1
|
Chen W, Wang YJ. Multifaceted roles of OCT4 in tumor microenvironment: biology and therapeutic implications. Oncogene 2025; 44:1213-1229. [PMID: 40229384 DOI: 10.1038/s41388-025-03408-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 03/28/2025] [Accepted: 04/04/2025] [Indexed: 04/16/2025]
Abstract
OCT4 (Octamer-binding transcription factor 4, encoded by the POU5F1 gene) is a master transcription factor for maintaining the self-renewal and pluripotency of pluripotent stem cells, as well as a pioneer factor regulating epigenetics-driven cell reprogramming and cell fate conversion. It is also detected in a variety of cancer tissues and particularly in a small subpopulation of cancer cells known as cancer stem cells (CSCs). Accumulating evidence has revealed that CSCs are a dynamic population, exhibiting shift between multipotency and differentiation states, or quiescence and proliferation states. Such cellular plasticity of CSCs is profoundly influenced by dynamic interplay between CSCs and the tumor microenvironment (TME). Here, we review recent evidence showing that OCT4 expressed in CSCs plays a multifaceted role in shaping the TME by interacting with the cellular TME components, including cancer-associated fibroblasts, tumor endothelial cells, tumor-infiltrating immune cells, as well as the non-cellular TME components, such as extracellular matrix (ECM), metabolites, soluble factors (e.g., growth factors, cytokines and chemokines), and intra-tumoral microbiota. Together, OCT4 regulates crucial processes encompassing ECM remodeling, epithelial-mesenchymal transition, metabolic reprogramming, angiogenesis, and immune responses. The complex and bidirectional interactions between OCT4-expressing CSCs and the TME create a supportive niche for tumor growth, invasion, and resistance to therapy. Better understanding OCT4's roles in such interactions can provide deeper insights into potential therapeutic strategies and targets for disrupting the supportive environment of tumors. The emerging therapies targeting OCT4 in CSCs might hold promise to resensitize therapeutic-resistant cancer cells, and to eradicate all cancer cells when combined with other therapies targeting the bulk of differentiated cancer cells as well as the TME.
Collapse
Affiliation(s)
- Wenjie Chen
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ying-Jie Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
2
|
Ruan Y, Huang X, Sun P, Yu X, Tan X, Song Y, Chen H, Liu Z. ZO-1 boosts the in vitro self-renewal of pre-haematopoietic stem cells from OCT4-reprogrammed human hair follicle mesenchymal stem cells through cytoskeleton remodeling. Stem Cell Res Ther 2024; 15:480. [PMID: 39696518 PMCID: PMC11658245 DOI: 10.1186/s13287-024-04080-w] [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: 02/12/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND The challenge of expanding haematopoietic stem/progenitor cells (HSPCs) in vitro has limited their clinical application. Human hair follicle mesenchymal stem cells (hHFMSCs) can be reprogrammed to generate intermediate stem cells by transducing OCT4 (hHFMSCsOCT4) and pre-inducing with FLT3LG/SCF, and differentiated into erythrocytes. These intermediate cells exhibit gene expression patterns similar to pre-HSCs, making them promising for artificial haematopoiesis. However, further investigation is required to elucidate the in vitro proliferation ability and mechanism underlying the self-renewal of pre-HSCs derived from hHFMSCs. METHODS hHFMSCsOCT4 were pre-treated with FLT3LG and SCF cytokines, followed by characterization and isolation of the floating cell subsets for erythroid differentiation through stimulation with hematopoietic cytokines and nutritional factors. Cell adhesion was assessed through disassociation and adhesion assays. OCT4 expression levels were measured using immunofluorescence staining, RT-qPCR, and Western blotting. RNA sequencing and Gene Ontology (GO) enrichment analysis were then conducted to identify proliferation-related biological processes. Proliferative capacity was evaluated through CCK-8, colony formation assays, Ki67 index, and cell cycle analysis. Cytoskeleton was observed through Wright‒Giemsa, Coomassie brilliant blue, and phalloidin staining. Expression of adherens junction (AJ) core members was confirmed through RT‒qPCR, Western blotting, and immunofluorescence staining before and after ZO-1 knockdown. A regulatory network was constructed to determine relationships among cytoskeleton, proliferation, and the AJ pathway. Student's t tests (GraphPad Prism 8.0.2) were used for group comparisons. The results were considered significant at P < 0.05. RESULTS Pre-treatment of hHFMSCsOCT4 with FLT3LG and SCF leads to the emergence of floating cell subsets exhibiting small, globoid morphology, suspended above adherent cells, forming colonies, and displaying minimal expression of CD45. Excessive OCT4 expression weakens adhesion in floating hHFMSCsOCT4. Floating cells moderately enhanced proliferation and undergo cytoskeleton remodelling, with increased contraction and aggregation of F-actin near the nucleus. The upregulation of ZO-1 could impact the expressions of F-actin, E-cadherin, and β-catenin genes, as well as the nuclear positioning of β-catenin, leading to variations in the cytoskeleton and cell cycle. Finally, a regulatory network revealed that the AJ pathway cored with ZO-1 critically bridges cytoskeletal remodelling and haematopoiesis-related proliferation in a β-catenin-dependent manner. CONCLUSIONS ZO-1 improved the self-renewal of pre-HSCs from OCT4-overexpressing hHFMSCs by remodeling the cytoskeleton via the ZO-1-regulated AJ pathway, suggesting floating hHFMSCsOCT4 as the promising seed cells for artificial hematopoiesis.
