1
|
Driscoll M, Sedore CA, Onken B, Coleman-Hulbert AL, Johnson E, Phillips PC, Lithgow G. NIA Caenorhabditis Intervention Testing Program: identification of robust and reproducible pharmacological interventions that promote longevity across experimentally accessible, genetically diverse populations. GeroScience 2025:10.1007/s11357-025-01627-4. [PMID: 40178707 DOI: 10.1007/s11357-025-01627-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 03/17/2025] [Indexed: 04/05/2025] Open
Abstract
A core facet of the National Institute on Aging's mission is to identify pharmacological interventions that can promote human healthy aging and long life. As part of the comprehensive effort toward that goal, the NIA Division of Biology of Aging established the Caenorhabditis Intervention Testing Program (CITP) in 2013. The C. elegans model (with an ~ 21 day lifespan) has led the field in dissection of longevity genetics and offers features that allow for relatively rapid testing and for the potential elaboration of biological mechanisms engaged by candidate geroprotectants. CITP builds on this foundation by utilizing a genetically diverse set of intervention test strains so that "subjects" represent genetic diversity akin to that that between mouse and humans. Another distinctive aspect of the CITP is a dedicated focus on reproducibility of longevity outcomes as labs at three independent test sites confirm positive outcomes. The overall goal of the Caenorhabditis Intervention Testing Program (CITP) is to identify robust and reproducible pro-longevity interventions affecting genetically diverse cohorts in the Caenorhabditis genus. A strong Data Collection Center supports data collection and dissemination. Pharmacological interventions tested by CITP can be nominated by the general public, directed by in-house screens, or supported by published scientific literature. As of December 2024, CITP tested > 75 compounds and conducted > 725,000 animal assays over 891 trials. We identified 12 compounds that confer a ≥ 20% increase in median lifespan to reproducibly and robustly extend lifespan across multiple strains and labs. Five of these interventions have pro-longevity impact reported in the mouse literature (most CITP positive interventions are not tested yet in mouse). As part of the celebration of the 50th Anniversary of the NIA, we review the development history and accomplishments of the CITP program, and we comment on translation and the promise of advancing understanding of fundamental aging biology that includes the pharmacological intervention/health interface.
Collapse
Affiliation(s)
- Monica Driscoll
- Rutgers, The State University of New Jersey, Piscataway, NJ, USA.
| | | | - Brian Onken
- Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | | | | | | | | |
Collapse
|
2
|
Banse SA, Coleman-Hulbert AL, Sedore CA, Johnson E, Lithgow GJ, Driscoll M, Phillips PC. Caenorhabditis Intervention Testing Program: all-trans retinoic acid-related compounds tamibarotene and bakuchiol do not extend lifespan in Caenorhabditis nematodes. MICROPUBLICATION BIOLOGY 2025; 2025:10.17912/micropub.biology.001517. [PMID: 40027526 PMCID: PMC11868923 DOI: 10.17912/micropub.biology.001517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 01/30/2025] [Accepted: 02/04/2025] [Indexed: 03/05/2025]
Abstract
The Caenorhabditis Intervention Testing Program recently characterized the longevity-promoting effects of the vitamin A derivative all-trans retinoic acid (atRA). Here, we test two atRA-related compounds, tamibarotene and bakuchiol, for longevity effects in three strains of Caenorhabditis species. Both tamibarotene, a potent RAR agonist, and bakuchiol, a meroterpene derived from Psoralea corylifolia , showed no significant increase in lifespan across a dosage range of six concentrations. Additionally, bakuchiol was broadly toxic at higher doses. These findings highlight the specificity of atRA's longevity effects and suggest that compounds related to atRA may not universally promote lifespan extension.
Collapse
Affiliation(s)
- Stephen A. Banse
- Institute of Ecology and Evolution, University of Oregon, Eugene, Oregon 97403, USA
| | | | - Christine A. Sedore
- Institute of Ecology and Evolution, University of Oregon, Eugene, Oregon 97403, USA
| | - Erik Johnson
- Institute of Ecology and Evolution, University of Oregon, Eugene, Oregon 97403, USA
| | - Gordon J. Lithgow
- The Buck Institute for Research on Aging, Novato, California 94945, USA
| | - Monica Driscoll
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Patrick C. Phillips
- Institute of Ecology and Evolution, University of Oregon, Eugene, Oregon 97403, USA
| |
Collapse
|
3
|
Zhang S, Liu Y, Qi J, Yan Y, Gao T, Zhang X, Sun D, Wang T, Zeng P. Accelerated aging as a mediator of the association between co-exposure to multiple air pollutants and risk of chronic kidney disease. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117582. [PMID: 39719816 DOI: 10.1016/j.ecoenv.2024.117582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 12/26/2024]
Abstract
BACKGROUND The association between co-exposure to multiple air pollutants and the occurrence of chronic kidney disease (CKD) was not well-established, and the mediating role of accelerated aging in this association remained uncertain. METHODS Using a cohort of 313,908 participants without CKD at baseline from the UK Biobank, we examined the potential association between co-exposure to multiple air pollutants, including PM2.5, PM10, PM2.5-10, NO2 and NOx, and the incidence of CKD by calculating an air pollution score. Mediation analyses were performed to examine the mediating role of accelerated aging (PhenoAgeAccel or KDM-BioAgeAccel) in this association. RESULTS During the median follow-up time of 12.9 years, 11,117 participants developed CKD. The results showed that per interquartile range (IQR) increment in air pollution score led to an approximately 9.0 % (6.6-11.4 %) elevated risk of occurring CKD. Compared to the first quartile (Q1) of air pollution score, those in the highest quartile (Q4) had a 21.2 % (14.8-27.9 %) higher risk of developing CKD (Ptrend<0.001). Mediation analyses suggested that PhenoAgeAccel and KDM-BioAgeAccel significantly mediated 1.5 % and 5.7 % of the association between air pollution score and incident CKD, respectively. CONCLUSION Co-exposure to multiple air pollutants could increase the risk of developing CKD, with accelerated aging serving as a partial mechanism in the relationship between air pollution and CKD. These findings highlight the importance of reducing air pollution, and suggest a possible mechanism from air pollution to CKD through accelerated aging.
Collapse
Affiliation(s)
- Shuo Zhang
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yuxin Liu
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Jike Qi
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yu Yan
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Tongyu Gao
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xin Zhang
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Dong Sun
- Department of Nephrology and Clinical Research Center for Kidney Disease, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China; Clinical Research Center for Kidney Disease, Xuzhou Medical University, Xuzhou 221004, China
| | - Ting Wang
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Ping Zeng
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Jiangsu Engineering Research Center of Biological Data Mining and Healthcare Transformation, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
4
|
Shibata Y, Tanaka Y, Mori S, Mitsuzumi K, Fujii S, Sasakura H, Morioka Y, Sugioka K, Takeuchi K, Nishiwaki K. Endogenous chondroitin extends lifespan by inhibiting VHA-7-mediated tubular lysosome formation. Sci Rep 2024; 14:29651. [PMID: 39609482 PMCID: PMC11605119 DOI: 10.1038/s41598-024-80242-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 11/18/2024] [Indexed: 11/30/2024] Open
Abstract
Chondroitin extends lifespan and healthspan in C. elegans, but the relationship between extracellular chondroitin and intracellular anti-aging mechanisms is unknown. The basement membrane (BM) that contains chondroitin proteoglycans is anchored to cells via hemidesmosomes (HDs), and it accumulates damage with aging. In this study, we found that chondroitin regulates aging through the formation of HDs and inhibition of tubular lysosomes (TLs). Reduction of chondroitin due to a mutation in sqv-5/Chondroitin synthase (ChSy) causes the earlier and excessive formation of TLs and leakage of the lysosomal nuclease in a manner dependent on VHA-7, the a-subunit of V-type ATPase. VHA-7, whose mutation suppresses the short lifespan of the sqv-5 mutant, is initially localized to the basal side of the hypodermal cells and transported to lysosomes with aging. These results demonstrate that endogenous chondroitin suppresses aging by inhibiting the earlier excessive formation of TLs. This is a novel anti-aging mechanism that is controlled by the BM.
Collapse
Affiliation(s)
- Yukimasa Shibata
- Department of Biomedical Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo, Japan.
