151
|
Jain SS, Barefoot ME, Varghese RS, Ressom HW. Cell-type Deconvolution and Age Estimation Using DNA Methylation Reveals NK Cell Deficiency in the Hepatocellular Carcinoma Microenvironment. PROCEEDINGS. IEEE INTERNATIONAL CONFERENCE ON BIOINFORMATICS AND BIOMEDICINE 2022; 2022:444-449. [PMID: 37663782 PMCID: PMC10473873 DOI: 10.1109/bibm55620.2022.9995041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Hepatocellular carcinoma (HCC) has been an approved indication for the administration of immunotherapy since 2017, but biomarkers that predict therapeutic response have remained limited. Understanding and characterizing the tumor immune microenvironment enables better classification of these tumors and may reveal biomarkers that predict immunotherapeutic efficacy. In this paper, we applied a cell-type deconvolution algorithm using DNA methylation array data to investigate the composition of the tumor microenvironment in HCC. Using two publicly available datasets with a total cohort size of 57 patients, each with tumor and matched normal tissue samples, we identified key differences in immune cell composition. We found that NK cell abundance was significantly decreased in HCC tumors compared to adjacent normal tissue. We also applied DNA methylation "clocks" which estimate phenotypic aging and compared these findings to expression-based determinations of cellular senescence. Senescence and epigenetic aging was significantly increased in HCC tumors, and the degree of age acceleration and senescence was strongly associated with decreased NK cell abundance. In summary, we found that NK cell infiltration in the tumor microenvironment is significantly diminished, and that this loss of NK abundance is strongly associated with increased senescence and age-related phenotype. These findings point to key interactions between NK cells and the senescent tumor microenvironment and offer insights into the pathogenesis of HCC as well as potential biomarkers of therapeutic efficacy.
Collapse
Affiliation(s)
- Sidharth S Jain
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Megan E Barefoot
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Rency S Varghese
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Habtom W Ressom
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
152
|
Saul D, Khosla S. Fracture Healing in the Setting of Endocrine Diseases, Aging, and Cellular Senescence. Endocr Rev 2022; 43:984-1002. [PMID: 35182420 PMCID: PMC9695115 DOI: 10.1210/endrev/bnac008] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Indexed: 11/19/2022]
Abstract
More than 2.1 million age-related fractures occur in the United States annually, resulting in an immense socioeconomic burden. Importantly, the age-related deterioration of bone structure is associated with impaired bone healing. Fracture healing is a dynamic process which can be divided into four stages. While the initial hematoma generates an inflammatory environment in which mesenchymal stem cells and macrophages orchestrate the framework for repair, angiogenesis and cartilage formation mark the second healing period. In the central region, endochondral ossification favors soft callus development while next to the fractured bony ends, intramembranous ossification directly forms woven bone. The third stage is characterized by removal and calcification of the endochondral cartilage. Finally, the chronic remodeling phase concludes the healing process. Impaired fracture healing due to aging is related to detrimental changes at the cellular level. Macrophages, osteocytes, and chondrocytes express markers of senescence, leading to reduced self-renewal and proliferative capacity. A prolonged phase of "inflammaging" results in an extended remodeling phase, characterized by a senescent microenvironment and deteriorating healing capacity. Although there is evidence that in the setting of injury, at least in some tissues, senescent cells may play a beneficial role in facilitating tissue repair, recent data demonstrate that clearing senescent cells enhances fracture repair. In this review, we summarize the physiological as well as pathological processes during fracture healing in endocrine disease and aging in order to establish a broad understanding of the biomechanical as well as molecular mechanisms involved in bone repair.
Collapse
Affiliation(s)
- Dominik Saul
- Kogod Center on Aging and Division of Endocrinology, Mayo Clinic, Rochester, Minnesota 55905, USA.,Department of Trauma, Orthopedics and Reconstructive Surgery, Georg-August-University of Goettingen, 37073 Goettingen, Germany
| | - Sundeep Khosla
- Kogod Center on Aging and Division of Endocrinology, Mayo Clinic, Rochester, Minnesota 55905, USA
| |
Collapse
|
153
|
Cogno N, Bauer R, Durante M. An Agent-Based Model of Radiation-Induced Lung Fibrosis. Int J Mol Sci 2022; 23:13920. [PMID: 36430398 PMCID: PMC9693125 DOI: 10.3390/ijms232213920] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/03/2022] [Accepted: 11/05/2022] [Indexed: 11/16/2022] Open
Abstract
Early- and late-phase radiation-induced lung injuries, namely pneumonitis and lung fibrosis (RILF), severely constrain the maximum dose and irradiated volume in thoracic radiotherapy. As the most radiosensitive targets, epithelial cells respond to radiation either by undergoing apoptosis or switching to a senescent phenotype that triggers the immune system and damages surrounding healthy cells. Unresolved inflammation stimulates mesenchymal cells' proliferation and extracellular matrix (ECM) secretion, which irreversibly stiffens the alveolar walls and leads to respiratory failure. Although a thorough understanding is lacking, RILF and idiopathic pulmonary fibrosis share multiple pathways and would mutually benefit from further insights into disease progression. Furthermore, current normal tissue complication probability (NTCP) models rely on clinical experience to set tolerance doses for organs at risk and leave aside mechanistic interpretations of the undergoing processes. To these aims, we implemented a 3D agent-based model (ABM) of an alveolar duct that simulates cell dynamics and substance diffusion following radiation injury. Emphasis was placed on cell repopulation, senescent clearance, and intra/inter-alveolar bystander senescence while tracking ECM deposition. Our ABM successfully replicates early and late fibrotic response patterns reported in the literature along with the ECM sigmoidal dose-response curve. Moreover, surrogate measures of RILF severity via a custom indicator show qualitative agreement with published fibrosis indices. Finally, our ABM provides a fully mechanistic alveolar survival curve highlighting the need to include bystander damage in lung NTCP models.
Collapse
Affiliation(s)
- Nicolò Cogno
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, 64291 Darmstadt, Germany
- Institute for Condensed Matter Physics, Technische Universität Darmstadt, 64289 Darmstadt, Germany
| | - Roman Bauer
- Department of Computer Science, University of Surrey, Guildford GU2 7XH, UK
| | - Marco Durante
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, 64291 Darmstadt, Germany
- Institute for Condensed Matter Physics, Technische Universität Darmstadt, 64289 Darmstadt, Germany
| |
Collapse
|
154
|
Zhou S, Zhu J, Zhou PK, Gu Y. Alveolar type 2 epithelial cell senescence and radiation-induced pulmonary fibrosis. Front Cell Dev Biol 2022; 10:999600. [PMID: 36407111 PMCID: PMC9666897 DOI: 10.3389/fcell.2022.999600] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/24/2022] [Indexed: 11/24/2022] Open
Abstract
Radiation-induced pulmonary fibrosis (RIPF) is a chronic and progressive respiratory tract disease characterized by collagen deposition. The pathogenesis of RIPF is still unclear. Type 2 alveolar epithelial cells (AT2), the essential cells that maintain the structure and function of lung tissue, are crucial for developing pulmonary fibrosis. Recent studies indicate the critical role of AT2 cell senescence during the onset and progression of RIPF. In addition, clearance of senescent AT2 cells and treatment with senolytic drugs efficiently improve lung function and radiation-induced pulmonary fibrosis symptoms. These findings indicate that AT2 cell senescence has the potential to contribute significantly to the innovative treatment of fibrotic lung disorders. This review summarizes the current knowledge from basic and clinical research about the mechanism and functions of AT2 cell senescence in RIPF and points to the prospects for clinical treatment by targeting senescent AT2 cells.
Collapse
Affiliation(s)
- Shenghui Zhou
- Hengyang Medical College, University of South China, Hengyang, China,Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, AMMS, Beijing, China
| | - Jiaojiao Zhu
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, AMMS, Beijing, China
| | - Ping-Kun Zhou
- Hengyang Medical College, University of South China, Hengyang, China,Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, AMMS, Beijing, China,*Correspondence: Yongqing Gu, ; Ping-Kun Zhou,
| | - Yongqing Gu
- Hengyang Medical College, University of South China, Hengyang, China,Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, AMMS, Beijing, China,*Correspondence: Yongqing Gu, ; Ping-Kun Zhou,
| |
Collapse
|
155
|
Chibaya L, Snyder J, Ruscetti M. Senescence and the tumor-immune landscape: Implications for cancer immunotherapy. Semin Cancer Biol 2022; 86:827-845. [PMID: 35143990 PMCID: PMC9357237 DOI: 10.1016/j.semcancer.2022.02.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/03/2022] [Indexed: 01/27/2023]
Abstract
Cancer therapies, including conventional chemotherapy, radiation, and molecularly targeted agents, can lead to tumor eradication through a variety of mechanisms. In addition to their effects on tumor cell growth and survival, these regimens can also influence the surrounding tumor-immune microenvironment in ways that ultimately impact therapy responses. A unique biological outcome of cancer therapy is induction of cellular senescence. Senescence is a damage-induced stress program that leads to both the durable arrest of tumor cells and remodeling the tumor-immune microenvironment through activation of a collection pleiotropic cytokines, chemokines, growth factors, and proteinases known as the senescence-associated secretory phenotype (SASP). Depending on the cancer context and the mechanism of action of the therapy, the SASP produced following therapy-induced senescence (TIS) can promote anti-tumor immunity that enhances therapeutic efficacy, or alternatively chronic inflammation that leads to therapy failure and tumor relapse. Thus, a deeper understanding of the mechanisms regulating the SASP and components necessary for robust anti-tumor immune surveillance in different cancer and therapy contexts are key to harnessing senescence for tumor control. Here we draw a roadmap to modulate TIS and its immune-stimulating features for cancer immunotherapy.
Collapse
Affiliation(s)
- Loretah Chibaya
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jarin Snyder
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Marcus Ruscetti
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA; Immunology and Microbiology Program, University of Massachusetts Chan Medical School, Worcester, MA, USA; Cancer Center, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
156
|
Abstract
PURPOSE OF REVIEW Systemic sclerosis (SSc) is a chronic rheumatic disease that is characterized by immune activation, vasculopathy and fibrosis of the skin and internal organs. It has been proposed that premature onset of ageing pathways and associated senescent changes in cells contribute to the clinical and pathological features of SSc. The aim of this review is to critically review recent insights into the involvement of cellular senescence in SSc. RECENT FINDINGS Cellular senescence plays a critical role in SSc pathogenesis, particularly involving endothelial cells and fibroblasts. Immunosenescence could also contribute to SSc pathogenesis by direct alteration of cellular functions or indirect promotion of defective immune surveillance. Molecular studies have shed some light on how cellular senescence contributes to fibrosis. Recent and planned proof-of-concept trials using senotherapeutics showed promising results in fibrotic diseases, including SSc. SUMMARY There is increasing evidence implicating cellular senescence in SSc. The mechanisms underlying premature cellular senescence in SSc, and its potential role in pathogenesis, merit further investigation. Emerging drugs targeting senescence-related pathways might be potential therapeutic options for SSc.
Collapse
Affiliation(s)
- Pei-Suen Tsou
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Scleroderma Program, University of Michigan, Ann Arbor, MI, USA
| | - Bo Shi
- Department of Dermatology, Northwestern University, Chicago, IL, USA
| | - John Varga
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Scleroderma Program, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
157
|
Onorati A, Havas AP, Lin B, Rajagopal J, Sen P, Adams PD, Dou Z. Upregulation of PD-L1 in Senescence and Aging. Mol Cell Biol 2022; 42:e0017122. [PMID: 36154662 PMCID: PMC9583718 DOI: 10.1128/mcb.00171-22] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/13/2022] [Accepted: 09/12/2022] [Indexed: 12/25/2022] Open
Abstract
Cellular senescence is a stable form of cell cycle arrest associated with proinflammatory responses. Senescent cells can be cleared by the immune system as a part of normal tissue homeostasis. However, senescent cells can also accumulate in aged and diseased tissues, contributing to inflammation and disease progression. The mechanisms mediating the impaired immune-mediated clearance of senescent cells are poorly understood. Here, we report that senescent cells upregulate the immune checkpoint molecule PD-L1, the ligand for PD-1 on immune cells, which drives immune cell inactivation. The induction of PD-L1 in senescence is dependent on the proinflammatory program. Furthermore, the secreted factors released by senescent cells are sufficient to upregulate PD-L1 in nonsenescent control cells, mediated by the JAK-STAT pathway. In addition, we show that prolongevity intervention rapamycin downregulates PD-L1 in senescent cells. Last, we found that PD-L1 is upregulated in several tissues in naturally aged mice and in the lungs of idiopathic pulmonary fibrosis patients. Together, our results report that senescence and aging are associated with upregulation of a major immune checkpoint molecule, PD-L1. Targeting PD-L1 may offer new therapeutic opportunities in treating senescence and age-associated diseases.
