151
|
|
152
|
Kee J, Thudium S, Renner DM, Glastad K, Palozola K, Zhang Z, Li Y, Lan Y, Cesare J, Poleshko A, Kiseleva AA, Truitt R, Cardenas-Diaz FL, Zhang X, Xie X, Kotton DN, Alysandratos KD, Epstein JA, Shi PY, Yang W, Morrisey E, Garcia BA, Berger SL, Weiss SR, Korb E. SARS-CoV-2 disrupts host epigenetic regulation via histone mimicry. Nature 2022; 610:381-388. [PMID: 36198800 PMCID: PMC9533993 DOI: 10.1038/s41586-022-05282-z] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 08/26/2022] [Indexed: 02/06/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged at the end of 2019 and caused the devastating global pandemic of coronavirus disease 2019 (COVID-19), in part because of its ability to effectively suppress host cell responses1-3. In rare cases, viral proteins dampen antiviral responses by mimicking critical regions of human histone proteins4-8, particularly those containing post-translational modifications required for transcriptional regulation9-11. Recent work has demonstrated that SARS-CoV-2 markedly disrupts host cell epigenetic regulation12-14. However, how SARS-CoV-2 controls the host cell epigenome and whether it uses histone mimicry to do so remain unclear. Here we show that the SARS-CoV-2 protein encoded by ORF8 (ORF8) functions as a histone mimic of the ARKS motifs in histone H3 to disrupt host cell epigenetic regulation. ORF8 is associated with chromatin, disrupts regulation of critical histone post-translational modifications and promotes chromatin compaction. Deletion of either the ORF8 gene or the histone mimic site attenuates the ability of SARS-CoV-2 to disrupt host cell chromatin, affects the transcriptional response to infection and attenuates viral genome copy number. These findings demonstrate a new function of ORF8 and a mechanism through which SARS-CoV-2 disrupts host cell epigenetic regulation. Further, this work provides a molecular basis for the finding that SARS-CoV-2 lacking ORF8 is associated with decreased severity of COVID-19.
Collapse
Affiliation(s)
- John Kee
- Department of Genetics at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Epigenetics Institute at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Samuel Thudium
- Department of Genetics at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Epigenetics Institute at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - David M Renner
- Department of Microbiology at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Penn Center for Research on Coronaviruses and Other Emerging Pathogens at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Karl Glastad
- Epigenetics Institute at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Katherine Palozola
- Department of Genetics at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Epigenetics Institute at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Zhen Zhang
- Epigenetics Institute at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Yize Li
- Department of Microbiology at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Penn Center for Research on Coronaviruses and Other Emerging Pathogens at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Yemin Lan
- Epigenetics Institute at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Joseph Cesare
- Epigenetics Institute at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Biochemistry and Biophysics at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Andrey Poleshko
- Department of Cell and Developmental Biology at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Anna A Kiseleva
- Department of Cell and Developmental Biology at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Penn Cardiovascular Institute at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Rachel Truitt
- Department of Medicine at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Fabian L Cardenas-Diaz
- Department of Medicine at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Xianwen Zhang
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Xuping Xie
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Darrell N Kotton
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, USA
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Konstantinos D Alysandratos
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, USA
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Jonathan A Epstein
- Department of Cell and Developmental Biology at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Penn Cardiovascular Institute at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Wenli Yang
- Department of Medicine at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Edward Morrisey
- Department of Medicine at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Penn-CHOP Lung Biology Institute at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Benjamin A Garcia
- Epigenetics Institute at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Biochemistry and Biophysics at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Shelley L Berger
- Epigenetics Institute at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Biology at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Susan R Weiss
- Department of Microbiology at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Penn Center for Research on Coronaviruses and Other Emerging Pathogens at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Erica Korb
- Department of Genetics at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
- Epigenetics Institute at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
153
|
Abstract
SARS-CoV-2, the virus that causes coronavirus disease (COVID)-19, has become a persistent global health threat. Individuals who are symptomatic for COVID-19 frequently exhibit respiratory illness, which is often accompanied by neurological symptoms of anosmia and fatigue. Mounting clinical data also indicate that many COVID-19 patients display long-term neurological disorders postinfection such as cognitive decline, which emphasizes the need to further elucidate the effects of COVID-19 on the central nervous system. In this review article, we summarize an emerging body of literature describing the impact of SARS-CoV-2 infection on central nervous system (CNS) health and highlight important areas of future investigation.
Collapse
Affiliation(s)
- Nick R. Natale
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia Health System, Charlottesville, VA, USA
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, USA
- Global Biothreats Graduate Training Program, University of Virginia, Charlottesville, VA, USA
| | - John R. Lukens
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, USA
- Global Biothreats Graduate Training Program, University of Virginia, Charlottesville, VA, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA, USA
| | - William A. Petri
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia Health System, Charlottesville, VA, USA
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, USA
- Global Biothreats Graduate Training Program, University of Virginia, Charlottesville, VA, USA
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Pathology, University of Virginia Health System, Charlottesville, VA, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA, USA
| |
Collapse
|
154
|
Sun Y, Li Q, Kirkland JL. Targeting senescent cells for a healthier longevity: the roadmap for an era of global aging. LIFE MEDICINE 2022; 1:103-119. [PMID: 36699942 PMCID: PMC9869767 DOI: 10.1093/lifemedi/lnac030] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/05/2022] [Indexed: 01/28/2023]
Abstract
Aging is a natural but relentless process of physiological decline, leading to physical frailty, reduced ability to respond to physical stresses (resilience) and, ultimately, organismal death. Cellular senescence, a self-defensive mechanism activated in response to intrinsic stimuli and/or exogenous stress, is one of the central hallmarks of aging. Senescent cells cease to proliferate, while remaining metabolically active and secreting numerous extracellular factors, a feature known as the senescence-associated secretory phenotype. Senescence is physiologically important for embryonic development, tissue repair, and wound healing, and prevents carcinogenesis. However, chronic accumulation of persisting senescent cells contributes to a host of pathologies including age-related morbidities. By paracrine and endocrine mechanisms, senescent cells can induce inflammation locally and systemically, thereby causing tissue dysfunction, and organ degeneration. Agents including those targeting damaging components of the senescence-associated secretory phenotype or inducing apoptosis of senescent cells exhibit remarkable benefits in both preclinical models and early clinical trials for geriatric conditions. Here we summarize features of senescent cells and outline strategies holding the potential to be developed as clinical interventions. In the long run, there is an increasing demand for safe, effective, and clinically translatable senotherapeutics to address healthcare needs in current settings of global aging.
Collapse
Affiliation(s)
- Yu Sun
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
- Department of Pharmacology, Institute of Aging Medicine, Binzhou Medical University, Yantai 264003, China
- Department of Medicine and VAPSHCS, University of Washington, Seattle, WA 98195, USA
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - James L Kirkland
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
155
|
Lupatov AY, Yarygin KN. Telomeres and Telomerase in the Control of Stem Cells. Biomedicines 2022; 10:biomedicines10102335. [PMID: 36289597 PMCID: PMC9598777 DOI: 10.3390/biomedicines10102335] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022] Open
Abstract
Stem cells serve as a source of cellular material in embryogenesis and postnatal growth and regeneration. This requires significant proliferative potential ensured by sufficient telomere length. Telomere attrition in the stem cells and their niche cells can result in the exhaustion of the regenerative potential of high-turnover organs, causing or contributing to the onset of age-related diseases. In this review, stem cells are examined in the context of the current telomere-centric theory of cell aging, which assumes that telomere shortening depends not just on the number of cell doublings (mitotic clock) but also on the influence of various internal and external factors. The influence of the telomerase and telomere length on the functional activity of different stem cell types, as well as on their aging and prospects of use in cell therapy applications, is discussed.
Collapse
|
156
|
SARS-CoV-2 Infection of Airway Epithelium Triggers Pulmonary Endothelial Cell Activation and Senescence Associated with Type I IFN Production. Cells 2022; 11:cells11182912. [PMID: 36139488 PMCID: PMC9496907 DOI: 10.3390/cells11182912] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/09/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Airway epithelial cells represent the main target of SARS-CoV-2 replication but several pieces of evidence suggest that endothelial cells (ECs), lining pulmonary blood vessels, are key players in lung injury in COVID-19 patients. Although in vivo evidence of SARS-CoV-2 affecting the vascular endothelium exists, in vitro data are limited. In the present study, we set up an organotypic model to dissect the crosstalk between airway epithelium and pulmonary endothelial cells during SARS-CoV-2 infection. We showed that SARS-CoV-2 infected airway epithelium triggers the induction of endothelial adhesion molecules in ECs, suggesting a bystander effect of dangerous soluble signals from the infected epithelium. The endothelial activation was correlated with inflammatory cytokines (IL-1β, IL-6, IL-8) and with the viral replication in the airway epithelium. Interestingly, SARS-CoV-2 infection determined a modulation of endothelial p21, which could be partially reversed by inhibiting IFN-β production from ECs when co-cultured with HAE. Altogether, we demonstrated that SARS-CoV-2 infected epithelium triggers activation/senescence processes in ECs involving type I IFN-β production, suggesting possible antiviral/damage mechanisms occurring in the endothelium.
Collapse
|
157
|
Rai PK, Sonne C, Song H, Kim KH. The effects of COVID-19 transmission on environmental sustainability and human health: Paving the way to ensure its sustainable management. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 838:156039. [PMID: 35595144 PMCID: PMC9113776 DOI: 10.1016/j.scitotenv.2022.156039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/02/2022] [Accepted: 05/14/2022] [Indexed: 05/02/2023]
Abstract
The transmission dynamics and health risks of coronavirus disease 2019 (COVID-19) pandemic are inextricably linked to ineract with environment, climate, air pollution, and meteorological conditions. The spread of COVID-19 infection can thus perturb the 'planetary health' and livelihood by exerting impacts on the temporal and spatial variabilities of environmental pollution. Prioritization of COVID-19 by the health-care sector has been posing a serious threat to economic progress while undermining the efforts to meet the United Nations' Sustainable Development Goals (SDGs) for environmental sustainability. Here, we review the multifaceted effects of COVID-19 with respect to environmental quality, climatic variables, SDGs, energy resilience, and sustainability programs. It is well perceived that COVID-19 may have long-lasting and profound effects on socio-economic systems, food security, livelihoods, and the 'nexus' indicators. To seek for the solution of these problems, consensus can be drawn to establish and ensure a sound health-care system, a sustainable environment, and a circular bioeconomy. A holistic analysis of COVID-19's effects on multiple sectors should help develop nature-based solutions, cleaner technologies, and green economic recovery plans to help maintain environmental sustainability, ecosystem resilience, and planetary health.
Collapse
Affiliation(s)
- Prabhat Kumar Rai
- Phyto-Technologies and Plant Invasion Lab, Department of Environmental Science, School of Earth Sciences and Natural Resources Management, Mizoram University, Aizawl, Mizoram, India
| | - C Sonne
- Department of Ecoscience, Arctic Research Centre, Aarhus University, Frederiksborgvej 399, DK-4000 Roskilde, Denmark
| | - H Song
- Department of Environment and Energy, Sejong University, Seoul 05006, Republic of Korea
| | - Ki-Hyun Kim
- Department of Civil and Environmental Engineering, Hanyang University, 222 Wangsimni-Ro, Seoul 04763, Republic of Korea.
| |
Collapse
|
158
|
Sfera A, Hazan S, Anton JJ, Sfera DO, Andronescu CV, Sasannia S, Rahman L, Kozlakidis Z. Psychotropic drugs interaction with the lipid nanoparticle of COVID-19 mRNA therapeutics. Front Pharmacol 2022; 13:995481. [PMID: 36160443 PMCID: PMC9503827 DOI: 10.3389/fphar.2022.995481] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/09/2022] [Indexed: 11/18/2022] Open
Abstract
The messenger RNA (mRNA) vaccines for COVID-19, Pfizer-BioNTech and Moderna, were authorized in the US on an emergency basis in December of 2020. The rapid distribution of these therapeutics around the country and the world led to millions of people being vaccinated in a short time span, an action that decreased hospitalization and death but also heightened the concerns about adverse effects and drug-vaccine interactions. The COVID-19 mRNA vaccines are of particular interest as they form the vanguard of a range of other mRNA therapeutics that are currently in the development pipeline, focusing both on infectious diseases as well as oncological applications. The Vaccine Adverse Event Reporting System (VAERS) has gained additional attention during the COVID-19 pandemic, specifically regarding the rollout of mRNA therapeutics. However, for VAERS, absence of a reporting platform for drug-vaccine interactions left these events poorly defined. For example, chemotherapy, anticonvulsants, and antimalarials were documented to interfere with the mRNA vaccines, but much less is known about the other drugs that could interact with these therapeutics, causing adverse events or decreased efficacy. In addition, SARS-CoV-2 exploitation of host cytochrome P450 enzymes, reported in COVID-19 critical illness, highlights viral interference with drug metabolism. For example, patients with severe psychiatric illness (SPI) in treatment with clozapine often displayed elevated drug levels, emphasizing drug-vaccine interaction.