- Gakuen Uegahara, Sanda, 669-1330, Japan.
| | - Yuri Tanaka
- Department of Biomedical Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo, Japan
| | - Shunsuke Mori
- Department of Biomedical Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo, Japan
| | - Kaito Mitsuzumi
- Department of Biomedical Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo, Japan
| | - Shion Fujii
- Department of Biomedical Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo, Japan
| | - Hiroyuki Sasakura
- Department of Medical Cell Biology, School of Medicine, Aichi Medical University, Nagakute, Aichi, Japan
| | - Yuki Morioka
- Department of Medical Cell Biology, School of Medicine, Aichi Medical University, Nagakute, Aichi, Japan
| | - Kenji Sugioka
- Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T1Z3, Canada
| | - Kosei Takeuchi
- Department of Medical Cell Biology, School of Medicine, Aichi Medical University, Nagakute, Aichi, Japan
| | - Kiyoji Nishiwaki
- Department of Biomedical Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo, Japan
| |
Collapse
|
5
|
Moriya A, Nakato E, Li JP, Nakato H. Chondroitin sulfate in invertebrate development. PROTEOGLYCAN RESEARCH 2024; 2:e70009. [PMID: 39664970 PMCID: PMC11632948 DOI: 10.1002/pgr2.70009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/16/2024] [Indexed: 12/13/2024]
Abstract
Chondroitin sulfate (CS) is one of the most evolutionarily conserved glycosaminoglycans (GAGs). Although CS's function in skeletal development is well established in vertebrates, CS exists in more primitive animal species with no cartilage or bone, such as C. elegans and Drosophila, indicating that the original role of CS was not in the skeletal system. In this review, we focus on the roles of CS and the mechanisms of action during development of two genetically trackable model organisms, C. elegans and Drosophila.
Collapse
Affiliation(s)
- Ayano Moriya
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota, USA
| | - Eriko Nakato
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jin-Ping Li
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Hiroshi Nakato
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
6
|
Jongsma E, Grigolon G, Baumann J, Weinkove D, Ewald CY, Wandrey F, Grothe T. Timut Pepper Extract Slows Age-Dependent Decline of Mobility and Collagen Loss and Promotes Longevity. Nutrients 2024; 16:2122. [PMID: 38999870 PMCID: PMC11243454 DOI: 10.3390/nu16132122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/14/2024] Open
Abstract
Investigations into human longevity are increasingly focusing on healthspan enhancement, not just lifespan extension. Lifestyle modifications and nutritional choices, including food supplements, can significantly affect aging and general health. Phytochemicals in centenarians' diets, such as those found in Timut pepper, a Nepalese spice with various medicinal properties, may contribute to their longevity. Similarly, Sichuan pepper, a related species, has demonstrated anti-inflammatory and neuroprotective activities. With the broader purpose of uncovering a novel treatment to address aging and its comorbidities, this study aims to investigate the potential lifespan- and healthspan-promoting effects of Timut pepper using the model organism Caenorhabditis elegans. We show that Timut pepper extract extends C. elegans' lifespan at different maintenance temperatures and increases the proportion of active nematodes in their early adulthood. In addition, we show that Timut pepper extract enhances speed and distance moved as the nematodes age. Finally, Timut pepper extract assures extracellular matrix homeostasis by slowing the age-dependent decline of collagen expression.
Collapse
Affiliation(s)
- Elisabeth Jongsma
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, 8603 Schwerzenbach, Switzerland
| | | | - Julia Baumann
- Mibelle Group Biochemistry, Mibelle AG, 5033 Buchs, Switzerland
| | - David Weinkove
- Magnitude Biosciences Ltd., NETPark Plexus, Thomas Wright Way, Sedgefield TS21 3FD, UK
- Department of Biosciences, Durham University, Stockton Road, Durham DH1 3LE, UK
| | - Collin Y. Ewald
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, 8603 Schwerzenbach, Switzerland
| | | | - Torsten Grothe
- Mibelle Group Biochemistry, Mibelle AG, 5033 Buchs, Switzerland
| |
Collapse
|
7
|
Chmielewski PP, Data K, Strzelec B, Farzaneh M, Anbiyaiee A, Zaheer U, Uddin S, Sheykhi-Sabzehpoush M, Mozdziak P, Zabel M, Dzięgiel P, Kempisty B. Human Aging and Age-Related Diseases: From Underlying Mechanisms to Pro-Longevity Interventions. Aging Dis 2024:AD.2024.0280. [PMID: 38913049 DOI: 10.14336/ad.2024.0280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/02/2024] [Indexed: 06/25/2024] Open
Abstract
As human life expectancy continues to rise, becoming a pressing global concern, it brings into focus the underlying mechanisms of aging. The increasing lifespan has led to a growing elderly population grappling with age-related diseases (ARDs), which strains healthcare systems and economies worldwide. While human senescence was once regarded as an immutable and inexorable phenomenon, impervious to interventions, the emerging field of geroscience now offers innovative approaches to aging, holding the promise of extending the period of healthspan in humans. Understanding the intricate links between aging and pathologies is essential in addressing the challenges presented by aging populations. A substantial body of evidence indicates shared mechanisms and pathways contributing to the development and progression of various ARDs. Consequently, novel interventions targeting the intrinsic mechanisms of aging have the potential to delay the onset of diverse pathological conditions, thereby extending healthspan. In this narrative review, we discuss the most promising methods and interventions aimed at modulating aging, which harbor the potential to mitigate ARDs in the future. We also outline the complexity of senescence and review recent empirical evidence to identify rational strategies for promoting healthy aging.
Collapse
Affiliation(s)
- Piotr Pawel Chmielewski
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Krzysztof Data
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Bartłomiej Strzelec
- 2nd Department of General Surgery and Surgical Oncology, Medical University Hospital, Wroclaw, Poland
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Amir Anbiyaiee
- Department of Surgery, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Uzma Zaheer
- School of Biosciences, Faculty of Health Sciences and Medicine, The University of Surrey, United Kingdom
| | - Shahab Uddin
- Translational Institute and Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Department of Biosciences, Integral University, Lucknow, Uttar Pradesh, India
| | | | - Paul Mozdziak
- Graduate Physiology Program, North Carolina State University, Raleigh, NC 27695, USA
| | - Maciej Zabel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, Wroclaw, Poland
- Division of Anatomy and Histology, The University of Zielona Góra, Poland
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, Wroclaw, Poland
| | - Bartosz Kempisty
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun, Poland
- Physiology Graduate Faculty, North Carolina State University, Raleigh, NC 27695, USA
- Center of Assisted Reproduction, Department of Obstetrics and Gynecology, University Hospital and Masaryk University, Brno, Czech Republic
| |
Collapse
|
8
|
Shibata Y, Tanaka Y, Sasakura H, Morioka Y, Sassa T, Fujii S, Mitsuzumi K, Ikeno M, Kubota Y, Kimura K, Toyoda H, Takeuchi K, Nishiwaki K. Endogenous chondroitin extends the lifespan and healthspan in C. elegans. Sci Rep 2024; 14:4813. [PMID: 38413743 PMCID: PMC10899230 DOI: 10.1038/s41598-024-55417-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/23/2024] [Indexed: 02/29/2024] Open
Abstract
Chondroitin, a class of glycosaminoglycan polysaccharides, is found as proteoglycans in the extracellular matrix, plays a crucial role in tissue morphogenesis during development and axonal regeneration. Ingestion of chondroitin prolongs the lifespan of C. elegans. However, the roles of endogenous chondroitin in regulating lifespan and healthspan mostly remain to be investigated. Here, we demonstrate that a gain-of-function mutation in MIG-22, the chondroitin polymerizing factor (ChPF), results in elevated chondroitin levels and a significant extension of both the lifespan and healthspan in C. elegans. Importantly, the remarkable longevity observed in mig-22(gf) mutants is dependent on SQV-5/chondroitin synthase (ChSy), highlighting the pivotal role of chondroitin in controlling both lifespan and healthspan. Additionally, the mig-22(gf) mutation effectively suppresses the reduced healthspan associated with the loss of MIG-17/ADAMTS metalloprotease, a crucial for factor in basement membrane (BM) remodeling. Our findings suggest that chondroitin functions in the control of healthspan downstream of MIG-17, while regulating lifespan through a pathway independent of MIG-17.
