1
|
Kleeblatt E, Lazki-Hagenbach P, Nabet E, Cohen R, Bahri R, Rogers N, Langton A, Bulfone-Paus S, Frenkel D, Sagi-Eisenberg R. p16 Ink4a-induced senescence in cultured mast cells as a model for ageing reveals significant morphological and functional changes. Immun Ageing 2024; 21:77. [PMID: 39529115 PMCID: PMC11552350 DOI: 10.1186/s12979-024-00478-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Mast cells (MCs) are tissue resident cells of the immune system, mainly known for their role in allergy. However, mounting evidence indicates their involvement in the pathology of age-related diseases, such as Alzheimer's disease, Parkinson's disease, and cancer. MC numbers increase in aged tissues, but how ageing affects MCs is poorly understood. RESULTS We show that MC ageing is associated with the increased expression of the cell cycle inhibitor p16 Ink4a, a marker and inducer of cellular senescence. Relying on this observation and the tight association of ageing with senescence, we developed a model of inducible senescence based on doxycycline-induced expression of p16Ink4a in cultured bone marrow derived MCs (BMMCs). Using this model, we show that senescent MCs upregulate IL-1β, TNF-α and VEGF-A. We also demonstrate that senescence causes marked morphological changes that impact MC function. Senescent MCs are larger, contain a larger number of secretory granules (SGs) and have less membrane protrusions. Particularly striking are the changes in their SGs, reflected in a significant reduction in the number of electron dense SGs with a concomitant increase in lucent SGs containing intraluminal vesicles. The changes in SG morphology are accompanied by changes in MC degranulation, including a significant increase in receptor-triggered release of CD63-positive extracellular vesicles (EVs) and the exteriorisation of proteoglycans, as opposed to a gradual inhibition of the release of β-hexosaminidase. CONCLUSIONS The inducible expression of p16Ink4a imposes MC senescence, providing a model for tracking the autonomous changes that occur in MCs during ageing. These changes include both morphological and functional alterations. In particular, the increased release of small EVs by senescent MCs suggests an enhanced ability to modulate neighbouring cells.
Collapse
Affiliation(s)
- Elisabeth Kleeblatt
- Department of Cell and Developmental Biology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Pia Lazki-Hagenbach
- Department of Cell and Developmental Biology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Ellon Nabet
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Reli Cohen
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Rajia Bahri
- Lydia Becker Institute of Immunology and Inflammation, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Nicholas Rogers
- Department of Environmental Studies, School of Mechanical Engineering, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Abigail Langton
- Lydia Becker Institute of Immunology and Inflammation, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Silvia Bulfone-Paus
- Lydia Becker Institute of Immunology and Inflammation, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Dan Frenkel
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 69978, Israel.
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel.
| | - Ronit Sagi-Eisenberg
- Department of Cell and Developmental Biology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, 69978, Israel.
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel.
| |
Collapse
|
2
|
Tsai YT, Seymen N, Thompson IR, Zou X, Mumtaz W, Gerlevik S, Mufti GJ, Karimi MM. Expression of most retrotransposons in human blood correlates with biological aging. eLife 2024; 13:RP96575. [PMID: 39417397 PMCID: PMC11486490 DOI: 10.7554/elife.96575] [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: 10/19/2024] Open
Abstract
Retrotransposons (RTEs) have been postulated to reactivate with age and contribute to aging through activated innate immune response and inflammation. Here, we analyzed the relationship between RTE expression and aging using published transcriptomic and methylomic datasets of human blood. Despite no observed correlation between RTE activity and chronological age, the expression of most RTE classes and families except short interspersed nuclear elements (SINEs) correlated with biological age-associated gene signature scores. Strikingly, we found that the expression of SINEs was linked to upregulated DNA repair pathways in multiple cohorts. We also observed DNA hypomethylation with aging and the significant increase in RTE expression level in hypomethylated RTEs except for SINEs. Additionally, our single-cell transcriptomic analysis suggested a role for plasma cells in aging mediated by RTEs. Altogether, our multi-omics analysis of large human cohorts highlights the role of RTEs in biological aging and suggests possible mechanisms and cell populations for future investigations.
Collapse
Affiliation(s)
- Yi-Ting Tsai
- Comprehensive Cancer Centre, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College LondonLondonUnited Kingdom
| | - Nogayhan Seymen
- Comprehensive Cancer Centre, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College LondonLondonUnited Kingdom
| | - I Richard Thompson
- Comprehensive Cancer Centre, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College LondonLondonUnited Kingdom
| | - Xinchen Zou
- MRC LMS, Imperial College LondonLondonUnited Kingdom
| | - Warisha Mumtaz
- Comprehensive Cancer Centre, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College LondonLondonUnited Kingdom
| | - Sila Gerlevik
- Comprehensive Cancer Centre, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College LondonLondonUnited Kingdom
| | - Ghulam J Mufti
- Comprehensive Cancer Centre, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College LondonLondonUnited Kingdom
| | - Mohammad M Karimi
- Comprehensive Cancer Centre, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College LondonLondonUnited Kingdom
| |
Collapse
|
3
|
Kar P, Sivasailam A, Lavarti R, Cai L, Thangaraju M, Nguyen E, Mundluru B, Raju RP. p53 dependence of senescence markers p21v1 and p21v2 in aging and acute injury. NPJ AGING 2024; 10:45. [PMID: 39402059 PMCID: PMC11473800 DOI: 10.1038/s41514-024-00175-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 09/29/2024] [Indexed: 10/17/2024]
Abstract
The senescence phenotype is heterogeneous, as observed by the context-dependent differential expression of senescence markers. Here, we provide evidence to demonstrate an inverse relationship in the expression pattern of the two murine variants of p21 (p21v1, and p21v2) in aging and hemorrhagic shock. While an upregulation of p21v1 was observed following hemorrhagic shock injury, p21v2 was upregulated in the aged mouse. We further show that the p21v1 response is, at least, partially independent of p53.
Collapse
Affiliation(s)
- Parmita Kar
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Ashok Sivasailam
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Rupa Lavarti
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Lun Cai
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Muthusamy Thangaraju
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Emma Nguyen
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Bhavishya Mundluru
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Raghavan Pillai Raju
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, USA.
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
4
|
Wu M, Yao Y, Chen R, Fu B, Sun Y, Yu Y, Liu Y, Feng H, Guo S, Yang Y, Zhang C. Effects of Melatonin and 3,5,3'-Triiodothyronine on the Development of Rat Granulosa Cells. Nutrients 2024; 16:3085. [PMID: 39339685 PMCID: PMC11435325 DOI: 10.3390/nu16183085] [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: 07/29/2024] [Revised: 09/05/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Melatonin, as an endocrine neurotransmitter, can promote the development of the ovary. Meanwhile, it also has protective effect on the ovary as an antioxidant. Thyroid hormone (TH) is essential for normal human reproductive function. Many studies have shown that 3,5,3'-triiodothyronine (T3) regulates the development of ovarian granulosa cells. However, little is known about the specific mechanisms by which melatonin combines with T3 to regulate granulosa cell development. The aim of present study was to investigate the effects and the possible mechanisms of melatonin and T3 on ovarian granulosa cell development. In the present study, cell development and apoptosis were detected by CCK8, EdU and TUNEL, respectively. The levels of related proteins were analyzed by Western blotting. The results showed that oxidative stress (OS) and reactive oxygen species (ROS) were induced by H2O2 in granulosa cells, and cell apoptosis was also increased accompanied with the decreased cellular proliferation and viability. Melatonin protects granulosa cells from H2O2-induced apoptosis and OS by downregulating ROS levels, especially in the presence of T3. Co-treatment of cell with melatonin and T3 also promotes the expression of GRP78 and AMH, while inhibiting CHOP, Caspase-3, and P16. It was demonstrated that melatonin alone or in combination with T3 had positive effect on the development of granulosa cells. In addition, the AMPK/SIRT1 signaling pathway is involved in the process of melatonin/T3 promoting granulosa cell development.
Collapse
Affiliation(s)
- Mingqi Wu
- College of Life Science, Capital Normal University, Beijing 100048, China
| | - Yilin Yao
- College of Life Science, Capital Normal University, Beijing 100048, China
| | - Rui Chen
- College of Life Science, Capital Normal University, Beijing 100048, China
| | - Baoqiang Fu
- College of Life Science, Capital Normal University, Beijing 100048, China
| | - Ying Sun
- College of Life Science, Capital Normal University, Beijing 100048, China
| | - Yakun Yu
- College of Life Science, Capital Normal University, Beijing 100048, China
| | - Yan Liu
- College of Life Science, Capital Normal University, Beijing 100048, China
| | - Haoyuan Feng
- College of Life Science, Capital Normal University, Beijing 100048, China
| | - Shuaitian Guo
- College of Life Science, Capital Normal University, Beijing 100048, China
| | - Yanzhou Yang
- Key Laboratory of Fertility Preservation and Maintenance, Ministry of Education, Key Laboratory of Reproduction and Genetics in Ningxia, Department of Histology and Embryology, Ningxia Medical University, Yinchuan 750004, China
| | - Cheng Zhang
- College of Life Science, Capital Normal University, Beijing 100048, China
| |
Collapse
|
5
|
Suda M, Paul KH, Tripathi U, Minamino T, Tchkonia T, Kirkland JL. Targeting Cell Senescence and Senolytics: Novel Interventions for Age-Related Endocrine Dysfunction. Endocr Rev 2024; 45:655-675. [PMID: 38500373 PMCID: PMC11405506 DOI: 10.1210/endrev/bnae010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 01/11/2024] [Accepted: 03/12/2024] [Indexed: 03/20/2024]
Abstract
Multiple changes occur in hormonal regulation with aging and across various endocrine organs. These changes are associated with multiple age-related disorders and diseases. A better understanding of responsible underling biological mechanisms could help in the management of multiple endocrine disorders over and above hormone replacement therapy (HRT). Cellular senescence is involved in multiple biological aging processes and pathologies common in elderly individuals. Cellular senescence, which occurs in many older individuals but also across the lifespan in association with tissue damage, acute and chronic diseases, certain drugs, and genetic syndromes, may contribute to such endocrine disorders as osteoporosis, metabolic syndrome, and type 2 diabetes mellitus. Drugs that selectively induce senescent cell removal, "senolytics,", and drugs that attenuate the tissue-destructive secretory state of certain senescent cells, "senomorphics," appear to delay the onset of or alleviate multiple diseases, including but not limited to endocrine disorders such as diabetes, complications of obesity, age-related osteoporosis, and cancers as well as atherosclerosis, chronic kidney disease, neurodegenerative disorders, and many others. More than 30 clinical trials of senolytic and senomorphic agents have already been completed, are underway, or are planned for a variety of indications. Targeting senescent cells is a novel strategy that is distinct from conventional therapies such as HRT, and thus might address unmet medical needs and can potentially amplify effects of established endocrine drug regimens, perhaps allowing for dose decreases and reducing side effects.
Collapse
Affiliation(s)
- Masayoshi Suda
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Karl H Paul
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Pharmacology, Karolinska Institutet, Solnavägen 9, 171 65 Solna, Sweden
| | - Utkarsh Tripathi
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Japan Agency for Medical Research and Development-Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, Tokyo, 100-0004, Japan
| | - Tamara Tchkonia
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - James L Kirkland
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
6
|
Elwakiel A, Gupta D, Rana R, Manoharan J, Al-Dabet MM, Ambreen S, Fatima S, Zimmermann S, Mathew A, Li Z, Singh K, Gupta A, Pal S, Sulaj A, Kopf S, Schwab C, Baber R, Geffers R, Götze T, Alo B, Lamers C, Kluge P, Kuenze G, Kohli S, Renné T, Shahzad K, Isermann B. Factor XII signaling via uPAR-integrin β1 axis promotes tubular senescence in diabetic kidney disease. Nat Commun 2024; 15:7963. [PMID: 39261453 PMCID: PMC11390906 DOI: 10.1038/s41467-024-52214-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 08/30/2024] [Indexed: 09/13/2024] Open
Abstract
Coagulation factor XII (FXII) conveys various functions as an active protease that promotes thrombosis and inflammation, and as a zymogen via surface receptors like urokinase-type plasminogen activator receptor (uPAR). While plasma levels of FXII are increased in diabetes mellitus and diabetic kidney disease (DKD), a pathogenic role of FXII in DKD remains unknown. Here we show that FXII is locally expressed in kidney tubular cells and that urinary FXII correlates with kidney dysfunction in DKD patients. F12-deficient mice (F12-/-) are protected from hyperglycemia-induced kidney injury. Mechanistically, FXII interacts with uPAR on tubular cells promoting integrin β1-dependent signaling. This signaling axis induces oxidative stress, persistent DNA damage and senescence. Blocking uPAR or integrin β1 ameliorates FXII-induced tubular cell injury. Our findings demonstrate that FXII-uPAR-integrin β1 signaling on tubular cells drives senescence. These findings imply previously undescribed diagnostic and therapeutic approaches to detect or treat DKD and possibly other senescence-associated diseases.
