1
|
Yu Y, Lin K, Wu H, Hu M, Yang X, Wang J, Grillari J, Chen J. Targeting senescent cells in aging and COVID-19: from cellular mechanisms to therapeutic opportunities. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:20. [PMID: 39358480 PMCID: PMC11447201 DOI: 10.1186/s13619-024-00201-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/10/2024] [Indexed: 10/04/2024]
Abstract
The COVID-19 pandemic has caused a global health crisis and significant social economic burden. While most individuals experience mild or non-specific symptoms, elderly individuals are at a higher risk of developing severe symptoms and life-threatening complications. Exploring the key factors associated with clinical severity highlights that key characteristics of aging, such as cellular senescence, immune dysregulation, metabolic alterations, and impaired regenerative potential, contribute to disruption of tissue homeostasis of the lung and worse clinical outcome. Senolytic and senomorphic drugs, which are anti-aging treatments designed to eliminate senescent cells or decrease the associated phenotypes, have shown promise in alleviating age-related dysfunctions and offer a novel approach to treating diseases that share certain aspects of underlying mechanisms with aging, including COVID-19. This review summarizes the current understanding of aging in COVID-19 progression, and highlights recent findings on anti-aging drugs that could be repurposed for COVID-19 treatment to complement existing therapies.
Collapse
Affiliation(s)
- Yuan Yu
- Center for Cell Lineage and Atlas, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kaixuan Lin
- Center for Cell Lineage and Atlas, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| | - Haoyu Wu
- Center for Cell Lineage and Atlas, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Mingli Hu
- Center for Cell Lineage and Atlas, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xuejie Yang
- Center for Cell Lineage and Atlas, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Jie Wang
- Center for Cell Lineage and Atlas, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Johannes Grillari
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Institute of Molecular Biotechnology, BOKU University, Vienna, Austria
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation With AUVA, 1200, Vienna, Austria
| | - Jiekai Chen
- Center for Cell Lineage and Atlas, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| |
Collapse
|
2
|
Mahmutović-Dizdarević I, Mesic A, Jerković-Mujkić A, Žujo B, Avdić M, Hukić M, Omeragić E, Osmanović A, Špirtović-Halilović S, Ahmetovski S, Mujkanović S, Pramenković E, Salihović M. Biological potential, chemical profiling, and molecular docking study of Morus alba L. extracts. Fitoterapia 2024; 177:106114. [PMID: 38971331 DOI: 10.1016/j.fitote.2024.106114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Morus alba L. is a plant with a long history of dietary and medicinal uses. We hypothesized that M. alba possesses a significant biological potential. In that sense, we aimed to generate the chemical, antimicrobial, toxicological, and molecular profile of M. alba leaf and fruit extracts. Our results showed that extracts were rich in vitamin C, phenols, and flavonoids, with quercetin and pterostilbene concentrated in the leaf, while fisetin, hesperidin, resveratrol, and luteolin were detected in fruit. Extracts exhibited antimicrobial activity against all tested bacteria, including multidrug-resistant strains. The widest inhibition zones were in Staphylococcus aureus ATCC 33591. The values of the minimum inhibitory concentration ranged from 15.62 μg/ml in Enterococcus faecalis to 500 μg/ml in several bacteria. Minimum bactericidal concentration ranged from 31.25 μg/ml to 1000 μg/ml. Extracts impacted the biofilm formation in a concentration-dependent and species-specific manner. A significant difference in the frequency of nucleoplasmic bridges between the methanolic extract of fruit (0.5 μg/ml, 1 μg/ml, 2 μg/ml), as well as for the frequency of micronuclei between ethanolic extract of leaf (2 μg/ml) and the control group was observed. Molecular docking suggested that hesperidin possesses the highest binding affinity for multidrug efflux transporter AcrB and acyl-PBP2a from MRSA, as well as for the SARS-CoV-2 Mpro. This study, by complementing previous research in this field, gives new insights that could be of great value in obtaining a more comprehensive picture of the Morus alba L. bioactive potential, chemical composition, antimicrobial and toxicological features, as well as molecular profile.
Collapse
Affiliation(s)
- Irma Mahmutović-Dizdarević
- University of Sarajevo-Faculty of Science, Department of Biology, Zmaja od Bosne 33-35, 71000 Sarajevo, Bosnia and Herzegovina.
| | - Aner Mesic
- University of Sarajevo-Faculty of Science, Department of Biology, Zmaja od Bosne 33-35, 71000 Sarajevo, Bosnia and Herzegovina
| | - Anesa Jerković-Mujkić
- University of Sarajevo-Faculty of Science, Department of Biology, Zmaja od Bosne 33-35, 71000 Sarajevo, Bosnia and Herzegovina
| | - Belma Žujo
- University of Sarajevo-Faculty of Science, Department of Biology, Zmaja od Bosne 33-35, 71000 Sarajevo, Bosnia and Herzegovina
| | - Monia Avdić
- International Burch University, Faculty of Engineering, Natural and Medical Sciences, Department of Genetics and Bioengineering, Francuske revolucije bb, 71210 Ilidža, Bosnia and Herzegovina; Academy of Sciences and Arts of Bosnia and Herzegovina, Center for Disease Control and Geohealth Studies, Bistrik 7, 71000 Sarajevo, Bosnia and Herzegovina
| | - Mirsada Hukić
- Academy of Sciences and Arts of Bosnia and Herzegovina, Center for Disease Control and Geohealth Studies, Bistrik 7, 71000 Sarajevo, Bosnia and Herzegovina; Institute for Biomedical Diagnostics and Research Nalaz, Čekaluša 69, 71000 Sarajevo, Bosnia and Herzegovina
| | - Elma Omeragić
- University of Sarajevo-Faculty of Pharmacy, Zmaja od Bosne 8, 71000 Sarajevo, Bosnia and Herzegovina
| | - Amar Osmanović
- University of Sarajevo-Faculty of Pharmacy, Zmaja od Bosne 8, 71000 Sarajevo, Bosnia and Herzegovina
| | - Selma Špirtović-Halilović
- University of Sarajevo-Faculty of Pharmacy, Zmaja od Bosne 8, 71000 Sarajevo, Bosnia and Herzegovina
| | - Sarah Ahmetovski
- University of Sarajevo-Faculty of Science, Department of Biology, Zmaja od Bosne 33-35, 71000 Sarajevo, Bosnia and Herzegovina
| | - Samra Mujkanović
- University of Sarajevo-Faculty of Science, Department of Biology, Zmaja od Bosne 33-35, 71000 Sarajevo, Bosnia and Herzegovina
| | - Emina Pramenković
- International Burch University, Faculty of Engineering, Natural and Medical Sciences, Department of Genetics and Bioengineering, Francuske revolucije bb, 71210 Ilidža, Bosnia and Herzegovina
| | - Mirsada Salihović
- University of Sarajevo-Faculty of Pharmacy, Zmaja od Bosne 8, 71000 Sarajevo, Bosnia and Herzegovina
| |
Collapse
|
3
|
Merigo F, Lagni A, Boschi F, Bernardi P, Conti A, Plebani R, Romano M, Sorio C, Lotti V, Sbarbati A. Loss of CFTR Reverses Senescence Hallmarks in SARS-CoV-2 Infected Bronchial Epithelial Cells. Int J Mol Sci 2024; 25:6185. [PMID: 38892373 PMCID: PMC11172982 DOI: 10.3390/ijms25116185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/28/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
SARS-CoV-2 infection has been recently shown to induce cellular senescence in vivo. A senescence-like phenotype has been reported in cystic fibrosis (CF) cellular models. Since the previously published data highlighted a low impact of SARS-CoV-2 on CFTR-defective cells, here we aimed to investigate the senescence hallmarks in SARS-CoV-2 infection in the context of a loss of CFTR expression/function. We infected WT and CFTR KO 16HBE14o-cells with SARS-CoV-2 and analyzed both the p21 and Ki67 expression using immunohistochemistry and viral and p21 gene expression using real-time PCR. Prior to SARS-CoV-2 infection, CFTR KO cells displayed a higher p21 and lower Ki67 expression than WT cells. We detected lipid accumulation in CFTR KO cells, identified as lipolysosomes and residual bodies at the subcellular/ultrastructure level. After SARS-CoV-2 infection, the situation reversed, with low p21 and high Ki67 expression, as well as reduced viral gene expression in CFTR KO cells. Thus, the activation of cellular senescence pathways in CFTR-defective cells was reversed by SARS-CoV-2 infection while they were activated in CFTR WT cells. These data uncover a different response of CF and non-CF bronchial epithelial cell models to SARS-CoV-2 infection and contribute to uncovering the molecular mechanisms behind the reduced clinical impact of COVID-19 in CF patients.
