1
|
Sizek H, Deritei D, Fleig K, Harris M, Regan PL, Glass K, Regan ER. Unlocking mitochondrial dysfunction-associated senescence (MiDAS) with NAD + - A Boolean model of mitochondrial dynamics and cell cycle control. Transl Oncol 2024; 49:102084. [PMID: 39163758 PMCID: PMC11380032 DOI: 10.1016/j.tranon.2024.102084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 05/14/2024] [Accepted: 05/25/2024] [Indexed: 08/22/2024] Open
Abstract
The steady accumulation of senescent cells with aging creates tissue environments that aid cancer evolution. Aging cell states are highly heterogeneous. 'Deep senescent' cells rely on healthy mitochondria to fuel a strong proinflammatory secretome, including cytokines, growth and transforming signals. Yet, the physiological triggers of senescence such as reactive oxygen species (ROS) can also trigger mitochondrial dysfunction, and sufficient energy deficit to alter their secretome and cause chronic oxidative stress - a state termed Mitochondrial Dysfunction-Associated Senescence (MiDAS). Here, we offer a mechanistic hypothesis for the molecular processes leading to MiDAS, along with testable predictions. To do this we have built a Boolean regulatory network model that qualitatively captures key aspects of mitochondrial dynamics during cell cycle progression (hyper-fusion at the G1/S boundary, fission in mitosis), apoptosis (fission and dysfunction) and glucose starvation (reversible hyper-fusion), as well as MiDAS in response to SIRT3 knockdown or oxidative stress. Our model reaffirms the protective role of NAD+ and external pyruvate. We offer testable predictions about the growth factor- and glucose-dependence of MiDAS and its reversibility at different stages of reactive oxygen species (ROS)-induced senescence. Our model provides mechanistic insights into the distinct stages of DNA-damage induced senescence, the relationship between senescence and epithelial-to-mesenchymal transition in cancer and offers a foundation for building multiscale models of tissue aging.
Collapse
Affiliation(s)
- Herbert Sizek
- Biochemistry and Molecular Biology, The College of Wooster, Wooster, OH 44691, USA
| | - Dávid Deritei
- Channing Division of Network Medicine, Brigham and Women's Hospital / Harvard Medical School, Boston, MA 02115, USA
| | - Katherine Fleig
- Neuroscience, The College of Wooster, Wooster, OH 44691, USA
| | - Marlayna Harris
- Biochemistry and Molecular Biology, The College of Wooster, Wooster, OH 44691, USA
| | - Peter L Regan
- Biochemistry and Molecular Biology, The College of Wooster, Wooster, OH 44691, USA
| | - Kimberly Glass
- Channing Division of Network Medicine, Brigham and Women's Hospital / Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
2
|
von Zglinicki T. Oxidative stress and cell senescence as drivers of ageing: chicken and egg. Ageing Res Rev 2024:102558. [PMID: 39454760 DOI: 10.1016/j.arr.2024.102558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
Oxidative stress and cell senescence are both important drivers of ageing and age-associated disease and disability. In vitro, they are closely interconnected in a chicken-and-egg relationship: Not only is oxidative stress an important cause of cell senescence, but senescent cells are also sources of oxidative stress, obscuring cause-effect relationships during the ageing process. We hypothesize that cell senescence is a significant cause of tissue and systemic oxidative stress during ageing. This review aims to critically summarize the available evidence for this hypothesis. After summarizing the cellular feedback mechanisms that make oxidative stress an integral part of the senescent phenotype, it critically reviews the existing evidence for a role of senescent cells as causes of oxidative stress during mammalian ageing in vivo, focussing on results from intervention experiments. It is concluded that while the available data are in agreement with this hypothesis, they are still too scarce to support a robust conclusion.
Collapse
Affiliation(s)
- Thomas von Zglinicki
- Ageing Research Laboratories, Biosciences Institute, Faculty of Medical Sciences, Campus for Ageing and Health, Newcastle University, UK.
| |
Collapse
|
3
|
Deritei D, Anamika WJ, Zhou X, Silverman EK, Regan ER, Glass K. HHIP's Dynamic Role in Epithelial Wound Healing Reveals a Potential Mechanism of COPD Susceptibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.05.611545. [PMID: 39416045 PMCID: PMC11482804 DOI: 10.1101/2024.09.05.611545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
A genetic variant near HHIP has been consistently identified as associated with increased risk for Chronic Obstructive Pulmonary Disease (COPD), the third leading cause of death worldwide. However HHIP's role in COPD pathogenesis remains elusive. Canonically, HHIP is a negative regulator of the hedgehog pathway and downstream GLI1 and GLI2 activation. The hedgehog pathway plays an important role in wound healing, specifically in activating transcription factors that drive the epithelial mesenchymal transition (EMT), which in its intermediate state (partial EMT) is necessary for the collective movement of cells closing the wound. Herein, we propose a mechanism to explain HHIP's role in faulty epithelial wound healing, which could contribute to the development of emphysema, a key feature of COPD. Using two different Boolean models compiled from the literature, we show dysfunctional HHIP results in a lack of negative feedback on GLI, triggering a full EMT, where cells become mesenchymal and do not properly close the wound. We validate these Boolean models with experimental evidence gathered from published scientific literature. We also experimentally test if low HHIP expression is associated with EMT at the edge of wounds by using a scratch assay in a human lung epithelial cell line. Finally, we show evidence supporting our hypothesis in bulk and single cell RNA-Seq data from different COPD cohorts. Overall, our analyses suggest that aberrant wound healing due to dysfunctional HHIP, combined with chronic epithelial damage through cigarette smoke exposure, may be a primary cause of COPD-associated emphysema.
Collapse
Affiliation(s)
- Dávid Deritei
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Wardatul Jannat Anamika
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Edwin K. Silverman
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02215, USA
| | | | - Kimberly Glass
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
4
|
Yang M, Huang Y, Tang A, Zhang Y, Liu Y, Fan Z, Li M. Research Hotspots in Mitochondria-Related Studies for AKI Treatment: A Bibliometric Study. Drug Des Devel Ther 2024; 18:4051-4063. [PMID: 39280255 PMCID: PMC11402358 DOI: 10.2147/dddt.s473426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/27/2024] [Indexed: 09/18/2024] Open
Abstract
Purpose Acute kidney injury (AKI) is a common clinical critical condition that has become a significant healthcare burden. In recent years, the relationship between AKI and mitochondria has attracted increasing attention. Protecting mitochondria or restoring their function has emerged as a novel therapeutic strategy for alleviating AKI. This study aims to analyze and summarize the current status, research trends, and hotspots in this field, providing references and directions for future research. Methods AKI and mitochondria-related literature from the Web of Science core collection were retrieved and collected. Bibliometric and visualization analyses were conducted using Microsoft Excel 2021, bibliometric tools (VosViewer, Citespace 6.3.R1, and the bibliometrix R package), R 4.3.2, and SCImagoGraphica software. Results A total of 2433 publications were included in this study. The number of annual publications in this field has increased year by year. China and the United States are the two most productive countries. Central South University is the most influential research institution in terms of research output, and Parikh SM, Schnellmann RG, and Dong Z are the most influential authors in this field. KIDNEY INTERNATIONAL, JOURNAL OF THE AMERICAN SOCIETY OF NEPHROLOGY, and AMERICAN JOURNAL OF PHYSIOLOGY-RENAL PHYSIOLOGY are the most influential journals. Initially, the research focused on keywords such as oxidative stress, ischemia-reperfusion injury, apoptosis, inflammation, and autophagy. In recent years, new research hotspots have emerged, including ferroptosis, aging, mitochondrial quality control, messenger RNA, mitochondrial-targeted antioxidants, extracellular vesicles, and nanodrug delivery. Conclusion Research on the relationship between mitochondria and AKI has broad developing prospects, and targeting mitochondrial regulation will become a focus of future AKI prevention and treatment research.
Collapse
Affiliation(s)
- Mengfan Yang
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Youqun Huang
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Anqi Tang
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| | - Yu Zhang
- Department of Nephrology, Shaanxi Provincial Hospital of Traditional Chinese Medicine, Xi’an, Shaanxi Provincial, People’s Republic of China
| | - Yu Liu
- Department of Nephrology, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, People’s Republic of China
| | - Zhenliang Fan
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Mingquan Li
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, People’s Republic of China
| |
Collapse
|
5
|
Kelly G, Kataura T, Panek J, Ma G, Salmonowicz H, Davis A, Kendall H, Brookes C, Ayine-Tora DM, Banks P, Nelson G, Dobby L, Pitrez PR, Booth L, Costello L, Richardson GD, Lovat P, Przyborski S, Ferreira L, Greaves L, Szczepanowska K, von Zglinicki T, Miwa S, Brown M, Flagler M, Oblong JE, Bascom CC, Carroll B, Reynisson J, Korolchuk VI. Suppressed basal mitophagy drives cellular aging phenotypes that can be reversed by a p62-targeting small molecule. Dev Cell 2024; 59:1924-1939.e7. [PMID: 38897197 DOI: 10.1016/j.devcel.2024.04.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/25/2023] [Accepted: 04/28/2024] [Indexed: 06/21/2024]
Abstract
Selective degradation of damaged mitochondria by autophagy (mitophagy) is proposed to play an important role in cellular homeostasis. However, the molecular mechanisms and the requirement of mitochondrial quality control by mitophagy for cellular physiology are poorly understood. Here, we demonstrated that primary human cells maintain highly active basal mitophagy initiated by mitochondrial superoxide signaling. Mitophagy was found to be mediated by PINK1/Parkin-dependent pathway involving p62 as a selective autophagy receptor (SAR). Importantly, this pathway was suppressed upon the induction of cellular senescence and in naturally aged cells, leading to a robust shutdown of mitophagy. Inhibition of mitophagy in proliferating cells was sufficient to trigger the senescence program, while reactivation of mitophagy was necessary for the anti-senescence effects of NAD precursors or rapamycin. Furthermore, reactivation of mitophagy by a p62-targeting small molecule rescued markers of cellular aging, which establishes mitochondrial quality control as a promising target for anti-aging interventions.
Collapse
Affiliation(s)
- George Kelly
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Tetsushi Kataura
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE4 5PL, UK; Department of Neurology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Johan Panek
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Gailing Ma
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Hanna Salmonowicz
- ReMedy International Research Agenda Unit, IMol Polish Academy of Sciences, Warsaw 02-247, Poland
| | - Ashley Davis
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Hannah Kendall
- Wellcome Centre for Mitochondrial Research, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Charlotte Brookes
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | | | - Peter Banks
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Glyn Nelson
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Laura Dobby
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Patricia R Pitrez
- FMUC - Faculty of Medicine, Pólo das Ciências da Saúde, Unidade Central Azinhaga de Santa Comba, Coimbra 3000-354, Portugal
| | - Laura Booth
- Translation and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Lydia Costello
- Department of Biosciences, Durham University, Durham DH1 3LE, UK
| | - Gavin D Richardson
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Penny Lovat
- Precision Medicine, Translation and Clinical Research Institute, Newcastle University Centre for Cancer, The Medical School, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | | | - Lino Ferreira
- FMUC - Faculty of Medicine, Pólo das Ciências da Saúde, Unidade Central Azinhaga de Santa Comba, Coimbra 3000-354, Portugal
| | - Laura Greaves
- Wellcome Centre for Mitochondrial Research, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Karolina Szczepanowska
- ReMedy International Research Agenda Unit, IMol Polish Academy of Sciences, Warsaw 02-247, Poland
| | - Thomas von Zglinicki
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Satomi Miwa
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Max Brown
- The Procter & Gamble Company, Cincinnati, OH 45040, USA
| | | | - John E Oblong
- The Procter & Gamble Company, Cincinnati, OH 45040, USA
| | | | | | - Jóhannes Reynisson
- School of Pharmacy and Bioengineering, Keele University, Newcastle under Lyme ST5 5BG, UK
| | - Viktor I Korolchuk
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE4 5PL, UK.
| |
Collapse
|
6
|
Quarato ER, Salama NA, Calvi LM. Interplay Between Skeletal and Hematopoietic Cells in the Bone Marrow Microenvironment in Homeostasis and Aging. Curr Osteoporos Rep 2024; 22:416-432. [PMID: 38782850 DOI: 10.1007/s11914-024-00874-2] [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] [Accepted: 05/05/2024] [Indexed: 05/25/2024]
Abstract
PURPOSE OF THE REVIEW In this review, we discuss the most recent scientific advances on the reciprocal regulatory interactions between the skeletal and hematopoietic stem cell niche, focusing on immunomodulation and its interplay with the cell's mitochondrial function, and how this impacts osteoimmune health during aging and disease. RECENT FINDINGS Osteoimmunology investigates interactions between cells that make up the skeletal stem cell niche and immune system. Much work has investigated the complexity of the bone marrow microenvironment with respect to the skeletal and hematopoietic stem cells that regulate skeletal formation and immune health respectively. It has now become clear that these cellular components cooperate to maintain homeostasis and that dysfunction in their interaction can lead to aging and disease. Having a deeper, mechanistic appreciation for osteoimmune regulation will lead to better research perspective and therapeutics with the potential to improve the aging process, skeletal and hematologic regeneration, and disease targeting.