Collapse
Affiliation(s)
- Ahmed Elwakiel
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany.
| | - Dheerendra Gupta
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Rajiv Rana
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Jayakumar Manoharan
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Moh'd Mohanad Al-Dabet
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
- Department of Medical Laboratory Sciences, School of Science, University of Jordan, Amman, Jordan
| | - Saira Ambreen
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Sameen Fatima
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Silke Zimmermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Akash Mathew
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Zhiyang Li
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Kunal Singh
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Anubhuti Gupta
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Surinder Pal
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Alba Sulaj
- Internal Medicine I and Clinical Chemistry, German Diabetes Center (DZD), University of Heidelberg, Heidelberg, Germany
| | - Stefan Kopf
- Internal Medicine I and Clinical Chemistry, German Diabetes Center (DZD), University of Heidelberg, Heidelberg, Germany
| | - Constantin Schwab
- Institute of pathology, University of Heidelberg, Heidelberg, Germany
| | - Ronny Baber
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
- Leipzig Medical Biobank, Leipzig University, Leipzig, Germany
| | - Robert Geffers
- Genome Analytics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Tom Götze
- Institute for Drug Discovery, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Bekas Alo
- Institute for Drug Discovery, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Christina Lamers
- Institute for Drug Discovery, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Paul Kluge
- Institute for Drug Discovery, Faculty of Medicine, Leipzig University, Leipzig, Germany
| | - Georg Kuenze
- Institute for Drug Discovery, Faculty of Medicine, Leipzig University, Leipzig, Germany
- Center for Scalable Data Analytics and Artificial Intelligence, Leipzig University, Leipzig, Germany
| | - Shrey Kohli
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, Mainz, Germany
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Khurrum Shahzad
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany
- National Centre of Excellence in Molecular Biology, University of the Punjab, 87-West Canal Bank Road, Lahore, Pakistan
| | - Berend Isermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig Medical Center, Leipzig, Germany.
| |
Collapse
|
7
|
Lho Y, Park Y, Do JY, Kim AY, Park YE, Kang SH. Empagliflozin attenuates epithelial-to-mesenchymal transition through senescence in peritoneal dialysis. Am J Physiol Renal Physiol 2024; 327:F363-F372. [PMID: 38961839 DOI: 10.1152/ajprenal.00028.2024] [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: 01/23/2024] [Revised: 06/25/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is considered as one of the senescence processes; reportedly, antisenescence therapies effectively reduce EMT. Some models have shown antisenescence effects with the use of sodium-glucose cotransporter 2 (SGLT2) inhibitor. Therefore, our study investigated the antisenescence effects of empagliflozin as an SGLT2 inhibitor in a peritoneal fibrosis model and their impact on EMT inhibition. For in vitro study, human peritoneal mesothelial cells (HPMCs) were isolated and grown in a 96-well plate. The cell media were exchanged with serum-free M199 medium with d-glucose, with or without empagliflozin. All animal experiments were carried out in male mice. Mice were randomly classified into three treatment groups based on peritoneal dialysis (PD) or empagliflozin. We evaluated changes in senescence and EMT markers in HPMCs and PD model. HPMCs treated with glucose transformed from cobblestone to spindle shape, resulting in EMT. Empagliflozin attenuated these morphological changes. Reactive oxygen species production, DNA damage, senescence, and EMT markers were increased by glucose treatment; however, cotreatment with glucose and empagliflozin attenuated these changes. For the mice with PD, an increase in thickness, collagen deposition, staining for senescence, or EMT markers of the parietal peritoneum was observed, which, however, was attenuated by cotreatment with empagliflozin. p53, p21, and p16 increased in mice with PD compared with those in the control group; however, these changes were decreased by empagliflozin. In conclusion, empagliflozin effectively attenuated glucose-induced EMT in HPMCs through a decrease in senescence. Cotreatment with empagliflozin improved peritoneal thickness and fibrosis in PD.NEW & NOTEWORTHY Epithelial-to-mesenchymal transition (EMT) is considered one of the senescence processes. Antisenescence therapies may effectively reduce EMT in peritoneal dialysis models. Human peritoneal mesothelial cells treated with glucose show an increase in senescence and EMT markers; however, empagliflozin attenuates these changes. Mice undergoing peritoneal dialysis exhibit increased senescence and EMT markers, which are decreased by empagliflozin. These findings suggest that empagliflozin may emerge as a novel strategy for prevention or treatment of peritoneal fibrosis.
Collapse
Affiliation(s)
- Yunmee Lho
- Senotherpy-Based Metabolic Disease Control Research Center, College of Medicine, Yeungnam University, Daegu, Republic of Korea
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Yeong Park
- Senotherpy-Based Metabolic Disease Control Research Center, College of Medicine, Yeungnam University, Daegu, Republic of Korea
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Jun Young Do
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - A-Young Kim
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Yong-Eun Park
- Department of Surgery, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Seok Hui Kang
- Senotherpy-Based Metabolic Disease Control Research Center, College of Medicine, Yeungnam University, Daegu, Republic of Korea
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| |
Collapse
|
8
|
Shi Y, Zhang Y, Zhang Y, Yao J, Guo J, Xu X, Wang L. Advances in Nanotherapy for Targeting Senescent Cells. Int J Nanomedicine 2024; 19:8797-8813. [PMID: 39220198 PMCID: PMC11365502 DOI: 10.2147/ijn.s469110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 08/17/2024] [Indexed: 09/04/2024] Open
Abstract
Aging is an inevitable process in the human body, and cellular senescence refers to irreversible cell cycle arrest caused by external aging-promoting mechanisms. Moreover, as age increases, the accumulation of senescent cells limits both the health of the body and lifespan and even accelerates the occurrence and progression of age-related diseases. Therefore, it is crucial to delay the periodic irreversible arrest and continuous accumulation of senescent cells to address the issue of aging. The fundamental solution is targeted therapy focused on eliminating senescent cells or reducing the senescence-associated secretory phenotype. Over the past few decades, the remarkable development of nanomaterials has revolutionized clinical drug delivery pathways. Their unique optical, magnetic, and electrical properties effectively compensate for the shortcomings of traditional drugs, such as low stability and short half-life, thereby maximizing the bioavailability and minimizing the toxicity of drug delivery. This article provides an overview of how nanomedicine systems control drug release and achieve effective diagnosis. By presenting and analyzing recent advances in nanotherapy for targeting senescent cells, the underlying mechanisms of nanomedicine for senolytic and senomorphic therapy are clarified, providing great potential for targeting senescent cells.
Collapse
Affiliation(s)
- Yurou Shi
- Geriatric Medicine Center, Department of Endocrinology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310015, People’s Republic of China
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, 310015, People’s Republic of China
| | - Yingjie Zhang
- Geriatric Medicine Center, Department of Endocrinology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310015, People’s Republic of China
| | - Yaxuan Zhang
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, 310015, People’s Republic of China
| | - Jiali Yao
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, 310015, People’s Republic of China
| | - Junping Guo
- Rainbowfish Rehabilitation and Nursing School, Hangzhou Vocational & Technical College, Hangzhou, 310018, People’s Republic of China
| | - Xiaoling Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, 310015, People’s Republic of China
| | - Lijun Wang
- Geriatric Medicine Center, Department of Endocrinology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310015, People’s Republic of China
| |
Collapse
|
9
|
Sudduth ER, López Ruiz A, Trautmann-Rodriguez M, Fromen CA. Age-dependent changes in phagocytic activity: in vivo response of mouse pulmonary antigen presenting cells to direct lung delivery of charged PEGDA nanoparticles. J Nanobiotechnology 2024; 22:476. [PMID: 39135064 PMCID: PMC11318229 DOI: 10.1186/s12951-024-02743-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 08/02/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Current needle-based vaccination for respiratory viruses is ineffective at producing sufficient, long-lasting local immunity in the elderly. Direct pulmonary delivery to the resident local pulmonary immune cells can create long-term mucosal responses. However, criteria for drug vehicle design rules that can overcome age-specific changes in immune cell functions have yet to be established. RESULTS Here, in vivo charge-based nanoparticle (NP) uptake was compared in mice of two age groups (2- and 16-months) within the four notable pulmonary antigen presenting cell (APC) populations: alveolar macrophages (AM), interstitial macrophages (IM), CD103+ dendritic cells (DCs), and CD11b+ DCs. Both macrophage populations exhibited preferential uptake of anionic nanoparticles but showed inverse rates of phagocytosis between the AM and IM populations across age. DC populations demonstrated preferential uptake of cationic nanoparticles, which remarkably did not significantly change in the aged group. Further characterization of cell phenotypes post-NP internalization demonstrated unique surface marker expression and activation levels for each APC population, showcasing heightened DC inflammatory response to NP delivery in the aged group. CONCLUSION The age of mice demonstrated significant preferences in the charge-based NP uptake in APCs that differed greatly between macrophages and DCs. Carefully balance of the targeting and activation of specific types of pulmonary APCs will be critical to produce efficient, age-based vaccines for the growing elderly population.
Collapse
Affiliation(s)
- Emma R Sudduth
- Chemical and Biomolecular Engineering Department, University of Delaware, 150 Academy St, Newark, DE, 19716, USA
| | - Aida López Ruiz
- Chemical and Biomolecular Engineering Department, University of Delaware, 150 Academy St, Newark, DE, 19716, USA
| | - Michael Trautmann-Rodriguez
- Chemical and Biomolecular Engineering Department, University of Delaware, 150 Academy St, Newark, DE, 19716, USA
| | - Catherine A Fromen
- Chemical and Biomolecular Engineering Department, University of Delaware, 150 Academy St, Newark, DE, 19716, USA.
| |
Collapse
|
10
|
Jain N, Li JL, Tong L, Jasmine F, Kibriya MG, Demanelis K, Oliva M, Chen LS, Pierce BL. DNA methylation correlates of chronological age in diverse human tissue types. Epigenetics Chromatin 2024; 17:25. [PMID: 39118140 PMCID: PMC11308253 DOI: 10.1186/s13072-024-00546-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/15/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND While the association of chronological age with DNA methylation (DNAm) in whole blood has been extensively studied, the tissue-specificity of age-related DNAm changes remains an active area of research. Studies investigating the association of age with DNAm in tissues such as brain, skin, immune cells, fat, and liver have identified tissue-specific and non-specific effects, thus, motivating additional studies of diverse human tissue and cell types. RESULTS Here, we performed an epigenome-wide association study, leveraging DNAm data (Illumina EPIC array) from 961 tissue samples representing 9 tissue types (breast, lung, colon, ovary, prostate, skeletal muscle, testis, whole blood, and kidney) from the Genotype-Tissue Expression (GTEx) project. We identified age-associated CpG sites (false discovery rate < 0.05) in 8 tissues (all except skeletal muscle, n = 47). This included 162,002 unique hypermethylated and 90,626 hypomethylated CpG sites across all tissue types, with 130,137 (80%) hypermethylated CpGs and 74,703 (82%) hypomethylated CpG sites observed in a single tissue type. While the majority of age-associated CpG sites appeared tissue-specific, the patterns of enrichment among genomic features, such as chromatin states and CpG islands, were similar across most tissues, suggesting common mechanisms underlying cellular aging. Consistent with previous findings, we observed that hypermethylated CpG sites are enriched in regions with repressed polycomb signatures and CpG islands, while hypomethylated CpG sites preferentially occurred in non-CpG islands and enhancers. To gain insights into the functional effects of age-related DNAm changes, we assessed the correlation between DNAm and local gene expression changes to identify age-related expression quantitative trait methylation (age-eQTMs). We identified several age-eQTMs present in multiple tissue-types, including in the CDKN2A, HENMT1, and VCWE regions. CONCLUSION Overall, our findings will aid future efforts to develop biomarkers of aging and understand mechanisms of aging in diverse human tissue types.
Collapse
Affiliation(s)
- Niyati Jain
- Department of Public Health Sciences, University of Chicago, Chicago, IL, 60637, USA
- Committee on Genetics, Genomics and Systems Biology, University of Chicago, Chicago, IL, 60637, USA
| | - James L Li
- Department of Public Health Sciences, University of Chicago, Chicago, IL, 60637, USA
- Interdisciplinary Scientist Training Program, University of Chicago, Chicago, IL, 60637, USA
| | - Lin Tong
- Department of Public Health Sciences, University of Chicago, Chicago, IL, 60637, USA
| | - Farzana Jasmine
- Institute for Population and Precision Health (IPPH), Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Muhammad G Kibriya
- Department of Public Health Sciences, University of Chicago, Chicago, IL, 60637, USA
| | - Kathryn Demanelis
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15232, USA
| | - Meritxell Oliva
- Department of Public Health Sciences, University of Chicago, Chicago, IL, 60637, USA
- Genomics Research Center, AbbVie, North Chicago, IL, 60064, USA
| | - Lin S Chen
- Department of Public Health Sciences, University of Chicago, Chicago, IL, 60637, USA
| | - Brandon L Pierce
- Department of Public Health Sciences, University of Chicago, Chicago, IL, 60637, USA.
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA.
- Comprehensive Cancer Center, University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
11
|
Mahmud S, Pitcher LE, Torbenson E, Robbins PD, Zhang L, Dong X. Developing transcriptomic signatures as a biomarker of cellular senescence. Ageing Res Rev 2024; 99:102403. [PMID: 38964507 PMCID: PMC11338099 DOI: 10.1016/j.arr.2024.102403] [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: 12/18/2023] [Revised: 06/07/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Cellular senescence is a cell fate driven by different types of stress, where damaged cells exit from the cell cycle and, in many cases, develop an inflammatory senescence-associated secretory phenotype (SASP). Senescence has often been linked to driving aging and the onset of multiple diseases conferred by the harmful SASP, which disrupts tissue homeostasis and impairs the regular function of many tissues. This phenomenon was first observed in vitro when fibroblasts halted replication after approximately 50 population doublings. In addition to replication-induced senescence, factors such as DNA damage and oncogene activation can induce cellular senescence both in culture and in vivo. Despite their contribution to aging and disease, identifying senescent cells in vivo has been challenging due to their heterogeneity. Although senescent cells can express the cell cycle inhibitors p16Ink4a and/or p21Cip1 and exhibit SA-ß-gal activity and evidence of a DNA damage response, there is no universal biomarker for these cells, regardless of inducer or cell type. Recent studies have analyzed the transcriptomic characteristics of these cells, leading to the identification of signature gene sets like CellAge, SeneQuest, and SenMayo. Advancements in single-cell and spatial RNA sequencing now allow for analyzing senescent cell heterogeneity within the same tissue and the development of machine learning algorithms, e.g., SenPred, SenSig, and SenCID, to discover cellular senescence using RNA sequencing data. Such insights not only deepen our understanding of the genetic pathways driving cellular senescence, but also promote the development of its quantifiable biomarkers. This review summarizes the current knowledge of transcriptomic signatures of cellular senescence and their potential as in vivo biomarkers.
Collapse
Affiliation(s)
- Shamsed Mahmud
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Twin Cities, 420 Washington Avenue SE, Minneapolis, MN 55455, USA; Department of Genetics, Cell Biology, and Development, University of Minnesota, Twin Cities, 420 Washington Avenue SE, Minneapolis, MN 55455, USA
| | - Louise E Pitcher
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Twin Cities, 420 Washington Avenue SE, Minneapolis, MN 55455, USA; Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Twin Cities, 420 Washington Avenue SE, Minneapolis, MN 55455, USA
| | - Elijah Torbenson
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Twin Cities, 420 Washington Avenue SE, Minneapolis, MN 55455, USA; Department of Genetics, Cell Biology, and Development, University of Minnesota, Twin Cities, 420 Washington Avenue SE, Minneapolis, MN 55455, USA
| | - Paul D Robbins
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Twin Cities, 420 Washington Avenue SE, Minneapolis, MN 55455, USA; Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Twin Cities, 420 Washington Avenue SE, Minneapolis, MN 55455, USA
| | - Lei Zhang
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Twin Cities, 420 Washington Avenue SE, Minneapolis, MN 55455, USA; Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Twin Cities, 420 Washington Avenue SE, Minneapolis, MN 55455, USA
| | - Xiao Dong
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Twin Cities, 420 Washington Avenue SE, Minneapolis, MN 55455, USA; Department of Genetics, Cell Biology, and Development, University of Minnesota, Twin Cities, 420 Washington Avenue SE, Minneapolis, MN 55455, USA.
| |
Collapse
|
12
|
Hense JD, Isola JVV, Garcia DN, Magalhães LS, Masternak MM, Stout MB, Schneider A. The role of cellular senescence in ovarian aging. NPJ AGING 2024; 10:35. [PMID: 39033161 PMCID: PMC11271274 DOI: 10.1038/s41514-024-00157-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 05/29/2024] [Indexed: 07/23/2024]
Abstract
This review explores the relationship between ovarian aging and senescent cell accumulation, as well as the efficacy of senolytics to improve reproductive longevity. Reproductive longevity is determined by the age-associated decline in ovarian reserve, resulting in reduced fertility and eventually menopause. Cellular senescence is a state of permanent cell cycle arrest and resistance to apoptosis. Senescent cells accumulate in several tissues with advancing age, thereby promoting chronic inflammation and age-related diseases. Ovaries also appear to accumulate senescent cells with age, which might contribute to aging of the reproductive system and whole organism through SASP production. Importantly, senolytic drugs can eliminate senescent cells and may present a potential intervention to mitigate ovarian aging. Herein, we review the current literature related to the efficacy of senolytic drugs for extending the reproductive window in mice.
