1
|
Eriksson M, Nylén S, Grönvik KO. Passive immunization of mice with IgY anti-H5N1 protects against experimental influenza virus infection and allows development of protective immunity. Vaccine 2024; 42:126133. [PMID: 39019655 DOI: 10.1016/j.vaccine.2024.07.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 06/24/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024]
Abstract
Influenza virus contributes substantially to the global human and animal disease burden. To protect individuals against disease, strategies are needed to minimize the time an individual is at risk of developing disease symptoms. Passive immunization using avian IgY antibodies can protect individuals against a variety of pathogens, including influenza virus. Yet the effect of IgY administration on generation of protective immunity is largely unknown. To address the effect of passive immunization on the host immune response development, adult or aged, male and female C57BL/6NCrl mice received chicken IgY anti-H5N1, normal IgY or PBS intranasally four hours before, and 20 hours after intranasal infection with H1N1 influenza A virus (PR8). The mice receiving cross-reactive IgY anti-H5N1 were protected from disease and developed influenza virus-specific memory T cells similar to control-treated mice. When re-challenged with PR8 35 days post primary infection IgY anti-H5N1-treated mice were fully protected. Moreover, when challenged with heterologous H3N2 influenza A virus (X-31) or with PR8 three months post infection the mice were protected against severe disease and death, albeit a slight transient weight loss was noted. The results show that passive immunization with IgY anti-H5N1 is safe and protects mice against disease induced by influenza virus without inhibiting development of protective immunity after virus exposure. This indicate that passive immunization can be used as prophylactic therapy in combination with immunization to prevent disease.
Collapse
Affiliation(s)
- Malin Eriksson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden; Department of Microbiology, Swedish Veterinary Agency, 751 89 Uppsala, Sweden.
| | - Susanne Nylén
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden.
| | - Kjell-Olov Grönvik
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden; Uppsala Immunobiology Lab, 752 37 Uppsala, Sweden.
| |
Collapse
|
2
|
Moussa AT, Cosenza MR, Wohlfromm T, Brobeil K, Hill A, Patrizi A, Müller-Decker K, Holland-Letz T, Jauch A, Kraft B, Krämer A. STIL overexpression shortens lifespan and reduces tumor formation in mice. PLoS Genet 2024; 20:e1011460. [PMID: 39466849 DOI: 10.1371/journal.pgen.1011460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/14/2024] [Indexed: 10/30/2024] Open
Abstract
Centrosomes are the major microtubule organizing centers of animal cells. Supernumerary centrosomes are a common feature of human tumors and associated with karyotype abnormalities and aggressive disease, but whether they are cause or consequence of cancer remains controversial. Here, we analyzed the consequences of centrosome amplification by generating transgenic mice in which centrosome numbers can be increased by overexpression of the structural centrosome protein STIL. We show that STIL overexpression induces centrosome amplification and aneuploidy, leading to senescence, apoptosis, and impaired proliferation in mouse embryonic fibroblasts, and microcephaly with increased perinatal lethality and shortened lifespan in mice. Importantly, both overall tumor formation in mice with constitutive, global STIL overexpression and chemical skin carcinogenesis in animals with inducible, skin-specific STIL overexpression were reduced, an effect that was not rescued by concomitant interference with p53 function. These results suggest that supernumerary centrosomes impair proliferation in vitro as well as in vivo, resulting in reduced lifespan and delayed spontaneous as well as carcinogen-induced tumor formation.
Collapse
Affiliation(s)
- Amira-Talaat Moussa
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
- Department of Pathology, Faculty of Veterinary Medicine, Zagazig University, Al Sharkia, Egypt
| | - Marco R Cosenza
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Timothy Wohlfromm
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Katharina Brobeil
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Anthony Hill
- Schaller Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Annarita Patrizi
- Schaller Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Karin Müller-Decker
- Core Facility Tumor Models, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tim Holland-Letz
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anna Jauch
- Institute of Human Genetics, University of Heidelberg, Heidelberg, Germany
| | - Bianca Kraft
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Alwin Krämer
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
3
|
Pratt EC, Mezzadra R, Kulick A, Kaminsky S, Samuels ZV, Loor A, de Stanchina E, Lowe SW, Lewis JS. uPAR Immuno-PET in Pancreatic Cancer, Aging, and Chemotherapy-Induced Senescence. J Nucl Med 2024:jnumed.124.268278. [PMID: 39362768 DOI: 10.2967/jnumed.124.268278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/13/2024] [Indexed: 10/05/2024] Open
Abstract
Identifying cancer therapy resistance is a key time-saving tool for physicians. Part of chemotherapy resistance includes senescence, a persistent state without cell division or cell death. Chemically inducing senescence with the combination of trametinib and palbociclib (TP) yields several tumorigenic and prometastatic factors in pancreatic cancer models with many potential antibody-based targets. In particular, urokinase plasminogen activator receptor (uPAR) has been shown to be a membrane-bound marker of senescence in addition to an oncology target. Methods: Here, 2 antibodies against murine uPAR and human uPAR were developed as immuno-PET agents to noninvasively track uPAR antigen abundance. Results: TP treatment increased cell uptake both in murine KPC cells and in human MiaPaCa2 cells. In vivo, subcutaneously implanted murine KPC tumors had high tumor uptake with the antimurine uPAR antibody independently of TP in young mice, yet uPAR uptake was maintained in aged mice on TP. Mice xenografted with human MiaPaCa2 tumors showed a significant increase in tumor uptake on TP therapy when imaged with the antihuman uPAR antibody. Imaging with either uPAR antibody was found to be more tumor-selective than imaging with [18F]FDG or [18F]F-DPA-714. Conclusion: The use of radiolabeled uPAR-targeting antibodies provides a new antibody-based PET imaging candidate for pancreatic cancer imaging as well as chemotherapy-induced senescence.
Collapse
Affiliation(s)
- Edwin C Pratt
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Riccardo Mezzadra
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Amanda Kulick
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Spencer Kaminsky
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Zachary V Samuels
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Angelique Loor
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elisa de Stanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Scott W Lowe
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- HHMI, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York;
- Department of Pharmacology, Weill Cornell Graduate School, New York, New York
| |
Collapse
|
4
|
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
|
5
|
Standley A, Xie J, Lau AW, Grote L, Gifford AJ. Working with Miraculous Mice: Mus musculus as a Model Organism. Curr Protoc 2024; 4:e70021. [PMID: 39435766 DOI: 10.1002/cpz1.70021] [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: 10/23/2024]
Abstract
The laboratory mouse has been described as a "miracle" model organism, providing a window by which we may gain an understanding of ourselves. Since the first recorded mouse experiment in 1664, the mouse has become the most used animal model in biomedical research. Mice are ideally suited as a model organism because of their small size, short gestation period, large litter size, and genetic similarity to humans. This article provides a broad overview of the laboratory mouse as a model organism and is intended for undergraduates and those new to working with mice. We delve into the history of the laboratory mouse and outline important terminology to accurately describe research mice. The types of laboratory mice available to researchers are reviewed, including outbred stocks, inbred strains, immunocompromised mice, and genetically engineered mice. The critical role mice have played in advancing knowledge in the areas of oncology, immunology, and pharmacology is highlighted by examining the significant contribution of mice to Nobel Prize winning research. International mouse mutagenesis programs and accurate phenotyping of mouse models are outlined. We also explain important considerations for working with mice, including animal ethics; the welfare principles of replacement, refinement, and reduction; and the choice of mouse model in experimental design. Finally, we present practical advice for maintaining a mouse colony, which involves adequate training of staff, the logistics of mouse housing, monitoring colony health, and breeding strategies. Useful resources for working with mice are also listed. The aim of this overview is to equip the reader with a broad appreciation of the enormous potential and some of the complexities of working with the laboratory mouse in a quest to improve human health. © 2024 The Author(s). Current Protocols published by Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Anick Standley
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Jinhan Xie
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Angelica Wy Lau
- Garvan Institute of Medical Research, St Vincent's Clinical School, Darlinghurst, NSW, Australia
| | - Lauren Grote
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Andrew J Gifford
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
- Anatomical Pathology, NSW Heath Pathology, Prince of Wales Hospital, Randwick, NSW, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
6
|
Kapadia CD, Williams N, Dawson KJ, Watson C, Yousefzadeh MJ, Le D, Nyamondo K, Cagan A, Waldvogel S, De La Fuente J, Leongamornlert D, Mitchell E, Florez MA, Aguilar R, Martell A, Guzman A, Harrison D, Niedernhofer LJ, King KY, Campbell PJ, Blundell J, Goodell MA, Nangalia J. Clonal dynamics and somatic evolution of haematopoiesis in mouse. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.17.613129. [PMID: 39345649 PMCID: PMC11429886 DOI: 10.1101/2024.09.17.613129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Haematopoietic stem cells maintain blood production throughout life. While extensively characterised using the laboratory mouse, little is known about how the population is sustained and evolves with age. We isolated stem cells and progenitors from young and old mice, identifying 221,890 somatic mutations genome-wide in 1845 single cell-derived colonies, and used phylogenetic analysis to infer the ontogeny and population dynamics of the stem cell pool. Mouse stem cells and progenitors accrue ~45 somatic mutations per year, a rate only about 2-fold greater than human progenitors despite the vastly different organismal sizes and lifespans. Phylogenetic patterns reveal that stem and multipotent progenitor cell pools are both established during embryogenesis, after which they independently self-renew in parallel over life. The stem cell pool grows steadily over the mouse lifespan to approximately 70,000 cells, self-renewing about every six weeks. Aged mice did not display the profound loss of stem cell clonal diversity characteristic of human haematopoietic ageing. However, targeted sequencing revealed small, expanded clones in the context of murine ageing, which were larger and more numerous following haematological perturbations and exhibited a selection landscape similar to humans. Our data illustrate both conserved features of population dynamics of blood and distinct patterns of age-associated somatic evolution in the short-lived mouse.
Collapse
Affiliation(s)
- Chiraag D. Kapadia
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
| | | | - Kevin J. Dawson
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Caroline Watson
- Early Cancer Institute, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Matthew J. Yousefzadeh
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Columbia Center for Translational Immunology, Columbia Center for Human Longevity, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Duy Le
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, TX, USA
| | - Kudzai Nyamondo
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
| | - Alex Cagan
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Departments of Genetics, Pathology & Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Sarah Waldvogel
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
| | - Josephine De La Fuente
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
| | | | - Emily Mitchell
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Marcus A. Florez
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, TX, USA
| | - Rogelio Aguilar
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
| | - Alejandra Martell
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
| | - Anna Guzman
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
| | | | - Laura J. Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Katherine Y. King
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, TX, USA
| | | | - Jamie Blundell
- Early Cancer Institute, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Margaret A. Goodell
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
| | - Jyoti Nangalia
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| |
Collapse
|
7
|
Fajac A, Simeonova I, Leemput J, Gabriel M, Morin A, Lejour V, Hamon A, Rakotopare J, Vaysse-Zinkhöfer W, Eldawra E, Pinskaya M, Morillon A, Bourdon JC, Bardot B, Toledo F. Mutant mice lacking alternatively spliced p53 isoforms unveil Ackr4 as a male-specific prognostic factor in Myc-driven B-cell lymphomas. eLife 2024; 13:RP92774. [PMID: 39298333 PMCID: PMC11412721 DOI: 10.7554/elife.92774] [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] [Indexed: 09/21/2024] Open
Abstract
The Trp53 gene encodes several isoforms of elusive biological significance. Here, we show that mice lacking the Trp53 alternatively spliced (AS) exon, thereby expressing the canonical p53 protein but not isoforms with the AS C-terminus, have unexpectedly lost a male-specific protection against Myc-induced B-cell lymphomas. Lymphomagenesis was delayed in Trp53+/+Eμ-Myc males compared to Trp53ΔAS/ΔAS Eμ-Myc males, but also compared to Trp53+/+Eμ-Myc and Trp53ΔAS/ΔAS Eμ-Myc females. Pre-tumoral splenic cells from Trp53+/+Eμ-Myc males exhibited a higher expression of Ackr4, encoding an atypical chemokine receptor with tumor suppressive effects. We identified Ackr4 as a p53 target gene whose p53-mediated transactivation is inhibited by estrogens, and as a male-specific factor of good prognosis relevant for murine Eμ-Myc-induced and human Burkitt lymphomas. Furthermore, the knockout of ACKR4 increased the chemokine-guided migration of Burkitt lymphoma cells. These data demonstrate the functional relevance of alternatively spliced p53 isoforms and reveal sex disparities in Myc-driven lymphomagenesis.
Collapse
Affiliation(s)
- Anne Fajac
- Genetics of Tumor Suppression, Institut CurieParisFrance
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
| | - Iva Simeonova
- Genetics of Tumor Suppression, Institut CurieParisFrance
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
| | - Julia Leemput
- Genetics of Tumor Suppression, Institut CurieParisFrance
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
| | - Marc Gabriel
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
- Non Coding RNA, Epigenetic and Genome Fluidity, Institut CurieParisFrance
| | - Aurélie Morin
- Genetics of Tumor Suppression, Institut CurieParisFrance
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
| | - Vincent Lejour
- Genetics of Tumor Suppression, Institut CurieParisFrance
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
| | - Annaïg Hamon
- Genetics of Tumor Suppression, Institut CurieParisFrance
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
| | - Jeanne Rakotopare
- Genetics of Tumor Suppression, Institut CurieParisFrance
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
| | - Wilhelm Vaysse-Zinkhöfer
- Genetics of Tumor Suppression, Institut CurieParisFrance
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
| | - Eliana Eldawra
- Genetics of Tumor Suppression, Institut CurieParisFrance
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
| | - Marina Pinskaya
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
- Non Coding RNA, Epigenetic and Genome Fluidity, Institut CurieParisFrance
| | - Antonin Morillon
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
- School of Medicine, Ninewells Hospital, University of DundeeDundeeUnited Kingdom
| | | | - Boris Bardot
- Genetics of Tumor Suppression, Institut CurieParisFrance
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
| | - Franck Toledo
- Genetics of Tumor Suppression, Institut CurieParisFrance
- CNRS UMR3244ParisFrance
- Sorbonne UniversityParisFrance
- PSL Research UniversityParisFrance
| |
Collapse
|
8
|
Carr LM, Mustafa S, Care A, Collins-Praino LE. More than a number: Incorporating the aged phenotype to improve in vitro and in vivo modeling of neurodegenerative disease. Brain Behav Immun 2024; 119:554-571. [PMID: 38663775 DOI: 10.1016/j.bbi.2024.04.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 03/04/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024] Open
Abstract
Age is the number one risk factor for developing a neurodegenerative disease (ND), such as Alzheimer's disease (AD) or Parkinson's disease (PD). With our rapidly ageing world population, there will be an increased burden of ND and need for disease-modifying treatments. Currently, however, translation of research from bench to bedside in NDs is poor. This may be due, at least in part, to the failure to account for the potential effect of ageing in preclinical modelling of NDs. While ageing can impact upon physiological response in multiple ways, only a limited number of preclinical studies of ND have incorporated ageing as a factor of interest. Here, we evaluate the aged phenotype and highlight the critical, but unmet, need to incorporate aspects of this phenotype into both the in vitro and in vivo models used in ND research. Given technological advances in the field over the past several years, we discuss how these could be harnessed to create novel models of ND that more readily incorporate aspects of the aged phenotype. This includes a recently described in vitro panel of ageing markers, which could help lead to more standardised models and improve reproducibility across studies. Importantly, we cannot assume that young cells or animals yield the same responses as seen in the context of ageing; thus, an improved understanding of the biology of ageing, and how to appropriately incorporate this into the modelling of ND, will ensure the best chance for successful translation of new therapies to the aged patient.