Collapse
Affiliation(s)
- Emily R Quarato
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA.
- James P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.
| | - Noah A Salama
- James P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA.
| | - Laura M Calvi
- James P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
7
|
Shao M, Qiu Y, Shen M, Liu W, Feng D, Luo Z, Zhou Y. Procyanidin C1 inhibits bleomycin-induced pulmonary fibrosis in mice by selective clearance of senescent myofibroblasts. FASEB J 2024; 38:e23749. [PMID: 38953707 DOI: 10.1096/fj.202302547rr] [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: 12/09/2023] [Revised: 05/05/2024] [Accepted: 06/06/2024] [Indexed: 07/04/2024]
Abstract
Pulmonary fibrosis is a formidable challenge in chronic and age-related lung diseases. Myofibroblasts secrete large amounts of extracellular matrix and induce pro-repair responses during normal wound healing. Successful tissue repair results in termination of myofibroblast activity via apoptosis; however, some myofibroblasts exhibit a senescent phenotype and escape apoptosis, causing over-repair that is characterized by pathological fibrotic scarring. Therefore, the removal of senescent myofibroblasts using senolytics is an important method for the treatment of pulmonary fibrosis. Procyanidin C1 (PCC1) has recently been discovered as a senolytic compound with very low toxicity and few side effects. This study aimed to determine whether PCC1 could improve lung fibrosis by promoting apoptosis in senescent myofibroblasts and to investigate the mechanisms involved. The results showed that PCC1 attenuates bleomycin (BLM)-induced pulmonary fibrosis in mice. In addition, we found that PCC1 inhibited extracellular matrix deposition and promoted the apoptosis of senescent myofibroblasts by increasing PUMA expression and activating the BAX signaling pathway. Our findings represent a new method of pulmonary fibrosis management and emphasize the potential of PCC1 as a senotherapeutic agent for the treatment of pulmonary fibrosis, providing hope for patients with pulmonary fibrosis worldwide. Our results advance our understanding of age-related diseases and highlight the importance of addressing cellular senescence in treatment.
Collapse
Affiliation(s)
- Min Shao
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Yujia Qiu
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Mengxia Shen
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Wei Liu
- Department of Community Nursing, Xiangya Nursing School, Central South University, Changsha, China
| | - Dandan Feng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Ziqiang Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
- Hunan Key Laboratory of Organ Fibrosis, Changsha, China
| | - Yan Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
8
|
Wang ZH, Wang ZJ, Liu HC, Wang CY, Wang YQ, Yue Y, Zhao C, Wang G, Wan JP. Targeting mitochondria for ovarian aging: new insights into mechanisms and therapeutic potential. Front Endocrinol (Lausanne) 2024; 15:1417007. [PMID: 38952389 PMCID: PMC11215021 DOI: 10.3389/fendo.2024.1417007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 05/29/2024] [Indexed: 07/03/2024] Open
Abstract
Ovarian aging is a complex process characterized by a decline in oocyte quantity and quality, directly impacting fertility and overall well-being. Recent researches have identified mitochondria as pivotal players in the aging of ovaries, influencing various hallmarks and pathways governing this intricate process. In this review, we discuss the multifaceted role of mitochondria in determining ovarian fate, and outline the pivotal mechanisms through which mitochondria contribute to ovarian aging. Specifically, we emphasize the potential of targeting mitochondrial dysfunction through innovative therapeutic approaches, including antioxidants, metabolic improvement, biogenesis promotion, mitophagy enhancement, mitochondrial transfer, and traditional Chinese medicine. These strategies hold promise as effective means to mitigate age-related fertility decline and preserve ovarian health. Drawing insights from advanced researches in the field, this review provides a deeper understanding of the intricate interplay between mitochondrial function and ovarian aging, offering valuable perspectives for the development of novel therapeutic interventions aimed at preserving fertility and enhancing overall reproductive health.
Collapse
Affiliation(s)
- Zi-Han Wang
- Department of Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Jinan Key Laboratory of Diagnosis and Treatment of Major Gynecological Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhen-Jing Wang
- Center for Reproductive Medicine, Shandong University, Jinan, China
| | - Huai-Chao Liu
- Department of Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Jinan Key Laboratory of Diagnosis and Treatment of Major Gynecological Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Chen-Yu Wang
- Department of Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Jinan Key Laboratory of Diagnosis and Treatment of Major Gynecological Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yu-Qi Wang
- Department of Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Jinan Key Laboratory of Diagnosis and Treatment of Major Gynecological Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yang Yue
- Department of Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Jinan Key Laboratory of Diagnosis and Treatment of Major Gynecological Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Chen Zhao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guoyun Wang
- Department of Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Jinan Key Laboratory of Diagnosis and Treatment of Major Gynecological Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ji-Peng Wan
- Department of Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Jinan Key Laboratory of Diagnosis and Treatment of Major Gynecological Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
9
|
Sizek H, Deritei D, Fleig K, Harris M, Regan PL, Glass K, Regan ER. Unlocking Mitochondrial Dysfunction-Associated Senescence (MiDAS) with NAD + - a Boolean Model of Mitochondrial Dynamics and Cell Cycle Control. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.18.572194. [PMID: 38187609 PMCID: PMC10769269 DOI: 10.1101/2023.12.18.572194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The steady accumulation of senescent cells with aging creates tissue environments that aid cancer evolution. Aging cell states are highly heterogeneous. 'Deep senescent' cells rely on healthy mitochondria to fuel a strong proinflammatory secretome, including cytokines, growth and transforming signals. Yet, the physiological triggers of senescence such as the reactive oxygen species (ROS) can also trigger mitochondrial dysfunction, and sufficient energy deficit to alter their secretome and cause chronic oxidative stress - a state termed Mitochondrial Dysfunction-Associated Senescence (MiDAS). Here, we offer a mechanistic hypothesis for the molecular processes leading to MiDAS, along with testable predictions. To do this we have built a Boolean regulatory network model that qualitatively captures key aspects of mitochondrial dynamics during cell cycle progression (hyper-fusion at the G1/S boundary, fission in mitosis), apoptosis (fission and dysfunction) and glucose starvation (reversible hyper-fusion), as well as MiDAS in response to SIRT3 knockdown or oxidative stress. Our model reaffirms the protective role of NAD + and external pyruvate. We offer testable predictions about the growth factor- and glucose-dependence of MiDAS and its reversibility at different stages of reactive oxygen species (ROS)-induced senescence. Our model provides mechanistic insights into the distinct stages of DNA-damage induced senescence, the relationship between senescence and epithelial-to-mesenchymal transition in cancer and offers a foundation for building multiscale models of tissue aging. Highlights Boolean regulatory network model reproduces mitochondrial dynamics during cell cycle progression, apoptosis, and glucose starvation. Model offers a mechanistic explanation for the positive feedback loop that locks in Mitochondrial Dysfunction-Associated Senescence (MiDAS), involving autophagy-resistant, hyperfused, dysfunctional mitochondria. Model reproduces ROS-mediated mitochondrial dysfunction and suggests that MiDAS is part of the early phase of damage-induced senescence. Model predicts that cancer-driving mutations that bypass the G1/S checkpoint generally increase the incidence of MiDAS, except for p53 loss.
Collapse
|
10
|
Cai Y, Han Z, Cheng H, Li H, Wang K, Chen J, Liu ZX, Xie Y, Lin Y, Zhou S, Wang S, Zhou X, Jin S. The impact of ageing mechanisms on musculoskeletal system diseases in the elderly. Front Immunol 2024; 15:1405621. [PMID: 38774874 PMCID: PMC11106385 DOI: 10.3389/fimmu.2024.1405621] [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: 03/23/2024] [Accepted: 04/22/2024] [Indexed: 05/24/2024] Open
Abstract
Ageing is an inevitable process that affects various tissues and organs of the human body, leading to a series of physiological and pathological changes. Mechanisms such as telomere depletion, stem cell depletion, macrophage dysfunction, and cellular senescence gradually manifest in the body, significantly increasing the incidence of diseases in elderly individuals. These mechanisms interact with each other, profoundly impacting the quality of life of older adults. As the ageing population continues to grow, the burden on the public health system is expected to intensify. Globally, the prevalence of musculoskeletal system diseases in elderly individuals is increasing, resulting in reduced limb mobility and prolonged suffering. This review aims to elucidate the mechanisms of ageing and their interplay while exploring their impact on diseases such as osteoarthritis, osteoporosis, and sarcopenia. By delving into the mechanisms of ageing, further research can be conducted to prevent and mitigate its effects, with the ultimate goal of alleviating the suffering of elderly patients in the future.
Collapse
Affiliation(s)
- Yijin Cai
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhongyu Han
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hong Cheng
- School of Automation Engineering, University of Electronic Science and Technology, Chengdu, China
| | - Hongpeng Li
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ke Wang
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jia Chen
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhi-Xiang Liu
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yulong Xie
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yumeng Lin
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shuwei Zhou
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Siyu Wang
- Department of Gastroenterology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Xiao Zhou
- Second Clinical Medical College, Heilongjiang University of Chinese Medicine, Heilongjiang, China
| | - Song Jin
- Department of Rehabilitation, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
11
|
Ansari MM, Ghosh M, Lee DS, Son YO. Senolytic therapeutics: An emerging treatment modality for osteoarthritis. Ageing Res Rev 2024; 96:102275. [PMID: 38494091 DOI: 10.1016/j.arr.2024.102275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/15/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
Osteoarthritis (OA), a chronic joint disease affecting millions of people aged over 65 years, is the main musculoskeletal cause of diminished joint mobility in the elderly. It is characterized by lingering pain and increasing deterioration of articular cartilage. Aging and accumulation of senescent cells (SCs) in the joints are frequently associated with OA. Apoptosis resistance; irreversible cell cycle arrest; increased p16INK4a expression, secretion of senescence-associated secretory phenotype factors, senescence-associated β-galactosidase levels, secretion of extracellular vesicles, and levels of reactive oxygen and reactive nitrogen species; and mitochondrial dysregulation are some common changes in cellular senescence in joint tissues. Development of OA correlates with an increase in the density of SCs in joint tissues. Senescence-associated secretory phenotype has been linked to OA and cartilage breakdown. Senolytics and therapeutic pharmaceuticals are being focused upon for OA management. SCs can be selectively eliminated or killed by senolytics to halt the pathogenesis and progression of OA. Comprehensive understanding of how aging affects joint dysfunction will benefit OA patients. Here, we discuss age-related mechanisms associated with OA pathogenesis and senolytics as an emerging modality in the management of age-related SCs and pathogenesis of OA in preclinical and clinical studies.
