1
|
Goyal A, Afzal M, Khan NH, Goyal K, Srinivasamurthy SK, Gupta G, Benod Kumar K, Ali H, Rana M, Wong LS, Kumarasamy V, Subramaniyan V. Targeting p53-p21 signaling to enhance mesenchymal stem cell regenerative potential. Regen Ther 2025; 29:352-363. [PMID: 40248767 PMCID: PMC12004386 DOI: 10.1016/j.reth.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/09/2025] [Accepted: 03/18/2025] [Indexed: 04/19/2025] Open
Abstract
Mesenchymal stem cells (MSCs) are properties of self-renewal and differentiation potentials and thus are very appealing to regenerative medicine. Nevertheless, their therapeutic potential is frequently constrained by senescence, limited proliferation, and stress-induced apoptosis. The key role of the p53-p21 biology in MSC biology resides in safeguarding genomic stability while promoting senescence and limiting regenerative capacity upon over-activation demonstrated. This pathway is a key point for improving MSC function and exploiting the inherent limitations. Recent advances indicate that senescence can be delayed by targeting the p53-p21 signaling and improved MSC proliferation and differentiation capacity. PFT-α pharmacological agents transiently inhibit p53 from increasing proliferation and lineage-specific differentiation, while antioxidants such as hydrogen-rich saline and epigallocatechin 3 gallate (EGCG) suppress oxidative stress and attenuate p53 p21 signaling. Genetic tools like CRISPR-Cas9 and RNA interference also precisely modulate TP53 and CDKN1A expression to optimize MSC functionality. The interplay of p53-p21 with pathways like Wnt/β-catenin and MAPK further highlights opportunities for combinatorial therapies to enhance MSC resilience and regenerative outcomes. This review aims to offer a holistic view of how p53-p21 targeting can further the regenerative potential of MSCs, resolving senescence, proliferation, and stress resilience towards advanced therapeutics built on MSCs.
Collapse
Affiliation(s)
- Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Nawaid Hussain Khan
- Faculty of Medicine, Ala-Too International University, Bishkek, Kyrgyz Republic
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - Suresh Kumar Srinivasamurthy
- Department of Pharmacology, Ras Al Khaimah College of Medical Sciences, Ras Al Khaimah Medical & Health Sciences University, P.O. Box 11172, Ras Al Khaimah, United Arab Emirates
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - K. Benod Kumar
- Department of General Surgery, Consultant Head and Neck Surgical Oncology, Dr.D.Y.Patil Medical College, Hospital and Research Centre, Pimpri, Pune, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Mohit Rana
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Ling Shing Wong
- Faculty of Health and Life Sciences, INTI International University, Nilai 71800, Malaysia
| | - Vinoth Kumarasamy
- Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Vetriselvan Subramaniyan
- Division of Pharmacology, Faculty of Medical and Life Sciences, Sunway University, Bandar Sunway, 47500 Selangor Darul Ehsan, Malaysia
| |
Collapse
|
2
|
Wan X, Xu P, Zhou X, Liu J, Yang Y, Liang C, Wang J, Wang W, Xu F, Wan X, Kang J, Tong P, Xia H. Qi-Gu capsule alleviates osteoporosis by inhibiting mesenchymal stem cell senescence via the HIF-1α/AMPK axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156764. [PMID: 40252437 DOI: 10.1016/j.phymed.2025.156764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 04/02/2025] [Accepted: 04/11/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Osteoporosis (OP) represents a systemic disease causing reduced bone mass and fragility fractures. Qigu Capsule (QGC), a traditional Chinese medicine, shows potential in alleviating human OP, but its precise mechanisms remain unclear, limiting clinical application. METHODS The bioactive components of QGC were analyzed using high-performance liquid chromatography (HPLC). An ovariectomy (OVX)-provoked OP rat model was established to evaluate QGC's effects on bone mass, trabecular architecture, and mechanical strength using micro-CT, histological staining, and biomechanical testing. RNA-seq analysis of human OP-derived mesenchymal stem cell (MSC) samples was performed to identify oxidative stress (OxS)- and senescence-associated gene changes. OxS-induced MSC senescence was modeled in vitro using H₂O₂, and QGC's effects on MSC proliferation, migration, and osteogenic differentiation were assessed. Network pharmacology (NP) was deployed to predict the key mechanisms behind the QGC treatment of OP. Further mechanistic studies utilized pharmacological inhibitors and siRNA-mediated gene knockdown to confirm the involvement of critical signaling pathways. RESULTS HPLC-MS analysis identified 505 unique bioactive compounds in QGC. In vivo, QGC significantly improved BMD, enhanced trabecular microarchitecture, and restored mechanical properties in OVX rats. ELISA, histological, and immunohistochemical analyses confirmed that QGC primarily enhanced osteoblast activity. RNA-seq analysis of GEO datasets revealed upregulation of senescence and OxS markers (P53, CDKN1A, and INOS) in human OP-derived MSCs. Both in vivo and in vitro QGC alleviated OxS-induced MSC senescence, reduced reactive oxygen species (ROS) levels, suppressed senescence and OxS marker, and promoted MSC proliferation, migration, and osteogenic differentiation. Moreover, NP predicted HIF-1α signaling as critical in QGC's regulation of MSC function during OP. Mechanistic studies demonstrated that QGC activated the HIF-1α/AMPK axis, and inhibition of either HIF-1α or AMPK abolished its therapeutic effects. CONCLUSION QGC mitigates OxS-induced MSC senescence and promotes osteogenesis through the HIF-1α/AMPK axis, highlighting its mechanistic basis in treating OP. These findings show QGC's potential as a therapeutic agent, not only by promoting osteogenesis but also by complementing or serving as an alternative to current OP treatments, offering valuable prospects for enhanced clinical management.
Collapse
Affiliation(s)
- Xuan Wan
- Department of Orthopedics, Affiliated hospital of Jiangxi University of Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi province 330004, China; University Medicine Rostock, University of Rostock, Parkstr. 6, Rostock 18057, Germany
| | - Pengchao Xu
- Zhejiang Provincial Chinese Medicine Hospital (First affiliated hospital of Zhejiang Chinese Medical University), Zhejiang Chinese Medicine University, No. 548, Binwen Road, Binjiang District, Hangzhou City, Zhejiang Province 310053, China
| | - Xing Zhou
- Zhejiang Provincial Chinese Medicine Hospital (First affiliated hospital of Zhejiang Chinese Medical University), Zhejiang Chinese Medicine University, No. 548, Binwen Road, Binjiang District, Hangzhou City, Zhejiang Province 310053, China; Department of Sports Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiaotong University, Shanghai 200080, China
| | - Jiangyuan Liu
- Zhejiang Provincial Chinese Medicine Hospital (First affiliated hospital of Zhejiang Chinese Medical University), Zhejiang Chinese Medicine University, No. 548, Binwen Road, Binjiang District, Hangzhou City, Zhejiang Province 310053, China; Department of Orthopedics, Affiliated hospital of Jiangxi University of Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi province 330004, China
| | - Yiwen Yang
- Zhejiang Provincial Chinese Medicine Hospital (First affiliated hospital of Zhejiang Chinese Medical University), Zhejiang Chinese Medicine University, No. 548, Binwen Road, Binjiang District, Hangzhou City, Zhejiang Province 310053, China
| | - Chaoyi Liang
- Department of Orthopedics, Affiliated hospital of Jiangxi University of Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi province 330004, China
| | - Jinglei Wang
- Zhejiang Provincial Chinese Medicine Hospital (First affiliated hospital of Zhejiang Chinese Medical University), Zhejiang Chinese Medicine University, No. 548, Binwen Road, Binjiang District, Hangzhou City, Zhejiang Province 310053, China
| | - Weixiang Wang
- Zhejiang Provincial Chinese Medicine Hospital (First affiliated hospital of Zhejiang Chinese Medical University), Zhejiang Chinese Medicine University, No. 548, Binwen Road, Binjiang District, Hangzhou City, Zhejiang Province 310053, China
| | - Fengjiao Xu
- Zhejiang Provincial Chinese Medicine Hospital (First affiliated hospital of Zhejiang Chinese Medical University), Zhejiang Chinese Medicine University, No. 548, Binwen Road, Binjiang District, Hangzhou City, Zhejiang Province 310053, China
| | - Xiaoming Wan
- Department of Orthopedics, Affiliated hospital of Jiangxi University of Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi province 330004, China
| | - Jian Kang
- Department of Orthopedics, Affiliated hospital of Jiangxi University of Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi province 330004, China.