Collapse
Affiliation(s)
- Angelique Onorati
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Aaron P. Havas
- Aging, Cancer and Immuno-oncology Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Brian Lin
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Developmental, Molecular & Chemical Biology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Jayaraj Rajagopal
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Klarman Cell Observatory, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, USA
| | - Payel Sen
- National Institute on Aging, National Institutes of Health, Laboratory of Genetics and Genomics, Baltimore, Maryland, USA
| | - Peter D. Adams
- Aging, Cancer and Immuno-oncology Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Zhixun Dou
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
158
|
Jin R, Chan AKY, Wu J, Lee TMC. Relationships between Inflammation and Age-Related Neurocognitive Changes. Int J Mol Sci 2022; 23:12573. [PMID: 36293430 PMCID: PMC9604276 DOI: 10.3390/ijms232012573] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 11/18/2022] Open
Abstract
The relationship between inflammation and age-related neurocognitive changes is significant, which may relate to the age-related immune dysfunctions characterized by the senescence of immune cells and elevated inflammatory markers in the peripheral circulation and the central nervous system. In this review, we discuss the potential mechanisms, including the development of vascular inflammation, neuroinflammation, organelle dysfunctions, abnormal cholesterol metabolism, and glymphatic dysfunctions as well as the role that the key molecules play in the immune-cognition interplay. We propose potential therapeutic pharmacological and behavioral strategies for ameliorating age-related neurocognitive changes associated with inflammation. Further research to decipher the multidimensional roles of chronic inflammation in normal and pathological aging processes will help unfold the pathophysiological mechanisms underpinning neurocognitive disorders. The insight gained will lay the path for developing cost-effective preventative measures and the buffering or delaying of age-related neurocognitive decline.
Collapse
Affiliation(s)
- Run Jin
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong 999077, China
- Laboratory of Neuropsychology and Human Neuroscience, The University of Hong Kong, Hong Kong 999077, China
| | - Aidan Kai Yeung Chan
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong 999077, China
- Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Jingsong Wu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350108, China
| | - Tatia Mei Chun Lee
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong 999077, China
- Laboratory of Neuropsychology and Human Neuroscience, The University of Hong Kong, Hong Kong 999077, China
| |
Collapse
|
159
|
Cellular senescence in ischemia/reperfusion injury. Cell Death Dis 2022; 8:420. [PMID: 36253355 PMCID: PMC9576687 DOI: 10.1038/s41420-022-01205-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/22/2022] [Accepted: 09/27/2022] [Indexed: 11/24/2022]
Abstract
Ischemia/reperfusion (IR) injury, a main reason of mortality and morbidity worldwide, occurs in many organs and tissues. As a result of IR injury, senescent cells can accumulate in multiple organs. Increasing evidence shows that cellular senescence is the underlying mechanism that transforms an acute organ injury into a chronic one. Several recent studies suggest senescent cells can be targeted for the prevention or elimination of acute and chronic organ injury induced by IR. In this review, we concisely introduce the underlying mechanism and the pivotal role of premature senescence in the transition from acute to chronic IR injuries. Special focus is laid on recent advances in the mechanisms as well as on the basic and clinical research, targeting cellular senescence in multi-organ IR injuries. Besides, the potential directions in this field are discussed in the end. Together, the recent advances reviewed here will act as a comprehensive overview of the roles of cellular senescence in IR injury, which could be of great significance for the design of related studies, or as a guide for potential therapeutic target.
Collapse
|
160
|
Marrella V, Facoetti A, Cassani B. Cellular Senescence in Immunity against Infections. Int J Mol Sci 2022; 23:11845. [PMID: 36233146 PMCID: PMC9570409 DOI: 10.3390/ijms231911845] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022] Open
Abstract
Cellular senescence is characterized by irreversible cell cycle arrest in response to different triggers and an inflammatory secretome. Although originally described in fibroblasts and cell types of solid organs, cellular senescence affects most tissues with advancing age, including the lymphoid tissue, causing chronic inflammation and dysregulation of both innate and adaptive immune functions. Besides its normal occurrence, persistent microbial challenge or pathogenic microorganisms might also accelerate the activation of cellular aging, inducing the premature senescence of immune cells. Therapeutic strategies counteracting the detrimental effects of cellular senescence are being developed. Their application to target immune cells might have the potential to improve immune dysfunctions during aging and reduce the age-dependent susceptibility to infections. In this review, we discuss how immune senescence influences the host's ability to resolve more common infections in the elderly and detail the different markers proposed to identify such senescent cells; the mechanisms by which infectious agents increase the extent of immune senescence are also reviewed. Finally, available senescence therapeutics are discussed in the context of their effects on immunity and against infections.
Collapse
Affiliation(s)
- Veronica Marrella
- UOS Milan Unit, Istituto di Ricerca Genetica e Biomedica (IRGB), CNR, 20138 Milan, Italy
- IRCCS Humanitas Research Hospital, 20089 Milan, Italy
| | - Amanda Facoetti
- Department of Biomedical Sciences, Humanitas University, 20090 Milan, Italy
| | - Barbara Cassani
- IRCCS Humanitas Research Hospital, 20089 Milan, Italy
- Department of Medical Biotechnologies and Translational Medicine, Università Degli Studi di Milano, 20089 Milan, Italy
| |
Collapse
|
161
|
Wyles SP, Tchkonia T, Kirkland JL. Targeting Cellular Senescence for Age-Related Diseases: Path to Clinical Translation. Plast Reconstr Surg 2022; 150:20S-26S. [PMID: 36170432 PMCID: PMC9529239 DOI: 10.1097/prs.0000000000009669] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
SUMMARY Beyond the palliative reach of today's medicines, medical therapies of tomorrow aim to treat the root cause of age-related diseases by targeting fundamental aging mechanisms. Pillars of aging include, among others, genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, dysregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication. The unitary theory of fundamental aging processes posits that by targeting one fundamental aging process, it may be feasible to impact several or all others given its interdependence. Indeed, pathologic accumulation of senescent cells is implicated in chronic diseases and age-associated morbidities, suggesting that senescent cells are a good target for whole-body aging intervention. Preclinical studies using senolytics, agents that selectively eliminate senescent cells, and senomorphics, agents that inhibit production or release of senescence-associated secretory phenotype factors, show promise in several aging and disease preclinical models. Early clinical trials using a senolytic combination (dasatinib and quercetin), and other senolytics including flavonoid, fisetin, and BCL-xL inhibitors, illustrate the potential of senolytics to alleviate age-related dysfunction and diseases including wound healing. Translation into clinical applications requires parallel clinical trials across institutions to validate senotherapeutics as a vanguard for delaying, preventing, or treating age-related disorders and aesthetic aging.
Collapse
Affiliation(s)
- Saranya P. Wyles
- Department of Dermatology, Mayo Clinic, Rochester, MN
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN
| | - Tamara Tchkonia
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN
| | - James L. Kirkland
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN
- Division of Geriatrics and Gerontology, Department of Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|
162
|
Maheshwari A. The Phylogeny, Ontogeny, and Organ-specific Differentiation of Macrophages in the Developing Intestine. NEWBORN (CLARKSVILLE, MD.) 2022; 1:340-355. [PMID: 36698382 PMCID: PMC9872774 DOI: 10.5005/jp-journals-11002-0044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Macrophages are large highly motile phagocytic leukocytes that appear early during embryonic development and have been conserved during evolution. The developmental roles of macrophages were first described nearly a century ago, at about the time these cells were being identified as central effectors in phagocytosis and elimination of microbes. Since then, we have made considerable progress in understanding the development of various subsets of macrophages and the diverse roles these cells play in both physiology and disease. This article reviews the phylogeny and the ontogeny of macrophages with a particular focus on the gastrointestinal tract, and the role of these mucosal macrophages in immune surveillance, innate immunity, homeostasis, tissue remodeling, angiogenesis, and repair of damaged tissues. We also discuss the importance of these macrophages in the inflammatory changes in neonatal necrotizing enterocolitis (NEC). This article presents a combination of our own peer-reviewed clinical and preclinical studies, with an extensive review of the literature using the databases PubMed, EMBASE, and Scopus.
Collapse
Affiliation(s)
- Akhil Maheshwari
- Global Newborn Society, Clarksville, Maryland, United States of America
| |
Collapse
|
163
|
Asaithamby A, Shay JW, Minna JD. Cellular senescence and lung cancer prognosis. Transl Lung Cancer Res 2022; 11:1982-1987. [PMID: 36386455 PMCID: PMC9641047 DOI: 10.21037/tlcr-22-678] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/17/2022] [Indexed: 01/25/2023]
Affiliation(s)
- Aroumougame Asaithamby
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA;,Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jerry W. Shay
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA;,Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - John D. Minna
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA;,Hamon Center for Therapeutic Oncology Research and the Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
164
|
Tang X, Deng B, Zang A, He X, Zhou Y, Wang D, Li D, Dai X, Chen J, Zhang X, Liu Y, Xu Y, Chen J, Zheng W, Zhang L, Gao C, Yang H, Li B, Wang X. Characterization of age-related immune features after autologous NK cell infusion: Protocol for an open-label and randomized controlled trial. Front Immunol 2022; 13:940577. [PMID: 36248873 PMCID: PMC9562930 DOI: 10.3389/fimmu.2022.940577] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 09/05/2022] [Indexed: 01/07/2023] Open
Abstract
Background Aging is usually accompanied by functional declines of the immune system, especially in T-cell responses. However, little is known about ways to alleviate this. Methods Here, 37 middle-aged healthy participants were recruited, among which 32 were intravenously administrated with expanded NK cells and 5 with normal saline. Then, we monitored changes of peripheral senescent and exhausted T cells within 4 weeks after infusion by flow cytometry, as well as serum levels of senescence-associated secretory phenotype (SASP)-related factors. In vitro co-culture assays were performed to study NK-mediated cytotoxic activity against senescent or exhausted T cells. Functional and phenotypic alteration of NK cells before and after expansion was finally characterized. Results After NK cell infusion, senescent CD28-, CD57+, CD28-CD57+, and CD28-KLRG1+ CD4+ and CD8+ T-cell populations decreased significantly, so did PD-1+ and TIM-3+ T cells. These changes were continuously observed for 4 weeks. Nevertheless, no significant changes were observed in the normal saline group. Moreover, SASP-related factors including IL-6, IL-8, IL-1α, IL-17, MIP-1α, MIP-1β, and MMP1 were significantly decreased after NK cell infusion. Further co-culture assays showed that expanded NK cells specifically and dramatically eliminated senescent CD4+ T cells other than CD28+CD4+ T cells. They also showed improved cytotoxic activity, with different expression patterns of activating and inhibitory receptors including NKG2C, NKG2A, KLRG1, LAG3, CD57, and TIM3. Conclusion Our findings imply that T-cell senescence and exhaustion is a reversible process in healthy individuals, and autologous NK cell administration can be introduced to alleviate the aging. Clinical Trial Registration ClinicalTrials.gov, ChiCTR-OOh-17011878.