Collapse
Affiliation(s)
- Md Meraj Ansari
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju-si 63243, Republic of Korea
| | - Mrinmoy Ghosh
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju-si 63243, Republic of Korea; Department of Biotechnology, School of Bio, Chemical and Processing Engineering (SBCE), Kalasalingam Academy of Research and Education, Krishnankoil 626126, India
| | - Dong-Sun Lee
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju-si 63243, Republic of Korea; Bio-Health Materials Core-Facility Center, Jeju National University, Jeju 63243, Republic of Korea; Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju-si 63243, Republic of Korea; Practical Translational Research Center, Jeju National University, Jeju 63243, Republic of Korea.
| | - Young-Ok Son
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju-si 63243, Republic of Korea; Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju-si 63243, Republic of Korea; Bio-Health Materials Core-Facility Center, Jeju National University, Jeju 63243, Republic of Korea; Practical Translational Research Center, Jeju National University, Jeju 63243, Republic of Korea.
| |
Collapse
|
12
|
Chen J, Zhang H, Yi X, Dou Q, Yang X, He Y, Chen J, Chen K. Cellular senescence of renal tubular epithelial cells in acute kidney injury. Cell Death Discov 2024; 10:62. [PMID: 38316761 PMCID: PMC10844256 DOI: 10.1038/s41420-024-01831-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 01/14/2024] [Accepted: 01/24/2024] [Indexed: 02/07/2024] Open
Abstract
Cellular senescence represents an irreversible state of cell-cycle arrest during which cells secrete senescence-associated secretory phenotypes, including inflammatory factors and chemokines. Additionally, these cells exhibit an apoptotic resistance phenotype. Cellular senescence serves a pivotal role not only in embryonic development, tissue regeneration, and tumor suppression but also in the pathogenesis of age-related degenerative diseases, malignancies, metabolic diseases, and kidney diseases. The senescence of renal tubular epithelial cells (RTEC) constitutes a critical cellular event in the progression of acute kidney injury (AKI). RTEC senescence inhibits renal regeneration and repair processes and, concurrently, promotes the transition of AKI to chronic kidney disease via the senescence-associated secretory phenotype. The mechanisms underlying cellular senescence are multifaceted and include telomere shortening or damage, DNA damage, mitochondrial autophagy deficiency, cellular metabolic disorders, endoplasmic reticulum stress, and epigenetic regulation. Strategies aimed at inhibiting RTEC senescence, targeting the clearance of senescent RTEC, or promoting the apoptosis of senescent RTEC hold promise for enhancing the renal prognosis of AKI. This review primarily focuses on the characteristics and mechanisms of RTEC senescence, and the impact of intervening RTEC senescence on the prognosis of AKI, aiming to provide a foundation for understanding the pathogenesis and providing potentially effective approaches for AKI treatment.
Collapse
Affiliation(s)
- Juan Chen
- Department of Nephrology, Daping Hospital, Army Medical University, 400042, Chongqing, China
| | - Huhai Zhang
- Department of Nephrology, Southwest Hospital, Army Medical University, 400042, Chongqing, China
| | - Xiangling Yi
- Department of Nephrology, Daping Hospital, Army Medical University, 400042, Chongqing, China
| | - Qian Dou
- Department of Nephrology, Daping Hospital, Army Medical University, 400042, Chongqing, China
| | - Xin Yang
- Department of Nephrology, Daping Hospital, Army Medical University, 400042, Chongqing, China
| | - Yani He
- Department of Nephrology, Daping Hospital, Army Medical University, 400042, Chongqing, China
| | - Jia Chen
- Department of Nephrology, Daping Hospital, Army Medical University, 400042, Chongqing, China.
| | - Kehong Chen
- Department of Nephrology, Daping Hospital, Army Medical University, 400042, Chongqing, China.
- State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, China.
| |
Collapse
|
13
|
Buss LG, Rheinheimer BA, Limesand KH. Radiation-induced changes in energy metabolism result in mitochondrial dysfunction in salivary glands. Sci Rep 2024; 14:845. [PMID: 38191641 PMCID: PMC10774336 DOI: 10.1038/s41598-023-50877-9] [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: 06/29/2023] [Accepted: 12/27/2023] [Indexed: 01/10/2024] Open
Abstract
Salivary glands are indirectly damaged during radiotherapy for head and neck cancer, resulting in acute and chronic hyposalivation. Current treatments for radiation-induced hyposalivation do not permanently restore function to the gland; therefore, more mechanistic understanding of the damage response is needed to identify therapeutic targets for lasting restoration. Energy metabolism reprogramming has been observed in cancer and wound healing models to provide necessary fuel for cell proliferation; however, there is limited understanding of alterations in energy metabolism reprogramming in tissues that fail to heal. We measured extracellular acidification and oxygen consumption rates, assessed mitochondrial DNA copy number, and tested fuel dependency of irradiated primary salivary acinar cells. Radiation treatment leads to increases in glycolytic flux, oxidative phosphorylation, and ATP production rate at acute and intermediate time points. In contrast, at chronic radiation time points there is a significant decrease in glycolytic flux, oxidative phosphorylation, and ATP production rate. Irradiated salivary glands exhibit significant decreases in spare respiratory capacity and increases in mitochondrial DNA copy number at days 5 and 30 post-treatment, suggesting a mitochondrial dysfunction phenotype. These results elucidate kinetic changes in energy metabolism reprogramming of irradiated salivary glands that may underscore the chronic loss of function phenotype.
Collapse
Affiliation(s)
- Lauren G Buss
- School of Nutritional Sciences and Wellness, University of Arizona, 1177 E 4th St, Shantz Building Room 421, Tucson, AZ, USA
| | - Brenna A Rheinheimer
- School of Nutritional Sciences and Wellness, University of Arizona, 1177 E 4th St, Shantz Building Room 421, Tucson, AZ, USA
| | - Kirsten H Limesand
- School of Nutritional Sciences and Wellness, University of Arizona, 1177 E 4th St, Shantz Building Room 421, Tucson, AZ, USA.
- University of Arizona Cancer Center, Tucson, AZ, USA.
| |
Collapse
|
14
|
Tenchov R, Sasso JM, Wang X, Zhou QA. Aging Hallmarks and Progression and Age-Related Diseases: A Landscape View of Research Advancement. ACS Chem Neurosci 2024; 15:1-30. [PMID: 38095562 PMCID: PMC10767750 DOI: 10.1021/acschemneuro.3c00531] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 01/04/2024] Open
Abstract
Aging is a dynamic, time-dependent process that is characterized by a gradual accumulation of cell damage. Continual functional decline in the intrinsic ability of living organisms to accurately regulate homeostasis leads to increased susceptibility and vulnerability to diseases. Many efforts have been put forth to understand and prevent the effects of aging. Thus, the major cellular and molecular hallmarks of aging have been identified, and their relationships to age-related diseases and malfunctions have been explored. Here, we use data from the CAS Content Collection to analyze the publication landscape of recent aging-related research. We review the advances in knowledge and delineate trends in research advancements on aging factors and attributes across time and geography. We also review the current concepts related to the major aging hallmarks on the molecular, cellular, and organismic level, age-associated diseases, with attention to brain aging and brain health, as well as the major biochemical processes associated with aging. Major age-related diseases have been outlined, and their correlations with the major aging features and attributes are explored. We hope this review will be helpful for apprehending the current knowledge in the field of aging mechanisms and progression, in an effort to further solve the remaining challenges and fulfill its potential.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS, a Division of the American Chemical
Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Janet M. Sasso
- CAS, a Division of the American Chemical
Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Xinmei Wang
- CAS, a Division of the American Chemical
Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Qiongqiong Angela Zhou
- CAS, a Division of the American Chemical
Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| |
Collapse
|
15
|
Woronzow V, Möhner J, Remane D, Zischler H. Generation of somatic de novo structural variation as a hallmark of cellular senescence in human lung fibroblasts. Front Cell Dev Biol 2023; 11:1274807. [PMID: 38152346 PMCID: PMC10751365 DOI: 10.3389/fcell.2023.1274807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 11/29/2023] [Indexed: 12/29/2023] Open
Abstract
Cellular senescence is characterized by replication arrest in response to stress stimuli. Senescent cells accumulate in aging tissues and can trigger organ-specific and possibly systemic dysfunction. Although senescent cell populations are heterogeneous, a key feature is that they exhibit epigenetic changes. Epigenetic changes such as loss of repressive constitutive heterochromatin could lead to subsequent LINE-1 derepression, a phenomenon often described in the context of senescence or somatic evolution. LINE-1 elements decode the retroposition machinery and reverse transcription generates cDNA from autonomous and non-autonomous TEs that can potentially reintegrate into genomes and cause structural variants. Another feature of cellular senescence is mitochondrial dysfunction caused by mitochondrial damage. In combination with impaired mitophagy, which is characteristic of senescent cells, this could lead to cytosolic mtDNA accumulation and, as a genomic consequence, integrations of mtDNA into nuclear DNA (nDNA), resulting in mitochondrial pseudogenes called numts. Thus, both phenomena could cause structural variants in aging genomes that go beyond epigenetic changes. We therefore compared proliferating and senescent IMR-90 cells in terms of somatic de novo numts and integrations of a non-autonomous composite retrotransposons - the so-called SVA elements-that hijack the retropositional machinery of LINE-1. We applied a subtractive and kinetic enrichment technique using proliferating cell DNA as a driver and senescent genomes as a tester for the detection of nuclear flanks of de novo SVA integrations. Coupled with deep sequencing we obtained a genomic readout for SVA retrotransposition possibly linked to cellular senescence in the IMR-90 model. Furthermore, we compared the genomes of proliferative and senescent IMR-90 cells by deep sequencing or after enrichment of nuclear DNA using AluScan technology. A total of 1,695 de novo SVA integrations were detected in senescent IMR-90 cells, of which 333 were unique. Moreover, we identified a total of 81 de novo numts with perfect identity to both mtDNA and nuclear hg38 flanks. In summary, we present evidence for possible age-dependent structural genomic changes by paralogization that go beyond epigenetic modifications. We hypothesize, that the structural variants we observe potentially impact processes associated with replicative aging of IMR-90 cells.
Collapse
Affiliation(s)
- Valentina Woronzow
- Division of Anthropology, Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Jonas Möhner
- Division of Anthropology, Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Daniel Remane
- Division of Anthropology, Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg University Mainz, Mainz, Germany
- HOX Life Science GmbH, Frankfurt, Hessen, Germany
| | - Hans Zischler
- Division of Anthropology, Institute of Organismic and Molecular Evolution, Faculty of Biology, Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
16
|
Buss LG, Rheinheimer BA, Limesand KH. Radiation-Induced Changes in Energy Metabolism Result in Mitochondrial Dysfunction in Salivary Glands. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.27.568879. [PMID: 38077038 PMCID: PMC10705263 DOI: 10.1101/2023.11.27.568879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Salivary glands are indirectly damaged during radiotherapy for head and neck cancer, resulting in acute and chronic hyposalivation. Current treatments for radiation-induced hyposalivation do not permanently restore function to the gland; therefore, more mechanistic understanding of the damage response is needed to identify therapeutic targets for lasting restoration. Energy metabolism reprogramming has been observed in cancer and wound healing models to provide necessary fuel for cell proliferation; however, there is limited understanding of alterations in energy metabolism reprogramming in tissues that fail to heal. We measured extracellular acidification and oxygen consumption rates, assessed mitochondrial DNA copy number, and tested fuel dependency of irradiated primary salivary acinar cells. Radiation treatment leads to increases in glycolytic flux, oxidative phosphorylation, and ATP production rate at acute and intermediate time points. In contrast, at chronic radiation time points there is a significant decrease in glycolytic flux, oxidative phosphorylation, and ATP production rate. Irradiated salivary glands exhibit significant decreases in spare respiratory capacity and increases in mitochondrial DNA copy number at days 5 and 30 post-treatment, suggesting a mitochondrial dysfunction phenotype. These results elucidate kinetic changes in energy metabolism reprogramming of irradiated salivary glands that may underscore the chronic loss of function phenotype.
