1
|
Kim HS, Jung CH. Impacts of Senolytic Phytochemicals on Gut Microbiota: A Comprehensive Review. J Microbiol Biotechnol 2024; 34:2166-2172. [PMID: 39603836 PMCID: PMC11637817 DOI: 10.4014/jmb.2408.08032] [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: 08/19/2024] [Revised: 10/21/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024]
Abstract
There is increasing interest in utilizing senolytics to selectively remove senescent cells from intestinal tissues, with the aim of maintaining a healthy gut environment during aging. This strategy underscores the potential of senolytics to enhance gut health by delaying intestinal aging and positively modulating gut microbiota. Certain plant-based phytochemicals have demonstrated promising senolytic effects. Beyond their ability to eliminate senescent cells, these compounds also exhibit antioxidant and anti-inflammatory properties, reducing oxidative stress and inflammation-key drivers of age-related diseases. By selectively removing senescent cells from the intestine, senolytic phytochemicals contribute to an improved intestinal inflammatory environment and promote the growth of a diverse microbial community. Ultimately, the dietary intake of these senolytic phytochemicals aids in maintaining a healthier intestinal microenvironment by targeting and clearing aged enterocytes.
Collapse
Affiliation(s)
- Hee Soo Kim
- Aging and Metabolism Research Group, Korea Food Research Institute, Wanju-gun, Jeollabuk-do 55365, Republic of Korea
- Department of Food Biotechnology, University of Science and Technology, Wanju-gun, Jeollabuk-do 55365, Republic of Korea
| | - Chang Hwa Jung
- Aging and Metabolism Research Group, Korea Food Research Institute, Wanju-gun, Jeollabuk-do 55365, Republic of Korea
- Department of Food Biotechnology, University of Science and Technology, Wanju-gun, Jeollabuk-do 55365, Republic of Korea
| |
Collapse
|
2
|
Imb M, Véghelyi Z, Maurer M, Kühnel H. Exploring Senolytic and Senomorphic Properties of Medicinal Plants for Anti-Aging Therapies. Int J Mol Sci 2024; 25:10419. [PMID: 39408750 PMCID: PMC11476546 DOI: 10.3390/ijms251910419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/09/2024] [Accepted: 09/21/2024] [Indexed: 10/20/2024] Open
Abstract
Senolytic and senomorphic therapies have gained more and more attention in the last decade. This kind of therapy is based on the killing of cellular senescent cells without harming the "normal" cells. Aging is not a disease. Clinical studies on healthy people will be difficult to conduct. Therefore, one possibility is to draw on the large repertoire of medicinal plants and use their senolytic properties to provide mild anti-aging therapies. Chamomile, goldenrod, reishi, and green tea were tested for their ability to trigger senolysis. Quercetin was used as control substance. Cellular senescence was induced with 25 µM etoposide in human dermal fibroblasts and established for at least 14 days. The plant extracts were tested for their antioxidant potential (DPPH assay) and their polyphenol content. Senolysis was determined by presto blue assay of young and etoposide-induced senescent cells, and SA-β-Gal assays were also performed. The senomorphic properties of the plants were investigated using IL-6 ELISA and qPCR. It turned out that chamomile triggers a kind of cytokine storm and causes the cytokine values in the ELISA and in the qPCR to rise extremely, and other senescence-associated phenotype (SASP) markers were also elevated. Goldenrod and quercetin tend to have a senolytic and senomorphic effect, respectively. Regarding the senolytic and senomorphic properties of herbs, we found that all tested herbs can have a senolytic effect, and a senomorphic effect of quercetin has also been discovered. With regard to the effect of chamomile, however, we can say that seemingly harmless tea products may have harmful effects, especially in combination with chemotherapy, at least in cell culture experiments. Nevertheless, inflammation is a double-bladed mechanism with positive effects, for example, in healing, but also known negative effects.
Collapse
Affiliation(s)
| | | | | | - Harald Kühnel
- Department of Applied Life Sciences, Bioengineering, University of Applied Sciences Campus Wien, Favoritenstraße 222, 1100 Vienna, Austria
| |
Collapse
|
3
|
Hense JD, Isola JVV, Garcia DN, Magalhães LS, Masternak MM, Stout MB, Schneider A. The role of cellular senescence in ovarian aging. NPJ AGING 2024; 10:35. [PMID: 39033161 PMCID: PMC11271274 DOI: 10.1038/s41514-024-00157-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 05/29/2024] [Indexed: 07/23/2024]
Abstract
This review explores the relationship between ovarian aging and senescent cell accumulation, as well as the efficacy of senolytics to improve reproductive longevity. Reproductive longevity is determined by the age-associated decline in ovarian reserve, resulting in reduced fertility and eventually menopause. Cellular senescence is a state of permanent cell cycle arrest and resistance to apoptosis. Senescent cells accumulate in several tissues with advancing age, thereby promoting chronic inflammation and age-related diseases. Ovaries also appear to accumulate senescent cells with age, which might contribute to aging of the reproductive system and whole organism through SASP production. Importantly, senolytic drugs can eliminate senescent cells and may present a potential intervention to mitigate ovarian aging. Herein, we review the current literature related to the efficacy of senolytic drugs for extending the reproductive window in mice.
Collapse
Affiliation(s)
- Jéssica D Hense
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Nutrition College, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - José V V Isola
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Driele N Garcia
- Nutrition College, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | | | - Michal M Masternak
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| | - Michael B Stout
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Augusto Schneider
- Nutrition College, Universidade Federal de Pelotas, Pelotas, RS, Brazil.
| |
Collapse
|
4
|
Tufail M, Huang YQ, Hu JJ, Liang J, He CY, Wan WD, Jiang CH, Wu H, Li N. Cellular Aging and Senescence in Cancer: A Holistic Review of Cellular Fate Determinants. Aging Dis 2024:AD.2024.0421. [PMID: 38913050 DOI: 10.14336/ad.2024.0421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 05/21/2024] [Indexed: 06/25/2024] Open
Abstract
This comprehensive review navigates the complex relationship between cellular aging, senescence, and cancer, unraveling the determinants of cellular fate. Beginning with an overview of cellular aging's significance in cancer, the review explores processes, changes, and molecular pathways influencing senescence. The review explores senescence as a dual mechanism in cancer, acting as a suppressor and contributor, focusing on its impact on therapy response. This review highlights opportunities for cancer therapies that target cellular senescence. The review further examines the senescence-associated secretory phenotype and strategies to modulate cellular aging to influence tumor behavior. Additionally, the review highlights the mechanisms of senescence escape in aging and cancer cells, emphasizing their impact on cancer prognosis and resistance to therapy. The article addresses current advances, unexplored aspects, and future perspectives in understanding cellular aging and senescence in cancer.
Collapse
Affiliation(s)
- Muhammad Tufail
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Yu-Qi Huang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Jia-Ju Hu
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Liang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Cai-Yun He
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Wen-Dong Wan
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
| | - Can-Hua Jiang
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hong Wu
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, 410083, China
| | - Ning Li
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Oral Precancerous Lesions, Central South University, Changsha, China
- Research Center of Oral and Maxillofacial Tumor, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
5
|
Snijckers RPM, Foks AC. Adaptive immunity and atherosclerosis: aging at its crossroads. Front Immunol 2024; 15:1350471. [PMID: 38686373 PMCID: PMC11056569 DOI: 10.3389/fimmu.2024.1350471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 03/28/2024] [Indexed: 05/02/2024] Open
Abstract
Adaptive immunity plays a profound role in atherosclerosis pathogenesis by regulating antigen-specific responses, inflammatory signaling and antibody production. However, as we age, our immune system undergoes a gradual functional decline, a phenomenon termed "immunosenescence". This decline is characterized by a reduction in proliferative naïve B- and T cells, decreased B- and T cell receptor repertoire and a pro-inflammatory senescence associated secretory profile. Furthermore, aging affects germinal center responses and deteriorates secondary lymphoid organ function and structure, leading to impaired T-B cell dynamics and increased autoantibody production. In this review, we will dissect the impact of aging on adaptive immunity and the role played by age-associated B- and T cells in atherosclerosis pathogenesis, emphasizing the need for interventions that target age-related immune dysfunction to reduce cardiovascular disease risk.
