1
|
Castro JP, Shindyapina AV, Barbieri A, Ying K, Strelkova OS, Paulo JA, Tyshkovskiy A, Meinl R, Kerepesi C, Petrashen AP, Mariotti M, Meer MV, Hu Y, Karamyshev A, Losyev G, Galhardo M, Logarinho E, Indzhykulian AA, Gygi SP, Sedivy JM, Manis JP, Gladyshev VN. Age-associated clonal B cells drive B cell lymphoma in mice. NATURE AGING 2024; 4:1403-1417. [PMID: 39117982 DOI: 10.1038/s43587-024-00671-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 06/19/2024] [Indexed: 08/10/2024]
Abstract
Although cancer is an age-related disease, how the processes of aging contribute to cancer progression is not well understood. In this study, we uncovered how mouse B cell lymphoma develops as a consequence of a naturally aged system. We show here that this malignancy is associated with an age-associated clonal B cell (ACBC) population that likely originates from age-associated B cells. Driven by c-Myc activation, promoter hypermethylation and somatic mutations, IgM+ ACBCs clonally expand independently of germinal centers and show increased biological age. ACBCs become self-sufficient and support malignancy when transferred into young recipients. Inhibition of mTOR or c-Myc in old mice attenuates pre-malignant changes in B cells during aging. Although the etiology of mouse and human B cell lymphomas is considered distinct, epigenetic changes in transformed mouse B cells are enriched for changes observed in human B cell lymphomas. Together, our findings characterize the spontaneous progression of cancer during aging through both cell-intrinsic and microenvironmental changes and suggest interventions for its prevention.
Collapse
Affiliation(s)
- José P Castro
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Aging and Aneuploidy Laboratory, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | | | | | - Kejun Ying
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Olga S Strelkova
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear Infirmary, Boston, MA, USA
- Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School, Boston, MA, USA
| | - João A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | | | - Rico Meinl
- Retro Biosciences, Redwood City, CA, USA
| | - Csaba Kerepesi
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Institute for Computer Science and Control (SZTAKI), Loránd Eötvös Research Network, Budapest, Hungary
| | - Anna P Petrashen
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Marco Mariotti
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Departament de Genètica, Microbiologia i Estadística, Universitat de Barcelona, Barcelona, Spain
| | - Margarita V Meer
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- San Diego Institute of Sciences, Altos Labs, San Diego, CA, USA
| | - Yan Hu
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Grigoriy Losyev
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mafalda Galhardo
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Elsa Logarinho
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Artur A Indzhykulian
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear Infirmary, Boston, MA, USA
- Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School, Boston, MA, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - John M Sedivy
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - John P Manis
- Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Vadim N Gladyshev
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Elies L, Guillaume E, Gorieu M, Neves P, Schorsch F. Historical Control Data of Spontaneous Pathological Findings in C57BL/6J Mice Used in 18-Month Dietary Carcinogenicity Assays. Toxicol Pathol 2024; 52:99-113. [PMID: 38757264 DOI: 10.1177/01926233241248658] [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: 05/18/2024]
Abstract
A retrospective analysis in C57BL6/J mice used in dietary carcinogenicity studies was performed to determine the survival rate, causes of death and incidences of spontaneous non-tumoral and tumoral findings. Data were collected from 1600 mice from control dose groups of sixteen 18-month carcinogenicity assays performed between 2003 and 2021 at the same test facility with similar environmental conditions and experimental procedures. The survival rate was high in both sexes (81%-85%) and the causes of humane euthanasia or death were mainly non-tumoral (chronic ulcerative dermatitis, atrial thrombosis). Benign tumors were more frequent than malignant tumors and females were more affected than males. Pituitary gland adenoma in females, lymphoma, bronchioloalveolar adenoma, and harderian gland adenoma in both sexes were the most common tumors. Systemic amyloidosis, the most frequent non-tumoral lesion, was observed variably across studies without sex predilection. The analysis by cohort (3 time periods of 6 years) showed a tendency toward higher incidences of lymphoma and pituitary gland adenoma and lower incidences of amyloidosis over time. The results presented here provide for the first time a robust set of control historical data in untreated C57BL/6J mice kept for 18 months contributing to build in depth knowledge of this animal model.
Collapse
Affiliation(s)
- Laëtitia Elies
- Charles River Laboratories, Saint-Germain-Nuelles, France
| | | | | | | | | |
Collapse
|
3
|
Mitchell SE, Togo J, Green CL, Derous D, Hambly C, Speakman JR. The Effects of Graded Levels of Calorie Restriction: XX. Impact of Long-Term Graded Calorie Restriction on Survival and Body Mass Dynamics in Male C57BL/6J Mice. J Gerontol A Biol Sci Med Sci 2023; 78:1953-1963. [PMID: 37354128 PMCID: PMC10613020 DOI: 10.1093/gerona/glad152] [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: 11/27/2022] [Indexed: 06/26/2023] Open
Abstract
Calorie restriction (CR) typically promotes a reduction in body mass, which correlates with increased lifespan. We evaluated the overall changes in survival, body mass dynamics, and body composition following long-term graded CR (580 days/19 months) in male C57BL/6J mice. Control mice (0% restriction) were fed ad libitum in the dark phase only (12-hour ad libitum [12AL]). CR groups were restricted by 10%-40% of their baseline food intake (10CR, 20CR, 30CR, and 40CR). Body mass was recorded daily, and body composition was measured at 8 time points. At 728 days/24 months, all surviving mice were culled. A gradation in survival rate over the CR groups was found. The pattern of body mass loss differed over the graded CR groups. Whereas the lower CR groups rapidly resumed an energy balance with no significant loss of fat or fat-free mass, changes in the 30 and 40CR groups were attributed to higher fat-free mass loss and protection of fat mass. Day-to-day changes in body mass were less variable under CR than for the 12AL group. There was no indication that body mass was influenced by external factors. Partial autocorrelation analysis examined the relationship between daily changes in body masses. A negative correlation between mass on Day 0 and Day +1 declined with age in the 12AL but not the CR groups. A reduction in the correlation with age suggested body mass homeostasis is a marker of aging that declines at the end of life and is protected by CR.
Collapse
Affiliation(s)
| | - Jacques Togo
- School of Biological Sciences, University of Aberdeen, Aberdeen, UK
| | - Cara L Green
- School of Biological Sciences, University of Aberdeen, Aberdeen, UK
| | - Davina Derous
- School of Biological Sciences, University of Aberdeen, Aberdeen, UK
| | - Catherine Hambly
- School of Biological Sciences, University of Aberdeen, Aberdeen, UK
| | - John R Speakman
- School of Biological Sciences, University of Aberdeen, Aberdeen, UK
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, P.R. China
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, P.R. China
- China Medical University, Shenyang, Liaoning, P.R. China
| |
Collapse
|
4
|
Padmanabhan P, Götz J. Clinical relevance of animal models in aging-related dementia research. NATURE AGING 2023; 3:481-493. [PMID: 37202516 DOI: 10.1038/s43587-023-00402-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 03/21/2023] [Indexed: 05/20/2023]
Abstract
Alzheimer's disease (AD) and other, less prevalent dementias are complex age-related disorders that exhibit multiple etiologies. Over the past decades, animal models have provided pathomechanistic insight and evaluated countless therapeutics; however, their value is increasingly being questioned due to the long history of drug failures. In this Perspective, we dispute this criticism. First, the utility of the models is limited by their design, as neither the etiology of AD nor whether interventions should occur at a cellular or network level is fully understood. Second, we highlight unmet challenges shared between animals and humans, including impeded drug transport across the blood-brain barrier, limiting effective treatment development. Third, alternative human-derived models also suffer from the limitations mentioned above and can only act as complementary resources. Finally, age being the strongest AD risk factor should be better incorporated into the experimental design, with computational modeling expected to enhance the value of animal models.
Collapse
Affiliation(s)
- Pranesh Padmanabhan
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, the University of Queensland, Brisbane, Queensland, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, the University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
5
|
Heller G, Bradbury AM, Sands MS, Bongarzone ER. Preclinical studies in Krabbe disease: A model for the investigation of novel combination therapies for lysosomal storage diseases. Mol Ther 2023; 31:7-23. [PMID: 36196048 PMCID: PMC9840155 DOI: 10.1016/j.ymthe.2022.09.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 08/16/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022] Open
Abstract
Krabbe disease (KD) is a lysosomal storage disease (LSD) caused by mutations in the galc gene. There are over 50 monogenetic LSDs, which largely impede the normal development of children and often lead to premature death. At present, there are no cures for LSDs and the available treatments are generally insufficient, short acting, and not without co-morbidities or long-term side effects. The last 30 years have seen significant advances in our understanding of LSD pathology as well as treatment options. Two gene therapy-based clinical trials, NCT04693598 and NCT04771416, for KD were recently started based on those advances. This review will discuss how our knowledge of KD got to where it is today, focusing on preclinical investigations, and how what was discovered may prove beneficial for the treatment of other LSDs.
Collapse
Affiliation(s)
- Gregory Heller
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, 808 S. Wood St M/C 512, Chicago, IL, USA.
| | - Allison M Bradbury
- Center for Gene Therapy, Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Abigail Wexner Research Institute Nationwide Children's Hospital Department of Pediatrics, The Ohio State University, Wexner Medical Center, Columbus, OH 43205, USA.
| | - Mark S Sands
- Department of Medicine, Washington University School of Medicine, 660 South Euclid Avenue Box 8007, St. Louis, MO, USA; Department of Genetics, Washington University School of Medicine, 660 South Euclid Avenue Box 8007, St. Louis, MO, USA.
| | - Ernesto R Bongarzone
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, 808 S. Wood St M/C 512, Chicago, IL, USA.
| |
Collapse
|
6
|
Balbi M, Blackmore DG, Padmanabhan P, Götz J. Ultrasound-Mediated Bioeffects in Senescent Mice and Alzheimer's Mouse Models. Brain Sci 2022; 12:775. [PMID: 35741660 PMCID: PMC9221310 DOI: 10.3390/brainsci12060775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 02/01/2023] Open
Abstract
Ultrasound is routinely used for a wide range of diagnostic imaging applications. However, given that ultrasound can operate over a wide range of parameters that can all be modulated, its applicability extends far beyond the bioimaging field. In fact, the modality has emerged as a hybrid technology that effectively assists drug delivery by transiently opening the blood-brain barrier (BBB) when combined with intravenously injected microbubbles, and facilitates neuromodulation. Studies in aged mice contributed to an insight into how low-intensity ultrasound brings about its neuromodulatory effects, including increased synaptic plasticity and improved cognitive functions, with a potential role for neurogenesis and the modulation of NMDA receptor-mediated neuronal signalling. This work is complemented by studies in mouse models of Alzheimer's disease (AD), a form of pathological ageing. Here, ultrasound was mainly employed as a BBB-opening tool that clears protein aggregates via microglial activation and neuronal autophagy, thereby restoring cognition. We discuss the currently available ultrasound approaches and how studies in senescent mice are relevant for AD and can accelerate the application of low-intensity ultrasound in the clinic.
Collapse
Affiliation(s)
- Matilde Balbi
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia; (M.B.); (D.G.B.); (P.P.)
| | - Daniel G. Blackmore
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia; (M.B.); (D.G.B.); (P.P.)
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Pranesh Padmanabhan
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia; (M.B.); (D.G.B.); (P.P.)
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jürgen Götz
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia; (M.B.); (D.G.B.); (P.P.)
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
7
|
Acosta-Rodríguez V, Rijo-Ferreira F, Izumo M, Xu P, Wight-Carter M, Green CB, Takahashi JS. Circadian alignment of early onset caloric restriction promotes longevity in male C57BL/6J mice. Science 2022; 376:1192-1202. [PMID: 35511946 PMCID: PMC9262309 DOI: 10.1126/science.abk0297] [Citation(s) in RCA: 169] [Impact Index Per Article: 84.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Caloric restriction (CR) prolongs lifespan, yet the mechanisms by which it does so remain poorly understood. Under CR, mice self-impose chronic cycles of 2-hour-feeding and 22-hour-fasting, raising the question whether calories, fasting, or time of day are causal. We show that 30%-CR is sufficient to extend lifespan 10%; however, a daily fasting interval and circadian-alignment of feeding act together to extend lifespan 35% in male C57BL/6J mice. These effects are independent of body weight. Aging induces widespread increases in gene expression associated with inflammation and decreases in expression of genes encoding components of metabolic pathways in liver from ad lib fed mice. CR at night ameliorates these aging-related changes. Thus, circadian interventions promote longevity and provide a perspective to further explore mechanisms of aging. Timed caloric restriction at night enhances longevity.