Collapse
Affiliation(s)
- Jéssica D Hense
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Nutrition College, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - José V V Isola
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Driele N Garcia
- Nutrition College, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | | | - Michal M Masternak
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| | - Michael B Stout
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Augusto Schneider
- Nutrition College, Universidade Federal de Pelotas, Pelotas, RS, Brazil.
| |
Collapse
|
13
|
Duarte LF, Villalobos V, Farías MA, Rangel-Ramírez MA, González-Madrid E, Navarro AJ, Carbone-Schellman J, Domínguez A, Alvarez A, Riedel CA, Bueno SM, Kalergis AM, Cáceres M, González PA. Asymptomatic herpes simplex virus brain infection elicits cellular senescence phenotypes in the central nervous system of mice suffering multiple sclerosis-like disease. Commun Biol 2024; 7:811. [PMID: 38965360 PMCID: PMC11224417 DOI: 10.1038/s42003-024-06486-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/21/2024] [Indexed: 07/06/2024] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a demyelinating disease affecting the central nervous system (CNS) in animals that parallels several clinical and molecular traits of multiple sclerosis in humans. Herpes simplex virus type 1 (HSV-1) infection mainly causes cold sores and eye diseases, yet eventually, it can also reach the CNS, leading to acute encephalitis. Notably, a significant proportion of healthy individuals are likely to have asymptomatic HSV-1 brain infection with chronic brain inflammation due to persistent latent infection in neurons. Because cellular senescence is suggested as a potential factor contributing to the development of various neurodegenerative disorders, including multiple sclerosis, and viral infections may induce a premature senescence state in the CNS, potentially increasing susceptibility to such disorders, here we examine the presence of senescence-related markers in the brains and spinal cords of mice with asymptomatic HSV-1 brain infection, EAE, and both conditions. Across all scenarios, we find a significant increases of senescence biomarkers in the CNS with some differences depending on the analyzed group. Notably, some senescence biomarkers are exclusively observed in mice with the combined conditions. These results indicate that asymptomatic HSV-1 brain infection and EAE associate with a significant expression of senescence biomarkers in the CNS.
Collapse
MESH Headings
- Animals
- Cellular Senescence
- Mice
- Brain/virology
- Brain/pathology
- Brain/metabolism
- Multiple Sclerosis/virology
- Multiple Sclerosis/pathology
- Multiple Sclerosis/metabolism
- Herpesvirus 1, Human/physiology
- Herpesvirus 1, Human/pathogenicity
- Herpes Simplex/virology
- Herpes Simplex/pathology
- Female
- Mice, Inbred C57BL
- Encephalomyelitis, Autoimmune, Experimental/virology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Phenotype
- Central Nervous System/virology
- Central Nervous System/metabolism
- Central Nervous System/pathology
- Spinal Cord/virology
- Spinal Cord/metabolism
- Spinal Cord/pathology
- Biomarkers/metabolism
- Encephalitis, Herpes Simplex/virology
- Encephalitis, Herpes Simplex/pathology
- Encephalitis, Herpes Simplex/metabolism
Collapse
Affiliation(s)
- Luisa F Duarte
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Ciencias Biológicas, Facultad de Ciencias de La Vida, Universidad Andrés Bello, Santiago, Chile
- Centro de Medicina Regenerativa, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Verónica Villalobos
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Mónica A Farías
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ma Andreina Rangel-Ramírez
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Ciencias Biológicas, Facultad de Ciencias de La Vida, Universidad Andrés Bello, Santiago, Chile
| | - Enrique González-Madrid
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Ciencias Biológicas, Facultad de Ciencias de La Vida, Universidad Andrés Bello, Santiago, Chile
| | - Areli J Navarro
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Javier Carbone-Schellman
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Angélica Domínguez
- Departamento de Salud Pública, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandra Alvarez
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A Riedel
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Ciencias Biológicas, Facultad de Ciencias de La Vida, Universidad Andrés Bello, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mónica Cáceres
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
14
|
Suryadevara V, Hudgins AD, Rajesh A, Pappalardo A, Karpova A, Dey AK, Hertzel A, Agudelo A, Rocha A, Soygur B, Schilling B, Carver CM, Aguayo-Mazzucato C, Baker DJ, Bernlohr DA, Jurk D, Mangarova DB, Quardokus EM, Enninga EAL, Schmidt EL, Chen F, Duncan FE, Cambuli F, Kaur G, Kuchel GA, Lee G, Daldrup-Link HE, Martini H, Phatnani H, Al-Naggar IM, Rahman I, Nie J, Passos JF, Silverstein JC, Campisi J, Wang J, Iwasaki K, Barbosa K, Metis K, Nernekli K, Niedernhofer LJ, Ding L, Wang L, Adams LC, Ruiyang L, Doolittle ML, Teneche MG, Schafer MJ, Xu M, Hajipour M, Boroumand M, Basisty N, Sloan N, Slavov N, Kuksenko O, Robson P, Gomez PT, Vasilikos P, Adams PD, Carapeto P, Zhu Q, Ramasamy R, Perez-Lorenzo R, Fan R, Dong R, Montgomery RR, Shaikh S, Vickovic S, Yin S, Kang S, Suvakov S, Khosla S, Garovic VD, Menon V, Xu Y, Song Y, Suh Y, Dou Z, Neretti N. SenNet recommendations for detecting senescent cells in different tissues. Nat Rev Mol Cell Biol 2024:10.1038/s41580-024-00738-8. [PMID: 38831121 DOI: 10.1038/s41580-024-00738-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2024] [Indexed: 06/05/2024]
Abstract
Once considered a tissue culture-specific phenomenon, cellular senescence has now been linked to various biological processes with both beneficial and detrimental roles in humans, rodents and other species. Much of our understanding of senescent cell biology still originates from tissue culture studies, where each cell in the culture is driven to an irreversible cell cycle arrest. By contrast, in tissues, these cells are relatively rare and difficult to characterize, and it is now established that fully differentiated, postmitotic cells can also acquire a senescence phenotype. The SenNet Biomarkers Working Group was formed to provide recommendations for the use of cellular senescence markers to identify and characterize senescent cells in tissues. Here, we provide recommendations for detecting senescent cells in different tissues based on a comprehensive analysis of existing literature reporting senescence markers in 14 tissues in mice and humans. We discuss some of the recent advances in detecting and characterizing cellular senescence, including molecular senescence signatures and morphological features, and the use of circulating markers. We aim for this work to be a valuable resource for both seasoned investigators in senescence-related studies and newcomers to the field.
Collapse
Affiliation(s)
- Vidyani Suryadevara
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, School of Medicine, Stanford, CA, USA
| | - Adam D Hudgins
- Department of Obstetrics and Gynecology, Columbia University, New York, NY, USA
| | - Adarsh Rajesh
- Sanford Burnham Prebys Medical Discovery Institute, Cancer Genome and Epigenetics Program, La Jolla, CA, USA
| | | | - Alla Karpova
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Amit K Dey
- National Institute on Aging, NIH, Baltimore, MD, USA
| | - Ann Hertzel
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Anthony Agudelo
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, USA
- Center on the Biology of Aging, Brown University, Providence, RI, USA
| | - Azucena Rocha
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, USA
- Center on the Biology of Aging, Brown University, Providence, RI, USA
| | - Bikem Soygur
- The Buck Institute for Research on Aging, Novato, CA, USA
| | | | - Chase M Carver
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
| | - Cristina Aguayo-Mazzucato
- Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center, Harvard Medical School, Boston, USA
| | - Darren J Baker
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - David A Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Diana Jurk
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Dilyana B Mangarova
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, School of Medicine, Stanford, CA, USA
| | - Ellen M Quardokus
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| | | | - Elizabeth L Schmidt
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Feng Chen
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Francesca E Duncan
- The Buck Institute for Research on Aging, Novato, CA, USA
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Gagandeep Kaur
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - George A Kuchel
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT, USA
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT, USA
| | - Gung Lee
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
| | - Heike E Daldrup-Link
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, School of Medicine, Stanford, CA, USA
| | - Helene Martini
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
| | - Hemali Phatnani
- New York Genome Center, New York, NY, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Iman M Al-Naggar
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT, USA
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Jia Nie
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - João F Passos
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
| | - Jonathan C Silverstein
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Judith Campisi
- The Buck Institute for Research on Aging, Novato, CA, USA
| | - Julia Wang
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Kanako Iwasaki
- Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center, Harvard Medical School, Boston, USA
| | - Karina Barbosa
- Sanford Burnham Prebys Medical Discovery Institute, Cancer Genome and Epigenetics Program, La Jolla, CA, USA
| | - Kay Metis
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kerem Nernekli
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, School of Medicine, Stanford, CA, USA
| | - Laura J Niedernhofer
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Li Ding
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Lichao Wang
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT, USA
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT, USA
| | - Lisa C Adams
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, School of Medicine, Stanford, CA, USA
| | - Liu Ruiyang
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Madison L Doolittle
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
- Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic, Rochester, MN, USA
| | - Marcos G Teneche
- Sanford Burnham Prebys Medical Discovery Institute, Cancer Genome and Epigenetics Program, La Jolla, CA, USA
| | - Marissa J Schafer
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Ming Xu
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT, USA
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT, USA
| | - Mohammadjavad Hajipour
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), Stanford University, School of Medicine, Stanford, CA, USA
| | | | | | - Nicholas Sloan
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Nikolai Slavov
- Center on the Biology of Aging, Brown University, Providence, RI, USA
- Department of Bioengineering, Northeastern University, Boston, MA, USA
- Department of Biology, Northeastern University, Boston, MA, USA
- Barnett Institute for Chemical and Biological Analysis, Northeastern University, Boston, MA, USA
| | - Olena Kuksenko
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Paul Robson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Institute for Systems Genomics, University of Connecticut, Farmington, CT, USA
| | - Paul T Gomez
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
| | - Periklis Vasilikos
- Department of Genetics and Development, Columbia University, New York, NY, USA
| | - Peter D Adams
- Sanford Burnham Prebys Medical Discovery Institute, Cancer Genome and Epigenetics Program, La Jolla, CA, USA
| | - Priscila Carapeto
- Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center, Harvard Medical School, Boston, USA
| | - Quan Zhu
- Center for Epigenomics, University of California, San Diego, CA, USA
| | | | | | - Rong Fan
- Yale-Center for Research on Aging, Yale School of Medicine, New Haven, CT, USA
| | - Runze Dong
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Graduate Program in Biological Physics, Structure and Design, University of Washington, Seattle, WA, USA
| | - Ruth R Montgomery
- Yale-Center for Research on Aging, Yale School of Medicine, New Haven, CT, USA
| | - Sadiya Shaikh
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Sanja Vickovic
- New York Genome Center, New York, NY, USA
- Herbert Irving Institute for Cancer Dynamics, Columbia University, New York, NY, USA
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Beijer Laboratory for Gene and Neuro Research, Uppsala University, Uppsala, Sweden
| | - Shanshan Yin
- Sanford Burnham Prebys Medical Discovery Institute, Cancer Genome and Epigenetics Program, La Jolla, CA, USA
| | - Shoukai Kang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Sonja Suvakov
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Sundeep Khosla
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Rochester, MN, USA
- Division of Endocrinology, Diabetes and Metabolism, Mayo Clinic, Rochester, MN, USA
| | - Vesna D Garovic
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, USA
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Vilas Menon
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Translational and Computational Neuroimmunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Yanxin Xu
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yizhe Song
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Yousin Suh
- Department of Obstetrics and Gynecology, Columbia University, New York, NY, USA
- Department of Genetics and Development, Columbia University, New York, NY, USA
| | - Zhixun Dou
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Nicola Neretti
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, USA.
- Center on the Biology of Aging, Brown University, Providence, RI, USA.
| |
Collapse
|
15
|
Abdelgawad IY, George B, Grant MKO, Huang Y, Shan Y, Huang RS, Zordoky BN. Sex-related differences in delayed doxorubicin-induced cardiac dysfunction in C57BL/6 mice. Arch Toxicol 2024; 98:1191-1208. [PMID: 38244039 DOI: 10.1007/s00204-023-03678-y] [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: 11/10/2023] [Accepted: 12/20/2023] [Indexed: 01/22/2024]
Abstract
Cancer survivors may experience long-term cardiovascular complications due to chemotherapeutic drugs such as doxorubicin (DOX). The exact mechanism of delayed DOX-induced cardiotoxicity has not been fully elucidated. Sex is an important risk factor for DOX-induced cardiotoxicity. In the current study, we identified sex differences in delayed DOX-induced cardiotoxicity and determined the underlying molecular determinants of the observed sexual dimorphism. Five-week-old male and female mice were administered intraperitoneal injections of DOX (4 mg/kg/week) or saline for 6 weeks. Echocardiography was performed 5 weeks after the last dose of DOX to evaluate cardiac function. Thereafter, mice were sacrificed and gene expression of markers of apoptosis, senescence, and inflammation was measured by PCR in hearts and livers. Proteomic profiling of the heart from both sexes was conducted to determine differentially expressed proteins (DEPs). Only DOX-treated male, but not female, mice demonstrated cardiac dysfunction, cardiac atrophy, and upregulated cardiac expression of Nppb and Myh7. No sex-related differences were observed in DOX-induced expression of most apoptotic, senescence, and pro-inflammatory markers. However, the gene expression of Trp53 was significantly reduced in hearts of DOX-treated female mice only. The anti-inflammatory marker Il-10 was significantly reduced in hearts of DOX-treated male mice only, while the pro-inflammatory marker Il-1α was significantly reduced in livers of DOX-treated female mice only. Gene expression of Tnf-α was reduced in hearts of both DOX-treated male and female mice. Proteomic analysis identified several DEPs after DOX treatment in a sex-specific manner, including anti-inflammatory acute phase proteins. This is the first study to assess sex-specific proteomic changes in a mouse model of delayed DOX-induced cardiotoxicity. Our proteomic analysis identified several sexually dimorphic DEPs, many of which are associated with the anti-inflammatory marker Il-10.