Collapse
Affiliation(s)
| | - Amanda C. Foks
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| |
Collapse
|
6
|
Dennery PA, Yao H. Emerging role of cellular senescence in normal lung development and perinatal lung injury. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:10-16. [PMID: 38567372 PMCID: PMC10987039 DOI: 10.1016/j.pccm.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Cellular senescence is a status of irreversible growth arrest, which can be triggered by the p53/p21cip1 and p16INK4/Rb pathways via intrinsic and external factors. Senescent cells are typically enlarged and flattened, and characterized by numerous molecular features. The latter consists of increased surfaceome, increased residual lysosomal activity at pH 6.0 (manifested by increased activity of senescence-associated beta-galactosidase [SA-β-gal]), senescence-associated mitochondrial dysfunction, cytoplasmic chromatin fragment, nuclear lamin b1 exclusion, telomere-associated foci, and the senescence-associated secretory phenotype. These features vary depending on the stressor leading to senescence and the type of senescence. Cellular senescence plays pivotal roles in organismal aging and in the pathogenesis of aging-related diseases. Interestingly, senescence can also both promote and inhibit wound healing processes. We recently report that senescence as a programmed process contributes to normal lung development. Lung senescence is also observed in Down Syndrome, as well as in premature infants with bronchopulmonary dysplasia and in a hyperoxia-induced rodent model of this disease. Furthermore, this senescence results in neonatal lung injury. In this review, we briefly discuss the molecular features of senescence. We then focus on the emerging role of senescence in normal lung development and in the pathogenesis of bronchopulmonary dysplasia as well as putative signaling pathways driving senescence. Finally, we discuss potential therapeutic approaches targeting senescent cells to prevent perinatal lung diseases.
Collapse
Affiliation(s)
- Phyllis A. Dennery
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI 02912, USA
- Department of Pediatrics, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Hongwei Yao
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI 02912, USA
| |
Collapse
|
7
|
Rad AN, Grillari J. Current senolytics: Mode of action, efficacy and limitations, and their future. Mech Ageing Dev 2024; 217:111888. [PMID: 38040344 DOI: 10.1016/j.mad.2023.111888] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/11/2023] [Accepted: 11/21/2023] [Indexed: 12/03/2023]
Abstract
Senescence is a cellular state characterized by its near-permanent halted cell cycle and distinct secretory phenotype. Although senescent cells have a variety of beneficial physiological functions, progressive accumulation of these cells due to aging or other conditions has been widely shown to provoke deleterious effects on the normal functioning of the same or higher-level biological organizations. Recently, erasing senescent cells in vivo, using senolytics, could ameliorate diseases identified with an elevated number of senescent cells. Since then, researchers have struggled to develop new senolytics each with different selectivity and potency. In this review, we have gathered and classified the proposed senolytics and discussed their mechanisms of action. Moreover, we highlight the heterogeneity of senolytics regarding their effect sizes, and cell type specificity as well as comment on the exploited strategies to improve these features. Finally, we suggest some prospective routes for the novel methods for ablation of senescent cells.
Collapse
Affiliation(s)
- Amirhossein Nayeri Rad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, P.O. Box 71468-64685, Shiraz, Iran.
| | - Johannes Grillari
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Donaueschingenstraße 13, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria; Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences Vienna, Muthgasse 18, 1190 Vienna, Austria.
| |
Collapse
|
8
|
Tagrida M, Palamae S, Saetang J, Ma L, Hong H, Benjakul S. Comparative Study of Quercetin and Hyperoside: Antimicrobial Potential towards Food Spoilage Bacteria, Mode of Action and Molecular Docking. Foods 2023; 12:4051. [PMID: 38002109 PMCID: PMC10670185 DOI: 10.3390/foods12224051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 11/26/2023] Open
Abstract
The antibacterial activities of quercetin and hyperoside were evaluated towards two major spoilage bacteria in fish, Pseudomonas aeruginosa (PA) and Shewanella putrefaciens (SP). Hyperoside showed a lower minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) towards both spoilage bacteria, PA and SP, than quercetin. Cell membrane morphology was affected when treated with hyperoside and quercetin. The release of content from the treated cells occurred, as ascertained by the release of potassium and magnesium ions and the increase in conductivity of the culture media. The morphology of cells was significantly changed, in which shrinkage and pores were obtained, when observed using SEM. Both compounds negatively affected the motility, both swimming and swarming, and the formation of extracellular polymeric substance (EPS), thus confirming antibiofilm activities. Agarose gel analysis revealed that both compounds could bind to or degrade the genomic DNA of both bacteria, thereby causing bacterial death. Molecular docking indicated that the compounds interacted with the minor groove of the DNA, favoring the adenine-thymine-rich regions. Thus, both quercetin and hyperoside could serve as potential antimicrobial agents to retard the spoilage of fish or perishable products.
Collapse
Affiliation(s)
- Mohamed Tagrida
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro-Industry, Prince of Songkla University, Hat Yai 90110, Songkhla, Thailand; (M.T.); (S.P.); (J.S.)
| | - Suriya Palamae
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro-Industry, Prince of Songkla University, Hat Yai 90110, Songkhla, Thailand; (M.T.); (S.P.); (J.S.)
| | - Jirakrit Saetang
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro-Industry, Prince of Songkla University, Hat Yai 90110, Songkhla, Thailand; (M.T.); (S.P.); (J.S.)
| | - Lukai Ma
- Key Laboratory of Green Processing and Intelligent Manufacturing of Lingnan Specialty Food of Ministry and Rural Affairs, College of Light Industry and Food, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China;
| | - Hui Hong
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China;
| | - Soottawat Benjakul
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro-Industry, Prince of Songkla University, Hat Yai 90110, Songkhla, Thailand; (M.T.); (S.P.); (J.S.)
- Department of Food and Nutrition, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
9
|
Dańczak‐Pazdrowska A, Gornowicz‐Porowska J, Polańska A, Krajka‐Kuźniak V, Stawny M, Gostyńska A, Rubiś B, Nourredine S, Ashiqueali S, Schneider A, Tchkonia T, Wyles SP, Kirkland JL, Masternak MM. Cellular senescence in skin-related research: Targeted signaling pathways and naturally occurring therapeutic agents. Aging Cell 2023; 22:e13845. [PMID: 37042069 PMCID: PMC10265178 DOI: 10.1111/acel.13845] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 03/25/2023] [Accepted: 03/27/2023] [Indexed: 04/13/2023] Open
Abstract
Despite the growing interest by researchers into cellular senescence, a hallmark of cellular aging, its role in human skin remains equivocal. The skin is the largest and most accessible human organ, reacting to the external and internal environment. Hence, it is an organ of choice to investigate cellular senescence and to target root-cause aging processes using senolytic and senomorphic agents, including naturally occurring plant-based derivatives. This review presents different aspects of skin cellular senescence, from physiology to pathology and signaling pathways. Cellular senescence can have both beneficial and detrimental effects on the skin, indicating that both prosenescent and antisenescent therapies may be desirable, based on the context. Knowledge of molecular mechanisms involved in skin cellular senescence may provide meaningful insights for developing effective therapeutics for senescence-related skin disorders, such as wound healing and cosmetic skin aging changes.
Collapse
Affiliation(s)
| | - Justyna Gornowicz‐Porowska
- Department and Division of Practical Cosmetology and Skin Diseases ProphylaxisPoznan University of Medical SciencesPoznanPoland
| | - Adriana Polańska
- Department of Dermatology and VenereologyPoznan University of Medical SciencesPoznanPoland
| | | | - Maciej Stawny
- Department of Pharmaceutical ChemistryPoznan University of Medical SciencesPoznanPoland
| | - Aleksandra Gostyńska
- Department of Pharmaceutical ChemistryPoznan University of Medical SciencesPoznanPoland
| | - Błażej Rubiś
- Department of Clinical Chemistry and Molecular DiagnosticsPoznan University of Medical SciencesPoznanPoland
| | - Sarah Nourredine
- Burnett School of Biomedical SciencesCollege of Medicine, University of Central FloridaOrlandoFloridaUSA
| | - Sarah Ashiqueali
- Burnett School of Biomedical SciencesCollege of Medicine, University of Central FloridaOrlandoFloridaUSA
| | | | - Tamara Tchkonia
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| | | | - James L. Kirkland
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| | - Michal M. Masternak
- Burnett School of Biomedical SciencesCollege of Medicine, University of Central FloridaOrlandoFloridaUSA
- Department of Head and Neck SurgeryPoznan University of Medical SciencesPoznanPoland
| |
Collapse
|
10
|
Abdelgawad IY, Agostinucci K, Sadaf B, Grant MKO, Zordoky BN. Metformin mitigates SASP secretion and LPS-triggered hyper-inflammation in Doxorubicin-induced senescent endothelial cells. FRONTIERS IN AGING 2023; 4:1170434. [PMID: 37168843 PMCID: PMC10164964 DOI: 10.3389/fragi.2023.1170434] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/13/2023] [Indexed: 05/13/2023]
Abstract
Introduction: Doxorubicin (DOX), a chemotherapeutic drug, induces senescence and increases the secretion of senescence-associated secretory phenotype (SASP) in endothelial cells (ECs), which contributes to DOX-induced inflammaging. Metformin, an anti-diabetic drug, demonstrates senomorphic effects on different models of senescence. However, the effects of metformin on DOX-induced endothelial senescence have not been reported before. Senescent ECs exhibit a hyper-inflammatory response to lipopolysachharide (LPS). Therefore, in our current work, we identified the effects of metformin on DOX-induced endothelial senescence and LPS-induced hyper-inflammation in senescent ECs. Methods: ECs were treated with DOX ± metformin for 24 h followed by 72 h incubation without DOX to establish senescence. Effects of metformin on senescence markers expression, SA-β-gal activity, and SASP secretion were assessed. To delineate the molecular mechanisms, the effects of metformin on major signaling pathways were determined. The effect of LPS ± metformin was determined by stimulating both senescent and non-senescent ECs with LPS for an additional 24 h. Results: Metformin corrected DOX-induced upregulation of senescence markers and decreased the secretion of SASP factors and adhesion molecules. These effects were associated with a significant inhibition of the JNK and NF-κB pathway. A significant hyper-inflammatory response to LPS was observed in DOX-induced senescent ECs compared to non-senescent ECs. Metformin blunted LPS-induced upregulation of pro-inflammatory SASP factors. Conclusion: Our study demonstrates that metformin mitigates DOX-induced endothelial senescence phenotype and ameliorates the hyper-inflammatory response to LPS. These findings suggest that metformin may protect against DOX-induced vascular aging and endothelial dysfunction and ameliorate infection-induced hyper-inflammation in DOX-treated cancer survivors.
