possible mechanism and delayed effect of tilapia skin collagen on skin aging for mice. MATERIALS AND METHODS Kunming (KM) mice were randomly divided into the aging model group, the normal group, the positive control group (vitamin E) and the low, medium, high dose tilapia skin collagen groups (2.0, 4.0, 8.0 mg/g). The normal group was only injected with saline at the back and the neck. The other groups were injected subcutaneously with 5% D-galactose and ultraviolet light jointly to establish the aging model. After modeling, the positive control group was treated with a dose of 10% vitamin E once a day, and the low, medium, high dose tilapia skin collagen group was separately applied 2.0, 4.0, 8.0 mg/g of tilapia skin collagen for 40 days. The changes of skin tissue morphology, water content, hydroxyproline (Hyp) content, and superoxide dismutase (SOD) activity in mice were studied at the day 10, 20, 30, 40, 50. RESULTS Compared with the normal group, the skin of mice in the aging model group was thinner, looser, and the skin moisture content, Hyp content, SOD activity were all decreased. For mice of the low, medium, high dose tilapia skin collagen group, the thickness of dermis increased, possessing close arrangement, and the moisture content, Hyp content, SOD activity were up-regulated significantly, which effectively alleviated the aging process of skin. The dose of tilapia skin collagen was directly proportional to the anti-aging effect. CONCLUSIONS Tilapia skin collagen has an obvious effect on improving skin aging.
Collapse
Affiliation(s)
- Xinyue Pu
- Weifang Medical University, Weifang, China
| | - Yan Qu
- Department of Dermatology, Yantai Yuhuangding Hospital, Yantai, China
| |
Collapse
|
11
|
Tokuda K, Lu SL, Zhang Z, Kato Y, Chen S, Noda K, Hirose K, Usami Y, Uzawa N, Murakami S, Toyosawa S, Fukuda M, Sun-Wada GH, Wada Y, Noda T. Rab32 and Rab38 maintain bone homeostasis by regulating intracellular traffic in osteoclasts. Cell Struct Funct 2023; 48:223-239. [PMID: 37793839 PMCID: PMC11496785 DOI: 10.1247/csf.23061] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 09/28/2023] [Indexed: 10/06/2023] Open
Abstract
Osteoclasts play a crucial role in bone homeostasis by forming resorption pits on bone surfaces, resulting in bone resorption. The osteoclast expression of Rab38 protein is highly induced during differentiation from macrophages. Here we generated mice with double knockout (DKO) of Rab38 and its paralogue, Rab32, to investigate the roles of these proteins in osteoclasts. Bone marrow-derived macrophages from Rab32/38 DKO mice differentiated normally into osteoclasts in vitro. However, DKO osteoclasts showed reduced bone resorption activity. These osteoclasts also demonstrated defective secretion of tartrate-resistant acid phosphatase and cathepsin K into culture medium. Furthermore, the plasma membrane localization of a3, an osteoclast-specific a subunit of V-ATPase, was abrogated in DKO mice, substantiating the reduced resorption activity. In vivo, Rab32- and Rab38-positive cells were attached to the bone surface. Eight-week-old DKO mice showed significantly thickened trabecular bones in micro-CT and histomorphometry analysis, as well as reduced serum levels of cross-linked C-telopeptide of type I collagen, indicating diminished bone resorption in vivo. In DKO male mice over 10 weeks of age, hyperostosis appeared at the talofibular syndesmosis, the distal junction of the tibia and fibula. Furthermore, middle-aged mice (10 to 12 months of age) exhibited kyphosis, which is not usually observed in wild-type male mice until around 24 months of age. These results indicate that Rab32 and Rab38 contribute to osteoclast function by supporting intracellular traffic, thereby maintaining normal bone homeostasis.Key words: Rab32, Rab38, osteoclast, lysosome-related organelle, secretory lysosome.
Collapse
Affiliation(s)
- Kanako Tokuda
- Graduate School of Frontier Biosciences, Osaka University, Osaka 565-0871, Japan
| | - Shiou-Ling Lu
- Department of Oral Cellular Biology, Center for Frontier Oral Science, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| | - Zidi Zhang
- Department of Oral Cellular Biology, Center for Frontier Oral Science, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| | - Yumiko Kato
- Department of Oral Cellular Biology, Center for Frontier Oral Science, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
- Department of Oral & Maxillofacial Oncology and Surgery, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| | - Siyu Chen
- Department of Oral Cellular Biology, Center for Frontier Oral Science, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| | - Kazuya Noda
- Department of Oral Cellular Biology, Center for Frontier Oral Science, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
- Department of Periodontology and Regenerative Dentistry, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| | - Katsutoshi Hirose
- Department of Oral and Maxillofacial Pathology, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| | - Yu Usami
- Department of Oral and Maxillofacial Pathology, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| | - Narikazu Uzawa
- Department of Oral & Maxillofacial Oncology and Surgery, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| | - Shinya Murakami
- Department of Periodontology and Regenerative Dentistry, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| | - Satoru Toyosawa
- Department of Oral and Maxillofacial Pathology, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| | - Mitsunori Fukuda
- Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Miyagi 980-8578, Japan
| | - Ge-Hong Sun-Wada
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Doshisha Women’s College, Kyoto 610-0395, Japan
| | - Yoh Wada
- Department of Biological Sciences, Institute of Scientific and Industrial Research, Osaka University, Osaka 565-0871, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka 565-0871, Japan
| | - Takeshi Noda
- Graduate School of Frontier Biosciences, Osaka University, Osaka 565-0871, Japan
- Department of Oral Cellular Biology, Center for Frontier Oral Science, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
12
|
Sri S, Greenstein A, Granata A, Collcutt A, Jochems ACC, McColl BW, Castro BD, Webber C, Reyes CA, Hall C, Lawrence CB, Hawkes C, Pegasiou-Davies CM, Gibson C, Crawford CL, Smith C, Vivien D, McLean FH, Wiseman F, Brezzo G, Lalli G, Pritchard HAT, Markus HS, Bravo-Ferrer I, Taylor J, Leiper J, Berwick J, Gan J, Gallacher J, Moss J, Goense J, McMullan L, Work L, Evans L, Stringer MS, Ashford MLJ, Abulfadl M, Conlon N, Malhotra P, Bath P, Canter R, Brown R, Ince S, Anderle S, Young S, Quick S, Szymkowiak S, Hill S, Allan S, Wang T, Quinn T, Procter T, Farr TD, Zhao X, Yang Z, Hainsworth AH, Wardlaw JM. A multi-disciplinary commentary on preclinical research to investigate vascular contributions to dementia. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2023; 5:100189. [PMID: 37941765 PMCID: PMC10628644 DOI: 10.1016/j.cccb.2023.100189] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/27/2023] [Accepted: 10/05/2023] [Indexed: 11/10/2023]
Abstract
Although dementia research has been dominated by Alzheimer's disease (AD), most dementia in older people is now recognised to be due to mixed pathologies, usually combining vascular and AD brain pathology. Vascular cognitive impairment (VCI), which encompasses vascular dementia (VaD) is the second most common type of dementia. Models of VCI have been delayed by limited understanding of the underlying aetiology and pathogenesis. This review by a multidisciplinary, diverse (in terms of sex, geography and career stage), cross-institute team provides a perspective on limitations to current VCI models and recommendations for improving translation and reproducibility. We discuss reproducibility, clinical features of VCI and corresponding assessments in models, human pathology, bioinformatics approaches, and data sharing. We offer recommendations for future research, particularly focusing on small vessel disease as a main underpinning disorder.
Collapse
Affiliation(s)
- Sarmi Sri
- UK Dementia Research Institute Headquarters, 6th Floor Maple House, London W1T 7NF, UK
| | - Adam Greenstein
- Division of Cardiovascular Sciences, The University of Manchester, Manchester M13 9PL, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Alessandra Granata
- Department of Clinical Neurosciences, Victor Phillip Dahdaleh Heart & Lung Research Institute, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Alex Collcutt
- UK Dementia Research Institute Headquarters, 6th Floor Maple House, London W1T 7NF, UK
| | - Angela C C Jochems
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
| | - Barry W McColl
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Blanca Díaz Castro
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Caleb Webber
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, UK
| | - Carmen Arteaga Reyes
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
| | - Catherine Hall
- School of Psychology and Sussex Neuroscience, University of Sussex, Falmer, Brighton, East Sussex, UK
| | - Catherine B Lawrence
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Cheryl Hawkes
- Biomedical and Life Sciences, Lancaster University, Lancaster, UK
| | | | - Claire Gibson
- School of Psychology, University of Nottingham, Nottingham NG7 2UH, UK
| | - Colin L Crawford
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Colin Smith
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Denis Vivien
- Physiopathology and Imaging of Neurological Disorders (PhIND), Normandie University, UNICAEN, INSERM UMR-S U1237, , GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), Caen, France
- Department of clinical research, Caen-Normandie University Hospital, Caen, France
| | - Fiona H McLean
- Division of Systems Medicine, School of Medicine, Ninewells Hospital & Medical School, University of Dundee, Dundee DD1 9SY, UK
| | - Frances Wiseman
- UK Dementia Research Institute, University College London, London WC1N 3BG, UK
| | - Gaia Brezzo
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Giovanna Lalli
- UK Dementia Research Institute Headquarters, 6th Floor Maple House, London W1T 7NF, UK
| | - Harry A T Pritchard
- Division of Cardiovascular Sciences, The University of Manchester, Manchester M13 9PL, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Hugh S Markus
- Stroke Research Group, Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Isabel Bravo-Ferrer
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Jade Taylor
- Division of Cardiovascular Sciences, The University of Manchester, Manchester M13 9PL, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - James Leiper
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Jason Berwick
- Department of Psychology, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
- Healthy Lifespan Institute, University of Sheffield, Sheffield, UK
| | - Jian Gan
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - John Gallacher
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, UK
| | - Jonathan Moss
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, UK
| | - Jozien Goense
- Neuroscience Program, University of Illinois, Urbana-Champaign, Urbana, IL, USA
- Department of Psychology, University of Illinois, Urbana-Champaign, Champaign, IL, USA
- Department of Bioengineering, University of Illinois, Urbana-Champaign, Urbana, IL, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL, USA
- School of Psychology and Neuroscience, University of Glasgow, UK
| | - Letitia McMullan
- School of Psychology and Sussex Neuroscience, University of Sussex, Falmer, Brighton, East Sussex, UK
| | - Lorraine Work
- School of Cardiovascular & Metabolic Health, College of Medical, Veterinary & Life Sciences, University of Glasgow; Glasgow; UK
| | - Lowri Evans
- Division of Cardiovascular Sciences, The University of Manchester, Manchester M13 9PL, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Michael S Stringer
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
| | - MLJ Ashford
- Division of Systems Medicine, School of Medicine, Ninewells Hospital & Medical School, University of Dundee, Dundee DD1 9SY, UK
| | - Mohamed Abulfadl
- Dementia Research Group, Department of Clinical Neurosciences, Bristol Medical School, University of Bristol, Bristol BS10 5NB, UK
| | - Nina Conlon
- Division of Cardiovascular Sciences, The University of Manchester, Manchester M13 9PL, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Paresh Malhotra
- Department of Brain Sciences, Imperial College London, London, UK
- Department of Neurology, Imperial College Healthcare NHS Trust, London, UK
- UK Dementia Research Institute Care Research and Technology Centre, Imperial College London and the University of Surrey, UK
| | - Philip Bath
- Stroke Trials Unit, University of Nottingham, Nottingham, UK; Stroke, Medicine Division, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Rebecca Canter
- Dementia Discovery Fund, SV Health Managers LLP, London, UK
| | - Rosalind Brown
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Selvi Ince
- Dementia Research Group, Department of Clinical Neurosciences, Bristol Medical School, University of Bristol, Bristol BS10 5NB, UK
| | - Silvia Anderle
- School of Psychology and Sussex Neuroscience, University of Sussex, Falmer, Brighton, East Sussex, UK
- Department of Neuroscience, Physiology and Pharmacology, University College London, UK
| | - Simon Young
- Dementias Platform UK, Department of Psychiatry, University of Oxford, Oxford OX3 7JX, UK
| | - Sophie Quick
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Stefan Szymkowiak
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, UK
| | - Steve Hill
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, UK
| | - Stuart Allan
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Tao Wang
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Division of Evolution, Infection and Genomic Sciences, Faculty of Biology Medicine and Health, School of Biological Sciences, The University of Manchester, Manchester, UK
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Terry Quinn
- College of Medical Veterinary and Life Sciences, University of Glasgow, Scotland, UK
| | - Tessa Procter
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
- Royal (Dick) School of Veterinary Studies, The University of Edinburgh, UK
| | - Tracy D Farr
- School of Life Sciences, Physiology, Pharmacology, and Neuroscience Division, Medical School, University of Nottingham, Nottingham NG7 2UH, UK
| | - Xiangjun Zhao
- Division of Evolution, Infection and Genomic Sciences, Faculty of Biology Medicine and Health, School of Biological Sciences, The University of Manchester, Manchester, UK
| | - Zhiyuan Yang
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, London, UK
| | - Atticus H Hainsworth
- Molecular and Clinical Sciences Research Institute, St George's University of London SW17 0RE, UK
- Department of Neurology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Joanna M Wardlaw
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute Edinburgh, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
13
|
Xie K, Ehninger D. Ageing-associated phenotypes in mice. Mech Ageing Dev 2023; 214:111852. [PMID: 37454704 DOI: 10.1016/j.mad.2023.111852] [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: 04/21/2023] [Revised: 06/22/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Ageing is a continuous process in life featuring progressive damage accumulation that leads to physiological decline, functional deterioration and ultimately death of an organism. Based on the relatively close anatomical and physiological similarity to humans, the mouse has been proven as a valuable model organism in ageing research over the last decades. In this review, we survey methods and tools currently in use to assess ageing phenotypes in mice. We summarize a range of ageing-associated alterations detectable at two major levels of analysis: (1) physiology and pathophysiology and (2) molecular biomarkers. Age-sensitive phenotypes provided in this article may serve to inform future studies targeting various aspects of organismal ageing in mice. In addition, we discuss conceptual and technical challenges faced by previous ageing studies in mice and, where possible, provide recommendations on how to resolve some of these issues.