Collapse
Affiliation(s)
- Yingchun Ruan
- Department of Pathology, Qingdao Municipal Hospital Group, 1 Jiaozhou Road, Qingdao, 266011, Shandong, China
| | - Xingang Huang
- Department of Pathology, Qingdao Municipal Hospital Group, 1 Jiaozhou Road, Qingdao, 266011, Shandong, China
| | - Pengpeng Sun
- Department of Critical Care Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital), 127 Siliunan Road, Qingdao, 266042, Shandong, China
| | - Xiaozhen Yu
- Department of Pathology, Qingdao Municipal Hospital Group, 1 Jiaozhou Road, Qingdao, 266011, Shandong, China
| | - Xiaohua Tan
- Department of Pathology, College of Basic Medical Sciences, Qingdao University, 308 Ningxia Road, Qingdao, 266071, Shandong, China
| | - Yaolin Song
- Department of Pathology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266000, Shandong, China
| | - Hua Chen
- Department of Pathology, Qingdao Municipal Hospital Group, 1 Jiaozhou Road, Qingdao, 266011, Shandong, China
| | - Zhijing Liu
- Department of Pathology, Qingdao Municipal Hospital Group, 1 Jiaozhou Road, Qingdao, 266011, Shandong, China.
| |
Collapse
|
3
|
Roessner PM, Seufert I, Chapaprieta V, Jayabalan R, Briesch H, Massoni-Badosa R, Boskovic P, Benckendorff J, Roider T, Arseni L, Coelho M, Chakraborty S, Vaca AM, Sivina M, Muckenhuber M, Rodriguez-Rodriguez S, Bonato A, Herbst SA, Zapatka M, Sun C, Kretzmer H, Naake T, Bruch PM, Czernilofsky F, ten Hacken E, Schneider M, Helm D, Yosifov DY, Kauer J, Danilov AV, Bewarder M, Heyne K, Schneider C, Stilgenbauer S, Wiestner A, Mallm JP, Burger JA, Efremov DG, Lichter P, Dietrich S, Martin-Subero JI, Rippe K, Seiffert M. T-bet suppresses proliferation of malignant B cells in chronic lymphocytic leukemia. Blood 2024; 144:510-524. [PMID: 38684038 PMCID: PMC11307267 DOI: 10.1182/blood.2023021990] [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: 07/31/2023] [Revised: 03/28/2024] [Accepted: 04/13/2024] [Indexed: 05/02/2024] Open
Abstract
ABSTRACT The T-box transcription factor T-bet is known as a master regulator of the T-cell response but its role in malignant B cells has not been sufficiently explored. Here, we conducted single-cell resolved multi-omics analyses of malignant B cells from patients with chronic lymphocytic leukemia (CLL) and studied a CLL mouse model with a genetic knockout of Tbx21. We found that T-bet acts as a tumor suppressor in malignant B cells by decreasing their proliferation rate. NF-κB activity, induced by inflammatory signals provided by the microenvironment, triggered T-bet expression, which affected promoter-proximal and distal chromatin coaccessibility and controlled a specific gene signature by mainly suppressing transcription. Gene set enrichment analysis identified a positive regulation of interferon signaling and negative control of proliferation by T-bet. In line, we showed that T-bet represses cell cycling and is associated with longer overall survival of patients with CLL. Our study uncovered a novel tumor suppressive role of T-bet in malignant B cells via its regulation of inflammatory processes and cell cycling, which has implications for the stratification and therapy of patients with CLL. Linking T-bet activity to inflammation explains the good prognostic role of genetic alterations in the inflammatory signaling pathways in CLL.