Collapse
|
17
|
Pal C. Small-molecule redox modulators with anticancer activity: A comprehensive mechanistic update. Free Radic Biol Med 2023; 209:211-227. [PMID: 37898387 DOI: 10.1016/j.freeradbiomed.2023.10.406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/27/2023] [Accepted: 10/25/2023] [Indexed: 10/30/2023]
Abstract
The pursuit of effective anticancer therapies has led to a burgeoning interest in the realm of redox modulation. This review provides a comprehensive exploration of the intricate mechanisms by which diverse anticancer molecules leverage redox pathways for therapeutic intervention. Redox modulation, encompassing the fine balance of oxidation-reduction processes within cells, has emerged as a pivotal player in cancer treatment. This review delves into the multifaceted mechanisms of action employed by various anticancer compounds, including small molecules and natural products, to disrupt cancer cell proliferation and survival. Beginning with an examination of the role of redox signaling in cancer development and resistance, the review highlights how aberrant redox dynamics can fuel tumorigenesis. It then meticulously dissects the strategies employed by anticancer agents to induce oxidative stress, perturb redox equilibrium, and trigger apoptosis within cancer cells. Furthermore, the review explores the challenges and potential side effects associated with redox-based treatments, along with the development of novel redox-targeted agents. In summary, this review offers a profound understanding of the dynamic interplay between redox modulation and anticancer molecules, presenting promising avenues to revolutionize cancer therapy and enhance patient outcomes.
Collapse
Affiliation(s)
- Chinmay Pal
- Department of Chemistry, Gobardanga Hindu College, North 24 Parganas, West Bengal, 743273, India.
| |
Collapse
|
18
|
Sullivan DI, Bello FM, Silva AG, Redding KM, Giordano L, Hinchie AM, Loughridge KE, Mora AL, Königshoff M, Kaufman BA, Jurczak MJ, Alder JK. Intact mitochondrial function in the setting of telomere-induced senescence. Aging Cell 2023; 22:e13941. [PMID: 37688329 PMCID: PMC10577573 DOI: 10.1111/acel.13941] [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: 04/17/2023] [Revised: 06/25/2023] [Accepted: 07/07/2023] [Indexed: 09/10/2023] Open
Abstract
Mitochondria play essential roles in metabolic support and signaling within all cells. Congenital and acquired defects in mitochondria are responsible for several pathologies, including premature entrance to cellar senescence. Conversely, we examined the consequences of dysfunctional telomere-driven cellular senescence on mitochondrial biogenesis and function. We drove senescence in vitro and in vivo by deleting the telomere-binding protein TRF2 in fibroblasts and hepatocytes, respectively. Deletion of TRF2 led to a robust DNA damage response, global changes in transcription, and induction of cellular senescence. In vitro, senescent cells had significant increases in mitochondrial respiratory capacity driven by increased cellular and mitochondrial volume. Hepatocytes with dysfunctional telomeres maintained their mitochondrial respiratory capacity in vivo, whether measured in intact cells or purified mitochondria. Induction of senescence led to the upregulation of overlapping and distinct genes in fibroblasts and hepatocytes, but transcripts related to mitochondria were preserved. Our results support that mitochondrial function and activity are preserved in telomere dysfunction-induced senescence, which may facilitate continued cellular functions.
Collapse
Affiliation(s)
- Daniel I. Sullivan
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, Division of Pulmonary, Allergy, and Critical Care MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Fiona M. Bello
- Division of Endocrinology and MetabolismUniversity of PittsburghPittsburghPennsylvaniaUSA
- Center for Metabolism and Mitochondrial MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Agustin Gil Silva
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, Division of Pulmonary, Allergy, and Critical Care MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Kevin M. Redding
- Center for Metabolism and Mitochondrial MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Heart, Lung, and Blood Vascular Medicine InstituteUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Luca Giordano
- Center for Metabolism and Mitochondrial MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Heart, Lung, and Blood Vascular Medicine InstituteUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Angela M. Hinchie
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, Division of Pulmonary, Allergy, and Critical Care MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Kelly E. Loughridge
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, Division of Pulmonary, Allergy, and Critical Care MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Ana L. Mora
- Division of Pulmonary, Critical Care and Sleep Medicine, Davis Heart Lung Research InstituteThe Ohio State UniversityColumbusOhioUSA
| | - Melanie Königshoff
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, Division of Pulmonary, Allergy, and Critical Care MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Brett A. Kaufman
- Center for Metabolism and Mitochondrial MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
- Heart, Lung, and Blood Vascular Medicine InstituteUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Michael J. Jurczak
- Division of Endocrinology and MetabolismUniversity of PittsburghPittsburghPennsylvaniaUSA
- Center for Metabolism and Mitochondrial MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Jonathan K. Alder
- Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, Division of Pulmonary, Allergy, and Critical Care MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
19
|
Victorelli S, Salmonowicz H, Chapman J, Martini H, Vizioli MG, Riley JS, Cloix C, Hall-Younger E, Machado Espindola-Netto J, Jurk D, Lagnado AB, Sales Gomez L, Farr JN, Saul D, Reed R, Kelly G, Eppard M, Greaves LC, Dou Z, Pirius N, Szczepanowska K, Porritt RA, Huang H, Huang TY, Mann DA, Masuda CA, Khosla S, Dai H, Kaufmann SH, Zacharioudakis E, Gavathiotis E, LeBrasseur NK, Lei X, Sainz AG, Korolchuk VI, Adams PD, Shadel GS, Tait SWG, Passos JF. Apoptotic stress causes mtDNA release during senescence and drives the SASP. Nature 2023; 622:627-636. [PMID: 37821702 PMCID: PMC10584674 DOI: 10.1038/s41586-023-06621-4] [Citation(s) in RCA: 102] [Impact Index Per Article: 102.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 09/07/2023] [Indexed: 10/13/2023]
Abstract
Senescent cells drive age-related tissue dysfunction partially through the induction of a chronic senescence-associated secretory phenotype (SASP)1. Mitochondria are major regulators of the SASP; however, the underlying mechanisms have not been elucidated2. Mitochondria are often essential for apoptosis, a cell fate distinct from cellular senescence. During apoptosis, widespread mitochondrial outer membrane permeabilization (MOMP) commits a cell to die3. Here we find that MOMP occurring in a subset of mitochondria is a feature of cellular senescence. This process, called minority MOMP (miMOMP), requires BAX and BAK macropores enabling the release of mitochondrial DNA (mtDNA) into the cytosol. Cytosolic mtDNA in turn activates the cGAS-STING pathway, a major regulator of the SASP. We find that inhibition of MOMP in vivo decreases inflammatory markers and improves healthspan in aged mice. Our results reveal that apoptosis and senescence are regulated by similar mitochondria-dependent mechanisms and that sublethal mitochondrial apoptotic stress is a major driver of the SASP. We provide proof-of-concept that inhibition of miMOMP-induced inflammation may be a therapeutic route to improve healthspan.
Collapse
Affiliation(s)
- Stella Victorelli
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Hanna Salmonowicz
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Biosciences Institute, Faculty of Medical Sciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
- ReMedy International Research Agenda Unit, IMol Polish Academy of Sciences, Warsaw, Poland
| | - James Chapman
- Biosciences Institute, Faculty of Medical Sciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Helene Martini
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Maria Grazia Vizioli
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Joel S Riley
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
- Institute of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Catherine Cloix
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Ella Hall-Younger
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | | - Diana Jurk
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Anthony B Lagnado
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Lilian Sales Gomez
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Joshua N Farr
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Dominik Saul
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Rebecca Reed
- Biosciences Institute, Faculty of Medical Sciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - George Kelly
- Biosciences Institute, Faculty of Medical Sciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Madeline Eppard
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Laura C Greaves
- Wellcome Centre for Mitochondrial Research, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Zhixun Dou
- Center for Regenerative Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Nicholas Pirius
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Karolina Szczepanowska
- ReMedy International Research Agenda Unit, IMol Polish Academy of Sciences, Warsaw, Poland
| | - Rebecca A Porritt
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Huijie Huang
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Timothy Y Huang
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Derek A Mann
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
- Department of Gastroenterology and Hepatology, School of Medicine, Koç University, Istanbul, Turkey
| | - Claudio Akio Masuda
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sundeep Khosla
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Haiming Dai
- Division of Oncology Research and Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Scott H Kaufmann
- Division of Oncology Research and Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Emmanouil Zacharioudakis
- Department of Biochemistry, Department of Medicine, Montefiore Einstein Cancer Center, Wilf Family Cardiovascular Research Institute, Institute for Aging Research, Albert Einstein College of Medicine, New York, NY, USA
| | - Evripidis Gavathiotis
- Department of Biochemistry, Department of Medicine, Montefiore Einstein Cancer Center, Wilf Family Cardiovascular Research Institute, Institute for Aging Research, Albert Einstein College of Medicine, New York, NY, USA
| | | | - Xue Lei
- Cancer Genome and Epigenetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Alva G Sainz
- Salk Institute for Biological Studies, La Jolla, CA, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Viktor I Korolchuk
- Biosciences Institute, Faculty of Medical Sciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Peter D Adams
- Cancer Genome and Epigenetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | | | - Stephen W G Tait
- Cancer Research UK Scotland Institute, Glasgow, UK.
- School of Cancer Sciences, University of Glasgow, Glasgow, UK.
| | - João F Passos
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
20
|
Kim JY, Atanassov I, Dethloff F, Kroczek L, Langer T. Time-resolved proteomic analyses of senescence highlight metabolic rewiring of mitochondria. Life Sci Alliance 2023; 6:e202302127. [PMID: 37321846 PMCID: PMC10272782 DOI: 10.26508/lsa.202302127] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 06/17/2023] Open
Abstract
Mitochondrial dysfunction and cellular senescence are hallmarks of aging. However, the relationship between these two phenomena remains incompletely understood. In this study, we investigated the rewiring of mitochondria upon development of the senescent state in human IMR90 fibroblasts. Determining the bioenergetic activities and abundance of mitochondria, we demonstrate that senescent cells accumulate mitochondria with reduced OXPHOS activity, resulting in an overall increase of mitochondrial activities in senescent cells. Time-resolved proteomic analyses revealed extensive reprogramming of the mitochondrial proteome upon senescence development and allowed the identification of metabolic pathways that are rewired with different kinetics upon establishment of the senescent state. Among the early responding pathways, the degradation of branched-chain amino acid was increased, whereas the one carbon folate metabolism was decreased. Late-responding pathways include lipid metabolism and mitochondrial translation. These signatures were confirmed by metabolic flux analyses, highlighting metabolic rewiring as a central feature of mitochondria in cellular senescence. Together, our data provide a comprehensive view on the changes in mitochondrial proteome in senescent cells and reveal how the mitochondrial metabolism is rewired in senescent cells.
Collapse
Affiliation(s)
- Jun Yong Kim
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Ilian Atanassov
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | | | - Lara Kroczek
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Thomas Langer
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| |
Collapse
|
21
|
Nehlin JO. Senolytic and senomorphic interventions to defy senescence-associated mitochondrial dysfunction. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 136:217-247. [PMID: 37437979 DOI: 10.1016/bs.apcsb.2023.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
The accumulation of senescent cells in the aging individual is associated with an increase in the occurrence of age-associated pathologies that contribute to poor health, frailty, and mortality. The number and type of senescent cells is viewed as a contributor to the body's senescence burden. Cellular models of senescence are based on induction of senescence in cultured cells in the laboratory. One type of senescence is triggered by mitochondrial dysfunction. There are several indications that mitochondria defects contribute to body aging. Senotherapeutics, targeting senescent cells, have been shown to induce their lysis by means of senolytics, or repress expression of their secretome, by means of senomorphics, senostatics or gerosuppressors. An outline of the mechanism of action of various senotherapeutics targeting mitochondria and senescence-associated mitochondria dysfunction will be here addressed. The combination of geroprotective interventions together with senotherapeutics will help to strengthen mitochondrial energy metabolism, biogenesis and turnover, and lengthen the mitochondria healthspan, minimizing one of several molecular pathways contributing to the aging phenotype.