Collapse
Affiliation(s)
- Yukimasa Shibata
- Department of Biomedical Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo, 669-1330, Japan.
| | - Yuri Tanaka
- Department of Biomedical Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo, 669-1330, Japan
| | - Hiroyuki Sasakura
- Department of Medical Cell Biology, School of Medicine, Aichi Medical University, Nagakute, Aichi, Japan
| | - Yuki Morioka
- Department of Medical Cell Biology, School of Medicine, Aichi Medical University, Nagakute, Aichi, Japan
| | | | - Shion Fujii
- Department of Biomedical Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo, 669-1330, Japan
| | - Kaito Mitsuzumi
- Department of Biomedical Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo, 669-1330, Japan
| | - Masashi Ikeno
- Department of Medical Cell Biology, School of Medicine, Aichi Medical University, Nagakute, Aichi, Japan
| | - Yukihiko Kubota
- Department of Biomedical Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo, 669-1330, Japan
- Department of Bioinformatics, College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Kenji Kimura
- Department of Biomedical Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo, 669-1330, Japan
| | - Hidenao Toyoda
- Laboratory of Bio-Analytical Chemistry, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Kosei Takeuchi
- Department of Medical Cell Biology, School of Medicine, Aichi Medical University, Nagakute, Aichi, Japan
| | - Kiyoji Nishiwaki
- Department of Biomedical Sciences, Kwansei Gakuin University, 1 Gakuen Uegahara, Sanda, Hyogo, 669-1330, Japan
| |
Collapse
|
9
|
Sharma A, Ewald CY. Clearance of extracellular human amyloid-β aggregates in C. elegans by nutraceutical and pharmaceutical interventions. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.000907. [PMID: 38287930 PMCID: PMC10823790 DOI: 10.17912/micropub.biology.000907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 12/20/2023] [Accepted: 01/03/2024] [Indexed: 01/31/2024]
Abstract
Numerous anti-amyloid therapies have seen recent clinical development and approval, such as the monoclonal antibodies aducanumab and lecanemab. However, in Alzheimer's disease patients, amyloid-β (Aβ) plaques are found embedded in the extracellular matrix and surrounded by collagens, which might hinder these antibodies from targeting the plaques. We reasoned that various different nutraceutical and pharmaceutical agents might induce collagen and extracellular matrix turnover and removal of these collagen-embedded amyloid-β (Aβ) plaques. To address this idea, here, we used a transgenic C. elegans strain, LSD2104 , expressing fluorescent human Aβ 1-42 as an in-vivo model for secreted amyloid aggregation in the extracellular matrix. We performed a screen of various nutraceuticals and pharmaceuticals along with different combinations, and we found that quercetin 350 µM and rifampicin 75 µM successfully cleared the extracellular amyloid plaque burden compared to the 0.2% DMSO control group, with a combination of the two agents producing the maximum effect compared to either drug alone. These results may implicate the exploration of combination therapeutics of nutraceuticals and pharmaceuticals in the clearance of amyloid-β (Aβ) plaques in Alzheimer's disease.
Collapse
Affiliation(s)
- Arastu Sharma
- Eidgenössische Technische Hochschule Zürich, Department of Health Sciences and Technology, Institute of Translational Medicine, 8603 Schwerzenbach-Zürich, Switzerland
- Johns Hopkins University, Baltimore, Maryland, United States
| | - Collin Y Ewald
- Eidgenössische Technische Hochschule Zürich, Department of Health Sciences and Technology, Institute of Translational Medicine, 8603 Schwerzenbach-Zürich, Switzerland
| |
Collapse
|
10
|
Teuscher AC, Statzer C, Goyala A, Domenig SA, Schoen I, Hess M, Hofer AM, Fossati A, Vogel V, Goksel O, Aebersold R, Ewald CY. Longevity interventions modulate mechanotransduction and extracellular matrix homeostasis in C. elegans. Nat Commun 2024; 15:276. [PMID: 38177158 PMCID: PMC10766642 DOI: 10.1038/s41467-023-44409-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 12/12/2023] [Indexed: 01/06/2024] Open
Abstract
Dysfunctional extracellular matrices (ECM) contribute to aging and disease. Repairing dysfunctional ECM could potentially prevent age-related pathologies. Interventions promoting longevity also impact ECM gene expression. However, the role of ECM composition changes in healthy aging remains unclear. Here we perform proteomics and in-vivo monitoring to systematically investigate ECM composition (matreotype) during aging in C. elegans revealing three distinct collagen dynamics. Longevity interventions slow age-related collagen stiffening and prolong the expression of collagens that are turned over. These prolonged collagen dynamics are mediated by a mechanical feedback loop of hemidesmosome-containing structures that span from the exoskeletal ECM through the hypodermis, basement membrane ECM, to the muscles, coupling mechanical forces to adjust ECM gene expression and longevity via the transcriptional co-activator YAP-1 across tissues. Our results provide in-vivo evidence that coordinated ECM remodeling through mechanotransduction is required and sufficient to promote longevity, offering potential avenues for interventions targeting ECM dynamics.
Collapse
Affiliation(s)
- Alina C Teuscher
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach, CH-8603, Switzerland
| | - Cyril Statzer
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach, CH-8603, Switzerland
| | - Anita Goyala
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach, CH-8603, Switzerland
| | - Seraina A Domenig
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach, CH-8603, Switzerland
| | - Ingmar Schoen
- School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Laboratory of Applied Mechanobiology, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| | - Max Hess
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach, CH-8603, Switzerland
| | - Alexander M Hofer
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach, CH-8603, Switzerland
| | - Andrea Fossati
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, Zurich, Switzerland
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, 94158, CA, USA
| | - Viola Vogel
- Laboratory of Applied Mechanobiology, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| | - Orcun Goksel
- Department of Information Technology and Electrical Engineering, ETH Zürich, Zürich, Switzerland
- Department of Information Technology, Uppsala University, Uppsala, Sweden
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, Zurich, Switzerland
| | - Collin Y Ewald
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach, CH-8603, Switzerland.
| |
Collapse
|
11
|
Li X, Cao X, Zhang J, Fu J, Mohedaner M, Zhuogadanzeng, Sun X, Yang G, Yang Z, Kuo CL, Chen X, Cohen AA, Liu Z. Accelerated aging mediates the associations of unhealthy lifestyles with cardiovascular disease, cancer, and mortality. J Am Geriatr Soc 2024; 72:181-193. [PMID: 37789775 PMCID: PMC11078652 DOI: 10.1111/jgs.18611] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/16/2023] [Accepted: 08/24/2023] [Indexed: 10/05/2023]
Abstract
BACKGROUND With two well-validated aging measures capturing mortality and morbidity risk, this study examined whether and to what extent aging mediates the associations of unhealthy lifestyles with adverse health outcomes. METHODS Data were from 405,944 adults (40-69 years) from UK Biobank (UKB) and 9972 adults (20-84 years) from the US National Health and Nutrition Examination Survey (NHANES). An unhealthy lifestyles score (range: 0-5) was constructed based on five factors (smoking, drinking, physical inactivity, unhealthy body mass index, and unhealthy diet). Two aging measures, Phenotypic Age Acceleration (PhenoAgeAccel) and Biological Age Acceleration (BioAgeAccel) were calculated using nine and seven blood biomarkers, respectively, with a higher value indicating the acceleration of aging. The outcomes included incident cardiovascular disease (CVD), incident cancer, and all-cause mortality in UKB; CVD mortality, cancer mortality, and all-cause mortality in NHANES. A general linear regression model, Cox proportional hazards model, and formal mediation analysis were performed. RESULTS The unhealthy lifestyles score was positively associated with PhenoAgeAccel (UKB: β = 0.741; NHANES: β = 0.874, all p < 0.001). We further confirmed the respective associations of PhenoAgeAccel and unhealthy lifestyles with the outcomes in UKB and NHANES. The mediation proportion of PhenoAgeAccel in associations of unhealthy lifestyles with incident CVD, incident cancer, and all-cause mortality were 20.0%, 17.8%, and 26.6% (all p < 0.001) in UKB, respectively. Similar results were found in NHANES. The findings were robust when using another aging measure-BioAgeAccel. CONCLUSIONS Accelerated aging partially mediated the associations of lifestyles with CVD, cancer, and mortality in UK and US populations. The findings reveal a novel pathway and the potential of geroprotective programs in mitigating health inequality in late life beyond lifestyle interventions.