Collapse
Affiliation(s)
- Laura M Carr
- School of Biomedicine, University of Adelaide, Adelaide, SA, Australia
| | - Sanam Mustafa
- School of Biomedicine, University of Adelaide, Adelaide, SA, Australia; Australian Research Council Centre of Excellence for Nanoscale Biophotonics, The University of Adelaide, Adelaide, SA, Australia; Davies Livestock Research Centre, The University of Adelaide, Roseworthy, SA, Australia
| | - Andrew Care
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Lyndsey E Collins-Praino
- School of Biomedicine, University of Adelaide, Adelaide, SA, Australia; Australian Research Council Centre of Excellence for Nanoscale Biophotonics, The University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
9
|
Nemoto S, Uchida K, Ohno H. Implications of Genetic Factors Underlying Mouse Hydronephrosis: Cautionary Considerations on Phenotypic Interpretation in Genetically Engineered Mice. Int J Mol Sci 2024; 25:7203. [PMID: 39000307 PMCID: PMC11241513 DOI: 10.3390/ijms25137203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Hydronephrosis, the dilation of kidneys due to abnormal urine retention, occurs spontaneously in certain inbred mouse strains. In humans, its occurrence is often attributed to acquired urinary tract obstructions in adults, whereas in children, it can be congenital. However, the genetic factors underlying hydronephrosis pathogenesis remain unclear. We investigated the cause of hydronephrosis by analyzing tetraspanin 7 (Tspan7) gene-modified mice, which had shown a high incidence of hydronephrosis-like symptoms. We found that these mice were characterized by low liver weights relative to kidney weights and elevated blood ammonia levels, suggesting liver involvement in hydronephrosis. Gene expression analysis of the liver suggested that dysfunction of ornithine transcarbamylase (OTC), encoded by the X chromosome gene Otc and involved in the urea cycle, may contribute as a congenital factor in hydronephrosis. This OTC dysfunction may be caused by genomic mutations in X chromosome genes contiguous to Otc, such as Tspan7, or via the genomic manipulations used to generate transgenic mice, including the introduction of Cre recombinase DNA cassettes and cleavage of loxP by Cre recombinase. Therefore, caution should be exercised in interpreting the hydronephrosis phenotype observed in transgenic mice as solely a physiological function of the target gene.
Collapse
Affiliation(s)
- Shino Nemoto
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Kazuyo Uchida
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
- Laboratory for Immune Regulation, Graduate School of Medical and Pharmaceutical Sciences, Chiba University, Chiba 260-0856, Japan
- Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama 230-0045, Japan
| |
Collapse
|
10
|
Elmore SA, Rehg JE, Schoeb TR, Everitt JI, Bolon B. Pathologists' perspective on the study design, analysis, and interpretation of proliferative lesions in a lifetime rodent carcinogenicity bioassay of sucralose. Food Chem Toxicol 2024; 188:114524. [PMID: 38428799 DOI: 10.1016/j.fct.2024.114524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/15/2024] [Accepted: 02/18/2024] [Indexed: 03/03/2024]
Abstract
Sucralose, a sugar substitute first approved for use in 1991, is a non-caloric sweetener regulated globally as a food additive. Based on numerous experimental animal studies (dating to the 1980s) and human epidemiology studies, international health agencies have determined that sucralose is safe when consumed as intended. A single lifetime rodent carcinogenicity bioassay conducted by the Ramazzini Institute (RI) reported that mice fed diets containing sucralose develop hematopoietic neoplasia, but controversy continues regarding the validity and relevance of these data for predicting health effects in humans. The present paper addresses the controversy by providing the perspective of experienced pathologists on sucralose-related animal toxicity and carcinogenicity data generally, and the RI carcinogenicity bioassay findings specifically, using results from publicly available papers and international regulatory authority decisions. In the authors' view, flaws in the design, methodology, data evaluation, and reporting of the RI carcinogenicity bioassay for sucralose diminish the value of the data as evidence that this agent represents a carcinogenic hazard to humans. This limitation will remain until the RI bioassay is repeated under Good Laboratory Practices and the design, data, and accuracy of the pathology diagnoses and interpretations are reviewed by qualified pathologists with experience in evaluating potential chemically-induced carcinogenic hazards.
Collapse
Affiliation(s)
| | - Jerold E Rehg
- Department of Pathology, Emeritus, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Trenton R Schoeb
- Department of Genetics and Animal Resources Program, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jeffrey I Everitt
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | | |
Collapse
|
11
|
Frost OG, Ramkilawan P, Rebbaa A, Stolzing A. A systematic review of lifespan studies in rodents using stem cell transplantations. Ageing Res Rev 2024; 97:102295. [PMID: 38588866 DOI: 10.1016/j.arr.2024.102295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 04/10/2024]
Abstract
Organismal aging involves the progressive decline in organ function and increased susceptibility to age-associated diseases. Regardless of its origin, cellular aging is consequently reflected at the level of organ and associated systems dysfunction. Aging of stem cell populations within the body and their decreased ability to self-renew, differentiate, and regenerate damaged tissues, is a key contributor to organismal decline. Based on this, supplementing young stem cells may delay tissue aging, improve frailty and extend health and lifespan. This review investigates studies in rodents using stem cell transplantation from either mice or human donors. The aim is to consolidate available information on the efficacy of stem cell therapies in rodent models and provide insights to guide further research efforts. Out of the 21 studies included in this review, the methodology varied significantly including the lifespan measurement. To enable comparison the median lifespan was calculated using WebPlotDigitizer 4.6 if not provided by the literature. A total of 18 out of 21 studies evidenced significant lifespan extension post stem cell transplant, with 7 studies demonstrating benefits in reduced frailty and other aging complications.
Collapse
Affiliation(s)
- Oliver G Frost
- Centre for Biological Engineering, School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough LE11 3TU, UK; SENS Research Foundation, Mountain View, CA 94041, USA
| | | | | | - Alexandra Stolzing
- Centre for Biological Engineering, School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough LE11 3TU, UK.
| |
Collapse
|
12
|
Poudel SB, Frikha-Benayed D, Ruff RR, Yildirim G, Dixit M, Korstanje R, Robinson L, Miller RA, Harrison DE, Strong JR, Schaffler MB, Yakar S. Targeting mitochondrial dysfunction using methylene blue or mitoquinone to improve skeletal aging. Aging (Albany NY) 2024; 16:4948-4964. [PMID: 38535998 PMCID: PMC11006499 DOI: 10.18632/aging.205147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/27/2023] [Indexed: 04/06/2024]
Abstract
Methylene blue (MB) is a well-established antioxidant that has been shown to improve mitochondrial function in both in vitro and in vivo settings. Mitoquinone (MitoQ) is a selective antioxidant that specifically targets mitochondria and effectively reduces the accumulation of reactive oxygen species. To investigate the effect of long-term administration of MB on skeletal morphology, we administered MB to aged (18 months old) female C57BL/J6 mice, as well as to adult male and female mice with a genetically diverse background (UM-HET3). Additionally, we used MitoQ as an alternative approach to target mitochondrial oxidative stress during aging in adult female and male UM-HET3 mice. Although we observed some beneficial effects of MB and MitoQ in vitro, the administration of these compounds in vivo did not alter the progression of age-induced bone loss. Specifically, treating 18-month-old female mice with MB for 6 or 12 months did not have an effect on age-related bone loss. Similarly, long-term treatment with MB from 7 to 22 months or with MitoQ from 4 to 22 months of age did not affect the morphology of cortical bone at the mid-diaphysis of the femur, trabecular bone at the distal-metaphysis of the femur, or trabecular bone at the lumbar vertebra-5 in UM-HET3 mice. Based on our findings, it appears that long-term treatment with MB or MitoQ alone, as a means to reduce skeletal oxidative stress, is insufficient to inhibit age-associated bone loss. This supports the notion that interventions solely with antioxidants may not provide adequate protection against skeletal aging.
Collapse
Affiliation(s)
- Sher Bahadur Poudel
- Department of Molecular Pathobiology, David B. Kriser Dental Center, New York University College of Dentistry, New York, NY 10010-4086, USA
| | - Dorra Frikha-Benayed
- Department of Biomedical Engineering, City College of New York, New York, NY 10031, USA
| | - Ryan R. Ruff
- Department of Epidemiology and Health Promotion, David B. Kriser Dental Center, New York University College of Dentistry, New York, NY 10010-4086, USA
| | - Gozde Yildirim
- Department of Molecular Pathobiology, David B. Kriser Dental Center, New York University College of Dentistry, New York, NY 10010-4086, USA
| | - Manisha Dixit
- Department of Molecular Pathobiology, David B. Kriser Dental Center, New York University College of Dentistry, New York, NY 10010-4086, USA
| | - Ron Korstanje
- Jackson Aging Center, Nathan Shock Center for Excellence in the Basic Biology of Aging, The Jackson’s Laboratories, Aging Center, Bar Harbor, ME 04609, USA
| | - Laura Robinson
- Jackson Aging Center, Nathan Shock Center for Excellence in the Basic Biology of Aging, The Jackson’s Laboratories, Aging Center, Bar Harbor, ME 04609, USA
| | - Richard A. Miller
- Department of Pathology and Geriatrics Center, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - John R. Strong
- Geriatric Research, Education and Clinical Center and Research Service, South Texas Veterans Health Care System, San Antonio, TX 78229, USA
- Department of Pharmacology, Barshop Institute for Longevity and Aging Studies, The University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Mitchell B. Schaffler
- Department of Biomedical Engineering, City College of New York, New York, NY 10031, USA
| | - Shoshana Yakar
- Department of Molecular Pathobiology, David B. Kriser Dental Center, New York University College of Dentistry, New York, NY 10010-4086, USA
| |
Collapse
|
13
|
Lagou MK, Argyris DG, Vodopyanov S, Gunther-Cummins L, Hardas A, Poutahidis T, Panorias C, DesMarais S, Entenberg C, Carpenter RS, Guzik H, Nishku X, Churaman J, Maryanovich M, DesMarais V, Macaluso FP, Karagiannis GS. Morphometric Analysis of the Thymic Epithelial Cell (TEC) Network Using Integrated and Orthogonal Digital Pathology Approaches. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.11.584509. [PMID: 38559037 PMCID: PMC10979902 DOI: 10.1101/2024.03.11.584509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The thymus, a central primary lymphoid organ of the immune system, plays a key role in T cell development. Surprisingly, the thymus is quite neglected with regards to standardized pathology approaches and practices for assessing structure and function. Most studies use multispectral flow cytometry to define the dynamic composition of the thymus at the cell population level, but they are limited by lack of contextual insight. This knowledge gap hinders our understanding of various thymic conditions and pathologies, particularly how they affect thymic architecture, and subsequently, immune competence. Here, we introduce a digital pathology pipeline to address these challenges. Our approach can be coupled to analytical algorithms and utilizes rationalized morphometric assessments of thymic tissue, ranging from tissue-wide down to microanatomical and ultrastructural levels. This pipeline enables the quantitative assessment of putative changes and adaptations of thymic structure to stimuli, offering valuable insights into the pathophysiology of thymic disorders. This versatile pipeline can be applied to a wide range of conditions that may directly or indirectly affect thymic structure, ranging from various cytotoxic stimuli inducing acute thymic involution to autoimmune diseases, such as myasthenia gravis. Here, we demonstrate applicability of the method in a mouse model of age-dependent thymic involution, both by confirming established knowledge, and by providing novel insights on intrathymic remodeling in the aged thymus. Our orthogonal pipeline, with its high versatility and depth of analysis, promises to be a valuable and practical toolset for both basic and translational immunology laboratories investigating thymic function and disease.
Collapse
Affiliation(s)
- Maria K Lagou
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Tumor Microenvironment and Metastasis Program, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, USA
| | - Dimitrios G Argyris
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Tumor Microenvironment and Metastasis Program, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, USA
- Integrated Imaging Program for Cancer Research, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, USA
| | - Stepan Vodopyanov
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Tumor Microenvironment and Metastasis Program, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, USA
| | - Leslie Gunther-Cummins
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Montefiore-Einstein Comprehensive Cancer, Center, Bronx, NY, USA
| | - Alexandros Hardas
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, North Mymms, Hatfield, United Kingdom
| | - Theofilos Poutahidis
- Laboratory of Pathology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Christos Panorias
- Division of Statistics and Operational Research, Department of Mathematics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Sophia DesMarais
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Conner Entenberg
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Randall S Carpenter
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Hillary Guzik
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY, USA
- Montefiore-Einstein Comprehensive Cancer, Center, Bronx, NY, USA
| | - Xheni Nishku
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY, USA
- Montefiore-Einstein Comprehensive Cancer, Center, Bronx, NY, USA
| | - Joseph Churaman
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY, USA
- Montefiore-Einstein Comprehensive Cancer, Center, Bronx, NY, USA
| | - Maria Maryanovich
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Cancer Dormancy and Tumor Microenvironment Institute, Montefiore-Einstein Comprehensive Cancer, Center, Bronx, NY, USA
| | - Vera DesMarais
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Montefiore-Einstein Comprehensive Cancer, Center, Bronx, NY, USA
| | - Frank P Macaluso
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Montefiore-Einstein Comprehensive Cancer, Center, Bronx, NY, USA
| | - George S Karagiannis
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Tumor Microenvironment and Metastasis Program, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, USA
- Integrated Imaging Program for Cancer Research, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Cancer Dormancy and Tumor Microenvironment Institute, Montefiore-Einstein Comprehensive Cancer, Center, Bronx, NY, USA
| |
Collapse
|
14
|
Martins C, Magalhães S, Almeida I, Neto V, Rebelo S, Nunes A. Metabolomics to Study Human Aging: A Review. Curr Mol Med 2024; 24:457-477. [PMID: 37026499 DOI: 10.2174/1566524023666230407123727] [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: 11/07/2022] [Revised: 02/17/2023] [Accepted: 02/17/2023] [Indexed: 04/08/2023]
Abstract
In the last years, with the increase in the average life expectancy, the world's population is progressively aging, which entails social, health and economic problems. In this sense, the need to better understand the physiology of the aging process becomes an urgent need. Since the study of aging in humans is challenging, cellular and animal models are widely used as alternatives. Omics, namely metabolomics, have emerged in the study of aging, with the aim of biomarker discovering, which may help to uncomplicate this complex process. This paper aims to summarize different models used for aging studies with their advantages and limitations. Also, this review gathers the published articles referring to biomarkers of aging already discovered using metabolomics approaches, comparing the results obtained in the different studies. Finally, the most frequently used senescence biomarkers are described, along with their importance in understanding aging.