Collapse
Affiliation(s)
- Flavia Merigo
- Anatomy and Histology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (F.M.); (P.B.); (A.C.); (A.S.)
| | - Anna Lagni
- Microbiology Section, Department of Diagnostic and Public Health, University of Verona, 37134 Verona, Italy;
| | - Federico Boschi
- Department of Engineering for Innovation Medicine, University of Verona, 37134 Verona, Italy;
| | - Paolo Bernardi
- Anatomy and Histology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (F.M.); (P.B.); (A.C.); (A.S.)
| | - Anita Conti
- Anatomy and Histology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (F.M.); (P.B.); (A.C.); (A.S.)
| | - Roberto Plebani
- Laboratory of Molecular Medicine, Center for Advanced Studies and Technology (CAST), Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (R.P.); (M.R.)
| | - Mario Romano
- Laboratory of Molecular Medicine, Center for Advanced Studies and Technology (CAST), Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (R.P.); (M.R.)
| | - Claudio Sorio
- General Pathology Section, Department of Medicine, University of Verona, 37134 Verona, Italy;
| | - Virginia Lotti
- Microbiology Section, Department of Diagnostic and Public Health, University of Verona, 37134 Verona, Italy;
| | - Andrea Sbarbati
- Anatomy and Histology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (F.M.); (P.B.); (A.C.); (A.S.)
| |
Collapse
|
4
|
Fang W, Du J, Nie M, Wang X. Recent advances in flavonoid compounds for the treatment of prostate cancer. Mol Biol Rep 2024; 51:653. [PMID: 38734766 DOI: 10.1007/s11033-024-09567-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/18/2024] [Indexed: 05/13/2024]
Abstract
Prostate cancer is a malignant epithelial tumor of the prostate gland and is the most common malignant tumor of the male genitourinary system. Pharmacological therapies, including chemotherapy and androgen deprivation therapy, play a key role in the treatment of prostate cancer. However, drug resistance and side effects limit the use of these drugs and so there is a need for new drug therapies for prostate cancer patients. Flavonoids, with their wide range of sources and diverse biological activities, have attracted much attention in the field of anti-tumor drug screening. In 2016, at least 58 flavonoids were reported to have anti-prostate cancer activity. In recent years, six additional flavonoid compounds have been found to have anti-prostate cancer potential. In this review, we have collected a large amount of evidence on the anti-prostate cancer effects of these six flavonoids, including a large number of cellular experiments and a small number of preclinical animal experiments. In addition, we predicted their drug-forming properties using Schrödinger's QikProp software and ADMETlab due to the lack of in vivo pharmacokinetic data for the six compounds. In conclusion, this review has fully confirmed the anti-prostate cancer effects of these six flavonoids, summarized their mechanisms of action and predicted their druggability. It provides a reference for the further development of these compounds into anti-prostate cancer drugs.
Collapse
Affiliation(s)
- Wenxuan Fang
- Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Guangxi University of Chinese Medicine, 13 Wuhe Road, Qingxiu District, Nanning, 530200, China
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning, 530200, China
| | - Junfang Du
- School of Yao Medicine, Guangxi University of Chinese Medicine, 179 Mingxiudong Road, Xixiangtang District, Nanning, 530001, China
| | - Mingyi Nie
- Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Guangxi University of Chinese Medicine, 13 Wuhe Road, Qingxiu District, Nanning, 530200, China
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning, 530200, China
| | - Xueni Wang
- Guangxi Zhuang Yao Medicine Center of Engineering and Technology, Guangxi University of Chinese Medicine, 13 Wuhe Road, Qingxiu District, Nanning, 530200, China.
| |
Collapse
|
5
|
Jacobson RM, Pignolo RJ, Lazaridis KN. Clinical Trials for Special Populations: Children, Older Adults, and Rare Diseases. Mayo Clin Proc 2024; 99:318-335. [PMID: 38309939 PMCID: PMC10842263 DOI: 10.1016/j.mayocp.2023.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/02/2023] [Accepted: 03/13/2023] [Indexed: 02/05/2024]
Abstract
Research cannot maximize population health unless it improves health for all members of the public, including special populations such as children, older adults, and people living with rare diseases. Each of these categories require special considerations when planning and performing clinical trials, and common threads of ethical conduct of research in vulnerable populations appear throughout. In this review, definitions of each of the three categories of special population (children, older adults, and rare diseases) are discussed in terms of US research regulations, the unique challenges to conducting clinical trials for these special populations, critical ethical issues, and opportunities for innovative ways to design and operationalize clinical trials in special populations. Additional critical attention is focused on factors that influence the generalizability of study results to reduce health disparities, as well as the importance of community engagement and advocacy groups that can help to educate potential trial participants of the benefits of clinical trial participation.
Collapse
Affiliation(s)
- Robert M Jacobson
- Department of Pediatric and Adolescent Medicine and Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA.
| | - Robert J Pignolo
- Department of Medicine and Divisions of Hospital Internal Medicine and Endocrinology, Home of Medical Excellence in Geriatric Medicine and Gerontology, Department of Physiology and Biomedical Engineering, and Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Konstantinos N Lazaridis
- Center for Individualized Medicine, Department of Internal Medicine, Division of Gastroenterology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
6
|
Dey AK, Banarjee R, Boroumand M, Rutherford DV, Strassheim Q, Nyunt T, Olinger B, Basisty N. Translating Senotherapeutic Interventions into the Clinic with Emerging Proteomic Technologies. BIOLOGY 2023; 12:1301. [PMID: 37887011 PMCID: PMC10604147 DOI: 10.3390/biology12101301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/28/2023]
Abstract
Cellular senescence is a state of irreversible growth arrest with profound phenotypic changes, including the senescence-associated secretory phenotype (SASP). Senescent cell accumulation contributes to aging and many pathologies including chronic inflammation, type 2 diabetes, cancer, and neurodegeneration. Targeted removal of senescent cells in preclinical models promotes health and longevity, suggesting that the selective elimination of senescent cells is a promising therapeutic approach for mitigating a myriad of age-related pathologies in humans. However, moving senescence-targeting drugs (senotherapeutics) into the clinic will require therapeutic targets and biomarkers, fueled by an improved understanding of the complex and dynamic biology of senescent cell populations and their molecular profiles, as well as the mechanisms underlying the emergence and maintenance of senescence cells and the SASP. Advances in mass spectrometry-based proteomic technologies and workflows have the potential to address these needs. Here, we review the state of translational senescence research and how proteomic approaches have added to our knowledge of senescence biology to date. Further, we lay out a roadmap from fundamental biological discovery to the clinical translation of senotherapeutic approaches through the development and application of emerging proteomic technologies, including targeted and untargeted proteomic approaches, bottom-up and top-down methods, stability proteomics, and surfaceomics. These technologies are integral for probing the cellular composition and dynamics of senescent cells and, ultimately, the development of senotype-specific biomarkers and senotherapeutics (senolytics and senomorphics). This review aims to highlight emerging areas and applications of proteomics that will aid in exploring new senescent cell biology and the future translation of senotherapeutics.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Nathan Basisty
- Translational Geroproteomics Unit, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA; (A.K.D.); (R.B.); (M.B.); (D.V.R.); (Q.S.); (T.N.); (B.O.)
| |
Collapse
|
7
|
Maurer GS, Clayton ZS. Anthracycline chemotherapy, vascular dysfunction and cognitive impairment: burgeoning topics and future directions. Future Cardiol 2023; 19:547-566. [PMID: 36354315 PMCID: PMC10599408 DOI: 10.2217/fca-2022-0086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/17/2022] [Indexed: 11/12/2022] Open
Abstract
Anthracyclines, chemotherapeutic agents used to treat common forms of cancer, increase cardiovascular (CV) complications, thereby necessitating research regarding interventions to improve the health of cancer survivors. Vascular dysfunction, which is induced by anthracycline chemotherapy, is an established antecedent to overt CV diseases. Potential treatment options for ameliorating vascular dysfunction have largely been understudied. Furthermore, patients treated with anthracyclines have impaired cognitive function and vascular dysfunction is an independent risk factor for the development of mild cognitive impairment. Here, we will focus on: anthracycline chemotherapy associated CV diseases risk; how targeting mechanisms underlying vascular dysfunction may be a means to improve both CV and cognitive health; and research gaps and potential future directions for the field of cardio-oncology.
Collapse
Affiliation(s)
- Grace S Maurer
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Zachary S Clayton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| |
Collapse
|
8
|
Caddye E, Pineau J, Reyniers J, Ronen I, Colasanti A. Lactate: A Theranostic Biomarker for Metabolic Psychiatry? Antioxidants (Basel) 2023; 12:1656. [PMID: 37759960 PMCID: PMC10526106 DOI: 10.3390/antiox12091656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/01/2023] [Accepted: 08/16/2023] [Indexed: 09/29/2023] Open
Abstract
Alterations in neurometabolism and mitochondria are implicated in the pathophysiology of psychiatric conditions such as mood disorders and schizophrenia. Thus, developing objective biomarkers related to brain mitochondrial function is crucial for the development of interventions, such as central nervous system penetrating agents that target brain health. Lactate, a major circulatory fuel source that can be produced and utilized by the brain and body, is presented as a theranostic biomarker for neurometabolic dysfunction in psychiatric conditions. This concept is based on three key properties of lactate that make it an intriguing metabolic intermediate with implications for this field: Firstly, the lactate response to various stimuli, including physiological or psychological stress, represents a quantifiable and dynamic marker that reflects metabolic and mitochondrial health. Second, lactate concentration in the brain is tightly regulated according to the sleep-wake cycle, the dysregulation of which is implicated in both metabolic and mood disorders. Third, lactate universally integrates arousal behaviours, pH, cellular metabolism, redox states, oxidative stress, and inflammation, and can signal and encode this information via intra- and extracellular pathways in the brain. In this review, we expand on the above properties of lactate and discuss the methodological developments and rationale for the use of functional magnetic resonance spectroscopy for in vivo monitoring of brain lactate. We conclude that accurate and dynamic assessment of brain lactate responses might contribute to the development of novel and personalized therapies that improve mitochondrial health in psychiatric disorders and other conditions associated with neurometabolic dysfunction.