Collapse
MESH Headings
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- T-Box Domain Proteins/genetics
- T-Box Domain Proteins/metabolism
- Animals
- Humans
- Cell Proliferation
- Mice
- B-Lymphocytes/pathology
- B-Lymphocytes/metabolism
- B-Lymphocytes/immunology
- Mice, Knockout
- Gene Expression Regulation, Leukemic
- NF-kappa B/metabolism
Collapse
Affiliation(s)
- Philipp M. Roessner
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
| | - Isabelle Seufert
- Division of Chromatin Networks, German Cancer Research Center and BioQuant, Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | | | - Ruparoshni Jayabalan
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Hannah Briesch
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Ramon Massoni-Badosa
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
- Single Cell Genomics, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Pavle Boskovic
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | | | - Tobias Roider
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Medicine Partnership Unit, Heidelberg, Germany
| | - Lavinia Arseni
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
| | - Mariana Coelho
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Supriya Chakraborty
- Molecular Hematology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Alicia M. Vaca
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Mariela Sivina
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Markus Muckenhuber
- Division of Chromatin Networks, German Cancer Research Center and BioQuant, Heidelberg, Germany
| | | | - Alice Bonato
- Molecular Hematology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Sophie A. Herbst
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Medicine Partnership Unit, Heidelberg, Germany
| | - Marc Zapatka
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
| | - Clare Sun
- Laboratory of Lymphoid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Helene Kretzmer
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Thomas Naake
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Peter-Martin Bruch
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Medicine Partnership Unit, Heidelberg, Germany
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Felix Czernilofsky
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Medicine Partnership Unit, Heidelberg, Germany
| | | | - Martin Schneider
- Proteomics Core Facility, German Cancer Research Center, Heidelberg, Germany
| | - Dominic Helm
- Proteomics Core Facility, German Cancer Research Center, Heidelberg, Germany
| | - Deyan Y. Yosifov
- Division of Chronic Lymphocytic Leukemia, Department of Internal Medicine III, Ulm University, Ulm, Germany
- Cooperation Unit Mechanisms of Leukemogenesis, German Cancer Research Center, Heidelberg, Germany
| | - Joseph Kauer
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Medicine Partnership Unit, Heidelberg, Germany
| | - Alexey V. Danilov
- Department of Hematology, City of Hope National Medical Center, Duarte, CA
| | - Moritz Bewarder
- José Carreras Center for Immuno- and Gene Therapy and Internal Medicine I, Saarland University Medical School, Homburg/Saar, Germany
| | - Kristina Heyne
- José Carreras Center for Immuno- and Gene Therapy and Internal Medicine I, Saarland University Medical School, Homburg/Saar, Germany
| | - Christof Schneider
- Division of Chronic Lymphocytic Leukemia, Department of Internal Medicine III, Ulm University, Ulm, Germany
| | - Stephan Stilgenbauer
- Division of Chronic Lymphocytic Leukemia, Department of Internal Medicine III, Ulm University, Ulm, Germany
| | - Adrian Wiestner
- Laboratory of Lymphoid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Jan-Philipp Mallm
- Division of Chromatin Networks, German Cancer Research Center and BioQuant, Heidelberg, Germany
| | - Jan A. Burger
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Dimitar G. Efremov
- Molecular Hematology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Peter Lichter
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
| | - Sascha Dietrich
- Department of Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- Molecular Medicine Partnership Unit, Heidelberg, Germany
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - José I. Martin-Subero
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - Karsten Rippe
- Division of Chromatin Networks, German Cancer Research Center and BioQuant, Heidelberg, Germany
| | - Martina Seiffert
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
4
|
Lai Z, Shu Q, Song Y, Tang A, Tian J. Effect of DNA methylation on the osteogenic differentiation of mesenchymal stem cells: concise review. Front Genet 2024; 15:1429844. [PMID: 39015772 PMCID: PMC11250479 DOI: 10.3389/fgene.2024.1429844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 06/10/2024] [Indexed: 07/18/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have promising potential for bone tissue engineering in bone healing and regeneration. They are regarded as such due to their capacity for self-renewal, multiple differentiation, and their ability to modulate the immune response. However, changes in the molecular pathways and transcription factors of MSCs in osteogenesis can lead to bone defects and metabolic bone diseases. DNA methylation is an epigenetic process that plays an important role in the osteogenic differentiation of MSCs by regulating gene expression. An increasing number of studies have demonstrated the significance of DNA methyltransferases (DNMTs), Ten-eleven translocation family proteins (TETs), and MSCs signaling pathways about osteogenic differentiation in MSCs. This review focuses on the progress of research in these areas.