Collapse
Affiliation(s)
- Claudia Martins
- Department of Medical Sciences, iBiMED: Institute of Biomedicine, University of Aveiro, Agra do Crasto, Aveiro 3810-193, Portugal
| | - Sandra Magalhães
- Department of Surgery and Physiology, Faculty of Medicine, UnIC@RISE, Cardiovascular Research & Development Centre, University of Porto, Alameda Prof. Hernâni Monteiro, Porto 4200-319, Portugal
| | - Idália Almeida
- Department of Medical Sciences, iBiMED: Institute of Biomedicine, University of Aveiro, Agra do Crasto, Aveiro 3810-193, Portugal
- CICECO: Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro 3810-193, Portugal
| | - Vanessa Neto
- Department of Medical Sciences, iBiMED: Institute of Biomedicine, University of Aveiro, Agra do Crasto, Aveiro 3810-193, Portugal
| | - Sandra Rebelo
- Department of Medical Sciences, iBiMED: Institute of Biomedicine, University of Aveiro, Agra do Crasto, Aveiro 3810-193, Portugal
| | - Alexandra Nunes
- Department of Medical Sciences, iBiMED: Institute of Biomedicine, University of Aveiro, Agra do Crasto, Aveiro 3810-193, Portugal
| |
Collapse
|
15
|
Nguyen DK, Vanden-Bossche A, Laroche N, Thomas M, Linossier MT, Peyroche S, Farlay D, Follet H, Laquerrière P, Lafage-Proust MH, Thomas T, Vico L, Marotte H, Rousseau M. Dietary supplementation with nacre reduces cortical bone loss in aged female mice. Exp Gerontol 2023; 184:112337. [PMID: 38006949 DOI: 10.1016/j.exger.2023.112337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 08/17/2023] [Accepted: 11/22/2023] [Indexed: 11/27/2023]
Abstract
Aging is associated with detrimental bone loss leading to fragility fractures in both men and women. Notably, a majority of bone loss with aging is cortical, as well as a large number of fractures are non-vertebral and at the non-hip sites. Nacre is a product of mollusks composed of calcium carbonate embedded in organic components. As our previous study demonstrated the protective effect of nacre supplementation on trabecular bone loss in ovariectomized rats, we sought to evaluate the effect of dietary nacre on bone loss related to aging in female mice which do not suffer true menopause as observed in women. The current study compared the effect of a 90-day long nacre-supplemented diet to that of Standard or CaCO3 diets on both bone mass and strength in 16-month-old C57BL/6 female mice. Multiple approaches were performed to assess the microarchitecture and mechanical properties of long bones, analyze trabecular histomorphometry, and measure bone cell-related gene expressions, and bone turnover markers. In the cortex, dietary nacre improved cortical bone strength in line with lower expression levels of genes reflecting osteoclasts activity compared to Standard or CaCO3 diets (p < 0.05). In the trabeculae, nacre-fed mice were characterized by a bone remodeling process more active than the other groups as shown by greater histomorphometric parameters and osteoblast-related gene expressions (p < 0.05). But these differences were not exhibited at the level of the trabecular microarchitecture at this age. Collectively, these data suggest that dietary nacre should be a potential candidate for reducing aging-associated cortical bone loss in the elderly.
Collapse
Affiliation(s)
- Dung Kim Nguyen
- Université Jean Monnet Saint-Étienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023 Saint-Étienne, France.
| | - Arnaud Vanden-Bossche
- Université Jean Monnet Saint-Étienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023 Saint-Étienne, France
| | - Norbert Laroche
- Université Jean Monnet Saint-Étienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023 Saint-Étienne, France
| | - Mireille Thomas
- Université Jean Monnet Saint-Étienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023 Saint-Étienne, France
| | - Marie-Thérèse Linossier
- Université Jean Monnet Saint-Étienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023 Saint-Étienne, France
| | - Sylvie Peyroche
- Université Jean Monnet Saint-Étienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023 Saint-Étienne, France
| | - Delphine Farlay
- INSERM, LYOS UMR 1033, Université Claude Bernard Lyon 1, 69008 Lyon, France
| | - Hélène Follet
- INSERM, LYOS UMR 1033, Université Claude Bernard Lyon 1, 69008 Lyon, France
| | - Patrice Laquerrière
- Université de Strasbourg, Strasbourg, France; CNRS, Institut Pluridisciplinaire Hubert Curien UMR 7178, Strasbourg, France
| | - Marie-Hélène Lafage-Proust
- Université Jean Monnet Saint-Étienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023 Saint-Étienne, France; Université Jean Monnet Saint-Étienne, Department of Rheumatology, CHU Saint-Etienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023, F-42055 Saint-Étienne, France
| | - Thierry Thomas
- Université Jean Monnet Saint-Étienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023 Saint-Étienne, France; Université Jean Monnet Saint-Étienne, Department of Rheumatology, CHU Saint-Etienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023, F-42055 Saint-Étienne, France
| | - Laurence Vico
- Université Jean Monnet Saint-Étienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023 Saint-Étienne, France
| | - Hubert Marotte
- Université Jean Monnet Saint-Étienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023 Saint-Étienne, France; Université Jean Monnet Saint-Étienne, Department of Rheumatology, CHU Saint-Etienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023, F-42055 Saint-Étienne, France
| | - Marthe Rousseau
- Université Jean Monnet Saint-Étienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023 Saint-Étienne, France; UMR5510 MATEIS, CNRS, Lyon University, INSA-Lyon, Lyon, France
| |
Collapse
|
16
|
Gobbo F, Zingariello M, Verachi P, Falchi M, Arciprete F, Martelli F, Peli A, Mazzarini M, Vierstra J, Mead-Harvey C, Dueck AC, Sarli G, Nava S, Sgalla G, Richeldi L, Migliaccio AR. GATA1-defective immune-megakaryocytes as possible drivers of idiopathic pulmonary fibrosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.20.542249. [PMID: 37425686 PMCID: PMC10327123 DOI: 10.1101/2023.06.20.542249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic lung disorder with limited therapeutic options. Insufficient understanding of driver mutations and poor fidelity of currently available animal models has limited the development of effective therapies. Since GATA1 deficient megakaryocytes sustain myelofibrosis, we hypothesized that they may also induce fibrosis in lungs. We discovered that lungs from IPF patients and Gata1low mice contain numerous GATA1negative immune-poised megakaryocytes that, in mice, have defective RNA-seq profiling and increased TGF-β1, CXCL1 and P-selectin content. With age, Gata1low mice develop fibrosis in lungs. Development of lung fibrosis in this model is prevented by P-selectin deletion and rescued by P-selectin, TGF-β1 or CXCL1 inhibition. Mechanistically, P-selectin inhibition decreases TGF-β1 and CXCL1 content and increases GATA1positive megakaryocytes while TGF-β1 or CXCL1 inhibition decreased CXCL1 only. In conclusion, Gata1low mice are a novel genetic-driven model for IPF and provide a link between abnormal immune-megakaryocytes and lung fibrosis.
Collapse
Affiliation(s)
- Francesca Gobbo
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell’Emilia (Bologna) 40064, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Maria Zingariello
- Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, University Campus Bio-Medico, Rome 00128, Italy
| | - Paola Verachi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Mario Falchi
- National Center HIV/AIDS Research, Istituto Superiore di Sanita, Rome 00161, Italy
| | - Francesca Arciprete
- Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, University Campus Bio-Medico, Rome 00128, Italy
| | - Fabrizio Martelli
- National Center for Preclinical and Clinical Research and Evaluation of Pharmaceutical Drugs, Istituto Superiore di Sanita, Rome 00161, Italy
| | - Angelo Peli
- Department for Life Quality Studies, University of Bologna, Rimini Campus, Rimini 47921, Italy
| | - Maria Mazzarini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- Altius Institute for Biomedical Sciences, Seattle, WA 98121, USA
| | - Jeff Vierstra
- Altius Institute for Biomedical Sciences, Seattle, WA 98121, USA
| | - Carolyn Mead-Harvey
- Mayo Clinic, Department of Quantitative Health Sciences, Division of Clinical Trials and Biostatistics, Scottsdale, AZ 85259, USA
| | - Amylou C. Dueck
- Mayo Clinic, Department of Quantitative Health Sciences, Division of Clinical Trials and Biostatistics, Scottsdale, AZ 85259, USA
| | - Giuseppe Sarli
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell’Emilia (Bologna) 40064, Italy
| | - Stefano Nava
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Respiratory and Critical Care Unit, Bologna 40138, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna 40138, Italy
| | - Giacomo Sgalla
- Department of Medical and Surgical Sciences Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Universita Cattolica del Sacro Cuore, Roma 00168, Italy
| | - Luca Richeldi
- Department of Medical and Surgical Sciences Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Universita Cattolica del Sacro Cuore, Roma 00168, Italy
| | - Anna Rita Migliaccio
- Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, University Campus Bio-Medico, Rome 00128, Italy
- Altius Institute for Biomedical Sciences, Seattle, WA 98121, USA
| |
Collapse
|
17
|
Gifford AJ. A Primer for Research Scientists on Assessing Mouse Gross and Histopathology Images in the Biomedical Literature. Curr Protoc 2023; 3:e891. [PMID: 37712877 DOI: 10.1002/cpz1.891] [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: 09/16/2023]
Abstract
Advances in genomic technologies have enabled the development of abundant mouse models of human disease, requiring accurate phenotyping to elucidate the consequences of genetic manipulation. Anatomic pathology, an important component of the mouse phenotyping pipeline, is ideally performed by human or veterinary pathologists; however, due to insufficient numbers of pathologists qualified to assess these mouse models morphologically, research scientists may perform "do-it-yourself" pathology, resulting in diagnostic error. In the biomedical literature, pathology data is commonly presented as images of tissue sections, stained with either hematoxylin and eosin or antibodies via immunohistochemistry, accompanied by a figure legend. Data presented in such images and figure legends may contain inaccuracies. Furthermore, there is limited guidance for non-pathologist research scientists concerning the elements required in an ideal pathology image and figure legend in a research publication. In this overview, the components of an ideal pathology image and figure legend are outlined and comprise image quality, image composition, and image interpretation. Background knowledge is important for producing accurate pathology images and critically assessing these images in the literature. This foundational knowledge includes understanding relevant human and mouse anatomy and histology and, for cancer researchers, an understanding of human and mouse tumor classification and morphology, mouse stain background lesions, and tissue processing artifacts. Accurate interpretation of immunohistochemistry is also vitally important and is detailed with emphasis on the requirement for tissue controls and the distribution, intensity, and intracellular location of staining. Common pitfalls in immunohistochemistry interpretation are outlined, and a checklist of questions is provided by which any pathology image may be critically examined. Collaboration with pathologist colleagues is encouraged. This overview aims to equip researchers to critically assess the quality and accuracy of pathology images in the literature to improve the reliability and reproducibility of published pathology data. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Andrew J Gifford
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, New South Wales, Australia
- Anatomical Pathology, NSW Health Pathology, Prince of Wales Hospital, Randwick, New South Wales, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
18
|
Xie K, Ehninger D. Ageing-associated phenotypes in mice. Mech Ageing Dev 2023; 214:111852. [PMID: 37454704 DOI: 10.1016/j.mad.2023.111852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/22/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Ageing is a continuous process in life featuring progressive damage accumulation that leads to physiological decline, functional deterioration and ultimately death of an organism. Based on the relatively close anatomical and physiological similarity to humans, the mouse has been proven as a valuable model organism in ageing research over the last decades. In this review, we survey methods and tools currently in use to assess ageing phenotypes in mice. We summarize a range of ageing-associated alterations detectable at two major levels of analysis: (1) physiology and pathophysiology and (2) molecular biomarkers. Age-sensitive phenotypes provided in this article may serve to inform future studies targeting various aspects of organismal ageing in mice. In addition, we discuss conceptual and technical challenges faced by previous ageing studies in mice and, where possible, provide recommendations on how to resolve some of these issues.
Collapse
Affiliation(s)
- Kan Xie
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany
| | - Dan Ehninger
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127 Bonn, Germany.
| |
Collapse
|
19
|
Perez-Lanzon M, Carbonnier V, Cordier P, De Palma FDE, Petrazzuolo A, Klein C, Arbaretaz F, Mangane K, Stoll G, Martins I, Fohrer Ting H, Paillet J, Mouillet-Richard S, Le Corre D, Xiao W, Sroussi M, Desdouets C, Laurent-Puig P, Pol J, Lopez-Otin C, Maiuri MC, Kroemer G. New hormone receptor-positive breast cancer mouse cell line mimicking the immune microenvironment of anti-PD-1 resistant mammary carcinoma. J Immunother Cancer 2023; 11:e007117. [PMID: 37344100 PMCID: PMC10314679 DOI: 10.1136/jitc-2023-007117] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2023] [Indexed: 06/23/2023] Open
Abstract
BACKGROUND Progress in breast cancer (BC) research relies on the availability of suitable cell lines that can be implanted in immunocompetent laboratory mice. The best studied mouse strain, C57BL/6, is also the only one for which multiple genetic variants are available to facilitate the exploration of the cancer-immunity dialog. Driven by the fact that no hormone receptor-positive (HR+) C57BL/6-derived mammary carcinoma cell lines are available, we decided to establish such cell lines. METHODS BC was induced in female C57BL/6 mice using a synthetic progesterone analog (medroxyprogesterone acetate, MPA) combined with a DNA damaging agent (7,12-dimethylbenz[a]anthracene, DMBA). Cell lines were established from these tumors and selected for dual (estrogen+progesterone) receptor positivity, as well as transplantability into C57BL/6 immunocompetent females. RESULTS One cell line, which we called B6BC, fulfilled these criteria and allowed for the establishment of invasive estrogen receptor-positive (ER+) tumors with features of epithelial to mesenchymal transition that were abundantly infiltrated by myeloid immune populations but scarcely by T lymphocytes, as determined by single-nucleus RNA sequencing and high-dimensional leukocyte profiling. Such tumors failed to respond to programmed cell death-1 (PD-1) blockade, but reduced their growth on treatment with ER antagonists, as well as with anthracycline-based chemotherapy, which was not influenced by T-cell depletion. Moreover, B6BC-derived tumors reduced their growth on CD11b blockade, indicating tumor sustainment by myeloid cells. The immune environment and treatment responses recapitulated by B6BC-derived tumors diverged from those of ER+ TS/A cell-derived tumors in BALB/C mice, and of ER- E0771 cell-derived and MPA/DMBA-induced tumors in C57BL/6 mice. CONCLUSIONS B6BC is the first transplantable HR+ BC cell line derived from C57BL/6 mice and B6BC-derived tumors recapitulate the complex tumor microenvironment of locally advanced HR+ BC naturally resistant to PD-1 immunotherapy.