Collapse
Affiliation(s)
| | | | | | | | - Beshay N. Zordoky
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN, United States
| |
Collapse
|
11
|
Admasu TD, Kim K, Rae M, Avelar R, Gonciarz RL, Rebbaa A, Pedro de Magalhães J, Renslo AR, Stolzing A, Sharma A. Selective ablation of primary and paracrine senescent cells by targeting iron dyshomeostasis. Cell Rep 2023; 42:112058. [PMID: 36753419 DOI: 10.1016/j.celrep.2023.112058] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 11/29/2022] [Accepted: 01/17/2023] [Indexed: 02/08/2023] Open
Abstract
Senescent cells can spread the senescent phenotype to other cells by secreting senescence-associated secretory phenotype factors. The resulting paracrine senescent cells make a significant contribution to the burden of senescent cell accumulation with age. Previous efforts made to characterize paracrine senescence are unreliable due to analyses being based on mixed populations of senescent and non-senescent cells. Here, we use dipeptidyl peptidase-4 (DPP4) as a surface maker to isolate senescent cells from mixed populations. Using this technique, we enrich the percentage of paracrine senescence from 40% to 85%. We then use this enriched culture to characterize DPP4+ primary and paracrine senescent cells. We observe ferroptosis dysregulation and ferrous iron accumulation as a common phenomenon in both primary and paracrine senescent cells. Finally, we identify ferroptosis induction and ferrous iron-activatable prodrug as a broad-spectrum senolytic approach to ablate multiple types of primary and paracrine senescent cells.
Collapse
Affiliation(s)
| | - Kristie Kim
- SENS Research Foundation, Mountain View, CA 94041, USA
| | - Michael Rae
- SENS Research Foundation, Mountain View, CA 94041, USA
| | - Roberto Avelar
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Ryan L Gonciarz
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | | | - João Pedro de Magalhães
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Adam R Renslo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Alexandra Stolzing
- Loughborough University, Centre for Biological Engineering, School of Mechanical, Electrical and Manufacturing Engineering, Epinal Way, Loughborough LE113TU, UK
| | - Amit Sharma
- SENS Research Foundation, Mountain View, CA 94041, USA.
| |
Collapse
|
12
|
Bone-targeted delivery of senolytics to eliminate senescent cells increases bone formation in senile osteoporosis. Acta Biomater 2023; 157:352-366. [PMID: 36470392 DOI: 10.1016/j.actbio.2022.11.056] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 10/31/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022]
Abstract
Systemic elimination of senescent cells using senolytic drugs presents therapeutic effects on age-related diseases, including senile osteoporosis. However, low bioavailability and potential side effects of senolytics restrict clinical application. Therefore, we developed a bone-targeted delivery system for senolytics to effective treatment of senile osteoporosis. In this study, quercetin was screened out as the ideal senolytics for eliminating senescent BMSCs. Treatment of quercetin efficiently decreased the senescence markers in senescent BMSCs models. After treatment with quercetin in vitro, cell mitosis and calcification staining assay confirmed that the proliferation and osteogenesis of the senescent BMSCs populations were enhanced. To enhance the effectiveness and minimize the side effect of treatment, liposomes decorated with bone affinity peptide (DSS)6 were constructed for bone-targeted delivery of quercetin. After administration of liposomes loading quercetin in two aged mice models, histological and cellular analysis confirmed that bone-targeted treatment with quercetin efficiently eliminated senescent cells in bone, restored the function of BMCSs, and promoted bone formation in aged mice models when compared to non-targeted treatment. Taken together, the bone-targeted delivery of senolytics efficiently eliminates senescent cells to recover bone mass and microarchitecture, showing an effective treatment for senile osteoporosis. STATEMENT OF SIGNIFICANCE: Senile osteoporosis, a common and hazardous chronic disease, has been still lacking effective therapy. How to effectively eliminate the hazards of senescent cells in skeleton to bone formation remains challenge. In this study, quercetin was screened out as the ideal senolytic drug for senescent BMSCs and could effectively eliminated senescent BMSCs to restore the cellular functions of senescent BMSCs models in vitro. Then, the bone-targeted liposomes were designed to encapsulate and deliver senolytics efficiently to senile bone tissue. Based on two aged mice models, we confirmed that bone-targeted delivery of quercetin efficiently eliminated senescent cells in skeleton and enhanced bone formation in vivo, suggesting the bone-targeted elimination of senescent cells is an effective treatment for senile osteoporosis.
Collapse
|
13
|
Endothelial senescence in vascular diseases: current understanding and future opportunities in senotherapeutics. Exp Mol Med 2023; 55:1-12. [PMID: 36599934 PMCID: PMC9898542 DOI: 10.1038/s12276-022-00906-w] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/25/2022] [Accepted: 10/31/2022] [Indexed: 01/05/2023] Open
Abstract
Senescence compromises the essential role that the endothelium plays in maintaining vascular homeostasis, so promoting endothelial dysfunction and the development of age-related vascular diseases. Their biological and clinical significance calls for strategies for identifying and therapeutically targeting senescent endothelial cells. While senescence and endothelial dysfunction have been studied extensively, distinguishing what is distinctly endothelial senescence remains a barrier to overcome for an effective approach to addressing it. Here, we review the mechanisms underlying endothelial senescence and the evidence for its clinical importance. Furthermore, we discuss the current state and the limitations in the approaches for the detection and therapeutic intervention of target cells, suggesting potential directions for future research.
Collapse
|
14
|
Al-Azab M, Safi M, Idiiatullina E, Al-Shaebi F, Zaky MY. Aging of mesenchymal stem cell: machinery, markers, and strategies of fighting. Cell Mol Biol Lett 2022; 27:69. [PMID: 35986247 PMCID: PMC9388978 DOI: 10.1186/s11658-022-00366-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 07/18/2022] [Indexed: 02/08/2023] Open
Abstract
Human mesenchymal stem cells (MSCs) are primary multipotent cells capable of differentiating into osteocytes, chondrocytes, and adipocytes when stimulated under appropriate conditions. The role of MSCs in tissue homeostasis, aging-related diseases, and cellular therapy is clinically suggested. As aging is a universal problem that has large socioeconomic effects, an improved understanding of the concepts of aging can direct public policies that reduce its adverse impacts on the healthcare system and humanity. Several studies of aging have been carried out over several years to understand the phenomenon and different factors affecting human aging. A reduced ability of adult stem cell populations to reproduce and regenerate is one of the main contributors to the human aging process. In this context, MSCs senescence is a major challenge in front of cellular therapy advancement. Many factors, ranging from genetic and metabolic pathways to extrinsic factors through various cellular signaling pathways, are involved in regulating the mechanism of MSC senescence. To better understand and reverse cellular senescence, this review highlights the underlying mechanisms and signs of MSC cellular senescence, and discusses the strategies to combat aging and cellular senescence.
Collapse
|
15
|
Pharmacological Activity of Quercetin: An Updated Review. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:3997190. [PMID: 36506811 PMCID: PMC9731755 DOI: 10.1155/2022/3997190] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/12/2022] [Accepted: 11/21/2022] [Indexed: 12/04/2022]
Abstract
Quercetin, a natural flavonoid compound with a widespread occurrence throughout the plant kingdom, exhibits a variety of pharmacological activities. Because of the wide spectrum of health-promoting effects, quercetin has attracted much attention of dietitians and medicinal chemists. An updated review of the literature on quercetin was performed using PubMed, Embase, and Science Direct databases. This article presents an overview of recent developments in pharmacological activities of quercetin including anti-SARS-CoV-2, antioxidant, anticancer, antiaging, antiviral, and anti-inflammatory activities as well as the mechanism of actions involved. The biological activities of quercetin were evaluated both in vitro and in vivo, involving a number of cell lines and animal models, but metabolic mechanisms of quercetin in the human body are not clear. Therefore, further large sample clinical studies are needed to determine the appropriate dosage and form of quercetin for the treatment of the disease.