Collapse
Affiliation(s)
- Victoria Acosta-Rodríguez
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Filipa Rijo-Ferreira
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mariko Izumo
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Pin Xu
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mary Wight-Carter
- Animal Resources Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Carla B Green
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Joseph S Takahashi
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
8
|
Lejnieks DV. Treatment of Ulcerative Dermatitis in Mice (Mus musculus) with Gabapentin: 14 cases 2011-2019. J Exot Pet Med 2022. [DOI: 10.1053/j.jepm.2022.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
9
|
Zhang B, Trapp A, Kerepesi C, Gladyshev VN. Emerging rejuvenation strategies-Reducing the biological age. Aging Cell 2022; 21:e13538. [PMID: 34972247 PMCID: PMC8761015 DOI: 10.1111/acel.13538] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 11/02/2021] [Accepted: 11/29/2021] [Indexed: 12/11/2022] Open
Abstract
Several interventions have recently emerged that were proposed to reverse rather than just attenuate aging, but the criteria for what it takes to achieve rejuvenation remain controversial. Distinguishing potential rejuvenation therapies from other longevity interventions, such as those that slow down aging, is challenging, and these anti-aging strategies are often referred to interchangeably. We suggest that the prerequisite for a rejuvenation intervention is a robust, sustained, and systemic reduction in biological age, which can be assessed by biomarkers of aging, such as epigenetic clocks. We discuss known and putative rejuvenation interventions and comparatively analyze them to explore underlying mechanisms.
Collapse
Affiliation(s)
- Bohan Zhang
- Division of GeneticsDepartment of MedicineHarvard Medical SchoolBrigham and Women’s HospitalBostonMassachusettsUSA
| | - Alexandre Trapp
- Division of GeneticsDepartment of MedicineHarvard Medical SchoolBrigham and Women’s HospitalBostonMassachusettsUSA
| | - Csaba Kerepesi
- Division of GeneticsDepartment of MedicineHarvard Medical SchoolBrigham and Women’s HospitalBostonMassachusettsUSA
| | - Vadim N. Gladyshev
- Division of GeneticsDepartment of MedicineHarvard Medical SchoolBrigham and Women’s HospitalBostonMassachusettsUSA
| |
Collapse
|
10
|
Metabolic and physical function are improved with lifelong 15% calorie restriction in aging male mice. Biogerontology 2022; 23:741-755. [PMID: 36315375 PMCID: PMC9722841 DOI: 10.1007/s10522-022-09996-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 10/12/2022] [Indexed: 12/14/2022]
Abstract
Chronic calorie restriction (CR) results in lengthened lifespan and reduced disease risk. Many previous studies have implemented 30-40% calorie restriction to investigate these benefits. The goal of our study was to investigate the effects of calorie restriction, beginning at 4 months of age, on metabolic and physical changes induced by aging. Male C57BL/6NCrl calorie restricted and ad libitum fed control mice were obtained from the National Institute on Aging (NIA) and studied at 10, 18, 26, and 28 months of age to better understand the metabolic changes that occur in response to CR in middle age and advanced age. Food intake was measured in ad libitum fed controls to assess the true degree of CR (15%) in these mice. We found that 15% CR decreased body mass and liver triglyceride content, improved oral glucose clearance, and increased all limb grip strength in 10- and 18-month-old mice. Glucose clearance in ad libitum fed 26- and 28-month-old mice is enhanced relative to younger mice but was not further improved by CR. CR decreased basal insulin concentrations in all age groups and improved insulin sensitivity and rotarod time to fall in 28-month-old mice. The results of our study demonstrate that even a modest reduction (15%) in caloric intake may improve metabolic and physical health. Thus, moderate calorie restriction may be a dietary intervention to promote healthy aging with improved likelihood for adherence in human populations.
Collapse
|
11
|
Rutten MG, Derks TG, Huijkman NC, Bos T, Kloosterhuis NJ, van de Kolk KC, Wolters JC, Koster MH, Bongiovanni L, Thomas RE, de Bruin A, van de Sluis B, Oosterveer MH. Modeling Phenotypic Heterogeneity of Glycogen Storage Disease Type 1a Liver Disease in Mice by Somatic CRISPR/CRISPR-associated protein 9-Mediated Gene Editing. Hepatology 2021; 74:2491-2507. [PMID: 34157136 PMCID: PMC8597008 DOI: 10.1002/hep.32022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 05/25/2021] [Accepted: 06/09/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS Patients with glycogen storage disease type 1a (GSD-1a) primarily present with life-threatening hypoglycemia and display severe liver disease characterized by hepatomegaly. Despite strict dietary management, long-term complications still occur, such as liver tumor development. Variations in residual glucose-6-phosphatase (G6PC1) activity likely contribute to phenotypic heterogeneity in biochemical symptoms and complications between patients. However, lack of insight into the relationship between G6PC1 activity and symptoms/complications and poor understanding of the underlying disease mechanisms pose major challenges to provide optimal health care and quality of life for GSD-1a patients. Currently available GSD-1a animal models are not suitable to systematically investigate the relationship between hepatic G6PC activity and phenotypic heterogeneity or the contribution of gene-gene interactions (GGIs) in the liver. APPROACH AND RESULTS To meet these needs, we generated and characterized a hepatocyte-specific GSD-1a mouse model using somatic CRISPR/CRISPR-associated protein 9 (Cas9)-mediated gene editing. Hepatic G6pc editing reduced hepatic G6PC activity up to 98% and resulted in failure to thrive, fasting hypoglycemia, hypertriglyceridemia, hepatomegaly, hepatic steatosis (HS), and increased liver tumor incidence. This approach was furthermore successful in simultaneously modulating hepatic G6PC and carbohydrate response element-binding protein, a transcription factor that is activated in GSD-1a and protects against HS under these conditions. Importantly, it also allowed for the modeling of a spectrum of GSD-1a phenotypes in terms of hepatic G6PC activity, fasting hypoglycemia, hypertriglyceridemia, hepatomegaly and HS. CONCLUSIONS In conclusion, we show that somatic CRISPR/Cas9-mediated gene editing allows for the modeling of a spectrum of hepatocyte-borne GSD-1a disease symptoms in mice and to efficiently study GGIs in the liver. This approach opens perspectives for translational research and will likely contribute to personalized treatments for GSD-1a and other genetic liver diseases.
Collapse
Affiliation(s)
- Martijn G.S. Rutten
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Terry G.J. Derks
- Section of Metabolic DiseasesBeatrix Children’s HospitalUniversity Medical Center GroningenGroningenThe Netherlands
| | - Nicolette C.A. Huijkman
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Trijnie Bos
- Department of Laboratory MedicineUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Niels J. Kloosterhuis
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Kees C.W.A. van de Kolk
- Central Animal FacilityGroningen Small Animal Imaging Facility (Gronsai)University Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Justina C. Wolters
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Mirjam H. Koster
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Laura Bongiovanni
- Dutch Molecular Pathology CenterFaculty of Veterinary MedicineUtrecht UniversityCL UtrechtThe Netherlands
| | - Rachel E. Thomas
- Dutch Molecular Pathology CenterFaculty of Veterinary MedicineUtrecht UniversityCL UtrechtThe Netherlands
| | - Alain de Bruin
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands,Dutch Molecular Pathology CenterFaculty of Veterinary MedicineUtrecht UniversityCL UtrechtThe Netherlands
| | - Bart van de Sluis
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Maaike H. Oosterveer
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| |
Collapse
|
12
|
Ono R, Abe M, Koike N, Inokawa H, Tsuchiya Y, Umemura Y, Sasawaki Y, Yamamoto T, Wakisaka S, Kanamura N, Yagita K. Quantitative morphometric analysis of molar teeth and alveolar bone using micro-computed tomography in aged mice. J Oral Biosci 2021; 63:265-270. [PMID: 34358700 DOI: 10.1016/j.job.2021.07.006] [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: 06/29/2021] [Revised: 07/22/2021] [Accepted: 07/27/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVES Irreversible morphological regressions of the teeth or related structures in older people can diminish their overall health. However, research on human aging in dentistry is complicated by several confounding factors. In this study, we conducted a morphometric analysis of the mandibular second molars and surrounding alveolar bone in C57BL/6 mice to evaluate age-related changes in the oral cavity. METHODS The animals were divided into five groups based on their age: 4 weeks (juvenile mice; n=5); 20 weeks (n=7), 50 weeks (n=5), 77 weeks (n=7), and 100 weeks (n=5); changes were evaluated using micro-computed tomography. RESULTS The molars of juvenile mice had sharp and pointed cusps and presented maximum heights. With age and occlusal wear, the cusp heights demonstrated a significant decrease (up to 75%) until the last stage of life. Conversely, apparent lesions were not observed on the basal portion of the crown, even in the most heavily worn teeth. The roots of the molars continued to grow in length at 4 weeks of age. Alveolar bone resorption begins to occur in middle age and continues throughout life. The proportion of vertical bone loss reached approximately 40% of the entire root length, demonstrating a remarkable increase between weeks 77 and 100. CONCLUSIONS Overall, these morphological changes were similar to those observed in humans. Therefore, it might be appropriate to use aged mice as an experimental model for basic and clinical research in geriatric dentistry.
Collapse
Affiliation(s)
- Ryutaro Ono
- Department of Dental Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan; Department of Physiology and Systems Bioscience, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Makoto Abe
- Department of Oral Anatomy and Developmental Biology, Graduate School of Dentistry, Osaka University, 1-8 Yamada-Oka, Suita, Osaka, 565-0871, Japan
| | - Nobuya Koike
- Department of Physiology and Systems Bioscience, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Hitoshi Inokawa
- Department of Physiology and Systems Bioscience, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan; Department of Human Nutrition, Faculty of Contemporary Human Life Science, Chugoku Gakuen University, 83 Niwase, Kita-ku, Okayama, 701-0197, Japan
| | - Yoshiki Tsuchiya
- Department of Physiology and Systems Bioscience, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Yasuhiro Umemura
- Department of Physiology and Systems Bioscience, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Yuh Sasawaki
- Department of Physiology and Systems Bioscience, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Toshiro Yamamoto
- Department of Dental Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Satoshi Wakisaka
- Department of Oral Anatomy and Developmental Biology, Graduate School of Dentistry, Osaka University, 1-8 Yamada-Oka, Suita, Osaka, 565-0871, Japan
| | - Narisato Kanamura
- Department of Dental Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Kazuhiro Yagita
- Department of Physiology and Systems Bioscience, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| |
Collapse
|
13
|
Therapeutic Ultrasound as a Treatment Modality for Physiological and Pathological Ageing Including Alzheimer's Disease. Pharmaceutics 2021; 13:pharmaceutics13071002. [PMID: 34371696 PMCID: PMC8309087 DOI: 10.3390/pharmaceutics13071002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/29/2021] [Accepted: 06/29/2021] [Indexed: 01/09/2023] Open
Abstract
Physiological and pathological ageing (as exemplified by Alzheimer's disease, AD) are characterized by a progressive decline that also includes cognition. How this decline can be slowed or even reversed is a critical question. Here, we discuss therapeutic ultrasound as a novel modality to achieve this goal. In our studies, we explored three fundamental strategies, (i) scanning ultrasound on its own (SUSonly), (ii) therapeutic ultrasound in concert with intravenously injected microbubbles (which transiently opens the blood-brain barrier, SUS+MB), and (iii) SUS+MB in combination with therapeutic antibodies (SUS+MB+mAb). These studies show SUS+MB effectively clears amyloid and restores memory in amyloid-depositing mice and partially clears Tau and ameliorates memory impairments in Tau transgenic mice, with additional improvements found in combination trials (SUS+MB+mAb). Interestingly, both SUSonly and SUS+MB restored the induction of long-term potentiation (LTP, electrophysiological correlate of memory) in senescent wild-type mice. Both lead to increased neurogenesis, and SUSonly, in particular, resulted in improved spatial memory. We discuss these findings side-by-side with our findings obtained in AD mouse models. We conclude that therapeutic ultrasound is a non-invasive, pleiotropic modality that may present a treatment option not only for AD but also for enhancing cognition in physiological ageing.