Collapse
Affiliation(s)
- Edward Caddye
- Clinical Imaging Sciences Centre, Brighton and Sussex Medical School, University of Sussex, Falmer BN1 9RR, UK
- Department of Clinical Neuroscience, Brighton and Sussex Medical School, University of Sussex, Falmer BN1 9RR, UK
| | - Julien Pineau
- Independent Researcher, Florianópolis 88062-300, Brazil
| | - Joshua Reyniers
- Department of Clinical Neuroscience, Brighton and Sussex Medical School, University of Sussex, Falmer BN1 9RR, UK
- School of Life Sciences, University of Sussex, Falmer BN1 9RR, UK
| | - Itamar Ronen
- Clinical Imaging Sciences Centre, Brighton and Sussex Medical School, University of Sussex, Falmer BN1 9RR, UK
| | - Alessandro Colasanti
- Clinical Imaging Sciences Centre, Brighton and Sussex Medical School, University of Sussex, Falmer BN1 9RR, UK
- Department of Clinical Neuroscience, Brighton and Sussex Medical School, University of Sussex, Falmer BN1 9RR, UK
| |
Collapse
|
9
|
Delval L, Hantute-Ghesquier A, Sencio V, Flaman JM, Robil C, Angulo FS, Lipskaia L, Çobanoğlu O, Lacoste AS, Machelart A, Danneels A, Corbin M, Deruyter L, Heumel S, Idziorek T, Séron K, Sauve F, Bongiovanni A, Prévot V, Wolowczuk I, Belouzard S, Saliou JM, Gosset P, Bernard D, Rouillé Y, Adnot S, Duterque-Coquillaud M, Trottein F. Removal of senescent cells reduces the viral load and attenuates pulmonary and systemic inflammation in SARS-CoV-2-infected, aged hamsters. NATURE AGING 2023; 3:829-845. [PMID: 37414987 PMCID: PMC10353934 DOI: 10.1038/s43587-023-00442-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 05/24/2023] [Indexed: 07/08/2023]
Abstract
Older age is one of the strongest risk factors for severe COVID-19. In this study, we determined whether age-associated cellular senescence contributes to the severity of experimental COVID-19. Aged golden hamsters accumulate senescent cells in the lungs, and the senolytic drug ABT-263, a BCL-2 inhibitor, depletes these cells at baseline and during SARS-CoV-2 infection. Relative to young hamsters, aged hamsters had a greater viral load during the acute phase of infection and displayed higher levels of sequelae during the post-acute phase. Early treatment with ABT-263 lowered pulmonary viral load in aged (but not young) animals, an effect associated with lower expression of ACE2, the receptor for SARS-CoV-2. ABT-263 treatment also led to lower pulmonary and systemic levels of senescence-associated secretory phenotype factors and to amelioration of early and late lung disease. These data demonstrate the causative role of age-associated pre-existing senescent cells on COVID-19 severity and have clear clinical relevance.
Collapse
Affiliation(s)
- Lou Delval
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017, Center for Infection and Immunity of Lille, Lille, France
| | - Aline Hantute-Ghesquier
- Université de Lille, CNRS, INSERM, CHU Lille, UMR9020-U1277, Institut Pasteur de Lille-CANTHER, Lille, France
| | - Valentin Sencio
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017, Center for Infection and Immunity of Lille, Lille, France
| | - Jean Michel Flaman
- Université de Lyon, CNRS, INSERM, U1052-UMR 5286, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Lyon, France
| | - Cyril Robil
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017, Center for Infection and Immunity of Lille, Lille, France
| | - Fabiola Silva Angulo
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017, Center for Infection and Immunity of Lille, Lille, France
| | - Larissa Lipskaia
- Université de Paris-Est Créteil, INSERM U955, Institut Mondor de Recherche Biomédicale, Créteil, France
| | - Ozmen Çobanoğlu
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017, Center for Infection and Immunity of Lille, Lille, France
| | - Anne-Sophie Lacoste
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, US 41-UAR 2014, Platforms Lille in Biology & Health, Lille, France
| | - Arnaud Machelart
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017, Center for Infection and Immunity of Lille, Lille, France
| | - Adeline Danneels
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017, Center for Infection and Immunity of Lille, Lille, France
| | - Mathieu Corbin
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017, Center for Infection and Immunity of Lille, Lille, France
| | - Lucie Deruyter
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017, Center for Infection and Immunity of Lille, Lille, France
| | - Séverine Heumel
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017, Center for Infection and Immunity of Lille, Lille, France
| | - Thierry Idziorek
- Université de Lille, CNRS, INSERM, CHU Lille, UMR9020-U1277, Institut Pasteur de Lille-CANTHER, Lille, France
| | - Karin Séron
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017, Center for Infection and Immunity of Lille, Lille, France
| | - Florent Sauve
- Université de Lille, INSERM, CHU Lille, U1172-UMR 9017, Lille Neuroscience & Cognition Research Center, Lille, France
| | - Antonino Bongiovanni
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, US 41-UAR 2014, Platforms Lille in Biology & Health, Lille, France
| | - Vincent Prévot
- Université de Lille, INSERM, CHU Lille, U1172-UMR 9017, Lille Neuroscience & Cognition Research Center, Lille, France
| | - Isabelle Wolowczuk
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017, Center for Infection and Immunity of Lille, Lille, France
| | - Sandrine Belouzard
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017, Center for Infection and Immunity of Lille, Lille, France
| | - Jean-Michel Saliou
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, US 41-UAR 2014, Platforms Lille in Biology & Health, Lille, France
| | - Philippe Gosset
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017, Center for Infection and Immunity of Lille, Lille, France
| | - David Bernard
- Université de Lyon, CNRS, INSERM, U1052-UMR 5286, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, Lyon, France
| | - Yves Rouillé
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017, Center for Infection and Immunity of Lille, Lille, France
| | - Serge Adnot
- Université de Paris-Est Créteil, INSERM U955, Institut Mondor de Recherche Biomédicale, Créteil, France
| | - Martine Duterque-Coquillaud
- Université de Lille, CNRS, INSERM, CHU Lille, UMR9020-U1277, Institut Pasteur de Lille-CANTHER, Lille, France
| | - François Trottein
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017, Center for Infection and Immunity of Lille, Lille, France.
| |
Collapse
|
10
|
Akter R, Rahman MR, Ahmed ZS, Afrose A. Plausibility of natural immunomodulators in the treatment of COVID-19-A comprehensive analysis and future recommendations. Heliyon 2023; 9:e17478. [PMID: 37366526 PMCID: PMC10284624 DOI: 10.1016/j.heliyon.2023.e17478] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 06/15/2023] [Accepted: 06/19/2023] [Indexed: 06/28/2023] Open
Abstract
The COVID-19 pandemic has inflicted millions of deaths worldwide. Despite the availability of several vaccines and some special drugs approved for emergency use to prevent or treat this disease still, there is a huge concern regarding their effectiveness, adverse effects, and most importantly, their efficacy against the new variants. A cascade of immune-inflammatory responses is involved with the pathogenesis and severe complications with COVID-19. People with dysfunctional and compromised immune systems display severe complications, including acute respiratory distress syndrome, sepsis, multiple organ failure etc., when they get infected with the SARS-CoV-2 virus. Plant-derived natural immune-suppressant compounds, such as resveratrol, quercetin, curcumin, berberine, luteolin, etc., have been reported to inhibit pro-inflammatory cytokines and chemokines. Therefore, natural products with immunomodulatory and anti-inflammatory potential could be plausible targets to treat this contagious disease. This review aims to delineate the clinical trials status and outcomes of natural compounds with immunomodulatory potential in COVID-19 patients along with the outcomes of their in-vivo studies. In clinical trials several natural immunomodulators resulted in significant improvement of COVID-19 patients by diminishing COVID-19 symptoms such as fever, cough, sore throat, and breathlessness. Most importantly, they reduced the duration of hospitalization and the need for supplemental oxygen therapy, improved clinical outcomes in patients with COVID-19, especially weakness, and eliminated acute lung injury and acute respiratory distress syndrome. This paper also discusses many potent natural immunomodulators yet to undergo clinical trials. In-vivo studies with natural immunomodulators demonstrated reduction of a wide range of proinflammatory cytokines. Natural immunomodulators that were found effective, safe, and well tolerated in small-scale clinical trials are warranted to undergo large-scale trials to be used as drugs to treat COVID-19 infections. Alongside, compounds yet to test clinically must undergo clinical trials to find their effectiveness and safety in the treatment of COVID-19 patients.