Collapse
Affiliation(s)
- Zhihao Lai
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qing Shu
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yue Song
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- College of Sports Medicine, Wuhan Sports University, Wuhan, China
| | - Ao Tang
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- College of Sports Medicine, Wuhan Sports University, Wuhan, China
| | - Jun Tian
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
5
|
R K, Kumar A, Vinod Kumar K, Sengupta A, Kundal K, Sharma S, Pawar A, Krishna PS, Alfatah M, Ray S, Tiwari B, Kumar R. AagingBase: a comprehensive database of anti-aging peptides. Database (Oxford) 2024; 2024:baae016. [PMID: 38470883 PMCID: PMC10930205 DOI: 10.1093/database/baae016] [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: 11/16/2023] [Revised: 01/07/2024] [Accepted: 03/05/2024] [Indexed: 03/14/2024]
Abstract
The process of aging is an intrinsic and inevitable aspect of life that impacts every living organism. As biotechnological advancements continue to shape our understanding of medicine, peptide therapeutics have emerged as a promising strategy for anti-aging interventions. This is primarily due to their favorable attributes, such as low immunogenicity and cost-effective production. Peptide-based treatments have garnered widespread acceptance and interest in aging research, particularly in the context of age-related therapies. To effectively develop anti-aging treatments, a comprehensive understanding of the physicochemical characteristics of anti-aging peptides is essential. Factors such as amino acid composition, instability index, hydrophobic areas and other relevant properties significantly determine their efficacy as potential therapeutic agents. Consequently, the creation of 'AagingBase', a comprehensive database for anti-aging peptides, aims to facilitate research on aging by leveraging the potential of peptide therapies. AagingBase houses experimentally validated 282 anti-aging peptides collected from 54 research articles and 236 patents. Employing state-of-the-art computational techniques, the acquired sequences have undergone rigorous physicochemical calculations. Furthermore, AagingBase presents users with various informative analyses highlighting atomic compositions, secondary structure fractions, tertiary structure, amino acid compositions and frequencies. The database also offers advanced search and filtering options and similarity search, thereby aiding researchers in understanding their biological functions. Hence, the database enables efficient identification and prioritization of potential peptide candidates in geriatric medicine and holds immense potential for advancing geriatric medicine research and innovations. AagingBase can be accessed without any restriction. Database URL: https://project.iith.ac.in/cgntlab/aagingbase/.
Collapse
Affiliation(s)
- Kunjulakshmi R
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
- Department of Biological Sciences, Indian Institute of Science Education and Research, Berhampur, Odisha 760010, India
| | - Ambuj Kumar
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| | - Keerthana Vinod Kumar
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| | - Avik Sengupta
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| | - Kavita Kundal
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| | - Simran Sharma
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| | - Ankita Pawar
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Clappana P.O., Kollam, Kerala 690525, India
| | - Pithani Sai Krishna
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Clappana P.O., Kollam, Kerala 690525, India
| | - Mohammad Alfatah
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, Matrix #07-01, Singapore 138671, Republic of Singapore
| | - Sandipan Ray
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| | - Bhavana Tiwari
- Department of Biological Sciences, Indian Institute of Science Education and Research, Berhampur, Odisha 760010, India
| | - Rahul Kumar
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| |
Collapse
|
6
|
Jiang N, Tian X, Wang Q, Hao J, Jiang J, Wang H. Regulation Mechanisms and Maintenance Strategies of Stemness in Mesenchymal Stem Cells. Stem Cell Rev Rep 2024; 20:455-483. [PMID: 38010581 DOI: 10.1007/s12015-023-10658-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2023] [Indexed: 11/29/2023]
Abstract
Stemness pertains to the intrinsic ability of mesenchymal stem cells (MSCs) to undergo self-renewal and differentiate into multiple lineages, while simultaneously impeding their differentiation and preserving crucial differentiating genes in a state of quiescence and equilibrium. Owing to their favorable attributes, including uncomplicated isolation protocols, ethical compliance, and ease of procurement, MSCs have become a focal point of inquiry in the domains of regenerative medicine and tissue engineering. As age increases or ex vivo cultivation is prolonged, the functionality of MSCs decreases and their stemness gradually diminishes, thereby limiting their potential therapeutic applications. Despite the existence of several uncertainties surrounding the comprehension of MSC stemness, considerable advancements have been achieved in the clarification of the potential mechanisms that lead to stemness loss, as well as the associated strategies for stemness maintenance. This comprehensive review provides a systematic overview of the factors influencing the preservation of MSC stemness, the molecular mechanisms governing it, the strategies for its maintenance, and the therapeutic potential associated with stemness. Finally, we underscore the obstacles and prospective avenues in present investigations, providing innovative perspectives and opportunities for the preservation and therapeutic utilization of MSC stemness.
Collapse
Affiliation(s)
- Nizhou Jiang
- Central Hospital of Dalian University of Technology Department of Spine Surgery, Dalian, China
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiliang Tian
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Quanxiang Wang
- Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, China
| | - Jiayu Hao
- Central Hospital of Dalian University of Technology Department of Spine Surgery, Dalian, China
| | - Jian Jiang
- Central Hospital of Dalian University of Technology Department of Spine Surgery, Dalian, China.
| | - Hong Wang
- Central Hospital of Dalian University of Technology Department of Spine Surgery, Dalian, China.