Collapse
|
14
|
Smith BRC, Nyström A, Nowell CJ, Hausser I, Gretzmeier C, Robertson SJ, Varigos GA, Has C, Kern JS, Pang KC. Mouse models for dominant dystrophic epidermolysis bullosa carrying common human point mutations recapitulate the human disease. Dis Model Mech 2021; 14:dmm048082. [PMID: 34085701 PMCID: PMC8214732 DOI: 10.1242/dmm.048082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 04/27/2021] [Indexed: 12/13/2022] Open
Abstract
Heterozygous missense mutations in the human COL7A1 gene - coding for collagen VII - lead to the rare, dominantly inherited skin disorder dominant dystrophic epidermolysis bullosa (DDEB), which is characterised by skin fragility, blistering, scarring and nail dystrophy. To better understand the pathophysiology of DDEB and develop more effective treatments, suitable mouse models for DDEB are required but to date none have existed. We identified the two most common COL7A1 mutations in DDEB patients (p.G2034R and p.G2043R) and used CRISPR-Cas9 to introduce the corresponding mutations into mouse Col7a1 (p.G2028R and p.G2037R). Dominant inheritance of either of these two alleles results in a phenotype that closely resembles that seen in DDEB patients. Specifically, mice carrying these alleles show recurrent blistering that is first observed transiently around the mouth and paws in the early neonatal period and then again around the digits from 5-10 weeks of age. Histologically, the mice show micro-blistering and reduced collagen VII immunostaining. Biochemically, collagen VII from these mice displays reduced thermal stability, which we also observed to be the case for DDEB patients carrying the analogous mutations. Unlike previous rodent models of epidermolysis bullosa, which frequently show early lethality and severe disease, these mouse models, which to our knowledge are the first for DDEB, show no reduction in growth and survival, and - together with a relatively mild phenotype - represent a practically and ethically tractable tool for better understanding and treating epidermolysis bullosa. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Blake R. C. Smith
- Murdoch Children's Research Institute, Parkville, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Alexander Nyström
- Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany
| | - Cameron J. Nowell
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Ingrid Hausser
- Institute of Pathology, University Hospital Heidelberg, Heidelberg 69120, Germany
| | - Christine Gretzmeier
- Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany
| | - Susan J. Robertson
- Murdoch Children's Research Institute, Parkville, VIC 3052, Australia
- Dermatology Department, Faculty of Medicine, Dentistry and Health Sciences, The Royal Melbourne Hospital, University of Melbourne, Parkville, VIC 3050, Australia
- Royal Children's Hospital, Parkville, VIC 3052, Australia
| | - George A. Varigos
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Dermatology Department, Faculty of Medicine, Dentistry and Health Sciences, The Royal Melbourne Hospital, University of Melbourne, Parkville, VIC 3050, Australia
| | - Cristina Has
- Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany
| | - Johannes S. Kern
- Dermatology Department, Faculty of Medicine, Dentistry and Health Sciences, The Royal Melbourne Hospital, University of Melbourne, Parkville, VIC 3050, Australia
| | - Ken C. Pang
- Murdoch Children's Research Institute, Parkville, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC 3052, Australia
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Royal Children's Hospital, Parkville, VIC 3052, Australia
| |
Collapse
|
15
|
Blackmore DG, Turpin F, Palliyaguru T, Evans HT, Chicoteau A, Lee W, Pelekanos M, Nguyen N, Song J, Sullivan RKP, Sah P, Bartlett PF, Götz J. Low-intensity ultrasound restores long-term potentiation and memory in senescent mice through pleiotropic mechanisms including NMDAR signaling. Mol Psychiatry 2021; 26:6975-6991. [PMID: 34040151 PMCID: PMC8760044 DOI: 10.1038/s41380-021-01129-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 03/31/2021] [Accepted: 04/14/2021] [Indexed: 12/20/2022]
Abstract
Advanced physiological aging is associated with impaired cognitive performance and the inability to induce long-term potentiation (LTP), an electrophysiological correlate of memory. Here, we demonstrate in the physiologically aged, senescent mouse brain that scanning ultrasound combined with microbubbles (SUS+MB), by transiently opening the blood-brain barrier, fully restores LTP induction in the dentate gyrus of the hippocampus. Intriguingly, SUS treatment without microbubbles (SUSonly), i.e., without the uptake of blood-borne factors, proved even more effective, not only restoring LTP, but also ameliorating the spatial learning deficits of the aged mice. This functional improvement is accompanied by an altered milieu of the aged hippocampus, including a lower density of perineuronal nets, increased neurogenesis, and synaptic signaling, which collectively results in improved spatial learning. We therefore conclude that therapeutic ultrasound is a non-invasive, pleiotropic modality that may enhance cognition in elderly humans.
Collapse
Affiliation(s)
- Daniel G. Blackmore
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Fabrice Turpin
- grid.1003.20000 0000 9320 7537Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Tishila Palliyaguru
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Harrison T. Evans
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Antony Chicoteau
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Wendy Lee
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Matthew Pelekanos
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Nghia Nguyen
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Jae Song
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Robert K. P. Sullivan
- grid.1003.20000 0000 9320 7537Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia
| | - Pankaj Sah
- grid.1003.20000 0000 9320 7537Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia ,grid.263817.90000 0004 1773 1790Joint Center for Neuroscience and Neural Engineering, and Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong Province P. R. China
| | - Perry F. Bartlett
- grid.1003.20000 0000 9320 7537Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia ,grid.1003.20000 0000 9320 7537Queensland Brain Institute, The University of Queensland, Brisbane, QLD Australia ,grid.263817.90000 0004 1773 1790Joint Center for Neuroscience and Neural Engineering, and Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong Province P. R. China
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
16
|
Coninx E, Chew YC, Yang X, Guo W, Coolkens A, Baatout S, Moons L, Verslegers M, Quintens R. Hippocampal and cortical tissue-specific epigenetic clocks indicate an increased epigenetic age in a mouse model for Alzheimer's disease. Aging (Albany NY) 2020; 12:20817-20834. [PMID: 33082299 PMCID: PMC7655172 DOI: 10.18632/aging.104056] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/27/2020] [Indexed: 04/17/2023]
Abstract
Epigenetic clocks are based on age-associated changes in DNA methylation of CpG-sites, which can accurately measure chronological age in different species. Recently, several studies have indicated that the difference between chronological and epigenetic age, defined as the age acceleration, could reflect biological age indicating functional decline and age-associated diseases. In humans, an epigenetic clock associated Alzheimer's disease (AD) pathology with an acceleration of the epigenetic age. In this study, we developed and validated two mouse brain region-specific epigenetic clocks from the C57BL/6J hippocampus and cerebral cortex. Both clocks, which could successfully estimate chronological age, were further validated in a widely used mouse model for AD, the triple transgenic AD (3xTg-AD) mouse. We observed an epigenetic age acceleration indicating an increased biological age for the 3xTg-AD mice compared to non-pathological C57BL/6J mice, which was more pronounced in the cortex as compared to the hippocampus. Genomic region enrichment analysis revealed that age-dependent CpGs were enriched in genes related to developmental, aging-related, neuronal and neurodegenerative functions. Due to the limited access of human brain tissues, these epigenetic clocks specific for mouse cortex and hippocampus might be important in further unravelling the role of epigenetic mechanisms underlying AD pathology or brain aging in general.
Collapse
Affiliation(s)
- Emma Coninx
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol 2400, Belgium
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven 3000, Belgium
| | - Yap Ching Chew
- Epigenetics Technologies, Zymo Research Corporation, Irvine, CA 92614, USA
| | - Xiaojing Yang
- Epigenetics Technologies, Zymo Research Corporation, Irvine, CA 92614, USA
| | - Wei Guo
- Epigenetics Technologies, Zymo Research Corporation, Irvine, CA 92614, USA
| | - Amelie Coolkens
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol 2400, Belgium
| | - Sarah Baatout
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol 2400, Belgium
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven 3000, Belgium
| | - Mieke Verslegers
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol 2400, Belgium
| | - Roel Quintens
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol 2400, Belgium
| |
Collapse
|
17
|
De Biase D, Esposito F, De Martino M, Pirozzi C, Luciano A, Palma G, Raso GM, Iovane V, Marzocco S, Fusco A, Paciello O. Characterization of inflammatory infiltrate of ulcerative dermatitis in C57BL/6NCrl-Tg(HMGA1P6)1Pg mice. Lab Anim 2018; 53:447-458. [PMID: 30522404 DOI: 10.1177/0023677218815718] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Ulcerative dermatitis (UD) is an idiopathic, spontaneous and progressive disease typically affecting C57BL/6 aged mice with an unknown aetiopathogenesis. For this study, we evaluated 25 cases of UD in C57BL/6NCrl-Tg(HMGA1P6)1Pg mice. Formalin-fixed, paraffin-embedded skin samples were submitted to morphological investigations. Immunohistochemical analysis was performed to characterize and quantify inflammatory cells using CD3, CD45/B220, CD4, CD8 and IL-17 antibodies. Mast cell-bound IgE was investigated by immunofluorescence, whereas serum and cryopreserved skin samples were collected for molecular analysis. Student's t-test (two-tailed) was performed to assess significant differences between the two groups. Affected skin showed extensive areas of ulceration and diffuse, severe and mixed inflammatory infiltrates. No relevant changes were observed in control mice. Immunohistochemical analysis showed a predominant CD3 + CD4 + leukocyte population with fewer CD45/B220 and IL-17 immunolabelled cells and mast cell-bound IgE. Increases in TNFα, IL-1β and Il-6 mRNA expression were observed in the skin of affected animals compared to controls. Serum TNFα and IL-6 did not vary between affected and control mice. Inflammatory infiltrates and cytokine expression were consistent with both Th2/IgE and Th17 differentiation, a typical pattern of a type I hypersensitivity reaction. Overall, our data suggest an allergic-based aetiopathogenesis of UD in C57BL/6NCrl-Tg(HMGA1P6)1Pg mice.
Collapse
Affiliation(s)
- Davide De Biase
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Italy
| | - Francesco Esposito
- CNR - Institute of Experimental Endocrinology and Oncology, c/o Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Italy
| | - Marco De Martino
- CNR - Institute of Experimental Endocrinology and Oncology, c/o Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Italy
| | - Claudio Pirozzi
- Department of Pharmacy, University of Naples Federico II, Italy
| | - Antonio Luciano
- S.S.D. Sperimentazione Animale, Istituto Nazionale Tumori-IRCCS-"Fondazione G. Pascale", Naples, Italy
| | - Giuseppe Palma
- S.S.D. Sperimentazione Animale, Istituto Nazionale Tumori-IRCCS-"Fondazione G. Pascale", Naples, Italy
| | | | | | | | - Alfredo Fusco
- CNR - Institute of Experimental Endocrinology and Oncology, c/o Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Italy
| | - Orlando Paciello
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Italy
| |
Collapse
|
18
|
Inhibition of glycosphingolipid synthesis reverses skin inflammation and hair loss in ApoE-/- mice fed western diet. Sci Rep 2018; 8:11463. [PMID: 30061606 PMCID: PMC6065400 DOI: 10.1038/s41598-018-28663-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 06/18/2018] [Indexed: 01/19/2023] Open
Abstract
Sphingolipids have been accorded numerous biological functions however, the effects of feeding a western diet (diet rich in cholesterol and fat) on skin phenotypes, and color is not known. Here, we observed that chronic high-fat and high-cholesterol diet intake in a mouse model of atherosclerosis (ApoE-/-) decreases the level of ceramides and glucosylceramide. At the expense of increased levels of lactosylceramide due to an increase in the expression of lactosylceramide synthase (GalT-V). This is accompanied with neutrophil infiltration into dermis, and enrichment of tumor necrosis factor-stimulated gene-6 (TSG-6) protein. This causes skin inflammation, hair discoloration and loss, in ApoE-/- mice. Conversely, inhibition of glycosphingolipid synthesis, by D-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol (D-PDMP), unbound or encapsulated in a biodegradable polymer (BPD) reversed these phenotypes. Thus, inhibition of glycosphingolipid synthesis represents a unique therapeutic approach relevant to human skin and hair Biology.
Collapse
|
19
|
The effects of graded levels of calorie restriction: XI. Evaluation of the main hypotheses underpinning the life extension effects of CR using the hepatic transcriptome. Aging (Albany NY) 2018; 9:1770-1824. [PMID: 28768896 PMCID: PMC5559174 DOI: 10.18632/aging.101269] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 07/27/2017] [Indexed: 12/15/2022]
Abstract
Calorie restriction (CR) may extend longevity by modulating the mechanisms involved in aging. Different hypotheses have been proposed for its main mode of action. We quantified hepatic transcripts of male C57BL/6 mice exposed to graded levels of CR (0% to 40% CR) for three months, and evaluated the responses relative to these various hypotheses. Of the four main signaling pathways implied to be linked to the impact of CR on lifespan (insulin/insulin like growth factor 1 (IGF-1), nuclear factor-kappa beta (NF-ĸB), mechanistic target of rapamycin (mTOR) and sirtuins (SIRTs)), all the pathways except SIRT were altered in a manner consistent with increased lifespan. However, the expression levels of SIRT4 and SIRT7 were decreased with increasing levels of CR. Changes consistent with altered fuel utilization under CR may reduce reactive oxygen species production, which was paralleled by reduced protection. Downregulated major urinary protein (MUP) transcription suggested reduced reproductive investment. Graded CR had a positive effect on autophagy and xenobiotic metabolism, and was protective with respect to cancer signaling. CR had no significant effect on fibroblast growth factor-21 (FGF21) transcription but affected transcription in the hydrogen sulfide production pathway. Responses to CR were consistent with several different hypotheses, and the benefits of CR on lifespan likely reflect the combined impact on multiple aging related processes.