Collapse
Affiliation(s)
- Adonis Sfera
- Patton State Hospital, San Bernardino, CA, United States
- Department of Psychiatry, University of California, Riverside, Riverside, CA, United States
| | - Sabine Hazan
- Department of Psychiatry, University of California, Riverside, Riverside, CA, United States
| | - Jonathan J. Anton
- Patton State Hospital, San Bernardino, CA, United States
- Department of Biology, California Baptist University, Riverside, CA, United States
| | - Dan O. Sfera
- Patton State Hospital, San Bernardino, CA, United States
| | | | | | - Leah Rahman
- Department of Medicine, University of Oregon, Eugene, OR, United States
| | - Zisis Kozlakidis
- International Agency For Research On Cancer (IARC), Lyon, France
| |
Collapse
|
159
|
L'Hôte V, Mann C, Thuret JY. From the divergence of senescent cell fates to mechanisms and selectivity of senolytic drugs. Open Biol 2022; 12:220171. [PMID: 36128715 PMCID: PMC9490338 DOI: 10.1098/rsob.220171] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Senescence is a cellular stress response that involves prolonged cell survival, a quasi-irreversible proliferative arrest and a modification of the transcriptome that sometimes includes inflammatory gene expression. Senescent cells are resistant to apoptosis, and if not eliminated by the immune system they may accumulate and lead to chronic inflammation and tissue dysfunction. Senolytics are drugs that selectively induce cell death in senescent cells, but not in proliferative or quiescent cells, and they have proved a viable therapeutic approach in multiple mouse models of pathologies in which senescence is implicated. As the catalogue of senolytic compounds is expanding, novel survival strategies of senescent cells are uncovered, and variations in sensitivity to senolysis between different types of senescent cells emerge. We propose herein a mechanistic classification of senolytic drugs, based on the level at which they target senescent cells: directly disrupting BH3 protein networks that are reorganized upon senescence induction; downregulating survival-associated pathways essential to senescent cells; or modulating homeostatic processes whose regulation is challenged in senescence. With this approach, we highlight the important diversity of senescent cells in terms of physiology and pathways of apoptosis suppression, and we describe possible avenues for the development of more selective senolytics.
Collapse
Affiliation(s)
- Valentin L'Hôte
- CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, Gif-sur-Yvette cedex, France
| | - Carl Mann
- CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, Gif-sur-Yvette cedex, France
| | - Jean-Yves Thuret
- CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, Gif-sur-Yvette cedex, France
| |
Collapse
|
160
|
Schmitt CA, Wang B, Demaria M. Senescence and cancer - role and therapeutic opportunities. Nat Rev Clin Oncol 2022; 19:619-636. [PMID: 36045302 PMCID: PMC9428886 DOI: 10.1038/s41571-022-00668-4] [Citation(s) in RCA: 390] [Impact Index Per Article: 130.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2022] [Indexed: 01/10/2023]
Abstract
Cellular senescence is a state of stable, terminal cell cycle arrest associated with various macromolecular changes and a hypersecretory, pro-inflammatory phenotype. Entry of cells into senescence can act as a barrier to tumorigenesis and, thus, could in principle constitute a desired outcome for any anticancer therapy. Paradoxically, studies published in the past decade have demonstrated that, in certain conditions and contexts, malignant and non-malignant cells with lastingly persistent senescence can acquire pro-tumorigenic properties. In this Review, we first discuss the major mechanisms involved in the antitumorigenic functions of senescent cells and then consider the cell-intrinsic and cell-extrinsic factors that participate in their switch towards a tumour-promoting role, providing an overview of major translational and emerging clinical findings. Finally, we comprehensively describe various senolytic and senomorphic therapies and their potential to benefit patients with cancer. The entry of cells into senescence can act as a barrier to tumorigenesis; however, in certain contexts senescent malignant and non-malignant cells can acquire pro-tumorigenic properties. The authors of this Review discuss the cell-intrinsic and cell-extrinsic mechanisms involved in both the antitumorigenic and tumour-promoting roles of senescent cells, and describe the potential of various senolytic and senomorphic therapeutic approaches in oncology. Cellular senescence is a natural barrier to tumorigenesis; senescent cells are widely detected in premalignant lesions from patients with cancer. Cellular senescence is induced by anticancer therapy and can contribute to some treatment-related adverse events (TRAEs). Senescent cells exert both protumorigenic and antitumorigenic effects via cell-autonomous and paracrine mechanisms. Pharmacological modulation of senescence-associated phenotypes has the potential to improve therapy efficacy and reduce the incidence of TRAEs.
Collapse
Affiliation(s)
- Clemens A Schmitt
- Charité Universitätsmedizin Berlin, Medical Department of Hematology, Oncology and Tumour Immunology, and Molekulares Krebsforschungszentrum-MKFZ, Campus Virchow Klinikum, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Johannes Kepler University, Linz, Austria.,Kepler University Hospital, Department of Hematology and Oncology, Linz, Austria.,Deutsches Konsortium für Translationale Krebsforschung (German Cancer Consortium), Partner site Berlin, Berlin, Germany
| | - Boshi Wang
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, the Netherlands
| | - Marco Demaria
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, the Netherlands.
| |
Collapse
|
161
|
Torrance BL, Haynes L. Cellular senescence is a key mediator of lung aging and susceptibility to infection. Front Immunol 2022; 13:1006710. [PMID: 36119079 PMCID: PMC9473698 DOI: 10.3389/fimmu.2022.1006710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 08/16/2022] [Indexed: 12/05/2022] Open
Abstract
Aging results in systemic changes that leave older adults at much higher risk for adverse outcomes following respiratory infections. Much work has been done over the years to characterize and describe the varied changes that occur with aging from the molecular/cellular up to the organismal level. In recent years, the systemic accumulation of senescent cells has emerged as a key mediator of many age-related declines and diseases of aging. Many of these age-related changes can impair the normal function of the respiratory system and its capability to respond appropriately to potential pathogens that are encountered daily. In this review, we aim to establish the effects of cellular senescence on the disruption of normal lung function with aging and describe how these effects compound to leave an aged respiratory system at great risk when exposed to a pathogen. We will also discuss the role cellular senescence may play in the inability of most vaccines to confer protection against respiratory infections when administered to older adults. We posit that cellular senescence may be the point of convergence of many age-related immunological declines. Enhanced investigation into this area could provide much needed insight to understand the aging immune system and how to effectively ameliorate responses to pathogens that continue to disproportionately harm this vulnerable population.
Collapse
Affiliation(s)
| | - Laura Haynes
- UConn Center on Aging and Department of Immunology, School of Medicine, University of Connecticut, Farmington, CT, United States
| |
Collapse
|
162
|
Retuerto M, Lledó A, Fernandez-Varas B, Guerrero-López R, Usategui A, Lalueza A, García-García R, Mancebo E, Paz-Artal E, Sastre L, Perona R, Pablos JL. Shorter telomere length is associated with COVID-19 hospitalization and with persistence of radiographic lung abnormalities. Immun Ageing 2022; 19:38. [PMID: 35996190 PMCID: PMC9394033 DOI: 10.1186/s12979-022-00294-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/11/2022] [Indexed: 11/10/2022]
Abstract
Abstract
Background
Age and comorbidity are the main determinants of COVID-19 outcome. Shorter leukocyte telomere length (TL), a hallmark of biological aging, has been associated with worse COVID-19 outcomes. We sought to determine TL in patients with severe COVID-19 requiring hospitalization to analyze whether clinical outcomes and post-COVID-19 manifestations are associated with shorter TL.
Results
We analyzed 251 patients with PCR-confirmed COVID-19, hospitalized in the first months of the pandemics. We determined TL in PBL at admission by quantitative-PCR (qPCR) analysis in patients. A healthy cohort from the same area with a similar age range (n = 169) was used to calculate TL Z-scores. After hospital discharge, 144 COVID-19 survivors were followed-up for persistent COVID-19 manifestations. A second TL determination was performed in a smaller group of 63 patients 1 year later and compared with baseline TL.
Hospitalized COVID-19 patients had a decreased baseline age-adjusted TL Z-score compared to the reference group. No differences in Z-scores were observed in patients with different COVID-19 outcomes, classified as WHO ordinal scores. In 144 patients, followed for a median of 8 months, post-COVID manifestations were not associated to differences in TL. Persistence of lung radiographic abnormalities was associated with shorter baseline TL. In patients with a second TL determination, further telomere shortening (TS) was observed in 35% and telomere lengthening in 49%. Patients with further TS had suffered a more severe disease.
Conclusion
Shorter TL is associated with COVID-19 hospitalization but not with hospital clinical outcomes nor with persistent post-COVID-19 manifestations. Delayed resolution of radiographic lung abnormalities was also associated with shorter TL.
Collapse
|
163
|
Lekva T, Ueland T, Halvorsen B, Murphy SL, Dyrhol-Riise AM, Tveita A, Finbråten AK, Mathiessen A, Müller KE, Aaløkken TM, Skjønsberg OH, Lerum TV, Aukrust P, Dahl TB. Markers of cellular senescence is associated with persistent pulmonary pathology after COVID-19 infection. Infect Dis (Lond) 2022; 54:918-923. [PMID: 35984738 DOI: 10.1080/23744235.2022.2113135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND The lungs are the organ most likely to sustain serious injury from coronavirus disease 2019 (COVID-19). However, the mechanisms for long-term complications are not clear. Patients with severe COVID-19 have shorter telomere lengths and higher levels of cellular senescence, and we hypothesized that circulating levels of the telomere-associated senescence markers chitotriosidase, β-galactosidase, cathelicidin antimicrobial peptide and stathmin 1 (STMN1) were elevated in hospitalized COVID-19 patients compared to controls and could be associated with pulmonary sequelae following hospitalization. METHODS Ninety-seven hospitalized patients with COVID-19 who underwent assessment for pulmonary sequelae at three-month follow-up were included in the study. β-Galactosidase and chitotriosidase were analysed by fluorescence; stathmin 1 and cathelicidin antimicrobial peptide were analysed by enzyme immuno-assay in plasma samples from the acute phase and after three-months. In addition, the classical senescence markers cyclin-dependent kinase inhibitor 1A and 2A were analysed by enzyme immuno-assay in peripheral blood mononuclear cell lysate after three months. RESULTS We found elevated plasma levels of the senescence markers chitotriosidase and stathmin 1 in patients three months after hospitalization with COVID-19, and these markers in addition to protein levels of cyclin-dependent kinase inhibitor 2A in cell lysate, were associated with pulmonary pathology. The elevated levels of these markers seem to reflect both age-dependent (chitotriosidase) and age-independent (stathmin 1, cyclin-dependent kinase inhibitor 2A) processes. CONCLUSIONS We suggest that accelerated ageing or senescence could be important for long-term pulmonary complications of COVID-19, and our findings may be relevant for future research exploring the pathophysiology and management of these patients.
Collapse
Affiliation(s)
- Tove Lekva
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Sarah Louise Murphy
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anne Ma Dyrhol-Riise
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Infectious Diseases, Oslo University Hospital Ullevål, Oslo, Norway
| | - Anders Tveita
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Medicine, Baerum Hospital, Vestre Viken Hospital Trust, Drammen, Norway
| | | | | | - Karl Erik Müller
- Department of Internal Medicine, Drammen Hospital, Vestre Viken Hospital Trust, Drammen, Norway
| | - Trond Mogens Aaløkken
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Radiology and Nuclear Medicine, Oslo University Hospital Ullevål, Oslo, Norway
| | - Ole Henning Skjønsberg
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Pulmonary Medicine, Oslo University Hospital Ullevål, Oslo, Norway
| | - Tøri Vigeland Lerum
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Pulmonary Medicine, Oslo University Hospital Ullevål, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway.,Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Tuva Børresdatter Dahl
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.,Division of Critical Care and Emergencies, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| |
Collapse
|
164
|
Ghamar Talepoor A, Doroudchi M. Immunosenescence in atherosclerosis: A role for chronic viral infections. Front Immunol 2022; 13:945016. [PMID: 36059478 PMCID: PMC9428721 DOI: 10.3389/fimmu.2022.945016] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/26/2022] [Indexed: 01/10/2023] Open
Abstract
Immune system is a versatile and dynamic body organ which offers survival and endurance of human beings in their hostile living environment. However, similar to other cells, immune cells are hijacked by senescence. The ageing immune cells lose their beneficial functions but continue to produce inflammatory mediators which draw other immune and non-immune cells to the senescence loop. Immunosenescence has been shown to be associated with different pathological conditions and diseases, among which atherosclerosis has recently come to light. There are common drivers of both immunosenescence and atherosclerosis; e.g. inflammation, reactive oxygen species (ROS), chronic viral infections, genomic damage, oxidized-LDL, hypertension, cigarette smoke, hyperglycaemia, and mitochondrial failure. Chronic viral infections induce inflammaging, sustained cytokine signaling, ROS generation and DNA damage which are associated with atherogenesis. Accumulating evidence shows that several DNA and RNA viruses are stimulators of immunosenescence and atherosclerosis in an interrelated network. DNA viruses such as CMV, EBV and HBV upregulate p16, p21 and p53 senescence-associated molecules; induce inflammaging, metabolic reprogramming of infected cells, replicative senescence and telomere shortening. RNA viruses such as HCV and HIV induce ROS generation, DNA damage, induction of senescence-associated secretory phenotype (SASP), metabolic reprogramming of infected cells, G1 cell cycle arrest, telomere shortening, as well as epigenetic modifications of DNA and histones. The newly emerged SARS-CoV-2 virus is also a potent inducer of cytokine storm and SASP. The spike protein of SARS-CoV-2 promotes senescence phenotype in endothelial cells by augmenting p16, p21, senescence-associated β-galactosidase (SA-β-Gal) and adhesion molecules expression. The impact of SARS-CoV-2 mega-inflammation on atherogenesis, however, remains to be investigated. In this review we focus on the common processes in immunosenescence and atherogenesis caused by chronic viral infections and discuss the current knowledge on this topic.