| | - Peijian Tong
- Zhejiang Provincial Chinese Medicine Hospital (First affiliated hospital of Zhejiang Chinese Medical University), Zhejiang Chinese Medicine University, No. 548, Binwen Road, Binjiang District, Hangzhou City, Zhejiang Province 310053, China.
| | - Hanting Xia
- Zhejiang Provincial Chinese Medicine Hospital (First affiliated hospital of Zhejiang Chinese Medical University), Zhejiang Chinese Medicine University, No. 548, Binwen Road, Binjiang District, Hangzhou City, Zhejiang Province 310053, China; Department of Orthopedics, Affiliated hospital of Jiangxi University of Chinese Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi province 330004, China.
| |
Collapse
|
3
|
Casorati B, Zafferri I, Castiglioni S, Maier JA. Replicative Senescence in Mesenchymal Stem Cells: An In Vitro Study on Mitochondrial Dynamics and Metabolic Alterations. Antioxidants (Basel) 2025; 14:446. [PMID: 40298797 PMCID: PMC12024194 DOI: 10.3390/antiox14040446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/03/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent progenitors capable of self-renewal and differentiation into various cell lineages, making them essential for tissue repair and regenerative medicine. However, their regenerative potential is constrained by replicative senescence, an irreversible growth arrest that occurs after a finite number of cell divisions. In this study, we serially passaged human bone marrow-derived MSCs (bMSCs) and compared young, pre-senescent, and senescent cells. The onset of senescence was accompanied by progressive alterations in mitochondrial dynamics, leading to a decline in mitochondrial membrane potential, and increased reactive oxygen species (ROS) production, alongside a diminished cellular antioxidant capacity. These mitochondrial defects play a role in metabolic reprogramming in senescent bMSCs. Our findings underscore the intricate interplay between ROS, mitochondrial dysfunction, and replicative senescence, offering valuable insights to guide the development of therapeutic strategies for preserving MSC functionality in aging and MSC-based therapies.
Collapse
Affiliation(s)
| | | | - Sara Castiglioni
- Department of Biomedical and Clinical Sciences, Università di Milano, 20157 Milano, Italy; (B.C.); (I.Z.); (J.A.M.)
| | | |
Collapse
|
4
|
Babu MA, Jyothi S R, Kaur I, Kumar S, Sharma N, Kumar MR, Rajput P, Ali H, Gupta G, Subramaniyan V, Wong LS, Kumarasamy V. The role of GATA4 in mesenchymal stem cell senescence: A new frontier in regenerative medicine. Regen Ther 2025; 28:214-226. [PMID: 39811069 PMCID: PMC11731776 DOI: 10.1016/j.reth.2024.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 11/21/2024] [Indexed: 01/16/2025] Open
Abstract
The Mesenchymal Stem Cell (MSC) is a multipotent progenitor cell with known differentiation potential towards various cell lineage, making it an appealing candidate for regenerative medicine. One major contributing factor to age-related MSC dysfunction is cellular senescence, which is the hallmark of relatively irreversible growth arrest and changes in functional properties. GATA4, a zinc-finger transcription factor, emerges as a critical regulator in MSC biology. Originally identified as a key regulator of heart development and specification, GATA4 has since been connected to several aspects of cellular processes, including stem cell proliferation and differentiation. Accumulating evidence suggests that the involvement of GATA4-nuclear signalizing in the process of MSC senescence-related traits may contribute to age-induced alterations in MSC behavior. GATA4 emerged as the central player in MSC senescence, interacting with several signaling pathways. Studies have shown that GATA4 expression is reduced with age in MSCs, which is associated with increased expression levels of senescence markers and impaired regenerative potential. At the mechanistic level, GATA4 regulates the expression of genes involved in cell cycle regulation, DNA repair, and oxidative stress response, thereby influencing the senescence phenotype in MSCs. The findings underscore the critical function of GATA4 in MSC homeostasis and suggest a promising new target to restore stem cell function during aging and disease. A better understanding of the molecular mechanisms that underlie GATA4 mediated modulation of MSC senescence would provide an opportunity to develop new therapies to revitalize old MSCs to increase their regenerative function for therapeutic purposes in regenerative medicine.
Collapse
Affiliation(s)
- M. Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, UP, India
| | - Renuka Jyothi S
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka, 560069, India
| | - Irwanjot Kaur
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Naveen Sharma
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali, 140307, Punjab, India
| | - M. Ravi Kumar
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | - Pranchal Rajput
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Vetriselvan Subramaniyan
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Malaysia
| | - Ling Shing Wong
- Faculty of Health and Life Sciences, INTI International University, Nilai, 71800, Malaysia
| | - Vinoth Kumarasamy
- Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000, Cheras, Kuala Lumpur, Malaysia
| |
Collapse
|
5
|
Lin ECY, Davis MP, Lee MS, Ma G, Xu W, Chang YI, Li WJ. Advancing immunomodulatory functions in mesenchymal stem/stromal cells through targeting the GATA6-mediated pathway. Cytotherapy 2025; 27:85-97. [PMID: 39207345 PMCID: PMC11668624 DOI: 10.1016/j.jcyt.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 07/18/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND AIMS The immunomodulatory capacity of mesenchymal stem/stromal cells (MSCs) is a key feature that makes them particularly valuable for regenerative medicine. However, this potential is affected by the chronological aging of the donors and the cell expansion procedures in culture. We have demonstrated that GATA binding protein 6 (GATA6) plays a pivotal role in the aging of MSCs and inhibiting GATA6 rejuvenates the characteristics of MSCs. METHODS In this study, we compared the immunomodulatory capabilities of young and old MSC models, using induced pluripotent stem cells-derived rejuvenated MSCs (rMSCs) and their parental MSCs (pMSCs), respectively, to identify a key mechanism involved in the differential regulation of these capabilities. Additionally, we explored the role of GATA6 in mediating the mechanism. RESULTS Our results demonstrated that rMSCs exhibited downregulated aging-associated regulators, including p53, p21 and GATA6, and showed enhanced suppression of T cell proliferation compared to pMSCs. Through analyzing our previous RNA-seq data and employing target gene knockdown, we determined both suppressors of cytokine signaling 3 (SOCS3) and interleukin 6 were involved in GATA6-induced regulation, collectively affecting the expression of programmed death ligand 1 (PDL1) in both pMSCs and rMSCs. CONCLUSIONS Our findings underline the significance of the GATA6/SOCS3/PDL1 pathway in regulating aging-associated changes in MSC immunomodulatory activity, providing valuable insights into the potential use of rMSCs in the treatment of immune diseases and regenerative medicine.
Collapse
Affiliation(s)
- Eric Chang-Yi Lin
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin, USA; Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei City, Taiwan
| | - Madison P Davis
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ming-Song Lee
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Gui Ma
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Wei Xu
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Yuan-I Chang
- Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei City, Taiwan
| | - Wan-Ju Li
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA.
| |
Collapse
|
6
|
Gao Q, Cekuc MS, Ergul YS, Pius AK, Shinohara I, Murayama M, Susuki Y, Ma C, Morita M, Chow SKH, Goodman SB. 3D Culture of MSCs for Clinical Application. Bioengineering (Basel) 2024; 11:1199. [PMID: 39768017 PMCID: PMC11726872 DOI: 10.3390/bioengineering11121199] [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: 10/30/2024] [Revised: 11/19/2024] [Accepted: 11/23/2024] [Indexed: 01/16/2025] Open
Abstract
Mesenchymal stem cells (MSCs) play an important role in regenerative medicine and drug discovery due to their multipotential differentiation capabilities and immunomodulatory effects. Compared with traditional 2D cultures of MSCs, 3D cultures of MSCs have emerged as an effective approach to enhance cell viability, proliferation, and functionality, and provide a more relevant physiological environment. Here, we review the therapeutic potential of 3D-cultured MSCs, highlighting their roles in tissue regeneration and repair and drug screening. We further summarize successful cases that apply 3D MSCs in modeling disease states, enabling the identification of novel therapeutic strategies. Despite these promising applications, we discuss challenges that remain in the clinical translation of 3D MSC technologies, including stability, cell heterogeneity, and regulatory issues. We conclude by addressing these obstacles and emphasizing the need for further research to fully exploit the potential of 3D MSCs in clinical practice.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94304, USA; (Q.G.)