Collapse
Affiliation(s)
- Ibrahim Y Abdelgawad
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN, 55455, USA
| | - Benu George
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN, 55455, USA
| | - Marianne K O Grant
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN, 55455, USA
| | - Yingbo Huang
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN, 55455, USA
| | - Yuting Shan
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN, 55455, USA
| | - R Stephanie Huang
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN, 55455, USA
| | - Beshay N Zordoky
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN, 55455, USA.
| |
Collapse
|
16
|
Ham M, Cho Y, Kang T, Oh T, Kim H, Kim K. Transcriptome-wide analysis reveals GYG2 as a mitochondria-related aging biomarker in human subcutaneous adipose tissue. Aging Cell 2024; 23:e14049. [PMID: 38062989 PMCID: PMC10861210 DOI: 10.1111/acel.14049] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 11/05/2023] [Accepted: 11/13/2023] [Indexed: 02/15/2024] Open
Abstract
Subcutaneous adipose tissue (SAT), a vital energy reservoir and endocrine organ for maintaining systemic glucose, lipid, and energy homeostasis, undergoes significant changes with age. However, among the existing aging-related markers, only few genes are associated with SAT aging. In this study, weighted gene co-expression network analysis was used on a transcriptome of SAT obtained from the Genotype-Tissue Expression portal to identify biologically relevant, SAT-specific, and age-related marker genes. We found modules that exhibited significant changes with age and identified GYG2 as a novel key aging associated gene. The link between GYG2 and mitochondrial function as well as brown/beige adipocytes was supported using additional bioinformatics and experimental analyses. Additionally, we identified PPARG as the transcription factor of GYG2 expression. The newly discovered GYG2 marker can be used to not only determine the age of SAT but also uncover new mechanisms underlying SAT aging.
Collapse
Affiliation(s)
- Mira Ham
- R&I Unit, Amorepacific CorporationGyeonggi‐doKorea
| | - Yeonju Cho
- R&I Unit, Amorepacific CorporationGyeonggi‐doKorea
| | - Tae‐Wook Kang
- Department of BioinformaticsThe Moagen Inc.DaejeonKorea
| | - Taeyun Oh
- Department of Internal Medicine, Institute of GastroenterologyYonsei University College of MedicineSeoulKorea
| | | | - Kyu‐Han Kim
- R&I Unit, Amorepacific CorporationGyeonggi‐doKorea
| |
Collapse
|
17
|
Orr ME. A Need for Refined Senescence Biomarkers and Measures of Senolytics in the Brain. J Alzheimers Dis 2024; 98:411-415. [PMID: 38461508 PMCID: PMC11063734 DOI: 10.3233/jad-231462] [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: 03/12/2024]
Abstract
Cellular senescence contributes to Alzheimer's disease (AD) pathogenesis. Treatments that remove senescent cells, senolytics, improve brain outcomes in AD mice with amyloid-β or tau deposition. 3xTgAD mice develop both AD neuropathologies; however, Ng et al. report low p16INK4a-associated senescence in the brain. Senolytic treatment by genetic removal; dasatinib with quercetin (D+Q), which enter the brain; and ABT-263 with limited brain penetrance all reduced AD neuropathology. Refined measures of senescence and brain exposure would help clarify the benefits of senolytics despite low p16INK4a-associated senescence and potential limited brain penetrance.
Collapse
Affiliation(s)
- Miranda E. Orr
- Department of Internal Medicine Section on Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Salisbury VA Medical Center, Salisbury, NC, USA
| |
Collapse
|
18
|
Scieszka D, Bolt AM, McCormick MA, Brigman JL, Campen MJ. Aging, longevity, and the role of environmental stressors: a focus on wildfire smoke and air quality. FRONTIERS IN TOXICOLOGY 2023; 5:1267667. [PMID: 37900096 PMCID: PMC10600394 DOI: 10.3389/ftox.2023.1267667] [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: 07/26/2023] [Accepted: 09/25/2023] [Indexed: 10/31/2023] Open
Abstract
Aging is a complex biological process involving multiple interacting mechanisms and is being increasingly linked to environmental exposures such as wildfire smoke. In this review, we detail the hallmarks of aging, emphasizing the role of telomere attrition, cellular senescence, epigenetic alterations, proteostasis, genomic instability, and mitochondrial dysfunction, while also exploring integrative hallmarks - altered intercellular communication and stem cell exhaustion. Within each hallmark of aging, our review explores how environmental disasters like wildfires, and their resultant inhaled toxicants, interact with these aging mechanisms. The intersection between aging and environmental exposures, especially high-concentration insults from wildfires, remains under-studied. Preliminary evidence, from our group and others, suggests that inhaled wildfire smoke can accelerate markers of neurological aging and reduce learning capabilities. This is likely mediated by the augmentation of circulatory factors that compromise vascular and blood-brain barrier integrity, induce chronic neuroinflammation, and promote age-associated proteinopathy-related outcomes. Moreover, wildfire smoke may induce a reduced metabolic, senescent cellular phenotype. Future interventions could potentially leverage combined anti-inflammatory and NAD + boosting compounds to counter these effects. This review underscores the critical need to study the intricate interplay between environmental factors and the biological mechanisms of aging to pave the way for effective interventions.
Collapse
Affiliation(s)
- David Scieszka
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Alicia M. Bolt
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Mark A. McCormick
- Department of Biochemistry and Molecular Biology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Jonathan L. Brigman
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Matthew J. Campen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| |
Collapse
|
19
|
Kumar A, Thirumurugan K. Understanding cellular senescence: pathways involved, therapeutics and longevity aiding. Cell Cycle 2023; 22:2324-2345. [PMID: 38031713 PMCID: PMC10730163 DOI: 10.1080/15384101.2023.2287929] [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: 02/09/2023] [Accepted: 11/06/2023] [Indexed: 12/01/2023] Open
Abstract
A normal somatic cell undergoes cycles of finite cellular divisions. The presence of surveillance checkpoints arrests cell division in response to stress inducers: oxidative stress from excess free radicals, oncogene-induced abnormalities, genotoxic stress, and telomere attrition. When facing such stress when undergoing these damages, there is a brief pause in the cell cycle to enable repair mechanisms. Also, the nature of stress determines whether the cell goes for repair or permanent arrest. As the cells experience transient or permanent stress, they subsequently choose the quiescence or senescence stage, respectively. Quiescence is an essential stage that allows the arrested/damaged cells to go through appropriate repair mechanisms and then revert to the mainstream cell cycle. However, senescent cells are irreversible and accumulate with age, resulting in inflammation and various age-related disorders. In this review, we focus on senescence-associated pathways and therapeutics understanding cellular senescence as a cascade that leads to aging, while discussing the recent details on the molecular pathways involved in regulating senescence and the benefits of therapeutic strategies against accumulated senescent cells and their secretions.
Collapse
Affiliation(s)
- Ashish Kumar
- Pearl Research Park, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Kavitha Thirumurugan
- Pearl Research Park, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| |
Collapse
|
20
|
Fang Y, Medina D, Stockwell R, McFadden S, Quinn K, Peck MR, Bartke A, Hascup KN, Hascup ER. Sexual dimorphic metabolic and cognitive responses of C57BL/6 mice to Fisetin or Dasatinib and quercetin cocktail oral treatment. GeroScience 2023; 45:2835-2850. [PMID: 37296266 PMCID: PMC10643448 DOI: 10.1007/s11357-023-00843-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Senolytic treatment in aged mice clears senescent cell burden leading to functional improvements. However, less is known regarding the effects of these compounds when administered prior to significant senescent cell accumulation. From 4-13 months of age, C57BL/6 male and female mice received monthly oral dosing of either 100 mg/kg Fisetin or a 5 mg/kg Dasatinib (D) plus 50 mg/kg Quercetin (Q) cocktail. During treatment, several aspects of healthy aging were assayed including glucose metabolism using an insulin and glucose tolerance test, cognitive performance using Morris water maze and novel object recognition, and energy metabolism using indirect calorimetry. Afterwards, mice were euthanized for plasma, tissue specific markers of senescence-associated secretory phenotype (SASP), and white adipose tissue accumulation (WAT). Sexually dimorphic treatment effects were observed. Fisetin treated male mice had reduced SASP, enhanced glucose and energy metabolism, improved cognitive performance, and increased mRNA expression of adiponectin receptor 1 and glucose transporter 4. D + Q treatment had minimal effects in male C57BL/6 mice, but was detrimental to females causing increased SASP expression along with accumulation of WAT depots. Reduced energy metabolism and cognitive performance were also noted. Fisetin treatment had no effect in female C57BL/6 mice potentially due to a slower rate of biological aging. In summary, the senolytic treatment in young adulthood, has beneficial, negligible, or detrimental effects in C57BL/6 mice dependent upon sex and treatment. These observations should serve as a note of caution in this rapidly evolving and expanding field of investigation. Male and female C57BL/6 mice were treated with once monthly oral doses of either Dasatinib (D) + Quercetin (Q) or Fisetin from 4-13 months of age. Males treated with Fisetin had reduced SASP markers (blue spheres) as well as improved metabolism (red flame) and cognition. Females treated with D + Q had increased adiposity and SASP markers (red spheres) along with decreased metabolism (blue flame) and cognitive performance. No effects were observed in females treated with Fisetin or males treated with D + Q.
Collapse
Affiliation(s)
- Yimin Fang
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - David Medina
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Robert Stockwell
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Sam McFadden
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Kathleen Quinn
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Mackenzie R Peck
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Kevin N Hascup
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA
| | - Erin R Hascup
- Department of Neurology, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA.
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, 62702, USA.
| |
Collapse
|
21
|
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
|
22
|
Jin J, Yang X, Gong H, Li X. Time- and Gender-Dependent Alterations in Mice during the Aging Process. Int J Mol Sci 2023; 24:12790. [PMID: 37628974 PMCID: PMC10454612 DOI: 10.3390/ijms241612790] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/01/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Compared to young people and adults, there are differences in the ability of elderly people to resist diseases or injuries, with some noticeable features being gender-dependent. However, gender differences in age-related viscera alterations are not clear. To evaluate a potential possibility of gender differences during the natural aging process, we used three age groups to investigate the impact on spleens, kidneys, and adrenal glands. The immunofluorescence results showed that male-specific p21 proteins were concentrated in the renal tubule epithelial cells of the kidney. Histological staining revealed an increase in the frequencies of fat vacuoles located in the renal tubule epithelial cells of the cortex, under the renal capsule in the kidneys of male mice with age. In female mice, we found that the width of the globular zone in the adrenal gland cortex was unchanged with age. On the contrary, the male displayed a reduction in width. Compared to females, the content of epinephrine in adrenal gland tissue according to ELISA analysis was higher in adults, and a greater decline was observed in aged males particularly. These data confirmed the age-dependent differences between female and male mice; therefore, gender should be considered one of the major factors for personalized treatment in clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Jing Jin
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China (H.G.)
| | - Xiaoquan Yang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China (H.G.)
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, HUST-Suzhou Institute for Brainsmatics, JITRI, Chinese Academy of Medical Sciences, Suzhou 215004, China
| | - Hui Gong
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China (H.G.)
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, HUST-Suzhou Institute for Brainsmatics, JITRI, Chinese Academy of Medical Sciences, Suzhou 215004, China
| | - Xiangning Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan 430074, China (H.G.)
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, HUST-Suzhou Institute for Brainsmatics, JITRI, Chinese Academy of Medical Sciences, Suzhou 215004, China
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou 570228, China
| |
Collapse
|
23
|
Holloway K, Neherin K, Dam KU, Zhang H. Cellular senescence and neurodegeneration. Hum Genet 2023; 142:1247-1262. [PMID: 37115318 DOI: 10.1007/s00439-023-02565-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023]
Abstract
Advancing age is a major risk factor of Alzheimer's disease (AD). The worldwide prevalence of AD is approximately 50 million people, and this number is projected to increase substantially. The molecular mechanisms underlying the aging-associated susceptibility to cognitive impairment in AD are largely unknown. As a hallmark of aging, cellular senescence is a significant contributor to aging and age-related diseases including AD. Senescent neurons and glial cells have been detected to accumulate in the brains of AD patients and mouse models. Importantly, selective elimination of senescent cells ameliorates amyloid beta and tau pathologies and improves cognition in AD mouse models, indicating a critical role of cellular senescence in AD pathogenesis. Nonetheless, the mechanisms underlying when and how cellular senescence contributes to AD pathogenesis remain unclear. This review provides an overview of cellular senescence and discusses recent advances in the understanding of the impact of cellular senescence on AD pathogenesis, with brief discussions of the possible role of cellular senescence in other neurodegenerative diseases including Down syndrome, Parkinson's disease, multiple sclerosis, and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Kristopher Holloway
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA, 01655, USA
| | - Kashfia Neherin
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA, 01655, USA
| | - Kha Uyen Dam
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA, 01655, USA
| | - Hong Zhang
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA, 01655, USA.
| |
Collapse
|
24
|
Nousis L, Kanavaros P, Barbouti A. Oxidative Stress-Induced Cellular Senescence: Is Labile Iron the Connecting Link? Antioxidants (Basel) 2023; 12:1250. [PMID: 37371980 DOI: 10.3390/antiox12061250] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Cellular senescence, a cell state characterized by a generally irreversible cell cycle arrest, is implicated in various physiological processes and a wide range of age-related pathologies. Oxidative stress, a condition caused by an imbalance between the production and the elimination of reactive oxygen species (ROS) in cells and tissues, is a common driver of cellular senescence. ROS encompass free radicals and other molecules formed as byproducts of oxygen metabolism, which exhibit varying chemical reactivity. A prerequisite for the generation of strong oxidizing ROS that can damage macromolecules and impair cellular function is the availability of labile (redox-active) iron, which catalyzes the formation of highly reactive free radicals. Targeting labile iron has been proven an effective strategy to counteract the adverse effects of ROS, but evidence concerning cellular senescence is sparse. In the present review article, we discuss aspects of oxidative stress-induced cellular senescence, with special attention to the potential implication of labile iron.
Collapse
Affiliation(s)
- Lambros Nousis
- Department of Hygiene and Epidemiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Panagiotis Kanavaros
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Alexandra Barbouti
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| |
Collapse
|
25
|
Liao KM, Chen CJ, Luo WJ, Hsu CW, Yu SL, Yang PC, Su KY. Senomorphic effect of diphenyleneiodonium through AMPK/MFF/DRP1 mediated mitochondrial fission. Biomed Pharmacother 2023; 162:114616. [PMID: 37004322 DOI: 10.1016/j.biopha.2023.114616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 03/22/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
With an aging population and the numerous health impacts associated with old age, the identification of anti-aging drugs has become an important new research direction. Although mitochondria have been recognized to affect aging, anti-aging drugs specifically targeting the mitochondria are less well characterized. In this study, diphenyleneiodonium (DPI) was identified as a potential senomorphic drug that functions by promoting mitochondrial fission. DPI significantly reduced the number of senescence-associated β-galactosidase (SA-β-gal) positive cells and increased the number of proliferating Ki-67 positive cells in BrdU or irradiation stress-induced senescent NIH3T3 cells or IMR90 cells and mouse embryonic fibroblasts (MEFs) replicative senescent cells. Cell cycle arrest genes and senescence-associated secretory phenotype (SASP) factors were downregulated with DPI treatment. In addition, the oxygen consumption rate (OCR) of mitochondrial respiration showed that DPI significantly reduced senescence-associated hyper OCR. Mechanistically, DPI promoted mitochondrial fission by enhancing AMPK/MFF phosphorylation and DRP1 mitochondrial translocation. Inhibition of DRP1 by Mdivi-1 abolished DPI-induced mitochondrial fission and the anti-senescence phenotype. Importantly, Eighty-eight-week-old mice treated with DPI had significantly reduced numbers of SA-β-gal positive cells and reduced expression of cell cycle arrest genes and SASP factors in their livers and kidneys. Pathological and functional assays showed DPI treatment not only reduced liver fibrosis and immune cell infiltration but also improved aged-related physical impairments in aged mice. Taken together, our study identified a potential anti-aging compound that exerts its effects through modulation of mitochondrial morphology.