| |
Collapse
|
7
|
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
|
8
|
ALZGHOUL YARA, ISSA HALAJBANI, SANAJLEH AHMADK, ALABDUH TAQWA, RABABAH FATIMAH, AL-SHDAIFAT MAHA, ABU-EL-RUB EJLAL, ALMAHASNEH FATIMAH, KHASAWNEH RAMADAR, ALZU’BI AYMAN, MAGABLEH HUTHAIFA. Therapeutic and regenerative potential of different sources of mesenchymal stem cells for cardiovascular diseases. BIOCELL 2024; 48:559-569. [DOI: 10.32604/biocell.2024.048056] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 01/16/2024] [Indexed: 09/03/2024]
|
9
|
Kim YH, Lee YK, Park SS, Park SH, Eom SY, Lee YS, Lee WJ, Jang J, Seo D, Kang HY, Kim JC, Lim SB, Yoon G, Kim HS, Kim JH, Park TJ. Mid-old cells are a potential target for anti-aging interventions in the elderly. Nat Commun 2023; 14:7619. [PMID: 37993434 PMCID: PMC10665435 DOI: 10.1038/s41467-023-43491-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 11/10/2023] [Indexed: 11/24/2023] Open
Abstract
The biological process of aging is thought to result in part from accumulation of senescent cells in organs. However, the present study identified a subset of fibroblasts and smooth muscle cells which are the major constituents of organ stroma neither proliferative nor senescent in tissues of the elderly, which we termed "mid-old status" cells. Upregulation of pro-inflammatory genes (IL1B and SAA1) and downregulation of anti-inflammatory genes (SLIT2 and CXCL12) were detected in mid-old cells. In the stroma, SAA1 promotes development of the inflammatory microenvironment via upregulation of MMP9, which decreases the stability of epithelial cells present on the basement membrane, decreasing epithelial cell function. Remarkably, the microenvironmental change and the functional decline of mid-old cells could be reversed by a young cell-originated protein, SLIT2. Our data identify functional reversion of mid-old cells as a potential method to prevent or ameliorate aspects of aging-related tissue dysfunction.
Collapse
Affiliation(s)
- Young Hwa Kim
- Inflamm-Aging Translational Research Center, Ajou University Medical Center, Suwon, 16499, Korea
| | - Young-Kyoung Lee
- Inflamm-Aging Translational Research Center, Ajou University Medical Center, Suwon, 16499, Korea
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, 16499, Korea
| | - Soon Sang Park
- Inflamm-Aging Translational Research Center, Ajou University Medical Center, Suwon, 16499, Korea
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, 16499, Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, 16499, Korea
| | - So Hyun Park
- Department of Pathology, Ajou University School of Medicine, Suwon, 16499, Korea
| | - So Yeong Eom
- Inflamm-Aging Translational Research Center, Ajou University Medical Center, Suwon, 16499, Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, 16499, Korea
- Department of Pathology, Ajou University School of Medicine, Suwon, 16499, Korea
| | - Young-Sam Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Korea
| | - Wonhee John Lee
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Korea
| | - Juhee Jang
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Korea
| | - Daeha Seo
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Korea
| | - Hee Young Kang
- Inflamm-Aging Translational Research Center, Ajou University Medical Center, Suwon, 16499, Korea
- Department of Dermatology, Ajou University School of Medicine, Suwon, 16499, Korea
| | - Jin Cheol Kim
- Inflamm-Aging Translational Research Center, Ajou University Medical Center, Suwon, 16499, Korea
- Department of Dermatology, Ajou University School of Medicine, Suwon, 16499, Korea
| | - Su Bin Lim
- Inflamm-Aging Translational Research Center, Ajou University Medical Center, Suwon, 16499, Korea
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, 16499, Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, 16499, Korea
| | - Gyesoon Yoon
- Inflamm-Aging Translational Research Center, Ajou University Medical Center, Suwon, 16499, Korea
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, 16499, Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, 16499, Korea
| | - Hong Seok Kim
- Department of Molecular Medicine, College of Medicine, Inha University, Incheon, 22212, Korea
| | - Jang-Hee Kim
- Inflamm-Aging Translational Research Center, Ajou University Medical Center, Suwon, 16499, Korea.
- Department of Pathology, Ajou University School of Medicine, Suwon, 16499, Korea.
| | - Tae Jun Park
- Inflamm-Aging Translational Research Center, Ajou University Medical Center, Suwon, 16499, Korea.
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, 16499, Korea.