Collapse
Affiliation(s)
- Jan O Nehlin
- Department of Clinical Research, Copenhagen University Hospital, Amager and Hvidovre, Hvidovre, Denmark.
| |
Collapse
|
22
|
Chen Q, Young L, Barsotti R. Mitochondria in cell senescence: A Friend or Foe? ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 136:35-91. [PMID: 37437984 DOI: 10.1016/bs.apcsb.2023.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Cell senescence denotes cell growth arrest in response to continuous replication or stresses damaging DNA or mitochondria. Mounting research suggests that cell senescence attributes to aging-associated failing organ function and diseases. Conversely, it participates in embryonic tissue maturation, wound healing, tissue regeneration, and tumor suppression. The acute or chronic properties and microenvironment may explain the double faces of senescence. Senescent cells display unique characteristics. In particular, its mitochondria become elongated with altered metabolomes and dynamics. Accordingly, mitochondria reform their function to produce more reactive oxygen species at the cost of low ATP production. Meanwhile, destructed mitochondrial unfolded protein responses further break the delicate proteostasis fostering mitochondrial dysfunction. Additionally, the release of mitochondrial damage-associated molecular patterns, mitochondrial Ca2+ overload, and altered NAD+ level intertwine other cellular organelle strengthening senescence. These findings further intrigue researchers to develop anti-senescence interventions. Applying mitochondrial-targeted antioxidants reduces cell senescence and mitigates aging by restoring mitochondrial function and attenuating oxidative stress. Metformin and caloric restriction also manifest senescent rescuing effects by increasing mitochondria efficiency and alleviating oxidative damage. On the other hand, Bcl2 family protein inhibitors eradicate senescent cells by inducing apoptosis to facilitate cancer chemotherapy. This review describes the different aspects of mitochondrial changes in senescence and highlights the recent progress of some anti-senescence strategies.
Collapse
Affiliation(s)
- Qian Chen
- Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States.
| | - Lindon Young
- Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States
| | - Robert Barsotti
- Philadelphia College of Osteopathic Medicine, Philadelphia, PA, United States
| |
Collapse
|
23
|
Salis HM. Genetic circuitry boosts cell longevity. Science 2023; 380:343. [PMID: 37104573 DOI: 10.1126/science.adh4872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Reprogramming cellular dynamics is used to study and delay the onset of aging in yeast.
Collapse
Affiliation(s)
- Howard M Salis
- Departments of Agricultural and Biological Engineering, Chemical Engineering, and Biomedical Engineering, Bioinformatics and Genomics Program, Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
24
|
Matsuyama S, Nakamura S, Minabe S, Sakatani M, Takenouchi N, Sasaki T, Inoue Y, Iwata H, Kimura K. Deterioration of mitochondrial biogenesis and degradation in the endometrium is a cause of subfertility in cows. Mol Reprod Dev 2023; 90:141-152. [PMID: 36645869 DOI: 10.1002/mrd.23670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 12/02/2022] [Accepted: 01/05/2023] [Indexed: 01/18/2023]
Abstract
To investigate possible causes of reproductive failure, we conducted global endometrial gene expression analyses in fertile and subfertile cows. Ingenuity pathway analysis showed that RICTOR and SIRT3 are significant upstream regulators for highly expressed genes in fertile cows, and are predicted to be activated upstream regulators of normal mitochondrial respiration. Canonical pathway analysis revealed that these highly expressed genes are involved in the activation of mitochondrial oxidative phosphorylation. Therefore, in subfertile cows, the inactivation of RICTOR and SIRT3 may correlate with decreased capacity of mitochondrial respiration. Furthermore, the expression levels of most mitochondrial DNA genes and nuclear genes encoding mitochondrial proteins were higher in subfertile cows. The mitochondrial DNA copy number was significantly higher in the endometrium of subfertile cows, whereas the ATP content did not differ between fertile and subfertile cows. Quantitative reverse transcription-PCR analysis demonstrated that the expression of PGC1a, TFAM, MFN1, FIS1, and BCL2L13 were significantly lower in subfertile cows. In addition, transmission electron microscopy images showed mitochondrial swelling in the endometrial cells of the subfertile cow. These results suggest that poor-quality mitochondria accumulate in the endometrium owing to a reduced capacity for mitochondrial biogenesis, fusion, fission, and degradation in subfertile cows, and may contribute to infertility.
Collapse
Affiliation(s)
- Shuichi Matsuyama
- Division of Animal Feeding and Management Research, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization (NARO), Nasushiobara, Japan
- Laboratory of Animal Production Science, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Sho Nakamura
- Division of Animal Feeding and Management Research, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization (NARO), Nasushiobara, Japan
- Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Japan
| | - Shiori Minabe
- Division of Animal Feeding and Management Research, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization (NARO), Nasushiobara, Japan
| | - Miki Sakatani
- Division of Animal Feeding and Management Research, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization (NARO), Nasushiobara, Japan
- Livestock and Grassland Research Division, Kyushu Okinawa Agricultural Research Center, National Agriculture and Food Research Organization (NARO), Kumamoto, Japan
| | - Naoki Takenouchi
- Livestock and Grassland Research Division, Kyushu Okinawa Agricultural Research Center, National Agriculture and Food Research Organization (NARO), Kumamoto, Japan
| | - Takuya Sasaki
- Laboratory of Animal Production Science, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
- Okayama A.I. Center, Livestock Improvement Association of Japan Inc., Maebashi, Japan
| | - Yuki Inoue
- Laboratory of Animal Reproduction, Department of Animal Science, Tokyo University of Agriculture, Setagaya, Kanagawa, Japan
| | - Hisataka Iwata
- Laboratory of Animal Reproduction, Department of Animal Science, Tokyo University of Agriculture, Setagaya, Kanagawa, Japan
| | - Koji Kimura
- Laboratory of Reproductive Physiology, Graduate School of Environmental and Life Science, Okayama University, Okayama, Japan
| |
Collapse
|
25
|
Nabuco Leva Ferreira de Freitas JA, Bischof O. Dynamic modeling of the cellular senescence gene regulatory network. Heliyon 2023; 9:e14007. [PMID: 36938415 PMCID: PMC10015196 DOI: 10.1016/j.heliyon.2023.e14007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 02/13/2023] [Accepted: 02/17/2023] [Indexed: 02/27/2023] Open
Abstract
Cellular senescence is a cell fate that prominently impacts physiological and pathophysiological processes. Diverse cellular stresses induce it, and dramatic gene expression changes accompany it. However, determining the interactions comprising the gene regulatory network (GRN) governing senescence remains challenging. Recent advances in signal processing techniques provide opportunities to reconstruct GRNs. Here, we describe a GRN for senescence integrating time-series transcriptome and transcription factor depletion datasets. Specifically, we infer a set of differential equations using the "Sparse Identification of Nonlinear Dynamics" (SINDy) algorithm, discriminate genes with potential hidden regulators, validate the inferred GRN for time-points not included in the training data, and comprehensively benchmark our approach. Our work is a proof of concept for a data-driven GRN reconstruction method, consolidating an iterative, powerful mathematical platform for senescence modeling that can be used to test hypotheses in silico and has the potential for future discoveries of clinical impact.
Collapse
Affiliation(s)
- José Américo Nabuco Leva Ferreira de Freitas
- IMRB, Mondor Institute for Biomedical Research, INSERM U955 – Université Paris Est Créteil, UPEC, Faculté de Médecine de Créteil 8, rue du Général Sarrail, 94010 Créteil
- Sorbonne Université, UMR 8256, Biological Adaptation and Ageing B2A–IBPS, F-75005, Paris, France
- INSERM U1164, F-75005, Paris, France
| | - Oliver Bischof
- IMRB, Mondor Institute for Biomedical Research, INSERM U955 – Université Paris Est Créteil, UPEC, Faculté de Médecine de Créteil 8, rue du Général Sarrail, 94010 Créteil
- Corresponding author.
| |
Collapse
|
26
|
Martini H, Passos JF. Cellular senescence: all roads lead to mitochondria. FEBS J 2023; 290:1186-1202. [PMID: 35048548 PMCID: PMC9296701 DOI: 10.1111/febs.16361] [Citation(s) in RCA: 99] [Impact Index Per Article: 99.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/11/2022] [Accepted: 01/18/2022] [Indexed: 01/10/2023]
Abstract
Senescence is a multi-functional cell fate, characterized by an irreversible cell-cycle arrest and a pro-inflammatory phenotype, commonly known as the senescence-associated secretory phenotype (SASP). Emerging evidence indicates that accumulation of senescent cells in multiple tissues drives tissue dysfunction and several age-related conditions. This has spurred the academic community and industry to identify new therapeutic interventions targeting this process. Mitochondrial dysfunction is an often-unappreciated hallmark of cellular senescence which plays important roles not only in the senescence growth arrest but also in the development of the SASP and resistance to cell-death. Here, we review the evidence that supports a role for mitochondria in the development of senescence and describe the underlying mechanisms. Finally, we propose that a detailed road map of mitochondrial biology in senescence will be crucial to guide the future development of senotherapies.
Collapse
Affiliation(s)
- Hélène Martini
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905 USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, 55905 USA
| | - João F. Passos
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905 USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, 55905 USA
| |
Collapse
|
27
|
Protasoni M, Serrano M. Targeting Mitochondria to Control Ageing and Senescence. Pharmaceutics 2023; 15:352. [PMID: 36839673 PMCID: PMC9960816 DOI: 10.3390/pharmaceutics15020352] [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: 11/21/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 01/24/2023] Open
Abstract
Ageing is accompanied by a progressive impairment of cellular function and a systemic deterioration of tissues and organs, resulting in increased vulnerability to multiple diseases. Here, we review the interplay between two hallmarks of ageing, namely, mitochondrial dysfunction and cellular senescence. The targeting of specific mitochondrial features in senescent cells has the potential of delaying or even reverting the ageing process. A deeper and more comprehensive understanding of mitochondrial biology in senescent cells is necessary to effectively face this challenge. Here, we discuss the main alterations in mitochondrial functions and structure in both ageing and cellular senescence, highlighting the differences and similarities between the two processes. Moreover, we describe the treatments available to target these pathways and speculate on possible future directions of anti-ageing and anti-senescence therapies targeting mitochondria.
Collapse
Affiliation(s)
- Margherita Protasoni
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Manuel Serrano
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
- Cambridge Institute of Science, Altos Labs, Granta Park, Cambridge CB21 6GP, UK
| |
Collapse
|
28
|
Abstract
Cellular senescence has become a subject of great interest within the ageing research field over the last 60 years, from the first observation in vitro by Leonard Hayflick and Paul Moorhead in 1961, to novel findings of phenotypic sub-types and senescence-like phenotype in post-mitotic cells. It has essential roles in wound healing, tumour suppression and the very first stages of human development, while causing widespread damage and dysfunction with age leading to a raft of age-related diseases. This chapter discusses these roles and their interlinking pathways, and how the observed accumulation of senescent cells with age has initiated a whole new field of ageing research, covering pathologies in the heart, liver, kidneys, muscles, brain and bone. This chapter will also examine how senescent cell accumulation presents in these different tissues, along with their roles in disease development. Finally, there is much focus on developing treatments for senescent cell accumulation in advanced age as a method of alleviating age-related disease. We will discuss here the various senolytic and senostatic treatment approaches and their successes and limitations, and the innovative new strategies being developed to address the differing effects of cellular senescence in ageing and disease.
Collapse
Affiliation(s)
- Rebecca Reed
- Biosciences Institute, Faculty of Medical Sciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Satomi Miwa
- Biosciences Institute, Faculty of Medical Sciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
29
|
Systems Biology of Ageing. Subcell Biochem 2023; 102:415-424. [PMID: 36600142 DOI: 10.1007/978-3-031-21410-3_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The ageing process is highly complex involving multiple processes operating at different biological levels. Systems Biology presents an approach using integrative computational and laboratory study that allows us to address such complexity. The approach relies on the computational analysis of knowledge and data to generate predictive models that may be validated with further laboratory experimentation. Our understanding of ageing is such that translational opportunities are within reach and systems biology offers a means to ensure that optimal decisions are made. We present an overview of the methods employed from bioinformatics and computational modelling and describe some of the insights into ageing that have been gained.