Collapse
|
16
|
Abstract
SUMMARY Skin aging is an outward manifestation of other cellular and molecular aging processes occurring elsewhere in the body. These processes are known collectively as the "hallmarks" of aging, which are a series of basic health maintenance mechanisms that fail over time. Cellular senescence is one of the most studied of the hallmarks of aging; senescent cells accumulate over time and are major drives of the aging process. Here, we discuss the impact of cellular senescence in the context of skin aging, and discuss the emerging landscape of interventions designed for their selective removal by targeted cell death (senolytics) or rejuvenation (senomorphics). We discuss the serotherapeutic strategies that are currently under investigation for systemic aging, which may bring eventual benefits for skin health. Next, we discuss a newly discovered hallmark of aging, dysregulated mRNA processing, which can be targeted for the senomorphic effect. Finally, we highlight a new modality for manipulation of disrupted mRNA processing, oligonucleotide therapeutics. The emerging field of senotherapeutics is set to revolutionize how we view and treat skin aging, and senotherapies are now poised to become a new class of skincare interventions.
Collapse
|
17
|
Pérez-Arancibia R, Ordoñez JL, Rivas A, Pihán P, Sagredo A, Ahumada U, Barriga A, Seguel I, Cárdenas C, Vidal RL, Hetz C, Delporte C. A phenolic-rich extract from Ugni molinae berries reduces abnormal protein aggregation in a cellular model of Huntington's disease. PLoS One 2021; 16:e0254834. [PMID: 34324551 PMCID: PMC8320977 DOI: 10.1371/journal.pone.0254834] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 07/02/2021] [Indexed: 12/21/2022] Open
Abstract
Accumulation of misfolded proteins in the brain is a common hallmark of most age-related neurodegenerative diseases. Previous studies from our group identified the presence of anti-inflammatory and antioxidant compounds in leaves derived from the Chilean berry Ugni molinae (murtilla), in addition to show a potent anti-aggregation activity in models of Alzheimer´s disease. However, possible beneficial effects of berry extracts of murtilla was not investigated. Here we evaluated the efficacy of fruit extracts from different genotypes of Chilean-native U. molinae on reducing protein aggregation using cellular models of Huntington´s disease and assess the correlation with their chemical composition. Berry extraction was performed by exhaustive maceration with increasing-polarity solvents. An unbiased automatic microscopy platform was used for cytotoxicity and protein aggregation studies in HEK293 cells using polyglutamine-EGFP fusion proteins, followed by secondary validation using biochemical assays. Phenolic-rich extracts from murtilla berries of the 19-1 genotype (ETE 19-1) significantly reduced polyglutamine peptide aggregation levels, correlating with the modulation in the expression levels of autophagy-related proteins. Using LC-MS and molecular network analysis we correlated the presence of flavonoids, phenolic acids, and ellagitannins with the protective effects of ETE 19-1 effects on protein aggregation. Overall, our results indicate the presence of bioactive components in ethanolic extracts from U. molinae berries that reduce the load of protein aggregates in living cells.
Collapse
Affiliation(s)
- Rodrigo Pérez-Arancibia
- Laboratorio de Productos Naturales, Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
- Instituto de Neurociencia Biomédica (BNI), Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Centro FONDAP de Gerociencia, Salud Mental y Metabolismo (GERO), Santiago, Chile
- Centro de Biología Integrativa, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Jose Luis Ordoñez
- Laboratorio de Productos Naturales, Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
- Laboratorio de Química Inorgánica y Analítica, Departamento de Química Inorgánica y Analítica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Alexis Rivas
- Instituto de Neurociencia Biomédica (BNI), Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Centro FONDAP de Gerociencia, Salud Mental y Metabolismo (GERO), Santiago, Chile
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Philippe Pihán
- Instituto de Neurociencia Biomédica (BNI), Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Centro FONDAP de Gerociencia, Salud Mental y Metabolismo (GERO), Santiago, Chile
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Alfredo Sagredo
- Instituto de Neurociencia Biomédica (BNI), Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Centro FONDAP de Gerociencia, Salud Mental y Metabolismo (GERO), Santiago, Chile
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Ulises Ahumada
- Centro FONDAP de Gerociencia, Salud Mental y Metabolismo (GERO), Santiago, Chile
- Centro de Biología Integrativa, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Andrés Barriga
- Unidad de Espectrometría de Masas, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Ivette Seguel
- Laboratorio de Productos Naturales, Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - César Cárdenas
- Centro FONDAP de Gerociencia, Salud Mental y Metabolismo (GERO), Santiago, Chile
- Centro de Biología Integrativa, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, CA, United States of America
- Buck Institute for Research on Aging, Novato, California, United States of America
| | - Rene L. Vidal
- Instituto de Neurociencia Biomédica (BNI), Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Centro FONDAP de Gerociencia, Salud Mental y Metabolismo (GERO), Santiago, Chile
- Centro de Biología Integrativa, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Claudio Hetz
- Instituto de Neurociencia Biomédica (BNI), Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Centro FONDAP de Gerociencia, Salud Mental y Metabolismo (GERO), Santiago, Chile
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
- Buck Institute for Research on Aging, Novato, California, United States of America
| | - Carla Delporte
- Laboratorio de Productos Naturales, Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| |
Collapse
|
18
|
Chondroprotection and Molecular Mechanism of Action of Phytonutraceuticals on Osteoarthritis. Molecules 2021; 26:molecules26082391. [PMID: 33924083 PMCID: PMC8074261 DOI: 10.3390/molecules26082391] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/16/2021] [Accepted: 04/17/2021] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease and an important cause of incapacitation. There is a lack of drugs and effective treatments that stop or slow the OA progression. Modern pharmacological treatments, such as analgesics, have analgesic effects but do not affect the course of OA. Long-term use of these drugs can lead to serious side effects. Given the OA nature, it is likely that lifelong treatment will be required to stop or slow its progression. Therefore, there is an urgent need for disease-modifying OA treatments that are also safe for clinical use over long periods. Phytonutraceuticals are herbal products that provide a therapeutic effect, including disease prevention, which not only have favorable safety characteristics but may have an alleviating effect on the OA and its symptoms. An estimated 47% of OA patients use alternative drugs, including phytonutraceuticals. The review studies the efficacy and action mechanism of widely used phytonutraceuticals, analyzes the available experimental and clinical data on the effect of some phytonutraceuticals (phytoflavonoids, polyphenols, and bioflavonoids) on OA, and examines the known molecular effect and the possibility of their use for chondroprotection.
Collapse
|
19
|
Dagher O, Mury P, Thorin-Trescases N, Noly PE, Thorin E, Carrier M. Therapeutic Potential of Quercetin to Alleviate Endothelial Dysfunction in Age-Related Cardiovascular Diseases. Front Cardiovasc Med 2021; 8:658400. [PMID: 33860002 PMCID: PMC8042157 DOI: 10.3389/fcvm.2021.658400] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/05/2021] [Indexed: 12/12/2022] Open
Abstract
The vascular endothelium occupies a catalog of functions that contribute to the homeostasis of the cardiovascular system. It is a physically active barrier between circulating blood and tissue, a regulator of the vascular tone, a biochemical processor and a modulator of coagulation, inflammation, and immunity. Given these essential roles, it comes to no surprise that endothelial dysfunction is prodromal to chronic age-related diseases of the heart and arteries, globally termed cardiovascular diseases (CVD). An example would be ischemic heart disease (IHD), which is the main cause of death from CVD. We have made phenomenal advances in treating CVD, but the aging endothelium, as it senesces, always seems to out-run the benefits of medical and surgical therapies. Remarkably, many epidemiological studies have detected a correlation between a flavonoid-rich diet and a lower incidence of mortality from CVD. Quercetin, a member of the flavonoid class, is a natural compound ubiquitously found in various food sources such as fruits, vegetables, seeds, nuts, and wine. It has been reported to have a wide range of health promoting effects and has gained significant attention over the years. A growing body of evidence suggests quercetin could lower the risk of IHD by mitigating endothelial dysfunction and its risk factors, such as hypertension, atherosclerosis, accumulation of senescent endothelial cells, and endothelial-mesenchymal transition (EndoMT). In this review, we will explore these pathophysiological cascades and their interrelation with endothelial dysfunction. We will then present the scientific evidence to quercetin's anti-atherosclerotic, anti-hypertensive, senolytic, and anti-EndoMT effects. Finally, we will discuss the prospect for its clinical use in alleviating myocardial ischemic injuries in IHD.