Collapse
|
26
|
Suda M, Paul KH, Minamino T, Miller JD, Lerman A, Ellison-Hughes GM, Tchkonia T, Kirkland JL. Senescent Cells: A Therapeutic Target in Cardiovascular Diseases. Cells 2023; 12:1296. [PMID: 37174697 PMCID: PMC10177324 DOI: 10.3390/cells12091296] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/27/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023] Open
Abstract
Senescent cell accumulation has been observed in age-associated diseases including cardiovascular diseases. Senescent cells lack proliferative capacity and secrete senescence-associated secretory phenotype (SASP) factors that may cause or worsen many cardiovascular diseases. Therapies targeting senescent cells, especially senolytic drugs that selectively induce senescent cell removal, have been shown to delay, prevent, alleviate, or treat multiple age-associated diseases in preclinical models. Some senolytic clinical trials have already been completed or are underway for a number of diseases and geriatric syndromes. Understanding how cellular senescence affects the various cell types in the cardiovascular system, such as endothelial cells, vascular smooth muscle cells, fibroblasts, immune cells, progenitor cells, and cardiomyocytes, is important to facilitate translation of senotherapeutics into clinical interventions. This review highlights: (1) the characteristics of senescent cells and their involvement in cardiovascular diseases, focusing on the aforementioned cardiovascular cell types, (2) evidence about senolytic drugs and other senotherapeutics, and (3) the future path and clinical potential of senotherapeutics for cardiovascular diseases.
Collapse
Affiliation(s)
- Masayoshi Suda
- Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Karl H. Paul
- Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
- Department of Physiology and Pharmacology, Karolinska Institutet, Solnavägen 9, 171 65 Solna, Sweden
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Japan Agency for Medical Research and Development-Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan
| | - Jordan D. Miller
- Division of Cardiovascular Surgery, Mayo Clinic College of Medicine, 200 First St., S.W., Rochester, MN 55905, USA
| | - Amir Lerman
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
| | - Georgina M. Ellison-Hughes
- Centre for Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, Faculty of Life Sciences & Medicine, Guy’s Campus, King’s College London, London SE1 1UL, UK
- Centre for Stem Cells and Regenerative Medicine, School of Basic and Medical Biosciences, Faculty of Life Sciences & Medicine, Guy’s Campus, King’s College London, London SE1 1UL, UK
| | - Tamar Tchkonia
- Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
| | - James L. Kirkland
- Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
| |
Collapse
|
27
|
Winsky-Sommerer R, King HA, Iadevaia V, Möller-Levet C, Gerber AP. A post-transcriptional regulatory landscape of aging in the female mouse hippocampus. Front Aging Neurosci 2023; 15:1119873. [PMID: 37122377 PMCID: PMC10135431 DOI: 10.3389/fnagi.2023.1119873] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/27/2023] [Indexed: 05/02/2023] Open
Abstract
Aging is associated with substantial physiological changes and constitutes a major risk factor for neurological disorders including dementia. Alterations in gene expression upon aging have been extensively studied; however, an in-depth characterization of post-transcriptional regulatory events remains elusive. Here, we profiled the age-related changes of the transcriptome and translatome in the female mouse hippocampus by RNA sequencing of total RNA and polysome preparations at four ages (3-, 6-, 12-, 20-month-old); and we implemented a variety of bioinformatics approaches to unravel alterations in transcript abundance, alternative splicing, and polyadenylation site selection. We observed mostly well-coordinated transcriptome and translatome expression signatures across age including upregulation of transcripts related to immune system processes and neuroinflammation, though transcripts encoding ribonucleoproteins or associated with mitochondrial functions, calcium signaling and the cell-cycle displayed substantial discordant profiles, suggesting translational control associated with age-related deficits in hippocampal-dependent behavior. By contrast, alternative splicing was less preserved, increased with age and was associated with distinct functionally-related transcripts encoding proteins acting at synapses/dendrites, RNA-binding proteins; thereby predicting regulatory roles for RBM3 and CIRBP. Only minor changes in polyadenylation site selection were identified, indicating pivotal 3'-end selection in young adults compared to older groups. Overall, our study provides a comprehensive resource of age-associated post-transcriptional regulatory events in the mouse hippocampus, enabling further examination of the molecular features underlying age-associated neurological diseases.
Collapse
Affiliation(s)
| | | | | | | | - André P. Gerber
- Faculty of Health and Medical Sciences, School of Biosciences and Medicine, University of Surrey, Guildford, Surrey, United Kingdom
| |
Collapse
|
28
|
Manna S, Mc Elwain CJ, Maher GM, Giralt Martín M, Musumeci A, McCarthy FP, McCarthy C. Heterogenous Differences in Cellular Senescent Phenotypes in Pre-Eclampsia and IUGR following Quantitative Assessment of Multiple Biomarkers of Senescence. Int J Mol Sci 2023; 24:ijms24043101. [PMID: 36834513 PMCID: PMC9963163 DOI: 10.3390/ijms24043101] [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: 11/15/2022] [Revised: 12/20/2022] [Accepted: 01/18/2023] [Indexed: 02/08/2023] Open
Abstract
Premature ageing of the placenta in pregnancy outcomes is associated with the persistent presence of oxidative stress and placental insufficiency reducing its functional capacity. In this study, we investigated cellular senescence phenotypes of pre-eclampsia and IUGR pregnancies by simultaneously measuring several biomarkers of senescence. Maternal plasma and placental samples were collected at term gestation from nulliparous women undergoing pre-labour elective caesarean section with pre-eclampsia without intrauterine growth restriction (PE; n = 5), pre-eclampsia associated with intrauterine growth restriction (n = 8), intrauterine growth restriction (IUGR < 10th centile; n = 6), and age-matched controls (n = 20). Placental absolute telomere length and senescence gene analysis was performed by RTqPCR. The expression of cyclin-dependent kinase inhibitors (p21 and p16) was determined by Western blot. Senescence-associated secretory phenotypes (SASPs) were evaluated in maternal plasma by multiplex ELISA assay. Placental expression of senescence-associated genes showed significant increases in CHEK1, PCNA, PTEN, CDKN2A, and CCNB-1 (p < 0.05) in pre-eclampsia, while TBX-2, PCNA, ATM, and CCNB-1 expression were evident (p < 0.05) and were significantly decreased in IUGR compared with controls. Placental p16 protein expression was significantly decreased in pre-eclampsia only compared with controls (p = 0.028). IL-6 was significantly increased in pre-eclampsia (0.54 pg/mL ± 0.271 vs. 0.3 pg/mL ± 0.102; p = 0.017) while IFN-γ was significantly increased in IUGR (4.6 pg/mL ± 2.2 vs. 2.17 pg/mL ± 0.8; p = 0.002) compared with controls. These results provide evidence of premature senescence in IUGR pregnancies, and while cell cycle checkpoint regulators are activated in pre-eclampsia, the cellular phenotype is one of cell repair and subsequent proliferation rather than progression to senescence. The heterogeneity of these cellular phenotypes highlights the complexity of characterising cellular senescence and may equally be indicative of the differing pathophysiological insults unique to each obstetric complication.
Collapse
Affiliation(s)
- Samprikta Manna
- Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, University College Cork, T12 YE02 Cork, Ireland
- INFANT Research Centre, University College Cork, T12 K8AF Cork, Ireland
| | - Colm J. Mc Elwain
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, T12 XF62 Cork, Ireland
| | - Gillian M. Maher
- INFANT Research Centre, University College Cork, T12 K8AF Cork, Ireland
- School of Public Health, Western Gateway Building, University College Cork, T12 XF62 Cork, Ireland
| | - Marta Giralt Martín
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, T12 XF62 Cork, Ireland
| | - Andrea Musumeci
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, T12 XF62 Cork, Ireland
| | - Fergus P. McCarthy
- Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, University College Cork, T12 YE02 Cork, Ireland
- INFANT Research Centre, University College Cork, T12 K8AF Cork, Ireland
- Correspondence:
| | - Cathal McCarthy
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, T12 XF62 Cork, Ireland
| |
Collapse
|
29
|
Perez K, Ciotlos S, McGirr J, Limbad C, Doi R, Nederveen JP, Nilsson MI, Winer DA, Evans W, Tarnopolsky M, Campisi J, Melov S. Single nuclei profiling identifies cell specific markers of skeletal muscle aging, frailty, and senescence. Aging (Albany NY) 2022; 14:9393-9422. [PMID: 36516485 PMCID: PMC9792217 DOI: 10.18632/aging.204435] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022]
Abstract
Aging is accompanied by a loss of muscle mass and function, termed sarcopenia, which causes numerous morbidities and economic burdens in human populations. Mechanisms implicated in age-related sarcopenia or frailty include inflammation, muscle stem cell depletion, mitochondrial dysfunction, and loss of motor neurons, but whether there are key drivers of sarcopenia are not yet known. To gain deeper insights into age-related muscle loss, we performed transcriptome profiling on lower limb muscle biopsies from 72 young, elderly, and frail human subjects using bulk RNA-seq (N = 72) and single-nuclei RNA-seq (N = 17). This combined approach revealed changes in gene expression that occur with age and frailty in multiple cell types comprising mature skeletal muscle. Notably, we found increased expression of the genes MYH8 and PDK4, and decreased expression of the gene IGFN1, in aged muscle. We validated several key genes changes in fixed human muscle tissue using digital spatial profiling. We also identified a small population of nuclei that express CDKN1A, present only in aged samples, consistent with p21cip1-driven senescence in this subpopulation. Overall, our findings identify unique cellular subpopulations in aged and sarcopenic skeletal muscle, which will facilitate the development of new therapeutic strategies to combat age-related frailty.
Collapse
Affiliation(s)
- Kevin Perez
- Buck Institute for Research on Aging, Novato, CA 94952, USA
| | - Serban Ciotlos
- Buck Institute for Research on Aging, Novato, CA 94952, USA
| | - Julia McGirr
- Buck Institute for Research on Aging, Novato, CA 94952, USA
| | | | - Ryosuke Doi
- Buck Institute for Research on Aging, Novato, CA 94952, USA
- Drug Discovery Research, Astellas Pharma, Tsukuba, Ibaraki, Japan
| | | | | | | | - William Evans
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720, USA
| | | | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA 94952, USA
| | - Simon Melov
- Buck Institute for Research on Aging, Novato, CA 94952, USA
| |
Collapse
|
30
|
Boe DM, Hulsebus HJ, Najarro KM, Mullen JE, Kim H, Tan AC, McMahan RH, Kovacs EJ. Advanced age is associated with changes in alveolar macrophages and their responses to the stress of traumatic injury. J Leukoc Biol 2022; 112:1371-1386. [PMID: 36120937 PMCID: PMC10150914 DOI: 10.1002/jlb.3hi0620-399rr] [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: 06/18/2020] [Revised: 08/14/2022] [Indexed: 01/04/2023] Open
Abstract
Alveolar macrophages (AMs) are tissue-resident cells of the lower airways that perform many homeostatic functions critical for pulmonary health and protection against pathogens. However, little is known about the factors that shape AMs during healthy aging. In these studies, we sought to characterize age-related changes in AM phenotype, function, and responses to a physiologic stressor, that is, distal injury. Age was associated with a wide range of changes in cell surface receptor and gene expression by AMs, reflecting a unique alternatively activated phenotype. AMs from aged mice also exhibited markers of cellular senescence along with down-regulation of genes involved in growth and cell cycle pathways relative to young controls. Furthermore, AMs from aged mice showed a stunted transcriptional response to distal injury compared with AMs from young mice. Many changes were found to involve glucocorticoid-regulated genes, and corticosteroid treatment of primary AMs ex vivo revealed diminished transcriptional responses in cells from aged animals. These results demonstrate that there is a complex age-dependent AM phenotype associated with dysregulated stress hormone signaling that may interfere with AM responses to physiologic stressors and could contribute to AM dysfunction and the decline of pulmonary immunity during healthy aging.
Collapse
Affiliation(s)
- Devin M. Boe
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Holly J. Hulsebus
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kevin M. Najarro
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Juliet E. Mullen
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Hyunmin Kim
- Department of Biostatistics and Bioinformatics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Aik Choon Tan
- Department of Biostatistics and Bioinformatics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, USA
| | - Rachel H. McMahan
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Elizabeth J. Kovacs
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
31
|
Boieri M, Marchese E, Pham QM, Azin M, Steidl LE, Malishkevich A, Demehri S. Thymic stromal lymphopoietin-stimulated CD4+ T cells induce senescence in advanced breast cancer. Front Cell Dev Biol 2022; 10:1002692. [PMID: 36467403 PMCID: PMC9714463 DOI: 10.3389/fcell.2022.1002692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/20/2022] [Indexed: 11/18/2022] Open
Abstract
Thymic Stromal Lymphopoietin (TSLP) plays a prominent role in inducing type 2 immune response, commonly associated with atopic diseases. TSLP-activated CD4+ T helper 2 cells block early carcinogenesis by inducing terminal differentiation in spontaneous breast and lung cancer models. However, the impact of TSLP induction on advanced cancer with altered cellular phenotypes is unclear. Using an established MMTV-PyMttg breast cancer cell line, we demonstrate that TSLP-stimulated CD4+ T cells possess an antitumor effect in advanced breast cancer. In contrast to early breast cancer suppression, the antitumor immunity mediated by TSLP-stimulated CD4+ T cells in advanced breast cancer is mediated by the induction of a senescent-like phenotype in cancer cells. Inflammatory CD4+ T cells drive breast cancer cells into senescence by releasing interferon-gamma and tumor necrosis factor-alpha, which directly bind to their receptors on cancer cells. Our findings reveal a novel mechanism of TSLP-activated CD4+ T cell immunity against advanced breast cancer, mediated by cellular senescence as a distinct effector mechanism for cancer immunotherapy.