| |
Collapse
|
7
|
Paola S, Lara G, Michela M, Silvia DC, Serena M, Rosalba P, Maria NA, Eleonora C, Fiorella C, Giulia G, Giovanna T, Giuseppe N, Federica S. When do the pathological signs become evident? Study of human mesenchymal stem cells in MDPL syndrome. Aging (Albany NY) 2024; 16:13505-13525. [PMID: 39611849 PMCID: PMC11723661 DOI: 10.18632/aging.206159] [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: 07/19/2024] [Accepted: 11/11/2024] [Indexed: 11/30/2024]
Abstract
Aging syndromes are rare genetic disorders sharing the features of accelerated senescence. Among these, Mandibular hypoplasia, Deafness and Progeroid features with concomitant Lipodystrophy (MDPL; OMIM #615381) is a rare autosomal dominant disease due to a de novo in-frame deletion in POLD1 gene, encoding the catalytic subunit of DNA polymerase delta. Here, we investigated how MSCs may contribute to the phenotypes and progression of premature aging syndromes such as MDPL. In human induced pluripotent stem cells (hiPSCs)-derived MSCs of three MDPL patients we detected several hallmarks of senescence, including (i) abnormal nuclear morphology, (ii) micronuclei presence, (iii) slow cell proliferation and cell cycle progression, (iv) reduced telomere length, and (v) increased levels of mitochondrial reactive oxygen species (ROS). We newly demonstrated that the pathological hallmarks of senescence manifest at an early stage of human development and represent a warning sign for the progression of the disease. Dissecting the mechanisms underlying stem cell dysfunction during aging can thereby contribute to the development of timely pharmacological therapies for ameliorating the pathological phenotype.
Collapse
Affiliation(s)
- Spitalieri Paola
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Guerrieri Lara
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Murdocca Michela
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Di Cesare Silvia
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Maccaroni Serena
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Pecorari Rosalba
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, Italy
| | | | - Candi Eleonora
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, Italy
| | - Colasuonno Fiorella
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, Italy
- Department of Science – LIME, Roma Tre University, Rome, Italy
| | - Gori Giulia
- Meyer Children’s Hospital IRCCS, Florence, Italy
| | | | - Novelli Giuseppe
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Sangiuolo Federica
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
8
|
Lee SS, Al Halawani A, Teo JD, Weiss AS, Yeo GC. The Matrix Protein Tropoelastin Prolongs Mesenchymal Stromal Cell Vitality and Delays Senescence During Replicative Aging. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402168. [PMID: 39120048 PMCID: PMC11497112 DOI: 10.1002/advs.202402168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/26/2024] [Indexed: 08/10/2024]
Abstract
Cellular senescence leads to the functional decline of regenerative cells such as mesenchymal stromal/stem cells (MSCs), which gives rise to chronic conditions and contributes to poor cell therapy outcomes. Aging tissues are associated with extracellular matrix (ECM) dysregulation, including loss of elastin. However, the role of the ECM in modulating senescence is underexplored. In this work, it is shown that tropoelastin, the soluble elastin precursor, is not only a marker of young MSCs but also actively preserves cell fitness and delays senescence during replicative aging. MSCs briefly exposed to tropoelastin exhibit upregulation of proliferative genes and concurrent downregulation of senescence genes. The seno-protective benefits of tropoelastin persist during continuous, long-term MSC culture, and significantly extend the MSC replicative lifespan. Tropoelastin-expanded MSCs further maintain youth-associated phenotype and function compared to age-matched controls, including preserved clonogenic potential, minimal senescence-associated beta-galactosidase activity, maintained cell sizes, reduced expression of senescence markers, suppressed secretion of senescence-associated factors, and increased production of youth-associated proteins. This work points to the utility of exogenously-supplemented tropoelastin for manufacturing MSCs that robustly maintain regenerative potential with age. It further reveals the active role of classical structural ECM proteins in driving cellular age-associated fitness, potentially leading to future interventions for aging-related pathologies.
Collapse
Affiliation(s)
- Sunny Shinchen Lee
- School of Life & Environmental Sciences and Charles Perkins CentreThe University of SydneyCamperdownNSW2006Australia
| | - Aleen Al Halawani
- School of Life & Environmental Sciences and Charles Perkins CentreThe University of SydneyCamperdownNSW2006Australia
| | - Jonathan D. Teo
- School of Medical Sciences and Charles Perkins CentreThe University of SydneyCamperdownNSW2006Australia
| | - Anthony S. Weiss
- School of Life & Environmental Sciences and Charles Perkins CentreThe University of SydneyCamperdownNSW2006Australia
- Sydney Nano InstituteThe University of SydneyCamperdownNSW2006Australia
| | - Giselle C. Yeo
- School of Life & Environmental Sciences and Charles Perkins CentreThe University of SydneyCamperdownNSW2006Australia
- Sydney Nano InstituteThe University of SydneyCamperdownNSW2006Australia
| |
Collapse
|
9
|
Lawton A, Tripodi N, Feehan J. Running on empty: Exploring stem cell exhaustion in geriatric musculoskeletal disease. Maturitas 2024; 188:108066. [PMID: 39089047 DOI: 10.1016/j.maturitas.2024.108066] [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: 02/29/2024] [Revised: 06/26/2024] [Accepted: 07/10/2024] [Indexed: 08/03/2024]
Abstract
Ageing populations globally are associated with increased musculoskeletal disease, including osteoporosis and sarcopenia. These conditions place a significant burden of disease on the individual, society and the economy. To address this, we need to understand the underpinning biological changes, including stem cell exhaustion, which plays a key role in the ageing of the musculoskeletal system. This review of the recent evidence provides an overview of the associated biological processes. The review utilised the PubMed/Medline, Science Direct, and Google Scholar databases. Mechanisms of ageing identified involve a reaction to the chronic inflammation and oxidative stress associated with ageing, resulting in progenitor cell senescence and adipogenic differentiation, leading to decreased mass and quality of both bone and muscle tissue. Although the mechanisms underpinning stem cell exhaustion are unclear, it remains a promising avenue through which to identify new strategies for prevention, detection and management.
Collapse
Affiliation(s)
- Amy Lawton
- Institute for Health and Sport, Victoria University, Melbourne, Australia; College of Sport, Health and Engineering, Victoria University, Melbourne, Australia
| | - Nicholas Tripodi
- Institute for Health and Sport, Victoria University, Melbourne, Australia; First Year College, Victoria University, Melbourne, Australia
| | - Jack Feehan
- Institute for Health and Sport, Victoria University, Melbourne, Australia; School of Health and Biomedical Sciences, STEM College, RMIT, Melbourne, Australia.
| |
Collapse
|
10
|
El Assaad N, Chebly A, Salame R, Achkar R, Bou Atme N, Akouch K, Rafoul P, Hanna C, Abou Zeid S, Ghosn M, Khalil C. Anti-aging based on stem cell therapy: A scoping review. World J Exp Med 2024; 14:97233. [PMID: 39312703 PMCID: PMC11372738 DOI: 10.5493/wjem.v14.i3.97233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/04/2024] [Accepted: 07/18/2024] [Indexed: 08/29/2024] Open
Abstract
Stem cells are present in the tissues and organs and remain in a quiescent and undifferentiated state until it is physiologically necessary to produce new descendant cells. Due to their multipotency property, mesenchymal stem cells have attracted considerable attention worldwide due to their immunomodulation and therapeutic function in tissue regeneration. Stem cells secrete components such as paracrine factors, extracellular vesicles, and exosomes which have been shown to have anti-inflammatory, anti-aging, reconstruction and wound healing potentials in many in vitro and in vivo models. The pluripotency and immunomodulatory features of stem cells could potentially be an effective tool in cell therapy and tissue repair. Aging affects the capacity for self-renewal and differentiation of stem cells, decreasing the potential for regeneration and the loss of optimal functions in organisms over time. Current progress in the field of cellular therapy and regenerative medicine has facilitated the evolution of particular guidelines and quality control approaches, which eventually lead to clinical trials. Cell therapy could potentially be one of the most promising therapies to control aging due to the fact that single stem cell transplantation can regenerate or substitute the injured tissue. To understand the involvement of stem cells not only in tissue maintenance and disease but also in the control of aging it is important to know and identify their properties, functions, and regulation in vivo, which are addressed in this review.