Collapse
|
20
|
Reinhard JR, Lin S, McKee KK, Meinen S, Crosson SC, Sury M, Hobbs S, Maier G, Yurchenco PD, Rüegg MA. Linker proteins restore basement membrane and correct LAMA2-related muscular dystrophy in mice. Sci Transl Med 2018; 9:9/396/eaal4649. [PMID: 28659438 DOI: 10.1126/scitranslmed.aal4649] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 03/03/2017] [Accepted: 05/04/2017] [Indexed: 12/12/2022]
Abstract
LAMA2-related muscular dystrophy (LAMA2 MD or MDC1A) is the most frequent form of early-onset, fatal congenital muscular dystrophies. It is caused by mutations in LAMA2, the gene encoding laminin-α2, the long arm of the heterotrimeric (α2, β1, and γ1) basement membrane protein laminin-211 (Lm-211). We establish that despite compensatory expression of laminin-α4, giving rise to Lm-411 (α4, β1, and γ1), muscle basement membrane is labile in LAMA2 MD biopsies. Consistent with this deficit, recombinant Lm-411 polymerized and bound to cultured myotubes only weakly. Polymerization and cell binding of Lm-411 were enhanced by addition of two specifically designed linker proteins. One, called αLNNd, consists of the N-terminal part of laminin-α1 and the laminin-binding site of nidogen-1. The second, called mini-agrin (mag), contains binding sites for laminins and α-dystroglycan. Transgenic expression of mag and αLNNd in a mouse model for LAMA2 MD fully restored basement membrane stability, recovered muscle force and size, increased overall body weight, and extended life span more than five times to a maximum survival beyond 2 years. These findings provide a mechanistic understanding of LAMA2 MD and establish a strong basis for a potential treatment.
Collapse
Affiliation(s)
| | - Shuo Lin
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Karen K McKee
- Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Sarina Meinen
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Stephanie C Crosson
- Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Maurizio Sury
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Samantha Hobbs
- Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | | | - Peter D Yurchenco
- Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Markus A Rüegg
- Biozentrum, University of Basel, 4056 Basel, Switzerland.
| |
Collapse
|
21
|
Mitchell SE, Delville C, Konstantopedos P, Derous D, Green CL, Wang Y, Han JDJ, Promislow DEL, Douglas A, Chen L, Lusseau D, Speakman JR. The effects of graded levels of calorie restriction: V. Impact of short term calorie and protein restriction on physical activity in the C57BL/6 mouse. Oncotarget 2017; 7:19147-70. [PMID: 27007156 PMCID: PMC4991372 DOI: 10.18632/oncotarget.8158] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 02/28/2016] [Indexed: 12/15/2022] Open
Abstract
Calorie restriction (CR) delays the onset of age-related disease and extends lifespan in a number of species. When faced with reduced energy supply animals need to lower energy demands, which may be achieved in part by reducing physical activity (PA). We monitored changes in PA using implanted transmitters in male C57BL/6 mice in response to graded levels of CR (10 to 40%) or matched levels of graded protein restriction (PR) for 3 months. Mice were fed at lights out and ad libitum controls were limited to dark-phase feeding (12AL) or 24hr/day. Total daily PA declined in a non-linear manner over the first 30 days of CR or PR, remaining stable thereafter. Total daily PA was not related to the level of CR or PR. Total daily PA over the last 20 days of restriction was related to circulating leptin, insulin, tumor necrosis factor-α (TNF-α) and insulin-like growth factor (IGF)-1 levels, measured after 3 months. Mice under restriction showed a high level of activity in the 2hrs before feeding (food anticipatory activity: FAA). FAA followed a complex pattern, peaking around day 20, falling on ∼day 37 then increasing again. FAA was also positively related to the level of restriction and inversely to leptin, insulin, TNF-α and IGF-1. Non-FAA, in contrast, declined over the period of restriction, generally more so in mice under greater restriction, thereby offsetting to some extent the increase in FAA. Mice under PR displayed no changes in PA over time or in comparison to 12AL, and showed no increase in FAA.
Collapse
Affiliation(s)
- Sharon E Mitchell
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Camille Delville
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Penelope Konstantopedos
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Davina Derous
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Cara L Green
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Yingchun Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Chaoyang, Beijing, China
| | - Jing-Dong J Han
- Key Laboratory of Computational Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Daniel E L Promislow
- Department of Pathology and Department of Biology, University of Washington, Seattle, Washington, USA
| | - Alex Douglas
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Luonan Chen
- Key Laboratory of Systems Biology, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - David Lusseau
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - John R Speakman
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK.,State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Chaoyang, Beijing, China
| |
Collapse
|
22
|
Brandt T, Mourier A, Tain LS, Partridge L, Larsson NG, Kühlbrandt W. Changes of mitochondrial ultrastructure and function during ageing in mice and Drosophila. eLife 2017; 6. [PMID: 28699890 PMCID: PMC5580880 DOI: 10.7554/elife.24662] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 06/19/2017] [Indexed: 02/04/2023] Open
Abstract
Ageing is a progressive decline of intrinsic physiological functions. We examined the impact of ageing on the ultrastructure and function of mitochondria in mouse and fruit flies (Drosophila melanogaster) by electron cryo-tomography and respirometry. We discovered distinct age-related changes in both model organisms. Mitochondrial function and ultrastructure are maintained in mouse heart, whereas subpopulations of mitochondria from mouse liver show age-related changes in membrane morphology. Subpopulations of mitochondria from young and old mouse kidney resemble those described for apoptosis. In aged flies, respiratory activity is compromised and the production of peroxide radicals is increased. In about 50% of mitochondria from old flies, the inner membrane organization breaks down. This establishes a clear link between inner membrane architecture and functional decline. Mitochondria were affected by ageing to very different extents, depending on the organism and possibly on the degree to which tissues within the same organism are protected against mitochondrial damage.
Collapse
Affiliation(s)
- Tobias Brandt
- Department of Structural Biology, Max-Planck-Institute of Biophysics, Frankfurt am Main, Germany
| | - Arnaud Mourier
- Department of Mitochondrial Biology, Max Planck Institute for Biology of Ageing, Cologne, Germany.,Institut de Biochimie et Génétique Cellulaires UMR 5095, Université de Bordeaux, Bordeaux, France.,CNRS, Institut de Biochimie et Génétique Cellulaires UMR 5095, Bordeaux, France
| | - Luke S Tain
- Department of Biological Mechanisms of Ageing, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Linda Partridge
- Department of Biological Mechanisms of Ageing, Max Planck Institute for Biology of Ageing, Cologne, Germany.,Institute of Healthy Ageing, Department of Genetics, Evolution, and Environment, University College London, London, United Kingdom
| | - Nils-Göran Larsson
- Department of Mitochondrial Biology, Max Planck Institute for Biology of Ageing, Cologne, Germany.,Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Werner Kühlbrandt
- Department of Structural Biology, Max-Planck-Institute of Biophysics, Frankfurt am Main, Germany
| |
Collapse
|
23
|
Lenart P, Bienertová-Vašků J. Keeping up with the Red Queen: the pace of aging as an adaptation. Biogerontology 2016; 18:693-709. [PMID: 28013399 DOI: 10.1007/s10522-016-9674-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 12/19/2016] [Indexed: 12/25/2022]
Abstract
For decades, a vast majority of biogerontologists assumed that aging is not and cannot be an adaptation. In recent years, however, several authors opposed this predominant view and repeatedly suggested that not only is aging an adaptation but that it is the result of a specific aging program. This issue almost instantaneously became somewhat controversial and many important authors produced substantial works refuting the notion of the aging program. In this article we review the current state of the debate and list the most important arguments proposed by both sides. Furthermore, although classical interpretations of the evolution of aging are in sharp contrast with the idea of programmed aging, we suggest that the truth might in fact very well lie somewhere in between. We also propose our own interpretation which states that although aging is in essence inevitable and results from damage accumulation rather than from a specific program, the actual rate of aging in nature may still be adaptive to some extent.
Collapse
Affiliation(s)
- Peter Lenart
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Building A18, 625 00, Brno, Czech Republic.
| | - Julie Bienertová-Vašků
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Building A18, 625 00, Brno, Czech Republic.,Research Centre for Toxic Compounds in the Environment, Faculty of Science, Masaryk University, Kamenice 5, Building A29, 625 00, Brno, Czech Republic
| |
Collapse
|
24
|
von Zglinicki T, Varela-Nieto I, Brites D, Karagianni N, Ortolano S, Georgopoulos S, Cardoso AL, Novella S, Lepperdinger G, Trendelenburg AU, van Os R. Frailty in mouse ageing: A conceptual approach. Mech Ageing Dev 2016; 160:34-40. [DOI: 10.1016/j.mad.2016.07.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 07/11/2016] [Accepted: 07/15/2016] [Indexed: 01/21/2023]
|
25
|
Kane AE, Hilmer SN, Mach J, Mitchell SJ, de Cabo R, Howlett SE. Animal models of frailty: current applications in clinical research. Clin Interv Aging 2016; 11:1519-1529. [PMID: 27822024 PMCID: PMC5089820 DOI: 10.2147/cia.s105714] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The ethical, logistical, and biological complications of working with an older population of people inherently limits clinical studies of frailty. The recent development of animal models of frailty, and tools for assessing frailty in animal models provides an invaluable opportunity for frailty research. This review summarizes currently published animal models of frailty including the interleukin-10 knock-out mouse, the mouse frailty phenotype assessment tool, and the mouse clinical frailty index. It discusses both current and potential roles of these models in research into mechanisms of frailty, interventions to prevent/delay frailty, and the effect of frailty on outcomes. Finally, this review discusses some of the challenges and opportunities of translating research findings from animals to humans.
Collapse
Affiliation(s)
- Alice E Kane
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Sarah N Hilmer
- Kolling Institute of Medical Research and Sydney Medical School, University of Sydney; Department of Clinical Pharmacology; Department of Aged Care, Royal North Shore Hospital, Sydney, NSW, Australia
| | - John Mach
- Kolling Institute of Medical Research and Sydney Medical School, University of Sydney; Department of Clinical Pharmacology; Department of Aged Care, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Sarah J Mitchell
- National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Rafael de Cabo
- National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Susan E Howlett
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
26
|
Mitchell SJ, Madrigal-Matute J, Scheibye-Knudsen M, Fang E, Aon M, González-Reyes JA, Cortassa S, Kaushik S, Gonzalez-Freire M, Patel B, Wahl D, Ali A, Calvo-Rubio M, Burón MI, Guiterrez V, Ward TM, Palacios HH, Cai H, Frederick DW, Hine C, Broeskamp F, Habering L, Dawson J, Beasley TM, Wan J, Ikeno Y, Hubbard G, Becker KG, Zhang Y, Bohr VA, Longo DL, Navas P, Ferrucci L, Sinclair DA, Cohen P, Egan JM, Mitchell JR, Baur JA, Allison DB, Anson RM, Villalba JM, Madeo F, Cuervo AM, Pearson KJ, Ingram DK, Bernier M, de Cabo R. Effects of Sex, Strain, and Energy Intake on Hallmarks of Aging in Mice. Cell Metab 2016; 23:1093-1112. [PMID: 27304509 PMCID: PMC4911707 DOI: 10.1016/j.cmet.2016.05.027] [Citation(s) in RCA: 315] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 05/27/2016] [Accepted: 05/31/2016] [Indexed: 01/10/2023]
Abstract
Calorie restriction (CR) is the most robust non-genetic intervention to delay aging. However, there are a number of emerging experimental variables that alter CR responses. We investigated the role of sex, strain, and level of CR on health and survival in mice. CR did not always correlate with lifespan extension, although it consistently improved health across strains and sexes. Transcriptional and metabolomics changes driven by CR in liver indicated anaplerotic filling of the Krebs cycle together with fatty acid fueling of mitochondria. CR prevented age-associated decline in the liver proteostasis network while increasing mitochondrial number, preserving mitochondrial ultrastructure and function with age. Abrogation of mitochondrial function negated life-prolonging effects of CR in yeast and worms. Our data illustrate the complexity of CR in the context of aging, with a clear separation of outcomes related to health and survival, highlighting complexities of translation of CR into human interventions.