Collapse
|
165
|
Pietzner M, Chua RL, Wheeler E, Jechow K, Willett JDS, Radbruch H, Trump S, Heidecker B, Zeberg H, Heppner FL, Eils R, Mall MA, Richards JB, Sander LE, Lehmann I, Lukassen S, Wareham NJ, Conrad C, Langenberg C. ELF5 is a potential respiratory epithelial cell-specific risk gene for severe COVID-19. Nat Commun 2022; 13:4484. [PMID: 35970849 PMCID: PMC9378714 DOI: 10.1038/s41467-022-31999-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 07/12/2022] [Indexed: 11/24/2022] Open
Abstract
Despite two years of intense global research activity, host genetic factors that predispose to a poorer prognosis of COVID-19 infection remain poorly understood. Here, we prioritise eight robust (e.g., ELF5) or suggestive but unreported (e.g., RAB2A) candidate protein mediators of COVID-19 outcomes by integrating results from the COVID-19 Host Genetics Initiative with population-based plasma proteomics using statistical colocalisation. The transcription factor ELF5 (ELF5) shows robust and directionally consistent associations across different outcome definitions, including a >4-fold higher risk (odds ratio: 4.88; 95%-CI: 2.47-9.63; p-value < 5.0 × 10-6) for severe COVID-19 per 1 s.d. higher genetically predicted plasma ELF5. We show that ELF5 is specifically expressed in epithelial cells of the respiratory system, such as secretory and alveolar type 2 cells, using single-cell RNA sequencing and immunohistochemistry. These cells are also likely targets of SARS-CoV-2 by colocalisation with key host factors, including ACE2 and TMPRSS2. In summary, large-scale human genetic studies together with gene expression at single-cell resolution highlight ELF5 as a risk gene for severe COVID-19, supporting a role of epithelial cells of the respiratory system in the adverse host response to SARS-CoV-2.
Collapse
Affiliation(s)
- Maik Pietzner
- Computational Medicine, Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany.
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK.
| | - Robert Lorenz Chua
- Center for Digital Health, Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Eleanor Wheeler
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Katharina Jechow
- Center for Digital Health, Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Julian D S Willett
- McGill Genome Centre, McGill University, Montréal, QC, Canada
- Lady Davis Institute, Jewish General Hospital, Montréal, QC, Canada
| | - Helena Radbruch
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin und Humboldt-Universität zu Berlin, Berlin, Germany
| | - Saskia Trump
- Molecular Epidemiology Unit, Center for Digital Health, Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Bettina Heidecker
- Department of Cardiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin und Humboldt-Universität zu Berlin, Berlin, Germany
| | - Hugo Zeberg
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Frank L Heppner
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin und Humboldt-Universität zu Berlin, Berlin, Germany
- Cluster of Excellence, NeuroCure, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
| | - Roland Eils
- Center for Digital Health, Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Health Data Science Unit, Heidelberg University Hospital and BioQuant, Heidelberg, Germany
- German Center for Lung Research (DZL), associated partner site, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Marcus A Mall
- German Center for Lung Research (DZL), associated partner site, Augustenburger Platz 1, 13353, Berlin, Germany
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - J Brent Richards
- McGill Genome Centre, McGill University, Montréal, QC, Canada
- Lady Davis Institute, Jewish General Hospital, Montréal, QC, Canada
- Departments of Medicine, Human Genetics, Epidemiology, Biostatistics and Occupational Health, McGill University, Montréal, QC, Canada
- Department of Twin Research, King's College London, London, United Kingdom
| | - Leif-Erik Sander
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Irina Lehmann
- Molecular Epidemiology Unit, Center for Digital Health, Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Center for Lung Research (DZL), associated partner site, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Sören Lukassen
- Center for Digital Health, Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Christian Conrad
- Center for Digital Health, Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany.
| | - Claudia Langenberg
- Computational Medicine, Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany.
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK.
| |
Collapse
|
166
|
Denholm M, Rintoul RC, Muñoz-Espín D. SARS-CoV-2-induced senescence as a potential therapeutic target. Eur Respir J 2022; 60:2201101. [PMID: 35777777 PMCID: PMC9248175 DOI: 10.1183/13993003.01101-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 06/17/2022] [Indexed: 11/12/2022]
Abstract
The global coronavirus disease 2019 (COVID-19) pandemic has caused major morbidity, mortality and socioeconomic disruption on an individual and collective level. Over 6 million COVID-19-related deaths have been reported, with total case numbers now well over 500 million worldwide [1]. Whilst the prompt and efficient design of effective vaccines has restored varying degrees of normal activity to some parts of world, the effects of the pandemic will be long in duration and far-reaching. Long implicated in the pathology of ageing, cancer and many other systemic diseases, cellular senescence is now emerging as a key factor in the pathogenesis of severe COVID-19, with implications for other viral illnesses. https://bit.ly/3bbmOuT
Collapse
Affiliation(s)
- Mary Denholm
- Early Cancer Institute, Dept of Oncology, University of Cambridge, Cambridge, UK
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Robert C Rintoul
- Royal Papworth Hospital NHS Foundation Trust, Cambridge, UK
- Cancer Research UK Cambridge Centre Thoracic Cancer Programme, Cambridge, UK
| | - Daniel Muñoz-Espín
- Early Cancer Institute, Dept of Oncology, University of Cambridge, Cambridge, UK
- Cancer Research UK Cambridge Centre Thoracic Cancer Programme, Cambridge, UK
| |
Collapse
|
167
|
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: 516] [Impact Index Per Article: 172.0] [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
|
168
|
Evangelou K, Veroutis D, Paschalaki K, Foukas PG, Lagopati N, Dimitriou M, Papaspyropoulos A, Konda B, Hazapis O, Polyzou A, Havaki S, Kotsinas A, Kittas C, Tzioufas AG, de Leval L, Vassilakos D, Tsiodras S, Stripp BR, Papantonis A, Blandino G, Karakasiliotis I, Barnes PJ, Gorgoulis VG. Pulmonary infection by SARS-CoV-2 induces senescence accompanied by an inflammatory phenotype in severe COVID-19: possible implications for viral mutagenesis. Eur Respir J 2022; 60:2102951. [PMID: 35086840 PMCID: PMC8796696 DOI: 10.1183/13993003.02951-2021] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 12/24/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection of the respiratory system can progress to a multisystemic disease with aberrant inflammatory response. Cellular senescence promotes chronic inflammation, named senescence-associated secretory phenotype (SASP). We investigated whether coronavirus disease 2019 (COVID-19) is associated with cellular senescence and SASP. METHODS Autopsy lung tissue samples from 11 COVID-19 patients and 43 age-matched non-COVID-19 controls with similar comorbidities were analysed by immunohistochemistry for SARS-CoV-2, markers of senescence and key SASP cytokines. Virally induced senescence was functionally recapitulated in vitro, by infecting epithelial Vero-E6 cells and a three-dimensional alveosphere system of alveolar type 2 (AT2) cells with SARS-CoV-2 strains isolated from COVID-19 patients. RESULTS SARS-CoV-2 was detected by immunocytochemistry and electron microscopy predominantly in AT2 cells. Infected AT2 cells expressed angiotensin-converting enzyme 2 and exhibited increased senescence (p16INK4A and SenTraGor positivity) and interleukin (IL)-1β and IL-6 expression. In vitro, infection of Vero-E6 cells with SARS-CoV-2 induced senescence (SenTraGor), DNA damage (γ-H2AX) and increased cytokine (IL-1β, IL-6, CXCL8) and apolipoprotein B mRNA-editing (APOBEC) enzyme expression. Next-generation sequencing analysis of progenies obtained from infected/senescent Vero-E6 cells demonstrated APOBEC-mediated SARS-CoV-2 mutations. Dissemination of the SARS-CoV-2-infection and senescence was confirmed in extrapulmonary sites (kidney and liver) of a COVID-19 patient. CONCLUSIONS We demonstrate that in severe COVID-19, AT2 cells infected by SARS-CoV-2 exhibit senescence and a proinflammatory phenotype. In vitro, SARS-CoV-2 infection induces senescence and inflammation. Importantly, infected senescent cells may act as a source of SARS-CoV-2 mutagenesis mediated by APOBEC enzymes. Therefore, SARS-CoV-2-induced senescence may be an important molecular mechanism of severe COVID-19, disease persistence and mutagenesis.
Collapse
Affiliation(s)
- Konstantinos Evangelou
- Molecular Carcinogenesis Group, Dept of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Contributed equally
| | - Dimitris Veroutis
- Molecular Carcinogenesis Group, Dept of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
- Contributed equally
| | - Koralia Paschalaki
- National Heart and Lung Institute, Imperial College London, London, UK
- Contributed equally
| | - Periklis G Foukas
- 2nd Dept of Pathology, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nefeli Lagopati
- Molecular Carcinogenesis Group, Dept of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Marios Dimitriou
- 2nd Dept of Pathology, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Laboratory of Biology, Dept of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Angelos Papaspyropoulos
- Molecular Carcinogenesis Group, Dept of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Bindu Konda
- Lung and Regenerative Medicine Institutes, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Orsalia Hazapis
- Molecular Carcinogenesis Group, Dept of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Aikaterini Polyzou
- Molecular Carcinogenesis Group, Dept of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Sophia Havaki
- Molecular Carcinogenesis Group, Dept of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanassios Kotsinas
- Molecular Carcinogenesis Group, Dept of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christos Kittas
- Molecular Carcinogenesis Group, Dept of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Athanasios G Tzioufas
- Dept of Pathophysiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Laurence de Leval
- Institute of Pathology, Lausanne University Hospital, Lausanne, Switzerland
| | - Demetris Vassilakos
- Molecular Carcinogenesis Group, Dept of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Sotirios Tsiodras
- 4th Dept of Internal Medicine, Attikon University Hospital, University of Athens Medical School, Athens, Greece
- Hellenic Centre for Disease Control and Prevention, Athens, Greece
| | - Barry R Stripp
- Lung and Regenerative Medicine Institutes, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Argyris Papantonis
- Translational Epigenetics Group, Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Giovanni Blandino
- Oncogenomic and Epigenetic Unit, IRCCS, Regina Elena National Cancer Institute, Rome, Italy
| | - Ioannis Karakasiliotis
- Laboratory of Biology, Dept of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Peter J Barnes
- National Heart and Lung Institute, Imperial College London, London, UK
- P.J. Barnes and V.G. Gorgoulis contributed equally to this article as lead authors and supervised the work
| | - Vassilis G Gorgoulis
- Molecular Carcinogenesis Group, Dept of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
- Faculty Institute for Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
- Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- P.J. Barnes and V.G. Gorgoulis contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
169
|
Sinha S, Castillo V, Espinoza CR, Tindle C, Fonseca AG, Dan JM, Katkar GD, Das S, Sahoo D, Ghosh P. COVID-19 lung disease shares driver AT2 cytopathic features with Idiopathic pulmonary fibrosis. EBioMedicine 2022; 82:104185. [PMID: 35870428 PMCID: PMC9297827 DOI: 10.1016/j.ebiom.2022.104185] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/06/2022] [Accepted: 07/06/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND In the aftermath of Covid-19, some patients develop a fibrotic lung disease, i.e., post-COVID-19 lung disease (PCLD), for which we currently lack insights into pathogenesis, disease models, or treatment options. METHODS Using an AI-guided approach, we analyzed > 1000 human lung transcriptomic datasets associated with various lung conditions using two viral pandemic signatures (ViP and sViP) and one covid lung-derived signature. Upon identifying similarities between COVID-19 and idiopathic pulmonary fibrosis (IPF), we subsequently dissected the basis for such similarity from molecular, cytopathic, and immunologic perspectives using a panel of IPF-specific gene signatures, alongside signatures of alveolar type II (AT2) cytopathies and of prognostic monocyte-driven processes that are known drivers of IPF. Transcriptome-derived findings were used to construct protein-protein interaction (PPI) network to identify the major triggers of AT2 dysfunction. Key findings were validated in hamster and human adult lung organoid (ALO) pre-clinical models of COVID-19 using immunohistochemistry and qPCR. FINDINGS COVID-19 resembles IPF at a fundamental level; it recapitulates the gene expression patterns (ViP and IPF signatures), cytokine storm (IL15-centric), and the AT2 cytopathic changes, e.g., injury, DNA damage, arrest in a transient, damage-induced progenitor state, and senescence-associated secretory phenotype (SASP). These immunocytopathic features were induced in pre-clinical COVID models (ALO and hamster) and reversed with effective anti-CoV-2 therapeutics in hamsters. PPI-network analyses pinpointed ER stress as one of the shared early triggers of both diseases, and IHC studies validated the same in the lungs of deceased subjects with COVID-19 and SARS-CoV-2-challenged hamster lungs. Lungs from tg-mice, in which ER stress is induced specifically in the AT2 cells, faithfully recapitulate the host immune response and alveolar cytopathic changes that are induced by SARS-CoV-2. INTERPRETATION Like IPF, COVID-19 may be driven by injury-induced ER stress that culminates into progenitor state arrest and SASP in AT2 cells. The ViP signatures in monocytes may be key determinants of prognosis. The insights, signatures, disease models identified here are likely to spur the development of therapies for patients with IPF and other fibrotic interstitial lung diseases. FUNDING This work was supported by the National Institutes for Health grants R01- GM138385 and AI155696 and funding from the Tobacco-Related disease Research Program (R01RG3780).