Collapse
Affiliation(s)
- Maria Perez-Lanzon
- Team "Metabolism, Cancer & Immunity", Centre de Recherche des Cordeliers, UMRS 1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
- Gustave Roussy Institute, Villejuif, France
| | - Vincent Carbonnier
- Team "Metabolism, Cancer & Immunity", Centre de Recherche des Cordeliers, UMRS 1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
- Gustave Roussy Institute, Villejuif, France
| | - Pierre Cordier
- Team 'Proliferation, Stress and Liver Physiopathology', Centre de Recherche des Cordeliers, Paris, France
| | - Fatima Domenica Elisa De Palma
- Team "Metabolism, Cancer & Immunity", Centre de Recherche des Cordeliers, UMRS 1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
- Gustave Roussy Institute, Villejuif, France
- Department of Molecular Medicine and Medical Biotechnologies, University of Napoli Federico II, Napoli, Italy
| | - Adriana Petrazzuolo
- Team "Metabolism, Cancer & Immunity", Centre de Recherche des Cordeliers, UMRS 1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
- Gustave Roussy Institute, Villejuif, France
| | - Christophe Klein
- Centre d'Histologie, d'Imagerie cellulaire et de Cytométrie (CHIC), Centre de Recherche des Cordeliers, Paris, France, UMRS1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
| | - Floriane Arbaretaz
- Centre d'Histologie, d'Imagerie cellulaire et de Cytométrie (CHIC), Centre de Recherche des Cordeliers, Paris, France, UMRS1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
| | - Khady Mangane
- Team "Metabolism, Cancer & Immunity", Centre de Recherche des Cordeliers, UMRS 1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
- Gustave Roussy Institute, Villejuif, France
| | - Gautier Stoll
- Team "Metabolism, Cancer & Immunity", Centre de Recherche des Cordeliers, UMRS 1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
- Gustave Roussy Institute, Villejuif, France
| | - Isabelle Martins
- Team "Metabolism, Cancer & Immunity", Centre de Recherche des Cordeliers, UMRS 1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
- Gustave Roussy Institute, Villejuif, France
| | - Helene Fohrer Ting
- Centre d'Histologie, d'Imagerie cellulaire et de Cytométrie (CHIC), Centre de Recherche des Cordeliers, Paris, France, UMRS1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
| | - Juliette Paillet
- Team "Metabolism, Cancer & Immunity", Centre de Recherche des Cordeliers, UMRS 1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
- Gustave Roussy Institute, Villejuif, France
| | - Sophie Mouillet-Richard
- Team 'Personalized medicine, pharmacogenomics, therapeutic optimization', Centre de Recherche des Cordeliers, Paris, France
| | - Delphine Le Corre
- Team 'Personalized medicine, pharmacogenomics, therapeutic optimization', Centre de Recherche des Cordeliers, Paris, France
| | - Wenjjin Xiao
- Team 'Personalized medicine, pharmacogenomics, therapeutic optimization', Centre de Recherche des Cordeliers, Paris, France
| | - Marine Sroussi
- Team 'Personalized medicine, pharmacogenomics, therapeutic optimization', Centre de Recherche des Cordeliers, Paris, France
| | - Chantal Desdouets
- Team 'Proliferation, Stress and Liver Physiopathology', Centre de Recherche des Cordeliers, Paris, France
| | - Pierre Laurent-Puig
- Team 'Personalized medicine, pharmacogenomics, therapeutic optimization', Centre de Recherche des Cordeliers, Paris, France
- Institut du Cancer Paris CARPEM, Institut Universitaire de France, Hôpital Européen Georges Pompidou, France-HP, Paris, France
| | - Jonathan Pol
- Team "Metabolism, Cancer & Immunity", Centre de Recherche des Cordeliers, UMRS 1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
- Gustave Roussy Institute, Villejuif, France
| | - Carlos Lopez-Otin
- Departamento de Bioquimica y Biologia Molecular, Instituto Universitario de Oncologia (IUOPA), University of Oviedo, Oviedo, Spain
| | - Maria Chiara Maiuri
- Team "Metabolism, Cancer & Immunity", Centre de Recherche des Cordeliers, UMRS 1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
- Gustave Roussy Institute, Villejuif, France
- Department of Molecular Medicine and Medical Biotechnologies, University of Napoli Federico II, Napoli, Italy
| | - Guido Kroemer
- Team "Metabolism, Cancer & Immunity", Centre de Recherche des Cordeliers, UMRS 1138, Inserm, Université Paris Cité, Sorbonne Université, Paris, France
- Gustave Roussy Institute, Villejuif, France
- Institut du Cancer Paris CARPEM, Institut Universitaire de France, Hôpital Européen Georges Pompidou, France-HP, Paris, France
| |
Collapse
|
20
|
Takasugi M, Yoshida Y, Hara E, Ohtani N. The role of cellular senescence and SASP in tumour microenvironment. FEBS J 2023; 290:1348-1361. [PMID: 35106956 DOI: 10.1111/febs.16381] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 12/04/2021] [Accepted: 01/31/2022] [Indexed: 01/01/2023]
Abstract
Cellular senescence refers to a state of irreversible cell cycle arrest that can be induced by various cellular stresses and is known to play a pivotal role in tumour suppression. While senescence-associated growth arrest can inhibit the proliferation of cancer-prone cells, the altered secretory profile of senescent cells, termed the senescence-associated secretory phenotype, can contribute to the microenvironment that promotes tumour development. Although the senescence-associated secretory phenotype and its effects on tumorigenesis are both highly context dependent, mechanisms underlying such diversity are becoming better understood, thereby allowing the creation of new strategies to effectively target the senescence-associated secretory phenotype and senescent cells for cancer therapy. In this review, we discuss the current knowledge on cellular senescence and the senescence-associated secretory phenotype to develop a structural understanding of their roles in the tumour microenvironment and provide perspectives for future research, including the possibility of senotherapy for the treatment of cancer.
Collapse
Affiliation(s)
- Masaki Takasugi
- Department of Pathophysiology, Graduate School of Medicine, Osaka City University, Japan
| | - Yuya Yoshida
- Department of Pathophysiology, Graduate School of Medicine, Osaka City University, Japan
| | - Eiji Hara
- Research Institute for Microbial Diseases, Osaka University, Japan.,Immunology Frontier Research Center (IFReC), Osaka University, Japan.,Center for Infectious Disease Education and Research (CiDER), Osaka University, Japan
| | - Naoko Ohtani
- Department of Pathophysiology, Graduate School of Medicine, Osaka City University, Japan.,AMED-CREST, AMED, Japan Agency for Medical Research and Development, Tokyo, Japan
| |
Collapse
|
21
|
Anton PE, Rutt LN, Capper C, Orlicky DJ, McCullough RL. Profiling the oxylipidome in aged mice after chronic ethanol feeding: Identifying lipid metabolites as drivers of hepatocyte stress. Alcohol 2023; 107:119-135. [PMID: 36150611 DOI: 10.1016/j.alcohol.2022.08.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/10/2022] [Accepted: 08/25/2022] [Indexed: 02/08/2023]
Abstract
The global population of people over the age of 65 is increasing and expected to reach 1.5 billion by 2050. While aging is associated with a number of chronic illnesses including dementia, the underlying contribution of alcohol misuse in the elderly is understudied. Long-term chronic alcohol misuse can lead to alcohol-associated liver disease, consisting of a spectrum of pathologies, including steatosis and cirrhosis; liver disease can be rapidly accelerated by non-resolving inflammation. Despite this knowledge, the mechanistic underpinnings of dysregulated host immunity and accelerated liver disease progression in the aged by alcohol is unknown. Alcohol misuse in the elderly is on the rise and aging is associated with progressive increases in pro-inflammatory cytokine production. The goals of the current study are to characterize bioactive lipid mediators of inflammation by making use of a murine model of ethanol-induced liver disease in 3-month-old and 20-month-old mice by quantitatively profiling selected oxylipins in liver, brain and plasma. Following chronic ethanol exposure, liver injury, steatosis, and senescence markers were robustly increased in aged mice compared to young adult mice. Expression of proinflammatory cytokines and lipid metabolizing enzymes were increased in liver by both age and ethanol feeding. Lipoxygenase-derived lipid metabolites 9- and 13-hydroxy-octadecadienoic acid and 15-hydroxyeicosatetraenoic acid were increased in liver and plasma in ethanol-fed aged mice and positively correlated with liver injury. In plasma, 9,10-dihydroxy-octadecenoic acid/epoxy-octadecenoic acid plasma ratios correlated with liver injury in ethanol-fed aged mice. Finally, 15-hydroxyeicosatetraenoic acid and 9,10-dihydroxy-octadecenoic acid positively correlated between liver and plasma. Importantly, leukotriene E4, 9,10-dihydroxy-octadecenoic acid and 15-hydroxyeicosatetraenoic acid increased lipid accumulation and ER stress in cultured AML12 hepatocytes. These data highlight the complexity of lipid metabolite networks but identify key mediators that may be used for diagnostic and prognostic markers in early stages of alcohol-related liver disease in patients of all ages.
Collapse
Affiliation(s)
- Paige E Anton
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Lauren N Rutt
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Courtney Capper
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - David J Orlicky
- Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Rebecca L McCullough
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States; Colorado University Alcohol Research Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States; GI and Liver Innate Immune Program, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.
| |
Collapse
|
22
|
Mazzarini M, Arciprete F, Picconi O, Valeri M, Verachi P, Martelli F, Migliaccio AR, Falchi M, Zingariello M. Single cell analysis of the localization of the hematopoietic stem cells within the bone marrow architecture identifies niche-specific proliferation dynamics. Front Med (Lausanne) 2023; 10:1166758. [PMID: 37188088 PMCID: PMC10175646 DOI: 10.3389/fmed.2023.1166758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Introduction Hematopoietic stem cells (HSC) reside in the bone marrow (BM) in specialized niches which provide support for their self-replication and differentiation into the blood cells. Recently, numerous studies using sophisticated molecular and microscopic technology have provided snap-shots information on the identity of the BM niches in mice. In adults, HSC are localized around arterioles and sinusoids/venules whereas in juvenile mice they are in close to the osteoblasts. However, although it is well recognized that in mice the nature of the hematopoietic niche change with age or after exposure to inflammatory insults, much work remains to be done to identify changes occurring under these conditions. The dynamic changes occurring in niche/HSC interactions as HSC enter into cycle are also poorly defined. Methods We exploit mice harboring the hCD34tTA/Tet-O-H2BGFP transgene to establish the feasibility to assess interactions of the HSC with their niche as they cycle. In this model, H2BGFP expression is driven by the TET trans-activator under the control of the human CD34 promoter which in mice is active only in the HSC. Since Doxycycline inhibits TET, HSC exposed to this drug no longer express H2BGFP and loose half of their label every division allowing establishing the dynamics of their first 1-3 divisions. To this aim, we first validated user-friendly confocal microscopy methods to determine HSC divisions by hemi-decrement changes in levels of GFP expression. We then tracked the interaction occurring in old mice between the HSC and their niche during the first HSC divisions. Results We determined that in old mice, most of the HSC are located around vessels, both arterioles which sustain quiescence and self-replication, and venules/sinusoids, which sustain differentiation. After just 1 week of exposure to Doxycycline, great numbers of the HSC around the venules lost most of their GFP label, indicating that they had cycled. By contrast, the few HSC surrounding the arterioles retained maximal levels of GFP expression, indicating that they are either dormant or cycle at very low rates. Conclusion These results reveal that in old mice, HSC cycle very dynamically and are biased toward interactions with the niche that instructs them to differentiate.
Collapse
Affiliation(s)
- Maria Mazzarini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- Altius Institute for Biomedical Sciences, Seattle, WA, United States
| | - Francesca Arciprete
- Unit of Microscopic and Ultrastructural Anatomy, University Campus Bio-Medico, Rome, Italy
| | - Orietta Picconi
- National Center for HIV/AIDS Research, Istituto Superiore di Sanità, Rome, Italy
| | - Mauro Valeri
- Center for Animal Experimentation and Well-Being, Istituto Superiore di Sanità, Rome, Italy
| | - Paola Verachi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Fabrizio Martelli
- National Center for Preclinical and Clinical Research and Evaluation of Pharmaceutical Drugs, Istituto Superiore di Sanità, Rome, Italy
| | - Anna Rita Migliaccio
- Altius Institute for Biomedical Sciences, Seattle, WA, United States
- Unit of Microscopic and Ultrastructural Anatomy, University Campus Bio-Medico, Rome, Italy
- *Correspondence: Anna Rita Migliaccio ;
| | - Mario Falchi
- National Center for HIV/AIDS Research, Istituto Superiore di Sanità, Rome, Italy
| | - Maria Zingariello
- Altius Institute for Biomedical Sciences, Seattle, WA, United States
| |
Collapse
|
23
|
Xie K, Fuchs H, Scifo E, Liu D, Aziz A, Aguilar-Pimentel JA, Amarie OV, Becker L, da Silva-Buttkus P, Calzada-Wack J, Cho YL, Deng Y, Edwards AC, Garrett L, Georgopoulou C, Gerlini R, Hölter SM, Klein-Rodewald T, Kramer M, Leuchtenberger S, Lountzi D, Mayer-Kuckuk P, Nover LL, Oestereicher MA, Overkott C, Pearson BL, Rathkolb B, Rozman J, Russ J, Schaaf K, Spielmann N, Sanz-Moreno A, Stoeger C, Treise I, Bano D, Busch DH, Graw J, Klingenspor M, Klopstock T, Mock BA, Salomoni P, Schmidt-Weber C, Weiergräber M, Wolf E, Wurst W, Gailus-Durner V, Breteler MMB, Hrabě de Angelis M, Ehninger D. Deep phenotyping and lifetime trajectories reveal limited effects of longevity regulators on the aging process in C57BL/6J mice. Nat Commun 2022; 13:6830. [PMID: 36369285 PMCID: PMC9652467 DOI: 10.1038/s41467-022-34515-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 10/27/2022] [Indexed: 11/13/2022] Open
Abstract
Current concepts regarding the biology of aging are primarily based on studies aimed at identifying factors regulating lifespan. However, lifespan as a sole proxy measure for aging can be of limited value because it may be restricted by specific pathologies. Here, we employ large-scale phenotyping to analyze hundreds of markers in aging male C57BL/6J mice. For each phenotype, we establish lifetime profiles to determine when age-dependent change is first detectable relative to the young adult baseline. We examine key lifespan regulators (putative anti-aging interventions; PAAIs) for a possible countering of aging. Importantly, unlike most previous studies, we include in our study design young treated groups of animals, subjected to PAAIs prior to the onset of detectable age-dependent phenotypic change. Many PAAI effects influence phenotypes long before the onset of detectable age-dependent change, but, importantly, do not alter the rate of phenotypic change. Hence, these PAAIs have limited effects on aging.