Collapse
Affiliation(s)
- Kan Xie
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Dan Ehninger
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany.
| |
Collapse
|
14
|
Prochownik EV, Wang H. Lessons in aging from Myc knockout mouse models. Front Cell Dev Biol 2023; 11:1244321. [PMID: 37621775 PMCID: PMC10446843 DOI: 10.3389/fcell.2023.1244321] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 07/31/2023] [Indexed: 08/26/2023] Open
Abstract
Despite MYC being among the most intensively studied oncogenes, its role in normal development has not been determined as Myc-/- mice do not survival beyond mid-gestation. Myc ± mice live longer than their wild-type counterparts and are slower to accumulate many age-related phenotypes. However, Myc haplo-insufficiency likely conceals other important phenotypes as many high-affinity Myc targets genes continue to be regulated normally. By delaying Myc inactivation until after birth it has recently been possible to study the consequences of its near-complete total body loss and thus to infer its normal function. Against expectation, these "MycKO" mice lived significantly longer than control wild-type mice but manifested a marked premature aging phenotype. This seemingly paradoxical behavior was potentially explained by a >3-fold lower lifetime incidence of cancer, normally the most common cause of death in mice and often Myc-driven. Myc loss accelerated the accumulation of numerous "Aging Hallmarks", including the loss of mitochondrial and ribosomal structural and functional integrity, the generation of reactive oxygen species, the acquisition of genotoxic damage, the detrimental rewiring of metabolism and the onset of senescence. In both mice and humans, normal aging in many tissues was accompaniued by the downregulation of Myc and the loss of Myc target gene regulation. Unlike most mouse models of premature aging, which are based on monogenic disorders of DNA damage recognition and repair, the MycKO mouse model directly impacts most Aging Hallmarks and may therefore more faithfully replicate the normal aging process of both mice and humans. It further establishes that the strong association between aging and cancer can be genetically separated and is maintained by a single gene.
Collapse
Affiliation(s)
- Edward V. Prochownik
- Division of Hematology/Oncology, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
- The Department of Microbiology and Molecular Genetics, UPMC, Pittsburgh, PA, United States
- The Hillman Cancer Center of UPMC, Pittsburgh, PA, United States
- The Pittsburgh Liver Research Center, UPMC, Pittsburgh, PA, United States
| | - Huabo Wang
- Division of Hematology/Oncology, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
15
|
Rezaeian AH, Dang F, Wei W. The circadian clock, aging and its implications in cancer. Neoplasia 2023; 41:100904. [PMID: 37148656 PMCID: PMC10192918 DOI: 10.1016/j.neo.2023.100904] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 04/28/2023] [Indexed: 05/08/2023]
Abstract
Circadian clock orchestrates the intergenic biochemical, physiological and behavioral changes to form an approximate 24h oscillation through the transcription-translation feedback loop (TTFL). Mechanistically, a heterodimer of transcriptional activator formed by BMAL1 and CLOCK, governs the expression of its transcriptional repressors, CRY, PER and REV-ERBα/β proteins, thereby controlling more than 50 % of protein encoding genes in human. There is also increasing evidence showing that tumor microenvironment can disrupt specific clock gene functions to facilitate tumorigenesis. Although there is great progress in understanding the molecular mechanisms of the circadian clock, aging and cancer, elucidating their complex relationships among these processes remains challenging. Herein, the optimization of the chronochemotherapy regimen has not been justified yet for treatment of cancer. Here, we discuss the hypothesis of relocalization of chromatin modifiers (RCM) along with function(s) of the circadian rhythm on aging and carcinogenesis. We will also introduce the function of the chromatin remodeling as a new avenue for rejuvenation of competent tissues to combat aging and cancer.
Collapse
Affiliation(s)
- Abdol-Hossein Rezaeian
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States.
| | - Fabin Dang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
16
|
Wilkie SE, Marcu DE, Carter RN, Morton NM, Gonzalo S, Selman C. Hepatic hydrogen sulfide levels are reduced in mouse model of Hutchinson-Gilford progeria syndrome. Aging (Albany NY) 2023; 15:5266-5278. [PMID: 37354210 PMCID: PMC10333079 DOI: 10.18632/aging.204835] [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: 02/10/2023] [Accepted: 06/09/2023] [Indexed: 06/26/2023]
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare human disease characterised by accelerated biological ageing. Current treatments are limited, and most patients die before 15 years of age. Hydrogen sulfide (H2S) is an important gaseous signalling molecule that it central to multiple cellular homeostasis mechanisms. Dysregulation of tissue H2S levels is thought to contribute to an ageing phenotype in many tissues across animal models. Whether H2S is altered in HGPS is unknown. We investigated hepatic H2S production capacity and transcript, protein and enzymatic activity of proteins that regulate hepatic H2S production and disposal in a mouse model of HGPS (G609G mice, mutated Lmna gene equivalent to a causative mutation in HGPS patients). G609G mice were maintained on either regular chow (RC) or high fat diet (HFD), as HFD has been previously shown to significantly extend lifespan of G609G mice, and compared to wild type (WT) mice maintained on RC. RC fed G609G mice had significantly reduced hepatic H2S production capacity relative to WT mice, with a compensatory elevation in mRNA transcripts associated with several H2S production enzymes, including cystathionine-γ-lyase (CSE). H2S levels and CSE protein were partially rescued in HFD fed G609G mice. As current treatments for patients with HGPS have failed to confer significant improvements to symptoms or longevity, the need for novel therapeutic targets is acute and the regulation of H2S through dietary or pharmacological means may be a promising new avenue for research.
Collapse
Affiliation(s)
- Stephen E. Wilkie
- Glasgow Ageing Research Network (GARNER), School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
- Division of Molecular Metabolism, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Solna 171 65, Sweden
| | - Diana E. Marcu
- Glasgow Ageing Research Network (GARNER), School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Roderick N. Carter
- Molecular Metabolism Group, University/BHF Centre for Cardiovascular Sciences, Queens Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Nicholas M. Morton
- Molecular Metabolism Group, University/BHF Centre for Cardiovascular Sciences, Queens Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Susana Gonzalo
- Department of Biochemistry and Molecular Biology, Edward A. Doisy Research Center, Saint Louis University School of Medicine, MO 63104, USA
| | - Colin Selman
- Glasgow Ageing Research Network (GARNER), School of Biodiversity, One Health and Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| |
Collapse
|
17
|
Purhonen J, Banerjee R, Wanne V, Sipari N, Mörgelin M, Fellman V, Kallijärvi J. Mitochondrial complex III deficiency drives c-MYC overexpression and illicit cell cycle entry leading to senescence and segmental progeria. Nat Commun 2023; 14:2356. [PMID: 37095097 PMCID: PMC10126100 DOI: 10.1038/s41467-023-38027-1] [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: 10/18/2022] [Accepted: 04/12/2023] [Indexed: 04/26/2023] Open
Abstract
Accumulating evidence suggests mitochondria as key modulators of normal and premature aging, yet whether primary oxidative phosphorylation (OXPHOS) deficiency can cause progeroid disease remains unclear. Here, we show that mice with severe isolated respiratory complex III (CIII) deficiency display nuclear DNA damage, cell cycle arrest, aberrant mitoses, and cellular senescence in the affected organs such as liver and kidney, and a systemic phenotype resembling juvenile-onset progeroid syndromes. Mechanistically, CIII deficiency triggers presymptomatic cancer-like c-MYC upregulation followed by excessive anabolic metabolism and illicit cell proliferation against lack of energy and biosynthetic precursors. Transgenic alternative oxidase dampens mitochondrial integrated stress response and the c-MYC induction, suppresses the illicit proliferation, and prevents juvenile lethality despite that canonical OXPHOS-linked functions remain uncorrected. Inhibition of c-MYC with the dominant-negative Omomyc protein relieves the DNA damage in CIII-deficient hepatocytes in vivo. Our results connect primary OXPHOS deficiency to genomic instability and progeroid pathogenesis and suggest that targeting c-MYC and aberrant cell proliferation may be therapeutic in mitochondrial diseases.
Collapse
Affiliation(s)
- Janne Purhonen
- Folkhälsan Research Center, Haartmaninkatu 8, 00290, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, P.O.Box 63, 00014, Helsinki, Finland
| | - Rishi Banerjee
- Folkhälsan Research Center, Haartmaninkatu 8, 00290, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, P.O.Box 63, 00014, Helsinki, Finland
| | - Vilma Wanne
- Folkhälsan Research Center, Haartmaninkatu 8, 00290, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, P.O.Box 63, 00014, Helsinki, Finland
| | - Nina Sipari
- Viikki Metabolomics Unit, University of Helsinki, P.O.Box 65, Helsinki, Finland
| | - Matthias Mörgelin
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, P.O.Box 117, 221 00, Lund, Sweden
- Colzyx AB, Scheelevägen 2, 22381, Lund, Sweden
| | - Vineta Fellman
- Folkhälsan Research Center, Haartmaninkatu 8, 00290, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, P.O.Box 63, 00014, Helsinki, Finland
- Department of Clinical Sciences, Lund, Pediatrics, Lund University, P.O.Box 117, 221 00, Lund, Sweden
- Children's Hospital, Clinicum, University of Helsinki, P.O. Box 22, 00014, Helsinki, Finland
| | - Jukka Kallijärvi
- Folkhälsan Research Center, Haartmaninkatu 8, 00290, Helsinki, Finland.
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, P.O.Box 63, 00014, Helsinki, Finland.
| |
Collapse
|
18
|
Prasad KN. A micronutrient mixture with collagen peptides, probiotics, cannabidiol, and diet may reduce aging, and development and progression of age-related alzheimer's disease, and improve its treatment. Mech Ageing Dev 2023; 210:111757. [PMID: 36460123 DOI: 10.1016/j.mad.2022.111757] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 11/30/2022]
Abstract
Human aging involves gradual decline in organ functions leading to organ specific age-related chronic diseases such as Alzheimer's disease (AD). Although advances in the development of new drugs, novel surgical procedures, improved diet and lifestyle, have resulted in doubling of lifespan of humans, the quality of life in many cases remains poor because of increased incidence of age-related chronic diseases. Using experimental models of accelerated aging, several cellular defects associated with aging and AD have been identified. Some cellular defects due to increased oxidative stress, chronic inflammation, autophagy defects, mitochondrial dysfunction, and imbalances in the composition probiotics in favor of harmful bacteria over beneficial bacteria are common to both aging and AD, while others such as telomere attrition, loss of collagen, elastin, and hyaluronic acid, failure of DNA repair system, and impaired immune function are unique to aging; and some such as increased production of beta-amyloids, hyperphosphorylation of tau protein, and abnormal behaviors are unique to AD. It is suggested that supplementation with a micronutrient mixture, probiotics, collagen peptides, CBD, and modifications in the diet and lifestyle may reduce the aging processes, and the development, progression of AD, and improve the treatments of this disease.