Collapse
Affiliation(s)
- Xueqin Li
- Center for Clinical Big Data and Analytics Second Affiliated Hospital and Department of Big Data in Health Science School of Public Health, the Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Xingqi Cao
- Center for Clinical Big Data and Analytics Second Affiliated Hospital and Department of Big Data in Health Science School of Public Health, the Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Jingyun Zhang
- Center for Clinical Big Data and Analytics Second Affiliated Hospital and Department of Big Data in Health Science School of Public Health, the Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Jinjing Fu
- Center for Clinical Big Data and Analytics Second Affiliated Hospital and Department of Big Data in Health Science School of Public Health, the Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Mayila Mohedaner
- Center for Clinical Big Data and Analytics Second Affiliated Hospital and Department of Big Data in Health Science School of Public Health, the Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Zhuogadanzeng
- Center for Clinical Big Data and Analytics Second Affiliated Hospital and Department of Big Data in Health Science School of Public Health, the Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Xiaoyi Sun
- Center for Clinical Big Data and Analytics Second Affiliated Hospital and Department of Big Data in Health Science School of Public Health, the Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Gan Yang
- Center for Clinical Big Data and Analytics Second Affiliated Hospital and Department of Big Data in Health Science School of Public Health, the Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Zhenqing Yang
- Center for Clinical Big Data and Analytics Second Affiliated Hospital and Department of Big Data in Health Science School of Public Health, the Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Chia-Ling Kuo
- Department of Community Medicine and Health Care, Connecticut Convergence Institute for Translation in Regenerative Engineering, Institute for Systems Genomics, University of Connecticut Health, Farmington, CT 06030, USA
| | - Xi Chen
- Department of Health Policy and Management, Yale School of Public Health, New Haven, CT 06520, USA
- Department of Economics, Yale University, New Haven, CT 06520, USA
| | - Alan A Cohen
- Department of Family Medicine, Research Centre on Aging, CHUS Research Centre, University of Sherbrooke, Sherbrooke, QC, Canada
- Butler Columbia Aging Center and Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY 10032, USA
| | - Zuyun Liu
- Center for Clinical Big Data and Analytics Second Affiliated Hospital and Department of Big Data in Health Science School of Public Health, the Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| |
Collapse
|
12
|
Lee MB, Blue B, Muir M, Kaeberlein M. The million-molecule challenge: a moonshot project to rapidly advance longevity intervention discovery. GeroScience 2023; 45:3103-3113. [PMID: 37432607 PMCID: PMC10643437 DOI: 10.1007/s11357-023-00867-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 06/30/2023] [Indexed: 07/12/2023] Open
Abstract
Targeting aging is the future of twenty-first century preventative medicine. Small molecule interventions that promote healthy longevity are known, but few are well-developed and discovery of novel, robust interventions has stagnated. To accelerate longevity intervention discovery and development, high-throughput systems are needed that can perform unbiased drug screening and directly measure lifespan and healthspan metrics in whole animals. C. elegans is a powerful model system for this type of drug discovery. Combined with automated data capture and analysis technologies, truly high-throughput longevity drug discovery is possible. In this perspective, we propose the "million-molecule challenge", an effort to quantitatively assess 1,000,000 interventions for longevity within five years. The WormBot-AI, our best-in-class robotics and AI data analysis platform, provides a tool to achieve the million-molecule challenge for pennies per animal tested.
Collapse
Affiliation(s)
- Mitchell B Lee
- Ora Biomedical, Inc., 12101 Tukwila International Blvd Suite 210, Seattle, WA, 98168, USA.
| | - Benjamin Blue
- Ora Biomedical, Inc., 12101 Tukwila International Blvd Suite 210, Seattle, WA, 98168, USA
| | - Michael Muir
- Ora Biomedical, Inc., 12101 Tukwila International Blvd Suite 210, Seattle, WA, 98168, USA
| | - Matt Kaeberlein
- Ora Biomedical, Inc., 12101 Tukwila International Blvd Suite 210, Seattle, WA, 98168, USA
- Optispan Geroscience, Seattle, WA, USA
| |
Collapse
|
13
|
Reich H, Savage-Dunn C. Signaling circuits and the apical extracellular matrix in aging: connections identified in the nematode Caenorhabditis elegans. Am J Physiol Cell Physiol 2023; 325:C1201-C1211. [PMID: 37721005 PMCID: PMC10861026 DOI: 10.1152/ajpcell.00195.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/24/2023] [Accepted: 09/11/2023] [Indexed: 09/19/2023]
Abstract
Numerous conserved signaling pathways play critical roles in aging, including insulin/IGF-1, TGF-β, and Wnt pathways. Some of these pathways also play prominent roles in the formation and maintenance of the extracellular matrix. The nematode Caenorhabditis elegans has been an enduringly productive system for the identification of conserved mechanisms of biological aging. Recent studies in C. elegans highlight the regulatory circuits between conserved signaling pathways and the extracellular matrix, revealing a bidirectional relationship between these factors and providing a platform to address how regulation of and by the extracellular matrix can impact lifespan and organismal health during aging. These discoveries provide new opportunities for clinical advances and novel therapeutic strategies.
Collapse
Affiliation(s)
- Hannah Reich
- Department of Biology, Queens College, City University of New York, Flushing, New York, United States
| | - Cathy Savage-Dunn
- Department of Biology, Queens College, City University of New York, Flushing, New York, United States
- PhD Program in Biology, The Graduate Center, City University of New York, New York, New York, United States
| |
Collapse
|
14
|
Jongsma E, Goyala A, Mateos JM, Ewald CY. Removal of extracellular human amyloid beta aggregates by extracellular proteases in C. elegans. eLife 2023; 12:e83465. [PMID: 37728486 PMCID: PMC10541181 DOI: 10.7554/elife.83465] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 09/19/2023] [Indexed: 09/21/2023] Open
Abstract
The amyloid beta (Aβ) plaques found in Alzheimer's disease (AD) patients' brains contain collagens and are embedded extracellularly. Several collagens have been proposed to influence Aβ aggregate formation, yet their role in clearance is unknown. To investigate the potential role of collagens in forming and clearance of extracellular aggregates in vivo, we created a transgenic Caenorhabditis elegans strain that expresses and secretes human Aβ1-42. This secreted Aβ forms aggregates in two distinct places within the extracellular matrix. In a screen for extracellular human Aβ aggregation regulators, we identified different collagens to ameliorate or potentiate Aβ aggregation. We show that a disintegrin and metalloprotease a disintegrin and metalloprotease 2 (ADM-2), an ortholog of ADAM9, reduces the load of extracellular Aβ aggregates. ADM-2 is required and sufficient to remove the extracellular Aβ aggregates. Thus, we provide in vivo evidence of collagens essential for aggregate formation and metalloprotease participating in extracellular Aβ aggregate removal.
Collapse
Affiliation(s)
- Elisabeth Jongsma
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH ZürichSchwerzenbachSwitzerland
| | - Anita Goyala
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH ZürichSchwerzenbachSwitzerland
| | - José Maria Mateos
- Center for Microscopy and Image Analysis, University of ZurichZurichSwitzerland
| | - Collin Yvès Ewald
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH ZürichSchwerzenbachSwitzerland
| |
Collapse
|
15
|
Ewald CY, Nyström A. Mechanotransduction through hemidesmosomes during aging and longevity. J Cell Sci 2023; 136:jcs260987. [PMID: 37522320 DOI: 10.1242/jcs.260987] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023] Open
Abstract
Hemidesmosomes are structural protein complexes localized at the interface of tissues with high mechanical demand and shear forces. Beyond tissue anchoring, hemidesmosomes have emerged as force-modulating structures important for translating mechanical cues into biochemical and transcriptional adaptation (i.e. mechanotransduction) across tissues. Here, we discuss the recent insights into the roles of hemidesmosomes in age-related tissue regeneration and aging in C. elegans, mice and humans. We highlight the emerging concept of preserved dynamic mechanoregulation of hemidesmosomes in tissue maintenance and healthy aging.