Collapse
Affiliation(s)
- Xiaofeng Tang
- Department of Blood Transfusion, Changzheng Hospital, Shanghai, China
| | - Biaolong Deng
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Aiping Zang
- Department of Research and Development, Shanghai Origincell Medical Technology Co., Ltd., Origincell Technology Group Co., Ltd., Shanghai, China
| | - Xiaowen He
- Department of Research and Development, Shanghai Origincell Medical Technology Co., Ltd., Origincell Technology Group Co., Ltd., Shanghai, China
| | - Ye Zhou
- Department of Blood Transfusion, Changzheng Hospital, Shanghai, China
| | - Daimeng Wang
- Department of Research and Development, Shanghai Origincell Medical Technology Co., Ltd., Origincell Technology Group Co., Ltd., Shanghai, China
| | - Dan Li
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xueyu Dai
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jieqiong Chen
- Department of Research and Development, Shanghai Affinity Biopharmaceutical Co., Ltd., Shanghai, China
| | - Xuhua Zhang
- Department of Research and Development, Shanghai Origincell Medical Technology Co., Ltd., Origincell Technology Group Co., Ltd., Shanghai, China
| | - Ye Liu
- Department of Blood Transfusion, Changzheng Hospital, Shanghai, China
| | - Yonghua Xu
- Department of Blood Transfusion, Changzheng Hospital, Shanghai, China
| | - Jingjing Chen
- Department of Blood Transfusion, Changzheng Hospital, Shanghai, China
| | - Weijie Zheng
- Department of Blood Transfusion, Changzheng Hospital, Shanghai, China
| | - Luding Zhang
- Department of Blood Transfusion, Changzheng Hospital, Shanghai, China
| | - Constance Gao
- Department of Biology, College of Science, Northeastern University, Boston, MA, United States
| | - Huanfeng Yang
- Department of Research and Development, Shanghai Origincell Medical Technology Co., Ltd., Origincell Technology Group Co., Ltd., Shanghai, China
| | - Bin Li
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xueqi Wang
- Department of Blood Transfusion, Changzheng Hospital, Shanghai, China
| |
Collapse
|
165
|
Conrad SJ, Raza T, Peterson EA, Liem J, Connor R, Nounamo B, Cannon M, Liu J. Myxoma virus lacking the host range determinant M062 stimulates cGAS-dependent type 1 interferon response and unique transcriptomic changes in human monocytes/macrophages. PLoS Pathog 2022; 18:e1010316. [PMID: 36103568 PMCID: PMC9473615 DOI: 10.1371/journal.ppat.1010316] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 08/04/2022] [Indexed: 11/19/2022] Open
Abstract
The evolutionarily successful poxviruses possess effective and diverse strategies to circumvent or overcome host defense mechanisms. Poxviruses encode many immunoregulatory proteins to evade host immunity to establish a productive infection and have unique means of inhibiting DNA sensing-dependent type 1 interferon (IFN-I) responses, a necessity given their dsDNA genome and exclusively cytoplasmic life cycle. We found that the key DNA sensing inhibition by poxvirus infection was dominant during the early stage of poxvirus infection before DNA replication. In an effort to identify the poxvirus gene products which subdue the antiviral proinflammatory responses (e.g., IFN-I response), we investigated the function of one early gene that is the known host range determinant from the highly conserved poxvirus host range C7L superfamily, myxoma virus (MYXV) M062. Host range factors are unique features of poxviruses that determine the species and cell type tropism. Almost all sequenced mammalian poxviruses retain at least one homologue of the poxvirus host range C7L superfamily. In MYXV, a rabbit-specific poxvirus, the dominant and broad-spectrum host range determinant of the C7L superfamily is the M062R gene. The M062R gene product is essential for MYXV infection in almost all cells tested from different mammalian species and specifically inhibits the function of host Sterile αMotif Domain-containing 9 (SAMD9), as M062R-null (ΔM062R) MYXV causes abortive infection in a SAMD9-dependent manner. In this study we investigated the immunostimulatory property of the ΔM062R. We found that the replication-defective ΔM062R activated host DNA sensing pathway during infection in a cGAS-dependent fashion and that knocking down SAMD9 expression attenuated proinflammatory responses. Moreover, transcriptomic analyses showed a unique feature of the host gene expression landscape that is different from the dsDNA alone-stimulated inflammatory state. This study establishes a link between the anti-neoplastic function of SAMD9 and the regulation of innate immune responses. Poxviruses encode a group of genes called host range determinants to maintain or expand their host tropism. The mechanism by which many viral host range factors function remains elusive. Some host range factors possess immunoregulatory functions responsible for evading or subduing host immune defense mechanisms. Most known immunoregulatory proteins encoded by poxviruses are dispensable for viral replication in vitro. The uniqueness of MYXV M062R is that it is essential for viral infection in vitro and belongs to one of the most conserved poxvirus host range families, the C7L superfamily. There is one known host target of the MYXV M062 protein, SAMD9. SAMD9 is constitutively expressed in mammalian cells and exclusively present in the cytoplasm with an anti-neoplastic function. Humans with deleterious mutations in SAMD9 present disease that ranges from lethality at a young age to a predisposition to myelodysplastic syndromes (MDS) that often require bone marrow transplantation. More importantly, SAMD9 serves as an important antiviral intrinsic molecule to many viruses. The cellular function of SAMD9 remains unclear mostly due to the difficulty of studying this protein, i.e., its large size, long half-life, and its constitutive expression in most cells. In this study we used M062R-null MYXV as a tool to study SAMD9 function and report a functional link between SAMD9 and the regulation of the proinflammatory responses triggered by cGAS-dependent DNA sensing.
Collapse
Affiliation(s)
- Steven J. Conrad
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences (UAMS), Little Rock, Arkansas, United States of America
| | - Tahseen Raza
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences (UAMS), Little Rock, Arkansas, United States of America
| | - Erich A. Peterson
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Jason Liem
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Richard Connor
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences (UAMS), Little Rock, Arkansas, United States of America
| | - Bernice Nounamo
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences (UAMS), Little Rock, Arkansas, United States of America
| | - Martin Cannon
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences (UAMS), Little Rock, Arkansas, United States of America
| | - Jia Liu
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences (UAMS), Little Rock, Arkansas, United States of America
- Center of Pathogenesis and Host Inflammatory Responses, University of Arkansas for Medical Sciences (UAMS), Little Rock, Arkansas, United States of America
- * E-mail:
| |
Collapse
|
166
|
Hernández-Silva D, Cantón-Sandoval J, Martínez-Navarro FJ, Pérez-Sánchez H, de Oliveira S, Mulero V, Alcaraz-Pérez F, Cayuela ML. Senescence-Independent Anti-Inflammatory Activity of the Senolytic Drugs Dasatinib, Navitoclax, and Venetoclax in Zebrafish Models of Chronic Inflammation. Int J Mol Sci 2022; 23:ijms231810468. [PMID: 36142384 PMCID: PMC9499634 DOI: 10.3390/ijms231810468] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Telomere shortening is the main molecular mechanism of aging, but not the only one. The adaptive immune system also ages, and older organisms tend to develop a chronic pro-inflammatory status with low-grade inflammation characterized by chronic activation of the innate immune system, called inflammaging. One of the main stimuli that fuels inflammaging is a high nutrient intake, triggering a metabolic inflammation process called metainflammation. In this study, we report the anti-inflammatory activity of several senolytic drugs in the context of chronic inflammation, by using two different zebrafish models: (i) a chronic skin inflammation model with a hypomorphic mutation in spint1a, the gene encoding the serine protease inhibitor, kunitz-type, 1a (also known as hai1a) and (ii) a non-alcoholic fatty liver disease/non-alcoholic steatohepatitis (NAFLD/NASH) model with inflammation induced by a high-fat diet. Our results show that, although these models do not manifest premature aging, the senolytic drugs dasatinib, navitoclax, and venetoclax have an anti-inflammatory effect that results in the amelioration of chronic inflammation.
Collapse
Affiliation(s)
- David Hernández-Silva
- Telomerase Cancer and Aging Group (TCAG), Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria-Arrixaca (IMIB-Arrixaca), 30120 Murcia, Spain
- Structural Bioinformatics and High-Performance Computing Research Group (BIO-HPC), Computer Engineering Department, Universidad Católica de Murcia (UCAM), Guadalupe, 30107 Murcia, Spain
| | - Joaquín Cantón-Sandoval
- Instituto Murciano de Investigación Biosanitaria-Arrixaca (IMIB-Arrixaca), 30120 Murcia, Spain
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 30100 Murcia, Spain
| | - Francisco Juan Martínez-Navarro
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Medicine (Hepatology), Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Horacio Pérez-Sánchez
- Structural Bioinformatics and High-Performance Computing Research Group (BIO-HPC), Computer Engineering Department, Universidad Católica de Murcia (UCAM), Guadalupe, 30107 Murcia, Spain
| | - Sofia de Oliveira
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Medicine (Hepatology), Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Victoriano Mulero
- Instituto Murciano de Investigación Biosanitaria-Arrixaca (IMIB-Arrixaca), 30120 Murcia, Spain
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, 30100 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 30100 Murcia, Spain
| | - Francisca Alcaraz-Pérez
- Telomerase Cancer and Aging Group (TCAG), Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria-Arrixaca (IMIB-Arrixaca), 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 30100 Murcia, Spain
- Correspondence: (F.A.-P.); (M.L.C.); Tel.: +34-968395328 (M.L.C.)
| | - María Luisa Cayuela
- Telomerase Cancer and Aging Group (TCAG), Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria-Arrixaca (IMIB-Arrixaca), 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 30100 Murcia, Spain
- Correspondence: (F.A.-P.); (M.L.C.); Tel.: +34-968395328 (M.L.C.)
| |
Collapse
|
167
|
Liu L, Yue X, Sun Z, Hambright WS, Feng Q, Cui Y, Huard J, Robbins PD, Wang Z, Mu X. Senolytic elimination of senescent macrophages restores muscle stem cell function in severely dystrophic muscle. Aging (Albany NY) 2022; 14:7650-7661. [PMID: 36084954 DOI: 10.18632/aging.204275] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/25/2022] [Indexed: 11/25/2022]
Abstract
The aging of the immune system, or immunosenescence, was recently verified to have a causal role in driving the aging of solid organs, while the senolytic elimination of senescent immune cells was found to effectively delay systemic aging. Our recent study also showed that immune cells in severely dystrophic muscles develop senescence-like phenotypes, including the increased expression of senescence-associated secretory phenotype (SASP) factors and senescence markers. Here we further investigated whether the specific clearance of senescent immune cells in dystrophic muscle may effectively improve the function of muscle stem cells and the phenotypes of dystrophic muscle. We observed increased percentage of senescent cells in macrophages from mdx/utro(-/-) mice (a murine model for muscular dystrophy disease, dystrophin-/-; utrophin-/-), while the treatment of mdx/utro(-/-) macrophages with senolytic drug fisetin resulted in reduced number of senescent cells. We administrated fisetin to mdx/utro(-/-) mice for 4 weeks, and observed obviously reduced number of senescent immune cells, restored number of muscle cells, and improve muscle phenotypes. In conclusion, our results reveal that senescent immune cells, such as macrophages, are greatly involved in the development of muscle dystrophy by impacting the function of muscle stem cells, and the senolytic ablation of these senescent cells with fisetin can be an effective therapeutic strategy for improving function of muscle stem cells and phenotypes of dystrophic muscles.
Collapse
Affiliation(s)
- Lei Liu
- Shandong The First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xianlin Yue
- Shandong The First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zewei Sun
- Shandong The First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - William S Hambright
- Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Qi Feng
- Shandong The First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yan Cui
- University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Johnny Huard
- Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Paul D Robbins
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN 55455, USA
| | - Zhihui Wang
- Shandong The First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiaodong Mu
- Shandong The First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
168
|
Jain C, Bilekova S, Lickert H. Targeting pancreatic β cells for diabetes treatment. Nat Metab 2022; 4:1097-1108. [PMID: 36131204 DOI: 10.1038/s42255-022-00618-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 07/13/2022] [Indexed: 11/09/2022]
Abstract
Insulin is a life-saving drug for patients with type 1 diabetes; however, even today, no pharmacotherapy can prevent the loss or dysfunction of pancreatic insulin-producing β cells to stop or reverse disease progression. Thus, pancreatic β cells have been a main focus for cell-replacement and regenerative therapies as a curative treatment for diabetes. In this Review, we highlight recent advances toward the development of diabetes therapies that target β cells to enhance proliferation, redifferentiation and protection from cell death and/or enable selective killing of senescent β cells. We describe currently available therapies and their mode of action, as well as insufficiencies of glucagon-like peptide 1 (GLP-1) and insulin therapies. We discuss and summarize data collected over the last decades that support the notion that pharmacological targeting of β cell insulin signalling might protect and/or regenerate β cells as an improved treatment of patients with diabetes.