Collapse
Affiliation(s)
- Kedar N Prasad
- Engage Global, Inc. 245 El Faisan Dr., San Rafael, CA 94903, USA.
| |
Collapse
|
19
|
Yang JH, Hayano M, Griffin PT, Amorim JA, Bonkowski MS, Apostolides JK, Salfati EL, Blanchette M, Munding EM, Bhakta M, Chew YC, Guo W, Yang X, Maybury-Lewis S, Tian X, Ross JM, Coppotelli G, Meer MV, Rogers-Hammond R, Vera DL, Lu YR, Pippin JW, Creswell ML, Dou Z, Xu C, Mitchell SJ, Das A, O'Connell BL, Thakur S, Kane AE, Su Q, Mohri Y, Nishimura EK, Schaevitz L, Garg N, Balta AM, Rego MA, Gregory-Ksander M, Jakobs TC, Zhong L, Wakimoto H, El Andari J, Grimm D, Mostoslavsky R, Wagers AJ, Tsubota K, Bonasera SJ, Palmeira CM, Seidman JG, Seidman CE, Wolf NS, Kreiling JA, Sedivy JM, Murphy GF, Green RE, Garcia BA, Berger SL, Oberdoerffer P, Shankland SJ, Gladyshev VN, Ksander BR, Pfenning AR, Rajman LA, Sinclair DA. Loss of epigenetic information as a cause of mammalian aging. Cell 2023; 186:305-326.e27. [PMID: 36638792 PMCID: PMC10166133 DOI: 10.1016/j.cell.2022.12.027] [Citation(s) in RCA: 252] [Impact Index Per Article: 252.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 08/09/2022] [Accepted: 12/15/2022] [Indexed: 01/13/2023]
Abstract
All living things experience an increase in entropy, manifested as a loss of genetic and epigenetic information. In yeast, epigenetic information is lost over time due to the relocalization of chromatin-modifying proteins to DNA breaks, causing cells to lose their identity, a hallmark of yeast aging. Using a system called "ICE" (inducible changes to the epigenome), we find that the act of faithful DNA repair advances aging at physiological, cognitive, and molecular levels, including erosion of the epigenetic landscape, cellular exdifferentiation, senescence, and advancement of the DNA methylation clock, which can be reversed by OSK-mediated rejuvenation. These data are consistent with the information theory of aging, which states that a loss of epigenetic information is a reversible cause of aging.
Collapse
Affiliation(s)
- Jae-Hyun Yang
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA, USA.
| | - Motoshi Hayano
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA, USA; Department of Ophthalmology, Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Patrick T Griffin
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA, USA
| | - João A Amorim
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA, USA; IIIUC-Institute of Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Michael S Bonkowski
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA, USA
| | - John K Apostolides
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Elias L Salfati
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA, USA
| | | | | | - Mital Bhakta
- Cantata/Dovetail Genomics, Scotts Valley, CA, USA
| | | | - Wei Guo
- Zymo Research Corporation, Irvine, CA, USA
| | | | - Sun Maybury-Lewis
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA, USA
| | - Xiao Tian
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA, USA
| | - Jaime M Ross
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA, USA
| | - Giuseppe Coppotelli
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA, USA
| | - Margarita V Meer
- Department of Medicine, Brigham and Women's Hospital, HMS, Boston, MA, USA
| | - Ryan Rogers-Hammond
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA, USA
| | - Daniel L Vera
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA, USA
| | - Yuancheng Ryan Lu
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA, USA
| | - Jeffrey W Pippin
- Division of Nephrology, University of Washington, Seattle, WA, USA
| | - Michael L Creswell
- Division of Nephrology, University of Washington, Seattle, WA, USA; Georgetown University School of Medicine, Washington, DC, USA
| | - Zhixun Dou
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Caiyue Xu
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Abhirup Das
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA, USA; Department of Pharmacology, UNSW, Sydney, NSW, Australia
| | | | - Sachin Thakur
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA, USA
| | - Alice E Kane
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA, USA
| | - Qiao Su
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Yasuaki Mohri
- Department of Stem Cell Biology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Emi K Nishimura
- Department of Stem Cell Biology, Tokyo Medical and Dental University, Tokyo, Japan
| | | | - Neha Garg
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA, USA
| | - Ana-Maria Balta
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA, USA
| | - Meghan A Rego
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA, USA
| | | | - Tatjana C Jakobs
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, HMS, Boston, MA, USA
| | - Lei Zhong
- The Massachusetts General Hospital Cancer Center, HMS, Boston, MA, USA
| | | | - Jihad El Andari
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty, University of Heidelberg, BioQuant, Heidelberg, Germany
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty, University of Heidelberg, BioQuant, Heidelberg, Germany
| | - Raul Mostoslavsky
- The Massachusetts General Hospital Cancer Center, HMS, Boston, MA, USA
| | - Amy J Wagers
- Paul F. Glenn Center for Biology of Aging Research, Harvard Stem Cell Institute, Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA; Joslin Diabetes Center, Boston, MA, USA
| | - Kazuo Tsubota
- Department of Ophthalmology, Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Stephen J Bonasera
- Division of Geriatrics, University of Nebraska Medical Center, Durham Research Center II, Omaha, NE, USA
| | - Carlos M Palmeira
- Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | | | | | - Norman S Wolf
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Jill A Kreiling
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - John M Sedivy
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - George F Murphy
- Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Richard E Green
- Department of Biomolecular Engineering, UCSC, Santa Cruz, CA, USA
| | - Benjamin A Garcia
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Shelley L Berger
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Vadim N Gladyshev
- Department of Medicine, Brigham and Women's Hospital, HMS, Boston, MA, USA
| | - Bruce R Ksander
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, HMS, Boston, MA, USA
| | - Andreas R Pfenning
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Luis A Rajman
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA, USA
| | - David A Sinclair
- Paul F. Glenn Center for Biology of Aging Research, Department of Genetics, Blavatnik Institute, Harvard Medical School (HMS), Boston, MA, USA.
| |
Collapse
|
20
|
Yazar V, Dawson VL, Dawson TM, Kang SU. DNA Methylation Signature of Aging: Potential Impact on the Pathogenesis of Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2023; 13:145-164. [PMID: 36710687 PMCID: PMC10041453 DOI: 10.3233/jpd-223517] [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: 01/25/2023]
Abstract
Regulation of gene expression by epigenetic modifications means lasting and heritable changes in the function of genes without alterations in the DNA sequence. Of all epigenetic mechanisms identified thus far, DNA methylation has been of particular interest in both aging and age-related disease research over the last decade given the consistency of site-specific DNA methylation changes during aging that can predict future health and lifespan. An increasing line of evidence has implied the dynamic nature of DNA (de)methylation events that occur throughout the lifespan has a role in the pathophysiology of aging and age-associated neurodegenerative conditions, including Parkinson's disease (PD). In this regard, PD methylome shows, to some extent, similar genome-wide changes observed in the methylome of healthy individuals of matching age. In this review, we start by providing a brief overview of studies outlining global patterns of DNA methylation, then its mechanisms and regulation, within the context of aging and PD. Considering diverging lines of evidence from different experimental and animal models of neurodegeneration and how they combine to shape our current understanding of tissue-specific changes in DNA methylome in health and disease, we report a high-level comparison of the genomic methylation landscapes of brain, with an emphasis on dopaminergic neurons in PD and in natural aging. We believe this will be particularly useful for systematically dissecting overlapping genome-wide alterations in DNA methylation during PD and healthy aging, and for improving our knowledge of PD-specific changes in methylation patterns independent of aging process.
Collapse
Affiliation(s)
- Volkan Yazar
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA, USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA, USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA, USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA, USA
| | - Sung-Ung Kang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
21
|
Butylina M, Föger-Samwald U, Gamsjaeger S, Wahl-Figlash K, Kothmayer M, Paschalis EP, Pusch O, Pietschmann P. Nothobranchius furzeri, the Turquoise Killifish: A Model of Age-Related Osteoporosis? Gerontology 2022; 68:1415-1427. [PMID: 35472763 PMCID: PMC9838087 DOI: 10.1159/000524300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 03/16/2022] [Indexed: 01/17/2023] Open
Abstract
INTRODUCTION Osteoporosis is a frequent age-related disease, which affects millions of people worldwide. Despite significant progress in the treatment of the disease, a high number of patients still are underdiagnosed and undertreated. Therefore, novel animal models for the investigation of the disease are necessary. Nothobranchius furzeri is the shortest-lived vertebrate (with a lifespan of 3-7 months) that can be kept in captivity. Although it is an established model for aging research, studies on bone are lacking. The aim of this study was therefore to characterize N. furzeri as a potential model for age-related osteoporosis. MATERIALS AND METHODS Bone properties of aging N. furzeri were investigated in male and female fish of the Gona Re Zhou strain, which were between 8 and 20 weeks old. Micro-computed tomography (Scanco Medical µCT35) was performed to determine the bone properties of the vertebral bodies. Bone structure and remodeling were investigated by different histological staining techniques and histomorphometry. The chemical composition of fish vertebrae and intervertebral discs was analyzed by Raman microspectroscopy. RESULTS Osteoblasts, mono- and multinucleated osteoclasts but no osteocytes could be observed in the vertebral area of N. furzeri. Histomorphometric evaluations revealed a significant decrease of the number of osteoblasts/bone perimeter and for osteoid volume/bone volume (BV) a trend toward a decrease in old male N. furzeri. Comparing male and female fish, males showed higher BV densities and cortical thickness. The relative values of the bone volume density of 20-week-old male N. furzeri were significantly lower than 10-week-old ones. The mineral to matrix ratio increased with age in male and female fish. In the intervertebral discs, proteoglycans in relation to the organic matrix were significantly lower in older female fish. CONCLUSION Our finding of a lack of osteocytes is in agreement with the fact that N. furzeri belongs to the evolutionarily advanced teleost fish. Furthermore, not only age-specific but also sex-specific differences were visible in the bone properties of N. furzeri, which can be taken into consideration for the study of gender aspects of age-related musculoskeletal diseases.
Collapse
Affiliation(s)
- Maria Butylina
- Institute for Pathophysiology and Allergy Research (IPA), Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Ursula Föger-Samwald
- Institute for Pathophysiology and Allergy Research (IPA), Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria,*Ursula Föger-Samwald,
| | - Sonja Gamsjaeger
- 1st Medical Department, Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of WGKK and AUVA Trauma Centre Meidling, Hanusch Hospital, Vienna, Austria
| | - Katharina Wahl-Figlash
- Institute for Pathophysiology and Allergy Research (IPA), Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Michael Kothmayer
- Center of Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Eleftherios P. Paschalis
- 1st Medical Department, Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of WGKK and AUVA Trauma Centre Meidling, Hanusch Hospital, Vienna, Austria
| | - Oliver Pusch
- Center of Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Peter Pietschmann
- Institute for Pathophysiology and Allergy Research (IPA), Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
22
|
Ward JM, Vogel P, Sundberg JP. Brain and spinal cord lesions in 28 inbred strains of aging mice. Vet Pathol 2022; 59:1047-1055. [PMID: 36062914 DOI: 10.1177/03009858221120009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Brain and spinal cord histopathology findings in male and female 20-month-old mice in a large-scale aging study of 28 inbred Jackson Laboratory mouse strains from 7 genetic families are described. Brain sections from selected strains at 12 and 24 months of age or older were also reviewed. Common lesions include axonal dystrophy in the gracile and/or cuneate nucleus in the sensory tract of the dorsal medulla and in the spinal cord in all strains. Hirano-like bodies were seen in 24/28 strains, and mineralization was observed in the thalamus of 9/28 strains. Less common lesions were also seen in the cerebellum, cerebral cortex, and other brain areas. No brain or spinal cord tumors were found. Evidence of an impairment of the ubiquitin-proteasome system (UPS) and/or suspected autophagy was manifested as medullary axonal dystrophy with intra-axonal granular eosinophilic bodies and LC3B immunohistochemistry in most strains. RIIIS/J, the most severely affected strain, showed moderate axonal dystrophy at 12 months, which progressed to severe lesions at 20 months. Comparative pathology in various species is discussed.