Collapse
Affiliation(s)
- Olina Dagher
- Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Surgery, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Center for Research, Montreal Heart Institute, Montreal, QC, Canada
| | - Pauline Mury
- Center for Research, Montreal Heart Institute, Montreal, QC, Canada
| | | | - Pierre Emmanuel Noly
- Department of Surgery, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Center for Research, Montreal Heart Institute, Montreal, QC, Canada
| | - Eric Thorin
- Department of Surgery, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Center for Research, Montreal Heart Institute, Montreal, QC, Canada
| | - Michel Carrier
- Department of Surgery, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Center for Research, Montreal Heart Institute, Montreal, QC, Canada
| |
Collapse
|
20
|
Targeting senescent cell clearance: An approach to delay aging and age-associated disorders. TRANSLATIONAL MEDICINE OF AGING 2021. [DOI: 10.1016/j.tma.2020.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
21
|
The use of geroprotectors to prevent multimorbidity: Opportunities and challenges. Mech Ageing Dev 2020; 193:111391. [PMID: 33144142 DOI: 10.1016/j.mad.2020.111391] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 12/14/2022]
Abstract
Over 60 % of people over the age of 65 will suffer from multiple diseases concomitantly but the common approach is to treat each disease separately. As age-associated diseases have common underlying mechanisms there is potential to tackle many diseases with the same pharmacological intervention. These are known as geroprotectors and could overcome the problems related to polypharmacy seen with the use of the single disease model. With some geroprotectors now reaching the end stage of preclinical studies and early clinical trials, there is a need to review the evidence and assess how they can be translated practically and effectively into routine practice. Despite promising evidence, there are many gaps and challenges in our understanding that must be addressed to make geroprotective medicine effective in the treatment of age-associated multimorbidity. Here we highlight the key barriers to clinical translation and discuss whether geroprotectors such as metformin, rapamycin and senolytics can tackle all age-associated diseases at the same dose, or whether a more nuanced approach is required. The evidence suggests that geroprotectors' mode of action may differ in different tissues or in response to different inducers of accelerating ageing, suggesting that a blunt 'one drug for many diseases' approach may not work. We make the case for the use of artificial intelligence to better understand multimorbidity, allowing identification of clusters and networks of diseases that are significantly associated beyond chance and the underpinning molecular pathway of ageing causal to each cluster. This will allow us to better understand the development of multimorbidity, select a more homogenous group of patients for intervention, match them with the appropriate geroprotector and identify biomarkers specific to the cluster.
Collapse
|
22
|
Cherif H, Bisson DG, Mannarino M, Rabau O, Ouellet JA, Haglund L. Senotherapeutic drugs for human intervertebral disc degeneration and low back pain. eLife 2020; 9:54693. [PMID: 32821059 PMCID: PMC7442487 DOI: 10.7554/elife.54693] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 08/03/2020] [Indexed: 12/12/2022] Open
Abstract
Cellular senescence is a contributor to intervertebral disc (IVD) degeneration and low back pain. Here, we found that RG-7112, a potent mouse double-minute two protein inhibitor, selectively kills senescent IVD cells through apoptosis. Gene expression pathway analysis was used to compare the functional networks of genes affected by RG-7112, a pure synthetic senolytic with o-Vanillin a natural and anti-inflammatory senolytic. Both affected a functional gene network related to cell death and survival. O-Vanillin also affected networks related to cell cycle progression as well as connective tissue development and function. Both senolytics effectively decreased the senescence-associated secretory phenotype (SASP) of IVD cells. Furthermore, bioavailability and efficacy were verified ex vivo in the physiological environment of degenerating intact human discs where a single dose improved disc matrix homeostasis. Matrix improvement correlated with a reduction in senescent cells and SASP, supporting a translational potential of targeting senescent cells as a therapeutic intervention. Pain in the lower back affects about four in five people during their lifetime. Over time, the discs that provide cushioning between the vertebrae of the spine can degenerate, which can be one of the major causes of lower back pain. It has been shown that when the cells of these discs are exposed to different stress factors, they stop growing and become irreversibly dormant. Such ‘senescent’ cells release a range of proteins and small molecules that lead to painful inflammation and further degeneration of the discs. Moreover, it is thought that a high number of senescent cells may be linked to other degenerative diseases such as arthritis. Current treatments can only reduce the severity of the symptoms, but they cannot prevent the degeneration from progressing. Now, Cherif et al. set out to test the effects of two different compounds on human disc cells grown in the laboratory. One of the molecules studied, RG-7112, is a synthetic drug that has been approved for safety by the US Food and Drug Administration and has been shown to remove senescent cells. The other, o-Vanillin, is a natural compound that has anti-inflammatory and anti-senescence properties. The results showed that both compounds were able to trigger changes to that helped new, healthy cells to grow and at the same time kill senescent cells. They also reduced the production of molecules linked to inflammation and pain. Further analyses revealed that the compounds were able to strengthen the fibrous matrix that surrounds and supports the discs. Cherif et al. hope that this could form the basis for a new family of drugs for back pain to slow the degeneration of the discs and reduce pain. This may also have benefits for other similar degenerative diseases caused by cell senescence, such as arthritis.
Collapse
Affiliation(s)
- Hosni Cherif
- Orthopaedic Research Lab, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, Canada.,McGill Scoliosis and Spine Group, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Daniel G Bisson
- Orthopaedic Research Lab, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, Canada.,McGill Scoliosis and Spine Group, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Matthew Mannarino
- Orthopaedic Research Lab, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, Canada.,McGill Scoliosis and Spine Group, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Oded Rabau
- McGill Scoliosis and Spine Group, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, Canada.,Shriner's Hospital for Children, 1003 Decarie Blvd, Montreal, Canada
| | - Jean A Ouellet
- McGill Scoliosis and Spine Group, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, Canada.,Shriner's Hospital for Children, 1003 Decarie Blvd, Montreal, Canada
| | - Lisbet Haglund
- Orthopaedic Research Lab, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, Canada.,McGill Scoliosis and Spine Group, Department of Surgery, McGill University and the Research Institute of the McGill University Health Centre, Montreal, Canada.,Shriner's Hospital for Children, 1003 Decarie Blvd, Montreal, Canada
| |
Collapse
|
23
|
Protective Effects of Polyphenols Present in Mediterranean Diet on Endothelial Dysfunction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2097096. [PMID: 32831990 PMCID: PMC7428938 DOI: 10.1155/2020/2097096] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/02/2020] [Accepted: 07/18/2020] [Indexed: 12/13/2022]
Abstract
Endothelial dysfunction tends to be the initial indicator in proinflammatory state and macro- and microvascular complications, such as atherosclerosis and cardiovascular diseases. It has been shown that certain compounds in diet can generate beneficial effects on cardiovascular disease due to its interactions with endothelial cells. Thus, this review is aimed at investigating whether certain polyphenols present in the Mediterranean diet, specifically catechin, quercetin, resveratrol, and urolithin, could exert positive effects on endothelial dysfunction. After analysis of numerous papers, we found that polyphenols aiding endothelial function is beneficial not only for patients with cardiovascular disease, diabetes, or endothelial dysfunction but for all people as it can improve the effects of aging on the endothelia. The additional benefit of these polyphenols on weight loss further improves health and lowers the risk of several diseases, including those caused by endothelial dysfunction. However, it is important to note that the dosages in the majorities of the studies mentioned in this review were of supplemental rather than nutritionally relevant quantities, and therefore, the recommended dosages are difficult to determine.
Collapse
|
24
|
Liu T, Yang Q, Zhang X, Qin R, Shan W, Zhang H, Chen X. Quercetin alleviates kidney fibrosis by reducing renal tubular epithelial cell senescence through the SIRT1/PINK1/mitophagy axis. Life Sci 2020; 257:118116. [PMID: 32702447 DOI: 10.1016/j.lfs.2020.118116] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/06/2020] [Accepted: 07/14/2020] [Indexed: 12/11/2022]
Abstract
Emerging evidence implicates accelerated renal tubular epithelial cell (RTEC) senescence in renal fibrosis progression. Mitophagy protects against kidney injury. However, the mechanistic interplay between cell senescence and mitophagy in RTECs is not clearly defined. The purpose of this study was to evaluate the inhibition of RTEC senescence and renal fibrosis by quercetin and explore the underlying mechanisms. We found that quercetin attenuated RTEC senescence induced by angiotensin II (AngII) in vitro and unilateral ureteral obstruction in vivo. Moreover, we demonstrated that mitochondrial abnormalities such as elevated reactive oxygen species, decreased membrane potential, and fragmentation and accumulation of mitochondrial mass, occurred in AngII-treated RTECs. Quercetin treatment reversed these effects. Furthermore, quercetin enhanced mitophagy in AngII-treated RTECs, which was markedly reduced by treatment with mitophagy-specific inhibitors. Sirtuin-1 (SIRT1) was involved in quercetin-mediated PTEN-induced kinase 1 (PINK1)/Parkin-associated mitophagy activation. Pharmacological antagonism of SIRT1 in AngII-treated RTECs blocked the effects of quercetin on mitophagy and cellular senescence. Finally, quercetin alleviated kidney fibrosis by reducing RTEC senescence via mitophagy. Collectively, the antifibrotic effect of quercetin involved inhibition of RTEC senescence, possibly through activation of SIRT1/PINK1/Parkin-mediated mitophagy. These findings suggest that pharmacological elimination of senescent cells and stimulation of mitophagy represent effective therapeutic strategies to prevent kidney fibrosis.