Collapse
Affiliation(s)
- Nassar El Assaad
- Faculty of Medicine, Saint Joseph University, Beirut 961, Lebanon
| | - Alain Chebly
- Centre Jacques Loiselet for Medical Genetics and Genomics (CGGM), Faculty of Medicine, Saint Joseph University, Beirut 961, Lebanon
- Higher Institute of Public Health, Saint Joseph University, Beirut 961, Lebanon
| | - Rawad Salame
- Stem Cell Therapy Lab, Reviva Regenerative Medicine Center, Beirut 961, Lebanon
| | - Robert Achkar
- Poz Pozan University of Medical Sciences, Pozan 034, Poland
| | - Nour Bou Atme
- Faculty of Medicine, Saint Joseph University, Beirut 961, Lebanon
| | - Khalil Akouch
- Stem Cell Therapy Lab, Reviva Regenerative Medicine Center, Beirut 961, Lebanon
| | - Paul Rafoul
- Department of Epidemiology and Biostatistics, Faculty of Public Health, Lebanese University, Beirut 961, Lebanon
| | - Colette Hanna
- School of Medicine, Lebanese American University, Beirut 961, Lebanon
| | - Samer Abou Zeid
- Faculty of Medicine, Saint Joseph University, Beirut 961, Lebanon
| | - Marwan Ghosn
- Faculty of Medicine, Saint Joseph University, Beirut 961, Lebanon
| | - Charbel Khalil
- Stem Cell Therapy Lab, Reviva Regenerative Medicine Center, Beirut 961, Lebanon
- School of Medicine, Lebanese American University, Beirut 961, Lebanon
- Bone Marrow Transplant Unit, Burjeel Medical City, Abu Dhabi 999041, United Arab Emirates
| |
Collapse
|
11
|
Wang J, Zhang M, Wang H. Emerging Landscape of Mesenchymal Stem Cell Senescence Mechanisms and Implications on Therapeutic Strategies. ACS Pharmacol Transl Sci 2024; 7:2306-2325. [PMID: 39144566 PMCID: PMC11320744 DOI: 10.1021/acsptsci.4c00284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/05/2024] [Accepted: 07/09/2024] [Indexed: 08/16/2024]
Abstract
Mesenchymal stem cells (MSCs) hold significant promise for regenerative medicine and tissue engineering due to their unique multipotent differentiation ability and immunomodulatory properties. MSC therapy is widely discussed and utilized in clinical treatment. However, during both in vitro expansion and in vivo transplantation, MSCs are prone to senescence, an irreversible growth arrest characterized by morphological, gene expression, and functional changes in genomic regulation. The microenvironment surrounding MSCs plays a crucial role in modulating their senescence phenotype, influenced by factors such as hypoxia, inflammation, and aging status. Numerous strategies targeting MSC senescence have been developed, including senolytics and senomorphic agents, antioxidant and exosome therapies, mitochondrial transfer, and niche modulation. Novel approaches addressing replicative senescence have also emerged. This paper comprehensively reviews the current molecular manifestations of MSC senescence, addresses the environmental impact on senescence, and highlights potential therapeutic strategies to mitigate senescence in MSC-based therapies. These insights aim to enhance the efficacy and understanding of MSC therapies.
Collapse
Affiliation(s)
- Jing Wang
- Department
of Cellular and Molecular Medicine, University
of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Muqing Zhang
- Institute
of Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, 21215, United States
| | - Hu Wang
- Institute
of Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, 21215, United States
| |
Collapse
|
12
|
Hamidi SH, Etebar N, Rahimzadegan M, Zali A, Roodsari SR, Niknazar S. Mesenchymal stem cells and their derived exosomes in multiple sclerosis disease: from paper to practice. Mol Cell Biochem 2024; 479:1643-1671. [PMID: 38977625 DOI: 10.1007/s11010-024-05051-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/10/2024] [Indexed: 07/10/2024]
Abstract
Multiple sclerosis (MS) is a chronic neurodegenerative, inflammatory, and demyelinating disease of the central nervous system (CNS). Current medicines are not sufficient to control the inflammation and progressive damage to the CNS that is known in MS. These drawbacks highlight the need for novel treatment options. Cell therapy can now be used to treat complex diseases when conventional therapies are ineffective. Mesenchymal stem cells (MSCs) are a diverse group of multipotential non-hematopoietic stromal cells which have immunomodulatory, neurogenesis, and remyelinating capacity. Their advantageous effects mainly rely on paracrine, cell-cell communication and differentiation properties which introduced them as excellent candidates for MS therapy. Exosomes, as one of the MSCs secretomes, have unique properties that make them highly promising candidates for innovative approach in regenerative medicine. This review discusses the therapeutic potential of MSCs and their derived exosomes as a novel treatment for MS, highlighting the differences between these two approaches.
Collapse
Affiliation(s)
- Seyed Hootan Hamidi
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Acharya BM Reddy College of Pharmacy, Rajiv Gandhi University of Health Sciences Bachelor of Pharmacy, Bangalore, India
| | - Negar Etebar
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, Cyprus
| | - Milad Rahimzadegan
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Zali
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Rahmati Roodsari
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Somayeh Niknazar
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Ei ZZ, Srithawirat T, Chunhacha P, Chaotham C, Arunmanee W, Phookphan P, Chanvorachote P. Resveratrol Shows Potent Senescence Reversal in Experimental Cellular Models of Particular Matter 2.5-induced Cellular Senescence in Human Dermal Papilla Cells. In Vivo 2024; 38:665-673. [PMID: 38418101 PMCID: PMC10905444 DOI: 10.21873/invivo.13487] [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: 11/10/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 03/01/2024]
Abstract
BACKGROUND/AIM Particular matter 2.5 (PM2.5) pollution is associated with senescence induction. Since the impact of PM2.5 on stem cell senescence and potential compounds capable of reversing this process are largely unknown, this study aimed to examine the senescence effects of PM2.5 on dermal papilla (DP) stem cells. Additionally, we explored the reversal of these effects using natural product-derived substances, such as resveratrol (Res) or Emblica fruits, soybean, and Thunbergia Laurifolia (EST) extract. MATERIALS AND METHODS Cell senescence was determined using the β-Galactosidase (SA-β-gal) assay. The senescence-associated secretory phenotype (SASP) was detected using real-time RT-PCR. For senescence markers, the mRNA and protein levels of p21 and p16 were measured using real-time RT-PCR and immunofluorescence analysis. RESULTS Subtoxic concentration of PM2.5 (50 μg/ml) induced senescence in DP cells. Resveratrol (50, 100 μM) and plant extracts (400, 800 μg/ml) reversed PM2.5-induced cell senescence. Treatment with Res or EST significantly decreased SA-β-gal staining in PM2.5-treated cells. Furthermore, Res and EST decreased the mRNA levels of SASP, including IL1α, IL7, IL8, and CXCL1. DP cells exposed to PM2.5 exhibited an increase in p21 and p16 mRNA and protein levels, which could be reversed by the addition of Res or EST. Res and EST could reduce p21 and p16 in senescent cells approximately 3- and 2-fold, respectively, compared to untreated senescent cells. CONCLUSION PM2.5 induced senescence in human DP stem cells. Res and EST extract potentially reverse the senescence phenotypes of such cells.
Collapse
Affiliation(s)
- Zin Zin Ei
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Thunwadee Srithawirat
- Department of Environmental Science, Pibulsongkram Rajabhat University, Phitsanulok, Thailand
| | - Preedakorn Chunhacha
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Chatchai Chaotham
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Wanatchaporn Arunmanee
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Preeyaphan Phookphan
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Pithi Chanvorachote
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand;
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
14
|
Kim SJ, Kwon S, Chung S, Lee EJ, Park SE, Choi SJ, Oh SY, Ryu GH, Jeon HB, Chang JW. Nervonic Acid Inhibits Replicative Senescence of Human Wharton's Jelly-Derived Mesenchymal Stem Cells. Int J Stem Cells 2024; 17:80-90. [PMID: 37822280 PMCID: PMC10899888 DOI: 10.15283/ijsc23101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/01/2023] [Accepted: 08/01/2023] [Indexed: 10/13/2023] Open
Abstract
Cellular senescence causes cell cycle arrest and promotes permanent cessation of proliferation. Since the senescence of mesenchymal stem cells (MSCs) reduces proliferation and multipotency and increases immunogenicity, aged MSCs are not suitable for cell therapy. Therefore, it is important to inhibit cellular senescence in MSCs. It has recently been reported that metabolites can control aging diseases. Therefore, we aimed to identify novel metabolites that regulate the replicative senescence in MSCs. Using a fecal metabolites library, we identified nervonic acid (NA) as a candidate metabolite for replicative senescence regulation. In replicative senescent MSCs, NA reduced senescence-associated β-galactosidase positive cells, the expression of senescence-related genes, as well as increased stemness and adipogenesis. Moreover, in non-senescent MSCs, NA treatment delayed senescence caused by sequential subculture and promoted proliferation. We confirmed, for the first time, that NA delayed and inhibited cellular senescence. Considering optimal concentration, duration, and timing of drug treatment, NA is a novel potential metabolite that can be used in the development of technologies that regulate cellular senescence.