Collapse
Affiliation(s)
- Jerrold M Ward
- The Jackson Laboratory, Bar Harbor, ME.,Global Vet Pathology, Montgomery Village, MD
| | - Peter Vogel
- St. Jude Children's Research Hospital, Memphis, TN
| | | |
Collapse
|
23
|
Salazar-Terreros MJ, Vernot JP. In Vitro and In Vivo Modeling of Normal and Leukemic Bone Marrow Niches: Cellular Senescence Contribution to Leukemia Induction and Progression. Int J Mol Sci 2022; 23:7350. [PMID: 35806354 PMCID: PMC9266537 DOI: 10.3390/ijms23137350] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 12/16/2022] Open
Abstract
Cellular senescence is recognized as a dynamic process in which cells evolve and adapt in a context dependent manner; consequently, senescent cells can exert both beneficial and deleterious effects on their surroundings. Specifically, senescent mesenchymal stromal cells (MSC) in the bone marrow (BM) have been linked to the generation of a supporting microenvironment that enhances malignant cell survival. However, the study of MSC's senescence role in leukemia development has been straitened not only by the availability of suitable models that faithfully reflect the structural complexity and biological diversity of the events triggered in the BM, but also by the lack of a universal, standardized method to measure senescence. Despite these constraints, two- and three dimensional in vitro models have been continuously improved in terms of cell culture techniques, support materials and analysis methods; in addition, research on animal models tends to focus on the development of techniques that allow tracking leukemic and senescent cells in the living organism, as well as to modify the available mice strains to generate individuals that mimic human BM characteristics. Here, we present the main advances in leukemic niche modeling, discussing advantages and limitations of the different systems, focusing on the contribution of senescent MSC to leukemia progression.
Collapse
Affiliation(s)
- Myriam Janeth Salazar-Terreros
- Grupo de Investigación Fisiología Celular y Molecular, Facultad de Medicina, Universidad Nacional de Colombia, Bogota 111321, Colombia;
| | - Jean-Paul Vernot
- Grupo de Investigación Fisiología Celular y Molecular, Facultad de Medicina, Universidad Nacional de Colombia, Bogota 111321, Colombia;
- Instituto de Investigaciones Biomédicas, Facultad de Medicina, Universidad Nacional de Colombia, Bogota 111321, Colombia
| |
Collapse
|
24
|
Rempuia V, Anima B, Jeremy M, Gurusubramanian G, Pankaj PP, Kharwar RK, Roy VK. Effects of metformin on the uterus of d-galactose-induced aging mice: Histomorphometric, immunohistochemical localization (B-cell lymphoma 2, Bcl2-associated X protein, and active capase3), and oxidative stress study. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART A, ECOLOGICAL AND INTEGRATIVE PHYSIOLOGY 2022; 337:600-611. [PMID: 35286779 DOI: 10.1002/jez.2592] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/18/2022] [Accepted: 02/23/2022] [Indexed: 06/14/2023]
Abstract
d-galactose (DG)-induced rodent aging model has widely been used for the study of age-related dysfunctions of various organs, including gonads and uterus. Antidiabetic drug metformin has gained an attention as antiaging drug in model organism and human but its effect on uterus has not been studied in relation to induced aging. Therefore, we investigated the effect of metformin on uterus of DG-induced aging mice model. Mice were randomly divided into three groups, that is, control (CN), DG-induced aging model and aging model treated with metformin. Histomorphometric results showed significantly decreased number of uterine glands, endometrial thickness, and increased luminal epithelium height in aging model. Furthermore, metformin resumed the number of uterine glands, endometrial thickness, and luminal epithelium height up to CN group. Metformin has also significantly decreased the age-associated oxidative stress (malondialdehyde and lipid hydroperoxide). Superoxide dismutase was significantly decreased in both treated groups compared to the CN group. However, catalase and glutathione peroxidase enzymes were significantly increased by metformin compared to the aging model. Immunostaining of active caspase3 and BAX were intense in the endometrium of aging model compare to CN- and metformin-treated groups. Localization of B-cell lymphoma 2 (Bcl2) showed intense immunostaining in the uterus of CN- and metformin-treated groups, with mild immunostaining in aging model. Our observations suggested that metformin treatment might be helpful for management of age-associated uterine dysfunctions. Moreover, it may be concluded that metformin might ameliorate uterine dysfunctions by reducing oxidative stress, suppressing apoptosis, and increasing the survival/antiapoptotic protein Bcl2.
Collapse
Affiliation(s)
- Vanlal Rempuia
- Department of Zoology, Mizoram University, Aizawl, Mizoram, India
| | - Borgohain Anima
- Department of Zoology, Mizoram University, Aizawl, Mizoram, India
| | | | | | - Pranay P Pankaj
- Department of Zoology, Nagaland University, Lumami, Nagaland, India
| | - Rajesh K Kharwar
- Department of Zoology, Kutir Post Graduate College, Chakkey, Jaunpur, India
| | - Vikas K Roy
- Department of Zoology, Mizoram University, Aizawl, Mizoram, India
| |
Collapse
|
25
|
Induction of Accelerated Aging in a Mouse Model. Cells 2022; 11:cells11091418. [PMID: 35563724 PMCID: PMC9102583 DOI: 10.3390/cells11091418] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 04/11/2022] [Accepted: 04/20/2022] [Indexed: 12/12/2022] Open
Abstract
With the global increase of the elderly population, the improvement of the treatment for various aging-related diseases and the extension of a healthy lifespan have become some of the most important current medical issues. In order to understand the developmental mechanisms of aging and aging-related disorders, animal models are essential to conduct relevant studies. Among them, mice have become one of the most prevalently used model animals for aging-related studies due to their high similarity to humans in terms of genetic background and physiological structure, as well as their short lifespan and ease of reproduction. This review will discuss some of the common and emerging mouse models of accelerated aging and related chronic diseases in recent years, with the aim of serving as a reference for future application in fundamental and translational research.
Collapse
|
26
|
Kudlova N, De Sanctis JB, Hajduch M. Cellular Senescence: Molecular Targets, Biomarkers, and Senolytic Drugs. Int J Mol Sci 2022; 23:ijms23084168. [PMID: 35456986 PMCID: PMC9028163 DOI: 10.3390/ijms23084168] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 11/16/2022] Open
Abstract
Cellular senescence is defined as irreversible cell cycle arrest caused by various processes that render viable cells non-functional, hampering normal tissue homeostasis. It has many endogenous and exogenous inducers, and is closely connected with age, age-related pathologies, DNA damage, degenerative disorders, tumor suppression and activation, wound healing, and tissue repair. However, the literature is replete with contradictory findings concerning its triggering mechanisms, specific biomarkers, and detection protocols. This may be partly due to the wide range of cellular and in vivo animal or human models of accelerated aging that have been used to study senescence and test senolytic drugs. This review summarizes recent findings concerning senescence, presents some widely used cellular and animal senescence models, and briefly describes the best-known senolytic agents.
Collapse
Affiliation(s)
- Natalie Kudlova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 77147 Olomouc, Czech Republic; (N.K.); (J.B.D.S.)
| | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 77147 Olomouc, Czech Republic; (N.K.); (J.B.D.S.)
- Institute of Molecular and Translational Medicine Czech Advanced Technologies and Research Institute, Palacky University, 77147 Olomouc, Czech Republic
| | - Marian Hajduch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 77147 Olomouc, Czech Republic; (N.K.); (J.B.D.S.)
- Institute of Molecular and Translational Medicine Czech Advanced Technologies and Research Institute, Palacky University, 77147 Olomouc, Czech Republic
- Correspondence: ; Tel.: +42-0-585632082
| |
Collapse
|
27
|
Walker RF. A Mechanistic Theory of Development-Aging Continuity in Humans and Other Mammals. Cells 2022; 11:cells11050917. [PMID: 35269539 PMCID: PMC8909351 DOI: 10.3390/cells11050917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 12/29/2022] Open
Abstract
There is consensus among biogerontologists that aging occurs either as the result of a purposeful genome-based, evolved program or due to spontaneous, randomly occurring, maladaptive events. Neither concept has yet identified a specific mechanism to explain aging’s emergence and acceleration during mid-life and beyond. Presented herein is a novel, unifying mechanism with empirical evidence that describes how aging becomes continuous with development. It assumes that aging emerges from deterioration of a regulatory process that directs morphogenesis and morphostasis. The regulatory system consists of a genome-wide “backbone” within which its specific genes are differentially expressed by the local epigenetic landscapes of cells and tissues within which they reside, thereby explaining its holistic nature. Morphostasis evolved in humans to ensure the nurturing of dependent offspring during the first decade of young adulthood when peak parental vitality prevails in the absence of aging. The strict redundancy of each morphostasis regulatory cycle requires sensitive dependence upon initial conditions to avoid initiating deterministic chaos behavior. However, when natural selection declines as midlife approaches, persistent, progressive, and specific DNA damage and misrepair changes the initial conditions of the regulatory process, thereby compromising morphostasis regulatory redundancy, instigating chaos, initiating senescence, and accelerating aging thereafter.
Collapse
|
28
|
Hey HWD, Lam WMR, Chan CX, Zhuo WH, Crombie EM, Tan TC, Chen WC, Cool S, Tsai SY. Paraspinal myopathy-induced intervertebral disc degeneration and thoracolumbar kyphosis in TSC1mKO mice model-a preliminary study. Spine J 2022; 22:483-494. [PMID: 34653636 DOI: 10.1016/j.spinee.2021.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND Increasing kyphosis of the spine in a human is a well-recognized clinical phenomenon that has been associated with back pain, poor physical performance and disability. The pathophysiology of age-related kyphosis is complex and has been associated with physiological changes in vertebrae, intervertebral disc (IVD) and paraspinal musculature, which current cross-sectional studies are unable to demonstrate. Creating an in vivo, paraspinal myopathic animal model for longitudinal study of these changes under controlled conditions is thus warranted. PURPOSE To confirm the TSC1 gene knockout effect on paraspinal muscle musculature; to analyze the development of spinal kyphosis, IVD degeneration and vertebra structural changes in a longitudinal manner to gain insights into the relationship between these processes. STUDY DESIGN A prospective cohort study of 28 female mice, divided into 4 groups-9-month-old TSC1mKO (n=7), 9-month-old control (n=4), 12-month-old TSC1mKO (n=8), and 12-month-old controls (n=9). METHODS High resolution micro-computed tomography was used to measure sagittal spinal alignment (Cobb's angle), vertebral height, vertebral body wedging, disc height index (DHI), disc wedge index (DWI), histomorphometry of trabecular bone and erector spinae muscle cross-sectional area. Paraspinal muscle specimens were harvested to assess for myopathic features with H&E stain, muscle fiber size, density of triangular fiber and central nucleus with WGA/DAPI stain, and percentage of fibers with PGC-1α stain. Intervertebral discs were evaluated for disc score using FAST stain. RESULTS Compared to controls, paraspinal muscle sections revealed features of myopathy in TSC1mKO mice similar to human sarcopenic paraspinal muscle. While there was significantly greater presence of small triangular fiber and density of central nucleus in 9-and 12-month-old TSC1mKO mice, significantly larger muscle fibers and decreased erector spinae muscle cross-sectional area were only found in 12-month-old TSC1mKO mice compared to controls. TSC1mKO mice developed accelerated thoracolumbar kyphosis, with significantly larger Cobb angles found only at 12 months old. Structural changes to the trabecular bone in terms of higher bone volume fraction and quality, as well as vertebral body wedging were observed only in 12-month-old TSC1mKO mice when compared to controls. Disc degeneration was observed as early as 9 months in TSC1mKO mice and corresponded with disc wedging. However, significant disc height loss was only observed when comparing 12-month-old TSC1mKO mice with controls. CONCLUSIONS This study successfully shows the TSC1 gene knockout effect on the development of paraspinal muscle myopathy in a mouse which is characteristic of sarcopenia. The TSC1mKO mice is by far the best model available to study the pathological consequence of sarcopenia on mice spine. With paraspinal muscle myopathy established as early as 9 months, TSC1mKO mice developed disc degeneration and disc wedging. This is followed by kyphosis of the spine at 12 months with concomitant disc height loss and vertebral body wedging due to bone remodeling. Age-related bone loss was not found in our study, suggesting osteoporosis and myopathy-induced vertebral body wedging are likely two independent processes. CLINICAL SIGNIFICANCE This is the first study to provide key insights on the early and late consequences of paraspinal myopathy on intervertebral disc degeneration, spinal kyphosis, and vertebral body changes. With this new understanding, future studies evaluating therapies for spinal degeneration may be performed to develop time-sensitive interventions.