Collapse
Affiliation(s)
- Raushanara Akter
- School of Pharmacy, Brac University, 66 Mohakhali, Dhaka, Bangladesh
| | - Md. Rashidur Rahman
- Department of Pharmacy, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Zainab Syed Ahmed
- School of Pharmacy, Brac University, 66 Mohakhali, Dhaka, Bangladesh
| | - Afrina Afrose
- School of Pharmacy, Brac University, 66 Mohakhali, Dhaka, Bangladesh
| |
Collapse
|
11
|
Halma MTJ, Plothe C, Marik P, Lawrie TA. Strategies for the Management of Spike Protein-Related Pathology. Microorganisms 2023; 11:1308. [PMID: 37317282 PMCID: PMC10222799 DOI: 10.3390/microorganisms11051308] [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: 03/16/2023] [Revised: 05/04/2023] [Accepted: 05/10/2023] [Indexed: 06/16/2023] Open
Abstract
In the wake of the COVID-19 crisis, a need has arisen to prevent and treat two related conditions, COVID-19 vaccine injury and long COVID-19, both of which can trace at least part of their aetiology to the spike protein, which can cause harm through several mechanisms. One significant mechanism of harm is vascular, and it is mediated by the spike protein, a common element of the COVID-19 illness, and it is related to receiving a COVID-19 vaccine. Given the significant number of people experiencing these two related conditions, it is imperative to develop treatment protocols, as well as to consider the diversity of people experiencing long COVID-19 and vaccine injury. This review summarizes the known treatment options for long COVID-19 and vaccine injury, their mechanisms, and their evidentiary basis.
Collapse
Affiliation(s)
| | - Christof Plothe
- Center for Biophysical Osteopathy, Am Wegweiser 27, 55232 Alzey, Germany
| | - Paul Marik
- Front Line COVID-19 Critical Care Alliance (FLCCC), 2001 L St. NW Suite 500, Washington, DC 20036, USA;
| | | |
Collapse
|
12
|
Du PY, Gandhi A, Bawa M, Gromala J. The ageing immune system as a potential target of senolytics. OXFORD OPEN IMMUNOLOGY 2023; 4:iqad004. [PMID: 37255929 PMCID: PMC10191675 DOI: 10.1093/oxfimm/iqad004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 06/01/2023] Open
Abstract
Ageing leads to a sharp decline in immune function, precipitating the development of inflammatory conditions. The combined impact of these processes renders older individuals at greater risk of inflammatory and immune-related diseases, such as cancer and infections. This is compounded by reduced efficacy in interventions aiming to limit disease impact, for instance vaccines being less effective in elderly populations. This state of diminished cellular function is driven by cellular senescence, a process where cells undergo stable growth arrest following exposure to stressful stimuli, and the associated pro-inflammatory secretory phenotype. Removing harmful senescent cells (SnCs) using senolytic therapies is an emerging field holding promise for patient benefit. Current senolytics have been developed either to specifically target SnCs, or repurposed from cancer therapies or vaccination protocols. Herein, we discuss recent developments in senolytic therapies, focusing on how senolytics could be used to combat the age-associated diminution of the immune system. In particular, exploring how these drugs may be used to promote immunity in the elderly, and highlighting recent trials of senolytics in idiopathic pulmonary fibrosis and diabetic kidney disease. Novel immunotherapeutic approaches including chimeric antigen receptor T-cells or monoclonal antibodies targeting SnCs are being investigated to combat the shortcomings of current senolytics and their adverse effects. The flexible nature of senolytic treatment modalities and their efficacy in safely removing harmful SnCs could have great potential to promote healthy immune function in ageing populations.
Collapse
Affiliation(s)
- Peter Yandi Du
- Correspondence address. Faculty of Medicine, Imperial College London, Level 2, Faculty Building, South Kensington Campus, London SW7 2AZ, UK. Tel: +44 (0)20 3313 8213, E-mail:
| | | | | | | |
Collapse
|
13
|
Schmitt CA, Tchkonia T, Niedernhofer LJ, Robbins PD, Kirkland JL, Lee S. COVID-19 and cellular senescence. Nat Rev Immunol 2023; 23:251-263. [PMID: 36198912 PMCID: PMC9533263 DOI: 10.1038/s41577-022-00785-2] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2022] [Indexed: 11/15/2022]
Abstract
The clinical severity of coronavirus disease 2019 (COVID-19) is largely determined by host factors. Recent advances point to cellular senescence, an ageing-related switch in cellular state, as a critical regulator of SARS-CoV-2-evoked hyperinflammation. SARS-CoV-2, like other viruses, can induce senescence and exacerbates the senescence-associated secretory phenotype (SASP), which is comprised largely of pro-inflammatory, extracellular matrix-degrading, complement-activating and pro-coagulatory factors secreted by senescent cells. These effects are enhanced in elderly individuals who have an increased proportion of pre-existing senescent cells in their tissues. SASP factors can contribute to a 'cytokine storm', tissue-destructive immune cell infiltration, endothelialitis (endotheliitis), fibrosis and microthrombosis. SASP-driven spreading of cellular senescence uncouples tissue injury from direct SARS-CoV-2-inflicted cellular damage in a paracrine fashion and can further amplify the SASP by increasing the burden of senescent cells. Preclinical and early clinical studies indicate that targeted elimination of senescent cells may offer a novel therapeutic opportunity to attenuate clinical deterioration in COVID-19 and improve resilience following infection with SARS-CoV-2 or other pathogens.
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.
- Faculty of Medicine, 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.
| | - Tamar Tchkonia
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Laura J Niedernhofer
- Institute on the Biology of Aging and Metabolism and the Department of Biochemistry, Molecular Biology, and Biochemistry, University of Minnesota, Minneapolis, MN, USA
| | - Paul D Robbins
- Institute on the Biology of Aging and Metabolism and the Department of Biochemistry, Molecular Biology, and Biochemistry, University of Minnesota, Minneapolis, MN, USA
| | - James L Kirkland
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Soyoung Lee
- 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.
- Faculty of Medicine, Johannes Kepler University, Linz, Austria.
| |
Collapse
|
14
|
Adnot S, Bernard D, Lipskaia L, Trottein F. [Cell senescence, a new target for respiratory viral infections: From influenza virus to SARS-CoV-2]. BULLETIN DE L'ACADEMIE NATIONALE DE MEDECINE 2023; 207:193-198. [PMID: 36624738 PMCID: PMC9812359 DOI: 10.1016/j.banm.2022.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 11/21/2022] [Indexed: 01/06/2023]
Abstract
The accumulation of senescent cells in tissues is a key process of aging and age-related diseases, including lung diseases such as chronic obstructive pulmonary disease, lung fibrosis, or cancer. In recent years, the spectrum of respiratory diseases associated with cellular senescence has been broadened, in particular acute viral pulmonary infections, foremost among which is coronavirus disease 2019 (COVID19), which is particularly severe in the elderly or in subjects with comorbidities. Influenza virus infection, which strikes more severely at the extreme ages of life, is also associated with severe pulmonary senescence. Cellular senescence potentially represents an original target for attacking these diseases, although its specific mechanisms remain largely misunderstood. New anti-senescent therapeutic approaches are thus proposed during severe viral pulmonary infections, with the aim of preventing acute effects and/or, in the longer term, pulmonary sequelae.
Collapse
Affiliation(s)
- S. Adnot
- Inserm U955 et département de physiologie, hôpital Henri-Mondor, AP–HP, FHU- SENEC, université Paris-Est Créteil (UPEC), 94010 Créteil, France,Auteur correspondant. Hôpital Henri Mondor, service de physiologie-explorations fonctionnelles, 94010 Créteil, France
| | - D. Bernard
- Université de Lyon, CNRS, Inserm, U1052 - UMR 5286, centre de recherche en cancérologie de Lyon, centre Léon Bérard, 69373 Lyon, France
| | - L. Lipskaia
- Inserm U955 et département de physiologie, hôpital Henri-Mondor, AP–HP, FHU- SENEC, université Paris-Est Créteil (UPEC), 94010 Créteil, France
| | - F. Trottein
- Université de Lille, CNRS, Inserm, CHU de Lille, institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, 59000 Lille, France
| |
Collapse
|
15
|
Chen JC, Wang R, Wei CC. Anti-aging effects of dietary phytochemicals: From Caenorhabditis elegans, Drosophila melanogaster, rodents to clinical studies. Crit Rev Food Sci Nutr 2023; 64:5958-5983. [PMID: 36597655 DOI: 10.1080/10408398.2022.2160961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Anti-aging research has become critical since the elderly population is increasing dramatically in this era. With the establishment of frailty phenotype and frailty index, the importance of anti-frailty research is concurrently enlightened. The application of natural phytochemicals against aging or frailty is always intriguing, and abundant related studies have been published. Various models are designed for biological research, and each model has its strength and weakness in deciphering the complex aging mechanisms. In this article, we attempt to show the potential of Caenorhabditis elegans in the study of phytochemicals' effects on anti-aging by comparing it to other animal models. In this review, the lifespan extension and anti-aging effects are demonstrated by various physical, cellular, or molecular biomarkers of dietary phytochemicals, including resveratrol, curcumin, urolithin A, sesamin, fisetin, quercetin, epigallocatechin-3-gallate, epicatechin, spermidine, sulforaphane, along with extracts of broccoli, cocoa, and blueberry. Meanwhile, the frequency of phytochemicals and models studied or presented in publications since 2010 were analyzed, and the most commonly mentioned animal models were rats, mice, and the nematode C. elegans. This up-to-date summary of the anti-aging effect of certain phytochemicals has demonstrated powerful potential for anti-aging or anti-frailty in the human population.