Collapse
Affiliation(s)
- Kan Xie
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Helmut Fuchs
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Enzo Scifo
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Dan Liu
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Ahmad Aziz
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany.,Department of Neurology, Faculty of Medicine, University of Bonn, Bonn, Germany
| | - Juan Antonio Aguilar-Pimentel
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Oana Veronica Amarie
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Lore Becker
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Patricia da Silva-Buttkus
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Julia Calzada-Wack
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Yi-Li Cho
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Yushuang Deng
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - A Cole Edwards
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Lillian Garrett
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany.,Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Christina Georgopoulou
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Raffaele Gerlini
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Sabine M Hölter
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany.,Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Tanja Klein-Rodewald
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | | | - Stefanie Leuchtenberger
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Dimitra Lountzi
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Phillip Mayer-Kuckuk
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Lena L Nover
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Manuela A Oestereicher
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Clemens Overkott
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Brandon L Pearson
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany.,Mailman School of Public Health, Columbia University, 630W. 168th St., New York, NY, 10032, USA
| | - Birgit Rathkolb
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany.,Member of German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany.,Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jan Rozman
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany.,Member of German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany.,Institute of Molecular Genetics of the Czech Academy of Sciences, Czech Centre for Phenogenomics, Prumyslova 595, Vestec, 252 50, Czech Republic
| | - Jenny Russ
- Nuclear Function Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Kristina Schaaf
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Nadine Spielmann
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Adrián Sanz-Moreno
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Claudia Stoeger
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Irina Treise
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Daniele Bano
- Aging and Neurodegeneration Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Dirk H Busch
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München, 81675, Munich, Germany
| | - Jochen Graw
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Martin Klingenspor
- Molecular Nutritional Medicine, Else Kröner-Fresenius Center, Technische Universität München, 85350, Freising-Weihenstephan, Germany
| | - Thomas Klopstock
- Friedrich-Baur-Institut, Department of Neurology, Ludwig-Maximilians-University Munich, 80336, Munich, Germany.,DZNE, German Center for Neurodegenerative Diseases, 80336, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), 80336, Munich, Germany
| | - Beverly A Mock
- Laboratory of Cancer Biology and Genetics, CCR, NCI, NIH, Bethesda, MD, 20892, USA
| | - Paolo Salomoni
- Nuclear Function Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany
| | - Carsten Schmidt-Weber
- Center of Allergy & Environment (ZAUM), Technische Universität München, and Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Marco Weiergräber
- Research Group Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices, 53175, Bonn, Germany
| | - Eckhard Wolf
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany.,DZNE, German Center for Neurodegenerative Diseases, 80336, Munich, Germany.,Chair of Developmental Genetics, TUM School of Life Sciences (SoLS), Technische Universität München, Freising, Germany
| | - Valérie Gailus-Durner
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany
| | - Monique M B Breteler
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany.,Institute for Medical Biometry, Informatics and Epidemiology, Faculty of Medicine, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Martin Hrabě de Angelis
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764, Neuherberg, Germany.,Member of German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany.,Chair of Experimental Genetics, TUM School of Life Sciences (SoLS), Technische Universität München, 85354, Freising, Germany
| | - Dan Ehninger
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1/99, 53127, Bonn, Germany.
| |
Collapse
|
24
|
Ward JM, Vogel P, Sundberg JP. Brain and spinal cord lesions in 28 inbred strains of aging mice. Vet Pathol 2022; 59:1047-1055. [PMID: 36062914 DOI: 10.1177/03009858221120009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Brain and spinal cord histopathology findings in male and female 20-month-old mice in a large-scale aging study of 28 inbred Jackson Laboratory mouse strains from 7 genetic families are described. Brain sections from selected strains at 12 and 24 months of age or older were also reviewed. Common lesions include axonal dystrophy in the gracile and/or cuneate nucleus in the sensory tract of the dorsal medulla and in the spinal cord in all strains. Hirano-like bodies were seen in 24/28 strains, and mineralization was observed in the thalamus of 9/28 strains. Less common lesions were also seen in the cerebellum, cerebral cortex, and other brain areas. No brain or spinal cord tumors were found. Evidence of an impairment of the ubiquitin-proteasome system (UPS) and/or suspected autophagy was manifested as medullary axonal dystrophy with intra-axonal granular eosinophilic bodies and LC3B immunohistochemistry in most strains. RIIIS/J, the most severely affected strain, showed moderate axonal dystrophy at 12 months, which progressed to severe lesions at 20 months. Comparative pathology in various species is discussed.
Collapse
Affiliation(s)
- Jerrold M Ward
- The Jackson Laboratory, Bar Harbor, ME.,Global Vet Pathology, Montgomery Village, MD
| | - Peter Vogel
- St. Jude Children's Research Hospital, Memphis, TN
| | | |
Collapse
|
25
|
O'Leary TP, Brown RE. Visuo-spatial learning and memory impairments in the 5xFAD mouse model of Alzheimer's disease: Effects of age, sex, albinism, and motor impairments. GENES, BRAIN, AND BEHAVIOR 2022; 21:e12794. [PMID: 35238473 PMCID: PMC9744519 DOI: 10.1111/gbb.12794] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 12/19/2021] [Indexed: 12/17/2022]
Abstract
The 5xFAD mouse model of Alzheimer's disease (AD) rapidly develops AD-related neuro-behavioral pathology. Learning and memory impairments in 5xFAD mice, however, are not always replicated and the size of impairments varies considerably across studies. To examine possible sources of this variability, we analyzed the effects of age, sex, albinism due to background genes (Tyrc , Oca2p ) and motor impairment on learning and memory performance of wild type and 5xFAD mice on the Morris water maze, from 3 to 15 months of age. The 5xFAD mice showed impaired learning at 6-9 months of age, but memory impairments were not detected with the test procedure used in this study. Performance of 5xFAD mice was profoundly impaired at 12-15 months of age, but was accompanied by slower swim speeds than wild-type mice and a frequent failure to locate the escape platform. Overall female mice performed worse than males, and reversal learning impairments in 5xFAD mice were more pronounced in females than males. Albino mice performed worse than pigmented mice, confirming that albinism can impair performance of 5xFAD mice independently of AD-related transgenes. Overall, these results show that 5xFAD mice have impaired learning performance at 6-9 months of age, but learning and memory performance at 12-15 months is confounded with motor impairments. Furthermore, sex and albinism should be controlled to provide an accurate assessment of AD-related transgenes on learning and memory. These results will help reduce variability across pre-clinical experiments with 5xFAD mice, and thus enhance the reliability of studies developing new therapeutics for AD.
Collapse
Affiliation(s)
- Timothy P. O'Leary
- Department of Psychology and NeuroscienceDalhousie UniversityHalifaxNova ScotiaCanada
| | - Richard E. Brown
- Department of Psychology and NeuroscienceDalhousie UniversityHalifaxNova ScotiaCanada
| |
Collapse
|
26
|
Goedeke L, Murt KN, Di Francesco A, Camporez JP, Nasiri AR, Wang Y, Zhang X, Cline GW, de Cabo R, Shulman GI. Sex- and strain-specific effects of mitochondrial uncoupling on age-related metabolic diseases in high-fat diet-fed mice. Aging Cell 2022; 21:e13539. [PMID: 35088525 PMCID: PMC8844126 DOI: 10.1111/acel.13539] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/17/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022] Open
Abstract
Mild uncoupling of oxidative phosphorylation is an intrinsic property of all mitochondria and may have evolved to protect cells against the production of damaging reactive oxygen species. Therefore, compounds that enhance mitochondrial uncoupling are potentially attractive anti‐aging therapies; however, chronic ingestion is associated with a number of unwanted side effects. We have previously developed a controlled‐release mitochondrial protonophore (CRMP) that is functionally liver‐directed and promotes oxidation of hepatic triglycerides by causing a subtle sustained increase in hepatic mitochondrial inefficiency. Here, we sought to leverage the higher therapeutic index of CRMP to test whether mild mitochondrial uncoupling in a liver‐directed fashion could reduce oxidative damage and improve age‐related metabolic disease and lifespan in diet‐induced obese mice. Oral administration of CRMP (20 mg/[kg‐day] × 4 weeks) reduced hepatic lipid content, protein kinase C epsilon activation, and hepatic insulin resistance in aged (74‐week‐old) high‐fat diet (HFD)‐fed C57BL/6J male mice, independently of changes in body weight, whole‐body energy expenditure, food intake, or markers of hepatic mitochondrial biogenesis. CRMP treatment was also associated with a significant reduction in hepatic lipid peroxidation, protein carbonylation, and inflammation. Importantly, long‐term (49 weeks) hepatic mitochondrial uncoupling initiated late in life (94–104 weeks), in conjugation with HFD feeding, protected mice against neoplastic disorders, including hepatocellular carcinoma (HCC), in a strain and sex‐specific manner. Taken together, these studies illustrate the complex variation of aging and provide important proof‐of‐concept data to support further studies investigating the use of liver‐directed mitochondrial uncouplers to promote healthy aging in humans.
Collapse
Affiliation(s)
- Leigh Goedeke
- Department of Internal Medicine Yale School of Medicine New Haven Connecticut USA
| | - Kelsey N. Murt
- Translational Gerontology Branch Intramural Research Program National Institute on Aging, NIH Baltimore Maryland USA
| | - Andrea Di Francesco
- Translational Gerontology Branch Intramural Research Program National Institute on Aging, NIH Baltimore Maryland USA
| | - João Paulo Camporez
- Department of Internal Medicine Yale School of Medicine New Haven Connecticut USA
- Department of Physiology Ribeirao Preto School of Medicine University of Sao Paulo São Paulo Brazil
| | - Ali R. Nasiri
- Department of Internal Medicine Yale School of Medicine New Haven Connecticut USA
| | - Yongliang Wang
- Department of Internal Medicine Yale School of Medicine New Haven Connecticut USA
| | - Xian‐Man Zhang
- Department of Internal Medicine Yale School of Medicine New Haven Connecticut USA
| | - Gary W. Cline
- Department of Internal Medicine Yale School of Medicine New Haven Connecticut USA
| | - Rafael de Cabo
- Translational Gerontology Branch Intramural Research Program National Institute on Aging, NIH Baltimore Maryland USA
| | - Gerald I. Shulman
- Department of Internal Medicine Yale School of Medicine New Haven Connecticut USA
- Department of Cellular and Molecular Physiology Yale School of Medicine New Haven Connecticut USA
| |
Collapse
|
27
|
Zingariello M, Verachi P, Gobbo F, Martelli F, Falchi M, Mazzarini M, Valeri M, Sarli G, Marinaccio C, Melo-Cardenas J, Crispino JD, Migliaccio AR. Resident Self-Tissue of Proinflammatory Cytokines Rather than Their Systemic Levels Correlates with Development of Myelofibrosis in Gata1low Mice. Biomolecules 2022; 12:biom12020234. [PMID: 35204735 PMCID: PMC8961549 DOI: 10.3390/biom12020234] [Citation(s) in RCA: 5] [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: 01/10/2022] [Revised: 01/27/2022] [Accepted: 01/27/2022] [Indexed: 02/05/2023] Open
Abstract
Serum levels of inflammatory cytokines are currently investigated as prognosis markers in myelofibrosis, the most severe Philadelphia-negative myeloproliferative neoplasm. We tested this hypothesis in the Gata1low model of myelofibrosis. Gata1low mice, and age-matched wild-type littermates, were analyzed before and after disease onset. We assessed cytokine serum levels by Luminex-bead-assay and ELISA, frequency and cytokine content of stromal cells by flow cytometry, and immunohistochemistry and bone marrow (BM) localization of GFP-tagged hematopoietic stem cells (HSC) by confocal microscopy. Differences in serum levels of 32 inflammatory-cytokines between prefibrotic and fibrotic Gata1low mice and their wild-type littermates were modest. However, BM from fibrotic Gata1low mice contained higher levels of lipocalin-2, CXCL1, and TGF-β1 than wild-type BM. Although frequencies of endothelial cells, mesenchymal cells, osteoblasts, and megakaryocytes were higher than normal in Gata1low BM, the cells which expressed these cytokines the most were malignant megakaryocytes. This increased bioavailability of proinflammatory cytokines was associated with altered HSC localization: Gata1low HSC were localized in the femur diaphysis in areas surrounded by microvessels, neo-bones, and megakaryocytes, while wild-type HSC were localized in the femur epiphysis around adipocytes. In conclusion, bioavailability of inflammatory cytokines in BM, rather than blood levels, possibly by reshaping the HSC niche, correlates with myelofibrosis in Gata1low mice.
Collapse
Affiliation(s)
| | - Paola Verachi
- Department of Biomedical and Neuromotorial Sciences, Alma Mater University, 40126 Bologna, Italy; (P.V.); (F.G.); (M.M.)
| | - Francesca Gobbo
- Department of Biomedical and Neuromotorial Sciences, Alma Mater University, 40126 Bologna, Italy; (P.V.); (F.G.); (M.M.)
- Department of Veterinary Medical Sciences, University of Bologna, 40126 Bologna, Italy;
| | - Fabrizio Martelli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Mario Falchi
- National Center HIV/AIDS Research, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Maria Mazzarini
- Department of Biomedical and Neuromotorial Sciences, Alma Mater University, 40126 Bologna, Italy; (P.V.); (F.G.); (M.M.)
| | - Mauro Valeri
- Center for Animal Experimentation and Well-Being, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Giuseppe Sarli
- Department of Veterinary Medical Sciences, University of Bologna, 40126 Bologna, Italy;
| | | | - Johanna Melo-Cardenas
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (J.M.-C.); (J.D.C.)
| | - John D. Crispino
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (J.M.-C.); (J.D.C.)
| | - Anna Rita Migliaccio
- Altius Institute for Biomedical Sciences, Seattle, WA 98121, USA
- Center for Integrated Biomedical Research, Campus Bio-Medico, 00128 Rome, Italy
- Correspondence:
| |
Collapse
|
28
|
Causal Biological Network Model for Inflammasome Signaling Applied for Interpreting Transcriptomic Changes in Various Inflammatory States. Int J Inflam 2022; 2022:4071472. [PMID: 35126992 PMCID: PMC8813300 DOI: 10.1155/2022/4071472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/27/2021] [Indexed: 11/17/2022] Open
Abstract
Virtually any stressor that alters the cellular homeostatic state may result in an inflammatory response. As a critical component of innate immunity, inflammasomes play a prominent role in the inflammatory response. The information on inflammasome biology is rapidly growing, thus creating the need for structuring it into a model that can help visualize and enhance the understanding of underlying biological processes. Causal biological network (CBN) models provide predictive power for novel disease mechanisms and treatment outcomes. We assembled the available literature information on inflammasome activation into the CBN model and scored it with publicly available transcriptomic datasets that address viral infection of the lungs, osteo- and rheumatoid arthritis, psoriasis, and aging. The scoring inferred pathway activation leading to NLRP3 inflammasome activation in these diverse conditions, demonstrating that the CBN model provides a platform for interpreting transcriptomic data in the context of inflammasome activation.
Collapse
|
29
|
Cooper TK, Meyerholz DK, Beck AP, Delaney MA, Piersigilli A, Southard TL, Brayton CF. Research-Relevant Conditions and Pathology of Laboratory Mice, Rats, Gerbils, Guinea Pigs, Hamsters, Naked Mole Rats, and Rabbits. ILAR J 2022; 62:77-132. [PMID: 34979559 DOI: 10.1093/ilar/ilab022] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/12/2021] [Indexed: 12/31/2022] Open
Abstract
Animals are valuable resources in biomedical research in investigations of biological processes, disease pathogenesis, therapeutic interventions, safety, toxicity, and carcinogenicity. Interpretation of data from animals requires knowledge not only of the processes or diseases (pathophysiology) under study but also recognition of spontaneous conditions and background lesions (pathology) that can influence or confound the study results. Species, strain/stock, sex, age, anatomy, physiology, spontaneous diseases (noninfectious and infectious), and neoplasia impact experimental results and interpretation as well as animal welfare. This review and the references selected aim to provide a pathology resource for researchers, pathologists, and veterinary personnel who strive to achieve research rigor and validity and must understand the spectrum of "normal" and expected conditions to accurately identify research-relevant experimental phenotypes as well as unusual illness, pathology, or other conditions that can compromise studies involving laboratory mice, rats, gerbils, guinea pigs, hamsters, naked mole rats, and rabbits.