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, 16499, Korea.
| |
Collapse
|
10
|
Fleifel D, Cook JG. G1 Dynamics at the Crossroads of Pluripotency and Cancer. Cancers (Basel) 2023; 15:4559. [PMID: 37760529 PMCID: PMC10526231 DOI: 10.3390/cancers15184559] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/29/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
G1 cell cycle phase dynamics are regulated by intricate networks involving cyclins, cyclin-dependent kinases (CDKs), and CDK inhibitors, which control G1 progression and ensure proper cell cycle transitions. Moreover, adequate origin licensing in G1 phase, the first committed step of DNA replication in the subsequent S phase, is essential to maintain genome integrity. In this review, we highlight the intriguing parallels and disparities in G1 dynamics between stem cells and cancer cells, focusing on their regulatory mechanisms and functional outcomes. Notably, SOX2, OCT4, KLF4, and the pluripotency reprogramming facilitator c-MYC, known for their role in establishing and maintaining stem cell pluripotency, are also aberrantly expressed in certain cancer cells. In this review, we discuss recent advances in understanding the regulatory role of these pluripotency factors in G1 dynamics in the context of stem cells and cancer cells, which may offer new insights into the interconnections between pluripotency and tumorigenesis.
Collapse
Affiliation(s)
| | - Jeanette Gowen Cook
- Department of Biochemistry & Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| |
Collapse
|
11
|
Soe ZC, Ei ZZ, Visuttijai K, Chanvorachote P. Potential Natural Products Regulation of Molecular Signaling Pathway in Dermal Papilla Stem Cells. Molecules 2023; 28:5517. [PMID: 37513389 PMCID: PMC10384366 DOI: 10.3390/molecules28145517] [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: 05/25/2023] [Revised: 07/12/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Stem cells have demonstrated significant potential for tissue engineering and repair, anti-aging, and rejuvenation. Hair follicle stem cells can be found in the dermal papilla at the base of the follicle and the bulge region, and they have garnered increased attention because of their potential to regenerate hair as well as their application for tissue repair. In recent years, these cells have been shown to affect hair restoration and prevent hair loss. These stem cells are endowed with mesenchymal characteristics and exhibit self-renewal and can differentiate into diverse cell types. As research in this field continues, it is probable that insights regarding stem cell maintenance, as well as their self-renewal and differentiation abilities, will benefit the application of these cells. In addition, an in-depth discussion is required regarding the molecular basis of cellular signaling and the influence of nature-derived compounds in stimulating the stemness properties of dermal papilla stem cells. This review summarizes (i) the potential of the mesenchymal cells component of the hair follicle as a target for drug action; (ii) the molecular mechanism of dermal papilla stem cells for maintenance of their stem cell function; and (iii) the positive effects of the natural product compounds in stimulating stemness in dermal papilla stem cells. Together, these insights may help facilitate the development of novel effective hair loss prevention and treatment.
Collapse
Affiliation(s)
- Zar Chi Soe
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Zin Zin Ei
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Kittichate Visuttijai
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Pithi Chanvorachote
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
12
|
Lee SS, Vũ TT, Weiss AS, Yeo GC. Stress-induced senescence in mesenchymal stem cells: Triggers, hallmarks, and current rejuvenation approaches. Eur J Cell Biol 2023; 102:151331. [PMID: 37311287 DOI: 10.1016/j.ejcb.2023.151331] [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: 01/30/2023] [Revised: 06/04/2023] [Accepted: 06/05/2023] [Indexed: 06/15/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have emerged as promising cell-based therapies in the treatment of degenerative and inflammatory conditions. However, despite accumulating evidence of the breadth of MSC functional potency, their broad clinical translation is hampered by inconsistencies in therapeutic efficacy, which is at least partly due to the phenotypic and functional heterogeneity of MSC populations as they progress towards senescence in vitro. MSC senescence, a natural response to aging and stress, gives rise to altered cellular responses and functional decline. This review describes the key regenerative properties of MSCs; summarises the main triggers, mechanisms, and consequences of MSC senescence; and discusses current cellular and extracellular strategies to delay the onset or progression of senescence, or to rejuvenate biological functions lost to senescence.
Collapse
Affiliation(s)
- Sunny Shinchen Lee
- Charles Perkins Centre, The University of Sydney, NSW 2006, Australia; School of Life and Environmental Sciences, The University of Sydney, NSW 2006, Australia
| | - Thu Thuy Vũ
- Vinmec Research Institute of Stem Cell and Gene Technology, Vinmec Healthcare System, Hanoi, Viet Nam
| | - Anthony S Weiss
- Charles Perkins Centre, The University of Sydney, NSW 2006, Australia; School of Life and Environmental Sciences, The University of Sydney, NSW 2006, Australia; Sydney Nano Institute, The University of Sydney, NSW 2006, Australia
| | - Giselle C Yeo
- Charles Perkins Centre, The University of Sydney, NSW 2006, Australia; School of Life and Environmental Sciences, The University of Sydney, NSW 2006, Australia.