Collapse
Affiliation(s)
- Chirag Jain
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Immunology Discovery, Genentech Inc., South San Francisco, CA, USA
| | - Sara Bilekova
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Chair of β-Cell Biology, Technische Universität München, School of Medicine, Klinikum Rechts der Isar, München, Germany.
| |
Collapse
|
169
|
Lascaris B, de Meijer VE, Porte RJ. Normothermic liver machine perfusion as a dynamic platform for regenerative purposes: What does the future have in store for us? J Hepatol 2022; 77:825-836. [PMID: 35533801 DOI: 10.1016/j.jhep.2022.04.033] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 04/19/2022] [Accepted: 04/27/2022] [Indexed: 01/06/2023]
Abstract
Liver transplantation has become an immense success; nevertheless, far more recipients are registered on waiting lists than there are available donor livers for transplantation. High-risk, extended criteria donor livers are increasingly used to reduce the discrepancy between organ demand and supply. Especially for high-risk livers, dynamic preservation using machine perfusion can decrease post-transplantation complications and may increase donor liver utilisation by improving graft quality and enabling viability testing before transplantation. To further increase the availability of donor livers suitable for transplantation, new strategies are required that make it possible to use organs that are initially too damaged to be transplanted. With the current progress in experimental liver transplantation research, (long-term) normothermic machine perfusion may be used in the future as a dynamic platform for regenerative medicine approaches, enabling repair and regeneration of injured donor livers. Currently explored therapeutics such as defatting cocktails, RNA interference, senolytics, and stem cell therapy may assist in the repair and/or regeneration of injured livers before transplantation. This review will provide a forecast of the future utility of normothermic machine perfusion in decreasing the imbalance between donor liver demand and supply by enabling the repair and regeneration of damaged donor livers.
Collapse
Affiliation(s)
- Bianca Lascaris
- Section of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Vincent E de Meijer
- Section of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Robert J Porte
- Section of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
170
|
Saito S, Deskin B, Rehan M, Yadav S, Matsunaga Y, Lasky JA, Thannickal VJ. Novel mediators of idiopathic pulmonary fibrosis. Clin Sci (Lond) 2022; 136:1229-1240. [PMID: 36043396 DOI: 10.1042/cs20210878] [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: 04/15/2022] [Revised: 08/04/2022] [Accepted: 08/08/2022] [Indexed: 11/17/2022]
Abstract
Fibrosis involving the lung may occur in many settings, including in association with known environmental agents, connective tissue diseases, and exposure to drugs or radiation therapy. The most common form is referred to as 'idiopathic' since a causal agent or specific association has not been determined; the strongest risk factor for idiopathic pulmonary fibrosis is aging. Emerging studies indicate that targeting certain components of aging biology may be effective in mitigating age-associated fibrosis. While transforming growth factor-β1 (TGF-β1) is a central mediator of fibrosis in almost all contexts, and across multiple organs, it is not feasible to target this canonical pathway at the ligand-receptor level due to the pleiotropic nature of its actions; importantly, its homeostatic roles as a tumor-suppressor and immune-modulator make this an imprudent strategy. However, defining targets downstream of its receptor(s) that mediate fibrogenesis, while relatively dispenable for tumor- and immune-suppressive functions may aid in developing safer and more effective therapies. In this review, we explore molecular targets that, although TGF-β1 induced/activated, may be relatively more selective in mediating tissue fibrosis. Additionally, we explore epigenetic mechanisms with global effects on the fibrogenic process, as well as metabolic pathways that regulate aging and fibrosis.
Collapse
Affiliation(s)
- Shigeki Saito
- Section of Pulmonary Diseases, Critical Care and Environmental Medicine, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, U.S.A
- John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, U.S.A, and the Southeast Louisiana Veterans Health Care System, New Orleans, LA, U.S.A
| | - Brian Deskin
- John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, U.S.A, and the Southeast Louisiana Veterans Health Care System, New Orleans, LA, U.S.A
| | - Mohammad Rehan
- John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, U.S.A, and the Southeast Louisiana Veterans Health Care System, New Orleans, LA, U.S.A
| | - Santosh Yadav
- John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, U.S.A, and the Southeast Louisiana Veterans Health Care System, New Orleans, LA, U.S.A
| | - Yasuka Matsunaga
- John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, U.S.A, and the Southeast Louisiana Veterans Health Care System, New Orleans, LA, U.S.A
| | - Joseph A Lasky
- Section of Pulmonary Diseases, Critical Care and Environmental Medicine, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, U.S.A
- John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, U.S.A, and the Southeast Louisiana Veterans Health Care System, New Orleans, LA, U.S.A
| | - Victor J Thannickal
- John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, U.S.A, and the Southeast Louisiana Veterans Health Care System, New Orleans, LA, U.S.A
| |
Collapse
|
171
|
Niveta JPS, Kumar MA, Parvathi VD. Telomere attrition and inflammation: the chicken and the egg story. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00335-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
AbstractThe challenge to improve human life span has progressed with the advent of health care services and technologies. This improvement poses a new challenge of an associated wave of diseases and pathologies that have not been observed or experienced. This has led to rise in geriatric population who are currently facing health challenges that needs to be addressed by the research community. This review focuses primarily on two mechanisms that have contributed to aging and associated pathologies: telomere attrition and inflammatory insults. A strong interplay appears to exist between telomere attrition and inflammation, and this could be the basis of many pathologies associated with increasing age. This creates a scientific dilemma as to what comes first: telomere attrition or inflammation. This review will enthuse the reader to the underlying molecules and mechanisms associated with telomere attrition and inflammation and their contribution to aging.
Collapse
|
172
|
Zhou Y, Xiao L, Long G, Cao J, Liu S, Tao Y, Zhou L, Tang J. Identification of senescence-related subtypes, establishment of a prognosis model, and characterization of a tumor microenvironment infiltration in breast cancer. Front Immunol 2022; 13:921182. [PMID: 36072578 PMCID: PMC9441960 DOI: 10.3389/fimmu.2022.921182] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 07/29/2022] [Indexed: 01/10/2023] Open
Abstract
Breast cancer is a malignancy with the highest incidence and mortality in women worldwide. Senescence is a model of arrest in the cell cycle, which plays an important role in tumor progression, while the prognostic value of cellular senescence-related genes (SRGs) in evaluating immune infiltration and clinical outcomes of breast cancer needs further investigation. In the present study, we identified two distinct molecular subtypes according to the expression profiles of 278 SRGs. We further explored the dysregulated pathways between the two subtypes and constructed a microenvironmental landscape of breast cancer. Subsequently, we established a senescence-related scoring signature based on the expression of four SRGs in the training set (GSE21653) and validated its accuracy in two validation sets (GSE20685 and GSE25055). In the training set, patients in the high-risk group had a worse prognosis than patients in the low-risk group. Multivariate Cox regression analysis showed that risk score was an independent prognostic indicator. Receiver operating characteristic curve (ROC) analysis proved the predictive accuracy of the signature. The prognostic value of this signature was further confirmed in the validation sets. We also observed that a lower risk score was associated with a higher pathological response rate in patients with neoadjuvant chemotherapy. We next performed functional experiments to validate the results above. Our study demonstrated that these cellular senescence patterns effectively grouped patients at low or high risk of disease recurrence and revealed their potential roles in the tumor–immune–stromal microenvironment. These findings enhanced our understanding of the tumor immune microenvironment and provided new insights for improving the prognosis of breast cancer patients.
Collapse
Affiliation(s)
- Yanling Zhou
- Department of Oncology, Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Liang Xiao
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Guo Long
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Jing Cao
- Department of Breast, Xiangya Hospital, Central South University, Changsha, China
| | - Shuang Liu
- Department of Oncology, Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yongguang Tao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis of the Ministry of Health, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, China
- Department of Thoracic Surgery, Hunan Key Laboratory of Tumor Models and Individualized Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| | - Ledu Zhou
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Jianing Tang
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Jianing Tang,
| |
Collapse
|
173
|
Wander SA, O’Brien N, Litchfield LM, O’Dea D, Morato Guimaraes C, Slamon DJ, Goel S. Targeting CDK4 and 6 in Cancer Therapy: Emerging Preclinical Insights Related to Abemaciclib. Oncologist 2022; 27:811-821. [PMID: 35917168 PMCID: PMC9526495 DOI: 10.1093/oncolo/oyac138] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/17/2022] [Indexed: 11/15/2022] Open
Abstract
Pharmacologic inhibitors of cyclin-dependent kinases 4 and 6 (CDK4 and 6) are approved for the treatment of subsets of patients with hormone receptor positive (HR+) breast cancer (BC). In metastatic disease, strategies involving endocrine therapy combined with CDK4 and 6 inhibitors (CDK4 and 6i) improve clinical outcomes in HR+ BCs. CDK4 and 6i prevent retinoblastoma tumor suppressor protein phosphorylation, thereby blocking the transcription of E2F target genes, which in turn inhibits both mitogen and estrogen-mediated cell proliferation. In this review, we summarize preclinical data pertaining to the use of CDK4 and 6i in BC, with a particular focus on several of the unique chemical, pharmacologic, and mechanistic properties of abemaciclib. As research efforts elucidate the novel mechanisms underlying abemaciclib activity, potential new applications are being identified. For example, preclinical studies have demonstrated abemaciclib can exert antitumor activity against multiple tumor types and can cross the blood-brain barrier. Abemaciclib has also demonstrated distinct activity as a monotherapeutic in the treatment of BC. Accordingly, we also discuss how a greater understanding of mechanisms related to CDK4 and 6 blockade highlight abemaciclib's unique in-class properties, and could pave new avenues for enhancing its therapeutic efficacy.
Collapse
Affiliation(s)
- Seth A Wander
- Seth Wander, MD, PhD, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA 02114, USA.
| | - Neil O’Brien
- Department of Medicine, Division of Hematology/Oncology, Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | | | | | - Dennis J Slamon
- Department of Medicine, Division of Hematology/Oncology, Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Shom Goel
- Corresponding author: Shom Goel, B Med Sci (Hons), MBBS (Hons), FRACP, PhD, Department of Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, VIC, 3000 Australia. Tel: +61 3 8559 8777; Fax: +61 3 8559 5039;
| |
Collapse
|
174
|
Wang Y, Dong C, Han Y, Gu Z, Sun C. Immunosenescence, aging and successful aging. Front Immunol 2022; 13:942796. [PMID: 35983061 PMCID: PMC9379926 DOI: 10.3389/fimmu.2022.942796] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/08/2022] [Indexed: 12/24/2022] Open
Abstract
Aging induces a series of immune related changes, which is called immunosenescence, playing important roles in many age-related diseases, especially neurodegenerative diseases, tumors, cardiovascular diseases, autoimmune diseases and coronavirus disease 2019(COVID-19). However, the mechanism of immunosenescence, the association with aging and successful aging, and the effects on diseases are not revealed obviously. In order to provide theoretical basis for preventing or controlling diseases effectively and achieve successful aging, we conducted the review and found that changes of aging-related phenotypes, deterioration of immune organ function and alterations of immune cell subsets participated in the process of immunosenescence, which had great effects on the occurrence and development of age-related diseases.