Collapse
Affiliation(s)
- Sun Jeong Kim
- Cell and Gene Therapy Institute, ENCell Co. Ltd., Seoul, Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
| | - Soojin Kwon
- Cell and Gene Therapy Institute, ENCell Co. Ltd., Seoul, Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
| | - Soobeen Chung
- Cell and Gene Therapy Institute, ENCell Co. Ltd., Seoul, Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
| | - Eun Joo Lee
- Cell and Gene Therapy Institute, ENCell Co. Ltd., Seoul, Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
| | - Sang Eon Park
- Cell and Gene Therapy Institute, ENCell Co. Ltd., Seoul, Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
| | - Suk-Joo Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Seoul, Korea
| | - Soo-Young Oh
- Department of Obstetrics and Gynecology, Samsung Medical Center, Seoul, Korea
| | - Gyu Ha Ryu
- Department of Medical Device Management and Research, SAIHST, Sungkyunkwan University, Seoul, Korea
- The Office of R&D Strategy & Planning, Samsung Medical Center, Seoul, Korea
| | - Hong Bae Jeon
- Cell and Gene Therapy Institute, ENCell Co. Ltd., Seoul, Korea
| | - Jong Wook Chang
- Cell and Gene Therapy Institute, ENCell Co. Ltd., Seoul, Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
15
|
Rasouli M, Naeimzadeh Y, Hashemi N, Hosseinzadeh S. Age-Related Alterations in Mesenchymal Stem Cell Function: Understanding Mechanisms and Seeking Opportunities to Bypass the Cellular Aging. Curr Stem Cell Res Ther 2024; 19:15-32. [PMID: 36642876 DOI: 10.2174/1574888x18666230113144016] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/28/2022] [Accepted: 11/23/2022] [Indexed: 01/17/2023]
Abstract
Undoubtedly, mesenchymal stem cells (MSCs) are the most common cell therapy candidates in clinical research and therapy. They not only exert considerable therapeutic effects to alleviate inflammation and promote regeneration, but also show low-immunogenicity properties, which ensure their safety following allogeneic transplantation. Thanks to the necessity of providing a sufficient number of MSCs to achieve clinically efficient outcomes, prolonged in vitro cultivation is indisputable. However, either following long-term in vitro expansion or aging in elderly individuals, MSCs face cellular senescence. Senescent MSCs undergo an impairment in their function and therapeutic capacities and secrete degenerative factors which negatively affect young MSCs. To this end, designing novel investigations to further elucidate cellular senescence and to pave the way toward finding new strategies to reverse senescence is highly demanded. In this review, we will concisely discuss current progress on the detailed mechanisms of MSC senescence and various inflicted changes following aging in MSC. We will also shed light on the examined strategies underlying monitoring and reversing senescence in MSCs to bypass the comprised therapeutic efficacy of the senescent MSCs.
Collapse
Affiliation(s)
- Mehdi Rasouli
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yasaman Naeimzadeh
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nader Hashemi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Simzar Hosseinzadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Wan Z, Chen YF, Pan Q, Wang Y, Yuan S, Chin HY, Wu HH, Lin WT, Cheng PY, Yang YJ, Wang YF, Kumta SM, Lee CW, Lee OKS. Single-cell transcriptome analysis reveals the effectiveness of cytokine priming irrespective of heterogeneity in mesenchymal stromal cells. Cytotherapy 2023; 25:1155-1166. [PMID: 37715776 DOI: 10.1016/j.jcyt.2023.08.006] [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: 04/12/2023] [Revised: 08/10/2023] [Accepted: 08/19/2023] [Indexed: 09/18/2023]
Abstract
BACKGROUND AIMS Mesenchymal stromal cells (MSCs) are recognized as a potential cell-based therapy for regenerative medicine. Short-term inflammatory cytokine pre-stimulation (cytokine priming) is a promising approach to enhance regenerative efficacy of MSCs. However, it is unclear whether their intrinsic heterogenic nature causes an unequal response to cytokine priming, which might blunt the accessibility of clinical applications. METHODS In this study, by analyzing the single-cell transcriptomic landscape of human bone marrow MSCs from a naïve to cytokine-primed state, we elucidated the potential mechanism of superior therapeutic potential in cytokine-primed MSCs. RESULTS We found that cytokine-primed MSCs had a distinct transcriptome landscape. Although substantial heterogeneity was identified within the population in both naïve and primed states, cytokine priming enhanced the several characteristics of MSCs associated with therapeutic efficacy irrespective of heterogeneity. After cytokine-priming, all sub-clusters of MSCs possessed high levels of immunoregulatory molecules, trophic factors, stemness-related genes, anti-apoptosis markers and low levels of multi-lineage and senescence signatures, which are critical for their therapeutic potency. CONCLUSIONS In conclusion, our results provide new insights into MSC heterogeneity under cytokine stimulation and suggest that cytokine priming reprogrammed MSCs independent of heterogeneity.
Collapse
Affiliation(s)
- Zihao Wan
- Department of Orthopaedics and Limb Reconstruction/Paediatric Orthopaedics, South China Hospital of Shenzhen University, Shenzhen, China; Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China; Hospital Authority, Hong Kong SAR, China
| | - Yu-Fan Chen
- Center for Translational Genomics & Regenerative Medicine Research, China Medical University Hospital, Taichung, Taiwan; Department of Biomedical Engineering, China Medical University, Taichung, Taiwan
| | - Qi Pan
- Department of Orthopaedics and Limb Reconstruction/Paediatric Orthopaedics, South China Hospital of Shenzhen University, Shenzhen, China
| | - Yiwei Wang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shuai Yuan
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Hui Yen Chin
- Hong Kong Hub of Paediatric Excellence, Hong Kong Children's Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hao-Hsiang Wu
- Center for Translational Genomics & Regenerative Medicine Research, China Medical University Hospital, Taichung, Taiwan
| | - Wei-Ting Lin
- Doctoral Degree Program of Translational Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Po-Yu Cheng
- Center for Translational Genomics & Regenerative Medicine Research, China Medical University Hospital, Taichung, Taiwan
| | - Yun-Jung Yang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Fan Wang
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shekhar Madhukar Kumta
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chien-Wei Lee
- Center for Translational Genomics & Regenerative Medicine Research, China Medical University Hospital, Taichung, Taiwan; Department of Biomedical Engineering, China Medical University, Taichung, Taiwan.
| | - Oscar Kuang-Sheng Lee
- Center for Translational Genomics & Regenerative Medicine Research, China Medical University Hospital, Taichung, Taiwan; Doctoral Degree Program of Translational Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Orthopedics, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
17
|
Lee SJ, Shin JW, Kwon MA, Lee KB, Kim HJ, Lee JH, Kang HS, Jun JK, Cho SY, Kim IG. Transglutaminase 2 Prevents Premature Senescence and Promotes Osteoblastic Differentiation of Mesenchymal Stem Cells through NRF2 Activation. Stem Cells Int 2023; 2023:8815888. [PMID: 37900967 PMCID: PMC10611545 DOI: 10.1155/2023/8815888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/31/2023] [Accepted: 09/12/2023] [Indexed: 10/31/2023] Open
Abstract
Transglutaminase 2 (TG2) is a multifunctional enzyme that exhibits transamidase, GTPase, kinase, and protein disulfide isomerase (PDI) activities. Of these, transamidase-mediated modification of proteins regulates apoptosis, differentiation, inflammation, and fibrosis. TG2 is highly expressed in mesenchymal stem cells (MSCs) compared with differentiated cells, suggesting a role of TG2 specific for MSC characteristics. In this study, we report a new function of TG2 in the regulation of MSC redox homeostasis. During in vitro MSC expansion, TG2 is required for cell proliferation and self-renewal by preventing premature senescence but has no effect on the expression of surface antigens and oxidative stress-induced cell death. Moreover, induction of differentiation upregulates TG2 that promotes osteoblastic differentiation. Molecular analyses revealed that TG2 mediates tert-butylhydroquinone, but not sulforaphane, -induced nuclear factor erythroid 2-related factor 2 (NRF2) activation in a transamidase activity-independent manner. Differences in the mechanism of action between two NRF2 activators suggest that PDI activity of TG2 may be implicated in the stabilization of NRF2. The role of TG2 in the regulation of antioxidant response was further supported by transcriptomic analysis of MSC. These results indicate that TG2 is a critical enzyme in eliciting antioxidant response in MSC through NRF2 activation, providing a target for optimizing MSC manufacturing processes to prevent premature senescence.