Collapse
Affiliation(s)
- Timothy K Cooper
- Department of Comparative Medicine, Penn State Hershey Medical Center, Hershey, PA, USA
| | - David K Meyerholz
- Department of Pathology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa, USA
| | - Amanda P Beck
- Department of Pathology, Yeshiva University Albert Einstein College of Medicine, Bronx, New York, USA
| | - Martha A Delaney
- Zoological Pathology Program, University of Illinois at Urbana-Champaign College of Veterinary Medicine, Urbana-Champaign, Illinois, USA
| | - Alessandra Piersigilli
- Laboratory of Comparative Pathology and the Genetically Modified Animal Phenotyping Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Teresa L Southard
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, New York, USA
| | - Cory F Brayton
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
30
|
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
|
31
|
FTIR Spectroscopy as a Tool to Study Age-Related Changes in Cardiac and Skeletal Muscle of Female C57BL/6J Mice. Molecules 2021; 26:molecules26216410. [PMID: 34770818 PMCID: PMC8587752 DOI: 10.3390/molecules26216410] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 11/30/2022] Open
Abstract
Studying aging is important to further understand the molecular mechanisms underlying this physiological process and, ideally, to identify a panel of aging biomarkers. Animals, in particular mice, are often used in aging studies, since they mimic important features of human aging, age quickly, and are easy to manipulate. The present work describes the use of Fourier Transform Infrared (FTIR) spectroscopy to identify an age-related spectroscopic profile of the cardiac and skeletal muscle tissues of C57BL/6J female mice. We acquired ATR-FTIR spectra of cardiac and skeletal muscle at four different ages: 6; 12; 17 and 24 months (10 samples at each age) and analyzed the data using multivariate statistical tools (PCA and PLS) and peak intensity analyses. The results suggest deep changes in protein secondary structure in 24-month-old mice compared to both tissues in 6-month-old mice. Oligomeric structures decreased with age in both tissues, while intermolecular β-sheet structures increased with aging in cardiac muscle but not in skeletal muscle. Despite FTIR spectroscopy being unable to identify the proteins responsible for these conformational changes, this study gives insights into the potential of FTIR to monitor the aging process and identify an age-specific spectroscopic signature.
Collapse
|
32
|
Yamashita Y, Sato T, Noishiki K, Kobayashi K, Uchiyama A, Izumi H, Tamura T, Shikamura M, Oinishi Y, Saito M, Kawamata S. Data on long-term survival of the NOD/Shi-scid IL-2Rγ null (NOG) mouse in two facilities. J Toxicol Sci 2021; 46:453-469. [PMID: 34602530 DOI: 10.2131/jts.46.453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The objective of this study was to investigate an appropriate observation period for an evaluation of tumorigenicity in NOD/Shi-scid IL-2 Rγnull (NOG) mice. At SNBL, 19 male and 19 female NOG mice were observed the general condition from 7 weeks old up to 68 weeks old and at FBRI, 7 male and 16 female NOG mice were observed the general condition throughout the lifespan from 7 weeks old. The survival rate started to decline rapidly around 54 to 56 weeks of age in both facilities without a facility difference. Based on these survival data, it seems reasonable to terminate a tumorigenicity study at 52 weeks of age.
Collapse
Affiliation(s)
- Yuji Yamashita
- Shin Nippon Biomedical Laboratories, Ltd. (SNBL), Drug Safety Research Laboratories
| | - Tamio Sato
- Shin Nippon Biomedical Laboratories, Ltd. (SNBL), Drug Safety Research Laboratories
| | - Kaori Noishiki
- Shin Nippon Biomedical Laboratories, Ltd. (SNBL), Drug Safety Research Laboratories
| | - Kinji Kobayashi
- Shin Nippon Biomedical Laboratories, Ltd. (SNBL), Drug Safety Research Laboratories
| | - Asako Uchiyama
- Shin Nippon Biomedical Laboratories, Ltd. (SNBL), Drug Safety Research Laboratories
| | - Hiroyuki Izumi
- Shin Nippon Biomedical Laboratories, Ltd. (SNBL), Drug Safety Research Laboratories
| | - Takashi Tamura
- Research & Development Center for Cell Therapy, Foundation for Biomedical Research and Innovation at Kobe (FBRI)
| | - Masayuki Shikamura
- Research & Development Center for Cell Therapy, Foundation for Biomedical Research and Innovation at Kobe (FBRI)
| | - Yu Oinishi
- Research & Development Center for Cell Therapy, Foundation for Biomedical Research and Innovation at Kobe (FBRI)
| | - Miho Saito
- Research & Development Center for Cell Therapy, Foundation for Biomedical Research and Innovation at Kobe (FBRI)
| | - Shin Kawamata
- Research & Development Center for Cell Therapy, Foundation for Biomedical Research and Innovation at Kobe (FBRI)
| |
Collapse
|
33
|
Saha J, Bae J, Wang SY, Lu H, Chappell LJ, Gopal P, Davis AJ. Ablating putative Ku70 phosphorylation sites results in defective DNA damage repair and spontaneous induction of hepatocellular carcinoma. Nucleic Acids Res 2021; 49:9836-9850. [PMID: 34428289 PMCID: PMC8464062 DOI: 10.1093/nar/gkab743] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 12/31/2022] Open
Abstract
Multiple pathways mediate the repair of DNA double-strand breaks (DSBs), with numerous mechanisms responsible for driving choice between the pathways. Previously, we reported that mutating five putative phosphorylation sites on the non-homologous end joining (NHEJ) factor, Ku70, results in sustained retention of human Ku70/80 at DSB ends and attenuation of DSB repair via homologous recombination (HR). In this study, we generated a knock-in mouse, in which the three conserved putative phosphorylation sites of Ku70 were mutated to alanine to ablate potential phosphorylation (Ku703A/3A), in order to examine if disrupting DSB repair pathway choice by modulating Ku70/80 dynamics at DSB ends results in enhanced genomic instability and tumorigenesis. The Ku703A/3A mice developed spontaneous and have accelerated chemical-induced hepatocellular carcinoma (HCC) compared to wild-type (Ku70+/+) littermates. The HCC tumors from the Ku703A/3A mice have increased γH2AX and 8-oxo-G staining, suggesting decreased DNA repair. Spontaneous transformed cell lines from Ku703A/3A mice are more radiosensitive, have a significant decrease in DNA end resection, and are more sensitive to the DNA cross-linking agent mitomycin C compared to cells from Ku70+/+ littermates. Collectively, these findings demonstrate that mutating the putative Ku70 phosphorylation sites results in defective DNA damage repair and disruption of this process drives genomic instability and accelerated development of HCC.
Collapse
Affiliation(s)
- Janapriya Saha
- Division of Molecular Radiation Biology, Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jinsung Bae
- Division of Molecular Radiation Biology, Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Shih-Ya Wang
- Division of Molecular Radiation Biology, Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Huiming Lu
- Division of Molecular Radiation Biology, Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX, USA
| | | | - Purva Gopal
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Anthony J Davis
- Division of Molecular Radiation Biology, Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
34
|
Wong ET, Luettich K, Krishnan S, Wong SK, Lim WT, Yeo D, Büttner A, Leroy P, Vuillaume G, Boué S, Hoeng J, Vanscheeuwijck P, Peitsch MC. Reduced Chronic Toxicity and Carcinogenicity in A/J Mice in Response to Life-Time Exposure to Aerosol From a Heated Tobacco Product Compared With Cigarette Smoke. Toxicol Sci 2021; 178:44-70. [PMID: 32780830 PMCID: PMC7657344 DOI: 10.1093/toxsci/kfaa131] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We conducted an inhalation study, in accordance with Organisation for Economic Co-operation and Development Test Guideline 453, exposing A/J mice to tobacco heating system (THS) 2.2 aerosol or 3R4F reference cigarette smoke (CS) for up to 18 months to evaluate chronic toxicity and carcinogenicity. All exposed mice showed lower thymus and spleen weight, blood lymphocyte counts, and serum lipid concentrations than sham mice, most likely because of stress and/or nicotine effects. Unlike THS 2.2 aerosol-exposed mice, CS-exposed mice showed increased heart weight, changes in red blood cell profiles and serum liver function parameters. Similarly, increased pulmonary inflammation, altered lung function, and emphysematous changes were observed only in CS-exposed mice. Histopathological changes in other respiratory tract organs were significantly lower in the THS 2.2 aerosol-exposed groups than in the CS-exposed group. Chronic exposure to THS 2.2 aerosol also did not increase the incidence or multiplicity of bronchioloalveolar adenomas or carcinomas relative to sham, whereas CS exposure did. Male THS 2.2 aerosol-exposed mice had a lower survival rate than sham mice, related to an increased incidence of urogenital issues that appears to be related to congenital factors rather than test item exposure. The lower impact of THS 2.2 aerosol exposure on tumor development and chronic toxicity is consistent with the significantly reduced levels of harmful and potentially harmful constituents in THS 2.2 aerosol relative to CS. The totality of the evidence from this study further supports the risk reduction potential of THS 2.2 for lung diseases in comparison with cigarettes.
Collapse
Affiliation(s)
- Ee Tsin Wong
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Karsta Luettich
- Department of Life Sciences, Systems Toxicology, PMI R&D, Philip Morris Products S.A, CH-2000 Neuchâtel, Switzerland
| | - Subash Krishnan
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Sin Kei Wong
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Wei Ting Lim
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Demetrius Yeo
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | | | - Patrice Leroy
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Grégory Vuillaume
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Stéphanie Boué
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Julia Hoeng
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Patrick Vanscheeuwijck
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| | - Manuel C Peitsch
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd, Science Park II, Singapore 117406, Singapore
| |
Collapse
|
35
|
Wu KC, Cain G, Corpuz J, Xu D, Ljumanovic N, Zarrin AA. Tpl2 kinase regulates inflammation but not tumorigenesis in mice. Toxicol Appl Pharmacol 2021; 418:115494. [PMID: 33722668 DOI: 10.1016/j.taap.2021.115494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 01/30/2021] [Accepted: 03/09/2021] [Indexed: 11/29/2022]
Abstract
Tumor progression locus 2 (Tpl2, gene name MAP3K8), a mitogen-activated protein kinase, is widely expressed in immune and non-immune cells to integrate tumor necrosis factor (TNF), toll-like receptors (TLRs), and interleukin-1 (IL1) receptor signaling to regulate inflammatory response. Given its central role in inflammatory response, Tpl2 is an attractive small molecule drug target. However, the role of Tpl2 as an oncogene or tumor suppressor gene remains controversial, and its function outside immune cells is not understood. We therefore utilized a Tpl2 kinase dead (Tpl2-KD) mouse model in an 18-month aging study to further elucidate Tpl2 effects on lifespan and chronic disease. Histopathological studies revealed the incidence and severity of spontaneous tumors and non-neoplastic lesions were comparable between wild type and Tpl2-KD mice. The only finding was that male Tpl2-KD mice had higher bodyweight and an increased incidence of liver steatosis, suggesting a sex-specific role for Tpl2 in hepatic lipid metabolism. In conclusion, loss of Tpl2 kinase activity did not lead to increased tumorigenesis over aging in mice but affected likely alterations in lipid metabolism in male animals.
Collapse
Affiliation(s)
- Kai Connie Wu
- Safety Assessment, Genentech, Inc., South San Francisco, CA 94080, USA.
| | - Gary Cain
- Safety Assessment, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Janice Corpuz
- Safety Assessment, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Daqi Xu
- Immunology Department, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Nina Ljumanovic
- Safety Assessment, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Ali A Zarrin
- Immunology Department, Genentech, Inc., South San Francisco, CA 94080, USA.
| |
Collapse
|
36
|
Singh K, Cornell CS, Jackson R, Kabiri M, Phipps M, Desai M, Fogle R, Ying X, Anarat-Cappillino G, Geller S, Johnson J, Roberts E, Malley K, Devlin T, DeRiso M, Berthelette P, Zhang YV, Ryan S, Rao S, Thurberg BL, Bangari DS, Kyostio-Moore S. CRISPR/Cas9 generated knockout mice lacking phenylalanine hydroxylase protein as a novel preclinical model for human phenylketonuria. Sci Rep 2021; 11:7254. [PMID: 33790381 PMCID: PMC8012645 DOI: 10.1038/s41598-021-86663-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 03/18/2021] [Indexed: 02/01/2023] Open
Abstract
Phenylketonuria (PKU) is an autosomal recessive inborn error of L-phenylalanine (Phe) metabolism. It is caused by a partial or complete deficiency of the enzyme phenylalanine hydroxylase (PAH), which is necessary for conversion of Phe to tyrosine (Tyr). This metabolic error results in buildup of Phe and reduction of Tyr concentration in blood and in the brain, leading to neurological disease and intellectual deficits. Patients exhibit retarded body growth, hypopigmentation, hypocholesterolemia and low levels of neurotransmitters. Here we report first attempt at creating a homozygous Pah knock-out (KO) (Hom) mouse model, which was developed in the C57BL/6 J strain using CRISPR/Cas9 where codon 7 (GAG) in Pah gene was changed to a stop codon TAG. We investigated 2 to 6-month-old, male, Hom mice using comprehensive behavioral and biochemical assays, MRI and histopathology. Age and sex-matched heterozygous Pah-KO (Het) mice were used as control mice, as they exhibit enough PAH enzyme activity to provide Phe and Tyr levels comparable to the wild-type mice. Overall, our findings demonstrate that 6-month-old, male Hom mice completely lack PAH enzyme, exhibit significantly higher blood and brain Phe levels, lower levels of brain Tyr and neurotransmitters along with lower myelin content and have significant behavioral deficit. These mice exhibit phenotypes that closely resemble PKU patients such as retarded body growth, cutaneous hypopigmentation, and hypocholesterolemia when compared to the age- and sex-matched Het mice. Altogether, biochemical, behavioral, and pathologic features of this novel mouse model suggest that it can be used as a reliable translational tool for PKU preclinical research and drug development.