Collapse
Affiliation(s)
- Margherita Boieri
- Center for Cancer Immunology, Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Emanuela Marchese
- Center for Cancer Immunology, Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Quan Minh Pham
- Center for Cancer Immunology, Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Marjan Azin
- Center for Cancer Immunology, Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Lauren E. Steidl
- Center for Cancer Immunology, Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Anna Malishkevich
- Center for Cancer Immunology, Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Shadmehr Demehri
- Center for Cancer Immunology, Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- *Correspondence: Shadmehr Demehri,
| |
Collapse
|
32
|
Wilkinson HN, Hardman MJ. Cellular Senescence in Acute and Chronic Wound Repair. Cold Spring Harb Perspect Biol 2022; 14:a041221. [PMID: 35817510 PMCID: PMC9620855 DOI: 10.1101/cshperspect.a041221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Cellular senescence, once thought an artifact of in vitro culture or passive outcome of aging, has emerged as fundamental to tissue development and function. The senescence mechanism importantly halts cell cycle progression to protect against tumor formation, while transiently present senescent cells produce a complex secretome (or SASP) of inflammatory mediators, proteases, and growth factors that guide developmental remodeling and tissue regeneration. Transiently present senescence is important for skin repair, where it accelerates extracellular matrix formation, limits fibrosis, promotes reepithelialization, and modulates inflammation. Unfortunately, advanced age and diabetes drive pathological accumulation of senescent cells in chronic wounds, which is perpetuated by a proinflammatory SASP, advanced glycation end-products, and oxidative damage. Although the biology of wound senescence remains incompletely understood, drugs that selectively target senescent cells are showing promise in clinical trials for diverse pathological conditions. It may not be long before senescence-targeted therapies will be available for the management, or perhaps even prevention, of chronic wounds.
Collapse
Affiliation(s)
- Holly N Wilkinson
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull HU6 7RX, United Kingdom
| | - Matthew J Hardman
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull HU6 7RX, United Kingdom
| |
Collapse
|
33
|
Heckenbach I, Mkrtchyan GV, Ezra MB, Bakula D, Madsen JS, Nielsen MH, Oró D, Osborne B, Covarrubias AJ, Idda ML, Gorospe M, Mortensen L, Verdin E, Westendorp R, Scheibye-Knudsen M. Nuclear morphology is a deep learning biomarker of cellular senescence. NATURE AGING 2022; 2:742-755. [PMID: 37118134 PMCID: PMC10154217 DOI: 10.1038/s43587-022-00263-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 07/01/2022] [Indexed: 04/30/2023]
Abstract
Cellular senescence is an important factor in aging and many age-related diseases, but understanding its role in health is challenging due to the lack of exclusive or universal markers. Using neural networks, we predict senescence from the nuclear morphology of human fibroblasts with up to 95% accuracy, and investigate murine astrocytes, murine neurons, and fibroblasts with premature aging in culture. After generalizing our approach, the predictor recognizes higher rates of senescence in p21-positive and ethynyl-2'-deoxyuridine (EdU)-negative nuclei in tissues and shows an increasing rate of senescent cells with age in H&E-stained murine liver tissue and human dermal biopsies. Evaluating medical records reveals that higher rates of senescent cells correspond to decreased rates of malignant neoplasms and increased rates of osteoporosis, osteoarthritis, hypertension and cerebral infarction. In sum, we show that morphological alterations of the nucleus can serve as a deep learning predictor of senescence that is applicable across tissues and species and is associated with health outcomes in humans.
Collapse
Affiliation(s)
- Indra Heckenbach
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
- Buck Institute for Research on Aging, Novato, CA, USA
- Tracked.bio, Copenhagen, Denmark
| | - Garik V Mkrtchyan
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Michael Ben Ezra
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
- Methods and Analysis, Statistics Denmark, Copenhagen, Denmark
| | - Daniela Bakula
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Jakob Sture Madsen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Brenna Osborne
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Anthony J Covarrubias
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - M Laura Idda
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Sassari, Italy
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Laust Mortensen
- Methods and Analysis, Statistics Denmark, Copenhagen, Denmark
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Rudi Westendorp
- Methods and Analysis, Statistics Denmark, Copenhagen, Denmark
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Morten Scheibye-Knudsen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark.
- Tracked.bio, Copenhagen, Denmark.
| |
Collapse
|
34
|
Lérida-Viso A, Estepa-Fernández A, Morellá-Aucejo Á, Lozano-Torres B, Alfonso M, Blandez JF, Bisbal V, Sepúlveda P, García-Fernández A, Orzáez M, Martínez-Máñez R. Pharmacological senolysis reduces doxorubicin-induced cardiotoxicity and improves cardiac function in mice. Pharmacol Res 2022; 183:106356. [PMID: 35843569 DOI: 10.1016/j.phrs.2022.106356] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/05/2022] [Accepted: 07/12/2022] [Indexed: 01/10/2023]
Abstract
Many anticancer agents used in clinics induce premature senescence in healthy tissues generating accelerated aging processes and adverse side-effects in patients. Cardiotoxicity is a well-known limiting factor of anticancer treatment with doxorubicin (DOX), a very effective anthracycline widely used as antitumoral therapy in clinical practice, that leads to long-term morbidity and mortality. DOX exposure severely affects the population of cardiac cells in both mice and human hearts by inducing premature senescence, which may represent the molecular basis of DOX-induced cardiomyopathy. Here, we demonstrate that senescence induction in the heart contributes to impaired cardiac function in mice upon DOX treatment. Concomitant elimination of senescent cells with the senolytic Navitoclax in different formulations produces a significant decrease in senescence and cardiotoxicity markers together with the restoration of the cardiac function in mice followed by echocardiography. These results evidence the potential clinical use of senolytic therapies to alleviate cardiotoxicities induced in chemotherapy-treated patients.
Collapse
Affiliation(s)
- Araceli Lérida-Viso
- Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València (UPV), Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av. Fernando Abril Martorell 106, Valencia 46026, Spain; Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Camino de Vera, s/n, Valencia 46022, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València-Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, Valencia 46012, Spain; CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Alejandra Estepa-Fernández
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Camino de Vera, s/n, Valencia 46022, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València-Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, Valencia 46012, Spain; CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Ángela Morellá-Aucejo
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Camino de Vera, s/n, Valencia 46022, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València-Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, Valencia 46012, Spain; CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Beatriz Lozano-Torres
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Camino de Vera, s/n, Valencia 46022, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València-Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, Valencia 46012, Spain; CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - María Alfonso
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Camino de Vera, s/n, Valencia 46022, Spain
| | - Juan F Blandez
- Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València (UPV), Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av. Fernando Abril Martorell 106, Valencia 46026, Spain; Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Camino de Vera, s/n, Valencia 46022, Spain; CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Viviana Bisbal
- Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, Valencia 46012, Spain
| | - Pilar Sepúlveda
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, Av. Fernando Abril Martorell 106, Valencia 46026, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Alba García-Fernández
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Camino de Vera, s/n, Valencia 46022, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València-Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, Valencia 46012, Spain; CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Spain.
| | - Mar Orzáez
- Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València-Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, Valencia 46012, Spain; Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, Valencia 46012, Spain.
| | - Ramón Martínez-Máñez
- Unidad Mixta de Investigación en Nanomedicina y Sensores, Universitat Politècnica de València (UPV), Instituto de Investigación Sanitaria La Fe (IIS La Fe), Av. Fernando Abril Martorell 106, Valencia 46026, Spain; Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Camino de Vera, s/n, Valencia 46022, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Universitat Politècnica de València-Centro de Investigación Príncipe Felipe, C/ Eduardo Primo Yúfera 3, Valencia 46012, Spain; CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Spain.
| |
Collapse
|
35
|
de Rijk MM, Wolf-Johnston A, Kullmann AF, Taiclet S, Kanai AJ, Shiva S, Birder LA. Aging-Associated Changes in Oxidative Stress Negatively Impacts the Urinary Bladder Urothelium. Int Neurourol J 2022; 26:111-118. [PMID: 35793989 PMCID: PMC9260322 DOI: 10.5213/inj.2142224.112] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/18/2020] [Indexed: 12/01/2022] Open
Abstract
PURPOSE Lower urinary tract symptoms are known to significantly increase with age, negatively impacting quality of life and self-reliance. The urothelium fulfills crucial tasks, serving as a barrier protecting the underlying bladder tissue from the harsh chemical composition of urine, and exhibits signaling properties via the release of mediators within the bladder wall that affect bladder functioning. Aging is associated with detrimental changes in cellular health, in part by increasing oxidative stress in the bladder mucosa, and more specifically the urothelium. This, in turn, may impact urothelial mitochondrial health and bioenergetics. METHODS We collected mucosal tissue samples from both young (3-4 months old) and aged (25-30 months old) rats. Tissue was evaluated for p21-Arc, nitrotyrosine, and cytochrome C expression by western immunoblotting. Urothelial cells were cultured for single-cell imaging to analyze basal levels of reactive oxygen species and the mitochondrial membrane potential. Mitochondrial bioenergetics and cellular respiration were investigated by the Seahorse assay, and measurements of adenosine triphosphate release were made using the luciferin-luciferase assay. RESULTS Aging was associated with a significant increase in biomarkers of cellular senescence, oxidative stress, and basal levels of reactive oxygen species. The mitochondrial membrane potential was significantly lower in urothelial cell cultures from aged animals, and cultures from aged animals showed a significant decrease in mitochondrial bioenergetics. CONCLUSION Aging-related increases in oxidative stress and excessive reactive oxygen species may be contributing factors underlying lower urinary tract symptoms in older adults. The mechanisms outlined in this study could be utilized to identify novel pharmaceutical targets to improve aging-associated bladder dysfunction.
Collapse
Affiliation(s)
- Mathijs M. de Rijk
- Department of Urology, School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
- Department of Urology, Maastricht University Medical Center+ (MUMC+), Maastricht, The Netherlands
| | - Amanda Wolf-Johnston
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Aura F. Kullmann
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stephanie Taiclet
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Anthony J. Kanai
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sruti Shiva
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lori A. Birder
- Department of Medicine, Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
36
|
Urolithin A attenuates auditory cell senescence by activating mitophagy. Sci Rep 2022; 12:7704. [PMID: 35546176 PMCID: PMC9095590 DOI: 10.1038/s41598-022-11894-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/28/2022] [Indexed: 11/08/2022] Open
Abstract
Aging of sensory organs is associated with a decline in mitochondrial function and the accumulation of dysfunctional mitochondria. Impaired mitophagy blocks the turnover of dysfunctional mitochondria and leads to their accumulation. Urolithin A (UA) induces mitophagy in various mammalian cells. This study was aimed at investigating the effect of the mitophagy activator, UA, on premature senescent auditory cells. The levels of cellular senescence-associated p53 and p21 significantly increased in H2O2-induced senescent House Ear Institute-Organ of Corti 1 (HEI-OC1) cells and cochlear explants. However, the levels of mitophagy-related molecules significantly decreased. UA significantly decreased the expression of senescence-associated p53 and p21, and increased the expression of mitophagy-related proteins, in H2O2-induced senescent cells and cochlear explants. The percentage of β-galactosidase-stained senescent cells also reduced in H2O2-treated cells and cochlear explants upon UA pre-treatment. The formation of mitophagosomes and mitophagolysosomes was restored upon UA pre-treatment of H2O2-induced senescent cells. The knockdown of mitophagy-related genes (Parkin and Bnip3) resulted in annulment of UA-induced anti-senescent activity. UA significantly increased the ATP content, mitochondrial DNA (mtDNA) integrity, and mitochondrial membrane potential in senescent HEI-OC1 cells. These findings indicate that UA counteracted mitophagy decline and prevented premature senescence in auditory cells. Hence, UA administration might be a promising strategy for preventing mitochondrial dysfunction in patients with age-related hearing loss.
Collapse
|
37
|
Fielder E, Wan T, Alimohammadiha G, Ishaq A, Low E, Weigand BM, Kelly G, Parker C, Griffin B, Jurk D, Korolchuk VI, von Zglinicki T, Miwa S. Short senolytic or senostatic interventions rescue progression of radiation-induced frailty and premature ageing in mice. eLife 2022; 11:75492. [PMID: 35507395 PMCID: PMC9154747 DOI: 10.7554/elife.75492] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 05/03/2022] [Indexed: 12/24/2022] Open
Abstract
Cancer survivors suffer from progressive frailty, multimorbidity, and premature morbidity. We hypothesise that therapy-induced senescence and senescence progression via bystander effects are significant causes of this premature ageing phenotype. Accordingly, the study addresses the question whether a short anti-senescence intervention is able to block progression of radiation-induced frailty and disability in a pre-clinical setting. Male mice were sublethally irradiated at 5 months of age and treated (or not) with either a senolytic drug (Navitoclax or dasatinib + quercetin) for 10 days or with the senostatic metformin for 10 weeks. Follow-up was for 1 year. Treatments commencing within a month after irradiation effectively reduced frailty progression (p<0.05) and improved muscle (p<0.01) and liver (p<0.05) function as well as short-term memory (p<0.05) until advanced age with no need for repeated interventions. Senolytic interventions that started late, after radiation-induced premature frailty was manifest, still had beneficial effects on frailty (p<0.05) and short-term memory (p<0.05). Metformin was similarly effective as senolytics. At therapeutically achievable concentrations, metformin acted as a senostatic neither via inhibition of mitochondrial complex I, nor via improvement of mitophagy or mitochondrial function, but by reducing non-mitochondrial reactive oxygen species production via NADPH oxidase 4 inhibition in senescent cells. Our study suggests that the progression of adverse long-term health and quality-of-life effects of radiation exposure, as experienced by cancer survivors, might be rescued by short-term adjuvant anti-senescence interventions.
Collapse
Affiliation(s)
- Edward Fielder
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, Newcastle, United Kingdom
| | - Tengfei Wan
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, Newcastle, United Kingdom
| | - Ghazaleh Alimohammadiha
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, Newcastle, United Kingdom
| | - Abbas Ishaq
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, Newcastle, United Kingdom
| | - Evon Low
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, Newcastle, United Kingdom
| | - B Melanie Weigand
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, Newcastle, United Kingdom
| | - George Kelly
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, Newcastle, United Kingdom
| | - Craig Parker
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, Newcastle, United Kingdom
| | - Brigid Griffin
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, Newcastle, United Kingdom
| | - Diana Jurk
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, Newcastle, United Kingdom
| | - Viktor I Korolchuk
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, Newcastle, United Kingdom
| | - Thomas von Zglinicki
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, Newcastle, United Kingdom
| | - Satomi Miwa
- Newcastle University Biosciences Institute, Newcastle University, Newcastle upon Tyne, Newcastle, United Kingdom
| |
Collapse
|
38
|
Ruiz-Aparicio PF, Vernot JP. Bone Marrow Aging and the Leukaemia-Induced Senescence of Mesenchymal Stem/Stromal Cells: Exploring Similarities. J Pers Med 2022; 12:jpm12050716. [PMID: 35629139 PMCID: PMC9147878 DOI: 10.3390/jpm12050716] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/20/2022] [Accepted: 04/27/2022] [Indexed: 12/17/2022] Open
Abstract
Bone marrow aging is associated with multiple cellular dysfunctions, including perturbed haematopoiesis, the propensity to haematological transformation, and the maintenance of leukaemia. It has been shown that instructive signals from different leukemic cells are delivered to stromal cells to remodel the bone marrow into a supportive leukemic niche. In particular, cellular senescence, a physiological program with both beneficial and deleterious effects on the health of the organisms, may be responsible for the increased incidence of haematological malignancies in the elderly and for the survival of diverse leukemic cells. Here, we will review the connection between BM aging and cellular senescence and the role that these processes play in leukaemia progression. Specifically, we discuss the role of mesenchymal stem cells as a central component of the supportive niche. Due to the specificity of the genetic defects present in leukaemia, one would think that bone marrow alterations would also have particular changes, making it difficult to envisage a shared therapeutic use. We have tried to summarize the coincident features present in BM stromal cells during aging and senescence and in two different leukaemias, acute myeloid leukaemia, with high frequency in the elderly, and B-acute lymphoblastic leukaemia, mainly a childhood disease. We propose that mesenchymal stem cells are similarly affected in these different leukaemias, and that the changes that we observed in terms of cellular function, redox balance, genetics and epigenetics, soluble factor repertoire and stemness are equivalent to those occurring during BM aging and cellular senescence. These coincident features may be used to explore strategies useful to treat various haematological malignancies.