| |
Collapse
|
13
|
Sung JH. Effective and economical cell therapy for hair regeneration. Biomed Pharmacother 2023; 157:113988. [PMID: 36370520 DOI: 10.1016/j.biopha.2022.113988] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 11/01/2022] [Accepted: 11/05/2022] [Indexed: 11/11/2022] Open
Abstract
We reviewed and summarized the latest reports on the characteristics of stem cells and follicular cells that are under development for hair loss treatment. Compared with conventional medicine, cell therapy could be effective in the long term with a single treatment while having mild adverse effects. Adipose-derived stem cells (ASCs) have the advantages of easy access and large isolation amount compared with dermal papilla cells (DPCs) and dermal sheath cup cells (DSCs), and promote hair growth through the paracrine effect. ASCs have a poor potential in hair neogenesis, therefore, methods to enhance trichogenecity of ASCs should be developed. DSCs can be isolated from the peribulbar dermal sheath cup, while having immune tolerance, and hair inductivity. Therefore, DSCs were first developed and finished the phase II clinical trial; however, the hair growth was not satisfactory. Considering that a single injection of DSCs is effective for at least 9 months in the clinical setting, they can be an alternative therapy for hair regeneration. Though DPCs are not yet studied in clinical trials, we should pay attention to DPCs, as hair loss is associated with gradual reduction of DPCs and DP cell numbers fluctuate over the hair cycle. DPCs could make new hair follicles with epidermal cells, and have an immunomodulatory function to enable allogeneic transplantation. In addition, we can expand large quantities of DPCs with hair inductivity using spheroid culture, hypoxia condition, and growth factor supplement. 'Off-the-shelf' DPC therapy could be effective and economical, and therefore promising for hair regeneration.
Collapse
Affiliation(s)
- Jong-Hyuk Sung
- Epi Biotech Co., Ltd., Incheon, South Korea; College of Pharmacy, Institute of Pharmaceutical Sciences, Yonsei University, Incheon, South Korea.
| |
Collapse
|
14
|
Wang X, Yu F, Ye L. Epigenetic control of mesenchymal stem cells orchestrates bone regeneration. Front Endocrinol (Lausanne) 2023; 14:1126787. [PMID: 36950693 PMCID: PMC10025550 DOI: 10.3389/fendo.2023.1126787] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 02/17/2023] [Indexed: 03/08/2023] Open
Abstract
Recent studies have revealed the vital role of MSCs in bone regeneration. In both self-healing bone regeneration processes and biomaterial-induced healing of bone defects beyond the critical size, MSCs show several functions, including osteogenic differentiation and thus providing seed cells. However, adverse factors such as drug intake and body senescence can significantly affect the functions of MSCs in bone regeneration. Currently, several modalities have been developed to regulate MSCs' phenotype and promote the bone regeneration process. Epigenetic regulation has received much attention because of its heritable nature. Indeed, epigenetic regulation of MSCs is involved in the pathogenesis of a variety of disorders of bone metabolism. Moreover, studies using epigenetic regulation to treat diseases are also being reported. At the same time, the effects of epigenetic regulation on MSCs are yet to be fully understood. This review focuses on recent advances in the effects of epigenetic regulation on osteogenic differentiation, proliferation, and cellular senescence in MSCs. We intend to illustrate how epigenetic regulation of MSCs orchestrates the process of bone regeneration.
Collapse
Affiliation(s)
- Xiaofeng Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fanyuan Yu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Fanyuan Yu, ; Ling Ye,
| | - Ling Ye
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Fanyuan Yu, ; Ling Ye,
| |
Collapse
|
15
|
Rawangkan A, Wongsirisin P, Pook-In G, Siriphap A, Yosboonruang A, Kiddee A, Chuerduangphui J, Reukngam N, Duangjai A, Saokaew S, Praphasawat R. Dinactin: A New Antitumor Antibiotic with Cell Cycle Progression and Cancer Stemness Inhibiting Activities in Lung Cancer. Antibiotics (Basel) 2022; 11:antibiotics11121845. [PMID: 36551502 PMCID: PMC9774622 DOI: 10.3390/antibiotics11121845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022] Open
Abstract
Lung cancer, especially non-small cell lung cancer (NSCLC), is one of the most complex diseases, despite the existence of effective treatments such as chemotherapy and immunotherapy. Since cancer stem cells (CSCs) are responsible for chemo- and radio-resistance, metastasis, and cancer recurrence, finding new therapeutic targets for CSCs is critical. Dinactin is a natural secondary metabolite produced by microorganisms. Recently, dinactin has been revealed as a promising antitumor antibiotic via various mechanisms. However, the evidence relating to cell cycle progression regulation is constrained, and effects on cancer stemness have not been elucidated. Therefore, the aim of this study is to evaluate the new function of dinactin in anti-NSCLC proliferation, focusing on cell cycle progression and cancer stemness properties in Lu99 and A549 cells. Flow cytometry and immunoblotting analyses revealed that 0.1-1 µM of dinactin suppresses cell growth through induction of the G0/G1 phase associated with down-regulation of cyclins A, B, and D3, and cdk2 protein expression. The tumor-sphere forming capacity was used to assess the effect of dinactin on the cancer stemness potential in NSCLC cells. At a concentration of 1 nM, dinactin reduced both the number and size of the tumor-spheres. The quantitative RT-PCR analyses indicated that dinactin suppressed sphere formation by significantly reducing expression of CSC markers (i.e., ALDH1A1, Nanog, Oct4, and Sox2) in Lu99 cells. Consequently, dinactin could be a promising strategy for NSCLC therapy targeting CSCs.