Collapse
Affiliation(s)
- Soo-Jin Lee
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ji-Woong Shin
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Mee-Ae Kwon
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ki Baek Lee
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Laboratory for Cellular Response to Oxidative Stress, Cell2in, Inc., Seoul, Republic of Korea
| | - Hyo-Jun Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jin-Haeng Lee
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Heun-Soo Kang
- Laboratory for Cellular Response to Oxidative Stress, Cell2in, Inc., Seoul, Republic of Korea
| | - Jong Kwan Jun
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sung-Yup Cho
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Medical Research Center, Genomic Medicine Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - In-Gyu Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute of Human-Environment Interface Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
18
|
Ei ZZ, Mutirangura A, Arunmanee W, Chanvorachote P. The Role of Box A of HMGB1 in Enhancing Stem Cell Properties of Human Mesenchymal Cells: A Novel Approach for the Pursuit of Anti-aging Therapy. In Vivo 2023; 37:2006-2017. [PMID: 37652483 PMCID: PMC10500522 DOI: 10.21873/invivo.13298] [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: 04/26/2023] [Revised: 06/25/2023] [Accepted: 06/26/2023] [Indexed: 09/02/2023]
Abstract
BACKGROUND/AIM Box A is a highly conserved DNA-binding domain of high-mobility group box 1 (HMGB1) and has been shown to reverse senescence and aging features in many cell models. We investigated whether the activation of box A can influence stem cell properties. MATERIALS AND METHODS Human dermal papilla (DP) cells and primary human white pre-adipocytes (HWPc) were employed as mesenchymal cell models. Box A-overexpressing plasmids were used to induce cellular box A expression. mRNA and protein levels of stemness markers POU class 5 homeobox 1 pseudogene 5 (OCT4, HGNC: 9221), Nanog homeobox (NANOG, HGNC: 20857), and SRY-box transcription factor 2 (SOX2, HGNC:11195) in DP cells and HWPc were measured by real-time polymerase chain reaction and immunofluorescence analysis, respectively. RESULTS Transfection efficiency of box A-overexpressing plasmid was 80% and 50% in DP cells and HWPc, respectively. The proliferative rate of both cell types significantly increased 72 h after transfection. Levels of OCT4, NANOG and SOX2 mRNA and protein expression were significantly increased in box A-transfected DP cells and HWPc compared to empty plasmid-transfected cells. Immunofluorescence analysis confirmed the induction of OCT4, NANOG and SOX2 protein expression in response to box A in DP cells and HWPc. OCT4 and SOX2 were expressed in both the nuclear and cytoplasmic compartments, while NANOG was intensely located in the nucleus of box A-transfected cells. CONCLUSION Our findings suggest that box A may potentially enhance stemness, which may have significant benefits in improving stem cell function due to aging processes and disease. This research may have implications for regenerative medicine applications.
Collapse
Affiliation(s)
- Zin Zin Ei
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Apiwat Mutirangura
- Center of Excellence in Molecular Genetics of Cancer and Human Disease, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Wanatchaporn Arunmanee
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Pithi Chanvorachote
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand;
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
19
|
Mokhtari T, Shayan M, Rezaei Rashnudi A, Hassanzadeh G, Mehran Nia K. Wharton's jelly mesenchymal stem cells attenuate global hypoxia-induced learning and memory impairment via preventing blood-brain barrier breakdown. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:1053-1060. [PMID: 37605722 PMCID: PMC10440140 DOI: 10.22038/ijbms.2023.70137.15250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 04/30/2023] [Indexed: 08/23/2023]
Abstract
Objectives Intracerebroventricular (ICV) injections of mesenchymal stem cells (MSCs) may improve the function and structure of blood-brain barrier (BBB), possibly by preserving the BBB integrity. This study examined the impact of Wharton's jelly (WJ)-MSCs on cognitive dysfunction and BBB disruption following a protracted hypoxic state. Materials and Methods Twenty-four male Wistar rats were randomly studied in four groups: Control (Co): Healthy animals, Sham (Sh): Rats were placed in the cage without hypoxia induction and with ICV injection of vehicle, Hypoxic (Hx)+vehicle: Hypoxic rats with ICV injection of vehicle (5 μl of PBS), and Hx+MSCs: Hypoxic rats with ICV injection of MSCs. Spatial learning and memory were evaluated one week after WJ-MSCs injection, and then animals were sacrificed for molecular research. Results Hypoxia increased latency and lowered the time and distance required reaching the target quarter, according to the findings. Furthermore, hypoxic rats had lower gene expression and protein levels of hippocampus vascular endothelial (VE)-cadherin, claudin 5, and tricellulin gene expression than Co and Sh animals (P<0.05). Finally, administering WJ-MSCs after long-term hypoxia effectively reversed the cognitive deficits and prevented the BBB breakdown via the upregulation of VE-cadherin, claudin 5, and tricellulin genes (P<0.05). Conclusion These findings suggest that prolonged hypoxia induces spatial learning and memory dysfunction and increases BBB disruption, the potential mechanism of which might be via reducing VE-cadherin, claudin 5, and tricellulin genes. Hence, appropriate treatment with WJ-MSCs could reverse ischemia adverse effects and protect the BBB integrity following prolonged hypoxia.
Collapse
Affiliation(s)
- Tahmineh Mokhtari
- Hubei Key Laboratory of Embryonic Stem Cell Research, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, People’s Republic of China
- Department of Histology and Embryology, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, People’s Republic of China
| | - Maryam Shayan
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Gholamreza Hassanzadeh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kobra Mehran Nia
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
20
|
Fraile M, Eiro N, Costa LA, Martín A, Vizoso FJ. Aging and Mesenchymal Stem Cells: Basic Concepts, Challenges and Strategies. BIOLOGY 2022; 11:1678. [PMID: 36421393 PMCID: PMC9687158 DOI: 10.3390/biology11111678] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/08/2022] [Accepted: 11/15/2022] [Indexed: 08/27/2023]
Abstract
Aging and frailty are complex processes implicating multifactorial mechanisms, such as replicative senescence, oxidative stress, mitochondrial dysfunction, or autophagy disorder. All of these mechanisms drive dramatic changes in the tissue environment, such as senescence-associated secretory phenotype factors and inflamm-aging. Thus, there is a demand for new therapeutic strategies against the devastating effects of the aging and associated diseases. Mesenchymal stem cells (MSC) participate in a "galaxy" of tissue signals (proliferative, anti-inflammatory, and antioxidative stress, and proangiogenic, antitumor, antifibrotic, and antimicrobial effects) contributing to tissue homeostasis. However, MSC are also not immune to aging. Three strategies based on MSC have been proposed: remove, rejuvenate, or replace the senescent MSC. These strategies include the use of senolytic drugs, antioxidant agents and genetic engineering, or transplantation of younger MSC. Nevertheless, these strategies may have the drawback of the adverse effects of prolonged use of the different drugs used or, where appropriate, those of cell therapy. In this review, we propose the new strategy of "Exogenous Restitution of Intercellular Signalling of Stem Cells" (ERISSC). This concept is based on the potential use of secretome from MSC, which are composed of molecules such as growth factors, cytokines, and extracellular vesicles and have the same biological effects as their parent cells. To face this cell-free regenerative therapy challenge, we have to clarify key strategy aspects, such as establishing tools that allow us a more precise diagnosis of aging frailty in order to identify the therapeutic requirements adapted to each case, identify the ideal type of MSC in the context of the functional heterogeneity of these cellular populations, to optimize the mass production and standardization of the primary materials (cells) and their secretome-derived products, to establish the appropriate methods to validate the anti-aging effects and to determine the most appropriate route of administration for each case.