Collapse
Affiliation(s)
- Kuldeep Singh
- grid.417555.70000 0000 8814 392XGlobal Discovery Pathology, Translational In-Vivo Models Research Platform, Sanofi, 5 The Mountain Road, Framingham, MA 01701 USA ,Present Address: WuXi AppTec Inc., 8th Floor, 55 Cambridge Parkway, Cambridge, MA 02142 USA
| | - Cathleen S. Cornell
- grid.417555.70000 0000 8814 392XGenomic Medicine Unit, Sanofi, 49 New York Avenue, Framingham, MA 01701 USA
| | - Robert Jackson
- grid.417555.70000 0000 8814 392XGenomic Medicine Unit, Sanofi, 49 New York Avenue, Framingham, MA 01701 USA
| | - Mostafa Kabiri
- grid.420214.1Transgenic Model and Technology, Translational In-Vivo Research Platform, Industrie Park Hoechst, Sanofi, Frankfurt, Germany
| | - Michael Phipps
- grid.417555.70000 0000 8814 392XTransgenic Model and Technology, Translational In-Vivo Models Research Platform, Sanofi, 5 The Mountain Road, Framingham, MA 01701 USA
| | - Mitul Desai
- grid.417555.70000 0000 8814 392XGlobal Bioimaging, Translational In-Vivo Models Research Platform, Sanofi, Framingham, MA 01701 USA
| | - Robert Fogle
- grid.417555.70000 0000 8814 392XGlobal Bioimaging, Translational In-Vivo Models Research Platform, Sanofi, Framingham, MA 01701 USA
| | - Xiaoyou Ying
- grid.417555.70000 0000 8814 392XGlobal Bioimaging, Translational In-Vivo Models Research Platform, Sanofi, Framingham, MA 01701 USA
| | - Gulbenk Anarat-Cappillino
- grid.417555.70000 0000 8814 392XPre-Development Sciences NA, Analytical R&D, Sanofi, Framingham, MA 01701 USA
| | - Sarah Geller
- grid.417555.70000 0000 8814 392XPre-Development Sciences NA, Analytical R&D, Sanofi, Framingham, MA 01701 USA
| | - Jennifer Johnson
- grid.417555.70000 0000 8814 392XGlobal Discovery Pathology, Translational In-Vivo Models Research Platform, Sanofi, 5 The Mountain Road, Framingham, MA 01701 USA
| | - Errin Roberts
- grid.417555.70000 0000 8814 392XGlobal Discovery Pathology, Translational In-Vivo Models Research Platform, Sanofi, 5 The Mountain Road, Framingham, MA 01701 USA
| | - Katie Malley
- grid.417555.70000 0000 8814 392XGlobal Discovery Pathology, Translational In-Vivo Models Research Platform, Sanofi, 5 The Mountain Road, Framingham, MA 01701 USA
| | - Tim Devlin
- grid.417555.70000 0000 8814 392XTransgenic Model and Technology, Translational In-Vivo Models Research Platform, Sanofi, 5 The Mountain Road, Framingham, MA 01701 USA
| | - Matthew DeRiso
- grid.417555.70000 0000 8814 392XTransgenic Model and Technology, Translational In-Vivo Models Research Platform, Sanofi, 5 The Mountain Road, Framingham, MA 01701 USA
| | - Patricia Berthelette
- grid.417555.70000 0000 8814 392XGenomic Medicine Unit, Sanofi, 49 New York Avenue, Framingham, MA 01701 USA
| | - Yao V. Zhang
- grid.417555.70000 0000 8814 392XGenomic Medicine Unit, Sanofi, 49 New York Avenue, Framingham, MA 01701 USA
| | - Susan Ryan
- grid.417555.70000 0000 8814 392XGlobal Discovery Pathology, Translational In-Vivo Models Research Platform, Sanofi, 5 The Mountain Road, Framingham, MA 01701 USA
| | - Srinivas Rao
- grid.417555.70000 0000 8814 392XTranslational In-Vivo Models Research Platform, Sanofi, 49 New York Avenue, Framingham, MA 01701 USA
| | - Beth L. Thurberg
- grid.417555.70000 0000 8814 392XGlobal Discovery Pathology, Translational In-Vivo Models Research Platform, Sanofi, 5 The Mountain Road, Framingham, MA 01701 USA
| | - Dinesh S. Bangari
- grid.417555.70000 0000 8814 392XGlobal Discovery Pathology, Translational In-Vivo Models Research Platform, Sanofi, 5 The Mountain Road, Framingham, MA 01701 USA
| | - Sirkka Kyostio-Moore
- grid.417555.70000 0000 8814 392XGenomic Medicine Unit, Sanofi, 49 New York Avenue, Framingham, MA 01701 USA
| |
Collapse
|
37
|
Gendron WH, Fertan E, Pelletier S, Roddick KM, O'Leary TP, Anini Y, Brown RE. Age related weight loss in female 5xFAD mice from 3 to 12 months of age. Behav Brain Res 2021; 406:113214. [PMID: 33677013 DOI: 10.1016/j.bbr.2021.113214] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/04/2021] [Accepted: 02/25/2021] [Indexed: 02/06/2023]
Abstract
In addition to cognitive decline, patients with Alzheimer's disease (AD) exhibit sensory, motor, and neuropsychiatric deficits. Many AD patients also show weight loss, suggesting that AD may involve a metabolic syndrome. The 5xFAD mouse model shows age-related weight loss compared to wildtype controls, and thus may exhibit metabolic dysfunction. This longitudinal study measured age-related weight loss in female 5xFAD and B6SJL/JF2 wild-type mice from 3 to 12 months of age, and examines some of the behavioural and physiological phenotypes in these mice that have been proposed to contribute to this weight loss. Because some mice had to be singly housed during the study, we also examined genotype by housing interactions. The 5xFAD mice weighed less and ate less than WT littermates starting at 6 months of age, exhibited less home cage activity, had higher frailty scores, less white adipose tissue, and lower leptin expression. At 9 and 12 months of age, heavier 5xFAD mice performed better on the rotarod, suggesting that metabolic deficits which begin between 6 and 9 months of age may exacerbate the behavioural deficits in 5xFAD mice. These results indicate that the 5xFAD mouse is a useful model to study the behavioural and metabolic changes in AD.
Collapse
Affiliation(s)
- William H Gendron
- Departments of Psychology and Neuroscience, Halifax, Nova Scotia, B3H 4R2, Canada
| | - Emre Fertan
- Departments of Psychology and Neuroscience, Halifax, Nova Scotia, B3H 4R2, Canada
| | - Stephanie Pelletier
- Departments of Psychology and Neuroscience, Halifax, Nova Scotia, B3H 4R2, Canada
| | - Kyle M Roddick
- Departments of Psychology and Neuroscience, Halifax, Nova Scotia, B3H 4R2, Canada
| | - Timothy P O'Leary
- Departments of Psychology and Neuroscience, Halifax, Nova Scotia, B3H 4R2, Canada
| | - Younes Anini
- Departments of Physiology and Biophysics, Halifax, Nova Scotia, B3H 4R2, Canada; Departments of Obstetrics and Gynecology, Dalhousie University, Halifax, Nova Scotia, B3H 4R2, Canada
| | - Richard E Brown
- Departments of Psychology and Neuroscience, Halifax, Nova Scotia, B3H 4R2, Canada; Departments of Physiology and Biophysics, Halifax, Nova Scotia, B3H 4R2, Canada.
| |
Collapse
|
38
|
Chan CS, Sun Y, Ke H, Zhao Y, Belete M, Zhang C, Feng Z, Levine AJ, Hu W. Genetic and stochastic influences upon tumor formation and tumor types in Li-Fraumeni mouse models. Life Sci Alliance 2021; 4:e202000952. [PMID: 33376133 PMCID: PMC7772779 DOI: 10.26508/lsa.202000952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 11/24/2022] Open
Abstract
p53 is the most frequently mutated gene in human cancers. Li-Fraumeni syndrome patients inheriting heterozygous p53 mutations often have a much-increased risk to develop cancer(s) at early ages. Recent studies suggest that some individuals inherited p53 mutations do not have the early onset or high frequency of cancers. These observations suggest that other genetic, environmental, immunological, epigenetic, or stochastic factors modify the penetrance of the cancerous mutant Tp53 phenotype. To test this possibility, this study explored dominant genetic modifiers of Tp53 mutations in heterozygous mice with different genetic backgrounds. Both genetic and stochastic effects upon tumor formation were observed in these mice. The genetic background of mice carrying Tp53 mutations has a strong influence upon the tissue type of the tumor produced and the number of tumors formed in a single mouse. The onset age of a tumor is correlated with the tissue type of that tumor, although identical tumor tissue types can occur at very different ages. These observations help to explain the great diversity of cancers in different Li-Fraumeni patients over lifetimes.
Collapse
Affiliation(s)
- Chang S Chan
- Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, NJ, USA
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers, State University of New Jersey, New Brunswick, NJ, USA
| | | | - Hua Ke
- Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, NJ, USA
| | - Yuhan Zhao
- Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, NJ, USA
| | - Merzu Belete
- Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, NJ, USA
| | - Cen Zhang
- Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, NJ, USA
| | - Zhaohui Feng
- Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, NJ, USA
| | - Arnold J Levine
- Simons Center for Systems Biology, Institute for Advanced Study, Princeton, NJ, USA
| | - Wenwei Hu
- Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
39
|
Chakravarti D, LaBella KA, DePinho RA. Telomeres: history, health, and hallmarks of aging. Cell 2021; 184:306-322. [PMID: 33450206 DOI: 10.1016/j.cell.2020.12.028] [Citation(s) in RCA: 263] [Impact Index Per Article: 87.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023]
Abstract
The escalating social and economic burden of an aging world population has placed aging research at center stage. The hallmarks of aging comprise diverse molecular mechanisms and cellular systems that are interrelated and act in concert to drive the aging process. Here, through the lens of telomere biology, we examine how telomere dysfunction may amplify or drive molecular biological processes underlying each hallmark of aging and contribute to development of age-related diseases such as neurodegeneration and cancer. The intimate link of telomeres to aging hallmarks informs preventive and therapeutic interventions designed to attenuate aging itself and reduce the incidence of age-associated diseases.
Collapse
Affiliation(s)
- Deepavali Chakravarti
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kyle A LaBella
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ronald A DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
40
|
Apelgren P, Amoroso M, Säljö K, Lindahl A, Brantsing C, Stridh Orrhult L, Markstedt K, Gatenholm P, Kölby L. Long-term in vivo integrity and safety of 3D-bioprinted cartilaginous constructs. J Biomed Mater Res B Appl Biomater 2021; 109:126-136. [PMID: 32633102 DOI: 10.1002/jbm.b.34687] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/31/2020] [Accepted: 06/23/2020] [Indexed: 11/07/2022]
Abstract
Long-term stability and biological safety are crucial for translation of 3D-bioprinting technology into clinical applications. Here, we addressed the long-term safety and stability issues associated with 3D-bioprinted constructs comprising a cellulose scaffold and human cells (chondrocytes and stem cells) over a period of 10 months in nude mice. Our findings showed that increasing unconfined compression strength over time significantly improved the mechanical stability of the cell-containing constructs relative to cell-free scaffolds. Additionally, the cell-free constructs exhibited a mean compressive stress and stiffness (compressive modulus) of 0.04 ± 0.05 MPa and 0.14 ± 0.18 MPa, respectively, whereas these values for the cell-containing constructs were 0.11 ± 0.08 MPa (p = .019) and 0.53 ± 0.59 MPa (p = .012), respectively. Moreover, histomorphologic analysis revealed that cartilage formed from the cell-containing constructs harbored an abundance of proliferating chondrocytes in clusters, and after 10 months, resembled native cartilage. Furthermore, extension of the experiment over the complete lifecycle of the animal model revealed no signs of ossification, fibrosis, necrosis, or implant-related tumor development in the 3D-bioprinted constructs. These findings confirm the in vivo biological safety and mechanical stability of 3D-bioprinted cartilaginous tissues and support their potential translation into clinical applications.
Collapse
Affiliation(s)
- Peter Apelgren
- Department of Plastic Surgery, Sahlgrenska University Hospital, University of Gothenburg, The Sahlgrenska Academy, Institute of Clinical Sciences, Göteborg, Sweden
| | - Matteo Amoroso
- Department of Plastic Surgery, Sahlgrenska University Hospital, University of Gothenburg, The Sahlgrenska Academy, Institute of Clinical Sciences, Göteborg, Sweden
| | - Karin Säljö
- Department of Plastic Surgery, Sahlgrenska University Hospital, University of Gothenburg, The Sahlgrenska Academy, Institute of Clinical Sciences, Göteborg, Sweden
| | - Anders Lindahl
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Camilla Brantsing
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Linnéa Stridh Orrhult
- 3D Bioprinting Centre, Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Göteborg, Sweden
| | - Kajsa Markstedt
- 3D Bioprinting Centre, Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Göteborg, Sweden
| | - Paul Gatenholm
- 3D Bioprinting Centre, Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Göteborg, Sweden
| | - Lars Kölby
- Department of Plastic Surgery, Sahlgrenska University Hospital, University of Gothenburg, The Sahlgrenska Academy, Institute of Clinical Sciences, Göteborg, Sweden
| |
Collapse
|
41
|
Delaney MA, Imai DM, Buffenstein R. Spontaneous Disease and Pathology of Naked Mole-Rats. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1319:353-380. [PMID: 34424525 DOI: 10.1007/978-3-030-65943-1_15] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Naked mole-rats are highly valuable research models and popular exhibition animals at zoos worldwide. Here, we provide comprehensive descriptions of common postmortem findings of naked mole-rats from both research colonies and populations managed in zoological institutions. Included are brief reviews of their natural history and related physiologic adaptations, unique anatomical features, gross and histologic lesions of common as well as rarely reported disease processes, and discussions of possible pathogeneses with recommendations for future investigations to fill knowledge gaps. Based on postmortem data of several hundreds of naked mole-rats in managed care, it is clear that cancer is extremely rare and infectious disease is infrequently reported. However, despite relatively benign aging phenotypes in this species, several degenerative processes have been nevertheless observed in older populations of naked mole-rats. As such, some potential diet and husbandry-related issues are discussed in addition to the one of the most prominent causes of morbidity and mortality, conspecific aggression and traumas. From this review of lesions and disease, it is clear that pathology, including histopathology, is integral to better understanding mechanisms of healthy aging and cancer resistance of these extraordinary rodents.
Collapse
Affiliation(s)
- Martha A Delaney
- Zoological Pathology Program, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| | - Denise M Imai
- Comparative Pathology Laboratory, School of Veterinary Medicine, University of California at Davis, Davis, CA, USA
| | - Rochelle Buffenstein
- Calico Life Sciences LLC, South San Francisco, CA, USA. .,Department of Pharmacology, University of Texas Health at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
42
|
Hoenerhoff MJ, Meyerholz DK, Brayton C, Beck AP. Challenges and Opportunities for the Veterinary Pathologist in Biomedical Research. Vet Pathol 2020; 58:258-265. [PMID: 33327888 DOI: 10.1177/0300985820974005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Animal models have critical roles in biomedical research in promoting understanding of human disease and facilitating development of new therapies and diagnostic techniques to improve human and animal health. In the study of myriad human conditions, each model requires in-depth characterization of its assets and limitations in order for it to be used to greatest advantage. Veterinary pathology expertise is critical in understanding the relevance and translational validity of animal models to conditions under study, assessing morbidity and mortality, and validating outcomes as relevant or not to the study interventions. Clear communication with investigators and education of research personnel on the use and interpretation of pathology endpoints in animal models are critical to the success of any research program. The veterinary pathologist is underutilized in biomedical research due to many factors including misconceptions about high fiscal costs, lack of perceived value, limited recognition of their expertise, and the generally low number of veterinary pathologists currently employed in biomedical research. As members of the multidisciplinary research team, veterinary pathologists have an important role to educate scientists, ensure accurate interpretation of pathology data, maximize rigor, and ensure reproducibility to provide the most reliable data for animal models in biomedical research.
Collapse
|
43
|
Westrick RJ, Røjkjær LP, Yang AY, Roh MH, Siebert AE, Ginsburg D. Deficiency of plasminogen activator inhibitor-2 results in accelerated tumor growth. J Thromb Haemost 2020; 18:2968-2975. [PMID: 32780555 PMCID: PMC7791406 DOI: 10.1111/jth.15054] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/20/2020] [Accepted: 08/03/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Upregulation of the plasminogen activation system, including urokinase plasminogen activator (uPA), has been observed in many malignancies, suggesting that co-opting the PA system is a common method by which tumor cells accomplish extracellular matrix proteolysis. PAI-2, a serine protease inhibitor, produced from the SERPINB2 gene, inhibits circulating and extracellular matrix-tethered uPA. Decreased SERPINB2 expression has been associated with increased tumor invasiveness and metastasis for several types of cancer. PAI-2 deficiency has not been reported in humans and PAI-2-deficient (SerpinB2-/- ) mice exhibit no apparent abnormalities. OBJECTIVES We investigated the role of PAI-2 deficiency on tumor growth and metastasis. METHODS To explore the long-term impact of PAI-2 deficiency, a cohort of SerpinB2-/- mice were aged to >18 months, with spontaneous malignancies observed in 4/9 animals, all of apparently vascular origin. To further investigate the role of PAI-2 deficiency in malignancy, SerpinB2-/- and wild-type control mice were injected with either B16 melanoma or Lewis lung carcinoma tumor cells, with markedly accelerated tumor growth observed in SerpinB2-/- mice for both cell lines. To determine the relative contributions of PAI-2 from hematopoietic or nonhematopoietically derived sources, bone marrow transplants between wild-type C57BL/6J and SerpinB2-/- mice were performed. RESULTS AND CONCLUSIONS Our results suggest that PAI-2 deficiency increases susceptibility to spontaneous tumorigenesis in the mouse, and demonstrate that SerpinB2 expression derived from a nonhematopoietic compartment is a key host factor in the regulation of tumor growth in both the B16 melanoma and Lewis lung carcinoma models.