Collapse
Affiliation(s)
- Sarah J Mitchell
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA
| | - Julio Madrigal-Matute
- Department of Developmental and Molecular Biology, Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Morten Scheibye-Knudsen
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA; Laboratory of Molecular Gerontology, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA; Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Evandro Fang
- Laboratory of Molecular Gerontology, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA
| | - Miguel Aon
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA
| | - José A González-Reyes
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Agrifood Campus of International Excellence, ceiA3, 14071 Córdoba, Spain
| | - Sonia Cortassa
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA
| | - Susmita Kaushik
- Department of Developmental and Molecular Biology, Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Marta Gonzalez-Freire
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA
| | - Bindi Patel
- Department of Developmental and Molecular Biology, Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Devin Wahl
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA
| | - Ahmed Ali
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA
| | - Miguel Calvo-Rubio
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Agrifood Campus of International Excellence, ceiA3, 14071 Córdoba, Spain
| | - María I Burón
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Agrifood Campus of International Excellence, ceiA3, 14071 Córdoba, Spain
| | - Vincent Guiterrez
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA
| | - Theresa M Ward
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA
| | - Hector H Palacios
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA
| | - Huan Cai
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA
| | - David W Frederick
- Department of Physiology, Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christopher Hine
- Department of Genetics and Complex Diseases, Harvard University, Boston, MA 02115, USA
| | - Filomena Broeskamp
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, and BioTechMed Graz, 8010 Graz, Austria
| | - Lukas Habering
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, and BioTechMed Graz, 8010 Graz, Austria
| | - John Dawson
- Department of Biostatistics, University of Alabama, Birmingham, AL 35294, USA; GRECC, Birmingham/Atlanta Veterans Administration Hospital, Birmingham, AL 35294, USA
| | - T Mark Beasley
- Department of Biostatistics, University of Alabama, Birmingham, AL 35294, USA; GRECC, Birmingham/Atlanta Veterans Administration Hospital, Birmingham, AL 35294, USA
| | - Junxiang Wan
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Yuji Ikeno
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX 78245-3207, USA
| | - Gene Hubbard
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX 78245-3207, USA
| | - Kevin G Becker
- Laboratory of Genetics, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA
| | - Yongqing Zhang
- Laboratory of Genetics, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA
| | - Vilhelm A Bohr
- Laboratory of Molecular Gerontology, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA
| | - Dan L Longo
- Laboratory of Genetics, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA
| | - Placido Navas
- Centro Andaluz de Biologia del Desarrollo, and CIBERER, Instituto de Salud Carlos III, Universidad Pablo de Olavide-CSIC, 41013 Sevilla, Spain
| | - Luigi Ferrucci
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA
| | - David A Sinclair
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Pinchas Cohen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Josephine M Egan
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA
| | - James R Mitchell
- Department of Genetics and Complex Diseases, Harvard University, Boston, MA 02115, USA
| | - Joseph A Baur
- Department of Physiology, Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David B Allison
- Department of Biostatistics, University of Alabama, Birmingham, AL 35294, USA; GRECC, Birmingham/Atlanta Veterans Administration Hospital, Birmingham, AL 35294, USA
| | - R Michael Anson
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA
| | - José M Villalba
- Department of Cell Biology, Physiology and Immunology, University of Córdoba, Agrifood Campus of International Excellence, ceiA3, 14071 Córdoba, Spain
| | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, and BioTechMed Graz, 8010 Graz, Austria
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Kevin J Pearson
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA; Graduate Center for Nutritional Sciences, University of Kentucky, C.T. Wethington Building, Room 591, 900 South Limestone, Lexington, KY 40536, USA
| | - Donald K Ingram
- Pennington Biomedical Research Center, Baton Rouge, LA 70809, USA
| | - Michel Bernier
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, 251 Bayview Boulevard, Suite 100, Baltimore, MD 21224, USA.
| |
Collapse
|
27
|
Shirakabe A, Ikeda Y, Sciarretta S, Zablocki DK, Sadoshima J. Aging and Autophagy in the Heart. Circ Res 2016; 118:1563-76. [PMID: 27174950 PMCID: PMC4869999 DOI: 10.1161/circresaha.116.307474] [Citation(s) in RCA: 307] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 02/15/2016] [Indexed: 12/15/2022]
Abstract
The aging population is increasing in developed countries. Because the incidence of cardiac disease increases dramatically with age, it is important to understand the molecular mechanisms through which the heart becomes either more or less susceptible to stress. Cardiac aging is characterized by the presence of hypertrophy, fibrosis, and accumulation of misfolded proteins and dysfunctional mitochondria. Macroautophagy (hereafter referred to as autophagy) is a lysosome-dependent bulk degradation mechanism that is essential for intracellular protein and organelle quality control. Autophagy and autophagic flux are generally decreased in aging hearts, and murine autophagy loss-of-function models develop exacerbated cardiac dysfunction that is accompanied by the accumulation of misfolded proteins and dysfunctional organelles. On the contrary, stimulation of autophagy generally improves cardiac function in mouse models of protein aggregation by removing accumulated misfolded proteins, dysfunctional mitochondria, and damaged DNA, thereby improving the overall cellular environment and alleviating aging-associated pathology in the heart. Increasing lines of evidence suggest that autophagy is required for many mechanisms that mediate lifespan extension, such as caloric restriction, in various organisms. These results raise the exciting possibility that autophagy may play an important role in combating the adverse effects of aging in the heart. In this review, we discuss the role of autophagy in the heart during aging, how autophagy alleviates age-dependent changes in the heart, and how the level of autophagy in the aging heart can be restored.
Collapse
Affiliation(s)
- Akihiro Shirakabe
- From the Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark (A.S., Y.I., S.S., D.K.Z., J.S.); Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan (Y.I.); Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy (S.S.); and Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S.)
| | - Yoshiyuki Ikeda
- From the Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark (A.S., Y.I., S.S., D.K.Z., J.S.); Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan (Y.I.); Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy (S.S.); and Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S.)
| | - Sebastiano Sciarretta
- From the Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark (A.S., Y.I., S.S., D.K.Z., J.S.); Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan (Y.I.); Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy (S.S.); and Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S.)
| | - Daniela K Zablocki
- From the Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark (A.S., Y.I., S.S., D.K.Z., J.S.); Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan (Y.I.); Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy (S.S.); and Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S.)
| | - Junichi Sadoshima
- From the Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark (A.S., Y.I., S.S., D.K.Z., J.S.); Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan (Y.I.); Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy (S.S.); and Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S.).
| |
Collapse
|
28
|
Walls GV, Stevenson M, Soukup BS, Lines KE, Grossman AB, Schmid HA, Thakker RV. Pasireotide Therapy of Multiple Endocrine Neoplasia Type 1-Associated Neuroendocrine Tumors in Female Mice Deleted for an Men1 Allele Improves Survival and Reduces Tumor Progression. Endocrinology 2016; 157:1789-98. [PMID: 26990064 PMCID: PMC4870877 DOI: 10.1210/en.2015-1965] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Pasireotide, a somatostatin analog, is reported to have anti-proliferative effects in neuroendocrine tumors (NETs). We therefore assessed the efficacy of pasireotide for treating pancreatic and pituitary NETs that develop in a mouse model of multiple endocrine neoplasia type 1 (MEN1). Men1(+/-) mice were treated from age 12 mo with 40 mg/kg pasireotide long-acting release formulation, or PBS, intramuscularly monthly for 9 mo. The Men1(+/-) mice had magnetic resonance imaging at 12 and 21 mo, and from 20 mo oral 5-bromo-2-deoxyuridine for 1 mo, to assess tumor development and proliferation, respectively. NETs were collected at age 21 mo, and proliferation and apoptosis assessed by immunohistochemistry and TUNEL assays, respectively. Pasireotide-treated Men1(+/-) mice had increased survival (pasireotide, 80.9% vs PBS, 65.2%; P < .05), with fewer mice developing pancreatic NETs (pasireotide, 86.9% vs PBS, 96.9%; P < .05) and smaller increases in pituitary NET volumes (pre-treated vs post-treated, 0.803 ± 0.058 mm(3) vs 2.872 ± 0.728 mm(3) [pasireotide] compared with 0.844 ± 0.066 mm(3) vs 8.847 ±1.948 mm(3) [PBS]; P < .01). In addition, pasireotide-treated mice had fewer pancreatic NETs compared with PBS-treated mice (2.36 ± 0.25 vs 3.72 ± 0.32, respectively; P < .001), with decreased proliferation in pancreatic NETs (pasireotide, 0.35 ± 0.03% vs PBS, 0.78 ± 0.08%; P < .0001) and pituitary NETs (pasireotide, 0.73 ±0.07% vs PBS, 1.81 ± 0.15%; P < .0001), but increased apoptosis in pancreatic NETs (pasireotide, 0.42 ± 0.05% vs PBS, 0.19 ± 0.03%; P < .001) and pituitary NETs (pasireotide, 14.75 ± 1.58% vs PBS, 2.35 ± 0.44%; P < .001). Thus, pasireotide increased survival and inhibited pancreatic and pituitary NET growth, thereby indicating its potential as an anti-proliferative and pro-apoptotic therapy.
Collapse
Affiliation(s)
- Gerard V Walls
- Academic Endocrine Unit (G.V.W., M.S., B.S.S., K.E.L., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; Nuffield Department of Surgical Sciences (G.V.W., B.S.S.), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom; Department of Endocrinology (A.B.G.), OCDEM, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; and Novartis Pharma AG (H.A.S.), Novartis Institutes for Biomedical Research, Oncology, CH-4057 Basel, Switzerland
| | - Mark Stevenson
- Academic Endocrine Unit (G.V.W., M.S., B.S.S., K.E.L., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; Nuffield Department of Surgical Sciences (G.V.W., B.S.S.), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom; Department of Endocrinology (A.B.G.), OCDEM, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; and Novartis Pharma AG (H.A.S.), Novartis Institutes for Biomedical Research, Oncology, CH-4057 Basel, Switzerland
| | - Benjamin S Soukup
- Academic Endocrine Unit (G.V.W., M.S., B.S.S., K.E.L., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; Nuffield Department of Surgical Sciences (G.V.W., B.S.S.), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom; Department of Endocrinology (A.B.G.), OCDEM, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; and Novartis Pharma AG (H.A.S.), Novartis Institutes for Biomedical Research, Oncology, CH-4057 Basel, Switzerland
| | - Kate E Lines
- Academic Endocrine Unit (G.V.W., M.S., B.S.S., K.E.L., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; Nuffield Department of Surgical Sciences (G.V.W., B.S.S.), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom; Department of Endocrinology (A.B.G.), OCDEM, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; and Novartis Pharma AG (H.A.S.), Novartis Institutes for Biomedical Research, Oncology, CH-4057 Basel, Switzerland
| | - Ashley B Grossman
- Academic Endocrine Unit (G.V.W., M.S., B.S.S., K.E.L., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; Nuffield Department of Surgical Sciences (G.V.W., B.S.S.), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom; Department of Endocrinology (A.B.G.), OCDEM, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; and Novartis Pharma AG (H.A.S.), Novartis Institutes for Biomedical Research, Oncology, CH-4057 Basel, Switzerland
| | - Herbert A Schmid
- Academic Endocrine Unit (G.V.W., M.S., B.S.S., K.E.L., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; Nuffield Department of Surgical Sciences (G.V.W., B.S.S.), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom; Department of Endocrinology (A.B.G.), OCDEM, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; and Novartis Pharma AG (H.A.S.), Novartis Institutes for Biomedical Research, Oncology, CH-4057 Basel, Switzerland
| | - Rajesh V Thakker
- Academic Endocrine Unit (G.V.W., M.S., B.S.S., K.E.L., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; Nuffield Department of Surgical Sciences (G.V.W., B.S.S.), University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom; Department of Endocrinology (A.B.G.), OCDEM, Churchill Hospital, Oxford, OX3 7LJ, United Kingdom; and Novartis Pharma AG (H.A.S.), Novartis Institutes for Biomedical Research, Oncology, CH-4057 Basel, Switzerland
| |
Collapse
|
29
|
A "Pedi" Cures All: Toenail Trimming and the Treatment of Ulcerative Dermatitis in Mice. PLoS One 2016; 11:e0144871. [PMID: 26735497 PMCID: PMC4703297 DOI: 10.1371/journal.pone.0144871] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 11/24/2015] [Indexed: 12/31/2022] Open
Abstract
Ulcerative Dermatitis (UD) is the most common cause of unplanned euthanasia in mice used in research, with prevalence rates reported between 4 and 21%. UD is characterized by a deep, ulcerative lesion that appears most commonly over the dorsal neck and is attendant with an intense pruritus. The underlying cause of UD is currently unknown, and as a consequence, there are no directed therapies that resolve lesions reliably. However, there is a growing body of evidence that suggests a behavioral component to the onset, maintenance, and progression of UD lesions. Scratching behavior in response to the intense pruritus associated with UD lesions may be an effective target for interventional therapies. We hypothesized that interfering with scratching behavior by trimming the toenails of mice with UD, would resolve UD lesions. To test this hypothesis, we first evaluated the efficacy of toenail trims with a single application of Vetericyn at the time of treatment versus our previous standard of care, topical Tresaderm applied daily. We found that toenail trims were significantly more effective at resolving lesions (n = 39 toenail trims, n = 100 Tresaderm, p<0.0001) with 93.3% of animals healing by 14 days (median time to lesion resolution). Furthermore, dorsal neck lesions did not recur by 42 days after a single toenail trim (n = 54); however, flank lesions did not resolve and the outcome of the two lesion distributions following treatment were significantly different (p<0.0001). Finally, we implemented toenail trims at an institutional level and found similar efficacies (approximately 90%) for toenail trims regardless of one-time topical supplement used (triple antibiotic ointment, Tresaderm, and Vetericyn, n = 55, 58, 18, p = 0.63). This is the first report of a highly effective treatment for one of the most serious welfare issues in laboratory mice.