Collapse
Affiliation(s)
- Hwee Weng Dennis Hey
- Department of Orthopaedic Surgery, National University Hospital, 1E Kent Ridge Road, NUHS Tower Block Level 11, Singapore 119228.
| | - Wing Moon Raymond Lam
- National University of Singapore Engineering Programme (NUSTEP), Department of Orthopedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore (NUS), 10 Medical Dr, Singapore 117597
| | - Chloe Xiaoyun Chan
- Department of Orthopaedic Surgery, National University Hospital, 1E Kent Ridge Road, NUHS Tower Block Level 11, Singapore 119228.
| | - Wen-Hai Zhuo
- National University of Singapore Engineering Programme (NUSTEP), Department of Orthopedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore (NUS), 10 Medical Dr, Singapore 117597
| | - Elisa Marie Crombie
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), 10 Medical Dr, Singapore 117597
| | - Tuan Chun Tan
- Institute of Medical Biology (IMB), Agency for Science, Technology and Research (A*STAR), 8a Biomedical Grove, Singapore 138648
| | - Way Cherng Chen
- Bruker Singapore Pte Ltd, Singapore, 30 Biopolis St, Singapore 138671
| | - Simon Cool
- Institute of Medical Biology (IMB), Agency for Science, Technology and Research (A*STAR), 8a Biomedical Grove, Singapore 138648
| | - Shih Yin Tsai
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), 10 Medical Dr, Singapore 117597
| |
Collapse
|
29
|
Sodium arsenite accelerates D-galactose-induced aging in the testis of the rat: Evidence for mitochondrial oxidative damage, NF-kB, JNK, and apoptosis pathways. Toxicology 2022; 470:153148. [DOI: 10.1016/j.tox.2022.153148] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 02/21/2022] [Accepted: 03/03/2022] [Indexed: 12/19/2022]
|
30
|
Riches-Suman K, Hussain A. Identifying and targeting the molecular signature of smooth muscle cells undergoing early vascular ageing. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166403. [DOI: 10.1016/j.bbadis.2022.166403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 03/25/2022] [Accepted: 03/25/2022] [Indexed: 10/18/2022]
|
31
|
Cooper TK, Meyerholz DK, Beck AP, Delaney MA, Piersigilli A, Southard TL, Brayton CF. Research-Relevant Conditions and Pathology of Laboratory Mice, Rats, Gerbils, Guinea Pigs, Hamsters, Naked Mole Rats, and Rabbits. ILAR J 2022; 62:77-132. [PMID: 34979559 DOI: 10.1093/ilar/ilab022] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/12/2021] [Indexed: 12/31/2022] Open
Abstract
Animals are valuable resources in biomedical research in investigations of biological processes, disease pathogenesis, therapeutic interventions, safety, toxicity, and carcinogenicity. Interpretation of data from animals requires knowledge not only of the processes or diseases (pathophysiology) under study but also recognition of spontaneous conditions and background lesions (pathology) that can influence or confound the study results. Species, strain/stock, sex, age, anatomy, physiology, spontaneous diseases (noninfectious and infectious), and neoplasia impact experimental results and interpretation as well as animal welfare. This review and the references selected aim to provide a pathology resource for researchers, pathologists, and veterinary personnel who strive to achieve research rigor and validity and must understand the spectrum of "normal" and expected conditions to accurately identify research-relevant experimental phenotypes as well as unusual illness, pathology, or other conditions that can compromise studies involving laboratory mice, rats, gerbils, guinea pigs, hamsters, naked mole rats, and rabbits.
Collapse
Affiliation(s)
- Timothy K Cooper
- Department of Comparative Medicine, Penn State Hershey Medical Center, Hershey, PA, USA
| | - David K Meyerholz
- Department of Pathology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa, USA
| | - Amanda P Beck
- Department of Pathology, Yeshiva University Albert Einstein College of Medicine, Bronx, New York, USA
| | - Martha A Delaney
- Zoological Pathology Program, University of Illinois at Urbana-Champaign College of Veterinary Medicine, Urbana-Champaign, Illinois, USA
| | - Alessandra Piersigilli
- Laboratory of Comparative Pathology and the Genetically Modified Animal Phenotyping Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Teresa L Southard
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, New York, USA
| | - Cory F Brayton
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
32
|
Nejmanová I, Vitverová B, Eissazadeh S, Tripská K, Igreja Sa IC, Hyšpler R, Němečkova I, Pericacho M, Nachtigal P. High Soluble Endoglin Levels Affect Aortic Vascular Function during Mice Aging. J Cardiovasc Dev Dis 2021; 8:jcdd8120173. [PMID: 34940528 PMCID: PMC8703792 DOI: 10.3390/jcdd8120173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/22/2021] [Accepted: 12/01/2021] [Indexed: 11/21/2022] Open
Abstract
Endoglin is a 180 kDa transmembrane glycoprotein that was demonstrated to be present in two different endoglin forms, namely membrane endoglin (Eng) and soluble endoglin (sEng). Increased sEng levels in the circulation have been detected in atherosclerosis, arterial hypertension, and type II diabetes mellitus. Moreover, sEng was shown to aggravate endothelial dysfunction when combined with a high-fat diet, suggesting it might be a risk factor for the development of endothelial dysfunction in combination with other risk factors. Therefore, this study hypothesized that high sEng levels exposure for 12 months combined with aging (an essential risk factor of atherosclerosis development) would aggravate vascular function in mouse aorta. Male transgenic mice with high levels of human sEng in plasma (Sol-Eng+) and their age-matched male transgenic littermates that do not develop high soluble endoglin (Control) on a chow diet were used. The aging process was initiated to contribute to endothelial dysfunction/atherosclerosis development, and it lasted 12 months. Wire myograph analysis showed impairment contractility in the Sol-Eng+ group when compared to the control group after KCl and PGF2α administration. Endothelium-dependent responsiveness to Ach was not significantly different between these groups. Western blot analysis revealed significantly decreased protein expression of Eng, p-eNOS, and ID1 expression in the Sol-Eng+ group compared to the control group suggesting reduced Eng signaling. In conclusion, we demonstrated for the first time that long-term exposure to high levels of sEng during aging results in alteration of vasoconstriction properties of the aorta, reduced eNOS phosphorylation, decreased Eng expression, and altered Eng signaling. These findings suggest that sEng can be considered a risk factor for the development of vascular dysfunction during aging and a potential therapeutical target for pharmacological intervention.
Collapse
Affiliation(s)
- Iveta Nejmanová
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (I.N.); (B.V.); (S.E.); (K.T.); (I.C.I.S.); (I.N.)
| | - Barbora Vitverová
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (I.N.); (B.V.); (S.E.); (K.T.); (I.C.I.S.); (I.N.)
| | - Samira Eissazadeh
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (I.N.); (B.V.); (S.E.); (K.T.); (I.C.I.S.); (I.N.)
| | - Katarina Tripská
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (I.N.); (B.V.); (S.E.); (K.T.); (I.C.I.S.); (I.N.)
| | - Ivone Cristina Igreja Sa
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (I.N.); (B.V.); (S.E.); (K.T.); (I.C.I.S.); (I.N.)
| | - Radomír Hyšpler
- Centrum for Research and Development, University Hospital, 500 05 Hradec Kralove, Czech Republic;
| | - Ivana Němečkova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (I.N.); (B.V.); (S.E.); (K.T.); (I.C.I.S.); (I.N.)
| | - Miguel Pericacho
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, Biomedical Research Institute of Salamanca (IBSAL), University of Salamanca, 37007 Salamanca, Spain;
| | - Petr Nachtigal
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic; (I.N.); (B.V.); (S.E.); (K.T.); (I.C.I.S.); (I.N.)
- Correspondence:
| |
Collapse
|
33
|
Hu Y, Zhu Y, Gerber SD, Osland JM, Chen M, Rao KA, Gu H, Yuan R. Deletion of Nrip1 delays skin aging by reducing adipose-derived mesenchymal stem cells (ADMSCs) senescence, and maintaining ADMSCs quiescence. GeroScience 2021; 43:1815-1833. [PMID: 33704619 PMCID: PMC8492836 DOI: 10.1007/s11357-021-00344-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 02/15/2021] [Indexed: 12/18/2022] Open
Abstract
Our previous studies found that deletion of nuclear receptor interacting protein 1 (Nrip1) extended longevity in female mice and delayed cell senescence. The current study investigates the role of NRIP1 in regulating functions of adipose-derived mesenchymal stem cells (ADMSCs) and explores the mechanisms of NRIP1 in skin aging. We first verified the skin aging phenotypes in young (6 months) and old (20 months) C57BL/6J (B6) mice and found deletion of Nrip1 can delay skin aging phenotypes, including reduced thickness of dermis and subcutaneous white adipose tissue (sWAT), as well as the accumulation of senescent cells in sWAT. In ADMSCs isolated from sWAT, we found that deletion of Nrip1 could decrease cell proliferation, prevent cell apoptosis, and suppress adipogenesis. Interestingly, deletion of Nrip1 also reduced cell senescence and maintain cell quiescence of ADMSCs. Moreover, the expressions of genes associated with senescence (p21, and p53), inflammation (p65, IL6, and IL1a), and growth factor (mTOR, Igf1) were reduced in Nrip1 knockout ADMSCs, as well as in siNrip1-treated ADMSCs. Suppression of Nrip1 by siNrip1 also decreased the expressions of mTOR, p-mTOR, p65, and p-p65 in ADMSCs. Reduced expressions of p65 and p-p65 were also confirmed in the skin of Nrip1 knockout mice. These findings suggest that NRIP1 plays an important role in delaying skin aging by reducing ADMSCs senescence and maintaining ADMSCs quiescence.
Collapse
Affiliation(s)
- Yu Hu
- Division of Geriatric Research, Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge Street Room 4361, Springfield, IL, 62702, USA
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 12 Jiangwangmiao Street, Nanjing, Jiangsu, China
| | - Yun Zhu
- Division of Geriatric Research, Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge Street Room 4361, Springfield, IL, 62702, USA
| | - Skyler D Gerber
- Division of Geriatric Research, Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge Street Room 4361, Springfield, IL, 62702, USA
| | - Jared M Osland
- Division of Geriatric Research, Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge Street Room 4361, Springfield, IL, 62702, USA
| | - Min Chen
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 12 Jiangwangmiao Street, Nanjing, Jiangsu, China
| | - Krishna A Rao
- Division of Hematology/Oncology, Department of Internal Medicine, Simmons Cancer Institute at Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Heng Gu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, 12 Jiangwangmiao Street, Nanjing, Jiangsu, China.
| | - Rong Yuan
- Division of Geriatric Research, Department of Internal Medicine, Southern Illinois University School of Medicine, 801 N. Rutledge Street Room 4361, Springfield, IL, 62702, USA.
| |
Collapse
|
34
|
Different Parts of the Chicken Embryo Egg Improve D-Galactose-Induced Aging in a Mice Model. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6654683. [PMID: 33997037 PMCID: PMC8099523 DOI: 10.1155/2021/6654683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/29/2021] [Accepted: 04/09/2021] [Indexed: 12/05/2022]
Abstract
Chick (CE) or duck embryo eggs are known for nutritional supplement foods in traditional East countries for physical fitness enhancement and postpartum conditioning for many years. In this study, we evaluated the effects of different parts of the 10-day CE (embryo: CEr, yolk: CEw, and chorioallantoic membrane: CEp) on the antifatigue and antiaging activities in a D-galactose- (D-gal) induced aging mice model. The results showed CEp obviously increased the muscle weight and the liver and muscle glycogen content and enhanced exercise performance. In the antiaging assay, CEp significantly increased the activity of superoxide dismutase (SOD) and Glutathione Peroxidase (GPx). Moreover, the immunohistochemistry results of NRF-2 and HO-1 were also detected in the livers of mice in the D-gal/CEp group. The only partially potential such as CEr might improve OFT function with TG level, and CEw had strange grip strength. Therefore, we suggest that CEp has a potent antifatigue ability and could minimize the occurrence of age-associated disorders, more than other parts of the 10 days chicken embryo egg.