Collapse
Affiliation(s)
- Randal J. Westrick
- Oakland University Department of Biological Sciences
- Oakland University Department of Bioengineering
- Oakland University Centers for Data Science and Big Data Analytics and Biomedical Research
- These authors contributed equally to this work
| | - Lisa Payne Røjkjær
- Life Sciences Institute, University of Michigan
- These authors contributed equally to this work
| | | | | | | | - David Ginsburg
- Life Sciences Institute, University of Michigan
- Howard Hughes Medical Institute, University of Michigan
- Departments of Human Genetics, Internal Medicine and Pediatrics, University of Michigan
| |
Collapse
|
44
|
Salloum JS, Garsetti DE, Rogers MB. Genetic background influences the impact of KLOTHO deficiency. Physiol Genomics 2020; 52:512-516. [PMID: 32956023 DOI: 10.1152/physiolgenomics.00094.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Genetic background is a key but sometimes overlooked factor that profoundly impacts disease susceptibility and presentation in both humans and disease models. Here we show that deficiency of KLOTHO protein, an important renal regulator of mineral homeostasis and a cofactor for FGF23, causes different phenotypes in 129S1/SvlmJ (129) and C57BL/6J (B6) mouse strains. The 129 strain is more severely affected, with decreased longevity, decreased body weight, and increased amounts of kidney calcification compared with B6 mice. Reciprocal F1 crosses of the strains also indicate a parentage effect on the Klotho phenotype with F1 KLOTHO-deficient progeny of B6 mothers and 129 fathers having more kidney calcification than progeny of 129 mothers and B6 fathers. Comparing and contrasting the genetic architecture leading to different phenotypes associated with specific inbred mouse strains may reveal previously unrecognized and important metabolic interactions affecting chronic kidney disease.
Collapse
Affiliation(s)
- Jawad S Salloum
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers - New Jersey Medical School, Newark, New Jersey
| | - Diane E Garsetti
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers - New Jersey Medical School, Newark, New Jersey
| | - Melissa B Rogers
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers - New Jersey Medical School, Newark, New Jersey
| |
Collapse
|
45
|
Abstract
The review covers select disease conditions most frequently described in aging rodents (rats, mice, hamsters, guinea pigs), rabbits, and ferrets. The conditions are categorized by general organ systems, infectious diseases, and neoplasms. Two data systems, the Veterinary Medical Teaching Hospital and Comparative Pathology Laboratory at the University of California, Davis and Zoo/Exotic Pathology Service, Citrus Heights, California were used in the determining disease conditions to describe.
Collapse
Affiliation(s)
- Drury R Reavill
- ZNLabs Veterinary Diagnostics, 7647 Wachtel Way, Citrus Heights, CA 95610, USA.
| | - Denise M Imai
- Comparative Pathology Laboratory, University of California, 1000 Old Davis Road, Building R1, Davis, CA 95616, USA
| |
Collapse
|
46
|
Arriola Apelo SI, Lin A, Brinkman JA, Meyer E, Morrison M, Tomasiewicz JL, Pumper CP, Baar EL, Richardson NE, Alotaibi M, Lamming DW. Ovariectomy uncouples lifespan from metabolic health and reveals a sex-hormone-dependent role of hepatic mTORC2 in aging. eLife 2020; 9:56177. [PMID: 32720643 PMCID: PMC7386906 DOI: 10.7554/elife.56177] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023] Open
Abstract
Inhibition of mTOR (mechanistic Target Of Rapamycin) signaling by rapamycin promotes healthspan and longevity more strongly in females than males, perhaps because inhibition of hepatic mTORC2 (mTOR Complex 2) specifically reduces the lifespan of males. Here, we demonstrate using gonadectomy that the sex-specific impact of reduced hepatic mTORC2 is not reversed by depletion of sex hormones. Intriguingly, we find that ovariectomy uncouples lifespan from metabolic health, with ovariectomized females having improved survival despite paradoxically having increased adiposity and decreased control of blood glucose levels. Further, ovariectomy unexpectedly promotes midlife survival of female mice lacking hepatic mTORC2, significantly increasing the survival of those mice that do not develop cancer. In addition to identifying a sex hormone-dependent role for hepatic mTORC2 in female longevity, our results demonstrate that metabolic health is not inextricably linked to lifespan in mammals, and highlight the importance of evaluating healthspan in mammalian longevity studies.
Collapse
Affiliation(s)
- Sebastian I Arriola Apelo
- Department of Dairy Science, University of Wisconsin-Madison, Madison, United States.,William S. Middleton Memorial Veterans Hospital, Madison, United States.,Department of Medicine, University of Wisconsin-Madison, Madison, United States
| | - Amy Lin
- Department of Dairy Science, University of Wisconsin-Madison, Madison, United States.,William S. Middleton Memorial Veterans Hospital, Madison, United States.,Department of Medicine, University of Wisconsin-Madison, Madison, United States
| | - Jacqueline A Brinkman
- William S. Middleton Memorial Veterans Hospital, Madison, United States.,Department of Medicine, University of Wisconsin-Madison, Madison, United States
| | - Emma Meyer
- Department of Dairy Science, University of Wisconsin-Madison, Madison, United States.,William S. Middleton Memorial Veterans Hospital, Madison, United States.,Department of Medicine, University of Wisconsin-Madison, Madison, United States
| | - Mark Morrison
- William S. Middleton Memorial Veterans Hospital, Madison, United States.,Department of Medicine, University of Wisconsin-Madison, Madison, United States
| | - Jay L Tomasiewicz
- William S. Middleton Memorial Veterans Hospital, Madison, United States
| | - Cassidy P Pumper
- William S. Middleton Memorial Veterans Hospital, Madison, United States.,Department of Medicine, University of Wisconsin-Madison, Madison, United States
| | - Emma L Baar
- William S. Middleton Memorial Veterans Hospital, Madison, United States.,Department of Medicine, University of Wisconsin-Madison, Madison, United States
| | - Nicole E Richardson
- William S. Middleton Memorial Veterans Hospital, Madison, United States.,Department of Medicine, University of Wisconsin-Madison, Madison, United States.,Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, United States
| | - Mohammed Alotaibi
- William S. Middleton Memorial Veterans Hospital, Madison, United States.,Department of Medicine, University of Wisconsin-Madison, Madison, United States.,Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, United States
| | - Dudley W Lamming
- William S. Middleton Memorial Veterans Hospital, Madison, United States.,Department of Medicine, University of Wisconsin-Madison, Madison, United States.,Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, United States.,University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, United States
| |
Collapse
|
47
|
Queen NJ, Hassan QN, Cao L. Improvements to Healthspan Through Environmental Enrichment and Lifestyle Interventions: Where Are We Now? Front Neurosci 2020; 14:605. [PMID: 32655354 PMCID: PMC7325954 DOI: 10.3389/fnins.2020.00605] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 05/18/2020] [Indexed: 12/11/2022] Open
Abstract
Environmental enrichment (EE) is an experimental paradigm that is used to explore how a complex, stimulating environment can impact overall health. In laboratory animal experiments, EE housing conditions typically include larger-than-standard cages, abundant bedding, running wheels, mazes, toys, and shelters which are rearranged regularly to further increase stimulation. EE has been shown to improve multiple aspects of health, including but not limited to metabolism, learning and cognition, anxiety and depression, and immunocompetence. Recent advances in lifespan have led some researchers to consider aging as a risk factor for disease. As such, there is a pressing need to understand the processes by which healthspan can be increased. The natural and predictable changes during aging can be reversed or decreased through EE and its underlying mechanisms. Here, we review the use of EE in laboratory animals to understand mechanisms involved in aging, and comment on relative areas of strength and weakness in the current literature. We additionally address current efforts toward applying EE-like lifestyle interventions to human health to extend healthspan. Although increasing lifespan is a clear goal of medical research, improving the quality of this added time also deserves significant attention. Despite hurdles in translating experimental results toward clinical application, we argue there is great potential in using features of EE toward improving human healthy life expectancy or healthspan, especially in the context of increased global longevity.
Collapse
Affiliation(s)
- Nicholas J. Queen
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Quais N. Hassan
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
- Medical Scientist Training Program, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Lei Cao
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, United States
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| |
Collapse
|
48
|
Razzoli M, Lindsay A, Law ML, Chamberlain CM, Southern WM, Berg M, Osborn J, Engeland WC, Metzger JM, Ervasti JM, Bartolomucci A. Social stress is lethal in the mdx model of Duchenne muscular dystrophy. EBioMedicine 2020; 55:102700. [PMID: 32192914 PMCID: PMC7251247 DOI: 10.1016/j.ebiom.2020.102700] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 02/10/2020] [Accepted: 02/18/2020] [Indexed: 12/19/2022] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is caused by the loss of dystrophin. Severe and ultimately lethal, DMD progresses relatively slowly in that patients become wheelchair bound only around age twelve with a survival expectancy reaching the third decade of life. Methods The mildly-affected mdx mouse model of DMD, and transgenic DysΔMTB-mdx and Fiona-mdx mice expressing dystrophin or utrophin, respectively, were exposed to either mild (scruffing) or severe (subordination stress) stress paradigms and profiled for their behavioral and physiological responses. A subgroup of mdx mice exposed to subordination stress were pretreated with the beta-blocker metoprolol. Findings Subordination stress caused lethality in ∼30% of mdx mice within 24 h and ∼70% lethality within 48 h, which was not rescued by metoprolol. Lethality was associated with heart damage, waddling gait and hypo-locomotion, as well as marked up-regulation of the hypothalamus-pituitary-adrenocortical axis. A novel cardiovascular phenotype emerged in mdx mice, in that scruffing caused a transient drop in arterial pressure, while subordination stress caused severe and sustained hypotension with concurrent tachycardia. Transgenic expression of dystrophin or utrophin in skeletal muscle protected mdx mice from scruffing and social stress-induced responses including mortality. Interpretation We have identified a robust new stress phenotype in the otherwise mildly affected mdx mouse that suggests relatively benign handling may impact the outcome of behavioural experiments, but which should also expedite the knowledge-based therapy development for DMD. Funding Greg Marzolf Jr. Foundation, Summer's Wish Fund, NIAMS, Muscular Dystrophy Association, University of Minnesota and John and Cheri Gunvalson Trust.
Collapse
Affiliation(s)
- Maria Razzoli
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Angus Lindsay
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Michelle L Law
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Christopher M Chamberlain
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN, United States
| | - William M Southern
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Madeleine Berg
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - John Osborn
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - William C Engeland
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Joseph M Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - James M Ervasti
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN, United States.
| | - Alessandro Bartolomucci
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States.
| |
Collapse
|
49
|
Lindhurst MJ, Brinster LR, Kondolf HC, Shwetar JJ, Yourick MR, Shiferaw H, Keppler-Noreuil KM, Elliot G, Rivas C, Garrett L, Gomez-Rodriguez J, Sebire NJ, Hewitt SM, Schwartzberg PL, Biesecker LG. A mouse model of Proteus syndrome. Hum Mol Genet 2020; 28:2920-2936. [PMID: 31194862 DOI: 10.1093/hmg/ddz116] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/20/2019] [Accepted: 05/22/2019] [Indexed: 01/29/2023] Open
Abstract
Proteus syndrome is a mosaic, progressive overgrowth disorder caused by a somatic activating variant c.49G > A p.(E17K) in AKT1. The presentation in affected individuals is variable, with a diversity of tissues demonstrating abnormalities. Common manifestations include skin and bony overgrowth, vascular malformations (VMs), cysts and benign tumors. We used two methods to create mouse models that had endogenously-regulated mosaic expression of the Proteus syndrome variant. Variant allele fractions (VAFs) ranged from 0% to 50% across numerous tissues in 44 Proteus syndrome mice. Mice were phenotypically heterogeneous with lesions rarely observed before 12 months of age. VMs were the most frequent finding with a total of 69 found in 29 of 44 Proteus syndrome mice. Twenty-eight cysts and ectasia, frequently biliary, were seen in 22 of 44 Proteus syndrome mice. Varying levels of mammary hyperplasia were seen in 10 of 16 female Proteus syndrome mice with other localized regions of hyperplasia and stromal expansion noted in several additional animals. Interestingly, 27 of 31 Proteus syndrome animals had non-zero blood VAF that is in contrast to the human disorder where it is rarely seen in peripheral blood. Identification of variant-positive cells by green fluorescent protein (GFP) staining in chimeric Proteus syndrome mice showed that in some lesions, hyperplastic cells were predominantly GFP/Akt1E17K-positive and showed increased pAKT signal compared to GFP-negative cells. However, hyperplastic mammary epithelium was a mixture of GFP/Akt1E17K-positive and negative cells with some GFP/Akt1E17K-negative cells also having increased pAKT signal suggesting that the variant-positive cells can induce lesion formation in a non-cell autonomous manner.
Collapse
Affiliation(s)
- Marjorie J Lindhurst
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - Lauren R Brinster
- Division of Veterinary Resources, Office of Research Services, NIH, Bethesda, MD, USA
| | - Hannah C Kondolf
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - Jasmine J Shwetar
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - Miranda R Yourick
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - Henoke Shiferaw
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - Kim M Keppler-Noreuil
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - Gene Elliot
- Genetic Disease Research Branch, NHGRI, NIH, Bethesda, MD, USA
| | - Cecilia Rivas
- Genetic Disease Research Branch, NHGRI, NIH, Bethesda, MD, USA
| | - Lisa Garrett
- Genetic Disease Research Branch, NHGRI, NIH, Bethesda, MD, USA
| | | | - Neil J Sebire
- Histopathology Department, Great Ormond Street Hospital NIHR Biomedical Research Centre, London, UK
| | - Stephen M Hewitt
- Laboratory of Pathology, National Cancer Institute, NIH, Bethesda, MD, USA
| | | | - Leslie G Biesecker
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
50
|
The Exacerbation of Aging and Oxidative Stress in the Epididymis of Sod1 Null Mice. Antioxidants (Basel) 2020; 9:antiox9020151. [PMID: 32054065 PMCID: PMC7071042 DOI: 10.3390/antiox9020151] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/13/2020] [Accepted: 02/10/2020] [Indexed: 12/11/2022] Open
Abstract
There is growing evidence that the quality of spermatozoa decreases with age and that children of older fathers have a higher incidence of birth defects and genetic mutations. The free radical theory of aging proposes that changes with aging are due to the accumulation of damage induced by exposure to excess reactive oxygen species. We showed previously that absence of the superoxide dismutase 1 (Sod1) antioxidant gene results in impaired mechanisms of repairing DNA damage in the testis in young Sod1−/− mice. In this study, we examined the effects of aging and the Sod−/− mutation on mice epididymal histology and the expression of markers of oxidative damage. We found that both oxidative nucleic acid damage (via 8-hydroxyguanosine) and lipid peroxidation (via 4-hydroxynonenal) increased with age and in Sod1−/− mice. These findings indicate that lack of SOD1 results in an exacerbation of the oxidative damage accumulation-related aging phenotype.
Collapse
|