Collapse
Affiliation(s)
- Maria Fraile
- Research Unit, Fundación Hospital de Jove, Avda. Eduardo Castro, 161, 33920 Gijon, Spain
| | - Noemi Eiro
- Research Unit, Fundación Hospital de Jove, Avda. Eduardo Castro, 161, 33920 Gijon, Spain
| | - Luis A. Costa
- Research Unit, Fundación Hospital de Jove, Avda. Eduardo Castro, 161, 33920 Gijon, Spain
| | - Arancha Martín
- Research Unit, Fundación Hospital de Jove, Avda. Eduardo Castro, 161, 33920 Gijon, Spain
- Department of Emergency, Hospital Universitario de Cabueñes, Los Prados, 395, 33394 Gijon, Spain
| | - Francisco J. Vizoso
- Research Unit, Fundación Hospital de Jove, Avda. Eduardo Castro, 161, 33920 Gijon, Spain
- Department of Surgery, Fundación Hospital de Jove, Avda. Eduardo Castro, 161, 33920 Gijon, Spain
| |
Collapse
|
21
|
Alves-Paiva RM, do Nascimento S, De Oliveira D, Coa L, Alvarez K, Hamerschlak N, Okamoto OK, Marti LC, Kondo AT, Kutner JM, Bortolini MAT, Castro R, de Godoy JAP. Senescence State in Mesenchymal Stem Cells at Low Passages: Implications in Clinical Use. Front Cell Dev Biol 2022; 10:858996. [PMID: 35445029 PMCID: PMC9015663 DOI: 10.3389/fcell.2022.858996] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 02/28/2022] [Indexed: 11/28/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells found in various tissues and are easily cultivated. For use in clinical protocols, MSCs must be expanded to obtain an adequate number of cells, but a senescence state may be instituted after some passages, reducing their replicative potential. In this study, we report a case where MSC derived from an elderly donor acquired a senescence state after three passages. The bone marrow was aspirated from a female patient submitted to a cell therapy for the incontinency urinary protocol; MSCs were cultivated with DMEM low glucose, supplemented with 10% autologous serum (AS) plus 1% L-glutamine and 1% antibiotic/antimycotic. Senescence analysis was performed by β-galactosidase staining after 24 and 48 h. Controls were established using BM-MSC from healthy donors and used for senescence and gene expression assays. Gene expression was performed using RT-PCR for pluripotency genes, such as SOX2, POU5F1, NANOG, and KLF4. MSC telomere length was measured by the Southern blotting technique, and MSCs were also analyzed for their capacity to differentiate into adipocytes, chondrocytes, and osteocytes. The patient’s MSC expansion using AS displayed an early senescence state. In order to understand the role of AS in senescence, MSCs were then submitted to two different culture conditions: 1) with AS or 2) with FBS supplementation. Senescence state was assessed after 24 h, and no statistical differences were observed between the two conditions. However, patients’ cells cultured with AS displayed a higher number of senescence cells than FBS medium after 48 h (p = 0.0018). Gene expression was performed in both conditions; increased expression of KLF4 was observed in the patient’s cells in comparison to healthy controls (p = 0.0016); reduced gene expression was observed for NANOG (p = 0.0016) and SOX2 (p = 0.0014) genes. Telomere length of the patient’s cells was shorter than that of a healthy donor and that of a patient of similar age. Osteocyte differentiation seemed to be more diffuse than that of the healthy donor and that of the patient of similar age. MSCs could enter a senescence state during expansion in early passages and can impact MSC quality for clinical applications, reducing their efficacy when administered.
Collapse
Affiliation(s)
- Raquel M Alves-Paiva
- Department of Hemotherapy and Cellular Therapy, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Sabrina do Nascimento
- Experimental Research Laboratory, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Denise De Oliveira
- Department of Hemotherapy and Cellular Therapy, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Larissa Coa
- Department of Hemotherapy and Cellular Therapy, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Kelen Alvarez
- Department of Hemotherapy and Cellular Therapy, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Nelson Hamerschlak
- Department of Hemotherapy and Cellular Therapy, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Oswaldo Keith Okamoto
- Department of Hemotherapy and Cellular Therapy, Hospital Israelita Albert Einstein, São Paulo, Brazil.,Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Biosciences Institute, University of São Paulo (USP), Sao Paulo, Brazil
| | - Luciana C Marti
- Experimental Research Laboratory, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Andrea T Kondo
- Department of Hemotherapy and Cellular Therapy, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Jose Mauro Kutner
- Department of Hemotherapy and Cellular Therapy, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | | | - Rodrigo Castro
- Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Juliana A Preto de Godoy
- Department of Hemotherapy and Cellular Therapy, Hospital Israelita Albert Einstein, São Paulo, Brazil
| |
Collapse
|
22
|
Liu Y, Schwam J, Chen Q. Senescence-Associated Cell Transition and Interaction (SACTAI): A Proposed Mechanism for Tissue Aging, Repair, and Degeneration. Cells 2022; 11:1089. [PMID: 35406653 PMCID: PMC8997723 DOI: 10.3390/cells11071089] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 02/01/2023] Open
Abstract
Aging is a broad process that occurs as a time-dependent functional decline and tissue degeneration in living organisms. On a smaller scale, aging also exists within organs, tissues, and cells. As the smallest functional unit in living organisms, cells "age" by reaching senescence where proliferation stops. Such cellular senescence is achieved through replicative stress, telomere erosion and stem cell exhaustion. It has been shown that cellular senescence is key to tissue degradation and cell death in aging-related diseases (ARD). However, senescent cells constitute only a small percentage of total cells in the body, and they are resistant to death during aging. This suggests that ARD may involve interaction of senescent cells with non-senescent cells, resulting in senescence-triggered death of non-senescent somatic cells and tissue degeneration in aging organs. Here, based on recent research evidence from our laboratory and others, we propose a mechanism-Senescence-Associated Cell Transition and Interaction (SACTAI)-to explain how cell heterogeneity arises during aging and how the interaction between somatic cells and senescent cells, some of which are derived from aging somatic cells, results in cell death and tissue degeneration.
Collapse
Affiliation(s)
| | | | - Qian Chen
- Laboratory of Molecular Biology and Nanomedicine, Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA; (Y.L.); (J.S.)
| |
Collapse
|
23
|
Bridger JM, Pereira RT, Pina C, Tosi S, Lewis A. Alterations to Genome Organisation in Stem Cells, Their Differentiation and Associated Diseases. Results Probl Cell Differ 2022; 70:71-102. [PMID: 36348105 DOI: 10.1007/978-3-031-06573-6_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The organisation of the genome in its home, the cell nucleus, is reliant on a number of different aspects to establish, maintain and alter its functional non-random positioning. The genome is dispersed throughout a cell nucleus in specific chromosome territories which are further divided into topologically associated domains (TADs), where regions of the genome from different and the same chromosomes come together. This organisation is both controlled by DNA and chromatin epigenetic modification and the association of the genome with nuclear structures such as the nuclear lamina, the nucleolus and nuclear bodies and speckles. Indeed, sequences that are associated with the first two structures mentioned are termed lamina-associated domains (LADs) and nucleolar-associated domains (NADs), respectively. The modifications and nuclear structures that regulate genome function are altered through a cell's life from stem cell to differentiated cell through to reversible quiescence and irreversible senescence, and hence impacting on genome organisation, altering it to silence specific genes and permit others to be expressed in a controlled way in different cell types and cell cycle statuses. The structures and enzymes and thus the organisation of the genome can also be deleteriously affected, leading to disease and/or premature ageing.
Collapse
Affiliation(s)
- Joanna M Bridger
- Division of Biosciences, Department of Life Sciences, Centre for Genome Engineering and Maintenance (cenGEM), College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UK.
| | - Rita Torres Pereira
- Division of Biosciences, Department of Life Sciences, Centre for Genome Engineering and Maintenance (cenGEM), College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UK
| | - Cristina Pina
- Division of Biosciences, Department of Life Sciences, Centre for Genome Engineering and Maintenance (cenGEM), College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UK
| | - Sabrina Tosi
- Division of Biosciences, Department of Life Sciences, Centre for Genome Engineering and Maintenance (cenGEM), College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UK
| | - Annabelle Lewis
- Division of Biosciences, Department of Life Sciences, Centre for Genome Engineering and Maintenance (cenGEM), College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UK
| |
Collapse
|
24
|
Guo X, Wang J, Zou W, Wei W, Guan X, Liu J. Exploring microenvironment strategies to delay mesenchymal stem cell senescence. Stem Cells Dev 2021; 31:38-52. [PMID: 34913751 DOI: 10.1089/scd.2021.0254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have recently emerged as an important candidate for cell therapy and tissue regeneration. However, some limitations in translational research and therapies still exist, such as insufficient cell supply, inadequate differentiation potential, and decreased immune capacity, all of which result from replicative senescence during long-term in vitro culture. In vitro, stem cells lack a protective microenvironment owing to the absence of physical and biochemical cues compared with the in vivo niche, which provides dynamic physicochemical and biological cues. This difference results in accelerated aging after long-term in vitro culture. Therefore, it remains a great challenge to delay replicative senescence in culture. Constructing a microenvironment to delay replicative senescence of MSCs by maintaining their phenotypes, properties, and functions is a feasible strategy to solve this problem and has made measurable progress both in preclinical studies and clinical trials. Here, we review the current knowledge on the characteristics of senescent MSCs, explore the molecular mechanisms of MSCs senescence, describe the niche of MSCs, and discuss some current microenvironment strategies to delay MSCs replicative senescence that can broaden their range of therapeutic applications.