Collapse
Affiliation(s)
- Ju-Chi Chen
- Institute of Food Safety and Health, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Reuben Wang
- Institute of Food Safety and Health, College of Public Health, National Taiwan University, Taipei, Taiwan
- Master of Public Health Program, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Chia-Cheng Wei
- Institute of Food Safety and Health, College of Public Health, National Taiwan University, Taipei, Taiwan
- Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
16
|
Yu X, Lobo JD, Sundermann E, Baker DJ, Tracy RP, Kuchel GA, Stephenson KE, Letendre SL, Brew B, Cysique LA, Dale SK, Wallen C, Kunisaki KM, Guaraldi G, Milic J, Winston A, Moore DJ, Margolick JB, Erlandson KM. Current Challenges and Solutions for Clinical Management and Care of People with HIV: Findings from the 12th Annual International HIV and Aging Workshop. AIDS Res Hum Retroviruses 2023; 39:1-12. [PMID: 36322713 PMCID: PMC9889016 DOI: 10.1089/aid.2022.0079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
People with HIV on combination antiretroviral therapy (ART) have longer life expectancy and are increasingly experiencing age-related comorbidities. Thus, aging with HIV has become a central issue in clinical care and research, which has been particularly challenging with the intersection of the ongoing coronavirus (COVID)-19 pandemic. Since 2009, the International Workshop on HIV and Aging has served as a multidisciplinary platform to share research findings from cross-disciplinary fields along with community advocates to address critical issues in HIV and aging. In this article, we summarize the key oral presentations from the 12th Annual International Workshop on HIV and Aging, held virtually on September 23rd and 24th, 2021. The topics ranged from basic science research on biological mechanisms of aging to quality of life and delivery of care under the COVID-19 pandemic. This workshop enriched our understanding of HIV and aging under the COVID-19 pandemic, identified challenges and opportunities to combat the impact of COVID-19 on HIV communities, and also provided updated research and future directions of the field to move HIV and aging research forward, with the ultimate goal of successful aging for older people with HIV.
Collapse
Affiliation(s)
- Xiaoying Yu
- Department of Biostatistics and Data Science, University of Texas Medical Branch, Galveston, Texas, USA
| | - Judith D. Lobo
- Department of Psychiatry, University of California San Diego, San Diego, California, USA
| | - Erin Sundermann
- Department of Psychiatry, University of California San Diego, San Diego, California, USA
| | - Darren J. Baker
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Russell P. Tracy
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Colchester, Vermont, USA
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Colchester, Vermont, USA
| | - George A. Kuchel
- UConn Center on Aging, Department of Medicine, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Kathryn E. Stephenson
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Scott L. Letendre
- Department of Psychiatry, University of California San Diego, San Diego, California, USA
- Department of Medicine, University of California San Diego, San Diego, California, USA
| | - Bruce Brew
- Department of Neurology and HIV Medicine, St. Vincent's Hospital, Sydney, Australia
- Department of Neurology, Peter Duncan Neurosciences Unit, St. Vincent's Centre for Applied Medical Research, Sydney, Australia
- Faculty of Medicine, Department of Neurology, University of New South Wales Sydney, Sydney, Australia
- University of Notre Dame Australia, Sydney, Australia
| | - Lucette A. Cysique
- Faculty of Science, Department of Psychology, School of Psychology, University of New South Wales Sydney, Sydney, Australia
- MAP Centre for Urban Health Solutions, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Canada
- Neuroscience Unit, St. Vincent's Hospital Centre for Applied Medical Research, Sydney, Australia
| | - Sannisha K. Dale
- Department of Psychology, University of Miami, Coral Gables, Florida, USA
| | - Chelsie Wallen
- Department of Psychology, University of Miami, Coral Gables, Florida, USA
| | - Ken M. Kunisaki
- Section of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Minneapolis VA Health Care System, Minneapolis, Minnesota, USA
- Department of Medicine; Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Giovanni Guaraldi
- Department of Surgical, Medical, Dental and Morphological Sciences, Modena HIV Metabolic Clinic (MHMC), University of Modena and Reggio Emilia, Modena, Italy
| | - Jovana Milic
- Department of Surgical, Medical, Dental and Morphological Sciences, Modena HIV Metabolic Clinic (MHMC), University of Modena and Reggio Emilia, Modena, Italy
| | - Alan Winston
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - David J. Moore
- Department of Psychiatry, University of California San Diego, San Diego, California, USA
| | - Joseph B. Margolick
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Kristine M. Erlandson
- Divisions of Infectious Diseases, Department of Medicine, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado, USA
- Divisions of Geriatric Medicine, Department of Medicine, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
17
|
Ma P, Seguin J, Ly KN, Henríquez LC, Plansart E, Hammad K, Gahoual R, Dhôtel H, Izabelle C, Saubamea B, Richard C, Escriou V, Mignet N, Corvis Y. Designing fisetin nanocrystals for enhanced in cellulo anti-angiogenic and anticancer efficacy. Int J Pharm X 2022; 4:100138. [DOI: 10.1016/j.ijpx.2022.100138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022] Open
|
18
|
Liao CM, Wulfmeyer VC, Chen R, Erlangga Z, Sinning J, von Mässenhausen A, Sörensen-Zender I, Beer K, von Vietinghoff S, Haller H, Linkermann A, Melk A, Schmitt R. Induction of ferroptosis selectively eliminates senescent tubular cells. Am J Transplant 2022; 22:2158-2168. [PMID: 35607817 DOI: 10.1111/ajt.17102] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 04/27/2022] [Accepted: 05/22/2022] [Indexed: 01/25/2023]
Abstract
The accumulation of senescent cells is an important contributor to kidney aging, chronic renal disease, and poor outcome after kidney transplantation. Approaches to eliminate senescent cells with senolytic compounds have been proposed as novel strategies to improve marginal organs. While most existing senolytics induce senescent cell clearance by apoptosis, we observed that ferroptosis, an iron-catalyzed subtype of regulated necrosis, might serve as an alternative way to ablate senescent cells. We found that murine kidney tubular epithelial cells became sensitized to ferroptosis when turning senescent. This was linked to increased expression of pro-ferroptotic lipoxygenase-5 and reduced expression of anti-ferroptotic glutathione peroxidase 4 (GPX4). In tissue slice cultures from aged kidneys low dose application of the ferroptosis-inducer RSL3 selectively eliminated senescent cells while leaving healthy tubular cells unaffected. Similar results were seen in a transplantation model, in which RSL3 reduced the senescent cell burden of aged donor kidneys and caused a reduction of damage and inflammatory cell infiltration during the early post-transplantation period. In summary, these data reveal an increased susceptibility of senescent tubular cells to ferroptosis with the potential to be exploited for selective reduction of renal senescence in aged kidney transplants.
Collapse
Affiliation(s)
- Chieh M Liao
- Department of Nephrology and Hypertension, Medical School Hannover, Hannover, Germany
| | - Vera C Wulfmeyer
- Department of Nephrology and Hypertension, Medical School Hannover, Hannover, Germany
| | - Rongjun Chen
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Medical School Hannover, Hannover, Germany
| | - Zulrahman Erlangga
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Medical School Hannover, Hannover, Germany
| | - Julius Sinning
- Department of Nephrology and Hypertension, Medical School Hannover, Hannover, Germany
| | - Anne von Mässenhausen
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische University of Dresden, Dresden, Germany
| | - Inga Sörensen-Zender
- Department of Nephrology and Hypertension, Medical School Hannover, Hannover, Germany
| | - Kristina Beer
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische University of Dresden, Dresden, Germany
| | - Sibylle von Vietinghoff
- Department of Nephrology and Hypertension, Medical School Hannover, Hannover, Germany.,Nephrology Section, First Medical Clinic, University Clinic and Rheinische Friedrich-Wilhelms Universität Bonn, Bonn, Germany
| | - Hermann Haller
- Department of Nephrology and Hypertension, Medical School Hannover, Hannover, Germany
| | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische University of Dresden, Dresden, Germany
| | - Anette Melk
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Medical School Hannover, Hannover, Germany
| | - Roland Schmitt
- Department of Nephrology and Hypertension, Medical School Hannover, Hannover, Germany
| |
Collapse
|
19
|
New Trends in Aging Drug Discovery. Biomedicines 2022; 10:biomedicines10082006. [PMID: 36009552 PMCID: PMC9405986 DOI: 10.3390/biomedicines10082006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/13/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
Aging is considered the main risk factor for many chronic diseases that frequently appear at advanced ages. However, the inevitability of this process is being questioned by recent research that suggests that senescent cells have specific features that differentiate them from younger cells and that removal of these cells ameliorates senescent phenotype and associated diseases. This opens the door to the design of tailored therapeutic interventions aimed at reducing and delaying the impact of senescence in life, that is, extending healthspan and treating aging as another chronic disease. Although these ideas are still far from reaching the bedside, it is conceivable that they will revolutionize the way we understand aging in the next decades. In this review, we analyze the main and well-validated cellular pathways and targets related to senescence as well as their implication in aging-associated diseases. In addition, the most relevant small molecules with senotherapeutic potential, with a special emphasis on their mechanism of action, ongoing clinical trials, and potential limitations, are discussed. Finally, a brief overview of alternative strategies that go beyond the small molecule field, together with our perspectives for the future of the field, is provided.