Collapse
|
35
|
Wirth A, Wolf B, Huang CK, Glage S, Hofer SJ, Bankstahl M, Bär C, Thum T, Kahl KG, Sigrist SJ, Madeo F, Bankstahl JP, Ponimaskin E. Novel aspects of age-protection by spermidine supplementation are associated with preserved telomere length. GeroScience 2021; 43:673-690. [PMID: 33517527 PMCID: PMC8110654 DOI: 10.1007/s11357-020-00310-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 12/02/2020] [Indexed: 12/17/2022] Open
Abstract
Ageing provokes a plethora of molecular, cellular and physiological deteriorations, including heart failure, neurodegeneration, metabolic maladaptation, telomere attrition and hair loss. Interestingly, on the molecular level, the capacity to induce autophagy, a cellular recycling and cleaning process, declines with age across a large spectrum of model organisms and is thought to be responsible for a subset of age-induced changes. Here, we show that a 6-month administration of the natural autophagy inducer spermidine in the drinking water to aged mice is sufficient to significantly attenuate distinct age-associated phenotypes. These include modulation of brain glucose metabolism, suppression of distinct cardiac inflammation parameters, decreased number of pathological sights in kidney and liver and decrease of age-induced hair loss. Interestingly, spermidine-mediated age protection was associated with decreased telomere attrition, arguing in favour of a novel cellular mechanism behind the anti-ageing effects of spermidine administration.
Collapse
Affiliation(s)
- Alexander Wirth
- Cellular Neurophysiology, Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Bettina Wolf
- Preclinical Molecular Imaging, Department of Nuclear Medicine, Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Cheng-Kai Huang
- Institute of Molecular and Translational Therapeutic Strategies, Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Silke Glage
- Institute for Laboratory Animal Science, Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Sebastian J Hofer
- Institute of Molecular Biosciences, Karl-Franzens-Universität Graz, Humboldtstraße 50/EG, 8010, Graz, Austria
| | - Marion Bankstahl
- Institute for Laboratory Animal Science, Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Christian Bär
- Institute of Molecular and Translational Therapeutic Strategies, Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
- REBIRTH Center for Translational Regenerative Medicine, Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
- REBIRTH Center for Translational Regenerative Medicine, Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Nikolai-Fuchs-Straße 1, 30625, Hannover, Germany
| | - Kai G Kahl
- Dept. of Psychiatry; Social Psychiatry and Psychotherapy, Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Stephan J Sigrist
- Freie University Berlin, Institute of Biology, Takusstraße 6, 14195, Berlin, Germany
| | - Frank Madeo
- Institute of Molecular Biosciences, Karl-Franzens-Universität Graz, Humboldtstraße 50/EG, 8010, Graz, Austria
| | - Jens P Bankstahl
- Preclinical Molecular Imaging, Department of Nuclear Medicine, Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.
| | - Evgeni Ponimaskin
- Cellular Neurophysiology, Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, Gagarin ave. 23, Nizhny Novgorod, Russian Federation, 603950.
| |
Collapse
|
36
|
Borchman D. Lipid conformational order and the etiology of cataract and dry eye. J Lipid Res 2021; 62:100039. [PMID: 32554545 PMCID: PMC7910524 DOI: 10.1194/jlr.tr120000874] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/15/2020] [Indexed: 12/12/2022] Open
Abstract
Lens and tear film lipids are as unique as the systems they reside in. The major lipid of the human lens is dihydrosphingomylein, found in quantity only in the lens. The lens contains a cholesterol to phospholipid molar ratio as high as 10:1, more than anywhere else in the body. Lens lipids contribute to maintaining lens clarity, and alterations in lens lipid composition due to age are likely to contribute to cataract. Lens lipid composition reflects adaptations to the unique characteristics of the lens: no turnover of lens lipids or proteins; the lowest amount of oxygen of any tissue; and contains almost no intracellular organelles. The tear film lipid layer (TFLL) is also unique. The TFLL is a thin (100 nm) layer of lipid on the surface of tears covering the cornea that contributes to tear film stability. The major lipids of the TFLL are wax esters and cholesterol esters that are not found in the lens. The hydrocarbon chains associated with the esters are longer than those found anywhere else in the body (as long as 32 carbons), and many are branched. Changes in the composition and structure of the 30,000 different moieties of TFLL contribute to the instability of tears. The focus of the current review is how spectroscopy has been used to elucidate the relationships between lipid composition, conformational order and function, and the etiology of cataract and dry eye.
Collapse
Affiliation(s)
- Douglas Borchman
- Department of Ophthalmology and Visual Sciences, University of Louisville, Louisville, KY 40202.
| |
Collapse
|
37
|
Regenerative and Antioxidant Properties of Autologous Platelet-Rich Plasma Can Reserve the Aging Process of the Cornea in the Rat Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4127959. [PMID: 33299525 PMCID: PMC7704147 DOI: 10.1155/2020/4127959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 09/14/2020] [Accepted: 11/07/2020] [Indexed: 12/19/2022]
Abstract
Aging is a natural progressive decline in the biological function of cells. Age-related changes in the cornea can affect its ability to refract light or repair itself. Platelet-rich plasma (PRP) has a promising role in regenerative medicine and evidenced its efficacy in multiple fields, but in corneal aging has not yet been elucidated. The present work was performed to estimate the regenerative antioxidant effect of PRP on corneal aging in rats. Rats were assigned into two main groups: (GI) adult group and (GII) aged group. The adult group was divided into GIa (adult rats), GIb (adult-saline treated), and GIc (adult-PRP treated). The aged group was divided into GIIa (aged rats) and GIIb (aged, PRP treated). PRP was administered by a single subconjunctival injection. After 10 days, histological, ultrastructural, immunohistochemical, and morphometrical investigations were carried out. Examination of the corneal sections of the aged group revealed corneal epithelial thinning, shedding of the surface epithelium with loss of desmosomal junction, and irregularity in Bowman's membrane. Disorganized widely spaced collagen bundles and neovascularization were detected in corneal stroma associated with thickening in Descemet's membrane. Ultrastructural examination revealed shrunken hyperchromatic nuclei, swollen mitochondria, and scanty cytoplasm with a strong nuclear reaction for caspase-3 immunostaining. Moreover, antioxidant/free radicals' imbalance was detected by the increase of malondialdehyde (MDA) level with a decrease of glutathione peroxidase (GPx) and superoxide dismutase (SOD) levels. In contrast, GIIb (aged, PRP treated) section examination revealed a restoration of the thickness of the corneal epithelial layer and Descemet's membrane with an amendment of collagen fiber regularity that is associated with weak nuclear reaction to caspase-3 and recovery of the balance in the redox state. These findings proved the effectiveness of PRP as a promising regenerative treatment for the age-associated changes in the cornea.
Collapse
|
38
|
Badawi Y, Nishimune H. Impairment Mechanisms and Intervention Approaches for Aged Human Neuromuscular Junctions. Front Mol Neurosci 2020; 13:568426. [PMID: 33328881 PMCID: PMC7717980 DOI: 10.3389/fnmol.2020.568426] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 10/16/2020] [Indexed: 12/19/2022] Open
Abstract
The neuromuscular junction (NMJ) is a chemical synapse formed between a presynaptic motor neuron and a postsynaptic muscle cell. NMJs in most vertebrate species share many essential features; however, some differences distinguish human NMJs from others. This review will describe the pre- and postsynaptic structures of human NMJs and compare them to NMJs of laboratory animals. We will focus on age-dependent declines in function and changes in the structure of human NMJs. Furthermore, we will describe insights into the aging process revealed from mouse models of accelerated aging. In addition, we will compare aging phenotypes to other human pathologies that cause impairments of pre- and postsynaptic structures at NMJs. Finally, we will discuss potential intervention approaches for attenuating age-related NMJ dysfunction and sarcopenia in humans.
Collapse
Affiliation(s)
- Yomna Badawi
- Department of Anatomy and Cell Biology, University of Kansas School of Medicine, Kansas City, KS, United States
| | - Hiroshi Nishimune
- Department of Anatomy and Cell Biology, University of Kansas School of Medicine, Kansas City, KS, United States.,Neurobiology of Aging, Tokyo Metropolitan Institute of Gerontology, Itabashi, Japan
| |
Collapse
|
39
|
Abstract
Cellular senescence is a cell cycle arrest in damaged or aged cells. Although this represents a critical mechanism of tumor suppression, persistence of senescent cells during aging induces chronic inflammation and tissue dysfunction through the adoption of the senescence-associated secretory phenotype (SASP). This has been shown to promote the progression of age-associated diseases such as Alzheimer's disease, pulmonary fibrosis, and atherosclerosis. As the global population ages, the role of cellular senescence in disease is becoming a more critical area of research. In this review, mechanisms, biomarkers, and pathology of cellular senescence and SASP are described with a brief discussion of literature supporting a role for cellular senescence in veterinary diseases. Cell culture and mouse models used in senescence studies are also reviewed including the senescence-accelerated mouse-prone (SAMP), senescence pathway knockout mice (p53, p21 [CDKN1A], and p16 [CDKN2A]), and the more recently developed senolysis mice, which allow for direct visualization and elimination (or lysis) of senescent cells in live mice (p16-3MR and INK-ATTAC). These and other mouse models have demonstrated the importance of cellular senescence in embryogenesis and wound healing but have also identified a therapeutic benefit for targeting persistent senescent cells in age-associated diseases including neurodegeneration, diabetes, and cardiac fibrosis.
Collapse
Affiliation(s)
- Jessica Beck
- Laboratory of Human Carcinogenesis, 313611National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Purdue University, West Lafayette, IN, USA
| | - Izumi Horikawa
- Laboratory of Human Carcinogenesis, 313611National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Curtis Harris
- Laboratory of Human Carcinogenesis, 313611National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
40
|
Jayarajan J, Milsom MD. The role of the stem cell epigenome in normal aging and rejuvenative therapy. Hum Mol Genet 2020; 29:R236-R247. [PMID: 32744315 DOI: 10.1093/hmg/ddaa167] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 07/27/2020] [Accepted: 07/27/2020] [Indexed: 12/21/2022] Open
Abstract
Adult stem cells are ultimately responsible for the lifelong maintenance of regenerating of tissues during both homeostasis and following injury. Hence, the functional attrition of adult stem cells is thought to be an important driving factor behind the progressive functional decline of tissues and organs that is observed during aging. The mechanistic cause underlying this age-associated exhaustion of functional stem cells is likely to be complex and multifactorial. However, it is clear that progressive remodeling of the epigenome and the resulting deregulation of gene expression programs can be considered a hallmark of aging, and is likely a key factor in mediating altered biological function of aged stem cells. In this review, we outline cell intrinsic and extrinsic mediators of epigenome remodeling during aging; discuss how such changes can impact on stem cell function; and describe how resetting the aged epigenome may rejuvenate some of the biological characteristics of stem cells.
Collapse
Affiliation(s)
- Jeyan Jayarajan
- Division of Experimental Hematology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Michael D Milsom
- Division of Experimental Hematology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM).,DKFZ-ZMBH Alliance, Heidelberg, Germany
| |
Collapse
|
41
|
Progeria, atherosclerosis and clonal hematopoiesis: links and future perspectives. Mech Ageing Dev 2020; 192:111365. [PMID: 33007346 DOI: 10.1016/j.mad.2020.111365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 08/26/2020] [Accepted: 09/22/2020] [Indexed: 11/20/2022]
Abstract
The main actors of this review are Hutchinson-Gilford progeria syndrome (HGPS) and atherosclerosis. HGPS is a very rare disease with no definitively approved specific drugs. Atherosclerosis is a very common disease with a more consolidated treatment strategy. Nevertheless, common mechanisms are shared by both these diseases, particularly related to inflammation, oxidative and endoplasmic reticulum (ER) stress. Pathways regulated by Nuclear factor E2 related factor (Nrf2), Nuclear factor kappa B (NF-kB) and related to the Unfolded Protein Response (UPR) and ER stress are receiving increasing attention. In HGPS "not omnia" happen(s) "cum tempore", that means that HGPS patients have atherosclerotic complications before their time. The third actor is clonal hematopoiesis: it constitutes a link between ageing and atherosclerosis. This review aims to analyse the current knowledge of atherosclerosis and clonal hematopoiesis in order to suggest therapeutic strategies to correct the timing of the atherosclerosis progression in HGPS. The goal for HGPS is a shift from "not omnia cum tempore" to "omnia cum tempore" in terms of significant lifespan extension by postponing atherosclerosis-related complications.