Collapse
|
30
|
Wedel S, Martic I, Guerrero Navarro L, Ploner C, Pierer G, Jansen‐Dürr P, Cavinato M. Depletion of growth differentiation factor 15 (GDF15) leads to mitochondrial dysfunction and premature senescence in human dermal fibroblasts. Aging Cell 2022; 22:e13752. [PMID: 36547021 PMCID: PMC9835581 DOI: 10.1111/acel.13752] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 11/13/2022] [Accepted: 11/19/2022] [Indexed: 12/24/2022] Open
Abstract
Growth differentiation factor 15 (GDF15) is a stress-responsive cytokine also known as a mitokine; however, its role in mitochondrial homeostasis and cellular senescence remained elusive. We show here that knocking down GDF15 expression in human dermal fibroblasts induced mitochondrial dysfunction and premature senescence, associated with a distinct senescence-associated secretory phenotype. Fibroblast-specific loss of GDF15 expression in a model of 3D reconstructed human skin induced epidermal thinning, a hallmark of skin aging. Our results suggest GDF15 to play a so far undisclosed role in mitochondrial homeostasis to delay both the onset of cellular senescence and the appearance of age-related changes in a 3D human skin model.
Collapse
Affiliation(s)
- Sophia Wedel
- Institute for Biochemical Aging ResearchUniversity of InnsbruckInnsbruckAustria,Center for Molecular Biosciences Innsbruck (CMBI)InnsbruckAustria
| | - Ines Martic
- Institute for Biochemical Aging ResearchUniversity of InnsbruckInnsbruckAustria,Center for Molecular Biosciences Innsbruck (CMBI)InnsbruckAustria
| | - Lena Guerrero Navarro
- Institute for Biochemical Aging ResearchUniversity of InnsbruckInnsbruckAustria,Center for Molecular Biosciences Innsbruck (CMBI)InnsbruckAustria
| | - Christian Ploner
- Department of Plastic, Reconstructive and Aesthetic SurgeryMedical University of InnsbruckInnsbruckAustria
| | - Gerhard Pierer
- Department of Plastic, Reconstructive and Aesthetic SurgeryMedical University of InnsbruckInnsbruckAustria
| | - Pidder Jansen‐Dürr
- Institute for Biochemical Aging ResearchUniversity of InnsbruckInnsbruckAustria,Center for Molecular Biosciences Innsbruck (CMBI)InnsbruckAustria
| | - Maria Cavinato
- Institute for Biochemical Aging ResearchUniversity of InnsbruckInnsbruckAustria,Center for Molecular Biosciences Innsbruck (CMBI)InnsbruckAustria
| |
Collapse
|
31
|
Yue R, Dutta A. Computational systems biology in disease modeling and control, review and perspectives. NPJ Syst Biol Appl 2022; 8:37. [PMID: 36192551 PMCID: PMC9528884 DOI: 10.1038/s41540-022-00247-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 09/05/2022] [Indexed: 02/02/2023] Open
Abstract
Omics-based approaches have become increasingly influential in identifying disease mechanisms and drug responses. Considering that diseases and drug responses are co-expressed and regulated in the relevant omics data interactions, the traditional way of grabbing omics data from single isolated layers cannot always obtain valuable inference. Also, drugs have adverse effects that may impair patients, and launching new medicines for diseases is costly. To resolve the above difficulties, systems biology is applied to predict potential molecular interactions by integrating omics data from genomic, proteomic, transcriptional, and metabolic layers. Combined with known drug reactions, the resulting models improve medicines' therapeutical performance by re-purposing the existing drugs and combining drug molecules without off-target effects. Based on the identified computational models, drug administration control laws are designed to balance toxicity and efficacy. This review introduces biomedical applications and analyses of interactions among gene, protein and drug molecules for modeling disease mechanisms and drug responses. The therapeutical performance can be improved by combining the predictive and computational models with drug administration designed by control laws. The challenges are also discussed for its clinical uses in this work.
Collapse
Affiliation(s)
- Rongting Yue
- Department of Electrical and Computer Engineering, University of Connecticut, 371 Fairfield Way, Storrs, CT, 06269, USA.
| | - Abhishek Dutta
- Department of Electrical and Computer Engineering, University of Connecticut, 371 Fairfield Way, Storrs, CT, 06269, USA
| |
Collapse
|
32
|
Guimera AM, Clark P, Wordsworth J, Anugula S, Rasmussen LJ, Shanley DP. Systems modelling predicts chronic inflammation and genomic instability prevent effective mitochondrial regulation during biological ageing. Exp Gerontol 2022; 166:111889. [PMID: 35811018 DOI: 10.1016/j.exger.2022.111889] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/30/2022] [Accepted: 07/04/2022] [Indexed: 11/15/2022]
Abstract
The regulation of mitochondrial turnover under conditions of stress occurs partly through the AMPK-NAD+-PGC1α-SIRT1 signalling pathway. This pathway can be affected by both genomic instability and chronic inflammation since these will result in an increased rate of NAD+ degradation through PARP1 and CD38 respectively. In this work we develop a computational model of this signalling pathway, calibrating and validating it against experimental data. The computational model is used to study mitochondrial turnover under conditions of stress and how it is affected by genomic instability, chronic inflammation and biological ageing in general. We report that the AMPK-NAD+-PGC1α-SIRT1 signalling pathway becomes less responsive with age and that this can prime for the accumulation of dysfunctional mitochondria.
Collapse
Affiliation(s)
- Alvaro Martinez Guimera
- Biosciences Institute, Ageing Research Laboratories, Campus for Ageing and Vitality, Newcastle University, United Kingdom
| | - Peter Clark
- Biosciences Institute, Ageing Research Laboratories, Campus for Ageing and Vitality, Newcastle University, United Kingdom
| | - James Wordsworth
- Biosciences Institute, Ageing Research Laboratories, Campus for Ageing and Vitality, Newcastle University, United Kingdom
| | - Sharath Anugula
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lene Juel Rasmussen
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Daryl P Shanley
- Biosciences Institute, Ageing Research Laboratories, Campus for Ageing and Vitality, Newcastle University, United Kingdom.
| |
Collapse
|
33
|
Evidence of Sex Differences in Cellular Senescence. Neurobiol Aging 2022; 120:88-104. [DOI: 10.1016/j.neurobiolaging.2022.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/30/2022] [Accepted: 08/30/2022] [Indexed: 11/20/2022]
|
34
|
Wang T, Huang S, He C. Senescent cells: A therapeutic target for osteoporosis. Cell Prolif 2022; 55:e13323. [DOI: 10.1111/cpr.13323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/21/2022] [Accepted: 07/25/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Tiantian Wang
- Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, West China Hospital Sichuan University Chengdu Sichuan China
- Institute of Rehabilitation Medicine, West China Hospital Sichuan University Chengdu Sichuan China
| | - Shishu Huang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital and West China School of Medicine Sichuan University Chengdu Sichuan China
| | - Chengqi He
- Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, West China Hospital Sichuan University Chengdu Sichuan China
- Institute of Rehabilitation Medicine, West China Hospital Sichuan University Chengdu Sichuan China
| |
Collapse
|
35
|
Tong Y, Zhang Z, Wang S. Role of Mitochondria in Retinal Pigment Epithelial Aging and Degeneration. FRONTIERS IN AGING 2022; 3:926627. [PMID: 35912040 PMCID: PMC9337215 DOI: 10.3389/fragi.2022.926627] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/21/2022] [Indexed: 12/17/2022]
Abstract
Retinal pigment epithelial (RPE) cells form a monolayer between the neuroretina and choroid. It has multiple important functions, including acting as outer blood-retina barrier, maintaining the function of neuroretina and photoreceptors, participating in the visual cycle and regulating retinal immune response. Due to high oxidative stress environment, RPE cells are vulnerable to dysfunction, cellular senescence, and cell death, which underlies RPE aging and age-related diseases, including age-related macular degeneration (AMD). Mitochondria are the powerhouse of cells and a major source of cellular reactive oxygen species (ROS) that contribute to mitochondrial DNA damage, cell death, senescence, and age-related diseases. Mitochondria also undergo dynamic changes including fission/fusion, biogenesis and mitophagy for quality control in response to stresses. The role of mitochondria, especially mitochondrial dynamics, in RPE aging and age-related diseases, is still unclear. In this review, we summarize the current understanding of mitochondrial function, biogenesis and especially dynamics such as morphological changes and mitophagy in RPE aging and age-related RPE diseases, as well as in the biological processes of RPE cellular senescence and cell death. We also discuss the current preclinical and clinical research efforts to prevent or treat RPE degeneration by restoring mitochondrial function and dynamics.
Collapse
Affiliation(s)
- Yao Tong
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
| | - Zunyi Zhang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
| | - Shusheng Wang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
- Department of Ophthalmology, Tulane University, New Orleans, LA, United States
- Tulane Personalized Health Institute, Tulane University, New Orleans, LA, United States
| |
Collapse
|
36
|
Miwa S, Kashyap S, Chini E, von Zglinicki T. Mitochondrial dysfunction in cell senescence and aging. J Clin Invest 2022; 132:158447. [PMID: 35775483 PMCID: PMC9246372 DOI: 10.1172/jci158447] [Citation(s) in RCA: 300] [Impact Index Per Article: 150.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Mitochondrial dysfunction and cell senescence are hallmarks of aging and are closely interconnected. Mitochondrial dysfunction, operationally defined as a decreased respiratory capacity per mitochondrion together with a decreased mitochondrial membrane potential, typically accompanied by increased production of oxygen free radicals, is a cause and a consequence of cellular senescence and figures prominently in multiple feedback loops that induce and maintain the senescent phenotype. Here, we summarize pathways that cause mitochondrial dysfunction in senescence and aging and discuss the major consequences of mitochondrial dysfunction and how these consequences contribute to senescence and aging. We also highlight the potential of senescence-associated mitochondrial dysfunction as an antiaging and antisenescence intervention target, proposing the combination of multiple interventions converging onto mitochondrial dysfunction as novel, potent senolytics.
Collapse
Affiliation(s)
- Satomi Miwa
- Newcastle University Biosciences Institute, Ageing Biology Laboratories, Newcastle upon Tyne, United Kingdom
| | - Sonu Kashyap
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, Florida, USA.,Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Eduardo Chini
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, Florida, USA.,Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Thomas von Zglinicki
- Newcastle University Biosciences Institute, Ageing Biology Laboratories, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
37
|
Aghali A, Koloko Ngassie ML, Pabelick CM, Prakash YS. Cellular Senescence in Aging Lungs and Diseases. Cells 2022; 11:cells11111781. [PMID: 35681476 PMCID: PMC9179897 DOI: 10.3390/cells11111781] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 05/23/2022] [Accepted: 05/26/2022] [Indexed: 12/10/2022] Open
Abstract
Cellular senescence represents a state of irreversible cell cycle arrest occurring naturally or in response to exogenous stressors. Following the initial arrest, progressive phenotypic changes define conditions of cellular senescence. Understanding molecular mechanisms that drive senescence can help to recognize the importance of such pathways in lung health and disease. There is increasing interest in the role of cellular senescence in conditions such as chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF) in the context of understanding pathophysiology and identification of novel therapies. Herein, we discuss the current knowledge of molecular mechanisms and mitochondrial dysfunction regulating different aspects of cellular senescence-related to chronic lung diseases to develop rational strategies for modulating the senescent cell phenotype in the lung for therapeutic benefit.
Collapse
Affiliation(s)
- Arbi Aghali
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; (A.A.); (C.M.P.)
| | - Maunick Lefin Koloko Ngassie
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands;
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Christina M. Pabelick
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; (A.A.); (C.M.P.)
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Y. S. Prakash
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA; (A.A.); (C.M.P.)