Collapse
Affiliation(s)
- Saptarshi Sinha
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Vanessa Castillo
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Celia R Espinoza
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Courtney Tindle
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Ayden G Fonseca
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jennifer M Dan
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Gajanan D Katkar
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Soumita Das
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA
| | - Debashis Sahoo
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA; Department of Computer Science and Engineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA 92093, USA.
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
170
|
Tsourdi E, Hofbauer LC, Rauner M. The Impact of COVID-19 in Bone Metabolism: Basic and Clinical Aspects. Horm Metab Res 2022; 54:540-548. [PMID: 35419776 DOI: 10.1055/a-1825-9641] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The use of standard procedures for the diagnosis of osteoporosis and assessment of fracture risk significantly decreased during the COVID-19 pandemic, while the incidence of fragility fractures was mostly unaltered. Both COVID-19 per se and its treatments are associated with a negative impact on bone health. Preclinical models show that mice infected with SARS-CoV2 even without symptoms display loss of trabecular bone mass two weeks post infection, due to increased numbers of osteoclasts. Osteoporosis medications do not aggravate the clinical course of COVID-19, while preclinical data suggests possible beneficial effects of some therapies. While vitamin D deficiency is clearly associated with a worse clinical course of COVID-19, evidence of improved patient outcome with vitamin D supplementation is lacking. Osteoporosis treatment should not be generally discontinued, and recommendations for substituting therapies are available. Osteoporosis therapies do not interfere with the efficacy or side-effect profiles of COVID-19 vaccines and should not be stopped or indefinitely delayed because of vaccination.
Collapse
Affiliation(s)
- Elena Tsourdi
- Department of Medicine III, University Clinic Dresden Medical Center, Dresden, Germany
- Center for Healthy Aging, University Clinic Dresden Medical Center, Dresden, Germany
| | - Lorenz C Hofbauer
- Department of Medicine III, University Clinic Dresden Medical Center, Dresden, Germany
- Center for Healthy Aging, University Clinic Dresden Medical Center, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III, University Clinic Dresden Medical Center, Dresden, Germany
- Center for Healthy Aging, University Clinic Dresden Medical Center, Dresden, Germany
| |
Collapse
|
171
|
Sinha S, Castillo V, Espinoza CR, Tindle C, Fonseca AG, Dan JM, Katkar GD, Das S, Sahoo D, Ghosh P. COVID-19 lung disease shares driver AT2 cytopathic features with Idiopathic pulmonary fibrosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2021.11.28.470269. [PMID: 34873597 PMCID: PMC8647648 DOI: 10.1101/2021.11.28.470269] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Background In the aftermath of Covid-19, some patients develop a fibrotic lung disease, i.e., p ost- C OVID-19 l ung d isease (PCLD), for which we currently lack insights into pathogenesis, disease models, or treatment options. Method Using an AI-guided approach, we analyzed > 1000 human lung transcriptomic datasets associated with various lung conditions using two viral pandemic signatures (ViP and sViP) and one covid lung-derived signature. Upon identifying similarities between COVID-19 and idiopathic pulmonary fibrosis (IPF), we subsequently dissected the basis for such similarity from molecular, cytopathic, and immunologic perspectives using a panel of IPF-specific gene signatures, alongside signatures of alveolar type II (AT2) cytopathies and of prognostic monocyte-driven processes that are known drivers of IPF. Transcriptome-derived findings were used to construct protein-protein interaction (PPI) network to identify the major triggers of AT2 dysfunction. Key findings were validated in hamster and human adult lung organoid (ALO) pre-clinical models of COVID-19 using immunohistochemistry and qPCR. Findings COVID-19 resembles IPF at a fundamental level; it recapitulates the gene expression patterns (ViP and IPF signatures), cytokine storm (IL15-centric), and the AT2 cytopathic changes, e.g., injury, DNA damage, arrest in a transient, damage-induced progenitor state, and senescence-associated secretory phenotype (SASP). These immunocytopathic features were induced in pre-clinical COVID models (ALO and hamster) and reversed with effective anti-CoV-2 therapeutics in hamsters. PPI-network analyses pinpointed ER stress as one of the shared early triggers of both diseases, and IHC studies validated the same in the lungs of deceased subjects with COVID-19 and SARS-CoV-2-challenged hamster lungs. Lungs from tg - mice, in which ER stress is induced specifically in the AT2 cells, faithfully recapitulate the host immune response and alveolar cytopathic changes that are induced by SARS-CoV-2. Interpretation Like IPF, COVID-19 may be driven by injury-induced ER stress that culminates into progenitor state arrest and SASP in AT2 cells. The ViP signatures in monocytes may be key determinants of prognosis. The insights, signatures, disease models identified here are likely to spur the development of therapies for patients with IPF and other fibrotic interstitial lung diseases. Funding This work was supported by the National Institutes for Health grants R01-GM138385 and AI155696 and funding from the Tobacco-Related disease Research Program (R01RG3780). One Sentence Summary Severe COVID-19 triggers cellular processes seen in fibrosing Interstitial Lung Disease. RESEARCH IN CONTEXT Evidence before this study: In its aftermath, the COVID-19 pandemic has left many survivors, almost a third of those who recovered, with a mysterious long-haul form of the disease which culminates in a fibrotic form of interstitial lung disease (post-COVID-19 ILD). Post-COVID-19 ILD remains a largely unknown entity. Currently, we lack insights into the core cytopathic features that drive this condition.Added value of this study: Using an AI-guided approach, which involves the use of sets of gene signatures, protein-protein network analysis, and a hamster model of COVID-19, we have revealed here that COVID-19 -lung fibrosis resembles IPF, the most common form of ILD, at a fundamental levelâ€"showing similar gene expression patterns in the lungs and blood, and dysfunctional AT2 processes (ER stress, telomere instability, progenitor cell arrest, and senescence). These findings are insightful because AT2 cells are known to contain an elegant quality control network to respond to intrinsic or extrinsic stress; a failure of such quality control results in diverse cellular phenotypes, of which ER stress appears to be a point of convergence, which appears to be sufficient to drive downstream fibrotic remodeling in the lung.Implications of all the available evidence: Because unbiased computational methods identified the shared fundamental aspects of gene expression and cellular processes between COVID-19 and IPF, the impact of our findings is likely to go beyond COVID-19 or any viral pandemic. The insights, tools (disease models, gene signatures, and biomarkers), and mechanisms identified here are likely to spur the development of therapies for patients with IPF and, other fibrotic interstitial lung diseases, all of whom have limited or no treatment options. To dissect the validated prognostic biomarkers to assess and track the risk of pulmonary fibrosis and develop therapeutics to halt fibrogenic progression.
Collapse
|
172
|
Bochyńska A, Stenzel AT, Boroujeni RS, Kuo CC, Barsoum M, Liang W, Bussmann P, Costa IG, Lüscher-Firzlaff J, Lüscher B. Induction of senescence upon loss of the Ash2l core subunit of H3K4 methyltransferase complexes. Nucleic Acids Res 2022; 50:7889-7905. [PMID: 35819198 PMCID: PMC9371893 DOI: 10.1093/nar/gkac591] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 05/30/2022] [Accepted: 06/28/2022] [Indexed: 12/12/2022] Open
Abstract
Gene expression is controlled in part by post-translational modifications of core histones. Methylation of lysine 4 of histone H3 (H3K4), associated with open chromatin and gene transcription, is catalyzed by type 2 lysine methyltransferase complexes that require WDR5, RBBP5, ASH2L and DPY30 as core subunits. Ash2l is essential during embryogenesis and for maintaining adult tissues. To expand on the mechanistic understanding of Ash2l, we generated mouse embryo fibroblasts (MEFs) with conditional Ash2l alleles. Upon loss of Ash2l, methylation of H3K4 and gene expression were downregulated, which correlated with inhibition of proliferation and cell cycle progression. Moreover, we observed induction of senescence concomitant with a set of downregulated signature genes but independent of SASP. Many of the signature genes are FoxM1 responsive. Indeed, exogenous FOXM1 was sufficient to delay senescence. Thus, although the loss of Ash2l in MEFs has broad and complex consequences, a distinct set of downregulated genes promotes senescence.
Collapse
Affiliation(s)
- Agnieszka Bochyńska
- Institute of Biochemistry and Molecular Biology, Faculty of Medicine, RWTH Aachen University, Pauwelsstrasse 30, 52057 Aachen, Germany
| | - Alexander T Stenzel
- Institute of Biochemistry and Molecular Biology, Faculty of Medicine, RWTH Aachen University, Pauwelsstrasse 30, 52057 Aachen, Germany
| | - Roksaneh Sayadi Boroujeni
- Institute of Biochemistry and Molecular Biology, Faculty of Medicine, RWTH Aachen University, Pauwelsstrasse 30, 52057 Aachen, Germany
| | - Chao-Chung Kuo
- Institute for Computational Genomics, Faculty of Medicine, RWTH Aachen University, Pauwelsstrasse 30, 52057 Aachen, Germany.,Interdisciplinary Center for Clinical Research (IZKF), Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Mirna Barsoum
- Institute of Biochemistry and Molecular Biology, Faculty of Medicine, RWTH Aachen University, Pauwelsstrasse 30, 52057 Aachen, Germany
| | - Weili Liang
- Institute of Biochemistry and Molecular Biology, Faculty of Medicine, RWTH Aachen University, Pauwelsstrasse 30, 52057 Aachen, Germany
| | - Philip Bussmann
- Institute of Biochemistry and Molecular Biology, Faculty of Medicine, RWTH Aachen University, Pauwelsstrasse 30, 52057 Aachen, Germany
| | - Ivan G Costa
- Institute for Computational Genomics, Faculty of Medicine, RWTH Aachen University, Pauwelsstrasse 30, 52057 Aachen, Germany
| | - Juliane Lüscher-Firzlaff
- Institute of Biochemistry and Molecular Biology, Faculty of Medicine, RWTH Aachen University, Pauwelsstrasse 30, 52057 Aachen, Germany
| | - Bernhard Lüscher
- Institute of Biochemistry and Molecular Biology, Faculty of Medicine, RWTH Aachen University, Pauwelsstrasse 30, 52057 Aachen, Germany
| |
Collapse
|
173
|
Xu S, Jin T, Weng J. Endothelial Cells as a Key Cell Type for Innate Immunity: A Focused Review on RIG-I Signaling Pathway. Front Immunol 2022; 13:951614. [PMID: 35865527 PMCID: PMC9294349 DOI: 10.3389/fimmu.2022.951614] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/06/2022] [Indexed: 12/25/2022] Open
Abstract
The vascular endothelium consists of a highly heterogeneous monolayer of endothelial cells (ECs) which are the primary target for bacterial and viral infections due to EC’s constant and close contact with the bloodstream. Emerging evidence has shown that ECs are a key cell type for innate immunity. Like macrophages, ECs serve as sentinels when sensing invading pathogens or microbial infection caused by viruses and bacteria. It remains elusive how ECs senses danger signals, transduce the signal and fulfil immune functions. Retinoic acid-inducible gene-I (RIG-I, gene name also known as DDX58) is an important member of RIG-I-like receptor (RLR) family that functions as an important pathogen recognition receptor (PRR) to execute immune surveillance and confer host antiviral response. Recent studies have demonstrated that virus infection, dsRNA, dsDNA, interferons, LPS, and 25-hydroxycholesterol (25-HC) can increase RIG-1 expression in ECs and propagate anti-viral response. Of translational significance, RIG-I activation can be inhibited by Panax notoginseng saponins, endogenous PPARγ ligand 15-PGJ2, tryptanthrin and 2-animopurine. Considering the pivotal role of inflammation and innate immunity in regulating endothelial dysfunction and atherosclerosis, here we provided a concise review of the role of RIG-I in endothelial cell function and highlight future direction to elucidate the potential role of RIG-I in regulating cardiovascular diseases as well as virus infectious disease, including COVID-19. Furthered understanding of RIG-I-mediated signaling pathways is important to control disorders associated with altered immunity and inflammation in ECs.
Collapse
Affiliation(s)
- Suowen Xu
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China
- Laboratory of Metabolics and Cardiovascular Diseases, Institute of Endocrine and Metabolic Diseases, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province , University of Science and Technology of China, Hefei, China
- *Correspondence: Suowen Xu, ; Jianping Weng,
| | - Tengchuan Jin
- Laboratory of Structural Immunology, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jianping Weng
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China
- Laboratory of Metabolics and Cardiovascular Diseases, Institute of Endocrine and Metabolic Diseases, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province , University of Science and Technology of China, Hefei, China
- *Correspondence: Suowen Xu, ; Jianping Weng,
| |
Collapse
|
174
|
Kawamoto S, Matsumoto T, Takasugi M, Hara E. The 6th international cell senescence association conference. Genes Cells 2022; 27:517-525. [PMID: 35726163 DOI: 10.1111/gtc.12970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/01/2022] [Accepted: 06/01/2022] [Indexed: 11/28/2022]
Abstract
The 6th conference of the international cell senescence association (ICSA) on the theme of "A New Era of Senescence Research: The Challenge of Controlling Aging and Cancer" was held on December 12-15, 2021 in Osaka, Japan as a Hybrid Meeting. The conference brought together basic and translational scientists to discuss recent developments in the field of cellular senescence research. In recent years, the study of cellular senescence has become a very hot field of research. It is clear that the ICSA, founded in 2015, has played an important role in this process. The 6th ICSA conference has provided another opportunity for exchanges and new connections between basic and translational scientists. The scientific program consisted of keynote lectures, invited talks, short talks selected from abstracts, a poster session, and luncheon seminars sponsored by the Japanese Society of Anti-Aging Medicine. In the Meet the Editor session, Dr Christoph Schmitt, Editor-in-Chief of Nature Metabolism, gave a short presentation about the journal and answered questions from the audience. Being a hybrid meeting, there was only so much that could be done, but we hope that the meeting was fruitful.