Collapse
|
42
|
Brunetti D, Bottani E, Segala A, Marchet S, Rossi F, Orlando F, Malavolta M, Carruba MO, Lamperti C, Provinciali M, Nisoli E, Valerio A. Targeting Multiple Mitochondrial Processes by a Metabolic Modulator Prevents Sarcopenia and Cognitive Decline in SAMP8 Mice. Front Pharmacol 2020; 11:1171. [PMID: 32848778 PMCID: PMC7411305 DOI: 10.3389/fphar.2020.01171] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/17/2020] [Indexed: 12/31/2022] Open
Abstract
The age-dependent declines of skeletal muscle and cognitive functions often coexist in elderly subjects. The underlying pathophysiological mechanisms share common features of mitochondrial dysfunction, which plays a central role in the development of overt sarcopenia and/or dementia. Dietary supplementation with formulations of essential and branched-chain amino acids (EAA-BCAA) is a promising preventive strategy because it can preserve mitochondrial biogenesis and function. The senescence-accelerated mouse prone 8 (SAMP8) is considered an accurate model of age-related muscular and cognitive alterations. Hence, we aimed to investigate the progression of mitochondrial dysfunctions during muscular and cognitive aging of SAMP8 mice and to study the effects of a novel EAA-BCAA-based metabolic modulator on these changes. We evaluated body condition, motor endurance, and working memory of SAMP8 mice at 5, 9, 12, and 15 months of age. Parallel changes in protein levels of mitochondrial respiratory chain subunits, regulators of mitochondrial biogenesis and dynamics, and the antioxidant response, as well as respiratory complex activities, were measured in the quadriceps femoris and the hippocampus. The same variables were assessed in 12-month-old SAMP8 mice that had received dietary supplementation with the novel EAA-BCAA formulation, containing tricarboxylic acid cycle intermediates and co-factors (PD-0E7, 1.5 mg/kg/body weight/day in drinking water) for 3 months. Contrary to untreated mice, which had a significant molecular and phenotypic impairment, PD-0E7-treated mice showed preserved healthy body condition, muscle weight to body weight ratio, motor endurance, and working memory at 12 months of age. The PD-0E7 mixture increased the protein levels and the enzymatic activities of mitochondrial complex I, II, and IV and the expression of proliferator-activated receptor γ coactivator-1α, optic atrophy protein 1, and nuclear factor, erythroid 2 like 2 in muscles and hippocampi. The mitochondrial amyloid-β-degrading pitrilysin metallopeptidase 1 was upregulated, while amyloid precursor protein was reduced in the hippocampi of PD-0E7 treated mice. In conclusion, we show that a dietary supplement tailored to boost mitochondrial respiration preserves skeletal muscle and hippocampal mitochondrial quality control and health. When administered at the early onset of age-related physical and cognitive decline, this novel metabolic inducer counteracts the deleterious effects of precocious aging in both domains.
Collapse
Affiliation(s)
- Dario Brunetti
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy.,Medical Genetics and Neurogenetics Unit, Fondazione IRCCS Istituto Neurologico C. Besta, Milan, Italy
| | - Emanuela Bottani
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.,Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Agnese Segala
- Medical Genetics and Neurogenetics Unit, Fondazione IRCCS Istituto Neurologico C. Besta, Milan, Italy
| | - Silvia Marchet
- Medical Genetics and Neurogenetics Unit, Fondazione IRCCS Istituto Neurologico C. Besta, Milan, Italy
| | - Fabio Rossi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Fiorenza Orlando
- Advanced Technology Center for Aging Research, Scientific Technological Area, IRCCS INRCA, Ancona, Italy
| | - Marco Malavolta
- Advanced Technology Center for Aging Research, Scientific Technological Area, IRCCS INRCA, Ancona, Italy
| | - Michele O Carruba
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy.,Center for Study and Research on Obesity, University of Milan, Milan, Italy
| | - Costanza Lamperti
- Medical Genetics and Neurogenetics Unit, Fondazione IRCCS Istituto Neurologico C. Besta, Milan, Italy
| | - Mauro Provinciali
- Advanced Technology Center for Aging Research, Scientific Technological Area, IRCCS INRCA, Ancona, Italy
| | - Enzo Nisoli
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy.,Center for Study and Research on Obesity, University of Milan, Milan, Italy
| | - Alessandra Valerio
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
43
|
The role of adipose tissue senescence in obesity- and ageing-related metabolic disorders. Clin Sci (Lond) 2020; 134:315-330. [PMID: 31998947 DOI: 10.1042/cs20190966] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 01/07/2020] [Accepted: 01/14/2020] [Indexed: 12/19/2022]
Abstract
Adipose tissue as the largest energy reservoir and endocrine organ is essential for maintenance of systemic glucose, lipid and energy homeostasis, but these metabolic functions decline with ageing and obesity. Adipose tissue senescence is one of the common features in obesity and ageing. Although cellular senescence is a defensive mechanism preventing tumorigenesis, its occurrence in adipose tissue causatively induces defective adipogenesis, inflammation, aberrant adipocytokines production and insulin resistance, leading to adipose tissue dysfunction. In addition to these paracrine effects, adipose tissue senescence also triggers systemic inflammation and senescence as well as insulin resistance in the distal metabolic organs, resulting in Type 2 diabetes and other premature physiological declines. Multiple cell types including mature adipocytes, immune cells, endothelial cells and progenitor cells gradually senesce at different levels in different fat depots with ageing and obesity, highlighting the heterogeneity and complexity of adipose tissue senescence. In this review, we discuss the causes and consequences of adipose tissue senescence, and the major cell types responsible for adipose tissue senescence in ageing and obesity. In addition, we summarize the pharmacological approaches and lifestyle intervention targeting adipose tissue senescence for the treatment of obesity- and ageing-related metabolic diseases.
Collapse
|
44
|
Cerebral Mitochondrial Function and Cognitive Performance during Aging: A Longitudinal Study in NMRI Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4060769. [PMID: 32377297 PMCID: PMC7180425 DOI: 10.1155/2020/4060769] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/19/2020] [Accepted: 03/11/2020] [Indexed: 12/12/2022]
Abstract
Brain aging is one of the major risk factors for the development of several neurodegenerative diseases. Therefore, mitochondrial dysfunction plays an important role in processes of both, brain aging and neurodegeneration. Aged mice including NMRI mice are established model organisms to study physiological and molecular mechanisms of brain aging. However, longitudinal data evaluated in one cohort are rare but are important to understand the aging process of the brain throughout life, especially since pathological changes early in life might pave the way to neurodegeneration in advanced age. To assess the longitudinal course of brain aging, we used a cohort of female NMRI mice and measured brain mitochondrial function, cognitive performance, and molecular markers every 6 months until mice reached the age of 24 months. Furthermore, we measured citrate synthase activity and respiration of isolated brain mitochondria. Mice at the age of three months served as young controls. At six months of age, mitochondria-related genes (complex IV, creb-1, β-AMPK, and Tfam) were significantly elevated. Brain ATP levels were significantly reduced at an age of 18 months while mitochondria respiration was already reduced in middle-aged mice which is in accordance with the monitored impairments in cognitive tests. mRNA expression of genes involved in mitochondrial biogenesis (cAMP response element-binding protein 1 (creb-1), peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1-α), nuclear respiratory factor-1 (Nrf-1), mitochondrial transcription factor A (Tfam), growth-associated protein 43 (GAP43), and synaptophysin 1 (SYP1)) and the antioxidative defense system (catalase (Cat) and superoxide dismutase 2 (SOD2)) was measured and showed significantly decreased expression patterns in the brain starting at an age of 18 months. BDNF expression reached, a maximum after 6 months. On the basis of longitudinal data, our results demonstrate a close connection between the age-related decline of cognitive performance, energy metabolism, and mitochondrial biogenesis during the physiological brain aging process.
Collapse
|
45
|
Saxena S, Kumar S. Pharmacotherapy to gene editing: potential therapeutic approaches for Hutchinson-Gilford progeria syndrome. GeroScience 2020; 42:467-494. [PMID: 32048129 PMCID: PMC7205988 DOI: 10.1007/s11357-020-00167-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/04/2020] [Indexed: 12/11/2022] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS), commonly called progeria, is an extremely rare disorder that affects only one child per four million births. It is characterized by accelerated aging in affected individuals leading to premature death at an average age of 14.5 years due to cardiovascular complications. The main cause of HGPS is a sporadic autosomal dominant point mutation in LMNA gene resulting in differently spliced lamin A protein known as progerin. Accumulation of progerin under nuclear lamina and activation of its downstream effectors cause perturbation in cellular morphology and physiology which leads to a systemic disorder that mainly impairs the cardiovascular system, bones, skin, and overall growth. Till now, no cure has been found for this catastrophic disorder; however, several therapeutic strategies are under development. The current review focuses on the overall progress in the field of therapeutic approaches for the management/cure of HGPS. We have also discussed the new disease models that have been developed for the study of this rare disorder. Moreover, we have highlighted the therapeutic application of extracellular vesicles derived from stem cells against aging and aging-related disorders and, therefore, suggest the same for the treatment of HGPS.
Collapse
Affiliation(s)
- Saurabh Saxena
- Department of Medical Laboratory Sciences, Lovely Professional University, Jalandhar - Delhi G.T. Road, Phagwara, Punjab, 144411, India.
| | - Sanjeev Kumar
- Faculty of Technology and Sciences, Lovely Professional University, Jalandhar - Delhi G.T. Road, Phagwara, Punjab, 144411, India
| |
Collapse
|
46
|
Yousefzadeh MJ, Zhao J, Bukata C, Wade EA, McGowan SJ, Angelini LA, Bank MP, Gurkar AU, McGuckian CA, Calubag MF, Kato JI, Burd CE, Robbins PD, Niedernhofer LJ. Tissue specificity of senescent cell accumulation during physiologic and accelerated aging of mice. Aging Cell 2020; 19:e13094. [PMID: 31981461 PMCID: PMC7059165 DOI: 10.1111/acel.13094] [Citation(s) in RCA: 187] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 11/11/2019] [Accepted: 12/07/2019] [Indexed: 12/27/2022] Open
Abstract
Senescent cells accumulate with age in vertebrates and promote aging largely through their senescence‐associated secretory phenotype (SASP). Many types of stress induce senescence, including genotoxic stress. ERCC1‐XPF is a DNA repair endonuclease required for multiple DNA repair mechanisms that protect the nuclear genome. Humans or mice with reduced expression of this enzyme age rapidly due to increased levels of spontaneous, genotoxic stress. Here, we asked whether this corresponds to an increased level of senescent cells. p16Ink4a and p21Cip1 mRNA were increased ~15‐fold in peripheral lymphocytes from 4‐ to 5‐month‐old Ercc1−/∆ and 2.5‐year‐old wild‐type (WT) mice, suggesting that these animals exhibit a similar biological age. p16Ink4a and p21Cip1 mRNA were elevated in 10 of 13 tissues analyzed from 4‐ to 5‐month‐old Ercc1−/∆ mice, indicating where endogenous DNA damage drives senescence in vivo. Aged WT mice had similar increases of p16Ink4a and p21Cip1 mRNA in the same 10 tissues as the mutant mice. Senescence‐associated β–galactosidase activity and p21Cip1 protein also were increased in tissues of the progeroid and aged mice, while Lamin B1 mRNA and protein levels were diminished. In Ercc1−/Δ mice with a p16Ink4a luciferase reporter, bioluminescence rose steadily with age, particularly in lung, thymus, and pancreas. These data illustrate where senescence occurs with natural and accelerated aging in mice and the relative extent of senescence among tissues. Interestingly, senescence was greater in male mice until the end of life. The similarities between Ercc1−/∆ and aged WT mice support the conclusion that the DNA repair‐deficient mice accurately model the age‐related accumulation of senescent cells, albeit six‐times faster.