Collapse
|
20
|
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
|
21
|
Clayton ZS, Craighead DH, Darvish S, Coppock M, Ludwig KR, Brunt VE, Seals DR, Rossman MJ. Promoting healthy cardiovascular aging: emerging topics. THE JOURNAL OF CARDIOVASCULAR AGING 2022; 2:43. [PMID: 36337728 PMCID: PMC9632540 DOI: 10.20517/jca.2022.27] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The development of age-related cardiovascular (CV) dysfunction increases the risk of CV disease as well as other chronic age-associated disorders, including chronic kidney disease, and Alzheimer's disease and related dementias. Major manifestations of age-associated CV dysfunction that increase disease risk are vascular dysfunction, primarily vascular endothelial dysfunction and arterial stiffening, and elevated systolic blood pressure. Declines in nitric oxide bioavailability secondary to increased oxidative stress and inflammation are established mechanisms of CV dysfunction with aging. Moreover, fundamental mechanisms of aging, termed the "hallmarks of aging" extend to the CV system and, as such, may be considered "hallmarks of CV aging". These mechanisms represent viable therapeutic targets for treating CV dysfunction with aging. Healthy lifestyle behaviors, such as regular aerobic exercise and certain dietary patterns, are considered "first-line" strategies to prevent and/or treat age-associated CV dysfunction. Despite the well-established benefits of these strategies, many older adults do not meet the recommended guidelines for exercise or consume a healthy diet. Therefore, it is important to establish alternative and/or complementary evidence-based approaches to prevent or reverse age-related CV dysfunction. Targeting fundamental mechanisms of CV aging with interventions such as time-efficient exercise training, food-derived molecules, termed nutraceuticals, or select synthetic pharmacological agents represents a promising approach. In the present review, we will highlight emerging topics in the field of healthy CV aging with a specific focus on how exercise, nutrition/dietary patterns, nutraceuticals and select synthetic pharmacological compounds may promote healthy CV aging, in part, by targeting the hallmarks of CV aging.
Collapse
Affiliation(s)
- Zachary S Clayton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Daniel H Craighead
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Sanna Darvish
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - McKinley Coppock
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Katelyn R Ludwig
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Vienna E Brunt
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Matthew J Rossman
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| |
Collapse
|
22
|
Affiliation(s)
- John W Rowe
- Robert Butler Columbia Aging Center, Mailman School of Public Health, Columbia University, New York, New York, USA
- Department of Health Policy and Management, Mailman School of Public Health, Columbia University, New York, New York, USA
| |
Collapse
|
23
|
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: 43] [Impact Index Per Article: 14.3] [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
|
24
|
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: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 05/06/2022] [Indexed: 01/15/2023]
Abstract
With aging, there is increased dysfunction of both innate and adaptive immune responses, which contributes to impaired immune responses to pathogens and greater mortality and morbidity. This age-related immune dysfunction is defined in general as immunosenescence and includes an increase in the number of memory T cells, loss of ability to respond to antigen and a lingering level of low-grade inflammation. However, certain features of immunosenescence are similar to cellular senescence, which is defined as the irreversible loss of proliferation in response to damage and stress. Importantly, senescence cells can develop an inflammatory senescence-associated secretory phenotype (SASP), that also drives non-autonomous cellular senescence and immune dysfunction. Interestingly, viral infection can increase the extent of immune senescence both directly and indirectly, leading to increased immune dysfunction and inflammation, especially in the elderly. This review focuses on age-related immune dysfunction, cellular senescence and the impaired immune response to pathogens.
Collapse
Affiliation(s)
| | | | | | | | - Paul D. Robbins
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
25
|
Roh JD, Kitchen RR, Guseh JS, McNeill JN, Aid M, Martinot AJ, Yu A, Platt C, Rhee J, Weber B, Trager LE, Hastings MH, Ducat S, Xia P, Castro C, Singh A, Atlason B, Churchill TW, Di Carli MF, Ellinor PT, Barouch DH, Ho JE, Rosenzweig A. Plasma Proteomics of COVID-19-Associated Cardiovascular Complications: Implications for Pathophysiology and Therapeutics. JACC Basic Transl Sci 2022; 7:425-441. [PMID: 35530264 PMCID: PMC9067411 DOI: 10.1016/j.jacbts.2022.01.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 12/30/2022]
Abstract
To gain insights into the mechanisms driving cardiovascular complications in COVID-19, we performed a case-control plasma proteomics study in COVID-19 patients. Our results identify the senescence-associated secretory phenotype, a marker of biological aging, as the dominant process associated with disease severity and cardiac involvement. FSTL3, an indicator of senescence-promoting Activin/TGFβ signaling, and ADAMTS13, the von Willebrand Factor-cleaving protease whose loss-of-function causes microvascular thrombosis, were among the proteins most strongly associated with myocardial stress and injury. Findings were validated in a larger COVID-19 patient cohort and the hamster COVID-19 model, providing new insights into the pathophysiology of COVID-19 cardiovascular complications with therapeutic implications.
Collapse
Affiliation(s)
- Jason D. Roh
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Robert R. Kitchen
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - J. Sawalla Guseh
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jenna N. McNeill
- Division of Pulmonary and Critical Care, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Malika Aid
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Amanda J. Martinot
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Department of Biomedical Sciences, Section of Pathology, Tufts University Cummings School of Veterinary Medicine, North Grafton, Massachusetts, USA
| | - Andy Yu
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Colin Platt
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - James Rhee
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Brittany Weber
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lena E. Trager
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Margaret H. Hastings
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sarah Ducat
- Department of Biomedical Sciences, Section of Pathology, Tufts University Cummings School of Veterinary Medicine, North Grafton, Massachusetts, USA
| | - Peng Xia
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Claire Castro
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Abhilasha Singh
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Bjarni Atlason
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Timothy W. Churchill
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Marcelo F. Di Carli
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Radiology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Patrick T. Ellinor
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Dan H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Jennifer E. Ho
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Anthony Rosenzweig
- Corrigan Minehan Heart Center, Division of Cardiology, Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
26
|
Gurwitz JH, Quinn CC, Abi-Elias IH, Adams AS, Bartel R, Bonner A, Boxer R, Delude C, Gifford D, Hanson B, Ito K, Jain P, Magaziner JS, Mazor KM, Mitchell SL, Mody L, Nace D, Ouslander J, Reifsnyder J, Resnick B, Zimmerman S. Advancing clinical trials in nursing homes: A proposed roadmap to success. Geriatr Nurs 2022; 45:230-234. [PMID: 35361514 PMCID: PMC8960155 DOI: 10.1016/j.gerinurse.2022.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
An effective clinical research effort in nursing homes to address prevention and treatment of COVID-19 faced overwhelming challenges. Under the Health Care Systems Research Network-Older Americans Independence Centers AGING Initiative, a multidisciplinary Stakeholder Advisory Panel was convened to develop recommendations to improve the capability of the clinical research enterprise in US nursing homes. The Panel considered the nursing home as a setting for clinical trials, reviewed the current state of clinical trials in nursing homes, and ultimately developed recommendations for the establishment of a nursing home clinical trials research network that would be centrally supported and administered. This report summarizes the Panel's recommendations, which were developed in alignment with the following core principles: build on available research infrastructure where appropriate; leverage existing productive partnerships of researchers with groups of nursing homes and nursing home corporations; encompass both efficacy and effectiveness clinical trials; be responsive to a broad range of stakeholders including nursing home residents and their care partners; be relevant to an expansive range of clinical and health care delivery research questions; be able to pivot as necessary to changing research priorities and circumstances; create a pathway for industry-sponsored research as appropriate; invest in strategies to increase diversity in study populations and the research workforce; and foster the development of the next generation of nursing home researchers.