Collapse
Affiliation(s)
- Collin Y Ewald
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Zürich, Schwerzenbach CH-8603, Switzerland
| | - Alexander Nyström
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, Freiburg DE-79104, Germany
- Freiburg Institute for Advanced Studies (FRIAS), Albertstraße 19, Freiburg im Breisgau DE-79104, Germany
| |
Collapse
|
16
|
Park JYC, King A, Björk V, English BW, Fedintsev A, Ewald CY. Strategic outline of interventions targeting extracellular matrix for promoting healthy longevity. Am J Physiol Cell Physiol 2023; 325:C90-C128. [PMID: 37154490 DOI: 10.1152/ajpcell.00060.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/28/2023] [Accepted: 04/28/2023] [Indexed: 05/10/2023]
Abstract
The extracellular matrix (ECM), composed of interlinked proteins outside of cells, is an important component of the human body that helps maintain tissue architecture and cellular homeostasis. As people age, the ECM undergoes changes that can lead to age-related morbidity and mortality. Despite its importance, ECM aging remains understudied in the field of geroscience. In this review, we discuss the core concepts of ECM integrity, outline the age-related challenges and subsequent pathologies and diseases, summarize diagnostic methods detecting a faulty ECM, and provide strategies targeting ECM homeostasis. To conceptualize this, we built a technology research tree to hierarchically visualize possible research sequences for studying ECM aging. This strategic framework will hopefully facilitate the development of future research on interventions to restore ECM integrity, which could potentially lead to the development of new drugs or therapeutic interventions promoting health during aging.
Collapse
Affiliation(s)
- Ji Young Cecilia Park
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach, Switzerland
| | - Aaron King
- Foresight Institute, San Francisco, California, United States
| | | | - Bradley W English
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | | | - Collin Y Ewald
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach, Switzerland
| |
Collapse
|
17
|
Statzer C, Park JYC, Ewald CY. Extracellular Matrix Dynamics as an Emerging yet Understudied Hallmark of Aging and Longevity. Aging Dis 2023; 14:670-693. [PMID: 37191434 DOI: 10.14336/ad.2022.1116] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/16/2022] [Indexed: 05/17/2023] Open
Abstract
The biomechanical properties of extracellular matrices (ECM) and their consequences for cellular homeostasis have recently emerged as a driver of aging. Here we review the age-dependent deterioration of ECM in the context of our current understanding of the aging processes. We discuss the reciprocal interactions of longevity interventions with ECM remodeling. And the relevance of ECM dynamics captured by the matrisome and the matreotypes associated with health, disease, and longevity. Furthermore, we highlight that many established longevity compounds promote ECM homeostasis. A large body of evidence for the ECM to qualify as a hallmark of aging is emerging, and the data in invertebrates is promising. However, direct experimental proof that activating ECM homeostasis is sufficient to slow aging in mammals is lacking. We conclude that further research is required and anticipate that a conceptual framework for ECM biomechanics and homeostasis will provide new strategies to promote health during aging.
Collapse
Affiliation(s)
- Cyril Statzer
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach CH-8603, Switzerland
| | - Ji Young Cecilia Park
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach CH-8603, Switzerland
| | - Collin Y Ewald
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach CH-8603, Switzerland
| |
Collapse
|
18
|
Singh AK, Peng BY, Chien ST, Chan CH, Deng YH, Pai HY, Wei HJ, Wang MF, Wang SH, Wu CY, Deng WP. Anti-aging biomaterial sturgeon chondroitin sulfate upregulating anti-oxidant and SIRT-1/c-fos gene expression to reprogram stem cell senescence and prolong longevity. Biomater Sci 2023. [PMID: 37158091 DOI: 10.1039/d2bm01997c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Aging involves tissue and cell potential dysfunction characterized by stem cell senescence and extracellular matrix microenvironment (ECM) alteration. Chondroitin sulfate (CS), found in the ECM of normal cells and tissues, aids in maintaining tissue homeostasis. Here, CS-derived biomaterial (CSDB) from sturgeon is extracted to investigate its antiaging effect in senescence-accelerated mouse prone-8 (SAMP8) mice and elucidate the underlying mechanism of its action. Although CSDB has been widely extracted from different sources and used as a scaffold, hydrogel, or drug carrier for the treatment of various pathological diseases, CSDB has not yet been used as a biomaterial for the amelioration of senescence and aging features. In this study, the extracted sturgeon CSDB showed a low molecular weight and comprised 59% 4-sulfated CS and 23% 6-sulfated CS. In an in vitro study, sturgeon CSDB promoted cell proliferation and reduced oxidative stress to inhibit stem cell senescence. In an ex vivo study, after oral CSDB treatment of SAMP8 mice, the stem cells were extracted to analyze the p16Ink4a and p19Arf gene-related pathways, which were inhibited and then SIRT-1 gene expression was upregulated to reprogram stem cells from a senescence state for retarding aging. In an in vivo study, CSDB also restored the aging-phenotype-related bone mineral density and skin morphology to prolong longevity. Thus, sturgeon CSDB may be useful for prolonging healthy longevity as an anti-aging drug.
Collapse
Affiliation(s)
- Abhinay Kumar Singh
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan.
| | - Bou-Yue Peng
- Department of Dentistry, Taipei Medical University Hospital, Taipei 110301, Taiwan
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Shaw-Ting Chien
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan.
| | - Chun-Hao Chan
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan.
| | - Yue-Hua Deng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan.
| | - Hsiao-Yu Pai
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan.
- Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei 110301, Taiwan
| | - Hong-Jian Wei
- Department of Radiation Oncology, Columbia University Irving Medical Center, 630 W. 168th Street, New York, NY, 10032, USA
| | - Ming-Fu Wang
- Department of Food and Nutrition, Providence University, Taichung 433303, Taiwan
| | - Shwu-Huey Wang
- Core Facility Center, Department of Research Development, Taipei Medical University, Taipei 11030, Taiwan
| | - Chia-Yu Wu
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Taipei Medical University Hospital, Taipei 110301, Taiwan.
| | - Win-Ping Deng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan.
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, Taipei 242062, Taiwan
| |
Collapse
|
19
|
Vidovic T, Dakhovnik A, Hrabovskyi O, MacArthur MR, Ewald CY. AI-Predicted mTOR Inhibitor Reduces Cancer Cell Proliferation and Extends the Lifespan of C. elegans. Int J Mol Sci 2023; 24:ijms24097850. [PMID: 37175557 PMCID: PMC10177929 DOI: 10.3390/ijms24097850] [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: 02/09/2023] [Revised: 04/21/2023] [Accepted: 04/22/2023] [Indexed: 05/15/2023] Open
Abstract
The mechanistic target of rapamycin (mTOR) kinase is one of the top drug targets for promoting health and lifespan extension. Besides rapamycin, only a few other mTOR inhibitors have been developed and shown to be capable of slowing aging. We used machine learning to predict novel small molecules targeting mTOR. We selected one small molecule, TKA001, based on in silico predictions of a high on-target probability, low toxicity, favorable physicochemical properties, and preferable ADMET profile. We modeled TKA001 binding in silico by molecular docking and molecular dynamics. TKA001 potently inhibits both TOR complex 1 and 2 signaling in vitro. Furthermore, TKA001 inhibits human cancer cell proliferation in vitro and extends the lifespan of Caenorhabditis elegans, suggesting that TKA001 is able to slow aging in vivo.
Collapse
Affiliation(s)
- Tinka Vidovic
- Tinka Therapeutics, Fra Ivana Rozica 7, 21276 Vrgorac, Croatia
| | - Alexander Dakhovnik
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, CH-8603 Schwerzenbach, Switzerland
| | - Oleksii Hrabovskyi
- Palladin Institute of Biochemistry of the NAS of Ukraine, 02000 Kyiv, Ukraine
| | - Michael R MacArthur
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08540, USA
| | - Collin Y Ewald
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, CH-8603 Schwerzenbach, Switzerland
| |
Collapse
|
20
|
Goyala A, Ewald CY. CRISPR-activated expression of collagen col-120 increases lifespan and heat tolerance. MICROPUBLICATION BIOLOGY 2023; 2023:10.17912/micropub.biology.000730. [PMID: 37122503 PMCID: PMC10133990 DOI: 10.17912/micropub.biology.000730] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 04/05/2023] [Accepted: 04/11/2023] [Indexed: 05/02/2023]
Abstract
Transgenic overexpression of collagen col-120 increases the lifespan of C. elegans . However, whether post-developmental enhancement of collagen expression could also increase the lifespan is unknown. Recently, we described a method to induce the expression of a target gene using catalytically dead Cas9 (dCas9)-engineered C. elegans via ingestion of bacteria expressing a pair of promoter-specific single guide RNAs (sgRNA). Here, we cloned col-120 promoter-specific sgRNA oligo pair into L4440-Biobrick-sgRNA and fed these bacteria to dCas9::VP64 transgenic C. elegans . We observed a similar percentage of lifespan extension by post-developmentally dCas9-induced expression of col-120 , as previously reported through transgenic overexpression of col-120 . Consistent with this result is that induction of another previously shown longevity-promoting collagen, col-10 , also increased lifespan. Furthermore, we found an enhanced resilience to heat stress and increased expression of hsp-16.2 upon dCas9-activated col-120 expression. Together, these results provide an orthogonal method to validate longevity by enhancing col-120 expression and point towards a potential role of collagen enhancement in thermotolerance.