Collapse
Affiliation(s)
- Paola Fernanda Ruiz-Aparicio
- Grupo de Investigación Fisiología Celular y Molecular, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 111321, Colombia;
| | - Jean-Paul Vernot
- Grupo de Investigación Fisiología Celular y Molecular, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 111321, Colombia;
- Instituto de Investigaciones Biomédicas, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 111321, Colombia
- Correspondence:
| |
Collapse
|
39
|
Hense JD, Garcia DN, Isola JV, Alvarado-Rincón JA, Zanini BM, Prosczek JB, Stout MB, Mason JB, Walsh PT, Brieño-Enríquez MA, Schadock I, Barros CC, Masternak MM, Schneider A. Senolytic treatment reverses obesity-mediated senescent cell accumulation in the ovary. GeroScience 2022; 44:1747-1759. [PMID: 35460445 DOI: 10.1007/s11357-022-00573-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 04/13/2022] [Indexed: 11/26/2022] Open
Abstract
Senescent cells are in a cell cycle arrest state and accumulate with aging and obesity, contributing to a chronic inflammatory state. Treatment with senolytic drugs dasatinib and quercetin (D + Q) can reduce senescent cell burden in several tissues, increasing lifespan. Despite this, there are few reports about senescent cells accumulating in female reproductive tissues. Therefore, the aim of the study was to characterize the ovarian reserve and its relationship with cellular senescence in genetically obese mice (ob/ob). In experiment 1, ob/ob (n = 5) and wild-type (WT) mice (n = 5) at 12 months of age were evaluated. In experiment 2, 2-month-old female ob/ob mice were treated with senolytics (D + Q, n = 6) or placebo (n = 6) during the 4 months. Obese mice had more senescent cells in ovaries, indicated by increased p21 and p16 and lipofuscin staining and macrophage infiltration. Treatment with D + Q significantly reduced senescent cell burden in ovaries of obese mice. Neither obesity nor treatment with D + Q affected the number of ovarian follicles. In conclusion, our data indicate that obesity due to leptin deficiency increases the load of senescent cells in the ovary, which is reduced by treatment by senolytics. However, neither obesity nor D + Q treatment affected the ovarian reserve.
Collapse
Affiliation(s)
- Jéssica D Hense
- Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, RS, Pelotas, Brazil
| | - Driele N Garcia
- Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - José V Isola
- Faculdade de Veterinária, Universidade Federal de Pelotas, Pelotas, RS, Brazil
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Joao A Alvarado-Rincón
- Facultad de Ciencias Agropecuarias, Universidad de La Salle, Campus Utopía, Yopal, Casanare, Colombia
| | - Bianka M Zanini
- Faculdade de Veterinária, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Juliane B Prosczek
- Faculdade de Nutrição, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1, Pelotas, RS, 96010-610, Brazil
| | - Michael B Stout
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Jeffrey B Mason
- Department of Animal, Dairy and Veterinary Sciences, Center for Integrated BioSystems, School of Veterinary Medicine, Utah State University, Logan, UT, USA
| | - Patrick T Walsh
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Miguel A Brieño-Enríquez
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ines Schadock
- Faculdade de Nutrição, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1, Pelotas, RS, 96010-610, Brazil
| | - Carlos C Barros
- Faculdade de Nutrição, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1, Pelotas, RS, 96010-610, Brazil
| | - Michal M Masternak
- College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
- Department of Head and Neck Surgery, Poznan University of Medical Sciences, Poznan, Poland
| | - Augusto Schneider
- Faculdade de Nutrição, Universidade Federal de Pelotas, Rua Gomes Carneiro, 1, Pelotas, RS, 96010-610, Brazil.
| |
Collapse
|
40
|
Imai M, Mizoguchi T, Wang M, Li Y, Hasegawa Y, Tonoki A, Itoh M. The guppy (Poecilia reticulata) is a useful model for analyzing age-dependent changes in metabolism, motor function, and gene expression. Exp Gerontol 2022; 160:111708. [DOI: 10.1016/j.exger.2022.111708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 01/13/2022] [Indexed: 11/04/2022]
|
41
|
Athanasopoulou S, Kapetanou M, Magouritsas MG, Mougkolia N, Taouxidou P, Papacharalambous M, Sakellaridis F, Gonos E. Antioxidant and Antiaging Properties of a Novel Synergistic Nutraceutical Complex: Readouts from an In Cellulo Study and an In Vivo Prospective, Randomized Trial. Antioxidants (Basel) 2022; 11:antiox11030468. [PMID: 35326118 PMCID: PMC8944750 DOI: 10.3390/antiox11030468] [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: 01/31/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 02/04/2023] Open
Abstract
Aging is a dynamic procedure that is developed in multiple layers and characterized by distinct hallmarks. The use of biomarkers that target different hallmarks of aging is substantial in predicting adverse outcomes during the aging process, implementing specifically designed antiaging interventions and monitoring responses to these interventions. The present study aimed to develop a novel composition of plant extracts, comprising identified active ingredients that synergistically target different hallmarks of aging in cellulo and in vivo. The selected single extracts and the developed composition were tested through a powerful set of biomarkers that we have previously identified and studied. The composition of selected extracts simultaneously increased cellular lifespan, reduced the cellular oxidative load and enhanced antioxidant defense mechanisms by increasing proteasome activity and content. In addition, the combination prevented telomere attrition and preserved optimum DNA methylation levels. Remarkably, biomarker profiling of healthy volunteers who received the identified combination in the form of a nutritional supplement within the frame of a prospective, randomized, controlled 3-month trial revealed an unprecedented antioxidant capacity in humans. In conclusion, our results support the notion that interventions with specifically designed combinations of natural compounds targeting multiple hallmarks of aging represent an effective way to improve healthspan and well-being.
Collapse
Affiliation(s)
- Sophia Athanasopoulou
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (S.A.); (M.K.); (N.M.)
- Faculty of Medicine, School of Health Sciences, University of Thessaly, 41334 Larisa, Greece
| | - Marianna Kapetanou
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (S.A.); (M.K.); (N.M.)
| | | | - Nikoletta Mougkolia
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (S.A.); (M.K.); (N.M.)
| | - Polykseni Taouxidou
- Department of Physical Education and Sport Science, Aristotle University, 57001 Thessaloniki, Greece;
| | | | | | - Efstathios Gonos
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (S.A.); (M.K.); (N.M.)
- Hellenic Pasteur Institute, 11521 Athens, Greece
- Correspondence: ; Tel.: +30-210-6478860
| |
Collapse
|
42
|
Dynamic Spatiotemporal Expression Pattern of the Senescence-Associated Factor p16Ink4a in Development and Aging. Cells 2022; 11:cells11030541. [PMID: 35159350 PMCID: PMC8833900 DOI: 10.3390/cells11030541] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 01/27/2022] [Accepted: 02/02/2022] [Indexed: 02/01/2023] Open
Abstract
A plethora of factors have been attributed to underly aging, including oxidative stress, telomere shortening and cellular senescence. Several studies have shown a significant role of the cyclin-dependent kinase inhibitor p16ink4a in senescence and aging. However, its expression in development has been less well documented. Therefore, to further clarify a potential role of p16 in development and aging, we conducted a developmental expression study of p16, as well as of p19ARF and p21, and investigated their expression on the RNA level in brain, heart, liver, and kidney of mice at embryonic, postnatal, adult, and old ages. P16 expression was further assessed on the protein level by immunohistochemistry. Expression of p16 was highly dynamic in all organs in embryonic and postnatal stages and increased dramatically in old mice. Expression of p19 and p21 was less variable and increased to a moderate extent at old age. In addition, we observed a predominant expression of p16 mRNA and protein in liver endothelial cells versus non-endothelial cells of old mice, which suggests a functional role specifically in liver endothelium of old subjects. Thus, p16 dynamic spatiotemporal expression might implicate p16 in developmental and physiological processes in addition to its well-known function in the build-up of senescence.
Collapse
|
43
|
Marquez-Exposito L, Tejedor-Santamaria L, Valentijn FA, Tejera-Muñoz A, Rayego-Mateos S, Marchant V, Rodrigues-Diez RR, Rubio-Soto I, Knoppert SN, Ortiz A, Ramos AM, Goldschmeding R, Ruiz-Ortega M. Oxidative Stress and Cellular Senescence Are Involved in the Aging Kidney. Antioxidants (Basel) 2022; 11:301. [PMID: 35204184 PMCID: PMC8868560 DOI: 10.3390/antiox11020301] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/28/2022] [Accepted: 01/29/2022] [Indexed: 01/10/2023] Open
Abstract
Chronic kidney disease (CKD) can be considered as a clinical model for premature aging. However, non-invasive biomarkers to detect early kidney damage and the onset of a senescent phenotype are lacking. Most of the preclinical senescence studies in aging have been done in very old mice. Furthermore, the precise characterization and over-time development of age-related senescence in the kidney remain unclear. To address these limitations, the age-related activation of cellular senescence-associated mechanisms and their correlation with early structural changes in the kidney were investigated in 3- to 18-month-old C57BL6 mice. Inflammatory cell infiltration was observed by 12 months, whereas tubular damage and collagen accumulation occurred later. Early activation of cellular-senescence-associated mechanisms was found in 12-month-old mice, characterized by activation of the DNA-damage-response (DDR), mainly in tubular cells; activation of the antioxidant NRF2 pathway; and klotho downregulation. However, induction of tubular-cell-cycle-arrest (CCA) and overexpression of renal senescent-associated secretory phenotype (SASP) components was only found in 18-month-old mice. In aging mice, both inflammation and oxidative stress (marked by elevated lipid peroxidation and NRF2 inactivation) remained increased. These findings support the hypothesis that prolonged DDR and CCA, loss of nephroprotective factors (klotho), and dysfunctional redox regulatory mechanisms (NRF2/antioxidant defense) can be early drivers of age-related kidney-damage progression.
Collapse
Affiliation(s)
- Laura Marquez-Exposito
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.M.-E.); (L.T.-S.); (A.T.-M.); (S.R.-M.); (V.M.); (I.R.-S.)
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, 28040 Madrid, Spain; (R.R.R.-D.); (A.O.); (A.M.R.)
| | - Lucia Tejedor-Santamaria
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.M.-E.); (L.T.-S.); (A.T.-M.); (S.R.-M.); (V.M.); (I.R.-S.)
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, 28040 Madrid, Spain; (R.R.R.-D.); (A.O.); (A.M.R.)
| | - Floris A. Valentijn
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (F.A.V.); (S.N.K.); (R.G.)
| | - Antonio Tejera-Muñoz
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.M.-E.); (L.T.-S.); (A.T.-M.); (S.R.-M.); (V.M.); (I.R.-S.)
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, 28040 Madrid, Spain; (R.R.R.-D.); (A.O.); (A.M.R.)
| | - Sandra Rayego-Mateos
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.M.-E.); (L.T.-S.); (A.T.-M.); (S.R.-M.); (V.M.); (I.R.-S.)
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, 28040 Madrid, Spain; (R.R.R.-D.); (A.O.); (A.M.R.)
| | - Vanessa Marchant
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.M.-E.); (L.T.-S.); (A.T.-M.); (S.R.-M.); (V.M.); (I.R.-S.)
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, 28040 Madrid, Spain; (R.R.R.-D.); (A.O.); (A.M.R.)
| | - Raul R. Rodrigues-Diez
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, 28040 Madrid, Spain; (R.R.R.-D.); (A.O.); (A.M.R.)
- Translational Immunology Laboratory, Health Research Institute of Asturias (ISPA), Av. Roma, s/n, 33011 Oviedo, Spain
| | - Irene Rubio-Soto
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.M.-E.); (L.T.-S.); (A.T.-M.); (S.R.-M.); (V.M.); (I.R.-S.)
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, 28040 Madrid, Spain; (R.R.R.-D.); (A.O.); (A.M.R.)
| | - Sebastiaan N. Knoppert
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (F.A.V.); (S.N.K.); (R.G.)
| | - Alberto Ortiz
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, 28040 Madrid, Spain; (R.R.R.-D.); (A.O.); (A.M.R.)
- Division of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Adrian M. Ramos
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, 28040 Madrid, Spain; (R.R.R.-D.); (A.O.); (A.M.R.)
- Division of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, H04.312, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands; (F.A.V.); (S.N.K.); (R.G.)
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.M.-E.); (L.T.-S.); (A.T.-M.); (S.R.-M.); (V.M.); (I.R.-S.)
- Red de Investigación Renal (REDinREN), Instituto de Salud Carlos III, 28040 Madrid, Spain; (R.R.R.-D.); (A.O.); (A.M.R.)
| |
Collapse
|
44
|
Liu X, Zhan Y, Xu W, Liu L, Liu X, Da J, Zhang K, Zhang X, Wang J, Liu Z, Jin H, Zhang B, Li Y. Characterization of transcriptional landscape in bone marrow-derived mesenchymal stromal cells treated with aspirin by RNA-seq. PeerJ 2022; 10:e12819. [PMID: 35127290 PMCID: PMC8793730 DOI: 10.7717/peerj.12819] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 12/30/2021] [Indexed: 01/10/2023] Open
Abstract
INTRODUCTION Aspirin is a common antipyretic, analgesic, and anti-inflammatory drug, which has been reported to extend life in animal models and application in the treatment of aging-related diseases. However, it remains unclear about the effects of aspirin on bone marrow-derived mesenchymal stromal cells (BM-MSCs). Here, we aimed to analyze the influence of aspirin on senescence and young BM-MSCs. METHODS BM-MSCs were serially passaged to construct a replicative senescence model. SA-β-gal staining, PCR, western blot, and RNA-sequencing were performed on BM-MSCs with or without aspirin treatment, to examine aspirin's impact on bone marrow-derived mesenchymal stem cells. RESULTS SA-β-gal staining, PCR, and western blot revealed that aspirin could alleviate the cellular expression of senescence-related indicators of BM-MSCs, including a decrease of SA-β-gal-positive cells and staining intensity, and downregulation of p16, p21, and p53 expression after aspirin treatment. RNA-sequencing results shown in the biological processes related to aging, aspirin could influence cellular immune response and lipid metabolism. CONCLUSION The efficacy of aspirin for retarding senescence of BM-MSCs was demonstrated. Our study indicated that the mechanisms of this delay might involve influencing immune response and lipid metabolism.