Collapse
Affiliation(s)
- Yunan Wang
- Department of Rheumatology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Chen Dong
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yudian Han
- Information Center, The First People’s Hospital of Nantong City, Nantong, China
| | - Zhifeng Gu
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong, China
- *Correspondence: Zhifeng Gu, ; Chi Sun,
| | - Chi Sun
- Department of Geriatrics, Affiliated Hospital of Nantong University, Nantong, China
- *Correspondence: Zhifeng Gu, ; Chi Sun,
| |
Collapse
|
175
|
Loureiro Salgado C, Mendéz Corea AF, Covre LP, De Matos Guedes HL, Falqueto A, Gomes DCO. Ageing impairs protective immunity and promotes susceptibility to murine visceral leishmaniasis. Parasitology 2022; 149:1249-1256. [PMID: 35670372 PMCID: PMC11010576 DOI: 10.1017/s0031182022000828] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/14/2022] [Accepted: 05/30/2022] [Indexed: 11/06/2022]
Abstract
It is well accepted that the impact of diseases is generally more detrimental in elderly individuals than in younger ones. Changes in the immune system due to ageing can directly affect the ability to respond effectively to infections and may contribute to the higher morbidities and mortalities in the elderly population. Leishmaniasis is a complex of clinically unique diseases caused by obligate intracellular protozoa belonging to genus Leishmania, wherein visceral leishmaniasis (VL) is the most severe form and is fatal if left untreated. In this study, aged mice (72 weeks old) presented increased susceptibility to L. infantum infection compared to younger mice (4–6-week-old), with notable parasitism in both the spleen and liver, as well as exhibiting hepatosplenomegaly. A pronounced inflammatory profile was observed in the aged-infected mice, with excessive production of TNF-α and nitrite, along with diminished IFN-γ production and reduced proliferative capacity of T cells (assessed by expression of the Ki67 marker). Additionally, both CD4+ and CD8+ T cells from the aged-infected mice presented increased expression of the inhibitory receptors PD-1 and KLRG1 that strongly correlated with the parasitism found in the liver and spleen of this group. Overall, the data reported in this study suggests for the first time that ageing may negatively impact the VL outcome and provides a perspective for new therapeutic strategies involving manipulation of immunosenescence features against Leishmania infection.
Collapse
Affiliation(s)
- Caio Loureiro Salgado
- Núcleo de Doenças Infecciosas, Universidade Federal do Espírito Santo, Vitoria, Brazil
| | | | | | - Herbet Leonel De Matos Guedes
- Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
- Instituto de Microbiologia Professor Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Aloisio Falqueto
- Departamento de Medicina Social, Universidade Federal do Espírito Santo, Vitoria, Brazil
| | - Daniel Cláudio Oliviera Gomes
- Núcleo de Doenças Infecciosas, Universidade Federal do Espírito Santo, Vitoria, Brazil
- Núcleo de Biotecnologia, Universidade Federal do Espírito Santo, Vitoria, Brazil
| |
Collapse
|
176
|
Zhang L, Pitcher LE, Yousefzadeh MJ, Niedernhofer LJ, Robbins PD, Zhu Y. Cellular senescence: a key therapeutic target in aging and diseases. J Clin Invest 2022; 132:e158450. [PMID: 35912854 PMCID: PMC9337830 DOI: 10.1172/jci158450] [Citation(s) in RCA: 252] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cellular senescence is a hallmark of aging defined by stable exit from the cell cycle in response to cellular damage and stress. Senescent cells (SnCs) can develop a characteristic pathogenic senescence-associated secretory phenotype (SASP) that drives secondary senescence and disrupts tissue homeostasis, resulting in loss of tissue repair and regeneration. The use of transgenic mouse models in which SnCs can be genetically ablated has established a key role for SnCs in driving aging and age-related disease. Importantly, senotherapeutics have been developed to pharmacologically eliminate SnCs, termed senolytics, or suppress the SASP and other markers of senescence, termed senomorphics. Based on extensive preclinical studies as well as small clinical trials demonstrating the benefits of senotherapeutics, multiple clinical trials are under way. This Review discusses the role of SnCs in aging and age-related diseases, strategies to target SnCs, approaches to discover and develop senotherapeutics, and preclinical and clinical advances of senolytics.
Collapse
Affiliation(s)
- Lei Zhang
- Institute on the Biology of Aging and Metabolism and the Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Louise E. Pitcher
- Institute on the Biology of Aging and Metabolism and the Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Matthew J. Yousefzadeh
- Institute on the Biology of Aging and Metabolism and the Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Laura J. Niedernhofer
- Institute on the Biology of Aging and Metabolism and the Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Paul D. Robbins
- Institute on the Biology of Aging and Metabolism and the Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Yi Zhu
- Robert and Arlene Kogod Center on Aging, and
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
177
|
Chaib S, Tchkonia T, Kirkland JL. Cellular senescence and senolytics: the path to the clinic. Nat Med 2022; 28:1556-1568. [PMID: 35953721 PMCID: PMC9599677 DOI: 10.1038/s41591-022-01923-y] [Citation(s) in RCA: 484] [Impact Index Per Article: 161.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/28/2022] [Indexed: 01/10/2023]
Abstract
Interlinked and fundamental aging processes appear to be a root-cause contributor to many disorders and diseases. One such process is cellular senescence, which entails a state of cell cycle arrest in response to damaging stimuli. Senescent cells can arise throughout the lifespan and, if persistent, can have deleterious effects on tissue function due to the many proteins they secrete. In preclinical models, interventions targeting those senescent cells that are persistent and cause tissue damage have been shown to delay, prevent or alleviate multiple disorders. In line with this, the discovery of small-molecule senolytic drugs that selectively clear senescent cells has led to promising strategies for preventing or treating multiple diseases and age-related conditions in humans. In this Review, we outline the rationale for senescent cells as a therapeutic target for disorders across the lifespan and discuss the most promising strategies-including recent and ongoing clinical trials-for translating small-molecule senolytics and other senescence-targeting interventions into clinical use.
Collapse
Affiliation(s)
- Selim Chaib
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Tamar Tchkonia
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - James L Kirkland
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
178
|
Lazaro-Pena MI, Ward ZC, Yang S, Strohm A, Merrill AK, Soto CA, Samuelson AV. HSF-1: Guardian of the Proteome Through Integration of Longevity Signals to the Proteostatic Network. FRONTIERS IN AGING 2022; 3:861686. [PMID: 35874276 PMCID: PMC9304931 DOI: 10.3389/fragi.2022.861686] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 06/13/2022] [Indexed: 12/15/2022]
Abstract
Discoveries made in the nematode Caenorhabditis elegans revealed that aging is under genetic control. Since these transformative initial studies, C. elegans has become a premier model system for aging research. Critically, the genes, pathways, and processes that have fundamental roles in organismal aging are deeply conserved throughout evolution. This conservation has led to a wealth of knowledge regarding both the processes that influence aging and the identification of molecular and cellular hallmarks that play a causative role in the physiological decline of organisms. One key feature of age-associated decline is the failure of mechanisms that maintain proper function of the proteome (proteostasis). Here we highlight components of the proteostatic network that act to maintain the proteome and how this network integrates into major longevity signaling pathways. We focus in depth on the heat shock transcription factor 1 (HSF1), the central regulator of gene expression for proteins that maintain the cytosolic and nuclear proteomes, and a key effector of longevity signals.
Collapse
Affiliation(s)
- Maria I. Lazaro-Pena
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, United States
| | - Zachary C. Ward
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, United States
| | - Sifan Yang
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, United States
- Department of Biology, University of Rochester, Rochester, NY, United States
| | - Alexandra Strohm
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, United States
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, United States
- Toxicology Training Program, University of Rochester Medical Center, Rochester, NY, United States
| | - Alyssa K. Merrill
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, United States
- Toxicology Training Program, University of Rochester Medical Center, Rochester, NY, United States
| | - Celia A. Soto
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, United States
- Cell Biology of Disease Graduate Program, University of Rochester Medical Center, Rochester, NY, United States
| | - Andrew V. Samuelson
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, United States
- *Correspondence: Andrew V. Samuelson,
| |
Collapse
|
179
|
Rivas M, Gupta G, Costanzo L, Ahmed H, Wyman AE, Geraghty P. Senescence: Pathogenic Driver in Chronic Obstructive Pulmonary Disease. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:817. [PMID: 35744080 PMCID: PMC9228143 DOI: 10.3390/medicina58060817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/09/2022] [Accepted: 06/15/2022] [Indexed: 01/10/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is recognized as a disease of accelerated lung aging. Over the past two decades, mounting evidence suggests an accumulation of senescent cells within the lungs of patients with COPD that contributes to dysregulated tissue repair and the secretion of multiple inflammatory proteins, termed the senescence-associated secretory phenotype (SASP). Cellular senescence in COPD is linked to telomere dysfunction, DNA damage, and oxidative stress. This review gives an overview of the mechanistic contributions and pathologic consequences of cellular senescence in COPD and discusses potential therapeutic approaches targeting senescence-associated signaling in COPD.
Collapse
Affiliation(s)
- Melissa Rivas
- Department of Medicine, State University of New York Downstate Medical Centre, Brooklyn, NY 11203, USA; (M.R.); (L.C.); (H.A.); (A.E.W.)
| | - Gayatri Gupta
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT 06520, USA;
| | - Louis Costanzo
- Department of Medicine, State University of New York Downstate Medical Centre, Brooklyn, NY 11203, USA; (M.R.); (L.C.); (H.A.); (A.E.W.)
| | - Huma Ahmed
- Department of Medicine, State University of New York Downstate Medical Centre, Brooklyn, NY 11203, USA; (M.R.); (L.C.); (H.A.); (A.E.W.)
| | - Anne E. Wyman
- Department of Medicine, State University of New York Downstate Medical Centre, Brooklyn, NY 11203, USA; (M.R.); (L.C.); (H.A.); (A.E.W.)
| | - Patrick Geraghty
- Department of Medicine, State University of New York Downstate Medical Centre, Brooklyn, NY 11203, USA; (M.R.); (L.C.); (H.A.); (A.E.W.)
| |
Collapse
|
180
|
Zha Y, Zhuang W, Yang Y, Zhou Y, Li H, Liang J. Senescence in Vascular Smooth Muscle Cells and Atherosclerosis. Front Cardiovasc Med 2022; 9:910580. [PMID: 35722104 PMCID: PMC9198250 DOI: 10.3389/fcvm.2022.910580] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/04/2022] [Indexed: 12/12/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) are the primary cell type involved in the atherosclerosis process; senescent VSMCs are observed in both aged vessels and atherosclerotic plaques. Factors associated with the atherosclerotic process, including oxidative stress, inflammation, and calcium-regulating factors, are closely linked to senescence in VSMCs. A number of experimental studies using traditional cellular aging markers have suggested that anti-aging biochemical agents could be used to treat atherosclerosis. However, doubt has recently been cast on such potential due to the increasingly apparent complexity of VSMCs status and an incomplete understanding of the role that these cells play in the atherosclerosis process, as well as a lack of specific or spectrum-limited cellular aging markers. The utility of anti-aging drugs in atherosclerosis treatment should be reevaluated. Promotion of a healthy lifestyle, exploring in depth the characteristics of each cell type associated with atherosclerosis, including VSMCs, and development of targeted drug delivery systems will ensure efficacy whilst evaluation of the safety and tolerability of drug use should be key aims of future anti-atherosclerosis research. This review summarizes the characteristics of VSMC senescence during the atherosclerosis process, the factors regulating this process, as well as an overview of progress toward the development and application of anti-aging drugs.
Collapse
Affiliation(s)
- Yiwen Zha
- Medical College, Yangzhou University, Yangzhou, China
| | - Wenwen Zhuang
- Medical College, Yangzhou University, Yangzhou, China
| | - Yongqi Yang
- Medical College, Yangzhou University, Yangzhou, China
| | - Yue Zhou
- Medical College, Yangzhou University, Yangzhou, China
| | - Hongliang Li
- Medical College, Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- *Correspondence: Hongliang Li,
| | - Jingyan Liang
- Medical College, Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
- Jingyan Liang,
| |
Collapse
|
181
|
Sabbatinelli J, Matacchione G, Giuliani A, Ramini D, Rippo MR, Procopio AD, Bonafè M, Olivieri F. Circulating biomarkers of inflammaging as potential predictors of COVID-19 severe outcomes. Mech Ageing Dev 2022; 204:111667. [PMID: 35341896 PMCID: PMC8949647 DOI: 10.1016/j.mad.2022.111667] [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: 11/24/2021] [Revised: 03/22/2022] [Accepted: 03/22/2022] [Indexed: 01/10/2023]
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 infection has been of unprecedented clinical and socio-economic worldwide relevance. The case fatality rate for COVID-19 grows exponentially with age and the presence of comorbidities. In the older patients, COVID-19 manifests predominantly as a systemic disease associated with immunological, inflammatory, and procoagulant responses. Timely diagnosis and risk stratification are crucial steps to define appropriate therapies and reduce mortality, especially in the older patients. Chronically and systemically activated innate immune responses and impaired antiviral responses have been recognized as the results of a progressive remodeling of the immune system during aging, which can be described by the words 'immunosenescence' and 'inflammaging'. These age-related features of the immune system were highlighted in patients affected by COVID-19 with the poorest clinical outcomes, suggesting that the mechanisms underpinning immunosenescence and inflammaging could be relevant for COVID-19 pathogenesis and progression. Increasing evidence suggests that senescent myeloid and endothelial cells are characterized by the acquisition of a senescence-associated pro-inflammatory phenotype (SASP), which is considered as the main culprit of both immunosenescence and inflammaging. Here, we reviewed this evidence and highlighted several circulating biomarkers of inflammaging that could provide additional prognostic information to stratify COVID-19 patients based on the risk of severe outcomes.