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Correspondence:
| |
Collapse
|
38
|
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
|
39
|
Mitochondrial fission factor promotes cisplatin resistancein hepatocellular carcinoma. Acta Biochim Biophys Sin (Shanghai) 2022; 54:301-310. [PMID: 35538029 PMCID: PMC9828151 DOI: 10.3724/abbs.2022007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver tumor and one of the leading causes of cancer-related death worldwide. Chemotherapeutic agents/regimens such as cisplatin (DDP) are frequently used for advanced HCC treatment. However, drug resistance remains a major hindrance and the underline mechanisms are not fully understood. In this study, we investigated the expression pattern and function of mitochondrial fission factor (Mff) in cisplatin-resistant HCC. We found that Mff is highly expressed in cisplatin-resistant HCC tissues and cell lines. Knockdown of Mff suppresses cell proliferation and promotes cell apoptosis of HCC/DDP cells. In addition, knockdown of Mff sensitizes Huh-7/DDP cells to cisplatin treatment, inhibits cell proliferation, migration and invasion, and enhances cell apoptosis. Confocal imaging showed that knockdown of Mff inhibits the mitochondrial fission and downregulates the expression of GTPase dynamin-related protein 1 (Drp1) in cisplatin-resistant Huh-7/DDP cells. Moreover, xenograft tumor model revealed that knockdown of Mff sensitizes Huh-7/DDP xenograft tumor to cisplatin treatment . In summary, our findings suggest that Mff regulates mitochondrial Drp1 expression and promotes cisplatin resistance in HCC, which provides a potential therapeutic target for the treatment of resistant HCC.
Collapse
|
40
|
Malvezzi H, Dobo C, Filippi RZ, Mendes do Nascimento H, Palmieri da Silva e Sousa L, Meola J, Piccinato CA, Podgaec S. Altered p16 Ink4a, IL-1β, and Lamin b1 Protein Expression Suggest Cellular Senescence in Deep Endometriotic Lesions. Int J Mol Sci 2022; 23:2476. [PMID: 35269619 PMCID: PMC8910415 DOI: 10.3390/ijms23052476] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/26/2022] [Accepted: 01/31/2022] [Indexed: 01/10/2023] Open
Abstract
Endometriosis causes immunological and cellular alterations. Endometriosis lesions have lower levels of lamin b1 than the endometrium. Moreover, high levels of pro-inflammatory markers are observed in the peritoneal fluid, follicular fluid, and serum in endometriosis lesions. Thus, we hypothesized that the accumulation of senescent cells in endometriosis tissues would facilitate endometriosis maintenance in an inflammatory microenvironment. To study senescent cell markers and the senescence-associated secretory phenotype (SASP) in endometriosis lesions, we conducted a cross-sectional study with 27 patients undergoing video laparoscopy for endometriosis resection and 19 patients without endometriosis. Endometriosis lesions were collected from patients with endometriosis, while eutopic endometrium was collected from patients both with and without endometriosis. Tissues were evaluated for senescence markers (p16Ink4a, lamin b1, and IL-1β) and interleukin concentrations. The expression of p16Ink4a increased in lesions compared to that in eutopic endometrium from endometriosis patients in the secretory phase. In the proliferative phase, lesions exhibited lower lamin b1 expression but higher IL-4 expression than the eutopic endometrium. Further, IL-1β levels were higher in the lesions than in the eutopic endometrium in both the secretory and proliferative phases. We believe that our findings may provide targets for better therapeutic interventions to alleviate the symptoms of endometriosis.
Collapse
Affiliation(s)
- Helena Malvezzi
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627, Morumbi 05652-900, SP, Brazil; (C.D.); (R.Z.F.); (H.M.d.N.); (L.P.d.S.e.S.); (C.A.P.); (S.P.)
| | - Cristine Dobo
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627, Morumbi 05652-900, SP, Brazil; (C.D.); (R.Z.F.); (H.M.d.N.); (L.P.d.S.e.S.); (C.A.P.); (S.P.)
| | - Renee Zon Filippi
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627, Morumbi 05652-900, SP, Brazil; (C.D.); (R.Z.F.); (H.M.d.N.); (L.P.d.S.e.S.); (C.A.P.); (S.P.)
| | - Helen Mendes do Nascimento
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627, Morumbi 05652-900, SP, Brazil; (C.D.); (R.Z.F.); (H.M.d.N.); (L.P.d.S.e.S.); (C.A.P.); (S.P.)
| | - Laura Palmieri da Silva e Sousa
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627, Morumbi 05652-900, SP, Brazil; (C.D.); (R.Z.F.); (H.M.d.N.); (L.P.d.S.e.S.); (C.A.P.); (S.P.)
| | - Juliana Meola
- School of Medicine of Ribeirão Preto, University of São Paulo, Gynecology and Obstetrics, Av. Bandeirantes, 3900, Vila Monte Alegre 14049-900, SP, Brazil;
| | - Carla Azevedo Piccinato
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627, Morumbi 05652-900, SP, Brazil; (C.D.); (R.Z.F.); (H.M.d.N.); (L.P.d.S.e.S.); (C.A.P.); (S.P.)
| | - Sérgio Podgaec
- Hospital Israelita Albert Einstein, Av. Albert Einstein 627, Morumbi 05652-900, SP, Brazil; (C.D.); (R.Z.F.); (H.M.d.N.); (L.P.d.S.e.S.); (C.A.P.); (S.P.)
| |
Collapse
|
41
|
Shin J, Nile A, Saini RK, Oh JW. Astaxanthin Sensitizes Low SOD2-Expressing GBM Cell Lines to TRAIL Treatment via Pathway Involving Mitochondrial Membrane Depolarization. Antioxidants (Basel) 2022; 11:antiox11020375. [PMID: 35204257 PMCID: PMC8869337 DOI: 10.3390/antiox11020375] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/09/2022] [Accepted: 02/11/2022] [Indexed: 02/05/2023] Open
Abstract
Carotenoids have been suggested to have either anti- or pro-oxidative effects in several cancer cells, and those effects can trigger an unbalanced reactive oxygen species (ROS) production resulting in an apoptotic response. Our study aimed to evaluate the effect of the well-known carotenoid 3, 3′-dihydroxy-β, β’-carotene-4, 4-dione (astaxanthin, AXT) on glioblastoma multiforme (GBM) cells, especially as a pretreatment of tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), that was previously shown to increase ROS and to induce apoptosis in cancer cells. We found that AXT by itself did not trigger apoptosis in four investigated GBM cell lines upon a 24 h treatment at various concentrations from 2.5 to 50 µM. However, in U251-MG and T98-MG GBM cells, pretreatment of 2.5 to 10 µM AXT sensitized cells to TRAIL treatment in a statistically significant manner (p < 0.05) while it did not affect CRT-MG and U87-MG GBM cells. We further compared AXT-sensitive U251-MG and -insensitive CRT-MG response to AXT and showed that 5 µM AXT treatment had a beneficial effect on both cell lines, as it enhanced mitochondrial potential and TRAIL treatment had the opposite effect, as it decreased mitochondrial potential. Interestingly, in U251-MG, 5 µM AXT pretreatment to TRAIL-treated cells mitochondrial potential further decreased compared to TRAIL alone cells. In addition, while 25 and 50 ng/mL TRAIL treatment increased ROS for both cell lines, pretreatment of 5 µM AXT induced a significant ROS decrease in CRT-MG (p < 0.05) while less effective in U251-MG. We found that in U251-MG, superoxide dismutase (SOD) 2 expression and enzymatic activity were lower compared to CRT-MG and that overexpression of SOD2 in U251-MG abolished AXT sensitization to TRAIL treatment. Taken together, these results suggest that while AXT acts as an ROS scavenger in GBM cell lines, it also has some role in decreasing mitochondrial potential together with TRAIL in a pathway that can be inhibited by SOD2.
Collapse
Affiliation(s)
- Juhyun Shin
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea; (J.S.); (A.N.)
| | - Arti Nile
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea; (J.S.); (A.N.)
| | | | - Jae-Wook Oh
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea; (J.S.); (A.N.)
- Correspondence:
| |
Collapse
|
42
|
Nayeri Rad A, Shams G, Avelar RA, Morowvat MH, Ghasemi Y. Potential senotherapeutic candidates and their combinations derived from transcriptional connectivity and network measures. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2022.100920] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
43
|
Weng Z, Wang Y, Ouchi T, Liu H, Qiao X, Wu C, Zhao Z, Li L, Li B. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:356-371. [PMID: 35485439 PMCID: PMC9052415 DOI: 10.1093/stcltm/szac004] [Citation(s) in RCA: 88] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/19/2021] [Indexed: 11/14/2022] Open
Affiliation(s)
| | | | - Takehito Ouchi
- Department of Physiology, Tokyo Dental College, Tokyo, Japan
| | - Hanghang Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Xianghe Qiao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Chenzhou Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Longjiang Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Bo Li
- Corresponding author: Bo Li, DDS, PhD, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, No.14, 3rd Section of Ren Min Nan Rd. Chengdu, Sichuan 610041, People’s Republic of China.
| |
Collapse
|
44
|
Guo J, Chiang WC. Mitophagy in aging and longevity. IUBMB Life 2021; 74:296-316. [PMID: 34889504 DOI: 10.1002/iub.2585] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 11/21/2021] [Indexed: 12/22/2022]
Abstract
The clearance of damaged or unwanted mitochondria by autophagy (also known as mitophagy) is a mitochondrial quality control mechanism postulated to play an essential role in cellular homeostasis, metabolism, and development and confers protection against a wide range of diseases. Proper removal of damaged or unwanted mitochondria is essential for organismal health. Defects in mitophagy are associated with Parkinson's, Alzheimer's disease, cancer, and other degenerative disorders. Mitochondria regulate organismal fitness and longevity via multiple pathways, including cellular senescence, stem cell function, inflammation, mitochondrial unfolded protein response (mtUPR), and bioenergetics. Thus, mitophagy is postulated to be pivotal for maintaining organismal healthspan and lifespan and the protection against aged-related degeneration. In this review, we will summarize recent understanding of the mechanism of mitophagy and aspects of mitochondrial functions. We will focus on mitochondria-related cellular processes that are linked to aging and examine current genetic evidence that supports the hypothesis that mitophagy is a pro-longevity mechanism.
Collapse
Affiliation(s)
- Jing Guo
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Chung Chiang
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
45
|
Wang L, Lu Z, Zhao J, Schank M, Cao D, Dang X, Nguyen LN, Nguyen LNT, Khanal S, Zhang J, Wu XY, El Gazzar M, Ning S, Moorman J, Yao ZQ. Selective oxidative stress induces dual damage to telomeres and mitochondria in human T cells. Aging Cell 2021; 20:e13513. [PMID: 34752684 PMCID: PMC8672791 DOI: 10.1111/acel.13513] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 09/27/2021] [Accepted: 10/31/2021] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress caused by excess reactive oxygen species (ROS) accelerates telomere erosion and mitochondrial injury, leading to impaired cellular functions and cell death. Whether oxidative stress‐mediated telomere erosion induces mitochondrial injury, or vice versa, in human T cells—the major effectors of host adaptive immunity against infection and malignancy—is poorly understood due to the pleiotropic effects of ROS. Here we employed a novel chemoptogenetic tool that selectively produces a single oxygen (1O2) only at telomeres or mitochondria in Jurkat T cells. We found that targeted 1O2 production at telomeres triggered not only telomeric DNA damage but also mitochondrial dysfunction, resulting in T cell apoptotic death. Conversely, targeted 1O2 formation at mitochondria induced not only mitochondrial injury but also telomeric DNA damage, leading to cellular crisis and apoptosis. Targeted oxidative stress at either telomeres or mitochondria increased ROS production, whereas blocking ROS formation during oxidative stress reversed the telomeric injury, mitochondrial dysfunction, and cellular apoptosis. Notably, the X‐ray repair cross‐complementing protein 1 (XRCC1) in the base excision repair (BER) pathway and multiple mitochondrial proteins in other cellular pathways were dysregulated by the targeted oxidative stress. By confining singlet 1O2 formation to a single organelle, this study suggests that oxidative stress induces dual injury in T cells via crosstalk between telomeres and mitochondria. Further identification of these oxidation pathways may offer a novel approach to preserve mitochondrial functions, protect telomere integrity, and maintain T cell survival, which can be exploited to combat various immune aging‐associated diseases.