Collapse
Affiliation(s)
- Shimpei Kawamoto
- Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Tomonori Matsumoto
- Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Masaki Takasugi
- Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Eiji Hara
- Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| |
Collapse
|
175
|
Ferrari D, Rubini M, Burns JS. The Potential of Purinergic Signaling to Thwart Viruses Including SARS-CoV-2. Front Immunol 2022; 13:904419. [PMID: 35784277 PMCID: PMC9248768 DOI: 10.3389/fimmu.2022.904419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/05/2022] [Indexed: 01/18/2023] Open
Abstract
A long-shared evolutionary history is congruent with the multiple roles played by purinergic signaling in viral infection, replication and host responses that can assist or hinder viral functions. An overview of the involvement of purinergic signaling among a range of viruses is compared and contrasted with what is currently understood for SARS-CoV-2. In particular, we focus on the inflammatory and antiviral responses of infected cells mediated by purinergic receptor activation. Although there is considerable variation in a patient's response to SARS-CoV-2 infection, a principle immediate concern in Coronavirus disease (COVID-19) is the possibility of an aberrant inflammatory activation causing diffuse lung oedema and respiratory failure. We discuss the most promising potential interventions modulating purinergic signaling that may attenuate the more serious repercussions of SARS-CoV-2 infection and aspects of their implementation.
Collapse
Affiliation(s)
- Davide Ferrari
- Section of Microbiology and Applied Pathology, University of Ferrara, Ferrara, Italy
- Department of Life Science and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Michele Rubini
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Jorge S. Burns
- Department of Life Science and Biotechnology, University of Ferrara, Ferrara, Italy
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
176
|
Khan A, Iqtadar S, Mumtaz SU, Heinrich M, Pascual-Figal DA, Livingstone S, Abaidullah S. Oral Co-Supplementation of Curcumin, Quercetin, and Vitamin D3 as an Adjuvant Therapy for Mild to Moderate Symptoms of COVID-19—Results From a Pilot Open-Label, Randomized Controlled Trial. Front Pharmacol 2022; 13:898062. [PMID: 35747751 PMCID: PMC9211374 DOI: 10.3389/fphar.2022.898062] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 04/27/2022] [Indexed: 12/15/2022] Open
Abstract
Background: Curcumin, quercetin, and vitamin D3 (cholecalciferol) are common natural ingredients of human nutrition and reportedly exhibit promising anti-inflammatory, immunomodulatory, broad-spectrum antiviral, and antioxidant activities. Objective: The present study aimed to investigate the possible therapeutic benefits of a single oral formulation containing supplements curcumin, quercetin, and cholecalciferol (combinedly referred to here as CQC) as an adjuvant therapy for early-stage of symptomatic coronavirus disease 2019 (COVID-19) in a pilot open-label, randomized controlled trial conducted at Mayo Hospital, King Edward Medical University, Lahore, Pakistan. Methods: Reverse transcriptase polymerase chain reaction (RT-PCR) confirmed, mild to moderate symptomatic COVID-19 outpatients were randomized to receive either the standard of care (SOC) (n = 25) (control arm) or a daily oral co-supplementation of 168 mg curcumin, 260 mg quercetin, and 9 µg (360 IU) of cholecalciferol, as two oral soft capsules b.i.d. as an add-on to the SOC (n = 25) (CQC arm) for 14 days. The SOC includes paracetamol with or without antibiotic (azithromycin). Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RT-PCR test, acute symptoms, and biochemistry including C-reactive protein (CRP), D-dimer, lactate dehydrogenase, ferritin, and complete blood count were evaluated at baseline and follow-up day seven. Results: Patients who received the CQC adjuvant therapy showed expedited negativization of the SARS-CoV-2 RT-PCR test, i.e., 15 (60.0%) vs. five (20.0%) of the control arm, p = 0.009. COVID-19- associated acute symptoms were rapidly resolved in the CQC arm, i.e., 15 (60.0%) vs. 10 (40.0%) of the control arm, p = 0.154. Patients in the CQC arm experienced a greater fall in serum CRP levels, i.e., from (median (IQR) 34.0 (21.0, 45.0) to 11.0 (5.0, 16.0) mg/dl as compared to the control arm, i.e., from 36.0 (28.0, 47.0) to 22.0 (15.0, 25.0) mg/dl, p = 0.006. The adjuvant therapy of co-supplementation of CQC was safe and well-tolerated by all 25 patients and no treatment-emergent effects, complications, side effects, or serious adverse events were reported. Conclusion: The co-supplementation of CQC may possibly have a therapeutic role in the early stage of COVID-19 infection including speedy negativization of the SARS-CoV-2 RT-PCR test, resolution of acute symptoms, and modulation of the hyperinflammatory response. In combination with routine care, the adjuvant co-supplementation of CQC may possibly help in the speedy recovery from early-stage mild to moderate symptoms of COVID-19. Further research is warranted. Clinical Trial Registration:Clinicaltrials.gov, identifier NCT05130671
Collapse
Affiliation(s)
- Amjad Khan
- INEOS Oxford Institute for AMR Research, University of Oxford, Oxford, United Kingdom
- Department of Biochemistry, Liaquat University of Medical and Health Sciences, Jamshoro, Pakistan
- *Correspondence: Amjad Khan,
| | - Somia Iqtadar
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | - Sami Ullah Mumtaz
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | - Michael Heinrich
- UCL School of Pharmacy, University of London, London, United Kingdom
| | - Domingo A. Pascual-Figal
- Department of Cardiology, University of Murcia Hospital Universitario Virgen de la Arrixaca Murcia, Murcia, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Sajid Abaidullah
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| |
Collapse
|
177
|
Schmitt CA. COVID-19: New disease mechanism is simultaneously a therapeutic target. MEMO 2022; 15:100-101. [PMID: 35693203 PMCID: PMC9172975 DOI: 10.1007/s12254-022-00813-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/25/2022] [Indexed: 06/15/2023]
Affiliation(s)
- Clemens A. Schmitt
- Department of Haematology and Oncology, Kepler University Hospital, Johannes Kepler University, Krankenhausstr. 7a, 4020 Linz, Austria
| |
Collapse
|
178
|
Lund Da Costa A, Mehta H, Mathur R. Promoting autophagy to mitigate coronavirus disease pathology in the elderly. CLINICAL AND TRANSLATIONAL DISCOVERY 2022; 2:e68. [PMID: 35942234 PMCID: PMC9347787 DOI: 10.1002/ctd2.68] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 04/25/2022] [Indexed: 11/11/2022]
Abstract
In this commentary, we highlight autophagy's important function, while identifying potential therapeutic targets for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the elderly. Autophagy's decline in the elderly causes increased cell senescence and a dysregulated immune system. As this demographic often faces decreased vaccine-provided immunity, coronavirus disease 2019 treatments must be developed. We discuss a recent study by Acharya et al. (2022) that found that SF2523 induced autophagy, reducing SARS-CoV-2 replication. Furthermore, across varying dosages, SF2523 was shown to have a synergistic effect with remdesivir or MU-UNMC. Consequently, we believe that SF2523, alone or with other anti-virals, is a promising potential therapeutic for preventing SARS-CoV-2-related mortalities.
Collapse
Affiliation(s)
- Abby Lund Da Costa
- Department of Geriatrics, School of Medicine and Health ScienceUniversity of North DakotaGrand ForksUSA
| | - Het Mehta
- Department of Geriatrics, School of Medicine and Health ScienceUniversity of North DakotaGrand ForksUSA
| | - Ramkumar Mathur
- Department of Geriatrics, School of Medicine and Health ScienceUniversity of North DakotaGrand ForksUSA
| |
Collapse
|
179
|
Dynamics of circulating calprotectin accurately predict the outcome of moderate COVID-19 patients. EBioMedicine 2022; 80:104077. [PMID: 35644124 PMCID: PMC9132728 DOI: 10.1016/j.ebiom.2022.104077] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/26/2022] [Accepted: 05/10/2022] [Indexed: 12/11/2022] Open
|
180
|
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
|
181
|
Kandhaya‐Pillai R, Yang X, Tchkonia T, Martin GM, Kirkland JL, Oshima J. TNF-α/IFN-γ synergy amplifies senescence-associated inflammation and SARS-CoV-2 receptor expression via hyper-activated JAK/STAT1. Aging Cell 2022; 21:e13646. [PMID: 35645319 PMCID: PMC9197409 DOI: 10.1111/acel.13646] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 05/01/2022] [Indexed: 12/12/2022] Open
Abstract
Older age and underlying conditions such as diabetes/obesity or immunosuppression are leading host risk factors for developing severe complications from COVID-19 infection. The pathogenesis of COVID-19-related cytokine storm, tissue damage, and fibrosis may be interconnected with fundamental aging processes, including dysregulated immune responses and cellular senescence. Here, we examined effects of key cytokines linked to cellular senescence on expression of SARS-CoV-2 viral entry receptors. We found exposure of human umbilical vein endothelial cells (HUVECs) to the inflammatory cytokines, TNF-α + IFN-γ or a cocktail of TNF-α + IFN-γ + IL-6, increased expression of ACE2/DPP4, accentuated the pro-inflammatory senescence-associated secretory phenotype (SASP), and decreased cellular proliferative capacity, consistent with progression towards a cellular senescence-like state. IL-6 by itself failed to induce substantial effects on viral entry receptors or SASP-related genes, while synergy between TNF-α and IFN-γ initiated a positive feedback loop via hyper-activation of the JAK/STAT1 pathway, causing SASP amplification. Breaking the interactive loop between senescence and cytokine secretion with JAK inhibitor ruxolitinib or antiviral drug remdesivir prevented hyper-inflammation, normalized SARS-CoV-2 entry receptor expression, and restored HUVECs proliferative capacity. This loop appears to underlie cytokine-mediated viral entry receptor activation and links with senescence and hyper-inflammation.
Collapse
Affiliation(s)
- Renuka Kandhaya‐Pillai
- Department of Laboratory Medicine & PathologyUniversity of WashingtonSeattleWashingtonUSA
| | - Xiaomeng Yang
- Department of Laboratory Medicine & PathologyUniversity of WashingtonSeattleWashingtonUSA
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Department of PhysiologyMayo ClinicRochesterMinnesotaUSA
| | - George M. Martin
- Department of Laboratory Medicine & PathologyUniversity of WashingtonSeattleWashingtonUSA
| | - James L. Kirkland
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Department of PhysiologyMayo ClinicRochesterMinnesotaUSA
- Department of MedicineMayo ClinicRochesterMinnesotaUSA
| | - Junko Oshima
- Department of Laboratory Medicine & PathologyUniversity of WashingtonSeattleWashingtonUSA
| |
Collapse
|
182
|
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: 120] [Impact Index Per Article: 40.0] [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
|
183
|
Licastro F. Special Issue Editorial: "Infections, Inflammation and Neurodegeneration in Alzheimer Disease" Infections, Neuronal Senescence, and Dementia. Int J Mol Sci 2022; 23:ijms23115865. [PMID: 35682542 PMCID: PMC9180241 DOI: 10.3390/ijms23115865] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/11/2022] [Accepted: 05/17/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease (AD) is a complex chronic disease of the brain characterized by several neurodegenerative mechanisms and is responsible for most dementia cases in the elderly. Declining immunity during ageing is often associated with peripheral chronic inflammation, and chronic neuroinflammation is a constant component of AD brain pathology. In the Special Issue published in 2021 eight papers were collected regarding different aspects of neurodegeneration associated with AD. Five papers presented and discussed infectious agents involved in brain AD pathology and three discussed data regarding receptors regulation and possible treatment of the disease. Below I will discuss and further elaborate on topics related to infections, inflammation, and neurodegenerative pathways in AD and brain senescence. The topic presented here may contribute to early intervention protocols for preventing or slowing the progression of cognitive deterioration in the elderly.