Collapse
Affiliation(s)
- Anita Goyala
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach CH-8603, Switzerland
| | - Collin Y. Ewald
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach CH-8603, Switzerland
| |
Collapse
|
21
|
Salamito M, Gillet B, Syx D, Vaganay E, Malbouyres M, Cerutti C, Tissot N, Exbrayat-Héritier C, Perez P, Jones C, Hughes S, Malfait F, Haydont V, Jäger S, Ruggiero F. NRF2 Shortage in Human Skin Fibroblasts Dysregulates Matrisome Gene Expression and Affects Collagen Fibrillogenesis. J Invest Dermatol 2023; 143:386-397.e12. [PMID: 38487918 DOI: 10.1016/j.jid.2022.07.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 10/14/2022]
Abstract
NRF2 is a master regulator of the antioxidative response that was recently proposed as a potential regulator of extracellular matrix (ECM) gene expression. Fibroblasts are major ECM producers in all connective tissues, including the dermis. A better understanding of NRF2-mediated ECM regulation in skin fibroblasts is thus of great interest for skin homeostasis maintenance and aging protection. In this study, we investigate the impact of NRF2 downregulation on matrisome gene expression and ECM deposits in human primary dermal fibroblasts. RNA-sequencing‒based transcriptome analysis of NRF2 silenced dermal fibroblasts shows that ECM genes are the most regulated gene sets, highlighting the relevance of the NRF2-mediated matrisome program in these cells. Using complementary light and electron microscopy methods, we show that NRF2 deprivation in dermal fibroblasts results in reduced collagen I biosynthesis and impacts collagen fibril deposition. Moreover, we identify ZNF469, a putative transcriptional regulator of collagen biosynthesis, as a target of NRF2. Both ZNF469 silenced fibroblasts and fibroblasts derived from Brittle Corneal Syndrome patients carrying variants in ZNF469 gene show reduced collagen I gene expression. Our study shows that NRF2 orchestrates matrisome expression in human skin fibroblasts through direct or indirect transcriptional mechanisms that could be prioritized to target dermal ECM homeostasis in health and disease.
Collapse
Affiliation(s)
- Mélanie Salamito
- Université de Lyon, École Normal Supérieure de Lyon (ENSL), Centre National de la Recherche Scientifique (CNRS), Institut de Génomique Fonctionnelle de Lyon (IGFL), Université Claude Bernard Lyon 1, Lyon, France; L'Oréal Research & Innovation, Aulnay-sous-Bois, France
| | - Benjamin Gillet
- Université de Lyon, École Normal Supérieure de Lyon (ENSL), Centre National de la Recherche Scientifique (CNRS), Institut de Génomique Fonctionnelle de Lyon (IGFL), Université Claude Bernard Lyon 1, Lyon, France
| | - Delfien Syx
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, Ghent University, Ghent, Belgium; Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Elisabeth Vaganay
- Université de Lyon, École Normal Supérieure de Lyon (ENSL), Centre National de la Recherche Scientifique (CNRS), Institut de Génomique Fonctionnelle de Lyon (IGFL), Université Claude Bernard Lyon 1, Lyon, France
| | - Marilyne Malbouyres
- Université de Lyon, École Normal Supérieure de Lyon (ENSL), Centre National de la Recherche Scientifique (CNRS), Institut de Génomique Fonctionnelle de Lyon (IGFL), Université Claude Bernard Lyon 1, Lyon, France
| | - Catherine Cerutti
- Université de Lyon, École Normal Supérieure de Lyon (ENSL), Centre National de la Recherche Scientifique (CNRS), Institut de Génomique Fonctionnelle de Lyon (IGFL), Université Claude Bernard Lyon 1, Lyon, France
| | | | - Chloé Exbrayat-Héritier
- Université de Lyon, École Normal Supérieure de Lyon (ENSL), Centre National de la Recherche Scientifique (CNRS), Institut de Génomique Fonctionnelle de Lyon (IGFL), Université Claude Bernard Lyon 1, Lyon, France
| | | | | | - Sandrine Hughes
- Université de Lyon, École Normal Supérieure de Lyon (ENSL), Centre National de la Recherche Scientifique (CNRS), Institut de Génomique Fonctionnelle de Lyon (IGFL), Université Claude Bernard Lyon 1, Lyon, France
| | - Fransiska Malfait
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, Ghent University, Ghent, Belgium; Ghent University Hospital, Ghent University, Ghent, Belgium
| | | | - Sibylle Jäger
- L'Oréal Research & Innovation, Aulnay-sous-Bois, France
| | - Florence Ruggiero
- Université de Lyon, École Normal Supérieure de Lyon (ENSL), Centre National de la Recherche Scientifique (CNRS), Institut de Génomique Fonctionnelle de Lyon (IGFL), Université Claude Bernard Lyon 1, Lyon, France.
| |
Collapse
|
22
|
Bennett DF, Goyala A, Statzer C, Beckett CW, Tyshkovskiy A, Gladyshev VN, Ewald CY, de Magalhães JP. Rilmenidine extends lifespan and healthspan in Caenorhabditis elegans via a nischarin I1-imidazoline receptor. Aging Cell 2023; 22:e13774. [PMID: 36670049 PMCID: PMC9924948 DOI: 10.1111/acel.13774] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 12/02/2022] [Accepted: 12/09/2022] [Indexed: 01/22/2023] Open
Abstract
Repurposing drugs capable of extending lifespan and health span has a huge untapped potential in translational geroscience. Here, we searched for known compounds that elicit a similar gene expression signature to caloric restriction and identified rilmenidine, an I1-imidazoline receptor agonist and prescription medication for the treatment of hypertension. We then show that treating Caenorhabditis elegans with rilmenidine at young and older ages increases lifespan. We also demonstrate that the stress-resilience, health span, and lifespan benefits of rilmenidine treatment in C. elegans are mediated by the I1-imidazoline receptor nish-1, implicating this receptor as a potential longevity target. Consistent with the shared caloric-restriction-mimicking gene signature, supplementing rilmenidine to calorically restricted C. elegans, genetic reduction of TORC1 function, or rapamycin treatment did not further increase lifespan. The rilmenidine-induced longevity required the transcription factors FOXO/DAF-16 and NRF1,2,3/SKN-1. Furthermore, we find that autophagy, but not AMPK signaling, was needed for rilmenidine-induced longevity. Moreover, transcriptional changes similar to caloric restriction were observed in liver and kidney tissues in mice treated with rilmenidine. Together, these results reveal a geroprotective and potential caloric restriction mimetic effect by rilmenidine that warrant fresh lines of inquiry into this compound.