Collapse
Affiliation(s)
- Jacopo Sabbatinelli
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy; Laboratory Medicine, AOU Ospedali Riuniti, Ancona, Italy
| | - Giulia Matacchione
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Angelica Giuliani
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Deborah Ramini
- Center of Clinical Pathology and Innovative Therapy, IRCCS INRCA, Ancona, Italy
| | - Maria Rita Rippo
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Antonio Domenico Procopio
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy; Center of Clinical Pathology and Innovative Therapy, IRCCS INRCA, Ancona, Italy
| | - Massimiliano Bonafè
- Department of Experimental, Diagnostic and Specialty Medicine, Università di Bologna, Bologna, Italy
| | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy; Center of Clinical Pathology and Innovative Therapy, IRCCS INRCA, Ancona, Italy.
| |
Collapse
|
182
|
Abstract
Senescence is a cellular response to a variety of stress signals that is characterized by a stable withdrawal from the cell cycle and major changes in cell morphology and physiology. While most research on senescence has been performed on non-cancer cells, it is evident that cancer cells can also mount a senescence response. In this Review, we discuss how senescence can be induced in cancer cells. We describe the distinctive features of senescent cancer cells and how these changes in cellular physiology might be exploited for the selective eradication of these cells (senolysis). We discuss activation of the host immune system as a particularly attractive way to clear senescent cancer cells. Finally, we consider the challenges and opportunities provided by a 'one-two punch' sequential treatment of cancer with pro-senescence therapy followed by senolytic therapy.
Collapse
Affiliation(s)
- Liqin Wang
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Lina Lankhorst
- Cancer, Stem Cells & Developmental Biology programme, Utrecht University, Utrecht, The Netherlands
| | - René Bernards
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
183
|
Inci N, Kamali D, Akyildiz EO, Tahir Turanli E, Bozaykut P. Translation of Cellular Senescence to Novel Therapeutics: Insights From Alternative Tools and Models. FRONTIERS IN AGING 2022; 3:828058. [PMID: 35821852 PMCID: PMC9261353 DOI: 10.3389/fragi.2022.828058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 04/12/2022] [Indexed: 01/10/2023]
Abstract
Increasing chronological age is the greatest risk factor for human diseases. Cellular senescence (CS), which is characterized by permanent cell-cycle arrest, has recently emerged as a fundamental mechanism in developing aging-related pathologies. During the aging process, senescent cell accumulation results in senescence-associated secretory phenotype (SASP) which plays an essential role in tissue dysfunction. Although discovered very recently, senotherapeutic drugs have been already involved in clinical studies. This review gives a summary of the molecular mechanisms of CS and its role particularly in the development of cardiovascular diseases (CVD) as the leading cause of death. In addition, it addresses alternative research tools including the nonhuman and human models as well as computational techniques for the discovery of novel therapies. Finally, senotherapeutic approaches that are mainly classified as senolytics and senomorphics are discussed.
Collapse
Affiliation(s)
- Nurcan Inci
- Graduate School of Natural and Applied Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Dilanur Kamali
- Graduate School of Natural and Applied Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Erdogan Oguzhan Akyildiz
- Graduate School of Natural and Applied Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Eda Tahir Turanli
- Graduate School of Natural and Applied Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Perinur Bozaykut
- Graduate School of Natural and Applied Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| |
Collapse
|
184
|
Sun Y, Wang X, Liu T, Zhu X, Pan X. The multifaceted role of the SASP in atherosclerosis: from mechanisms to therapeutic opportunities. Cell Biosci 2022; 12:74. [PMID: 35642067 PMCID: PMC9153125 DOI: 10.1186/s13578-022-00815-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 05/15/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The global population of older individuals is growing, and ageing is a key risk factor for atherosclerotic cardiovascular diseases. Abnormal accumulation of senescent cells can cause potentially deleterious effects on the organism with age. As a vital marker of cellular senescence, the senescence-associated secretory phenotype (SASP) is a novel mechanism to link cellular senescence with atherosclerosis. MAIN BODY In this review, we concretely describe the characteristics of the SASP and its regulation mechanisms. Importantly, we provide novel perspectives on how the SASP can promote atherosclerosis. The SASP from different types of senescent cells have vital roles in atherosclerosis progression. As a significant mediator of the harmful effects of senescent cells, it can play a pro-atherogenic role by producing inflammation and immune dysfunction. Furthermore, the SASP can deliver senescence signals to the surrounding vascular cells, gradually contributing to the development of atherosclerosis. Finally, we focus on a variety of novel therapeutic strategies aimed to reduce the burden of atherosclerosis in elderly individuals by targeting senescent cells and inhibiting the regulatory mechanisms of the SASP. CONCLUSION This review systematically summarizes the multiple roles of the SASP in atherosclerosis and can contribute to the exploration of new therapeutic opportunities.
Collapse
Affiliation(s)
- Yu Sun
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xia Wang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Tianwei Liu
- Institute of Cerebrovascular Diseases, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiaoyan Zhu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
185
|
Lee KA, Flores RR, Jang IH, Saathoff A, Robbins PD. Immune Senescence, Immunosenescence and Aging. FRONTIERS IN AGING 2022; 3:900028. [PMID: 35821850 PMCID: PMC9261375 DOI: 10.3389/fragi.2022.900028] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 05/06/2022] [Indexed: 01/15/2023]
Abstract
With aging, there is increased dysfunction of both innate and adaptive immune responses, which contributes to impaired immune responses to pathogens and greater mortality and morbidity. This age-related immune dysfunction is defined in general as immunosenescence and includes an increase in the number of memory T cells, loss of ability to respond to antigen and a lingering level of low-grade inflammation. However, certain features of immunosenescence are similar to cellular senescence, which is defined as the irreversible loss of proliferation in response to damage and stress. Importantly, senescence cells can develop an inflammatory senescence-associated secretory phenotype (SASP), that also drives non-autonomous cellular senescence and immune dysfunction. Interestingly, viral infection can increase the extent of immune senescence both directly and indirectly, leading to increased immune dysfunction and inflammation, especially in the elderly. This review focuses on age-related immune dysfunction, cellular senescence and the impaired immune response to pathogens.
Collapse
Affiliation(s)
| | | | | | | | - Paul D. Robbins
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
186
|
Puspitasari YM, Ministrini S, Schwarz L, Karch C, Liberale L, Camici GG. Modern Concepts in Cardiovascular Disease: Inflamm-Aging. Front Cell Dev Biol 2022; 10:882211. [PMID: 35663390 PMCID: PMC9158480 DOI: 10.3389/fcell.2022.882211] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/13/2022] [Indexed: 11/18/2022] Open
Abstract
The improvements in healthcare services and quality of life result in a longer life expectancy and a higher number of aged individuals, who are inevitably affected by age-associated cardiovascular (CV) diseases. This challenging demographic shift calls for a greater effort to unravel the molecular mechanisms underlying age-related CV diseases to identify new therapeutic targets to cope with the ongoing aging "pandemic". Essential for protection against external pathogens and intrinsic degenerative processes, the inflammatory response becomes dysregulated with aging, leading to a persistent state of low-grade inflammation known as inflamm-aging. Of interest, inflammation has been recently recognized as a key factor in the pathogenesis of CV diseases, suggesting inflamm-aging as a possible driver of age-related CV afflictions and a plausible therapeutic target in this context. This review discusses the molecular pathways underlying inflamm-aging and their involvement in CV disease. Moreover, the potential of several anti-inflammatory approaches in this context is also reviewed.
Collapse
Affiliation(s)
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Internal Medicine, Angiology and Atherosclerosis, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Lena Schwarz
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Caroline Karch
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Luca Liberale
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa—Italian Cardiovascular Network, Genoa, Italy
| | - Giovanni G. Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
- Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
187
|
Villar-Álvarez F, Rosa-Carrillo DDL, Fariñas-Guerrero F, Jiménez-Ruiz CA. Immunosenescence, immune fitness and vaccination schedule in the adult respiratory patient. OPEN RESPIRATORY ARCHIVES 2022. [PMID: 37496575 PMCID: PMC9109993 DOI: 10.1016/j.opresp.2022.100181] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Immunosenescence is the gradual deterioration of the immune system caused by advancing age. It is associated with a reduced ability to respond to infections and develop long-term immune memory. It plays a key role in the development of respiratory diseases that are more common in older people, such as asthma, COPD, diffuse interstitial disease and respiratory infections in the elderly. We call immune fitness the establishment of lifestyle habits that can improve our immune capacity. We now know that good eating habits, good social relationships, not smoking, limiting alcohol consumption, exercising, controlling stress levels and establishing a proper vaccination programme can slow down the process of immunosenescence. Influenza and pneumococcal vaccines (PCV13 and PPSV23 conjugate) are well established in the adult vaccination schedule. The new pneumococcal vaccines PCV15 and PCV20 will help to extend protection against pneumococcal disease in adults. The vaccine against COVID-19 is currently the most useful tool to prevent the disease and reduce its pathogenicity. COPD patients and others with respiratory diseases may benefit from prevention of herpes zoster and Bordetella pertussis through vaccination. Respiratory syncytial virus (RSV) vaccine may be another vaccine to be added to the schedule, pending the results of its studies.
Collapse
|
188
|
Chandra A, Lagnado AB, Farr JN, Doolittle M, Tchkonia T, Kirkland JL, LeBrasseur NK, Robbins PD, Niedernhofer LJ, Ikeno Y, Passos JF, Monroe DG, Pignolo RJ, Khosla S. Targeted clearance of p21- but not p16-positive senescent cells prevents radiation-induced osteoporosis and increased marrow adiposity. Aging Cell 2022; 21:e13602. [PMID: 35363946 PMCID: PMC9124310 DOI: 10.1111/acel.13602] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/09/2022] [Accepted: 03/20/2022] [Indexed: 12/14/2022] Open
Abstract
Cellular senescence, which is a major cause of tissue dysfunction with aging and multiple other conditions, is known to be triggered by p16Ink4a or p21Cip1 , but the relative contributions of each pathway toward inducing senescence are unclear. Here, we directly addressed this issue by first developing and validating a p21-ATTAC mouse with the p21Cip1 promoter driving a "suicide" transgene encoding an inducible caspase-8 which, upon induction, selectively kills p21Cip1 -expressing senescent cells. Next, we used the p21-ATTAC mouse and the established p16-INK-ATTAC mouse to directly compare the contributions of p21Cip1 versus p16Ink4a in driving cellular senescence in a condition where a tissue phenotype (bone loss and increased marrow adiposity) is clearly driven by cellular senescence-specifically, radiation-induced osteoporosis. Using RNA in situ hybridization, we confirmed the reduction in radiation-induced p21Cip1 - or p16Ink4a -driven transcripts following senescent cell clearance in both models. However, only clearance of p21Cip1 +, but not p16Ink4a +, senescent cells prevented both radiation-induced osteoporosis and increased marrow adiposity. Reduction in senescent cells with dysfunctional telomeres following clearance of p21Cip1 +, but not p16Ink4a +, senescent cells also reduced several of the radiation-induced pro-inflammatory senescence-associated secretory phenotype factors. Thus, by directly comparing senescent cell clearance using two parallel genetic models, we demonstrate that radiation-induced osteoporosis is driven predominantly by p21Cip1 - rather than p16Ink4a -mediated cellular senescence. Further, this approach can be used to dissect the contributions of these pathways in other senescence-associated conditions, including aging across tissues.