Collapse
Affiliation(s)
- Federico Licastro
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
184
|
Kim C, Lee SG, Lim S, Jung M, Kwon SP, Hong J, Kang M, Sohn HS, Go S, Moon S, Lee SJ, Kim JS, Kim BS. A Senolytic-Eluting Coronary Stent for the Prevention of In-Stent Restenosis. ACS Biomater Sci Eng 2022; 8:1921-1929. [PMID: 35416659 DOI: 10.1021/acsbiomaterials.1c01611] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The vast majority of drug-eluting stents (DES) elute either sirolimus or one of its analogues. While limus drugs stymie vascular smooth muscle cell (VSMC) proliferation to prevent in-stent restenosis, their antiproliferative nature is indiscriminate and limits healing of the endothelium in stented vessels, increasing the risk of late-stent thrombosis. Oxidative stress, which is associated with vascular injury from stent implantation, can induce VSMCs to undergo senescence, and senescent VSMCs can produce pro-inflammatory cytokines capable of inducing proliferation of neighboring nonsenescent VSMCs. We explored the potential of senolytic therapy, which involves the selective elimination of senescent cells, in the form of a senolytic-eluting stent (SES) for interventional cardiology. Oxidative stress was modeled in vitro by exposing VSMCs to H2O2, and H2O2-mediated senescence was evaluated by cytochemical staining of senescence-associated β-galactosidase activity and qRT-PCR. Quiescent VSMCs were then treated with the conditioned medium (CM) of H2O2-treated VSMCs. Proliferative effects of CM were analyzed by staining for proliferating cell nuclear antigen. Senolytic effects of the first-generation senolytic ABT263 were observed in vitro, and the effects of ABT263 on endothelial cells were also investigated through an in vitro re-endothelialization assay. SESs were prepared by dip coating. Iliofemoral arteries of hypercholesteremic rabbits were implanted with SES, everolimus-eluting stents (EESs), or bare-metal stents (BMSs), and the area of stenosis was measured 4 weeks post-implantation using optical coherence tomography. We found that a portion of H2O2-treated VSMCs underwent senescence, and that CM of H2O2-treated senescent VSMCs triggered the proliferation of quiescent VSMCs. ABT263 reverted H2O2-mediated senescence and the proliferative capacity of senescent VSMC CM. Unlike everolimus, ABT263 did not affect endothelial cell migration and/or proliferation. SES, but not EES, significantly reduced stenosis area in vivo compared with bare-metal stents (BMSs). This study shows the potential of SES as an alternative to current forms of DES.
Collapse
Affiliation(s)
- Cheesue Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Seul-Gee Lee
- Yonsei Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Songhyun Lim
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Mungyo Jung
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Sung Pil Kwon
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Jihye Hong
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Mikyung Kang
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Hee Su Sohn
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Seokhyeong Go
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Sangjun Moon
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Seung-Jun Lee
- Cardiology Division, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jung-Sun Kim
- Yonsei Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.,Cardiology Division, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea.,Interdisciplinary Program for Bioengineering, Seoul National University, Seoul 08826, Republic of Korea.,Institute of Chemical Processes, Institute of Engineering Research, and BioMAX, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
185
|
Vasquez-Rios G, Nadkarni GN. Proteomic Analyses Unveil Actionable Disease Pathways in COVID-19: A Step Toward Targeted Therapies. JACC Basic Transl Sci 2022; 7:442-444. [PMID: 35663631 PMCID: PMC9156428 DOI: 10.1016/j.jacbts.2022.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- George Vasquez-Rios
- Barbara T. Murphy Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Girish N. Nadkarni
- Barbara T. Murphy Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Division of Data-Driven and Digital Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Address for correspondence: Dr Girish N. Nadkarni, Barbara T. Murphy Division of Nephrology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1243, New York, New York 10029, USA.
| |
Collapse
|
186
|
Cortese K, Holland G, Möller L, Gagliani MC, Barisione E, Ball L, Pelosi P, Grillo F, Mastracci L, Fiocca R, Laue M. Ultrastructural examination of lung "cryobiopsies" from a series of fatal COVID-19 cases hardly revealed infected cells. Virchows Arch 2022; 480:967-977. [PMID: 35294603 PMCID: PMC8924574 DOI: 10.1007/s00428-022-03308-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/24/2022] [Accepted: 03/07/2022] [Indexed: 11/10/2022]
Abstract
Ultrastructural analysis of autopsy samples from COVID-19 patients usually suffers from significant structural impairment possibly caused by the rather long latency between death of the patient and an appropriate sample fixation. To improve structural preservation of the tissue, we obtained samples from ventilated patients using a trans-bronchial "cryobiopsy" within 30 min after their death and fixed them immediately for electron microscopy. Samples of six COVID-19 patients with a documented histopathology were systematically investigated by thin section electron microscopy. The different samples and areas inspected revealed the ultrastructural correlates of the different phases of diffuse alveolar damage, including detachment of the alveolar epithelium, hyperplasia of type 2 cells, exudates, and accumulation of extracellular material, such as the hyaline membranes and fibrin. Macrophages and neutrophilic granulocytes were regularly detected. Structural integrity of endothelium was intact in regions where the alveolar epithelium was already detached. Aggregates of erythrocytes, leukocytes with fibrin, and thrombocytes were not observed. Coronavirus particles were only found in and around very few cells in one of the six patient samples. The type and origin of these cells could not be assessed although the overall structural preservation of the samples allowed the identification of pulmonary cell types. Hence, the observed alveolar damage is not associated with virus presence or structural impairment due to ongoing replication at later stages of the disease in fatal cases, which implies that the lung damage in these patients is at least propagated by alternative mechanisms, perhaps, an inappropriate immune or stress response.
Collapse
Affiliation(s)
- Katia Cortese
- Cellular Electron Microscopy Laboratory, Department of Experimental Medicine (DIMES), Human Anatomy, University of Genoa, Genoa, Italy
| | - Gudrun Holland
- National Consultant Laboratory for Electron Microscopy of Infectious Pathogens, Centre for Biological Threats and Special Pathogens 4 (ZBS 4), Robert Koch Institute, Seestr. 10, 13353, Berlin, Germany
| | - Lars Möller
- National Consultant Laboratory for Electron Microscopy of Infectious Pathogens, Centre for Biological Threats and Special Pathogens 4 (ZBS 4), Robert Koch Institute, Seestr. 10, 13353, Berlin, Germany
| | - Maria Cristina Gagliani
- Cellular Electron Microscopy Laboratory, Department of Experimental Medicine (DIMES), Human Anatomy, University of Genoa, Genoa, Italy
| | - Emanuela Barisione
- Interventional Pneumology Unit, Polyclinic San Martino University Hospital, Genoa, Italy
- IRCCS Policlinico San Martino University Hospital, Genoa, Italy
| | - Lorenzo Ball
- IRCCS Policlinico San Martino University Hospital, Genoa, Italy
- Anesthesia and Intensive Care Unit, Department of Surgical Science and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Paolo Pelosi
- IRCCS Policlinico San Martino University Hospital, Genoa, Italy
- Anesthesia and Intensive Care Unit, Department of Surgical Science and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Federica Grillo
- IRCCS Policlinico San Martino University Hospital, Genoa, Italy
- Anatomic Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Luca Mastracci
- IRCCS Policlinico San Martino University Hospital, Genoa, Italy
- Anatomic Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Roberto Fiocca
- IRCCS Policlinico San Martino University Hospital, Genoa, Italy
| | - Michael Laue
- National Consultant Laboratory for Electron Microscopy of Infectious Pathogens, Centre for Biological Threats and Special Pathogens 4 (ZBS 4), Robert Koch Institute, Seestr. 10, 13353, Berlin, Germany.
| |
Collapse
|
187
|
Levy G, Guglielmelli P, Langmuir P, Constantinescu S. JAK inhibitors and COVID-19. J Immunother Cancer 2022; 10:jitc-2021-002838. [PMID: 35459733 PMCID: PMC9035837 DOI: 10.1136/jitc-2021-002838] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2022] [Indexed: 12/11/2022] Open
Abstract
During SARS-CoV-2 infection, the innate immune response can be inhibited or delayed, and the subsequent persistent viral replication can induce emergency signals that may culminate in a cytokine storm contributing to the severe evolution of COVID-19. Cytokines are key regulators of the immune response and virus clearance, and, as such, are linked to the—possibly altered—response to the SARS-CoV-2. They act via a family of more than 40 transmembrane receptors that are coupled to one or several of the 4 Janus kinases (JAKs) coded by the human genome, namely JAK1, JAK2, JAK3, and TYK2. Once activated, JAKs act on pathways for either survival, proliferation, differentiation, immune regulation or, in the case of type I interferons, antiviral and antiproliferative effects. Studies of graft-versus-host and systemic rheumatic diseases indicated that JAK inhibitors (JAKi) exert immunosuppressive effects that are non-redundant with those of corticotherapy. Therefore, they hold the potential to cut-off pathological reactions in COVID-19. Significant clinical experience already exists with several JAKi in COVID-19, such as baricitinib, ruxolitinib, tofacitinib, and nezulcitinib, which were suggested by a meta-analysis (Patoulias et al.) to exert a benefit in terms of risk reduction concerning major outcomes when added to standard of care in patients with COVID-19. Yet, only baricitinib is recommended in first line for severe COVID-19 treatment by the WHO, as it is the only JAKi that has proven efficient to reduce mortality in individual randomized clinical trials (RCT), especially the Adaptive COVID-19 Treatment Trial (ACTT-2) and COV-BARRIER phase 3 trials. As for secondary effects of JAKi treatment, the main caution with baricitinib consists in the induced immunosuppression as long-term side effects should not be an issue in patients treated for COVID-19. We discuss whether a class effect of JAKi may be emerging in COVID-19 treatment, although at the moment the convincing data are for baricitinib only. Given the key role of JAK1 in both type I IFN action and signaling by cytokines involved in pathogenic effects, establishing the precise timing of treatment will be very important in future trials, along with the control of viral replication by associating antiviral molecules.
Collapse
Affiliation(s)
- Gabriel Levy
- Signal Transduction and Molecular Hematology, Ludwig Institute for Cancer Research, Brussels, Belgium.,Signal Transduction on Molecular Hematology, de Duve Institute, Université Catholique de Louvain, Bruxelles, Belgium.,WELBIO, Walloon Excellence in Life Sciences and Biotechnology, Brussels, Belgium
| | - Paola Guglielmelli
- Department of Clinical and Experimental Medicine, University of Florence, Firenze, Italy.,Center of Research and Innovation for Myeloproliferative Neoplasms (CRIMM), Azienda Ospedaliero Universitaria Careggi, Firenze, Italy
| | - Peter Langmuir
- Oncology Targeted Therapeutics, Incyte Corp, Wilmington, Delaware, USA
| | - Stefan Constantinescu
- Signal Transduction and Molecular Hematology, Ludwig Institute for Cancer Research, Brussels, Belgium .,Signal Transduction on Molecular Hematology, de Duve Institute, Université Catholique de Louvain, Bruxelles, Belgium.,WELBIO, Walloon Excellence in Life Sciences and Biotechnology, Brussels, Belgium.,Nuffield Department of Medicine, Oxford University, Ludwig Institute for Cancer Research, Oxford, UK
| |
Collapse
|
188
|
Pawar A, Russo M, Rani I, Goswami K, Russo GL, Pal A. A critical evaluation of risk to reward ratio of quercetin supplementation for COVID-19 and associated comorbid conditions. Phytother Res 2022; 36:2394-2415. [PMID: 35393674 PMCID: PMC9111035 DOI: 10.1002/ptr.7461] [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: 06/18/2021] [Revised: 03/19/2022] [Accepted: 03/26/2022] [Indexed: 01/08/2023]
Abstract
The interim results of the large, multinational trials on coronavirus disease 2019 (COVID‐19) using a combination of antiviral drugs appear to have little to no effect on the 28‐day mortality or the in‐hospital course. Therefore, there is a still vivid interest in finding alternate re‐purposed drugs and nutrition supplements, which can halt or slow the disease severity. We review here the multiple preclinical studies, partially supported by clinical evidence showing the quercetin's possible therapeutic/prophylaxis efficacy against severe acute respiratory syndrome coronavirus (SARS‐CoV) as well as comorbidities like chronic obstructive pulmonary disease (COPD), diabetes mellitus, obesity, coagulopathy, and hypertension. Currently, 14 interventional clinical trials are underway assessing the efficacy of quercetin along with other antiviral drugs/nutritional supplements as prophylaxis/treatment option against COVID‐19. The present review is tempting to suggest that, based on circumstantial scientific evidence and preliminary clinical data, the flavonoid quercetin can ameliorate COVID‐19 infection and symptoms acting in concert on two parallel and independent paths: inhibiting key factors responsible for SARS‐CoV‐2 infections and mitigating the clinical manifestations of the disease in patients with comorbid conditions. Despite the broad therapeutic properties of quercetin, further high power randomized clinical trials are needed to firmly establish its clinical efficacy against COVID‐19.
Collapse
Affiliation(s)
- Anil Pawar
- Department of Zoology, DAV University, Jalandhar, India
| | - Maria Russo
- National Research Council, Institute of Food Sciences, Avellino, Italy
| | - Isha Rani
- Department of Biochemistry, Maharishi Markandeshwar Institute of Medical Sciences and Research (MMIMSR), Maharishi Markandeshwar University (MMU), Ambala, India
| | | | - Gian Luigi Russo
- National Research Council, Institute of Food Sciences, Avellino, Italy
| | - Amit Pal
- Department of Biochemistry, AIIMS, Kalyani, India
| |
Collapse
|
189
|
Lv N, Zhao Y, Liu X, Ye L, Liang Z, Kang Y, Dong Y, Wang W, Kolliputi N, Shi L. Dysfunctional telomeres through mitostress-induced cGAS/STING activation to aggravate immune senescence and viral pneumonia. Aging Cell 2022; 21:e13594. [PMID: 35313074 PMCID: PMC9009109 DOI: 10.1111/acel.13594] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/28/2022] [Accepted: 03/05/2022] [Indexed: 12/16/2022] Open
Abstract
Disproportionately high incidence and mortality of respiratory infection such as influenza A virus (IAV) and SARS-CoV-2 have been evidenced in the elderly, but the role and the mechanism of age-associated immune deregulation in disease exacerbation are not well defined. Using a late generation of mice deficient in telomerase RNA (Terc-/- ), we herein demonstrated that aged mice were exquisitely susceptible to respiratory viral infection, with excessive inflammation and increased mortality. Furthermore, we identified the cGAS/STING pathway, which was essentially induced by the leaked mitochondrial DNA, as a biologically relevant mechanism contributing to exaggerated inflammation in Terc-/- mice following viral infection. Innate immune cells, mainly, macrophages with shortened telomeres, exhibited hallmarks of cellular senescence, mitochondrial distress, and aberrant activation of STING and NLRP3 inflammasome pathways, which predisposed mice to severe viral pneumonia during commonly mild infections. Application of STING inhibitor and, more importantly, senolytic agent, reduced the burden of stressed macrophages, improved mitochondrial integrity, and suppressed STING activation, thereby conferring the protection for Terc-/- mice against respiratory infection. Together, the findings expand our understanding of innate immune senescence and reveal the potential of the senolytics as a promising treatment to alleviate the symptom of viral pneumonia, particularly for the older population.