Collapse
Affiliation(s)
- Xunhui Guo
- First Affiliated Hospital of Dalian Medical University, 74710, Stem Cell Clinical Research Center, Dalian, China;
| | - Jiayi Wang
- First Affiliated Hospital of Dalian Medical University, 74710, Stem Cell Clinical Research Center, Dalian, Dalian, China;
| | - Wei Zou
- Liaoning Normal University, 66523, College of Life Sciences, Dalian, China;
| | - Wenjuan Wei
- First Affiliated Hospital of Dalian Medical University, 74710, Dalian, China, 116011;
| | - Xin Guan
- First Affiliated Hospital of Dalian Medical University, 74710, Dalian, China, 116011;
| | - Jing Liu
- First Affiliated Hospital of Dalian Medical University, 74710, Dalian, China, 116011;
| |
Collapse
|
25
|
Deng J, Ouyang P, Li W, Zhong L, Gu C, Shen L, Cao S, Yin L, Ren Z, Zuo Z, Deng J, Yan Q, Yu S. Curcumin Alleviates the Senescence of Canine Bone Marrow Mesenchymal Stem Cells during In Vitro Expansion by Activating the Autophagy Pathway. Int J Mol Sci 2021; 22:ijms222111356. [PMID: 34768788 PMCID: PMC8583405 DOI: 10.3390/ijms222111356] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/16/2021] [Accepted: 10/17/2021] [Indexed: 12/16/2022] Open
Abstract
Senescence in mesenchymal stem cells (MSCs) not only hinders the application of MSCs in regenerative medicine but is also closely correlated with biological aging and the development of degenerative diseases. In this study, we investigated the anti-aging effects of curcumin (Cur) on canine bone marrow-derived MSCs (cBMSCs), and further elucidated the potential mechanism of action based on the modulation of autophagy. cBMSCs were expanded in vitro with standard procedures to construct a cell model of premature senescence. Our evidence indicates that compared with the third passage of cBMSCs, many typical senescence-associated phenotypes were observed in the sixth passage of cBMSCs. Cur treatment can improve cBMSC survival and retard cBMSC senescence according to observations that Cur (1 μM) treatment can improve the colony-forming unit-fibroblasts (CFU-Fs) efficiency and upregulated the mRNA expression of pluripotent transcription factors (SOX-2 and Nanog), as well as inhibiting the senescence-associated beta-galactosidase (SA-β-gal) activities and mRNA expression of the senescence-related markers (p16 and p21) and pro-inflammatory molecules (tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6)). Furthermore, Cur (0.1 μM~10 μM) was observed to increase autophagic activity, as identified by upregulation of microtubule-associated protein 1 light chain 3 (LC3), unc51-like autophagy-activating kinase-1 (ULK1), autophagy-related gene (Atg) 7 and Atg12, and the generation of type II of light chain 3 (LC3-II), thereby increasing autophagic vacuoles and acidic vesicular organelles, as well as causing a significant decrease in the p62 protein level. Moreover, the autophagy activator rapamycin (RAP) and Cur were found to partially ameliorate the senescent features of cBMSCs, while the autophagy inhibitor 3-methyladenine (3-MA) was shown to aggravate cBMSCs senescence and Cur treatment was able to restore the suppressed autophagy and counteract 3-MA-induced cBMSC senescence. Hence, our study highlights the important role of Cur-induced autophagy and its effects for ameliorating cBMSC senescence and provides new insight for delaying senescence and improving the therapeutic potential of MSCs.
Collapse
Affiliation(s)
- Jiaqiang Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (J.D.); (P.O.); (W.L.); (L.Z.); (C.G.); (L.S.); (S.C.); (L.Y.); (Z.R.); (Z.Z.); (J.D.)
- College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Ping Ouyang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (J.D.); (P.O.); (W.L.); (L.Z.); (C.G.); (L.S.); (S.C.); (L.Y.); (Z.R.); (Z.Z.); (J.D.)
| | - Weiyao Li
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (J.D.); (P.O.); (W.L.); (L.Z.); (C.G.); (L.S.); (S.C.); (L.Y.); (Z.R.); (Z.Z.); (J.D.)
| | - Lijun Zhong
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (J.D.); (P.O.); (W.L.); (L.Z.); (C.G.); (L.S.); (S.C.); (L.Y.); (Z.R.); (Z.Z.); (J.D.)
| | - Congwei Gu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (J.D.); (P.O.); (W.L.); (L.Z.); (C.G.); (L.S.); (S.C.); (L.Y.); (Z.R.); (Z.Z.); (J.D.)
- Laboratory Animal Centre, Southwest Medical University, Luzhou 646000, China
| | - Liuhong Shen
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (J.D.); (P.O.); (W.L.); (L.Z.); (C.G.); (L.S.); (S.C.); (L.Y.); (Z.R.); (Z.Z.); (J.D.)
| | - Suizhong Cao
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (J.D.); (P.O.); (W.L.); (L.Z.); (C.G.); (L.S.); (S.C.); (L.Y.); (Z.R.); (Z.Z.); (J.D.)
| | - Lizi Yin
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (J.D.); (P.O.); (W.L.); (L.Z.); (C.G.); (L.S.); (S.C.); (L.Y.); (Z.R.); (Z.Z.); (J.D.)
| | - Zhihua Ren
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (J.D.); (P.O.); (W.L.); (L.Z.); (C.G.); (L.S.); (S.C.); (L.Y.); (Z.R.); (Z.Z.); (J.D.)
| | - Zhicai Zuo
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (J.D.); (P.O.); (W.L.); (L.Z.); (C.G.); (L.S.); (S.C.); (L.Y.); (Z.R.); (Z.Z.); (J.D.)
| | - Junliang Deng
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (J.D.); (P.O.); (W.L.); (L.Z.); (C.G.); (L.S.); (S.C.); (L.Y.); (Z.R.); (Z.Z.); (J.D.)
| | - Qigui Yan
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (J.D.); (P.O.); (W.L.); (L.Z.); (C.G.); (L.S.); (S.C.); (L.Y.); (Z.R.); (Z.Z.); (J.D.)
- Correspondence: (Q.Y.); (S.Y.); Tel.: +86-139-8160-8208 (Q.Y.); +86-189-8057-3629 (S.Y.)
| | - Shumin Yu
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (J.D.); (P.O.); (W.L.); (L.Z.); (C.G.); (L.S.); (S.C.); (L.Y.); (Z.R.); (Z.Z.); (J.D.)
- Correspondence: (Q.Y.); (S.Y.); Tel.: +86-139-8160-8208 (Q.Y.); +86-189-8057-3629 (S.Y.)
| |
Collapse
|
26
|
Cellular Senescence in Lung Fibrosis. Int J Mol Sci 2021; 22:ijms22137012. [PMID: 34209809 PMCID: PMC8267738 DOI: 10.3390/ijms22137012] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/13/2021] [Accepted: 06/24/2021] [Indexed: 12/19/2022] Open
Abstract
Fibrosing interstitial lung diseases (ILDs) are chronic and ultimately fatal age-related lung diseases characterized by the progressive and irreversible accumulation of scar tissue in the lung parenchyma. Over the past years, significant progress has been made in our incomplete understanding of the pathobiology underlying fibrosing ILDs, in particular in relation to diverse age-related processes and cell perturbations that seem to lead to maladaptation to stress and susceptibility to lung fibrosis. Growing evidence suggests that a specific biological phenomenon known as cellular senescence plays an important role in the initiation and progression of pulmonary fibrosis. Cellular senescence is defined as a cell fate decision caused by the accumulation of unrepairable cellular damage and is characterized by an abundant pro-inflammatory and pro-fibrotic secretome. The senescence response has been widely recognized as a beneficial physiological mechanism during development and in tumour suppression. However, recent evidence strengthens the idea that it also drives degenerative processes such as lung fibrosis, most likely by promoting molecular and cellular changes in chronic fibrosing processes. Here, we review how cellular senescence may contribute to lung fibrosis pathobiology, and we highlight current and emerging therapeutic approaches to treat fibrosing ILDs by targeting cellular senescence.
Collapse
|