Collapse
|
30
|
Dai N, Zhao L, Wrighting D, Krämer D, Majithia A, Wang Y, Cracan V, Borges-Rivera D, Mootha VK, Nahrendorf M, Thorburn DR, Minichiello L, Altshuler D, Avruch J. IGF2BP2/IMP2-Deficient mice resist obesity through enhanced translation of Ucp1 mRNA and Other mRNAs encoding mitochondrial proteins. Cell Metab 2015; 21:609-21. [PMID: 25863250 PMCID: PMC4663978 DOI: 10.1016/j.cmet.2015.03.006] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 09/28/2014] [Accepted: 03/02/2015] [Indexed: 01/01/2023]
Abstract
Although variants in the IGF2BP2/IMP2 gene confer risk for type 2 diabetes, IMP2, an RNA binding protein, is not known to regulate metabolism. Imp2(-/-) mice gain less lean mass after weaning and have increased lifespan. Imp2(-/-) mice are highly resistant to diet-induced obesity and fatty liver and display superior glucose tolerance and insulin sensitivity, increased energy expenditure, and better defense of core temperature on cold exposure. Imp2(-/-) brown fat and Imp2(-/-) brown adipocytes differentiated in vitro contain more UCP1 polypeptide than Imp2(+/+) despite similar levels of Ucp1 mRNA; the Imp2(-/-)adipocytes also exhibit greater uncoupled oxygen consumption. IMP2 binds the mRNAs encoding Ucp1 and other mitochondrial components, and most exhibit increased translational efficiency in the absence of IMP2. In vitro IMP2 inhibits translation of mRNAs bearing the Ucp1 untranslated segments. Thus IMP2 limits longevity and regulates nutrient and energy metabolism in the mouse by controlling the translation of its client mRNAs.
Collapse
Affiliation(s)
- Ning Dai
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Diabetes Unit, Medical Services, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Liping Zhao
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Diabetes Unit, Medical Services, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Diedra Wrighting
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Dana Krämer
- European Molecular Biology Laboratory, Mouse Biology Unit, 00015-Monterotondo, Italy
| | - Amit Majithia
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Diabetes Unit, Medical Services, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Yanqun Wang
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Valentin Cracan
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Diego Borges-Rivera
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Vamsi K Mootha
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Diabetes Unit, Medical Services, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Matthias Nahrendorf
- Department of Radiology and the Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Radiology, Harvard Medical School, Boston, MA 02115, USA
| | - David R Thorburn
- Murdoch Childrens Research Institute and Victorian Clinical Genetics Services, Royal Children's Hospital, Flemington Road, Parkville, Melbourne, VIC 3052, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Liliana Minichiello
- European Molecular Biology Laboratory, Mouse Biology Unit, 00015-Monterotondo, Italy; Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - David Altshuler
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Diabetes Unit, Medical Services, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| | - Joseph Avruch
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Diabetes Unit, Medical Services, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
31
|
Walsh ME, Sloane LB, Fischer KE, Austad SN, Richardson A, Van Remmen H. Use of Nerve Conduction Velocity to Assess Peripheral Nerve Health in Aging Mice. J Gerontol A Biol Sci Med Sci 2014; 70:1312-9. [PMID: 25477428 DOI: 10.1093/gerona/glu208] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 10/01/2014] [Indexed: 12/13/2022] Open
Abstract
Nerve conduction velocity (NCV), the speed at which electrical signals propagate along peripheral nerves, is used in the clinic to evaluate nerve function in humans. A decline in peripheral nerve function is associated with a number of age-related pathologies. While several studies have shown that NCV declines with age in humans, there is little information on the effect of age on NCV in peripheral nerves in mice. In this study, we evaluated NCV in male and female C57Bl/6 mice ranging from 4 to 32 months of age. We observed a decline in NCV in both male and female mice after 20 months of age. Sex differences were detected in sensory NCV as well as the rate of decline during aging in motor nerves; female mice had slower sensory NCV and a slower age-related decline in motor nerves compared with male mice. We also tested the effect of dietary restriction on NCV in 30-month-old female mice. Dietary restriction prevented the age-related decline in sciatic NCV but not other nerves. Because NCV is clinically relevant to the assessment of nerve function, we recommend that NCV be used to evaluate healthspan in assessing genetic and pharmacological interventions that increase the life span of mice.
Collapse
Affiliation(s)
- Michael E Walsh
- Department of Cellular and Structural Biology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Lauren B Sloane
- Department of Biology, State University of New York at Delhi, Delhi, New York
| | - Kathleen E Fischer
- Department of Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Steven N Austad
- Department of Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Arlan Richardson
- Oklahoma City VA Medical Center, Oklahoma City, Oklahoma. University of Oklahoma Health Science Center, Oklahoma City, Oklahoma
| | - Holly Van Remmen
- Oklahoma City VA Medical Center, Oklahoma City, Oklahoma. Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma.
| |
Collapse
|
32
|
Haussig JM, Burgold J, Hafalla JCR, Matuschewski K, Kooij TWA. Signatures of malaria vaccine efficacy in ageing murine immune memory. Parasite Immunol 2014; 36:199-206. [PMID: 24495208 DOI: 10.1111/pim.12104] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 01/27/2014] [Indexed: 02/06/2023]
Abstract
Malaria transmission occurs by mosquito bite. Thereafter, Plasmodium sporozoites specifically invade the liver, where they develop into thousands of merozoites that initiate blood-stage infection and clinical malaria. The pre-erythrocytic phase of a Plasmodium infection is the target of experimental whole-parasite vaccines against malaria. Repeated immunizations with high doses of live, metabolically active sporozoites can induce protracted protection against Plasmodium reinfection. Parasites lacking a Plasmodium-specific apicoplast protein, termed PALM, arrest very late during intrahepatic development just prior to liver merozoite release and can elicit sterile protection with two immunization doses only. In this report, we show in the robust Plasmodium berghei-C57BL/6 model that partial protection extends beyond 1 year after the last immunization. In ageing mice, intracellular cytokine staining of Plasmodium peptide-stimulated intrahepatic CD8+ T cells revealed elevated levels of interferon gamma in vaccinated mice. We conclude that antigen-specific T cells persist in the target organ and are critical signatures of lasting protection. Our data also support the notions that memory T-cell responses generated early in life remain largely intact well into old age and that murine Plasmodium vaccination and infection models are suitable to study the mechanisms of maintenance and efficiency of adaptive immunity during immunosenescence.
Collapse
Affiliation(s)
- J M Haussig
- Max Planck Institute for Infection Biology, Parasitology Unit, Berlin, Germany
| | | | | | | | | |
Collapse
|
33
|
Ballak SB, Degens H, de Haan A, Jaspers RT. Aging related changes in determinants of muscle force generating capacity: a comparison of muscle aging in men and male rodents. Ageing Res Rev 2014; 14:43-55. [PMID: 24495393 DOI: 10.1016/j.arr.2014.01.005] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 01/20/2014] [Accepted: 01/24/2014] [Indexed: 12/25/2022]
Abstract
Human aging is associated with a progressive decline in skeletal muscle mass and force generating capacity, however the exact mechanisms underlying these changes are not fully understood. Rodents models have often been used to enhance our understanding of mechanisms of age-related changes in human skeletal muscle. However, to what extent age-related alterations in determinants of muscle force generating capacity observed in rodents resemble those in humans has not been considered thoroughly. This review compares the effect of aging on muscle force generating determinants (muscle mass, fiber size, fiber number, fiber type distribution and muscle specific tension), in men and male rodents at similar relative age. It appears that muscle aging in male F344*BN rat resembles that in men most; 32-35-month-old rats exhibit similar signs of muscle weakness to those of 70-80-yr-old men, and the decline in 36-38-month-old rats is similar to that in men aged over 80 yrs. For male C57BL/6 mice, age-related decline in muscle force generating capacity seems to occur only at higher relative age than in men. We conclude that the effects on determinants of muscle force differ between species as well as within species, but qualitatively show the same pattern as that observed in men.
Collapse
Affiliation(s)
- Sam B Ballak
- School of Healthcare Science, Cognitive Motor Function Research Group, Manchester Metropolitan University, Manchester M1 5GD, United Kingdom; Laboratory for Myology, Move Research Institute Amsterdam, Faculty of Human Movement Sciences, VU University Amsterdam, Amsterdam 1081 BT, The Netherlands.
| | - Hans Degens
- School of Healthcare Science, Cognitive Motor Function Research Group, Manchester Metropolitan University, Manchester M1 5GD, United Kingdom
| | - Arnold de Haan
- School of Healthcare Science, Cognitive Motor Function Research Group, Manchester Metropolitan University, Manchester M1 5GD, United Kingdom; Laboratory for Myology, Move Research Institute Amsterdam, Faculty of Human Movement Sciences, VU University Amsterdam, Amsterdam 1081 BT, The Netherlands
| | - Richard T Jaspers
- Laboratory for Myology, Move Research Institute Amsterdam, Faculty of Human Movement Sciences, VU University Amsterdam, Amsterdam 1081 BT, The Netherlands
| |
Collapse
|
34
|
Phelps M, Pettan-Brewer C, Ladiges W, Yablonka-Reuveni Z. Decline in muscle strength and running endurance in klotho deficient C57BL/6 mice. Biogerontology 2013; 14:729-39. [PMID: 24030242 DOI: 10.1007/s10522-013-9447-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 07/23/2013] [Indexed: 12/01/2022]
Abstract
Alpha klotho (known as klotho) is a multifunctional protein that may be linked to age-associated decline in tissue homeostasis. The original klotho hypomorphic (klotho (hm) ) mouse, produced on a mixed C57BL/6 and C3H background, is short lived and exhibits extensive aging-like deterioration of several body systems. Differently, klotho (hm) mice on a pure C57BL/6 background do not appear sickly nor die young, which has permitted us to gain insight into the effect of klotho deficiency in adult life. First, analyzing klotho transcript levels in the kidney, the main site of klotho production, we demonstrated a 71-fold decline in klotho (hm) females compared to wildtype females versus only a 4-fold decline in mutant males. We then examined the effect of klotho deficiency on muscle-related attributes in adult mice, focusing on 7-11 month old females. Body weight and forelimb grip strength were significantly reduced in klotho (hm) mice compared to wildtype and klotho overexpressing mice. The female mice were also subjected to voluntary wheel running for a period of 6 days. Running endurance was markedly reduced in klotho (hm) mice, which exhibited a sporadic running pattern that may be characteristic of repeated bouts of exhaustions. When actually running, klotho (hm) females ran at the same speed as wildtype and klotho overexpressing mice, but spent about 65 % less time running compared to the other two groups. Our novel results suggest an important link between klotho deficiency and muscle performance. This study provides a foundation for further research on klotho involvement as a potential inhibitor of age-associated muscle deterioration.
Collapse
Affiliation(s)
- Michael Phelps
- Department of Biological Structure, School of Medicine, University of Washington, Health Sciences Building, Room G520, 1959 NE Pacific Street, Box 35740, Seattle, WA, 98195, USA
| | | | | | | |
Collapse
|
35
|
Yang L, Zhang J, Zheng K, Shen H, Chen X. Long-term Ginsenoside Rg1 Supplementation Improves Age-Related Cognitive Decline by Promoting Synaptic Plasticity Associated Protein Expression in C57BL/6J Mice. J Gerontol A Biol Sci Med Sci 2013; 69:282-94. [DOI: 10.1093/gerona/glt091] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
36
|
Komarova EA, Antoch MP, Novototskaya LR, Chernova OB, Paszkiewicz G, Leontieva OV, Blagosklonny MV, Gudkov AV. Rapamycin extends lifespan and delays tumorigenesis in heterozygous p53+/- mice. Aging (Albany NY) 2013; 4:709-14. [PMID: 23123616 PMCID: PMC3517941 DOI: 10.18632/aging.100498] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The TOR (Target of Rapamycin) pathway accelerates cellular and organismal aging. Similar to rapamycin, p53 can inhibit the mTOR pathway in some mammalian cells. Mice lacking one copy of p53 (p53+/− mice) have an increased cancer incidence and a shorter lifespan. We hypothesize that rapamycin can delay cancer in heterozygous p53+/− mice. Here we show that rapamycin (given in a drinking water) extended the mean lifespan of p53+/− mice by 10% and when treatment started early in life (at the age less than 5 months) by 28%. In addition, rapamycin decreased the incidence of spontaneous tumors. This observation may have applications in management of Li-Fraumeni syndrome patients characterized by heterozygous mutations in the p53 gene.