Collapse
Affiliation(s)
- Matthew J. Yousefzadeh
- Institute on the Biology of Aging and Metabolism University of Minnesota Minneapolis MN USA
- Department of Biochemistry, Molecular Biology and Biophysics University of Minnesota Minneapolis MN USA
- Department of Molecular Medicine Scripps Research Institute Jupiter FL USA
| | - Jing Zhao
- Department of Molecular Medicine Scripps Research Institute Jupiter FL USA
| | - Christina Bukata
- Department of Molecular Medicine Scripps Research Institute Jupiter FL USA
- Harriet L. Wilkes Honors CollegeFlorida Atlantic University Jupiter FL USA
| | - Erin A. Wade
- Department of Molecular Medicine Scripps Research Institute Jupiter FL USA
- Harriet L. Wilkes Honors CollegeFlorida Atlantic University Jupiter FL USA
| | - Sara J. McGowan
- Institute on the Biology of Aging and Metabolism University of Minnesota Minneapolis MN USA
- Department of Biochemistry, Molecular Biology and Biophysics University of Minnesota Minneapolis MN USA
- Department of Molecular Medicine Scripps Research Institute Jupiter FL USA
| | - Luise A. Angelini
- Institute on the Biology of Aging and Metabolism University of Minnesota Minneapolis MN USA
- Department of Biochemistry, Molecular Biology and Biophysics University of Minnesota Minneapolis MN USA
- Department of Molecular Medicine Scripps Research Institute Jupiter FL USA
| | - Michael P. Bank
- Department of Molecular Medicine Scripps Research Institute Jupiter FL USA
- Charles E. Schmidt College of Medicine Florida Atlantic University Boca Raton FL USA
| | - Aditi U. Gurkar
- Department of Molecular Medicine Scripps Research Institute Jupiter FL USA
- Department of Medicine University of Pittsburgh Pittsburgh PA USA
| | - Collin A. McGuckian
- Institute on the Biology of Aging and Metabolism University of Minnesota Minneapolis MN USA
- Department of Biochemistry, Molecular Biology and Biophysics University of Minnesota Minneapolis MN USA
- Department of Molecular Medicine Scripps Research Institute Jupiter FL USA
| | - Mariah F. Calubag
- Department of Molecular Medicine Scripps Research Institute Jupiter FL USA
- Harriet L. Wilkes Honors CollegeFlorida Atlantic University Jupiter FL USA
| | - Jonathan I. Kato
- Department of Molecular Medicine Scripps Research Institute Jupiter FL USA
- Harriet L. Wilkes Honors CollegeFlorida Atlantic University Jupiter FL USA
| | - Christin E. Burd
- Departments of Molecular Genetics and Cancer Biology and Genetics The Ohio State University Columbus OH USA
| | - Paul D. Robbins
- Institute on the Biology of Aging and Metabolism University of Minnesota Minneapolis MN USA
- Department of Biochemistry, Molecular Biology and Biophysics University of Minnesota Minneapolis MN USA
- Department of Molecular Medicine Scripps Research Institute Jupiter FL USA
| | - Laura J. Niedernhofer
- Institute on the Biology of Aging and Metabolism University of Minnesota Minneapolis MN USA
- Department of Biochemistry, Molecular Biology and Biophysics University of Minnesota Minneapolis MN USA
- Department of Molecular Medicine Scripps Research Institute Jupiter FL USA
| |
Collapse
|
47
|
Montano M, Bhasin S, D'Aquila RT, Erlandson KM, Evans WJ, Funderburg NT, Justice A, Ndhlovu LC, Ojikutu B, Pahor M, Pahwa S, Ryan AS, Schrack J, Schultz MB, Sebastiani P, Sinclair DA, Tripp J, Walker B, Womack JA, Yung R, Reeves RK. Harvard HIV and Aging Workshop: Perspectives and Priorities from Claude D. Pepper Centers and Centers for AIDS Research. AIDS Res Hum Retroviruses 2019; 35:999-1012. [PMID: 31456412 DOI: 10.1089/aid.2019.0130] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
People aging with HIV (PAWH) infection experience greater impairments in physical and cognitive function, in addition to higher rates of peripheral comorbid conditions (e.g., renal failure, diabetes, bone fracture, hypertension, cardiovascular disease, polypharmacy, and multimorbidity). While multifactorial drivers, including HIV infection itself, antiretroviral therapy-related toxicities, disparities in care, and biobehavioral factors, likely contribute, there remains an overarching question as to what are the relevant age-related mechanisms and models that could inform interventions that promote health span and life span in PAWH? This workshop was convened to hear from experts on the biology of aging and HIV researchers studying PAWH to focus on advancing investigations at the interface of HIV and Aging. In this study, we summarize the discussions from the Harvard Center for AIDS Research and Boston Claude D. Pepper cosponsored workshop on HIV and Aging, which took place in October 2018.
Collapse
Affiliation(s)
- Monty Montano
- Boston Pepper OAIC, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Men's Health: Aging and Metabolism, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Shalender Bhasin
- Boston Pepper OAIC, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Men's Health: Aging and Metabolism, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | | - William J Evans
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California
| | - Nicholas T Funderburg
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, Ohio State University, Columbus, Ohio
| | - Amy Justice
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
- VA Connecticut Healthcare System, West Haven, Connecticut
| | - Lishomwa C Ndhlovu
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii
| | - Bisola Ojikutu
- Division of Global Health Equity, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Marco Pahor
- Institute on Aging, Department of Aging and Geriatric Research, College of Medicine, University of Florida, Gainesville, Florida
| | - Savita Pahwa
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, Florida
| | - Alice S Ryan
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
- Baltimore Veterans Affairs Geriatric Research Education and Clinical Center and Research and Development Service, Baltimore, Maryland
| | - Jennifer Schrack
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Michael B Schultz
- Department of Genetics, Paul F. Glenn Labs for the Biology of Aging, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
| | - Paola Sebastiani
- Department of Biostatistics, Boston University, Boston, Massachusetts
| | - David A Sinclair
- Department of Genetics, Paul F. Glenn Labs for the Biology of Aging, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
| | - Julia Tripp
- Harvard University Center for AIDS Research, Cambridge, Massachusetts
| | - Bruce Walker
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts
| | - Julie A Womack
- VA Connecticut Healthcare System, West Haven, Connecticut
- Yale School of Nursing, West Haven, Connecticut
| | - Raymond Yung
- Division of Geriatric and Palliative Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - R Keith Reeves
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
48
|
Nagarajan P, Agudelo Garcia PA, Iyer CC, Popova LV, Arnold WD, Parthun MR. Early-onset aging and mitochondrial defects associated with loss of histone acetyltransferase 1 (Hat1). Aging Cell 2019; 18:e12992. [PMID: 31290578 PMCID: PMC6718594 DOI: 10.1111/acel.12992] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 05/07/2019] [Accepted: 05/27/2019] [Indexed: 12/18/2022] Open
Abstract
Histone acetyltransferase 1 (Hat1) is responsible for the acetylation of newly synthesized histone H4 on lysines 5 and 12 during the process of chromatin assembly. To understand the broader biological role of Hat1, we have generated a conditional mouse knockout model of this enzyme. We previously reported that Hat1 is required for viability and important for mammalian development and genome stability. In this study, we show that haploinsufficiency of Hat1 results in a significant decrease in lifespan. Defects observed in Hat1+/− mice are consistent with an early‐onset aging phenotype. These include lordokyphosis (hunchback), muscle atrophy, minor growth retardation, reduced subcutaneous fat, cancer, and paralysis. In addition, the expression of Hat1 is linked to the normal aging process as Hat1 mRNA and protein becomes undetectable in many tissues in old mice. At the cellular level, fibroblasts from Hat1 haploinsufficient embryos undergo early senescence and accumulate high levels of p21. Hat1+/− mouse embryonic fibroblasts (MEFs) display modest increases in endogenous DNA damage but have significantly higher levels of reactive oxygen species (ROS). Consistently, further studies show that Hat1−/− MEFs exhibit mitochondrial defects suggesting a critical role for Hat1 in mitochondrial function. Taken together, these data show that loss of Hat1 induces multiple hallmarks of early‐onset aging.
Collapse
Affiliation(s)
- Prabakaran Nagarajan
- Department of Biological Chemistry and Pharmacology, The Ohio State University Columbus Ohio
| | - Paula A. Agudelo Garcia
- Department of Biological Chemistry and Pharmacology, The Ohio State University Columbus Ohio
| | - Chitra C. Iyer
- Department of Neurology The Ohio State University Columbus Ohio
| | - Liudmila V. Popova
- Department of Biological Chemistry and Pharmacology, The Ohio State University Columbus Ohio
| | | | - Mark R. Parthun
- Department of Biological Chemistry and Pharmacology, The Ohio State University Columbus Ohio
| |
Collapse
|
49
|
Folgueras AR, Freitas-Rodríguez S, Velasco G, López-Otín C. Mouse Models to Disentangle the Hallmarks of Human Aging. Circ Res 2019; 123:905-924. [PMID: 30355076 DOI: 10.1161/circresaha.118.312204] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Model organisms have provided fundamental evidence that aging can be delayed and longevity extended. These findings gave rise to a new era in aging research aimed at elucidating the pathways and networks controlling this complex biological process. The identification of 9 hallmarks of aging has established a framework to evaluate the relative contribution of each hallmark and the interconnections among them. In this review, we revisit these hallmarks with the information obtained exclusively through the generation of genetically modified mouse models that have a significant impact on the aging process. We discuss within each hallmark those interventions that accelerate aging or that have been successful at increasing lifespan, with the final goal of identifying the most promising antiaging avenues based on the current knowledge provided by in vivo models.
Collapse
Affiliation(s)
- Alicia R Folgueras
- From the Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Spain
| | - Sandra Freitas-Rodríguez
- From the Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Spain
| | - Gloria Velasco
- From the Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Spain
| | - Carlos López-Otín
- From the Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Spain
| |
Collapse
|
50
|
Sundermann EE, Erlandson KM, Pope CN, Rubtsova A, Montoya J, Moore AA, Marzolini C, O'Brien KK, Pahwa S, Payne BA, Rubin LH, Walmsley S, Haughey NJ, Montano M, Karris MY, Margolick JB, Moore DJ. Current Challenges and Solutions in Research and Clinical Care of Older Persons Living with HIV: Findings Presented at the 9th International Workshop on HIV and Aging. AIDS Res Hum Retroviruses 2019; 35:985-998. [PMID: 31373216 PMCID: PMC6862962 DOI: 10.1089/aid.2019.0100] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
In the era of effective antiretroviral therapy, the number of older people with HIV (PWH) is increasing, and those aging with HIV are experiencing an increasing burden of age-associated comorbidities. Life expectancy among older PWH is approaching that of demographically comparable HIV-uninfected (HIV-) adults. With this changing demographic of PWH come new challenges for researchers and clinicians in how to identify, address, and manage the complex interplay of treated HIV infection and aging-associated factors. In response to these challenges, the annual International Workshop on HIV and Aging was initiated in 2009 as a multidisciplinary platform for scientific discourse on the research and clinical complications arising from the aging population of PWH. The multidisciplinary nature of the workshop has resulted in a wide range of topics addressed over the past 9 years, from basic mechanisms in aging and HIV pathogenesis, to epidemiology of aging within large cohorts, interventions, and implementation of clinical programs. Herein, we summarize the key topics discussed at the 9th Annual International Workshop on HIV and Aging 2018, including "inflammaging," mitochondrial dysfunction, exercise interventions, HIV-associated neurocognitive impairment, metabolic dysfunction, menopause, and polypharmacy. In addition to recent developments in research and clinical care, we discuss open questions and future research directions required to better understand the interaction of HIV and aging.
Collapse
Affiliation(s)
- Erin E. Sundermann
- Department of Psychiatry, University of California, San Diego, La Jolla, California
| | - Kristine M. Erlandson
- Department of Medicine, University of Colorado-Anschutz Medical Center, Aurora, Colorado
- Department of Epidemiology, School of Public Health, University of Colorado-Anschutz Medical Center, Aurora, Colorado
| | - Caitlin N. Pope
- Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Anna Rubtsova
- Department of Behavioral Sciences and Health Education, Emory University Rollins School of Public Health, Atlanta, Georgia
| | - Jessica Montoya
- Department of Psychiatry, University of California, San Diego, La Jolla, California
| | - Alison A. Moore
- Division of Geriatrics and Gerontology, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Catia Marzolini
- Division of Infectious Diseases and Hospital Epidemiology, Departments of Medicine and Clinical Research, University Hospital Basel and University of Basel, Basel, Switzerland
- The Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Kelly K. O'Brien
- Department of Physical Therapy, University of Toronto, Toronto, Ontario, Canada
- Institute of Health Policy, Management and Evaluation (IHPME), University of Toronto, Toronto, Ontario, Canada
- The Rehabilitation Sciences Institute (RSI), University of Toronto, Toronto, Ontario, Canada
| | - Savita Pahwa
- Department of Microbiology and Immunology and the Miami CFAR, University of Miami Miller School of Medicine, Miami, Florida
| | - Brendan A.I. Payne
- Wellcome Centre for Mitochondrial Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Leah H. Rubin
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Sharon Walmsley
- Toronto General Hospital, University Health Network, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Canada
| | - Norman J. Haughey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Monty Montano
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Maile Y. Karris
- Division of Infectious Diseases and Global Public Health, Department of Medicine, Unviersity of California San Diego, San Diego, California
| | - Joseph B. Margolick
- Department of Molecular Microbiology and Immunology, Environmental Health and Engineering, and Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - David J. Moore
- Department of Psychiatry, University of California, San Diego, La Jolla, California
| |
Collapse
|