Collapse
Affiliation(s)
- Tao Liu
- Department of Pharmacology, College of Pharmacy and Laboratory Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Qunfang Yang
- Department of Pharmacology, College of Pharmacy and Laboratory Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Xuan Zhang
- Department of Pharmacology, College of Pharmacy and Laboratory Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Rongxing Qin
- Department of Pharmacology, College of Pharmacy and Laboratory Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Wenjun Shan
- Department of Pharmacology, College of Pharmacy and Laboratory Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Haigang Zhang
- Department of Pharmacology, College of Pharmacy and Laboratory Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Xiaohong Chen
- Department of Pharmacology, College of Pharmacy and Laboratory Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| |
Collapse
|
25
|
Hughes MJ, McGettrick HM, Sapey E. Shared mechanisms of multimorbidity in COPD, atherosclerosis and type-2 diabetes: the neutrophil as a potential inflammatory target. Eur Respir Rev 2020; 29:190102. [PMID: 32198215 PMCID: PMC9488696 DOI: 10.1183/16000617.0102-2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 11/02/2019] [Indexed: 12/17/2022] Open
Abstract
Multimorbidity is increasingly common and current healthcare strategies are not always aligned to treat this complex burden of disease. COPD, type-2 diabetes mellitus (T2D) and cardiovascular disease, especially atherosclerosis, occur more frequently together than expected, even when risk factors such as smoking, obesity, inactivity and poverty are considered. This supports the possibility of unifying mechanisms that contribute to the pathogenesis or progression of each condition.Neutrophilic inflammation is causally associated with COPD, and increasingly recognised in the pathogenesis of atherosclerosis and T2D, potentially forming an aetiological link between conditions. This link might reflect an overspill of inflammation from one affected organ into the systemic circulation, exposing all organs to an increased milieu of proinflammatory cytokines. Additionally, increasing evidence supports the involvement of other processes in chronic disease pathogenesis, such as cellular senescence or changes in cellular phenotypes.This review explores the current scientific evidence for inflammation, cellular ageing and cellular processes, such as reactive oxygen species production and phenotypic changes in the pathogenesis of COPD, T2D and atherosclerosis; highlighting common mechanisms shared across these diseases. We identify emerging therapeutic approaches that target these areas, but also where more work is still required to improve our understanding of the underlying cellular biology in a multimorbid disease setting.
Collapse
Affiliation(s)
- Michael J Hughes
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Helen M McGettrick
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Elizabeth Sapey
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| |
Collapse
|
26
|
Abstract
Multiple interventions in the aging process have been discovered to extend the healthspan of model organisms. Both industry and academia are therefore exploring possible transformative molecules that target aging and age-associated diseases. In this overview, we summarize the presented talks and discussion points of the 5th Annual Aging and Drug Discovery Forum 2018 in Basel, Switzerland. Here academia and industry came together, to discuss the latest progress and issues in aging research. The meeting covered talks about the mechanistic cause of aging, how longevity signatures may be highly conserved, emerging biomarkers of aging, possible interventions in the aging process and the use of artificial intelligence for aging research and drug discovery. Importantly, a consensus is emerging both in industry and academia, that molecules able to intervene in the aging process may contain the potential to transform both societies and healthcare.
Collapse
|
27
|
|
28
|
Lewinska A, Adamczyk-Grochala J, Bloniarz D, Olszowka J, Kulpa-Greszta M, Litwinienko G, Tomaszewska A, Wnuk M, Pazik R. AMPK-mediated senolytic and senostatic activity of quercetin surface functionalized Fe 3O 4 nanoparticles during oxidant-induced senescence in human fibroblasts. Redox Biol 2019; 28:101337. [PMID: 31622846 PMCID: PMC6812309 DOI: 10.1016/j.redox.2019.101337] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 09/22/2019] [Accepted: 10/01/2019] [Indexed: 01/23/2023] Open
Abstract
Cellular senescence may contribute to aging and age-related diseases and senolytic drugs that selectively kill senescent cells may delay aging and promote healthspan. More recently, several categories of senolytics have been established, namely HSP90 inhibitors, Bcl-2 family inhibitors and natural compounds such as quercetin and fisetin. However, senolytic and senostatic potential of nanoparticles and surface-modified nanoparticles has never been addressed. In the present study, quercetin surface functionalized Fe3O4 nanoparticles (MNPQ) were synthesized and their senolytic and senostatic activity was evaluated during oxidative stress-induced senescence in human fibroblasts in vitro. MNPQ promoted AMPK activity that was accompanied by non-apoptotic cell death and decreased number of stress-induced senescent cells (senolytic action) and the suppression of senescence-associated proinflammatory response (decreased levels of secreted IL-8 and IFN-β, senostatic action). In summary, we have shown for the first time that MNPQ may be considered as promising candidates for senolytic- and senostatic-based anti-aging therapies. Quercetin surface functionalized magnetite nanoparticles (MNPQ) were synthesized. MNPQ eliminated hydrogen peroxide-induced senescent human fibroblasts. MNPQ limited senescence-associated proinflammatory responses. Senotherapeutic action of MNPQ was accompanied by increased activity of AMPK. MNPQ may be useful for senolytic- and senostatic-based anti-aging therapies.
Collapse
Affiliation(s)
- Anna Lewinska
- Department of Cell Biochemistry, Faculty of Biotechnology, University of Rzeszow, Pigonia 1, 35-310, Rzeszow, Poland
| | - Jagoda Adamczyk-Grochala
- Department of Cell Biochemistry, Faculty of Biotechnology, University of Rzeszow, Pigonia 1, 35-310, Rzeszow, Poland
| | - Dominika Bloniarz
- Department of Perinatology, Institute of Midwifery and Medical Emergency, Faculty of Medicine, University of Rzeszow, Pigonia 6, 35-310, Rzeszow, Poland
| | - Jakub Olszowka
- Department of Genetics, Faculty of Biotechnology, University of Rzeszow, Pigonia 1, 35-310, Rzeszow, Poland
| | - Magdalena Kulpa-Greszta
- Faculty of Chemistry, Rzeszow University of Technology, Powstancow Warszawy 12, 35-959, Rzeszow, Poland
| | | | - Anna Tomaszewska
- Department of Medicinal Chemistry and Nanomaterials, Faculty of Biotechnology, University of Rzeszow, Pigonia 1, 35-310, Rzeszow, Poland
| | - Maciej Wnuk
- Department of Genetics, Faculty of Biotechnology, University of Rzeszow, Pigonia 1, 35-310, Rzeszow, Poland.
| | - Robert Pazik
- Department of Medicinal Chemistry and Nanomaterials, Faculty of Biotechnology, University of Rzeszow, Pigonia 1, 35-310, Rzeszow, Poland.
| |
Collapse
|
29
|
Li W, Qin L, Feng R, Hu G, Sun H, He Y, Zhang R. Emerging senolytic agents derived from natural products. Mech Ageing Dev 2019; 181:1-6. [PMID: 31077707 DOI: 10.1016/j.mad.2019.05.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/05/2019] [Accepted: 05/07/2019] [Indexed: 01/10/2023]
Abstract
Cellular senescence is a hallmark of aging, it is a permanent state of cell cycle arrest induced by cellular stresses. During the aging process, senescent cells (SCs) increasingly accumulate in tissues, causing a loss of tissue-repair capacity because of cell cycle arrest in progenitor cells and produce proinflammatory and matrix-degrading molecules which are known as the senescence-associated secretory phenotype (SASP), and thereby contribute to the development of various age-related diseases. Genetic evidence has demonstrated that clearance of SCs can delay aging and extend healthspan. Senolytics, small molecules that can selectively kill SCs, have been developed to treat various age-related diseases. In recent years, emerging natural compounds have been discovered to be effective senolytic agents, such as quercetin, fisetin, piperlongumine and the curcumin analog. Some of the compounds have been validated in animal models and have great potential to be pushed to clinical applications. In this review, we will discuss cellular senescence and its potential as a target for treating age-related diseases, and summarize the known natural compounds as senolytic agents and their applications.