Collapse
Affiliation(s)
- Elena A Komarova
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Neuhaus B, Niessen CM, Mesaros A, Withers DJ, Krieg T, Partridge L. Experimental analysis of risk factors for ulcerative dermatitis in mice. Exp Dermatol 2013; 21:712-3. [PMID: 22897579 DOI: 10.1111/j.1600-0625.2012.01558.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Ulcerative dermatitis (UD) is a severe inflammatory skin disorder with an unknown aetiology. Recently, insulin receptor substrate 1 KO mice were shown to be fully resistant to UD. In this study, we showed that high-fat diet (HFD) feeding significantly increased incidence of UD in wild type (WT) C57BL/6 mice, as did lithium-mediated inhibition of GSK3-β, which is a key negative regulator of IRS1. In contrast to WT mice, resistance to UD was fully preserved in HFD-fed Irs1-KO mice. Our results identify IRS1 as a key determinant of UD pathogenesis and establish a direct link between diet composition, obesity-induced inflammation and chronic ulceration.
Collapse
Affiliation(s)
- Brit Neuhaus
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | | | | | | | | | | |
Collapse
|
38
|
Cameron KM, Miwa S, Walker C, von Zglinicki T. Male mice retain a metabolic memory of improved glucose tolerance induced during adult onset, short-term dietary restriction. LONGEVITY & HEALTHSPAN 2012; 1:3. [PMID: 24764509 PMCID: PMC3886267 DOI: 10.1186/2046-2395-1-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 01/06/2012] [Indexed: 01/14/2023]
Abstract
BACKGROUND Chronic dietary restriction (DR) has been shown to have beneficial effects on glucose homeostasis and insulin sensitivity. These factors show rapid and robust improvements when rodents were crossed over from an ad libitum (AL) diet to DR in mid life. We aimed to determine whether the beneficial effects induced by short-term exposure to DR can be retained as a 'metabolic memory' when AL feeding is resumed (AL-DR-AL) and vice versa: whether the effects of long-term DR can be reversed by a period of AL feeding (DR-AL-DR). C57BL/6 male and female mice were used to examine sex differences (N = 10/sex/group). Mice were fed AL or DR from 3 until 15 months (baseline) and each dietary crossover lasted approximately 5 months. RESULTS In females, body and fat mass were proportional to the changes in feeding regime and plasma insulin and glucose tolerance were unaffected by the crossovers. However, in male mice, glucose tolerance and plasma insulin levels were reversed within 6 to 12 weeks. When males returned to AL intake following 5 months DR (AL-DR-AL), body mass was maintained below baseline, proportional to changes in fat mass. Glucose tolerance was also significantly better compared to baseline. CONCLUSIONS Male mice retained a metabolic memory of 5 months of DR feeding in terms of reduced body mass and improved glucose tolerance. This implies that some of the beneficial effects induced by a period of DR in adult life may be beneficial, even when free feeding is resumed at least in males. However, under continuous DR, lifespan extension was more prominent in females than in males.
Collapse
Affiliation(s)
- Kerry M Cameron
- Ageing Research Laboratories, Centre for Integrated Systems Biology of Ageing and Nutrition, Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Satomi Miwa
- Ageing Research Laboratories, Centre for Integrated Systems Biology of Ageing and Nutrition, Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Cornelia Walker
- Ageing Research Laboratories, Centre for Integrated Systems Biology of Ageing and Nutrition, Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Thomas von Zglinicki
- Ageing Research Laboratories, Centre for Integrated Systems Biology of Ageing and Nutrition, Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| |
Collapse
|
39
|
Aires DJ, Rockwell G, Wang T, Frontera J, Wick J, Wang W, Tonkovic-Capin M, Lu J, E L, Zhu H, Swerdlow RH. Potentiation of dietary restriction-induced lifespan extension by polyphenols. Biochim Biophys Acta Mol Basis Dis 2012; 1822:522-6. [PMID: 22265987 DOI: 10.1016/j.bbadis.2012.01.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 01/04/2012] [Indexed: 10/14/2022]
Abstract
Dietary restriction (DR) extends lifespan across multiple species including mouse. Antioxidant plant extracts rich in polyphenols have also been shown to increase lifespan. We hypothesized that polyphenols might potentiate DR-induced lifespan extension. Twenty week old C57BL/6 mice were placed on one of three diets: continuous feeding (control), alternate day chow (Intermittent fed, IF), or IF supplemented with polyphenol antioxidants (PAO) from blueberry, pomegranate, and green tea extracts (IF+PAO). Both IF and IF+PAO groups outlived the control group and the IF+PAO group outlived the IF group (all p<0.001). In the brain, IF induced the expression of inflammatory genes and p38 MAPK phosphorylation, while the addition of PAO reduced brain inflammatory gene expression and p38 MAPK phosphorylation. Our data indicate that while IF overall promotes longevity, some aspects of IF-induced stress may paradoxically lessen this effect. Polyphenol compounds, in turn, may potentiate IF-induced longevity by minimizing specific components of IF-induced cell stress.
Collapse
Affiliation(s)
- Daniel J Aires
- Division of Dermatology, University of Kansas, KS 66160, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Parks RJ, Fares E, MacDonald JK, Ernst MC, Sinal CJ, Rockwood K, Howlett SE. A Procedure for Creating a Frailty Index Based on Deficit Accumulation in Aging Mice. ACTA ACUST UNITED AC 2011; 67:217-27. [DOI: 10.1093/gerona/glr193] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
41
|
Na B, Huang Z, Wang Q, Qi Z, Tian Y, Lu CC, Yu J, Hanes MA, Kakar S, Huang EJ, Ou JHJ, Liu L, Yen TSB. Transgenic expression of entire hepatitis B virus in mice induces hepatocarcinogenesis independent of chronic liver injury. PLoS One 2011; 6:e26240. [PMID: 22022578 PMCID: PMC3192172 DOI: 10.1371/journal.pone.0026240] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 09/22/2011] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC), the third leading cause of cancer deaths worldwide, is most commonly caused by chronic hepatitis B virus (HBV) infection. However, whether HBV plays any direct role in carcinogenesis, other than indirectly causing chronic liver injury by inciting the host immune response, remains unclear. We have established two independent transgenic mouse lines expressing the complete genome of a mutant HBV ("preS2 mutant") that is found at much higher frequencies in people with HCC than those without. The transgenic mice show evidence of stress in the endoplasmic reticulum (ER) and overexpression of cyclin D1 in hepatocytes. These mice do not show any evidence of chronic liver injury, but by 2 years of age a majority of the male mice develop hepatocellular neoplasms, including HCC. Unexpectedly, we also found a significant increase in hepatocarcinogenesis independent of necroinflammation in a transgenic line expressing the entire wildtype HBV. As in the mutant HBV mice, HCC was found only in aged--2-year-old--mice of the wildtype HBV line. The karyotype in all the three transgenic lines appears normal and none of the integration sites of the HBV transgene in the mice is near an oncogene or tumor suppressor gene. The significant increase of HCC incidence in all the three transgenic lines--expressing either mutant or wildtype HBV--therefore argues strongly that in absence of chronic necroinflammation, HBV can contribute directly to the development of HCC.
Collapse
Affiliation(s)
- Bing Na
- Pathology Service, Veterans Administration Medical Center, San Francisco, California, United States of America
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California, United States of America
| | - Zhiming Huang
- Pathology Service, Veterans Administration Medical Center, San Francisco, California, United States of America
| | - Qian Wang
- Pathology Service, Veterans Administration Medical Center, San Francisco, California, United States of America
| | - Zhongxia Qi
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, California, United States of America
| | - Yongjun Tian
- Department of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, California, United States of America
| | - Cheng-Chan Lu
- Department of Pathology, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Jingwei Yu
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, California, United States of America
| | - Martha A. Hanes
- Department of Laboratory Animal Resources, University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Sanjay Kakar
- Pathology Service, Veterans Administration Medical Center, San Francisco, California, United States of America
- Department of Pathology, University of California, San Francisco, San Francisco, California, United States of America
| | - Eric J. Huang
- Pathology Service, Veterans Administration Medical Center, San Francisco, California, United States of America
- Department of Pathology, University of California, San Francisco, San Francisco, California, United States of America
| | - J.-H. James Ou
- Department of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, California, United States of America
| | - Limin Liu
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, California, United States of America
| | - T. S. Benedict Yen
- Pathology Service, Veterans Administration Medical Center, San Francisco, California, United States of America
- Department of Pathology, University of California, San Francisco, San Francisco, California, United States of America
| |
Collapse
|
42
|
Pettan-Brewer C, Treuting PM. Practical pathology of aging mice. PATHOBIOLOGY OF AGING & AGE RELATED DISEASES 2011; 1:PBA-1-7202. [PMID: 22953032 PMCID: PMC3417704 DOI: 10.3402/pba.v1i0.7202] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2011] [Revised: 05/10/2011] [Accepted: 05/10/2011] [Indexed: 11/30/2022]
Abstract
Old mice will have a subset of lesions as part of the progressive decline in organ function that defines aging. External and palpable lesions will be noted by the research, husbandry, or veterinary staff during testing, cage changing, or physical exams. While these readily observable lesions may cause alarm, not all cause undue distress or are life-threatening. In aging research, mice are maintained until near end of life that, depending on strain and genetic manipulation, can be upwards of 33 months. Aging research has unique welfare issues related to age-related decline, debilitation, fragility, and associated pain of chronic diseases. An effective aging research program includes the collaboration and education of the research, husbandry, and veterinary staff, and of the members of the institution animal care and use committee. This collaborative effort is critical to humanely maintaining older mice and preventing excessive censorship due to non-lethal diseases. Part of the educational process is becoming familiar with how old mice appear clinically, at necropsy and histopathologically. This baseline knowledge is important in making the determination of humane end points, defining health span, contributing causes of death and effects of interventions. The goal of this paper is to introduce investigators to age-associated diseases and lesion patterns in mice from clinical presentation to pathologic assessment. To do so, we present and illustrate the common clinical appearances, necropsy and histopathological lesions seen in subsets of the aging colonies maintained at the University of Washington.
Collapse
|
43
|
Deng XH, Bertini G, Palomba M, Xu YZ, Bonaconsa M, Nygård M, Bentivoglio M. Glial transcripts and immune-challenged glia in the suprachiasmatic nucleus of young and aged mice. Chronobiol Int 2010; 27:742-67. [PMID: 20560709 DOI: 10.3109/07420521003681498] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Biological rhythms are frequently disturbed with advancing age, and aging-related changes of glia in the hypothalamic suprachiasmatic nucleus (SCN), the master circadian pacemaker, require special attention. In particular, astrocytes contribute to SCN function, and aging is associated with increased inflammatory activity in the brain, in which microglia could be especially implicated. On this basis, we investigated in the SCN of young and old mice glial transcripts and cell features, and the glial cell response to a central inflammatory challenge. Quantitative real-time reverse transcriptase-polymerase chain reaction (RT-PCR) was used to analyze the expression of mRNAs encoding the astrocytic glial fibrillary acidic protein and the microglial antigen CD11b. Both these transcripts, here investigated in the SCN for the first time, were significantly increased in the old SCN. Glial cell phenotyping with immunohistochemistry revealed hypertrophic and intensely stained astrocytes and microglia in the aged SCN. In both age groups, microglia were scattered throughout the SCN and astrocytes were prominent in the ventral portion, where retinal fibers are densest; in the aged SCN, astrocytes were also numerous in the dorsal portion. After intracerebroventricular injections of a mixture of interferon-gamma and tumor necrosis factor-alpha, or phosphate-buffered saline as control, immunolabeling was evaluated with stereological cell counts and confocal microscopy. Phenotypic features of astrocyte and microglia activation in response to cytokine injections were markedly enhanced in the aged SCN. Subregional variations in glial cell density were also documented in the aged compared to the young SCN. Altogether, the findings show increases in the expression of glial transcripts and hypertrophy of astrocytes and microglia in the aged SCN, as well as age-dependent variation in the responses of immune-challenged SCN glia. The data thus point out an involvement of glia in aging-related changes of the biological clock.
Collapse
Affiliation(s)
- Xiao-Hua Deng
- Department of Neuroscience, University of Verona, Italy
| | | | | | | | | | | | | |
Collapse
|
44
|
Jacobson J, Lambert AJ, Portero-Otín M, Pamplona R, Magwere T, Miwa S, Driege Y, Brand MD, Partridge L. Biomarkers of aging in Drosophila. Aging Cell 2010; 9:466-477. [PMID: 20367621 PMCID: PMC4467031 DOI: 10.1111/j.1474-9726.2010.00573.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Low environmental temperature and dietary restriction (DR) extend lifespan in diverse organisms. In the fruit fly Drosophila, switching flies between temperatures alters the rate at which mortality subsequently increases with age but does not reverse mortality rate. In contrast, DR acts acutely to lower mortality risk; flies switched between control feeding and DR show a rapid reversal of mortality rate. Dietary restriction thus does not slow accumulation of aging-related damage. Molecular species that track the effects of temperatures on mortality but are unaltered with switches in diet are therefore potential biomarkers of aging-related damage. However, molecular species that switch upon instigation or withdrawal of DR are thus potential biomarkers of mechanisms underlying risk of mortality, but not of aging-related damage. Using this approach, we assessed several commonly used biomarkers of aging-related damage. Accumulation of fluorescent advanced glycation end products (AGEs) correlated strongly with mortality rate of flies at different temperatures but was independent of diet. Hence, fluorescent AGEs are biomarkers of aging-related damage in flies. In contrast, five oxidized and glycated protein adducts accumulated with age, but were reversible with both temperature and diet, and are therefore not markers either of acute risk of dying or of aging-related damage. Our approach provides a powerful method for identification of biomarkers of aging.