Collapse
Affiliation(s)
- Abhishek Chandra
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
| | - Anthony B. Lagnado
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
| | - Joshua N. Farr
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
| | - Madison Doolittle
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
| | - Tamara Tchkonia
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
| | - James L. Kirkland
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
| | - Nathan K. LeBrasseur
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Department of Physical Medicine and RehabilitationMayo ClinicRochesterMinnesotaUSA
| | - Paul D. Robbins
- Institute on the Biology of Aging and MetabolismDepartment of Biochemistry, Molecular Biology and BiophysicsUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Laura J. Niedernhofer
- Institute on the Biology of Aging and MetabolismDepartment of Biochemistry, Molecular Biology and BiophysicsUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Yuji Ikeno
- Department of Pathology and Laboratory MedicineUniversity of Texas Health Science CenterSan AntonioTexasUSA
| | - João F. Passos
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
| | - David G. Monroe
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
| | - Robert J. Pignolo
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
| | - Sundeep Khosla
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
189
|
Mato-Basalo R, Lucio-Gallego S, Alarcón-Veleiro C, Sacristán-Santos M, Quintana MDPM, Morente-López M, de Toro FJ, Silva-Fernández L, González-Rodríguez A, Arufe MC, Labora JAF. Action Mechanisms of Small Extracellular Vesicles in Inflammaging. Life (Basel) 2022; 12:life12040546. [PMID: 35455036 PMCID: PMC9028066 DOI: 10.3390/life12040546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 02/05/2023] Open
Abstract
The accumulation process of proinflammatory components in the body due to aging influences intercellular communication and is known as inflammaging. This biological mechanism relates the development of inflammation to the aging process. Recently, it has been reported that small extracellular vesicles (sEVs) are mediators in the transmission of paracrine senescence involved in inflammatory aging. For this reason, their components, as well as mechanisms of action of sEVs, are relevant to develop a new therapy called senodrugs (senolytics and senomorphic) that regulates the intercellular communication of inflammaging. In this review, we include the most recent and relevant studies on the role of sEVs in the inflammatory aging process and in age-related diseases such as cancer and type 2 diabetes.
Collapse
|
190
|
Feunteun J, Ostyn P, Delaloge S. TUMOR CELL MALIGNANCY: A COMPLEX TRAIT BUILT THROUGH RECIPROCAL INTERACTIONS BETWEEN TUMORS AND TISSUE-BODY SYSTEM. iScience 2022; 25:104217. [PMID: 35494254 PMCID: PMC9044163 DOI: 10.1016/j.isci.2022.104217] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Since the discovery of oncogenes and tumor suppressor genes in the late past century, cancer research has been overwhelmingly focused on the genetics and biology of tumor cells and hence has addressed mostly cell-autonomous processes with emphasis on traditional driver/passenger genetic models. Nevertheless, over that same period, multiple seminal observations have accumulated highlighting the role of non-cell autonomous effectors in tumor growth and metastasis. However, given that cell autonomous and non-autonomous events are observed together at the time of diagnosis, it is in fact impossible to know whether the malignant transformation is initiated by cell autonomous oncogenic events or by non-cell autonomous conditions generated by alterations of the tissue-body ecosystem. This review aims at addressing this issue by taking the option of defining malignancy as a complex genetic trait incorporating genetically determined reciprocal interactions between tumor cells and tissue-body ecosystem.
Collapse
Affiliation(s)
- Jean Feunteun
- INSERM U981, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- UMR 9019, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- Corresponding author
| | - Pauline Ostyn
- UMR 9019, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Suzette Delaloge
- Breast Cancer Group, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
191
|
Dorigatti AO, Riordan R, Yu Z, Ross G, Wang R, Reynolds-Lallement N, Magnusson K, Galvan V, Perez VI. Brain cellular senescence in mouse models of Alzheimer's disease. GeroScience 2022; 44:1157-1168. [PMID: 35249206 PMCID: PMC9135905 DOI: 10.1007/s11357-022-00531-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 02/16/2022] [Indexed: 01/05/2023] Open
Abstract
The accumulation of senescent cells contributes to aging pathologies, including neurodegenerative diseases, and its selective removal improves physiological and cognitive function in wild-type mice as well as in Alzheimer's disease (AD) models. AD models recapitulate some, but not all components of disease and do so at different rates. Whether brain cellular senescence is recapitulated in some or all AD models and whether the emergence of cellular senescence in AD mouse models occurs before or after the expected onset of AD-like cognitive deficits in these models are not yet known. The goal of this study was to identify mouse models of AD and AD-related dementias that develop measurable markers of cellular senescence in brain and thus may be useful to study the role of cellular senescence in these conditions. We measured the levels of cellular senescence markers in the brains of P301S(PS19), P301L, hTau, and 3xTg-AD mice that model amyloidopathy and/or tauopathy in AD and related dementias and in wild-type, age-matched control mice for each strain. Expression of cellular senescence markers in brains of transgenic P301L and 3xTg-AD mice was largely indistinguishable from that in WT control age-matched mice. In contrast, markers of cellular senescence were differentially increased in brains of transgenic hTau and P301S(PS19) mice as compared to WT control mice before the onset of AD-like cognitive deficits. Taken together, our data suggest that P301S(PS19) and hTau mice may be useful models for the study of brain cellular senescence in tauopathies including, but not limited to, AD.
Collapse
Affiliation(s)
- Angela O Dorigatti
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Ruben Riordan
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR, USA
- Department of Biochemistry and Biophysics, Linus Pauling Institute, Oregon State University, 351 Linus Pauling Science Center, Corvallis, OR, 97331, USA
| | - Zhen Yu
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR, USA
- Department of Biochemistry and Biophysics, Linus Pauling Institute, Oregon State University, 351 Linus Pauling Science Center, Corvallis, OR, 97331, USA
| | - Grace Ross
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR, USA
- Department of Biochemistry and Biophysics, Linus Pauling Institute, Oregon State University, 351 Linus Pauling Science Center, Corvallis, OR, 97331, USA
| | - Rong Wang
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR, USA
- Department of Biochemistry and Biophysics, Linus Pauling Institute, Oregon State University, 351 Linus Pauling Science Center, Corvallis, OR, 97331, USA
| | - Nadjalisse Reynolds-Lallement
- Department of Biochemistry and Biophysics, Linus Pauling Institute, Oregon State University, 351 Linus Pauling Science Center, Corvallis, OR, 97331, USA
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, USA
| | - Kathy Magnusson
- Department of Biochemistry and Biophysics, Linus Pauling Institute, Oregon State University, 351 Linus Pauling Science Center, Corvallis, OR, 97331, USA
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, USA
| | - Veronica Galvan
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
- South Texas Veterans Health Care System, San Antonio, TX, USA.
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd, BMSB 853, Oklahoma City, OK, 73104, USA.
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Viviana I Perez
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, OR, USA.
- Department of Biochemistry and Biophysics, Linus Pauling Institute, Oregon State University, 351 Linus Pauling Science Center, Corvallis, OR, 97331, USA.
| |
Collapse
|
192
|
Abdelgawad IY, Agostinucci K, Zordoky BN. Cardiovascular ramifications of therapy-induced endothelial cell senescence in cancer survivors. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166352. [PMID: 35041996 PMCID: PMC8844223 DOI: 10.1016/j.bbadis.2022.166352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/22/2021] [Accepted: 01/07/2022] [Indexed: 12/15/2022]
Abstract
Cancer survivorship has remarkably improved over the past decades; nevertheless, cancer survivors are burdened with multiple health complications primarily caused by their cancer therapy. Therapy-induced senescence is recognized as a fundamental mechanism contributing to adverse health complications in cancer survivors. In this mini-review, we will discuss the recent literature describing the mechanisms of cancer therapy-induced senescence. We will focus on endothelial cell senescence since it has been shown to be a key player in numerous cardiovascular complications. We will also discuss novel senotherapeutic approaches that have the potential to combat therapy-induced endothelial cell senescence.
Collapse
Affiliation(s)
- Ibrahim Y Abdelgawad
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA.
| | - Kevin Agostinucci
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA.
| | - Beshay N Zordoky
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA.
| |
Collapse
|
193
|
Fang H, Deng Z, Liu J, Chen S, Deng Z, Li W. The Mechanism of Bone Remodeling After Bone Aging. Clin Interv Aging 2022; 17:405-415. [PMID: 35411139 PMCID: PMC8994557 DOI: 10.2147/cia.s349604] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/29/2022] [Indexed: 01/02/2023] Open
Abstract
Senescence mainly manifests as a series of degenerative changes in the morphological structure and function of the body. Osteoporosis is a systemic bone metabolic disease characterized by destruction of bone microstructure, low bone mineral content, decreased bone strength, and increased brittleness and fracture susceptibility. Osteoblasts, osteoclasts and osteocytes are the main cellular components of bones. However, in the process of aging, due to various self or environmental factors, the body’s function and metabolism are disordered, and osteoporosis will appear in the bones. Here, we summarize the mechanism of aging, and focus on the impact of aging on bone remodeling homeostasis, including the mechanism of ion channels on bone remodeling. Finally, we summarized the current clinical medications, targets and defects for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Huankun Fang
- Hand and Foot Surgery Department, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, 518035, People’s Republic of China
- Medical College, Shantou University, Shantou, Guangdong, 515041, People’s Republic of China
| | - Zhiqin Deng
- Hand and Foot Surgery Department, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, 518035, People’s Republic of China
| | - Jianquan Liu
- Hand and Foot Surgery Department, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, 518035, People’s Republic of China
| | - Siyu Chen
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, 518035, People’s Republic of China
| | - Zhenhan Deng
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, 518035, People’s Republic of China
- Correspondence: Zhenhan Deng, Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, 3002 Sungang West Road, Shenzhen City, 518025, People’s Republic of China, Tel +86 13928440786, Fax +86 755-83366388, Email
| | - Wencui Li
- Hand and Foot Surgery Department, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, 518035, People’s Republic of China
- Wencui Li, Department of Hand and Foot Surgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, 3002 Sungang West Road, Shenzhen City, 518025, People’s Republic of China, Tel +86 13923750767, Email
| |
Collapse
|
194
|
Sakellakis M, Flores L, Ramachandran S. Patterns of indolence in prostate cancer (Review). Exp Ther Med 2022; 23:351. [PMID: 35493432 PMCID: PMC9019743 DOI: 10.3892/etm.2022.11278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/11/2022] [Indexed: 11/20/2022] Open
Abstract
Although prostate cancer is a major cause of cancer-related mortality worldwide, most patients will have a relatively indolent clinical course. Contrary to most other types of cancer, even the diagnosis of locally advanced or metastatic disease is not always lethal. The present review aimed to summarize what is known regarding the underlying mechanisms related to the indolent course of subsets of prostate cancer, at various stages. The data suggested that no specific gene alteration by itself was responsible for carcinogenesis or disease aggressiveness. However, pathway analysis identified genetic aberrations in multiple critical pathways that tend to accumulate over the course of the disease. The progression from indolence into aggressive disease is associated with a complex interplay in which genetic and epigenetic factors are involved. The effect of the immune tumor microenvironment is also very important. Emerging evidence has suggested that the upregulation of pathways related to cellular aging and senescence can identify patients with indolent disease. In addition, a number of tumors enter a long-lasting quiescent state. Further research will determine whether halting tumor evolution is a feasible option, and whether the life of patients can be markedly prolonged by inducing tumor senescence or long-term dormancy.
Collapse
Affiliation(s)
- Minas Sakellakis
- Fourth Oncology Department and Comprehensive Clinical Trials Center, Metropolitan Hospital, 18547 Athens, Greece
| | - Laura Flores
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, University of Texas, Houston, TX 77025, USA
| | - Sumankalai Ramachandran
- Department of Genitourinary Oncology, MD Anderson Cancer Center, University of Texas, Houston, TX 77025, USA
| |
Collapse
|
195
|
Stoczynska-Fidelus E, Węgierska M, Kierasińska A, Ciunowicz D, Rieske P. Role of Senescence in Tumorigenesis and Anticancer Therapy. JOURNAL OF ONCOLOGY 2022; 2022:5969536. [PMID: 35342397 PMCID: PMC8956409 DOI: 10.1155/2022/5969536] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 01/18/2022] [Accepted: 02/05/2022] [Indexed: 12/20/2022]
Abstract
Although the role of senescence in many physiological and pathological processes is becoming more identifiable, many aspects of senescence are still enigmatic. A special attention is paid to the role of this phenomenon in tumor development and therapy. This review mainly deals with a large spectrum of oncological issues, beginning with therapy-induced senescence and ending with oncogene-induced senescence. Moreover, the role of senescence in experimental approaches, such as primary cancer cell culture or reprogramming into stem cells, is also beginning to receive further consideration. Additional focus is made on senescence resulting from mitotic catastrophe processes triggered by events occurring during mitosis and jeopardizing chromosomal stability. It has to be also realized that based on recent findings, the basics of senescent cell property interpretation, such as irreversibility of proliferation blockade, can be undermined. It shows that the definition of senescence probably requires updating. Finally, the role of senescence is lately more understandable in the immune system, especially since senescence can diminish the effectiveness of the chimeric antigen receptor T-cell (CAR-T) therapy. In this review, we summarize the current knowledge regarding all these issues.