Collapse
Affiliation(s)
- Wen Li
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, China; Department of Endocrinology, The Third People's Hospital of Yunnan Province, Kunming, Yunnan 650011, China
| | - Lin Qin
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, China; Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650000, China
| | - Rennan Feng
- Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Guangrong Hu
- Department of Emergency, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Hui Sun
- Department of Emergency, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Yonghan He
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming 650223, China.
| | - Rongping Zhang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan 650500, China.
| |
Collapse
|
30
|
Wu L, Li Q, Liu S, An X, Huang Z, Zhang B, Yuan Y, Xing C. Protective effect of hyperoside against renal ischemia-reperfusion injury via modulating mitochondrial fission, oxidative stress, and apoptosis. Free Radic Res 2019; 53:727-736. [PMID: 31130024 DOI: 10.1080/10715762.2019.1623883] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Ischemia/reperfusion (IR) is a common cause of acute kidney injury (AKI). However, effective therapies for IR-induced AKI are lacking. Hyperoside is an active constituent in the flowers of Abelmoschus manihot (L.) Medic, which is a traditional Chinese herbal medicine for the treatment of various ischemic brain and heart diseases. Our previous study demonstrated that hyperoside inhibited adriamycin induced podocyte injury both in vivo and in vitro. The aim of this study is to investigate the effect of hyperoside in IR-induced AKI. In mice, pretreatment of hyperoside could markedly attenuate IR-induced AKI, tubular cell apoptosis, and oxidative stress in the kidneys. Meanwhile, we found hyperoside inhibited IR-induced mitochondrial fission by suppressing OMA1 mediated proteolysis of optic atrophy 1 (OPA1). Consistently, in human proximal tubular epithelial cells, hyperoside might inhibit CoCl2-induced mitochondrial fission, oxidative stress, and apoptosis by regulating OMA1-OPA1 axis. Taken together, our results support the idea that OMA1-OPA1 mediated mitochondrial fission can be used for the prevention of AKI. Hyperoside might have novel therapeutic potential in the treatment of AKI.
Collapse
Affiliation(s)
- Lin Wu
- a Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital , Nanjing , PR China
| | - Qing Li
- a Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital , Nanjing , PR China
| | - Simeng Liu
- a Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital , Nanjing , PR China
| | - Xiaofei An
- b Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine , Nanjing , PR China
| | - Zhimin Huang
- a Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital , Nanjing , PR China
| | - Bo Zhang
- a Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital , Nanjing , PR China
| | - Yanggang Yuan
- a Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital , Nanjing , PR China
| | - Changying Xing
- a Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital , Nanjing , PR China
| |
Collapse
|
31
|
Abstract
Cellular senescence (CS) is one of hallmarks of aging and accumulation of senescent cells (SCs) with age contributes to tissue or organismal aging, as well as the pathophysiologies of diverse age-related diseases (ARDs). Genetic ablation of SCs in tissues lengthened health span and reduced the risk of age-related pathologies in a mouse model, suggesting a direct link between SCs, longevity, and ARDs. Therefore, senotherapeutics, medicines targeting SCs, might be an emerging strategy for the extension of health span, and prevention or treatment of ARDs. Senotherapeutics are classified as senolytics which kills SCs selectively; senomorphics which modulate functions and morphology of SCs to those of young cells, or delays the progression of young cells to SCs in tissues; and immune-system mediators of the clearance of SCs. Some senolytics and senomorphics have been proven to markedly prevent or treat ARDs in animal models. This review will present the current status of the development of senotherapeutics, in relation to aging itself and ARDs. Finally, future directions and opportunities for senotherapeutics use will discussed. This knowledge will provide information that can be used to develop novel senotherapeutics for health span and ARDs. [BMB Reports 2019; 52(1): 47-55].
Collapse
Affiliation(s)
- Eok-Cheon Kim
- Department of Biochemistry and Molecular Biology, Smart-aging Convergence Research Center, College of Medicine, Yeungnam University, Daegu 42415, Korea
| | - Jae-Ryong Kim
- Department of Biochemistry and Molecular Biology, Smart-aging Convergence Research Center, College of Medicine, Yeungnam University, Daegu 42415, Korea
| |
Collapse
|
32
|
Skaria T, Bachli E, Schoedon G. Gene Ontology Analysis for Drug Targets of the Whole Genome Transcriptome of Human Vascular Endothelial Cells in Response to Proinflammatory IL-1. Front Pharmacol 2019; 10:414. [PMID: 31068815 PMCID: PMC6491677 DOI: 10.3389/fphar.2019.00414] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 04/01/2019] [Indexed: 12/31/2022] Open
Affiliation(s)
- Tom Skaria
- Inflammation Research Unit, Division of Internal Medicine, University Hospital Zürich, Zurich, Switzerland
| | - Esther Bachli
- Department of Medicine, Uster Hospital, Uster, Switzerland
| | - Gabriele Schoedon
- Inflammation Research Unit, Division of Internal Medicine, University Hospital Zürich, Zurich, Switzerland
| |
Collapse
|
33
|
Hwang HV, Lin Y, Rebuffatti MN, Tran DT, Lee L, Gomes AV, Li CS, Knowlton AA. Impaired proteostasis in senescent vascular endothelial cells: a perspective on estrogen and oxidative stress in the aging vasculature. Am J Physiol Heart Circ Physiol 2018; 316:H421-H429. [PMID: 30499713 DOI: 10.1152/ajpheart.00318.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The heat shock response is an important cytoprotective mechanism for protein homeostasis and is an essential protective response to cellular stress and injury. Studies on changes in the heat shock response with aging have been mixed with regard to whether it is inhibited, and this, at least in part, reflects different tissues and different models. Cellular senescence is a key feature in aging, but work on the heat shock response in cultured senescent (SEN) cells has largely been limited to fibroblasts. Given the prevalence of oxidative injury in the aging cardiovascular system, we investigated whether SEN primary human coronary artery endothelial cells have a diminished heat shock response and impaired proteostasis. In addition, we tested whether this downregulation of heat shock response can be mitigated by 17β-estradiol (E2), which has a critical cardioprotective role in women, as we have previously reported that E2 improves the heat shock response in endothelial cells (Hamilton KL, Mbai FN, Gupta S, Knowlton AA. Arterioscler Thromb Vasc Biol 24: 1628-1633, 2004). We found that SEN endothelial cells, despite their unexpectedly increased proteasome activity, had a diminished heat shock response and had more protein aggregation than early passage cells. SEN cells had increased oxidative stress, which promoted protein aggregation. E2 treatment did not decrease protein aggregation or improve the heat shock response in either early passage or SEN cells. In summary, cellular senescence in adult human endothelial cells is accompanied by increased oxidative stress and a blunting of proteostasis, and E2 did not mitigate these changes. NEW & NOTEWORTHY Senescent human endothelial cells have a diminished heat shock response and increased protein aggregates. Senescent human endothelial cells have increased basal oxidative stress, which increases protein aggregates. Physiological level of 17β-estradiol did not improve proteostasis in endothelial cells.
Collapse
Affiliation(s)
- HyunTae V Hwang
- Molecular and Cellular Cardiology, Cardiovascular Division, Department of Internal Medicine, University of California, Davis, California
| | - Yun Lin
- Molecular and Cellular Cardiology, Cardiovascular Division, Department of Internal Medicine, University of California, Davis, California
| | - Michelle N Rebuffatti
- Molecular and Cellular Cardiology, Cardiovascular Division, Department of Internal Medicine, University of California, Davis, California
| | - Darlene T Tran
- Molecular and Cellular Cardiology, Cardiovascular Division, Department of Internal Medicine, University of California, Davis, California
| | - Lily Lee
- Molecular and Cellular Cardiology, Cardiovascular Division, Department of Internal Medicine, University of California, Davis, California
| | - Aldrin V Gomes
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, California.,Department of Physiology and Membrane Biology, University of California, Davis, California
| | - Chin-Shang Li
- School of Nursing, The State University of New York, University at Buffalo, Buffalo, New York
| | - Anne A Knowlton
- Veterans Affairs Medical Center, Sacramento, California.,Molecular and Cellular Cardiology, Cardiovascular Division, Department of Internal Medicine, University of California, Davis, California.,Department of Pharmacology, University of California, Davis, California
| |
Collapse
|
34
|
Jang SA, Park DW, Sohn EH, Lee SR, Kang SC. Hyperoside suppresses tumor necrosis factor α-mediated vascular inflammatory responses by downregulating mitogen-activated protein kinases and nuclear factor-κB signaling. Chem Biol Interact 2018; 294:48-55. [DOI: 10.1016/j.cbi.2018.08.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/13/2018] [Accepted: 08/17/2018] [Indexed: 01/06/2023]
|
35
|
Oost W, Talma N, Meilof JF, Laman JD. Targeting senescence to delay progression of multiple sclerosis. J Mol Med (Berl) 2018; 96:1153-1166. [PMID: 30229272 PMCID: PMC6208951 DOI: 10.1007/s00109-018-1686-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 07/18/2018] [Accepted: 08/09/2018] [Indexed: 12/11/2022]
Abstract
Multiple sclerosis (MS) is a chronic and often progressive, demyelinating disease of the central nervous system (CNS) white and gray matter and the single most common cause of disability in young adults. Age is one of the factors most strongly influencing the course of progression in MS. One of the hallmarks of aging is cellular senescence. The elimination of senescent cells with senolytics has very recently been shown to delay age-related dysfunction in animal models for other neurological diseases. In this review, the possible link between cellular senescence and the progression of MS is discussed, and the potential use of senolytics as a treatment for progressive MS is explored. Currently, there is no cure for MS and there are limited treatment options to slow the progression of MS. Current treatment is based on immunomodulatory approaches. Various cell types present in the CNS can become senescent and thus potentially contribute to MS disease progression. We propose that, after cellular senescence has indeed been shown to be directly implicated in disease progression, administration of senolytics should be tested as a potential therapeutic approach for the treatment of progressive MS.