Collapse
Affiliation(s)
- Jake Jacobson
- Institute for Healthy Ageing, Department of Genes, Evolution and Environment, University College London, London WC1E 6BT, UK; Max Planck Institute for Biology of Ageing, Gleueler Strasse. 50 a, D-50931, Cologne, Germany
| | | | - Manuel Portero-Otín
- Metabolic Pathophysiology Research Group, Department of Experimental Medicine, University of Lleida-IRBLLEIDA, Lleida 25008, Spain
| | - Reinald Pamplona
- Metabolic Pathophysiology Research Group, Department of Experimental Medicine, University of Lleida-IRBLLEIDA, Lleida 25008, Spain
| | - Tapiwanashe Magwere
- Institute for Healthy Ageing, Department of Genes, Evolution and Environment, University College London, London WC1E 6BT, UK; Max Planck Institute for Biology of Ageing, Gleueler Strasse. 50 a, D-50931, Cologne, Germany
| | - Satomi Miwa
- MRC Mitochondrial Biology Unit, Hills Rd, Cambridge CB2 2XY, UK
| | - Yasmine Driege
- Institute for Healthy Ageing, Department of Genes, Evolution and Environment, University College London, London WC1E 6BT, UK; Max Planck Institute for Biology of Ageing, Gleueler Strasse. 50 a, D-50931, Cologne, Germany
| | - Martin D. Brand
- MRC Mitochondrial Biology Unit, Hills Rd, Cambridge CB2 2XY, UK
| | - Linda Partridge
- Institute for Healthy Ageing, Department of Genes, Evolution and Environment, University College London, London WC1E 6BT, UK; Max Planck Institute for Biology of Ageing, Gleueler Strasse. 50 a, D-50931, Cologne, Germany
| |
Collapse
|
45
|
Spindler SR. Caloric restriction: from soup to nuts. Ageing Res Rev 2010; 9:324-53. [PMID: 19853062 DOI: 10.1016/j.arr.2009.10.003] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2009] [Revised: 10/07/2009] [Accepted: 10/09/2009] [Indexed: 12/25/2022]
Abstract
Caloric restriction (CR), reduced protein, methionine, or tryptophan diets; and reduced insulin and/or IGFI intracellular signaling can extend mean and/or maximum lifespan and delay deleterious age-related physiological changes in animals. Mice and flies can shift readily between the control and CR physiological states, even at older ages. Many health benefits are induced by even brief periods of CR in flies, rodents, monkeys, and humans. In humans and nonhuman primates, CR produces most of the physiologic, hematologic, hormonal, and biochemical changes it produces in other animals. In primates, CR provides protection from type 2 diabetes, cardiovascular and cerebral vascular diseases, immunological decline, malignancy, hepatotoxicity, liver fibrosis and failure, sarcopenia, inflammation, and DNA damage. It also enhances muscle mitochondrial biogenesis, affords neuroprotection; and extends mean and maximum lifespan. CR rapidly induces antineoplastic effects in mice. Most claims of lifespan extension in rodents by drugs or nutrients are confounded by CR effects. Transcription factors and co-activators involved in the regulation of mitochondrial biogenesis and energy metabolism, including SirT1, PGC-1alpha, AMPK and TOR may be involved in the lifespan effects of CR. Paradoxically, low body weight in middle aged and elderly humans is associated with increased mortality. Thus, enhancement of human longevity may require pharmaceutical interventions.
Collapse
|
46
|
Wei W, Li B, Hanes MA, Kakar S, Chen X, Liu L. S-nitrosylation from GSNOR deficiency impairs DNA repair and promotes hepatocarcinogenesis. Sci Transl Med 2010; 2:19ra13. [PMID: 20371487 DOI: 10.1126/scitranslmed.3000328] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human hepatocellular carcinoma (HCC) is associated with elevated expression of inducible nitric oxide synthase (iNOS), but the role of nitric oxide in the pathogenesis of HCC remains unknown. We found that the abundance and activity of S-nitrosoglutathione reductase (GSNOR), a protein critical for control of protein S-nitrosylation, were significantly decreased in approximately 50% of patients with HCC. GSNOR-deficient mice were very susceptible to spontaneous and carcinogen-induced HCC. During inflammatory responses, the livers of GSNOR-deficient mice exhibited substantial S-nitrosylation and proteasomal degradation of the key DNA repair protein O(6)-alkylguanine-DNA alkyltransferase. As a result, repair of carcinogenic O(6)-alkylguanines in GSNOR-deficient mice was significantly impaired. Predisposition to HCC, S-nitrosylation and depletion of alkylguanine-DNA alkyltransferase, and accumulation of O(6)-alkylguanines were all abolished in mice deficient in both GSNOR and iNOS. Thus, our data suggest that GSNOR deficiency, through dysregulated S-nitrosylation, may promote HCC, possibly by inactivating a DNA repair system.
Collapse
Affiliation(s)
- Wei Wei
- Department of Microbiology and Immunology, University of California, San Francisco, CA 94143, USA
| | | | | | | | | | | |
Collapse
|
47
|
Conover CA, Bale LK, Mader JR, Mason MA, Keenan KP, Marler RJ. Longevity and age-related pathology of mice deficient in pregnancy-associated plasma protein-A. J Gerontol A Biol Sci Med Sci 2010; 65:590-9. [PMID: 20351075 DOI: 10.1093/gerona/glq032] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The pregnancy-associated plasma protein-A knockout (PAPP-A KO) mouse is a model of reduced local insulin-like growth factor (IGF)-I activity with normal circulating IGF-I levels. In this study, PAPP-A KO mice had significantly increased mean (27%), median (27%), and maximum (35%) life span compared with wild-type (WT) littermates. End-of-life pathology indicated that the incidence of neoplastic disease was not significantly different in the two groups of mice; however, it occurred in older aged PAPP-A KO compared with WT mice. Furthermore, PAPP-A KO mice were less likely to show degenerative changes of age. Scheduled pathologies at 78, 104, and 130 weeks of age indicated that WT mice, in general, had more degenerative changes and tumors earlier than PAPP-A KO mice. This was particularly true for abnormalities in heart, testes, brain, kidney, spleen, and thymus. In summary, the major contributors to the extended life span of PAPP-A KO mice are delayed occurrence of fatal neoplasias and decreased incidence of age-related degenerative changes.
Collapse
Affiliation(s)
- Cheryl A Conover
- The Division of Endocrinology and Metabolism, Department of Medicine, Mayo Clinic, 200 First Street SW, 5-194 Joseph, Rochester, MN 55905, USA.
| | | | | | | | | | | |
Collapse
|
48
|
Harper JM, Wilkinson JE, Miller RA. Macrophage migration inhibitory factor-knockout mice are long lived and respond to caloric restriction. FASEB J 2010; 24:2436-42. [PMID: 20219983 DOI: 10.1096/fj.09-152223] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Macrophage migration inhibitory factor (MIF) affects inflammation, glucose homeostasis, and cellular proliferation in mammals. Previously, we found that MIF was significantly elevated in multiple long-lived mouse models, including calorie restriction (CR), which led us to hypothesize that MIF might be important in the control of mammalian life span and be necessary for the life-extending effects of CR. To test this hypothesis, we examined the life span of mice with a targeted deletion of the Mif gene on a segregating B6 x 129/Sv background (MIF-KO) under ad libitum (AL) feeding and CR conditions. Control mice were generated by mating C57BL/6J females with 129/SvJ males to make an F1 hybrid, and crossing F1 males to F1 females to produce segregating F2 mice homozygous for the normal MIF allele. Not only did MIF-KO mice show a life span extension in response to CR, they were, unexpectedly, longer lived than controls under standard AL conditions. MIF-KO mice were significantly protected against lethal hemangiosarcoma, but more likely than controls to die of disseminated amyloid, an age-related inflammatory syndrome. Overall, these data refute the suggestion that MIF is required for the CR effect on life span, but raise the possibility that MIF may limit life span in normal mice.
Collapse
Affiliation(s)
- James M Harper
- Department of Pathology and Geriatrics Center, University of Michigan, Ann Arbor, Michigan, USA.
| | | | | |
Collapse
|
49
|
Regulation of cytokine signaling and T-cell recruitment in the aging mouse brain in response to central inflammatory challenge. Brain Behav Immun 2010; 24:138-52. [PMID: 19765643 DOI: 10.1016/j.bbi.2009.09.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Revised: 09/04/2009] [Accepted: 09/11/2009] [Indexed: 01/26/2023] Open
Abstract
Aging is often accompanied by increased levels of inflammatory molecules in the organism, but age-related changes in the brain response to inflammatory challenges still require clarification. We here investigated in mice whether cytokine signaling and T-cell neuroinvasion undergo age-related changes. We first analyzed the expression of molecules involved in T-cell infiltration and cytokine signaling regulation in the septum and hippocampus of 2-3 months and 20- to 24-month-old mice at 4h after intracerebroventricular injections of tumor necrosis factor (TNF)-alpha or interferon-gammaversus saline injections. Transcripts of the chemokine CXCL9, intercellular adhesion molecule (ICAM)-1 and suppressor of cytokine signaling molecules (SOCS) 1 and 3 were increased in both age groups after cytokine injection; microglia-derived matrix metalloproteinase (MMP) 12 mRNA was induced in old mice also after control saline injections. Age-related changes in ICAM-1 protein expression and T-cell infiltration were then analyzed in mice of 3-4, 8-9 and 15-16 months at 48h after TNF-alpha injections. ICAM-1 immunoreactivity, and Western blotting in striatum, septum, hippocampus and hypothalamus showed progressive age-related enhancement of TNF-alpha-elicited ICAM-1 upregulation. Double immunofluorescence revealed ICAM-1 expression in microglia and astrocytic processes. CD3(+), CD4(+) and CD8(+) T-cells exhibited progressive age-related increases in brain parenchyma and choroid plexus after cytokine exposure. The findings indicate that the brain responses to inflammatory challenges are not only preserved with advancing age, but also include gradual amplification of ICAM-1 expression and T-cell recruitment. The data highlight molecular and cellular correlates of age-related increase of brain sensitivity to inflammatory stimuli, which could be involved in altered brain vulnerability during aging.
Collapse
|
50
|
Fahlström A, Yu Q, Ulfhake B. Behavioral changes in aging female C57BL/6 mice. Neurobiol Aging 2009; 32:1868-80. [PMID: 20005598 DOI: 10.1016/j.neurobiolaging.2009.11.003] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2009] [Revised: 10/21/2009] [Accepted: 11/03/2009] [Indexed: 12/22/2022]
Abstract
Using a range of tests we have studied alterations in behavior with advancing age in female C57BL/6 (of Jackson origin), the golden standard on which most genetically engineered mice are back-crossed. In parallel, growth and survival data were collected. In a protected environment the 90% and 75% cohort survival age was 20 and 25 months, respectively, and the 50% cohort survival was 32 months. In mice, body weight increases continuously until 15-20 months of age, while in advanced age whole body weight drops. The body mass loss in senescence is associated with emergence of other aged phenotype features such as kyphosis, balding and loss of fur-color. Our behavioral data show that aging modulates certain aspects of basic behavior in a continuous manner, like explorative and locomotor activities. Advanced age associates with an acceleration of behavioral impairments evident in most of the tests used, including motor skill acquisition and memory consolidation. However, certain domains of mouse behavior were well preserved also in advanced age such as thermal noxious threshold and working memory as assessed by an object recognition task. The decreased drive to explore is suggested to be a key factor underlying many aspects of reduced performance including cognitive capacity during aging. Behavioral aging affects genetically closely related individuals housed under strictly standardized conditions differentially (Collier, T.J., Coleman, P.D., 1991. Divergence of biological and chronological aging: evidence from rodent studies. Neurobiol. Aging, 12, 685-693; Ingram, D.K., 1988. Motor performance variability during aging in rodents. Assessment of reliability and validity of individual differences. Ann. N.Y. Acad. Sci., 515, 70-96). Consistent with this a subpopulation of the 28-month-old mice showed an explorative activity similar to young-adult mice and a significantly stronger preference for a novel object than aged mice with a less explorative behavior. Thus, subtle environmental factors and epigenetic modifications may be important modulators of aging.
Collapse
Affiliation(s)
- Andreas Fahlström
- Experimental Neurogerontology, Department of Neuroscience, Karolinska Institutet, Retzius väg 8, 171 77 Stockholm, Sweden
| | | | | |
Collapse
|