Collapse
Affiliation(s)
- Jerry H Gurwitz
- Meyers Health Care Institute, Worcester, MA, USA; UMass Chan Medical School, Worcester, MA, USA.
| | | | | | - Alyce S Adams
- Stanford University School of Medicine, Stanford, CA, USA
| | - Rosie Bartel
- AGING Patient/Caregiver Advisory Council, Worcester, MA, USA
| | - Alice Bonner
- Institute for Healthcare Improvement, Boston, MA, USA; Johns Hopkins University School of Nursing, Baltimore, MD, USA
| | | | | | - David Gifford
- American Health Care Association, Washington, DC, USA
| | - Bruce Hanson
- AGING Patient/Caregiver Advisory Council, Worcester, MA, USA
| | - Kouta Ito
- Meyers Health Care Institute, Worcester, MA, USA; UMass Chan Medical School, Worcester, MA, USA
| | - Paavani Jain
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jay S Magaziner
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kathleen M Mazor
- Meyers Health Care Institute, Worcester, MA, USA; UMass Chan Medical School, Worcester, MA, USA
| | - Susan L Mitchell
- Hebrew SeniorLife Hinda and Arthur Marcus Institute for Aging Research, Boston, MA, USA
| | - Lona Mody
- University of Michigan, Ann Arbor, MI, USA
| | - David Nace
- University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | - Barbara Resnick
- University of Maryland School of Nursing, Baltimore, MD, USA
| | | |
Collapse
|
27
|
Mato-Basalo R, Lucio-Gallego S, Alarcón-Veleiro C, Sacristán-Santos M, Quintana MDPM, Morente-López M, de Toro FJ, Silva-Fernández L, González-Rodríguez A, Arufe MC, Labora JAF. Action Mechanisms of Small Extracellular Vesicles in Inflammaging. Life (Basel) 2022; 12:life12040546. [PMID: 35455036 PMCID: PMC9028066 DOI: 10.3390/life12040546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 02/05/2023] Open
Abstract
The accumulation process of proinflammatory components in the body due to aging influences intercellular communication and is known as inflammaging. This biological mechanism relates the development of inflammation to the aging process. Recently, it has been reported that small extracellular vesicles (sEVs) are mediators in the transmission of paracrine senescence involved in inflammatory aging. For this reason, their components, as well as mechanisms of action of sEVs, are relevant to develop a new therapy called senodrugs (senolytics and senomorphic) that regulates the intercellular communication of inflammaging. In this review, we include the most recent and relevant studies on the role of sEVs in the inflammatory aging process and in age-related diseases such as cancer and type 2 diabetes.
Collapse
|
28
|
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
|
29
|
Gurwitz JH, Quinn CC, Abi-Elias IH, Adams AS, Bartel R, Bonner A, Boxer R, Delude C, Gifford D, Hanson B, Ito K, Jain P, Magaziner JS, Mazor KM, Mitchell SL, Mody L, Nace D, Ouslander J, Reifsnyder J, Resnick B, Zimmerman S. Advancing Clinical Trials in Nursing Homes: A Proposed Roadmap to Success. J Am Geriatr Soc 2022; 70:701-708. [PMID: 35195276 PMCID: PMC8910690 DOI: 10.1111/jgs.17696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 02/01/2022] [Indexed: 02/02/2023]
Abstract
An effective clinical research effort in nursing homes to address prevention and treatment of COVID-19 faced overwhelming challenges. Under the Health Care Systems Research Network-Older Americans Independence Centers AGING Initiative, a multidisciplinary Stakeholder Advisory Panel was convened to develop recommendations to improve the capability of the clinical research enterprise in US nursing homes. The Panel considered the nursing home as a setting for clinical trials, reviewed the current state of clinical trials in nursing homes, and ultimately developed recommendations for the establishment of a nursing home clinical trials research network that would be centrally supported and administered. This report summarizes the Panel's recommendations, which were developed in alignment with the following core principles: build on available research infrastructure where appropriate; leverage existing productive partnerships of researchers with groups of nursing homes and nursing home corporations; encompass both efficacy and effectiveness clinical trials; be responsive to a broad range of stakeholders including nursing home residents and their care partners; be relevant to an expansive range of clinical and health care delivery research questions; be able to pivot as necessary to changing research priorities and circumstances; create a pathway for industry-sponsored research as appropriate; invest in strategies to increase diversity in study populations and the research workforce; and foster the development of the next generation of nursing home researchers.
Collapse
Affiliation(s)
- Jerry H. Gurwitz
- Meyers Health Care Institute, Worcester, MA, USA
- UMass Chan Medical School, Worcester, MA, USA
| | | | | | - Alyce S. Adams
- Stanford University School of Medicine, Stanford, CA, USA
| | - Rosie Bartel
- AGING Patient/Caregiver Advisory Council, Worcester, MA, USA
| | - Alice Bonner
- Institute for Healthcare Improvement, Boston, MA, USA
- Johns Hopkins University School of Nursing, Baltimore, MD, USA
| | | | | | - David Gifford
- American Health Care Association, Washington, DC, USA
| | - Bruce Hanson
- AGING Patient/Caregiver Advisory Council, Worcester, MA, USA
| | - Kouta Ito
- Meyers Health Care Institute, Worcester, MA, USA
- UMass Chan Medical School, Worcester, MA, USA
| | - Paavani Jain
- University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Kathleen M. Mazor
- Meyers Health Care Institute, Worcester, MA, USA
- UMass Chan Medical School, Worcester, MA, USA
| | - Susan L. Mitchell
- Hebrew SeniorLife Hinda and Arthur Marcus Institute for Aging Research, Boston, MA, USA
| | - Lona Mody
- University of Michigan, Ann Arbor, MI, USA
| | - David Nace
- University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | - Barbara Resnick
- University of Maryland School of Nursing, Baltimore, MD, USA
| | | |
Collapse
|
30
|
Gurwitz JH, Quinn CC, Abi-Elias IH, Adams AS, Bartel R, Bonner A, Boxer R, Delude C, Gifford D, Hanson B, Ito K, Jain P, Magaziner JS, Mazor KM, Mitchell SL, Mody L, Nace D, Ouslander J, Reifsnyder J, Resnick B, Zimmerman S. Advancing Clinical Trials in Nursing Homes: A Proposed Roadmap to Success. J Am Med Dir Assoc 2022; 23:345-349. [PMID: 34953784 PMCID: PMC8692165 DOI: 10.1016/j.jamda.2021.11.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 11/26/2021] [Indexed: 12/26/2022]
Abstract
An effective clinical research effort in nursing homes to address prevention and treatment of COVID-19 faced overwhelming challenges. Under the Health Care Systems Research Network-Older Americans Independence Centers AGING Initiative, a multidisciplinary Stakeholder Advisory Panel was convened to develop recommendations to improve the capability of the clinical research enterprise in US nursing homes. The Panel considered the nursing home as a setting for clinical trials, reviewed the current state of clinical trials in nursing homes, and ultimately developed recommendations for the establishment of a nursing home clinical trials research network that would be centrally supported and administered. This report summarizes the Panel's recommendations, which were developed in alignment with the following core principles: build on available research infrastructure where appropriate; leverage existing productive partnerships of researchers with groups of nursing homes and nursing home corporations; encompass both efficacy and effectiveness clinical trials; be responsive to a broad range of stakeholders including nursing home residents and their care partners; be relevant to an expansive range of clinical and health care delivery research questions; be able to pivot as necessary to changing research priorities and circumstances; create a pathway for industry-sponsored research as appropriate; invest in strategies to increase diversity in study populations and the research workforce; and foster the development of the next generation of nursing home researchers.
Collapse
Affiliation(s)
- Jerry H Gurwitz
- Meyers Health Care Institute, Worcester, MA, USA; UMass Chan Medical School, Worcester, MA, USA.