Collapse
Affiliation(s)
- Nianyin Lv
- Department of Immunology and Medical Microbiology Nanjing University of Chinese Medicine Nanjing Jiangsu China
| | - Yufang Zhao
- Department of Basic Medicine Jiangxi Medical College Nanchang Jiangxi China
| | - Xiaoyi Liu
- Department of Immunology and Medical Microbiology Nanjing University of Chinese Medicine Nanjing Jiangsu China
- Department of Pharmacology and Physiology University of Rochester School of Medicine and Dentistry Rochester New York USA
| | - Lusha Ye
- Department of Immunology and Medical Microbiology Nanjing University of Chinese Medicine Nanjing Jiangsu China
| | - Zihao Liang
- Department of Immunology and Medical Microbiology Nanjing University of Chinese Medicine Nanjing Jiangsu China
| | - Yanhua Kang
- Department of Immunology and Medical Microbiology Nanjing University of Chinese Medicine Nanjing Jiangsu China
| | - Yeping Dong
- Department of Immunology and Medical Microbiology Nanjing University of Chinese Medicine Nanjing Jiangsu China
- Institute of Translational Medicine Zhejiang Shuren University Hangzhou Zhejiang China
| | - Wei Wang
- Department of Clinical Laboratory the Tongde Hospital Affiliated to Zhejiang TCM University Hangzhou Zhejiang China
| | - Narasaiah Kolliputi
- Division of Allergy and Immunology Department of Internal Medicine Morsani College of Medicine University of South Florida Tampa Florida USA
| | - Liyun Shi
- Department of Immunology and Medical Microbiology Nanjing University of Chinese Medicine Nanjing Jiangsu China
- Institute of Translational Medicine Zhejiang Shuren University Hangzhou Zhejiang China
| |
Collapse
|
190
|
Pence B, Zhang Y, Antwi I, Cory TJ. Senescent macrophages alter fibroblast fibrogenesis in response to SARS-CoV-2 infection. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2022; 1:37-42. [PMID: 36534613 PMCID: PMC9726213 DOI: 10.1515/nipt-2022-0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/28/2022] [Indexed: 06/17/2023]
Abstract
SARS-CoV-2 has, since its emergence in 2019, become a global pandemic. Disease outcomes are worsened in older patients who are infected. The causes for this is multifactorial, but one potential cause for this disparity is increased rates of cellular senescence in older individuals, particularly in immune cells. Cellular senescence, the accumulation of factors resulting in cell growth arrest and apoptosis resistance, increases as individuals age. In immune cells, senescence is associated with increased inflammation, and alterations in immune response. We utilized a co-culture system consisting of senescent or non-senescent macrophages directly cultured with fibroblasts, and infected with SARS-CoV-2. We assessed the expression of collagen and fibronectin, important molecules in the extracellular matrix, as well as a number of fibrogenic factors. We observed that infection with SARS-CoV-2 induced collagen production in co-cultures with senescent, but not non-senescent macrophages. Fibronectin expression was decreased in both co-culture conditions. While significant results were not observed, concentrations of other fibrogenic molecules were consistent with the collagen results. These data demonstrate that senescence in macrophages alters the production of fibrotic molecules from fibroblasts in a SARS-CoV-2 infection model. As collagen and fibronectin expression are generally directly correlated, this suggests that senescence dysregulates fibrogenesis in response to infection with SARS-CoV-2. There is a need to further investigate the mechanisms for these changes.
Collapse
Affiliation(s)
- Brandt Pence
- University of Memphis College of Health Sciences, Memphis, TN, USA
| | - Yufeng Zhang
- University of Memphis College of Health Sciences, Memphis, TN, USA
| | - Ivy Antwi
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center College of Pharmacy, Memphis, TN, USA
| | - Theodore James Cory
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center College of Pharmacy, Memphis, TN, USA
| |
Collapse
|
191
|
Hu L, Li H, Zi M, Li W, Liu J, Yang Y, Zhou D, Kong QP, Zhang Y, He Y. Why Senescent Cells Are Resistant to Apoptosis: An Insight for Senolytic Development. Front Cell Dev Biol 2022; 10:822816. [PMID: 35252191 PMCID: PMC8890612 DOI: 10.3389/fcell.2022.822816] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 01/26/2022] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence is a process that leads to a state of irreversible cell growth arrest induced by a variety of intrinsic and extrinsic stresses. Senescent cells (SnCs) accumulate with age and have been implicated in various age-related diseases in part via expressing the senescence-associated secretory phenotype. Elimination of SnCs has the potential to delay aging, treat age-related diseases and extend healthspan. However, once cells becoming senescent, they are more resistant to apoptotic stimuli. Senolytics can selectively eliminate SnCs by targeting the SnC anti-apoptotic pathways (SCAPs). They have been developed as a novel pharmacological strategy to treat various age-related diseases. However, the heterogeneity of the SnCs indicates that SnCs depend on different proteins or pathways for their survival. Thus, a better understanding of the underlying mechanisms for apoptotic resistance of SnCs will provide new molecular targets for the development of cell-specific or broad-spectrum therapeutics to clear SnCs. In this review, we discussed the latest research progresses and challenge in senolytic development, described the significance of regulation of senescence and apoptosis in aging, and systematically summarized the SCAPs involved in the apoptotic resistance in SnCs.
Collapse
Affiliation(s)
- Li Hu
- Department of Geriatrics, The Second Affiliated Hospital of Hainan Medical University, Haikou, China.,State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.,College of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China
| | - Huiqin Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Meiting Zi
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Wen Li
- Department of Endocrinology, The Third People's Hospital of Yunnan Province, Kunming, China
| | - Jing Liu
- Lab of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, China
| | - Yang Yang
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Daohong Zhou
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, United States
| | - Qing-Peng Kong
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yunxia Zhang
- Department of Geriatrics, The Second Affiliated Hospital of Hainan Medical University, Haikou, China.,College of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China
| | - Yonghan He
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
192
|
Hong X, Wang L, Zhang K, Liu J, Liu JP. Molecular Mechanisms of Alveolar Epithelial Stem Cell Senescence and Senescence-Associated Differentiation Disorders in Pulmonary Fibrosis. Cells 2022; 11:877. [PMID: 35269498 PMCID: PMC8909789 DOI: 10.3390/cells11050877] [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: 02/10/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/04/2023] Open
Abstract
Pulmonary senescence is accelerated by unresolved DNA damage response, underpinning susceptibility to pulmonary fibrosis. Recently it was reported that the SARS-Cov-2 viral infection induces acute pulmonary epithelial senescence followed by fibrosis, although the mechanism remains unclear. Here, we examine roles of alveolar epithelial stem cell senescence and senescence-associated differentiation disorders in pulmonary fibrosis, exploring the mechanisms mediating and preventing pulmonary fibrogenic crisis. Notably, the TGF-β signalling pathway mediates alveolar epithelial stem cell senescence by mechanisms involving suppression of the telomerase reverse transcriptase gene in pulmonary fibrosis. Alternatively, telomere uncapping caused by stress-induced telomeric shelterin protein TPP1 degradation mediates DNA damage response, pulmonary senescence and fibrosis. However, targeted intervention of cellular senescence disrupts pulmonary remodelling and fibrosis by clearing senescent cells using senolytics or preventing senescence using telomere dysfunction inhibitor (TELODIN). Studies indicate that the development of senescence-associated differentiation disorders is reprogrammable and reversible by inhibiting stem cell replicative senescence in pulmonary fibrosis, providing a framework for targeted intervention of the molecular mechanisms of alveolar stem cell senescence and pulmonary fibrosis. Abbreviations: DPS, developmental programmed senescence; IPF, idiopathic pulmonary fibrosis; OIS, oncogene-induced replicative senescence; SADD, senescence-associated differentiation disorder; SALI, senescence-associated low-grade inflammation; SIPS, stress-induced premature senescence; TERC, telomerase RNA component; TERT, telomerase reverse transcriptase; TIFs, telomere dysfunction-induced foci; TIS, therapy-induced senescence; VIS, virus-induced senescence.
Collapse
Affiliation(s)
- Xiaojing Hong
- Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou 311121, China; (X.H.); (L.W.); (K.Z.); (J.L.)
| | - Lihui Wang
- Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou 311121, China; (X.H.); (L.W.); (K.Z.); (J.L.)
| | - Kexiong Zhang
- Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou 311121, China; (X.H.); (L.W.); (K.Z.); (J.L.)
| | - Jun Liu
- Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou 311121, China; (X.H.); (L.W.); (K.Z.); (J.L.)
| | - Jun-Ping Liu
- Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou 311121, China; (X.H.); (L.W.); (K.Z.); (J.L.)
- Department of Immunology and Pathology, Monash University Faculty of Medicine, Prahran, VIC 3181, Australia
- Hudson Institute of Medical Research, Monash University Department of Molecular and Translational Science, Clayton, VIC 3168, Australia
| |
Collapse
|
193
|
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
|
194
|
Park J, Foox J, Hether T, Danko DC, Warren S, Kim Y, Reeves J, Butler DJ, Mozsary C, Rosiene J, Shaiber A, Afshin EE, MacKay M, Rendeiro AF, Bram Y, Chandar V, Geiger H, Craney A, Velu P, Melnick AM, Hajirasouliha I, Beheshti A, Taylor D, Saravia-Butler A, Singh U, Wurtele ES, Schisler J, Fennessey S, Corvelo A, Zody MC, Germer S, Salvatore S, Levy S, Wu S, Tatonetti NP, Shapira S, Salvatore M, Westblade LF, Cushing M, Rennert H, Kriegel AJ, Elemento O, Imielinski M, Rice CM, Borczuk AC, Meydan C, Schwartz RE, Mason CE. System-wide transcriptome damage and tissue identity loss in COVID-19 patients. Cell Rep Med 2022; 3:100522. [PMID: 35233546 PMCID: PMC8784611 DOI: 10.1016/j.xcrm.2022.100522] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 12/22/2021] [Accepted: 01/16/2022] [Indexed: 01/07/2023]
Abstract
The molecular mechanisms underlying the clinical manifestations of coronavirus disease 2019 (COVID-19), and what distinguishes them from common seasonal influenza virus and other lung injury states such as acute respiratory distress syndrome, remain poorly understood. To address these challenges, we combine transcriptional profiling of 646 clinical nasopharyngeal swabs and 39 patient autopsy tissues to define body-wide transcriptome changes in response to COVID-19. We then match these data with spatial protein and expression profiling across 357 tissue sections from 16 representative patient lung samples and identify tissue-compartment-specific damage wrought by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, evident as a function of varying viral loads during the clinical course of infection and tissue-type-specific expression states. Overall, our findings reveal a systemic disruption of canonical cellular and transcriptional pathways across all tissues, which can inform subsequent studies to combat the mortality of COVID-19 and to better understand the molecular dynamics of lethal SARS-CoV-2 and other respiratory infections.