Collapse
Affiliation(s)
- Xinpeng Liu
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuanbo Zhan
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China,The Second Affiliated Hospital of Harbin Medical University, Department of Periodontology and Oral Mucosa, Harbin, China
| | - Wenxia Xu
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lixue Liu
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoyao Liu
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Junlong Da
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kai Zhang
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinjian Zhang
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jianqun Wang
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ziqi Liu
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Han Jin
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bin Zhang
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China,Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Ying Li
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
45
|
El Hadri K, Smith R, Duplus E, El Amri C. Inflammation, Oxidative Stress, Senescence in Atherosclerosis: Thioredoxine-1 as an Emerging Therapeutic Target. Int J Mol Sci 2021; 23:ijms23010077. [PMID: 35008500 PMCID: PMC8744732 DOI: 10.3390/ijms23010077] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/19/2021] [Accepted: 12/19/2021] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis is a leading cause of cardiovascular diseases (CVD) worldwide and intimately linked to aging. This pathology is characterized by chronic inflammation, oxidative stress, gradual accumulation of low-density lipoproteins (LDL) particles and fibrous elements in focal areas of large and medium arteries. These fibrofatty lesions in the artery wall become progressively unstable and thrombogenic leading to heart attack, stroke or other severe heart ischemic syndromes. Elevated blood levels of LDL are major triggering events for atherosclerosis. A cascade of molecular and cellular events results in the atherosclerotic plaque formation, evolution, and rupture. Moreover, the senescence of multiple cell types present in the vasculature were reported to contribute to atherosclerotic plaque progression and destabilization. Classical therapeutic interventions consist of lipid-lowering drugs, anti-inflammatory and life style dispositions. Moreover, targeting oxidative stress by developing innovative antioxidant agents or boosting antioxidant systems is also a well-established strategy. Accumulation of senescent cells (SC) is also another important feature of atherosclerosis and was detected in various models. Hence, targeting SCs appears as an emerging therapeutic option, since senolytic agents favorably disturb atherosclerotic plaques. In this review, we propose a survey of the impact of inflammation, oxidative stress, and senescence in atherosclerosis; and the emerging therapeutic options, including thioredoxin-based approaches such as anti-oxidant, anti-inflammatory, and anti-atherogenic strategy with promising potential of senomodulation.
Collapse
|
46
|
Lynch SM, Guo G, Gibson DS, Bjourson AJ, Rai TS. Role of Senescence and Aging in SARS-CoV-2 Infection and COVID-19 Disease. Cells 2021; 10:3367. [PMID: 34943875 PMCID: PMC8699414 DOI: 10.3390/cells10123367] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/17/2021] [Accepted: 11/23/2021] [Indexed: 02/07/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has resulted in a global pandemic associated with substantial morbidity and mortality worldwide, with particular risk for severe disease and mortality in the elderly population. SARS-CoV-2 infection is driven by a pathological hyperinflammatory response which results in a dysregulated immune response. Current advancements in aging research indicates that aging pathways have fundamental roles in dictating healthspan in addition to lifespan. Our review discusses the aging immune system and highlights that senescence and aging together, play a central role in COVID-19 pathogenesis. In our review, we primarily focus on the immune system response to SARS-CoV-2 infection, the interconnection between severe COVID-19, immunosenescence, aging, vaccination, and the emerging problem of Long-COVID. We hope to highlight the importance of identifying specific senescent endotypes (or "sendotypes"), which can used as determinants of COVID-19 severity and mortality. Indeed, identified sendotypes could be therapeutically exploited for therapeutic intervention. We highlight that senolytics, which eliminate senescent cells, can target aging-associated pathways and therefore are proving attractive as potential therapeutic options to alleviate symptoms, prevent severe infection, and reduce mortality burden in COVID-19 and thus ultimately enhance healthspan.
Collapse
Affiliation(s)
| | | | | | | | - Taranjit Singh Rai
- Northern Ireland Centre for Stratified Medicine, School of Biomedical Sciences, Ulster University, C-TRIC Building, Altnagelvin Area Hospital, Glenshane Road, Derry BT47 6SB, UK; (S.M.L.); (G.G.); (D.S.G.); (A.J.B.)
| |
Collapse
|
47
|
Parella KJ, Manhardt C, Capucilli D, Moyer B, Colegrove H, Moody KJ, Sleeper M, Banas A, Rebbaa A, Wolfe AJ. Fluorescence-Based Detection of Ferrous Iron in Senescent Cells. Rejuvenation Res 2021; 24:456-463. [PMID: 34841899 DOI: 10.1089/rej.2021.0075] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
A major limitation in aging research is the lack of reliable biomarkers to assess phenotypic changes with age or monitor response to antiaging interventions. This study investigates the role of intracellular ferrous iron (Fe2+) as a potential biomarker of senescence. Iron is known to accumulate in various tissues with age and recent studies have demonstrated that its level increases dramatically in senescent cells. The current techniques used to measure the accumulation of iron are cumbersome and only measure total iron not specific isotopes such as the redox reactive Fe2+. It is still to be determined whether the damaging form of iron (Fe2+) is specifically elevated in senescent cells. In this study, we assessed the potential use of a newly discovered Fe2+ reactive probe (SiRhoNox-1) for selective labeling of senescent cells in vitro. For this we have generated various senescent cell models and subjected them to SiRhoNox-1 labeling. Our results indicate that SiRhoNox-1 selectivity labels live senescent cells and was more specific and faster than current staining such as SA-βGal or a derived fluorescent probe C12FDG. Together these findings suggest that SiRhoNox-1 may serve as a convenient tool to detect senescent cells based on their ferrous iron level.
Collapse
Affiliation(s)
- Kyle J Parella
- Ichor Life Sciences, Inc., Lafayette, New York, USA.,Lewis School of Health Sciences, Clarkson University, Potsdam, New York, USA.,Department of Chemistry, Clarkson University, Potsdam, New York, USA
| | | | | | | | | | - Kelsey J Moody
- Ichor Life Sciences, Inc., Lafayette, New York, USA.,Lewis School of Health Sciences, Clarkson University, Potsdam, New York, USA.,Department of Chemistry, Clarkson University, Potsdam, New York, USA.,FoxBio, Inc., Lafayette, New York, USA
| | | | - Andrew Banas
- Ichor Life Sciences, Inc., Lafayette, New York, USA
| | | | - Aaron J Wolfe
- Ichor Life Sciences, Inc., Lafayette, New York, USA.,Lewis School of Health Sciences, Clarkson University, Potsdam, New York, USA.,Department of Chemistry, Clarkson University, Potsdam, New York, USA.,FoxBio, Inc., Lafayette, New York, USA
| |
Collapse
|
48
|
Klinngam W, Rungkamoltip P, Thongin S, Joothamongkhon J, Khumkhrong P, Khongkow M, Namdee K, Tepaamorndech S, Chaikul P, Kanlayavattanakul M, Lourith N, Piboonprai K, Ruktanonchai U, Asawapirom U, Iempridee T. Polymethoxyflavones from Kaempferia parviflora ameliorate skin aging in primary human dermal fibroblasts and ex vivo human skin. Biomed Pharmacother 2021; 145:112461. [PMID: 34839253 DOI: 10.1016/j.biopha.2021.112461] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 11/13/2021] [Accepted: 11/19/2021] [Indexed: 12/12/2022] Open
Abstract
Skin aging is accompanied by an increase in the number of senescent cells, resulting in various pathological outcomes. These include inflammation, impaired barrier function, and susceptibility to skin disorders such as cancer. Kaempferia parviflora (Thai black ginger), a medicinal plant native to Thailand, has been shown to counteract inflammation, cancer, and senescence. This study demonstrates that polymethoxyflavones (5,7-dimethoxyflavone, 5,7,4'-trimethoxyflavone, and 3,5,7,3',4'-pentamethoxyflavone) purified from K. parviflora rhizomes suppressed cellular senescence, reactive oxygen species, and the senescence-associated secretory phenotype in primary human dermal fibroblasts. In addition, they increased tropocollagen synthesis and alleviated free radical-induced cellular and mitochondrial damage. Moreover, the compounds mitigated chronological aging in a human ex vivo skin model by attenuating senescence and restoring expression of essential components of the extracellular matrix, including collagen type I, fibrillin-1, and hyaluronic acid. Finally, we report that polymethoxyflavones enhanced epidermal thickness and epidermal-dermal stability, while blocking age-related inflammation in skin explants. Our findings support the use of polymethoxyflavones from K. parviflora as natural anti-aging agents, highlighting their potential as active ingredients in cosmeceutical and nutraceutical products.
Collapse
Affiliation(s)
- Wannita Klinngam
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Phetploy Rungkamoltip
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Saowarose Thongin
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Jaruwan Joothamongkhon
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Phattharachanok Khumkhrong
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Mattaka Khongkow
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Katawut Namdee
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Surapun Tepaamorndech
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Puxvadee Chaikul
- Phytocosmetics and Cosmeceuticals Research Group, School of Cosmetic Science, Mae Fah Luang University, Chiang Rai, Thailand; School of Cosmetic Science, Mae Fah Luang University, Chiang Rai 57100, Thailand
| | - Mayuree Kanlayavattanakul
- Phytocosmetics and Cosmeceuticals Research Group, School of Cosmetic Science, Mae Fah Luang University, Chiang Rai, Thailand; School of Cosmetic Science, Mae Fah Luang University, Chiang Rai 57100, Thailand
| | - Nattaya Lourith
- Phytocosmetics and Cosmeceuticals Research Group, School of Cosmetic Science, Mae Fah Luang University, Chiang Rai, Thailand; School of Cosmetic Science, Mae Fah Luang University, Chiang Rai 57100, Thailand
| | - Kitiya Piboonprai
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand; Laboratory of Host Defense, The World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan
| | - Uracha Ruktanonchai
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Udom Asawapirom
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Tawin Iempridee
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand.
| |
Collapse
|
49
|
Huang P, Zhou Y, Tang W, Ren C, Jiang A, Wang X, Qian X, Zhou Z, Gong A. Long-term treatment of Nicotinamide mononucleotide improved age-related diminished ovary reserve through enhancing the mitophagy level of granulosa cells in mice. J Nutr Biochem 2021; 101:108911. [PMID: 34801690 DOI: 10.1016/j.jnutbio.2021.108911] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/25/2021] [Accepted: 10/22/2021] [Indexed: 12/17/2022]
Abstract
Ovarian aging affects the reproductive health of elderly women due to decline in oocyte quality, which is closely related to mitochondrial dysfunction. Nicotinamide mononucleotide (NMN), as a precursor of NAD+, effectively regulate mitochondria metabolism in mice. However, roles of NMN in improving age-related diminished ovary reserve remain to be determined. In present study, 4, 8, 12, 24, 40-week old female ICR mice were collected and a 20-week-long administration of NMN was conducted to 40-week-old mice (60WN), meanwhile the control group is given water (60WC). First, we found that 20-week-long administration of NMN to 40-week-old mice exhibited anti-aging and anti-inflammatory effects on organ structures, along with the improvement of estrus cycle condition and endocrine function. The number of primordial, primary, secondary, antral follicles and corpora luteum of ovaries in 60WN group was significantly increased compared with those in 60WC group. Additionally, the protein and gene expressions of P16 of ovaries were significantly reduced in 60WN group than in 60WC group. the mitochondria biogenesis, autophagy level, and proteases activity enhanced in granulosa cells after 20-week-administration of NMN. Present results indicate that NMN has the potential to save diminished ovary reserve by long-term treatment, providing a basis for exploring the role of NMN in anti-ovarian aging by enhancing the mitophagy level of granulosa cells.
Collapse
Affiliation(s)
- Pan Huang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Yan Zhou
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Weihong Tang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Caifang Ren
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Anqi Jiang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Xuxin Wang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Xin Qian
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Zhengrong Zhou
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, China.
| | - Aihua Gong
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, China.
| |
Collapse
|
50
|
Thapa D, Valente JDS, Barrett B, Smith MJ, Argunhan F, Lee SY, Nikitochkina S, Kodji X, Brain SD. Dysfunctional TRPM8 signalling in the vascular response to environmental cold in ageing. eLife 2021; 10:70153. [PMID: 34726597 PMCID: PMC8592571 DOI: 10.7554/elife.70153] [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] [Received: 05/07/2021] [Accepted: 11/02/2021] [Indexed: 12/16/2022] Open
Abstract
Ageing is associated with increased vulnerability to environmental cold exposure. Previously, we identified the role of the cold-sensitive transient receptor potential (TRP) A1, M8 receptors as vascular cold sensors in mouse skin. We hypothesised that this dynamic cold-sensor system may become dysfunctional in ageing. We show that behavioural and vascular responses to skin local environmental cooling are impaired with even moderate ageing, with reduced TRPM8 gene/protein expression especially. Pharmacological blockade of the residual TRPA1/TRPM8 component substantially diminished the response in aged, compared with young mice. This implies the reliance of the already reduced cold-induced vascular response in ageing mice on remaining TRP receptor activity. Moreover, sympathetic-induced vasoconstriction was reduced with downregulation of the α2c adrenoceptor expression in ageing. The cold-induced vascular response is important for sensing cold and retaining body heat and health. These findings reveal that cold sensors, essential for this neurovascular pathway, decline as ageing onsets.
Collapse
Affiliation(s)
- Dibesh Thapa
- Section of Vascular Biology and Inflammation, School of Cardiovascular Medicine and Sciences, BHF Centre of Research Excellence, King's College London, London, United Kingdom
| | - Joäo de Sousa Valente
- Section of Vascular Biology and Inflammation, School of Cardiovascular Medicine and Sciences, BHF Centre of Research Excellence, King's College London, London, United Kingdom
| | - Brentton Barrett
- Section of Vascular Biology and Inflammation, School of Cardiovascular Medicine and Sciences, BHF Centre of Research Excellence, King's College London, London, United Kingdom
| | - Matthew John Smith
- Section of Vascular Biology and Inflammation, School of Cardiovascular Medicine and Sciences, BHF Centre of Research Excellence, King's College London, London, United Kingdom
| | - Fulye Argunhan
- Section of Vascular Biology and Inflammation, School of Cardiovascular Medicine and Sciences, BHF Centre of Research Excellence, King's College London, London, United Kingdom
| | - Sheng Y Lee
- Section of Vascular Biology and Inflammation, School of Cardiovascular Medicine and Sciences, BHF Centre of Research Excellence, King's College London, London, United Kingdom.,Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Sofya Nikitochkina
- Section of Vascular Biology and Inflammation, School of Cardiovascular Medicine and Sciences, BHF Centre of Research Excellence, King's College London, London, United Kingdom
| | - Xenia Kodji
- Section of Vascular Biology and Inflammation, School of Cardiovascular Medicine and Sciences, BHF Centre of Research Excellence, King's College London, London, United Kingdom.,Skin Research Institute, Agency of Science, Technology, and Research (A*STAR), Singapore, Singapore
| | - Susan D Brain
- Section of Vascular Biology and Inflammation, School of Cardiovascular Medicine and Sciences, BHF Centre of Research Excellence, King's College London, London, United Kingdom
| |
Collapse
|