Collapse
Affiliation(s)
- Wendy Oost
- University of Groningen, Groningen, The Netherlands
| | - Nynke Talma
- European Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Neuroscience, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jan F Meilof
- Department of Neurology, Martini Hospital, Groningen, The Netherlands.,MS Center Noord Nederland (MSCNN), University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jon D Laman
- Department of Neuroscience, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands. .,MS Center Noord Nederland (MSCNN), University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
36
|
Gurău F, Baldoni S, Prattichizzo F, Espinosa E, Amenta F, Procopio AD, Albertini MC, Bonafè M, Olivieri F. Anti-senescence compounds: A potential nutraceutical approach to healthy aging. Ageing Res Rev 2018; 46:14-31. [PMID: 29742452 DOI: 10.1016/j.arr.2018.05.001] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 04/02/2018] [Accepted: 05/03/2018] [Indexed: 01/10/2023]
Abstract
The desire of eternal youth seems to be as old as mankind. However, the increasing life expectancy experienced by populations in developed countries also involves a significantly increased incidence of the most common age-related diseases (ARDs). Senescent cells (SCs) have been identified as culprits of organismal aging. Their number rises with age and their senescence-associated secretory phenotype fuels the chronic, pro-inflammatory systemic state (inflammaging) that characterizes aging, impairing the regenerative ability of stem cells and increasing the risk of developing ARDs. A variegated class of molecules, including synthetic senolytic compounds and natural compounds contained in food, have been suggested to possess anti-senescence activity. Senolytics are attracting growing interest, and their safety and reliability as anti-senescence drugs are being assessed in human clinical trials. Notably, since SCs spread inflammation at the systemic level through pro-oxidant and pro-inflammatory signals, foods rich in polyphenols, which exert antioxidant and anti-inflammatory actions, have the potential to be harnessed as "anti-senescence foods" in a nutraceutical approach to healthier aging. We discuss the beneficial effects of polyphenol-rich foods in relation to the Mediterranean diet and the dietary habits of long-lived individuals, and examine their ability to modulate bacterial genera in the gut.
Collapse
Affiliation(s)
- Felicia Gurău
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - Simone Baldoni
- School of Medicinal Sciences and Health Products, University of Camerino, Camerino, Italy
| | | | - Emma Espinosa
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - Francesco Amenta
- School of Medicinal Sciences and Health Products, University of Camerino, Camerino, Italy
| | - Antonio Domenico Procopio
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy; Center of Clinical Pathology and Innovative Therapy, INRCA-IRCCS National Institute, Ancona, Italy
| | | | - Massimiliano Bonafè
- DIMES- Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum, Bologna, Italy; Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Forlì, Italy.
| | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy; Center of Clinical Pathology and Innovative Therapy, INRCA-IRCCS National Institute, Ancona, Italy.
| |
Collapse
|
37
|
Skaria T, Bachli E, Schoedon G. RSPO3 impairs barrier function of human vascular endothelial monolayers and synergizes with pro-inflammatory IL-1. Mol Med 2018; 24:45. [PMID: 30157748 PMCID: PMC6116367 DOI: 10.1186/s10020-018-0048-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 08/15/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Endothelial barrier dysfunction characterized by hyperpermeability of the vascular endothelium is a key factor in the pathogenesis of chronic inflammatory diseases and affects clinical outcomes. In states of chronic inflammation, mediators secreted by activated immune cells or vascular endothelium may affect the barrier function and permeability of the vascular endothelium. The matricellular R-spondin family member RSPO3 is produced by inflammatory-activated human monocytes and vascular endothelial cells, but its effects in the regulation of vascular endothelial barrier function remains elusive. METHODS The present study investigates the effects of RSPO3 on the barrier function of adult human primary macro- and micro- vascular endothelial monolayers. Tight monolayers of primary endothelial cells from human coronary and pulmonary arteries, and cardiac, brain, and dermal microvascular beds were treated with RSPO3 either alone or in combination with pro-inflammatory mediator IL-1β. Endothelial barrier function was assessed non-invasively in real-time using Electric Cell-substrate Impedance Sensing. RESULTS RSPO3 treatment critically affected barrier function by enhancing the permeability of all vascular endothelial monolayers investigated. To confer hyperpermeable phenotype in vascular endothelial monolayers, RSPO3 induced inter-endothelial gap formation by disrupting the β-catenin and VE-cadherin alignment at adherens junctions. RSPO3 synergistically enhanced the barrier impairing properties of the pro-inflammatory mediator IL-1β. CONCLUSION Here, we show that the matricellular protein RSPO3 is a mediator of endothelial hyperpermeability that can act in synergy with the inflammatory mediator IL-1β. This finding stimulates further studies to delineate the endothelial barrier impairing properties of RSPO3 and its synergistic interaction with IL-1β in chronic inflammatory diseases.
Collapse
Affiliation(s)
- Tom Skaria
- Inflammation Research Unit, Division of Internal Medicine, University Hospital Zürich, Rämistrasse 100, CH-8091, Zürich, Switzerland
| | - Esther Bachli
- Department of Medicine, Uster Hospital, Brunnenstrasse 42, CH-8610, Uster, Switzerland
| | - Gabriele Schoedon
- Inflammation Research Unit, Division of Internal Medicine, University Hospital Zürich, Rämistrasse 100, CH-8091, Zürich, Switzerland.
| |
Collapse
|
38
|
Myrianthopoulos V, Evangelou K, Vasileiou PVS, Cooks T, Vassilakopoulos TP, Pangalis GA, Kouloukoussa M, Kittas C, Georgakilas AG, Gorgoulis VG. Senescence and senotherapeutics: a new field in cancer therapy. Pharmacol Ther 2018; 193:31-49. [PMID: 30121319 DOI: 10.1016/j.pharmthera.2018.08.006] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Cellular senescence is a stress response mechanism ensuring homeostasis. Its temporal activation during embryonic development or normal adult life is linked with beneficial properties. In contrast, persistent (chronic) senescence seems to exert detrimental effects fostering aging and age-related disorders, such as cancer. Due to the lack of a reliable marker able to detect senescence in vivo, its precise impact in age-related diseases is to a large extent still undetermined. A novel reagent termed GL13 (SenTraGorTM) that we developed, allowing senescence recognition in any type of biological material, emerges as a powerful tool to study the phenomenon of senescence in vivo. Exploiting the advantages of this novel methodological approach, scientists will be able to detect and connect senescence with aggressive behavior in human malignancies, such as tolerance to chemotherapy in classical Hodgkin Lymphoma and Langerhans Cell Histiocytosis. The latter depicts the importance of developing the new and rapidly expanding field of senotherapeutic agents targeting and driving to cell death senescent cells. We discuss in detail the current progress of this exciting area of senotherapeutics and suggest its future perspectives and applications.
Collapse
Affiliation(s)
- Vassilios Myrianthopoulos
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece; Division of Pharmaceutical Chemistry, School of Pharmacy, National and Kapodistrian University of Athens, Greece; PharmaInformatics Unit, Athena Research Center, Greece
| | - Konstantinos Evangelou
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece; Department of Anatomy-Histology-Embryology, Medical School, University of Ioannina, Ioannina, Greece
| | - Panagiotis V S Vasileiou
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Tomer Cooks
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Theodoros P Vassilakopoulos
- Department of Haematology and Bone Marrow Transplantation, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Myrsini Kouloukoussa
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece; Museum of Anthropology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christos Kittas
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Alexandros G Georgakilas
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Athens, Greece.
| | - Vassilis G Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece; Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece; Faculty Institute for Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK; Biomedical Research Foundation, Academy of Athens, Athens, Greece.
| |
Collapse
|