Collapse
Affiliation(s)
- Dominic F. Bennett
- Integrative Genomics of Ageing GroupInstitute of Ageing and Chronic Disease, University of LiverpoolLiverpoolUK
| | - Anita Goyala
- Department of Health Sciences and Technology, Laboratory of Extracellular Matrix RegenerationInstitute of Translational Medicine, ETH ZürichSchwerzenbachSwitzerland
| | - Cyril Statzer
- Department of Health Sciences and Technology, Laboratory of Extracellular Matrix RegenerationInstitute of Translational Medicine, ETH ZürichSchwerzenbachSwitzerland
| | - Charles W. Beckett
- Integrative Genomics of Ageing GroupInstitute of Ageing and Chronic Disease, University of LiverpoolLiverpoolUK
| | - Alexander Tyshkovskiy
- Division of Genetics, Department of MedicineBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA,Belozersky Institute of Physico‐Chemical BiologyMoscow State UniversityMoscowRussia
| | - Vadim N. Gladyshev
- Division of Genetics, Department of MedicineBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Collin Y. Ewald
- Department of Health Sciences and Technology, Laboratory of Extracellular Matrix RegenerationInstitute of Translational Medicine, ETH ZürichSchwerzenbachSwitzerland
| | - João Pedro de Magalhães
- Integrative Genomics of Ageing GroupInstitute of Ageing and Chronic Disease, University of LiverpoolLiverpoolUK,Present address:
Institute of Inflammation and AgeingUniversity of Birmingham, Queen Elizabeth HospitalBirminghamUK
| |
Collapse
|
23
|
López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. Hallmarks of aging: An expanding universe. Cell 2023; 186:243-278. [PMID: 36599349 DOI: 10.1016/j.cell.2022.11.001] [Citation(s) in RCA: 2196] [Impact Index Per Article: 1098.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/19/2022] [Accepted: 11/01/2022] [Indexed: 01/05/2023]
Abstract
Aging is driven by hallmarks fulfilling the following three premises: (1) their age-associated manifestation, (2) the acceleration of aging by experimentally accentuating them, and (3) the opportunity to decelerate, stop, or reverse aging by therapeutic interventions on them. We propose the following twelve hallmarks of aging: genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, disabled macroautophagy, deregulated nutrient-sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, altered intercellular communication, chronic inflammation, and dysbiosis. These hallmarks are interconnected among each other, as well as to the recently proposed hallmarks of health, which include organizational features of spatial compartmentalization, maintenance of homeostasis, and adequate responses to stress.
Collapse
Affiliation(s)
- Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
| | - Maria A Blasco
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain
| | - Linda Partridge
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London, London, UK; Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Manuel Serrano
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain; Altos Labs, Cambridge, UK
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France; Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| |
Collapse
|
24
|
Loose JA, Amrit FRG, Patil T, Yanowitz JL, Ghazi A. Meiotic dysfunction accelerates somatic aging in Caenorhabditis elegans. Aging Cell 2022; 21:e13716. [PMID: 36176234 PMCID: PMC9649607 DOI: 10.1111/acel.13716] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 08/07/2022] [Accepted: 08/31/2022] [Indexed: 01/25/2023] Open
Abstract
An expanding body of evidence, from studies in model organisms to human clinical data, reveals that reproductive health influences organismal aging. However, the impact of germline integrity on somatic aging is poorly understood. Moreover, assessing the causal relationship of such an impact is challenging to address in human and vertebrate models. Here, we demonstrate that disruption of meiosis, a germline restricted process, shortened lifespan, impaired individual aspects of healthspan, and accelerated somatic aging in Caenorhabditis elegans. Young meiotic mutants exhibited transcriptional profiles that showed remarkable overlap with the transcriptomes of old worms and shared similarities with transcriptomes of aging human tissues as well. We found that meiosis dysfunction caused increased expression of functionally relevant longevity determinants whose inactivation enhanced the lifespan of normal animals. Further, meiotic mutants manifested destabilized protein homeostasis and enhanced proteasomal activity partially rescued the associated lifespan defects. Our study demonstrates a role for meiotic integrity in controlling somatic aging and reveals proteostasis control as a potential mechanism through which germline status impacts overall organismal health.
Collapse
Affiliation(s)
- Julia A. Loose
- Department of Pediatrics, John G. Rangos Sr. Research CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Francis R. G. Amrit
- Department of Pediatrics, John G. Rangos Sr. Research CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Thayjas Patil
- Department of Pediatrics, John G. Rangos Sr. Research CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Judith L. Yanowitz
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Magee‐Womens Research InstituteUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Arjumand Ghazi
- Department of Pediatrics, John G. Rangos Sr. Research CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA,Department of Developmental Biology, John G. Rangos Sr. Research CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA,Department of Cell Biology & PhysiologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| |
Collapse
|
25
|
Vidović T, Ewald CY. Longevity-Promoting Pathways and Transcription Factors Respond to and Control Extracellular Matrix Dynamics During Aging and Disease. FRONTIERS IN AGING 2022; 3:935220. [PMID: 35874275 PMCID: PMC9301135 DOI: 10.3389/fragi.2022.935220] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 05/27/2022] [Indexed: 05/28/2023]
Abstract
Aging is one of the largest risk factors for cancer, type 2 diabetes, osteoarthritis, cardiovascular diseases, and other age-related pathologies. Here, we give a detailed description of the interplay of chronic age-related pathologies with the remodeling of the extracellular matrix during disease development and progression. Longevity-promoting signaling pathways slow or prevent age-related diseases. In particular, we focus on the mTOR signaling pathway, sirtuins, and canonical longevity-promoting transcription factors, such as FOXO, NF-κB, and Nrf2. We extend our analysis using chromatin immunoprecipitation (ChIP) sequencing and transcriptomic data and report that many established and emerging longevity-promoting transcription factors, such as CREB1, FOXO1,3, GATA1,2,3,4, HIF1A, JUN, KLF4, MYC, NFE2L2/Nrf2, RELA/NF-κB, REST, STAT3,5A, and TP53/p53, directly regulate many extracellular matrix genes and remodelers. We propose that modulation of these pathways increases lifespan and protects from age-related diseases in part due to their effects on extracellular matrix remodeling. Therefore, to successfully treat age-related diseases, it is necessary to better understand the connection between extracellular matrix components and longevity pathways.
Collapse
Affiliation(s)
| | - Collin Y. Ewald
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
26
|
Meron E, Thaysen M, Angeli S, Antebi A, Barzilai N, Baur JA, Bekker-Jensen S, Birkisdottir M, Bischof E, Bruening J, Brunet A, Buchwalter A, Cabreiro F, Cai S, Chen BH, Ermolaeva M, Ewald CY, Ferrucci L, Florian MC, Fortney K, Freund A, Georgievskaya A, Gladyshev VN, Glass D, Golato T, Gorbunova V, Hoejimakers J, Houtkooper RH, Jager S, Jaksch F, Janssens G, Jensen MB, Kaeberlein M, Karsenty G, de Keizer P, Kennedy B, Kirkland JL, Kjaer M, Kroemer G, Lee KF, Lemaitre JM, Liaskos D, Longo VD, Lu YX, MacArthur MR, Maier AB, Manakanatas C, Mitchell SJ, Moskalev A, Niedernhofer L, Ozerov I, Partridge L, Passegué E, Petr MA, Peyer J, Radenkovic D, Rando TA, Rattan S, Riedel CG, Rudolph L, Ai R, Serrano M, Schumacher B, Sinclair DA, Smith R, Suh Y, Taub P, Trapp A, Trendelenburg AU, Valenzano DR, Verburgh K, Verdin E, Vijg J, Westendorp RGJ, Zonari A, Bakula D, Zhavoronkov A, Scheibye-Knudsen M. Meeting Report: Aging Research and Drug Discovery. Aging (Albany NY) 2022. [PMID: 35089871 PMCID: PMC8833115 DOI: 10.18632/aging.203859] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Aging is the single largest risk factor for most chronic diseases, and thus possesses large socioeconomic interest to continuously aging societies. Consequently, the field of aging research is expanding alongside a growing focus from the industry and investors in aging research. This year’s 8th Annual Aging Research and Drug Discovery (ARDD) meeting was organized as a hybrid meeting from August 30th to September 3rd 2021 with more than 130 attendees participating on-site at the Ceremonial Hall at University of Copenhagen, Denmark, and 1800 engaging online. The conference comprised of presentations from 75 speakers focusing on new research in topics including mechanisms of aging and how these can be modulated as well as the use of AI and new standards of practices within aging research. This year, a longevity workshop was included to build stronger connections with the clinical community.