Collapse
Affiliation(s)
- Jiwoon Park
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Jonathan Foox
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | | | - David C. Danko
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- Tri-Institutional Computational Biology & Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | | | - Youngmi Kim
- NanoString Technologies, Inc., Seattle, WA, USA
| | | | - Daniel J. Butler
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - Christopher Mozsary
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - Joel Rosiene
- New York Genome Center, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Alon Shaiber
- New York Genome Center, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Evan E. Afshin
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Matthew MacKay
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - André F. Rendeiro
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- Englander Institute for Precision Medicine and the Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Yaron Bram
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | | | - Arryn Craney
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Priya Velu
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Ari M. Melnick
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Iman Hajirasouliha
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- Englander Institute for Precision Medicine and the Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Afshin Beheshti
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Deanne Taylor
- Department of Biomedical and Health Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amanda Saravia-Butler
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, USA
- Logyx, LLC, Mountain View, CA, USA
| | - Urminder Singh
- Bioinformatics and Computational Biology Program, Center for Metabolic Biology, Department of Genetics, Development and Cell Biology Iowa State University, Ames, IA, USA
| | - Eve Syrkin Wurtele
- Bioinformatics and Computational Biology Program, Center for Metabolic Biology, Department of Genetics, Development and Cell Biology Iowa State University, Ames, IA, USA
| | - Jonathan Schisler
- McAllister Heart Institute at The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, and Department of Pathology and Lab Medicine at The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | | | | | | - Steven Salvatore
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Shawn Levy
- HudsonAlpha Discovery Institute, Huntsville, AL, USA
| | - Shixiu Wu
- Hangzhou Cancer Institute, Hangzhou Cancer Hospital, Hangzhou, China
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, China
| | - Nicholas P. Tatonetti
- Department of Biomedical Informatics, Department of Systems Biology, Department of Medicine, Institute for Genomic Medicine, Columbia University, New York, NY, USA
| | - Sagi Shapira
- Department of Biomedical Informatics, Department of Systems Biology, Department of Medicine, Institute for Genomic Medicine, Columbia University, New York, NY, USA
| | - Mirella Salvatore
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Lars F. Westblade
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Melissa Cushing
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Hanna Rennert
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Alison J. Kriegel
- Department of Physiology, Cardiovascular Center, Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Olivier Elemento
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
- Tri-Institutional Computational Biology & Medicine Program, Weill Cornell Medicine, New York, NY, USA
- Englander Institute for Precision Medicine and the Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Marcin Imielinski
- New York Genome Center, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Charles M. Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Alain C. Borczuk
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Cem Meydan
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Robert E. Schwartz
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Christopher E. Mason
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
195
|
Novel SARS-CoV-2 therapeutic targets: RNA proofreading complex and virus-induced senescence. Cell Death Differ 2022; 29:263-265. [PMID: 35039647 PMCID: PMC8761837 DOI: 10.1038/s41418-021-00909-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/17/2021] [Accepted: 11/17/2021] [Indexed: 01/30/2023] Open
|
196
|
Bogdanov VY, Khirmanov VN. SARS-CoV-2, platelets, and endothelium: coexistence in space and time, or a pernicious ménage à trois? VASCULAR BIOLOGY (BRISTOL, ENGLAND) 2022; 4:R35-R43. [PMID: 35949299 PMCID: PMC9354055 DOI: 10.1530/vb-22-0004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/08/2022] [Indexed: 11/30/2022]
Abstract
As we enter year 3 of SARS-CoV-2 pandemic, long-term consequences of COVID-19 have become a major public health issue worldwide; however, the molecular and cellular underpinnings of 'long COVID' remain very poorly understood. A paradigm has recently emerged that thrombo-inflammatory consequences of SARS-CoV-2's impact on endothelial cells and platelets likely play a significant role in the development of chronic symptomatology associated with COVID-19. In this brief overview, we discuss the recent findings pertaining to the detection of SARS-CoV-2 virions in vascular cell subtypes, the contribution of the coagulation system to the development of 'long COVID', and the potential role of stem/progenitor cells in the viral and thrombotic dissemination in this disorder.
Collapse
Affiliation(s)
- Vladimir Y Bogdanov
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Vladimir N Khirmanov
- Department of Cardiovascular Medicine, Nikiforov’s All-Russian Center for Emergency and Radiation Medicine, Saint Petersburg, Russia
| |
Collapse
|
197
|
Tsuji S, Minami S, Hashimoto R, Konishi Y, Suzuki T, Kondo T, Sasai M, Torii S, Ono C, Shichinohe S, Sato S, Wakita M, Okumura S, Nakano S, Matsudaira T, Matsumoto T, Kawamoto S, Yamamoto M, Watanabe T, Matsuura Y, Takayama K, Kobayashi T, Okamoto T, Hara E. SARS-CoV-2 infection triggers paracrine senescence and leads to a sustained senescence-associated inflammatory response. NATURE AGING 2022; 2:115-124. [PMID: 37117754 PMCID: PMC10154207 DOI: 10.1038/s43587-022-00170-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 01/06/2022] [Indexed: 04/30/2023]
Abstract
Reports of post-acute COVID-19 syndrome, in which the inflammatory response persists even after SARS-CoV-2 has disappeared, are increasing1, but the underlying mechanisms of post-acute COVID-19 syndrome remain unknown. Here, we show that SARS-CoV-2-infected cells trigger senescence-like cell-cycle arrest2,3 in neighboring uninfected cells in a paracrine manner via virus-induced cytokine production. In cultured human cells or bronchial organoids, these SASR-CoV-2 infection-induced senescent cells express high levels of a series of inflammatory factors known as senescence-associated secretory phenotypes (SASPs)4 in a sustained manner, even after SARS-CoV-2 is no longer detectable. We also show that the expression of the senescence marker CDKN2A (refs. 5,6) and various SASP factor4 genes is increased in the pulmonary cells of patients with severe post-acute COVID-19 syndrome. Furthermore, we find that mice exposed to a mouse-adapted strain of SARS-CoV-2 exhibit prolonged signs of cellular senescence and SASP in the lung at 14 days after infection when the virus was undetectable, which could be substantially reduced by the administration of senolytic drugs7. The sustained infection-induced paracrine senescence described here may be involved in the long-term inflammation caused by SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Shunya Tsuji
- Department of Molecular Microbiology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Shohei Minami
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Rina Hashimoto
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Yusuke Konishi
- Department of Molecular Microbiology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Tatsuya Suzuki
- Division of Infectious Diseases, Institute for Advanced Co-Creation Studies, Osaka University, Suita, Japan
| | - Tamae Kondo
- Department of Molecular Microbiology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Miwa Sasai
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Shiho Torii
- Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Chikako Ono
- Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Shintaro Shichinohe
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Shintaro Sato
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masahiro Wakita
- Department of Molecular Microbiology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Shintaro Okumura
- Department of Molecular Microbiology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Sosuke Nakano
- Department of Molecular Microbiology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Tatsuyuki Matsudaira
- Department of Molecular Microbiology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Tomonori Matsumoto
- Department of Molecular Microbiology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Shimpei Kawamoto
- Department of Molecular Microbiology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Immunology Frontier Research Center, Osaka University, Suita, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Japan
| | - Tokiko Watanabe
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Japan
| | - Yoshiharu Matsuura
- Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Japan
| | - Kazuo Takayama
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Takeshi Kobayashi
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Japan
| | - Toru Okamoto
- Division of Infectious Diseases, Institute for Advanced Co-Creation Studies, Osaka University, Suita, Japan
| | - Eiji Hara
- Department of Molecular Microbiology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan.
- Immunology Frontier Research Center, Osaka University, Suita, Japan.
- Center for Infectious Disease Education and Research, Osaka University, Suita, Japan.
| |
Collapse
|
198
|
Jakob F, Herrmann M. Gestörte Geweberegeneration durch entzündliche
Prozesse bei Alterung, Seneszenz und degenerativen Erkrankungen –
Interaktionen mit dem COVID-19-induzierten Zytokin-Sturm des angeborenen
Immunsystems. OSTEOLOGIE 2022; 31:7-16. [DOI: 10.1055/a-1725-8211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
ZusammenfassungEntzündung ist Bestandteil einer jeglichen Geweberegeneration. Verletzung
und Schädigung von Geweben - inklusive exogene virale und bakterielle
Infektionen - induzieren eine frühe pro-inflammatorische Phase, die
durch Aktivierung von residenten und aus dem peripheren Blut und Knochenmark
rekrutierten Zellen des angeborenen Immunsystems weiter propagiert wird. Diese
Phase dient auch dem Clearing der Umgebung von vorgeschädigten Zellen
und cell debris. Um eine erfolgreiche Geweberegeneration zu erreichen ist es
essentiell, die Auflösung der Entzündung durch zeitgerechte
Einleitung einer anti-inflammatorischen Phase der Geweberegeneration zu
ermöglichen. Dieser Phase kann dann die Gewebeneubildung folgen, am
Beispiel der Frakturheilung als „Modeling“ bezeichnet. Das
schnell gebildete neue Gewebe wird in der letzten Phase der Regeneration an die
physikalischen Bedingungen im Gewebeverband angepasst, bei der Frakturheilung
„Remodeling“ genannt. Kann die zeitgerechte Auflösung
der Entzündung nicht erfolgen, verhindert die persistierende
Entzündung das Eintreten in die Phase der Gewebeneubildung und damit die
erfolgreiche Regeneration. Es erfolgt dann entweder als
„Notlösung“ eine Narbenheilung oder im Falle weiter
ausufernder Entzündung eine Zerstörung des Gewebes. Die mit dem
Alter sich verschlechternde Regenerationskapazität vieler Gewebe
inklusive Knochen, Muskel und Sehnen ist unter anderem eine Folge der
subklinischen chronischen Entzündung von Geweben, die Alterung
(„Inflammaging“) propagiert. Die Entzündung im
Mikromillieu involviert neben den gewebe-typischen Zellen und deren adulten
Progenitoren auch die Zellen des gewebeeigenen (residenten) angeborenen
Immunsystems, allen voran Makrophagen. Auch diese unterliegen
Alters-assoziierten Veränderungen wie Zellalterung und eine gesteigerte
Suszeptibilität für pro-inflammatorische Überreaktionen.
Chronische Inflammation mündet letztlich in die zelluläre
Seneszenz, die begleitet ist von einem Seneszenz-assoziierten sekretorischen
Phänotyp (SASP) mit hoher Produktion von Interleukinen 1, 6, 8, und
anderen Zytokinen. Solange solche Zellen nicht in den geregelten Zelltod gehen,
unterhalten sie die chronische Entzündung und damit die Voraussetzungen
für insuffiziente Geweberegeneration. Eine COVID-19 Infektion triggert
und unterhält identische inflammatorische Mechanismen und induziert
zusätzlich Seneszenz. Dies kann in der Summe zu einem Zytokin-Sturm
führen, der in einem circulus vitiosus eine zerstörerische
Hyperinflammation unterhält und der umso schwerwiegender
ausfällt je höher die Vorlast an seneszenten Zellen ist, wie das
in den COVID-Risikopopulationen der Fall ist. Deren Zusammensetzung
überlappt sehr stark mit unseren Risikopopulationen für
degenerative muskuloskelettale Erkrankungen wie Osteoporose und Sarkopenie.
Collapse
Affiliation(s)
- Franz Jakob
- Bernhard-Heine-Centrum für Bewegungsforschung,
Julius-Maximilians-Universität Würzburg, Wurzburg,
Germany
| | - Marietta Herrmann
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University
Hospital Wurzburg, Wurzburg, Germany
| |
Collapse
|
199
|
Teissier T, Boulanger E, Cox LS. Interconnections between Inflammageing and Immunosenescence during Ageing. Cells 2022; 11:359. [PMID: 35159168 PMCID: PMC8834134 DOI: 10.3390/cells11030359] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/13/2022] [Accepted: 01/15/2022] [Indexed: 02/04/2023] Open
Abstract
Acute inflammation is a physiological response to injury or infection, with a cascade of steps that ultimately lead to the recruitment of immune cells to clear invading pathogens and heal wounds. However, chronic inflammation arising from the continued presence of the initial trigger, or the dysfunction of signalling and/or effector pathways, is harmful to health. While successful ageing in older adults, including centenarians, is associated with low levels of inflammation, elevated inflammation increases the risk of poor health and death. Hence inflammation has been described as one of seven pillars of ageing. Age-associated sterile, chronic, and low-grade inflammation is commonly termed inflammageing-it is not simply a consequence of increasing chronological age, but is also a marker of biological ageing, multimorbidity, and mortality risk. While inflammageing was initially thought to be caused by "continuous antigenic load and stress", reports from the last two decades describe a much more complex phenomenon also involving cellular senescence and the ageing of the immune system. In this review, we explore some of the main sources and consequences of inflammageing in the context of immunosenescence and highlight potential interventions. In particular, we assess the contribution of cellular senescence to age-associated inflammation, identify patterns of pro- and anti-inflammatory markers characteristic of inflammageing, describe alterations in the ageing immune system that lead to elevated inflammation, and finally assess the ways that diet, exercise, and pharmacological interventions can reduce inflammageing and thus, improve later life health.
Collapse
Affiliation(s)
- Thibault Teissier
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK;
| | - Eric Boulanger
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167—RID-AGE—Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, F-59000 Lille, France;
| | - Lynne S. Cox
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK;
| |
Collapse
|
200
|
Promising Effects of 3-Month Period of Quercetin Phytosome ® Supplementation in the Prevention of Symptomatic COVID-19 Disease in Healthcare Workers: A Pilot Study. Life (Basel) 2022; 12:life12010066. [PMID: 35054459 PMCID: PMC8780248 DOI: 10.3390/life12010066] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/23/2021] [Accepted: 01/02/2022] [Indexed: 12/13/2022] Open
Abstract
Quercetin, for its crucial properties, fulfills the need for a multifactor action that is useful for the potential counterbalance of a COVID-19 infection. Given this background, the aim of the study was to evaluate the potential effect of 3 months’ supplementation with Quercetin Phytosome® (250 mg twice a day) as prevention against symptomatic COVID-19. In total, 120 subjects were enrolled (males, 63; females, 57; age 49 ± 12), with 60 in the supplementation group and 60 in the placebo group. No significant differences were detected between groups in terms of gender, smoking, and chronic disease. Subjects underwent rapid COVID-19 diagnostic tests every 3 weeks. During our study, 5 subjects had COVID-19, 1 out of 60 subjects in the quercetin group and 4 out of 60 in the control group. Complete clinical remission was recorded at 7 and 15 days in the quercetin and placebo groups, respectively. Analysis showed that, at 5 months, the COVID free survival function (risk of infection) was 99.8% in subjects under quercetin supplementation and 96.5% in control group. As shown by the value of EXP(B), those who had taken the supplement had a protection factor of 14% more to not contract the COVID-19 infection than that of those who had taken a placebo. Obtained results are encouraging, but further studies are required to add quercetin as regular prophylaxis.
Collapse
|