Collapse
Affiliation(s)
- Ewelina Stoczynska-Fidelus
- Department of Molecular Biology, Chair of Medical Biology, Medical University of Lodz, Zeligowskiego 7/9 St., 90-752 Lodz, Poland
| | - Marta Węgierska
- Department of Tumor Biology, Chair of Medical Biology, Medical University of Lodz, Zeligowskiego 7/9 St., 90-752 Lodz, Poland
| | - Amelia Kierasińska
- Department of Tumor Biology, Chair of Medical Biology, Medical University of Lodz, Zeligowskiego 7/9 St., 90-752 Lodz, Poland
| | - Damian Ciunowicz
- Department of Molecular Biology, Chair of Medical Biology, Medical University of Lodz, Zeligowskiego 7/9 St., 90-752 Lodz, Poland
| | - Piotr Rieske
- Department of Tumor Biology, Chair of Medical Biology, Medical University of Lodz, Zeligowskiego 7/9 St., 90-752 Lodz, Poland
| |
Collapse
|
196
|
Cytokine-Induced Senescence in the Tumor Microenvironment and Its Effects on Anti-Tumor Immune Responses. Cancers (Basel) 2022; 14:cancers14061364. [PMID: 35326515 PMCID: PMC8946098 DOI: 10.3390/cancers14061364] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/04/2022] [Accepted: 03/04/2022] [Indexed: 02/04/2023] Open
Abstract
In contrast to surgical excision, chemotherapy or radiation therapy, immune checkpoint blockade therapies primarily influence cells in the tumor microenvironment, especially the tumor-associated lymphocytes and antigen-presenting cells. Besides complete remission of tumor lesions, in some patients, early tumor regression is followed by a consolidation phase where residing tumors remain dormant. Whereas the cytotoxic mechanisms of the regression phase (i.e., apoptosis, necrosis, necroptosis, and immune cell-mediated cell death) have been extensively described, the mechanisms underlying the dormant state are still a matter of debate. Here, we propose immune-mediated induction of senescence in cancers as one important player. Senescence can be achieved by tumor-associated antigen-specific T helper 1 cells, cytokines or antibodies targeting immune checkpoints. This concept differs from cytotoxic treatment, which often targets the genetic makeup of cancer cells. The immune system's ability to establish "defensive walls" around tumors also places the tumor microenvironment into the fight against cancer. Those "defensive walls" isolate the tumor cells instead of increasing the selective pressure. They also keep the tumor cells in a non-proliferating state, thereby correcting the derailed tissue homeostasis. In conclusion, strengthening the senescence surveillance of tumors by the immune cells of the microenvironment is a future goal to dampen this life-threatening disease.
Collapse
|
197
|
Kim K, Admasu TD, Stolzing A, Sharma A. Enhanced co-culture and enrichment of human natural killer cells for the selective clearance of senescent cells. Aging (Albany NY) 2022; 14:2131-2147. [PMID: 35245208 PMCID: PMC8954966 DOI: 10.18632/aging.203931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 02/22/2022] [Indexed: 11/26/2022]
Abstract
In the context of aging and age-associated diseases, Natural Killer (NK) cells have been revealed as a key cell type responsible for the immune clearance of senescent cells. Subsequently, NK cell-based therapies have emerged as promising alternatives to drug-based therapeutic interventions for the prevention and treatment of age-related disease and debility. Given the promise of NK cell-mediated immunotherapies as a safe and effective treatment strategy, we outline an improved method by which primary NK cells can be efficiently enriched from human peripheral blood across multiple donors (ages 20-42 years old), with a practical protocol that reliably enhances both CD56dim and CD56bright NK cells by 15-fold and 3-fold, respectively. Importantly, we show that our co-culture protocol can be used as an easily adaptable tool to assess highly efficient and selective killing of senescent cells by primary NK cells enriched via our method using longer co-culture durations and a low target to effector ratio, which may be more physiological than has been achieved in previous literature.
Collapse
Affiliation(s)
- Kristie Kim
- SENS Research Foundation, Mountain View, CA 94041, USA
| | | | - Alexandra Stolzing
- SENS Research Foundation, Mountain View, CA 94041, USA.,Loughborough University, Centre for Biological Engineering, Wolfson School of Electrical, Material and Manufacturing Engineering, Loughborough, UK
| | - Amit Sharma
- SENS Research Foundation, Mountain View, CA 94041, USA
| |
Collapse
|
198
|
Role of senescence in the chronic health consequences of COVID-19. Transl Res 2022; 241:96-108. [PMID: 34695606 PMCID: PMC8532377 DOI: 10.1016/j.trsl.2021.10.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/28/2021] [Accepted: 10/19/2021] [Indexed: 02/07/2023]
Abstract
While the full impact of COVID-19 is not yet clear, early studies have indicated that upwards of 10% of patients experience COVID-19 symptoms longer than 3 weeks, known as Long-Hauler's Syndrome or PACS (postacute sequelae of SARS-CoV-2 infection). There is little known about risk factors or predictors of susceptibility for Long-Hauler's Syndrome, but older adults are at greater risk for severe outcomes and mortality from COVID-19. The pillars of aging (including cellular senescence, telomere dysfunction, impaired proteostasis, mitochondrial dysfunction, deregulated nutrient sensing, genomic instability, progenitor cell exhaustion, altered intercellular communication, and epigenetic alterations) that contribute to age-related dysfunction and chronic diseases (the "Geroscience Hypothesis") may interfere with defenses against viral infection and consequences of these infections. Heightening of the low-grade inflammation that is associated with aging may generate an exaggerated response to an acute COVID-19 infection. Innate immune system dysfunction that leads to decreased senescent cell removal and/or increased senescent cell formation could contribute to accumulation of senescent cells with both aging and viral infections. These processes may contribute to increased risk for long-term COVID-19 sequelae in older or chronically ill patients. Hence, senolytics and other geroscience interventions that may prolong healthspan and alleviate chronic diseases and multimorbidity linked to fundamental aging processes might be an option for delaying, preventing, or alleviating Long-Hauler's Syndrome.
Collapse
Key Words
- ampk, amp-activated protein kinase
- covid-19, coronavirus disease 2019
- covid-fis, a phase 2 placebo-controlled pilot study in covid-19 of fisetin to alleviate dysfunction and excessive inflammatory response in older adults in nursing homes
- cr, caloric restriction
- fga, facility for geroscience analysis
- icu, intensive care unit
- if, intermittent fasting
- ltcf, long-term care facility
- mcc, multiple chronic conditions
- mers-cov, middle east respiratory syndrome coronavirus
- mtor, mammalian target of rapamycin
- nad+, nicotinamide adenine dinucleotide
- nmn, nicotinamide mononucleotide
- nr, nicotinamide riboside
- pacs, postacute sequalae of sars-cov-2 infection
- pamps, pathogen-associated molecular profile factors
- ros, reactive oxygen species
- sars, severe acute respiratory syndrome
- sars-cov-1, severe acute respiratory syndrome coronavirus 1
- sars-cov-2, severe acute respiratory syndrome coronavirus 2
- sasp, senescence-associated secretory phenotype
- snf, skilled nursing facility
- tgn, translational geroscience network
- who, world health organization
Collapse
|
199
|
Zeidler JD, Hogan KA, Agorrody G, Peclat TR, Kashyap S, Kanamori KS, Gomez LS, Mazdeh DZ, Warner GM, Thompson KL, Chini CCS, Chini EN. The CD38 glycohydrolase and the NAD sink: implications for pathological conditions. Am J Physiol Cell Physiol 2022; 322:C521-C545. [PMID: 35138178 PMCID: PMC8917930 DOI: 10.1152/ajpcell.00451.2021] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/12/2022] [Accepted: 01/12/2022] [Indexed: 02/07/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD) acts as a cofactor in several oxidation-reduction (redox) reactions and is a substrate for a number of nonredox enzymes. NAD is fundamental to a variety of cellular processes including energy metabolism, cell signaling, and epigenetics. NAD homeostasis appears to be of paramount importance to health span and longevity, and its dysregulation is associated with multiple diseases. NAD metabolism is dynamic and maintained by synthesis and degradation. The enzyme CD38, one of the main NAD-consuming enzymes, is a key component of NAD homeostasis. The majority of CD38 is localized in the plasma membrane with its catalytic domain facing the extracellular environment, likely for the purpose of controlling systemic levels of NAD. Several cell types express CD38, but its expression predominates on endothelial cells and immune cells capable of infiltrating organs and tissues. Here we review potential roles of CD38 in health and disease and postulate ways in which CD38 dysregulation causes changes in NAD homeostasis and contributes to the pathophysiology of multiple conditions. Indeed, in animal models the development of infectious diseases, autoimmune disorders, fibrosis, metabolic diseases, and age-associated diseases including cancer, heart disease, and neurodegeneration are associated with altered CD38 enzymatic activity. Many of these conditions are modified in CD38-deficient mice or by blocking CD38 NADase activity. In diseases in which CD38 appears to play a role, CD38-dependent NAD decline is often a common denominator of pathophysiology. Thus, understanding dysregulation of NAD homeostasis by CD38 may open new avenues for the treatment of human diseases.
Collapse
Affiliation(s)
- Julianna D Zeidler
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Kelly A Hogan
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Guillermo Agorrody
- Departamento de Fisiopatología, Hospital de Clínicas, Montevideo, Uruguay
- Laboratorio de Patologías del Metabolismo y el Envejecimiento, Instituto Pasteur de Montevideo, Montevideo, Uruguay
| | - Thais R Peclat
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Sonu Kashyap
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, Florida
| | - Karina S Kanamori
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Lilian Sales Gomez
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Delaram Z Mazdeh
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Gina M Warner
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Katie L Thompson
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Claudia C S Chini
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, Florida
| | - Eduardo Nunes Chini
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center, Department of Anesthesiology and Perioperative Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, Florida
| |
Collapse
|
200
|
Trussoni CE, O'Hara SP, LaRusso NF. Cellular senescence in the cholangiopathies: a driver of immunopathology and a novel therapeutic target. Semin Immunopathol 2022; 44:527-544. [PMID: 35178659 DOI: 10.1007/s00281-022-00909-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/12/2022] [Indexed: 12/12/2022]
Abstract
The cholangiopathies are a group of liver diseases that affect cholangiocytes, the epithelial cells that line the bile ducts. Biliary atresia (BA), primary biliary cholangitis (PBC), and primary sclerosing cholangitis (PSC) are three cholangiopathies with significant immune-mediated pathogenesis where chronic inflammation and fibrosis lead to obliteration of bile ducts and eventual liver cirrhosis. Cellular senescence is a state of cell cycle arrest in which cells become resistant to apoptosis and profusely secrete a bioactive secretome. Recent evidence indicates that cholangiocyte senescence contributes to the pathogenesis of BA, PBC, and PSC. This review explores the role of cholangiocyte senescence in BA, PBC, and PSC, ascertains how cholangiocyte senescence may promote a senescence-associated immunopathology in these cholangiopathies, and provides the rationale for therapeutically targeting senescence as a treatment option for BA, PBC, and PSC.
Collapse
Affiliation(s)
- Christy E Trussoni
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, MN, USA
| | - Steven P O'Hara
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, MN, USA
| | - Nicholas F LaRusso
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, MN, USA. .,Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, 200 First Street, SW, Rochester, MN, 55905, USA.
| |
Collapse
|