Collapse
Affiliation(s)
- Esther Meron
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Maria Thaysen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Suzanne Angeli
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Adam Antebi
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Nir Barzilai
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Institute for Aging Research, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Joseph A Baur
- Smilow Center for Translational Research, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Simon Bekker-Jensen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Maria Birkisdottir
- Department of Molecular Genetics, Erasmus MC, Rotterdam, Netherlands.,Department of Neuroscience, Erasmus MC, Rotterdam, Netherlands
| | - Evelyne Bischof
- Shanghai University of Medicine and Health Sciences, College of Clinical Medicine, Shanghai, China
| | - Jens Bruening
- Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Anne Brunet
- Department of Genetics, Stanford School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Abigail Buchwalter
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Filipe Cabreiro
- Institute of Clinical Sciences, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK.,CECAD Research Center, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Shiqing Cai
- Institute of Neuroscience, Chinese Academy of Science, Shanghai, China
| | - Brian H Chen
- FOXO Technologies Inc, Minneapolis, MN 55402, USA.,The Herbert Wertheim School of Public Health and Human Longevity Science, UC San Diego, La Jolla, CA 92093, USA
| | | | - Collin Y Ewald
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach CH-8603, Switzerland
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | | | | | - Adam Freund
- Arda Therapeutics, San Carlos, CA 94070, USA
| | | | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - David Glass
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | | | - Vera Gorbunova
- Departments of Biology and Medicine, University of Rochester, Rochester, NY 14627, USA
| | - Jan Hoejimakers
- Department of Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Sibylle Jager
- L'Oréal Research and Innovation, Aulnay-sous-Bois, France
| | | | - Georges Janssens
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Matt Kaeberlein
- Departments of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Gerard Karsenty
- Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Peter de Keizer
- Department of Molecular Cancer Research, Center for Molecular Medicine, Division of Biomedical Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Brian Kennedy
- Buck Institute for Research on Aging, Novato, CA 94945, USA.,Departments of Biochemistry and Physiology, Yong Loo Lin School of Medicine, National University Singapore, Singapore.,Center for Healthy Longevity, National University Health System, Singapore
| | - James L Kirkland
- Division of General Internal Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Michael Kjaer
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Université de Paris, Sorbonne Université, Inserm U1138, Paris, France
| | - Kai-Fu Lee
- Sinovation Ventures and Sinovation AI Institute, Beijing, China
| | - Jean-Marc Lemaitre
- Institute for Regenerative Medicine and Biotherapies, INSERM UMR 1183, Montpellier, France
| | | | - Valter D Longo
- USC Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Yu-Xuan Lu
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Michael R MacArthur
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Andrea B Maier
- Center for Healthy Longevity, National University Health System, Singapore.,Department of Human Movement Sciences, @AgeAmsterdam, Faculty of Behavioural and Movement Sciences, Amsterdam Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Department of Medicine, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | | | - Sarah J Mitchell
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Alexey Moskalev
- Institute of Biology of FRC Komi Science Center of Ural Division of RAS, Syktyvkar, Russia.,Russian Clinical and Research Center of Gerontology, Moscow, Russia
| | - Laura Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ivan Ozerov
- Insilico Medicine, Hong Kong Science and Technology Park, Hong Kong
| | - Linda Partridge
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | | | - Michael A Petr
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark.,Tracked.bio, Copenhagen, Denmark
| | | | - Dina Radenkovic
- Hooke London by Health and Longevity Optimisation, London, UK
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences and Paul F. Glenn Center for Biology of Aging, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Suresh Rattan
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Christian G Riedel
- Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden
| | | | - Ruixue Ai
- Department of Clinical Molecular Biology
- UiO, University of Oslo and Akershus University Hospital, Norway
| | - Manuel Serrano
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Björn Schumacher
- CECAD Research Center, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - David A Sinclair
- Blavatnik Institute, Department of Genetics, Paul F. Glenn Center for Biology of Aging Research at Harvard Medical School, Boston, MA 94107, USA
| | | | - Yousin Suh
- Departments of Obstetrics and Gynecology, Genetics and Development, Columbia University, New York, NY 10027, USA
| | - Pam Taub
- Division of Cardiovascular Medicine, University of California, San Diego, CA 92093, USA
| | - Alexandre Trapp
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Dario Riccardo Valenzano
- Max Planck Institute for Biology of Ageing, Cologne, Germany.,Leibniz Institute on Aging, Jena, Germany
| | | | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Jan Vijg
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | - Daniela Bakula
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Alex Zhavoronkov
- Insilico Medicine, Hong Kong Science and Technology Park, Hong Kong
| | - Morten Scheibye-Knudsen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
27
|
Shen X, Wu B, Jiang W, Li Y, Zhang Y, Zhao K, Nie N, Gong L, Liu Y, Zou X, Liu J, Jin J, Ouyang H. Scale bar of aging trajectories for screening personal rejuvenation treatments. Comput Struct Biotechnol J 2022; 20:5750-5760. [DOI: 10.1016/j.csbj.2022.10.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/15/2022] [Accepted: 10/15/2022] [Indexed: 11/27/2022] Open
|
28
|
Carlston C, Weinmann R, Stec N, Abbatemarco S, Schwager F, Wang J, Ouyang H, Ewald CY, Gotta M, Hammell CM. PQN-59 antagonizes microRNA-mediated repression during post-embryonic temporal patterning and modulates translation and stress granule formation in C. elegans. PLoS Genet 2021; 17:e1009599. [PMID: 34807903 PMCID: PMC8648105 DOI: 10.1371/journal.pgen.1009599] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 12/06/2021] [Accepted: 10/27/2021] [Indexed: 12/14/2022] Open
Abstract
microRNAs (miRNAs) are potent regulators of gene expression that function in a variety of developmental and physiological processes by dampening the expression of their target genes at a post-transcriptional level. In many gene regulatory networks (GRNs), miRNAs function in a switch-like manner whereby their expression and activity elicit a transition from one stable pattern of gene expression to a distinct, equally stable pattern required to define a nascent cell fate. While the importance of miRNAs that function in this capacity are clear, we have less of an understanding of the cellular factors and mechanisms that ensure the robustness of this form of regulatory bistability. In a screen to identify suppressors of temporal patterning phenotypes that result from ineffective miRNA-mediated target repression, we identified pqn-59, an ortholog of human UBAP2L, as a novel factor that antagonizes the activities of multiple heterochronic miRNAs. Specifically, we find that depletion of pqn-59 can restore normal development in animals with reduced lin-4 and let-7-family miRNA activity. Importantly, inactivation of pqn-59 is not sufficient to bypass the requirement of these regulatory RNAs within the heterochronic GRN. The pqn-59 gene encodes an abundant, cytoplasmically-localized, unstructured protein that harbors three essential "prion-like" domains. These domains exhibit LLPS properties in vitro and normally function to limit PQN-59 diffusion in the cytoplasm in vivo. Like human UBAP2L, PQN-59's localization becomes highly dynamic during stress conditions where it re-distributes to cytoplasmic stress granules and is important for their formation. Proteomic analysis of PQN-59 complexes from embryonic extracts indicates that PQN-59 and human UBAP2L interact with orthologous cellular components involved in RNA metabolism and promoting protein translation and that PQN-59 additionally interacts with proteins involved in transcription and intracellular transport. Finally, we demonstrate that pqn-59 depletion reduces protein translation and also results in the stabilization of several mature miRNAs (including those involved in temporal patterning). These data suggest that PQN-59 may ensure the bistability of some GRNs that require miRNA functions by promoting miRNA turnover and, like UBAP2L, enhancing protein translation.
Collapse
Affiliation(s)
- Colleen Carlston
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, United States of America
| | - Robin Weinmann
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, United States of America
| | - Natalia Stec
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, United States of America
| | - Simona Abbatemarco
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Francoise Schwager
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Jing Wang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, United States of America
| | - Huiwu Ouyang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, United States of America
| | - Collin Y. Ewald
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach, Switzerland
| | - Monica Gotta
- Department of Cellular Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | |
Collapse
|
29
|
Ewald CY. Drug Screening Implicates Chondroitin Sulfate as a Potential Longevity Pill. FRONTIERS IN AGING 2021; 2:741843. [PMID: 35821992 PMCID: PMC9261418 DOI: 10.3389/fragi.2021.741843] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 08/26/2021] [Indexed: 11/13/2022]
Abstract
Discovering compounds that promote health during aging ("geroprotectors") is key to the retardation of age-related pathologies and the prevention of chronic age-related diseases. In in-silico and model organisms' lifespan screens, chondroitin sulfate has emerged as a geroprotective compound. Chondroitin sulfate is a glycosaminoglycan attached to extracellular matrix proteins and is naturally produced by our body. Oral supplementation of chondroitin sulfate shows a high tolerance in humans, preferable pharmacokinetics, a positive correlation with healthy human longevity, and efficacy in deceleration of age-related diseases in randomized clinical trials. We have recently shown that chondroitin sulfate supplementation increases the lifespan of C. elegans. Thus, chondroitin sulfate holds the potential to become a geroprotective strategy to promote health during human aging. This review discusses the two major potential mechanisms of action, extracellular matrix homeostasis and inhibition of inflammation, that counteract age-related pathologies upon chondroitin sulfate supplementation.
Collapse
|