Collapse
Affiliation(s)
- Ling Wang
- Center of Excellence in Inflammation, Infectious Disease and Immunity Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
- Division of Infectious, Inflammatory and Immunologic Diseases Department of Internal Medicine Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
| | - Zeyuan Lu
- Center of Excellence in Inflammation, Infectious Disease and Immunity Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
- Division of Infectious, Inflammatory and Immunologic Diseases Department of Internal Medicine Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
| | - Juan Zhao
- Center of Excellence in Inflammation, Infectious Disease and Immunity Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
- Division of Infectious, Inflammatory and Immunologic Diseases Department of Internal Medicine Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
| | - Madison Schank
- Center of Excellence in Inflammation, Infectious Disease and Immunity Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
- Division of Infectious, Inflammatory and Immunologic Diseases Department of Internal Medicine Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
| | - Dechao Cao
- Center of Excellence in Inflammation, Infectious Disease and Immunity Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
- Division of Infectious, Inflammatory and Immunologic Diseases Department of Internal Medicine Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
| | - Xindi Dang
- Center of Excellence in Inflammation, Infectious Disease and Immunity Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
- Division of Infectious, Inflammatory and Immunologic Diseases Department of Internal Medicine Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
| | - Lam Nhat Nguyen
- Center of Excellence in Inflammation, Infectious Disease and Immunity Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
- Division of Infectious, Inflammatory and Immunologic Diseases Department of Internal Medicine Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
| | - Lam Ngoc Thao Nguyen
- Center of Excellence in Inflammation, Infectious Disease and Immunity Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
- Division of Infectious, Inflammatory and Immunologic Diseases Department of Internal Medicine Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
| | - Sushant Khanal
- Center of Excellence in Inflammation, Infectious Disease and Immunity Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
- Division of Infectious, Inflammatory and Immunologic Diseases Department of Internal Medicine Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
| | - Jinyu Zhang
- Center of Excellence in Inflammation, Infectious Disease and Immunity Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
- Division of Infectious, Inflammatory and Immunologic Diseases Department of Internal Medicine Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
| | - Xiao Y. Wu
- Center of Excellence in Inflammation, Infectious Disease and Immunity Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
- Division of Infectious, Inflammatory and Immunologic Diseases Department of Internal Medicine Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
| | - Mohamed El Gazzar
- Center of Excellence in Inflammation, Infectious Disease and Immunity Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
- Division of Infectious, Inflammatory and Immunologic Diseases Department of Internal Medicine Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
| | - Shunbin Ning
- Center of Excellence in Inflammation, Infectious Disease and Immunity Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
- Division of Infectious, Inflammatory and Immunologic Diseases Department of Internal Medicine Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
| | - Jonathan P. Moorman
- Center of Excellence in Inflammation, Infectious Disease and Immunity Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
- Division of Infectious, Inflammatory and Immunologic Diseases Department of Internal Medicine Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
- Hepatitis (HCV/HBV/HIV) Program Department of Veterans Affairs James H. Quillen VA Medical Center Johnson City Tennessee USA
| | - Zhi Q. Yao
- Center of Excellence in Inflammation, Infectious Disease and Immunity Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
- Division of Infectious, Inflammatory and Immunologic Diseases Department of Internal Medicine Quillen College of Medicine East Tennessee State University Johnson City Tennessee USA
- Hepatitis (HCV/HBV/HIV) Program Department of Veterans Affairs James H. Quillen VA Medical Center Johnson City Tennessee USA
| |
Collapse
|
46
|
Gondal MN, Chaudhary SU. Navigating Multi-Scale Cancer Systems Biology Towards Model-Driven Clinical Oncology and Its Applications in Personalized Therapeutics. Front Oncol 2021; 11:712505. [PMID: 34900668 PMCID: PMC8652070 DOI: 10.3389/fonc.2021.712505] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 10/26/2021] [Indexed: 12/19/2022] Open
Abstract
Rapid advancements in high-throughput omics technologies and experimental protocols have led to the generation of vast amounts of scale-specific biomolecular data on cancer that now populates several online databases and resources. Cancer systems biology models built using this data have the potential to provide specific insights into complex multifactorial aberrations underpinning tumor initiation, development, and metastasis. Furthermore, the annotation of these single- and multi-scale models with patient data can additionally assist in designing personalized therapeutic interventions as well as aid in clinical decision-making. Here, we have systematically reviewed the emergence and evolution of (i) repositories with scale-specific and multi-scale biomolecular cancer data, (ii) systems biology models developed using this data, (iii) associated simulation software for the development of personalized cancer therapeutics, and (iv) translational attempts to pipeline multi-scale panomics data for data-driven in silico clinical oncology. The review concludes that the absence of a generic, zero-code, panomics-based multi-scale modeling pipeline and associated software framework, impedes the development and seamless deployment of personalized in silico multi-scale models in clinical settings.
Collapse
Affiliation(s)
- Mahnoor Naseer Gondal
- Biomedical Informatics Research Laboratory, Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, United States
| | - Safee Ullah Chaudhary
- Biomedical Informatics Research Laboratory, Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| |
Collapse
|
47
|
Csekes E, Račková L. Skin Aging, Cellular Senescence and Natural Polyphenols. Int J Mol Sci 2021; 22:12641. [PMID: 34884444 PMCID: PMC8657738 DOI: 10.3390/ijms222312641] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/13/2021] [Accepted: 11/18/2021] [Indexed: 01/10/2023] Open
Abstract
The skin, being the barrier organ of the body, is constitutively exposed to various stimuli impacting its morphology and function. Senescent cells have been found to accumulate with age and may contribute to age-related skin changes and pathologies. Natural polyphenols exert many health benefits, including ameliorative effects on skin aging. By affecting molecular pathways of senescence, polyphenols are able to prevent or delay the senescence formation and, consequently, avoid or ameliorate aging and age-associated pathologies of the skin. This review aims to provide an overview of the current state of knowledge in skin aging and cellular senescence, and to summarize the recent in vitro studies related to the anti-senescent mechanisms of natural polyphenols carried out on keratinocytes, melanocytes and fibroblasts. Aged skin in the context of the COVID-19 pandemic will be also discussed.
Collapse
Affiliation(s)
- Erika Csekes
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Dúbravská Cesta 9, 841 04 Bratislava, Slovakia
| | - Lucia Račková
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Dúbravská Cesta 9, 841 04 Bratislava, Slovakia
| |
Collapse
|
48
|
Udomsinprasert W, Sobhonslidsuk A, Jittikoon J, Honsawek S, Chaikledkaew U. Cellular senescence in liver fibrosis: Implications for age-related chronic liver diseases. Expert Opin Ther Targets 2021; 25:799-813. [PMID: 34632912 DOI: 10.1080/14728222.2021.1992385] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION New insights indicate a causative link between cellular senescence and liver fibrosis. Senescent hepatic stellate cells (HSCs) facilitate fibrosis resolution, while senescence in hepatocytes and cholangiocytes acts as a potent mechanism driving liver fibrogenesis. In many clinical studies, telomeres and mitochondrial DNA contents, which are both aging biomarkers, were reportedly associated with a degree of liver fibrosis in patients with chronic liver diseases (CLDs); this highlights their potential as biomarkers for liver fibrogenesis. A deeper understanding of mechanisms underlying multi-step progression of senescence may yield new therapeutic strategies for age-related chronic liver pathologies. AREAS COVERED This review examines the recent findings from preclinical and clinical studies on mechanisms of senescence in liver fibrogenesis and its involvement in liver fibrosis. A comprehensive literature search in electronic databases consisting of PubMed and Scopus from inception to 31 August 2021 was performed. EXPERT OPINION Cellular senescence has diagnostic, prognostic, and therapeutic potential in progressive liver complications, especially liver fibrosis. Stimulating or reinforcing the immune response against senescent cells may be a promising and forthright biotherapeutic strategy. This approach will need a deeper understanding of the immune system's ability to eliminate senescent cells and the molecular and cellular mechanisms underlying this process.
Collapse
Affiliation(s)
| | - Abhasnee Sobhonslidsuk
- Division of Gastroenterology and Hepatology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Jiraphun Jittikoon
- Department of Biochemistry, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Sittisak Honsawek
- Department of Biochemistry, Osteoarthritis and Musculoskeleton Research Unit, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Usa Chaikledkaew
- Social and Administrative Pharmacy Division, Department of Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand.,Mahidol University Health Technology Assessment (MUHTA) Graduate Program, Mahidol University, Bangkok, Thailand
| |
Collapse
|
49
|
Narasimhan A, Flores RR, Robbins PD, Niedernhofer LJ. Role of Cellular Senescence in Type II Diabetes. Endocrinology 2021; 162:6345039. [PMID: 34363464 PMCID: PMC8386762 DOI: 10.1210/endocr/bqab136] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Indexed: 01/10/2023]
Abstract
Cellular senescence is a cell fate that occurs in response to numerous types of stress and can promote tissue repair or drive inflammation and disruption of tissue homeostasis depending on the context. Aging and obesity lead to an increase in the senescent cell burden in multiple organs. Senescent cells release a myriad of senescence-associated secretory phenotype factors that directly mediate pancreatic β-cell dysfunction, adipose tissue dysfunction, and insulin resistance in peripheral tissues, which promote the onset of type II diabetes mellitus. In addition, hyperglycemia and metabolic changes seen in diabetes promote cellular senescence. Diabetes-induced cellular senescence contributes to various diabetic complications. Thus, type II diabetes is both a cause and consequence of cellular senescence. This review summarizes recent studies on the link between aging, obesity, and diabetes, focusing on the role of cellular senescence in disease processes.
Collapse
Affiliation(s)
- Akilavalli Narasimhan
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, 55455, USA
| | - Rafael R Flores
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, 55455, USA
| | - Paul D Robbins
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, 55455, USA
| | - Laura J Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, 55455, USA
- Correspondence: Laura J. Niedernhofer, MD, PhD, Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Medical School, 6-155 Jackson Hall, 321 Church Street, SE, Minneapolis, MN 55455, USA.
| |
Collapse
|
50
|
Teulière J, Bernard C, Bapteste E. Interspecific interactions that affect ageing: Age-distorters manipulate host ageing to their own evolutionary benefits. Ageing Res Rev 2021; 70:101375. [PMID: 34082078 DOI: 10.1016/j.arr.2021.101375] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/22/2021] [Accepted: 05/26/2021] [Indexed: 02/07/2023]
Abstract
Genetic causes for ageing are traditionally investigated within a species. Yet, the lifecycles of many organisms intersect. Additional evolutionary and genetic causes of ageing, external to a focal species/organism, may thus be overlooked. Here, we introduce the phrase and concept of age-distorters and its evidence. Age-distorters carry ageing interfering genes, used to manipulate the biological age of other entities upon which the reproduction of age-distorters relies, e.g. age-distorters bias the reproduction/maintenance trade-offs of cells/organisms for their own evolutionary interests. Candidate age-distorters include viruses, parasites and symbionts, operating through specific, genetically encoded interferences resulting from co-evolution and arms race between manipulative non-kins and manipulable species. This interference results in organismal ageing when age-distorters prompt manipulated organisms to favor their reproduction at the expense of their maintenance, turning these hosts into expanded disposable soma. By relying on reproduction/maintenance trade-offs affecting disposable entities, which are left ageing to the reproductive benefit of other physically connected lineages with conflicting evolutionary interests, the concept of age-distorters expands the logic of the Disposable Soma theory beyond species with fixed germen/soma distinctions. Moreover, acknowledging age-distorters as external sources of mutation accumulation and antagonistic pleiotropic genes expands the scope of the mutation accumulation and of the antagonistic pleiotropy theories.
Collapse
Affiliation(s)
- Jérôme Teulière
- Institut de Systématique, Evolution, Biodiversité (ISYEB), Sorbonne Université, CNRS, Museum National d'Histoire Naturelle, EPHE, Université des Antilles, Paris, France
| | - Charles Bernard
- Institut de Systématique, Evolution, Biodiversité (ISYEB), Sorbonne Université, CNRS, Museum National d'Histoire Naturelle, EPHE, Université des Antilles, Paris, France
| | - Eric Bapteste
- Institut de Systématique, Evolution, Biodiversité (ISYEB), Sorbonne Université, CNRS, Museum National d'Histoire Naturelle, EPHE, Université des Antilles, Paris, France.
| |
Collapse
|