| | | | | | - Alyce S Adams
- Stanford University School of Medicine, Stanford, CA, USA
| | - Rosie Bartel
- AGING Patient/Caregiver Advisory Council, Worcester, MA, USA
| | - Alice Bonner
- Institute for Healthcare Improvement, Boston, MA, USA; Johns Hopkins University School of Nursing, Baltimore, MD, USA
| | | | | | - David Gifford
- American Health Care Association, Washington, DC, USA
| | - Bruce Hanson
- AGING Patient/Caregiver Advisory Council, Worcester, MA, USA
| | - Kouta Ito
- Meyers Health Care Institute, Worcester, MA, USA; UMass Chan Medical School, Worcester, MA, USA
| | - Paavani Jain
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jay S Magaziner
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kathleen M Mazor
- Meyers Health Care Institute, Worcester, MA, USA; UMass Chan Medical School, Worcester, MA, USA
| | - Susan L Mitchell
- Hebrew SeniorLife Hinda and Arthur Marcus Institute for Aging Research, Boston, MA, USA
| | - Lona Mody
- University of Michigan, Ann Arbor, MI, USA
| | - David Nace
- University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | - Barbara Resnick
- University of Maryland School of Nursing, Baltimore, MD, USA
| | | |
Collapse
|
31
|
Delude C, Abi‐Elias IH, Quinn CC, Adams AS, Magaziner JS, Ito K, Jain P, Gurwitz JH, Mazor KM. Stakeholders’ Views on Priorities Essential for Establishing a Supportive Environment for Clinical Trials in Nursing Homes. J Am Geriatr Soc 2022; 70:950-959. [PMID: 35188222 PMCID: PMC8986625 DOI: 10.1111/jgs.17710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND The U.S. clinical research enterprise in nursing homes was unprepared to mount clinical trials in nursing homes to address urgent questions relevant to prevention and treatment during the COVID-19 pandemic. We identify priorities essential for establishing a supportive environment for future clinical trials in nursing homes. METHODS Two cross-sectional online questionnaires were administered between January and February 2021. One was administered to nursing home providers, researchers, and policymakers; respondents rated the importance of attributes of researchers, facilities, leaders and staff for conducting clinical trials in nursing homes. Because importance may depend on trial type, respondents rated each attribute for efficacy trials (testing an intervention in ideal circumstances) and effectiveness trials (testing an intervention in "real world" circumstances). We calculated the attribute rating means and standard deviations, and used content analysis to characterize open-ended responses. The second questionnaire for resident family members and advocates included open-ended questions about nursing home research, and factors influencing willingness to participate. RESULTS The attributes rated as most essential for conducting efficacy and effectiveness trials in nursing homes are research team attributes, that is, that researchers recognize regulatory constraints; understand and adapt to nursing home workflow; and work collaboratively with nursing home leaders to identify priorities. Resident and facility diversity emerged as essential for effectiveness trials; important dimensions included resident race, ethnicity and income, as well as nursing home urban/rural location, quality ratings, geography, staffing ratios, size, and profit status. Caregivers and resident advocates stressed the importance of communication among participants, researchers, and nursing home leadership and staff at all stages of a trial. CONCLUSION Developing a robust U.S. clinical research enterprise capable of efficiently mounting future clinical trials in nursing homes will require a reimagining of the relationships that exist between researchers, facilities, nursing home leaders, and residents, with a research infrastructure specifically focused on supporting and fostering these connections.
Collapse
Affiliation(s)
| | | | | | | | | | - Kouta Ito
- Meyers Health Care Institute
- UMass Chan Medical School
| | | | | | | |
Collapse
|
32
|
Lorenzo GD, Scafuri L, Costabile F, Pepe L, Scognamiglio A, Crocetto F, Guerra G, Buonerba C. Fisetin as an adjuvant treatment in prostate cancer patients receiving androgen-deprivation therapy. Future Sci OA 2022; 8:FSO784. [PMID: 35251698 PMCID: PMC8890115 DOI: 10.2144/fsoa-2022-0002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 01/17/2021] [Indexed: 11/23/2022] Open
Affiliation(s)
- Giuseppe Di Lorenzo
- Oncology Unit, Hospital ‘Andrea Tortora’, ASL Salerno, Pagani, Italy
- Associazione O.R.A., Somma Vesuviana, Naples, Italy
- Department of Medicine & Health Science, University of Molise, Campobasso, Italy
| | - Luca Scafuri
- Oncology Unit, Hospital ‘Andrea Tortora’, ASL Salerno, Pagani, Italy
- Associazione O.R.A., Somma Vesuviana, Naples, Italy
| | - Ferdinando Costabile
- Oncology Unit, Hospital ‘Andrea Tortora’, ASL Salerno, Pagani, Italy
- Associazione O.R.A., Somma Vesuviana, Naples, Italy
| | - Liuba Pepe
- Department of Medicine & Health Science, University of Molise, Campobasso, Italy
| | - Anna Scognamiglio
- Department of Medicine & Health Science, University of Molise, Campobasso, Italy
| | - Felice Crocetto
- Department of Neurosciences, Reproductive Sciences & Odontostomatology, Federico II University, Naples, Italy
| | - Germano Guerra
- Department of Medicine & Health Science, University of Molise, Campobasso, Italy
| | - Carlo Buonerba
- Oncology Unit, Hospital ‘Andrea Tortora’, ASL Salerno, Pagani, Italy
- Associazione O.R.A., Somma Vesuviana, Naples, Italy
| |
Collapse
|
33
|
Raffaele M, Vinciguerra M. The costs and benefits of senotherapeutics for human health. THE LANCET. HEALTHY LONGEVITY 2022; 3:e67-e77. [PMID: 36098323 DOI: 10.1016/s2666-7568(21)00300-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/09/2021] [Accepted: 11/21/2021] [Indexed: 12/24/2022] Open
Abstract
Cellular senescence is a major contributor to age-related diseases in humans; however, it also has a beneficial role in physiological and pathological processes, including wound healing, host immunity, and tumour suppression. Reducing the burden of cell senescence in animal models of cardiometabolic disorders, inflammatory conditions, neurodegenerative diseases, and cancer using pharmaceutical approaches that selectively target senescent cells (ie, senolytics) or that suppress senescence-associated secretory phenotype (ie, senomorphics) holds great promise for the management of chronic age-associated conditions. Although studies have provided evidence that senolytics or senomorphics are effective at decreasing the number of senescent cells in humans, the short-term and long-term side-effects of these therapies are largely unknown. In this Review, we systematically discuss the senolytics and senomorphics that have been investigated in clinical trials or have been used off-label, presenting their various adverse effects. Despite the potential of senotherapeutics to transform anti-ageing medicine, a cautionary approach regarding unwanted dose-dependent side-effects should be adopted.
Collapse
Affiliation(s)
- Marco Raffaele
- International Clinical Research Center, St Anne's University Hospital, Brno, Czech Republic
| | - Manlio Vinciguerra
- International Clinical Research Center, St Anne's University Hospital, Brno, Czech Republic; Division of Medicine, University College London, London, UK; Research Institute of the Medical University of Varna, Varna, Bulgaria.
| |
Collapse
|
34
|
Lynch SM, Guo G, Gibson DS, Bjourson AJ, Rai TS. Role of Senescence and Aging in SARS-CoV-2 Infection and COVID-19 Disease. Cells 2021; 10:3367. [PMID: 34943875 PMCID: PMC8699414 DOI: 10.3390/cells10123367] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/17/2021] [Accepted: 11/23/2021] [Indexed: 02/07/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has resulted in a global pandemic associated with substantial morbidity and mortality worldwide, with particular risk for severe disease and mortality in the elderly population. SARS-CoV-2 infection is driven by a pathological hyperinflammatory response which results in a dysregulated immune response. Current advancements in aging research indicates that aging pathways have fundamental roles in dictating healthspan in addition to lifespan. Our review discusses the aging immune system and highlights that senescence and aging together, play a central role in COVID-19 pathogenesis. In our review, we primarily focus on the immune system response to SARS-CoV-2 infection, the interconnection between severe COVID-19, immunosenescence, aging, vaccination, and the emerging problem of Long-COVID. We hope to highlight the importance of identifying specific senescent endotypes (or "sendotypes"), which can used as determinants of COVID-19 severity and mortality. Indeed, identified sendotypes could be therapeutically exploited for therapeutic intervention. We highlight that senolytics, which eliminate senescent cells, can target aging-associated pathways and therefore are proving attractive as potential therapeutic options to alleviate symptoms, prevent severe infection, and reduce mortality burden in COVID-19 and thus ultimately enhance healthspan.
Collapse
Affiliation(s)
| | | | | | | | - Taranjit Singh Rai
- Northern Ireland Centre for Stratified Medicine, School of Biomedical Sciences, Ulster University, C-TRIC Building, Altnagelvin Area Hospital, Glenshane Road, Derry BT47 6SB, UK; (S.M.L.); (G.G.); (D.S.G.); (A.J.B.)
| |
Collapse
|
35
|
Kaempferol, Myricetin and Fisetin in Prostate and Bladder Cancer: A Systematic Review of the Literature. Nutrients 2021; 13:nu13113750. [PMID: 34836005 PMCID: PMC8621729 DOI: 10.3390/nu13113750] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/16/2021] [Accepted: 10/22/2021] [Indexed: 02/06/2023] Open
Abstract
Prostate and bladder cancer represent the two most frequently diagnosed genito-urinary malignancies. Diet has been implicated in both prostate and bladder cancer. Given their prolonged latency and high prevalence rates, both prostate and bladder cancer represent attractive candidates for dietary preventive measures, including the use of nutritional supplements. Flavonols, a class of flavonoids, are commonly found in fruit and vegetables and are known for their protective effect against diabetes and cardiovascular diseases. Furthermore, a higher dietary intake of flavonols was associated with a lower risk of both bladder and prostate cancer in epidemiological studies. In this systematic review, we gathered all available evidence supporting the anti-cancer potential of selected flavonols (kaempferol, fisetin and myricetin) against bladder and prostate cancer. A total of 21, 15 and 7 pre-clinical articles on bladder or prostate cancer reporting on kaempferol, fisetin and myricetin, respectively, were found, while more limited evidence was available from animal models and epidemiological studies or clinical trials. In conclusion, the available evidence supports the potential use of these flavonols in prostate and bladder cancer, with a low expected toxicity, thus providing the rationale for clinical trials that explore dosing, settings for clinical use as well as their use in combination with other pharmacological and non-pharmacological interventions.
Collapse
|