Collapse
Affiliation(s)
- Anchalee Rawangkan
- Division of Microbiology and Parasitology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand
- UNIt of Excellence on Clinical Outcomes Research and IntegratioN (UNICORN), School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Pattama Wongsirisin
- Department of Medical Services, National Cancer Institute, Bangkok 10400, Thailand
| | - Grissana Pook-In
- Division of Microbiology and Parasitology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Achiraya Siriphap
- Division of Microbiology and Parasitology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Atchariya Yosboonruang
- Division of Microbiology and Parasitology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Anong Kiddee
- Division of Microbiology and Parasitology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand
| | | | - Nanthawan Reukngam
- Laboratory of Organic Synthesis, Chulabhorn Research Institute, Bangkok 10210, Thailand
| | - Acharaporn Duangjai
- Division of Physiology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Surasak Saokaew
- UNIt of Excellence on Clinical Outcomes Research and IntegratioN (UNICORN), School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
- Division of Social and Administrative Pharmacy, Department of Pharmaceutical Care, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Ratsada Praphasawat
- Department of Pathology, School of Medicine, University of Phayao, Phayao 56000, Thailand
- Correspondence: ; Tel.: +66-54466666 (ext. 3824) or +66-86-926-2448
| |
Collapse
|
16
|
Shin JY, Kim DY, Lee J, Shin YJ, Kim YS, Lee PH. Priming mesenchymal stem cells with α-synuclein enhances neuroprotective properties through induction of autophagy in Parkinsonian models. STEM CELL RESEARCH & THERAPY 2022; 13:483. [PMID: 36153562 PMCID: PMC9509608 DOI: 10.1186/s13287-022-03139-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 08/14/2022] [Indexed: 11/30/2022]
Abstract
Background Mesenchymal stem cells (MSCs) may be one of candidates for disease-modifying therapy in Parkinsonian diseases. As knowledge regarding the therapeutic properties of MSCs accumulates, some obstacles still remain to be overcome, especially, successful clinical translation requires the development of culture systems that mimic the natural MSC niche, while allowing clinical-scale cell expansion without compromising quality and function of the cells. In recent years, priming approaches using bioactive peptide or complement components have been investigated to enhance the therapeutic potential of MSCs. Methods We investigated an innovative priming strategy by conditioning the MSCs with α-synuclein (α-syn). To induce priming, MSCs were treated with different concentrations of α-syn and various time course. We evaluated whether α-syn enhances stemness properties of MSCs and priming MSCs with α-syn would modulate autophagy-related gene expression profiles. Results Treatment of naïve MSCs with α-syn upregulated transcriptional factors responsible for regulation of stemness, which was associated with the elevated expression of genes involved in glycolysis and cell re-programming. Primed MSCs with α-syn enhanced the expression of autophagy-regulating miRNA, and exosomes derived from primed MSCs were packed with autophagy-associated miRNA. In α-syn-overexpressing neuronal cells, primed MSCs with α-syn enhanced neuronal viability relative to naïve MSCs, through the induction of autophagy and lysosome activity. Animal study using an α-syn-overexpressing mice showed that the pro-survival effect of MSCs on dopaminergic neurons was more prominent in primed MSC-treated mice compared with that in naïve MSC-treated mice. Conclusions The present data suggest that MSC priming with α-syn exerts neuroprotective effects through augmented stemness and possibly the enhancement of autophagy-mediated α-syn modulation in Parkinsonian models. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03139-w.
Collapse
|
17
|
Jay Sarkar T, Hermsmeier M, L. Ross J, Scott Herron G. Genetic and Epigenetic Influences on Cutaneous Cellular Senescence. Physiology (Bethesda) 2022. [DOI: 10.5772/intechopen.101152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Skin is the largest human organ system, and its protective function is critical to survival. The epithelial, dermal, and subcutaneous compartments are heterogeneous mixtures of cell types, yet they all display age-related skin dysfunction through the accumulation of an altered phenotypic cellular state called senescence. Cellular senescence is triggered by complex and dynamic genetic and epigenetic processes. A senescence steady state is achieved in different cell types under various and overlapping conditions of chronological age, toxic injury, oxidative stress, replicative exhaustion, DNA damage, metabolic dysfunction, and chromosomal structural changes. These inputs lead to outputs of cell-cycle withdrawal and the appearance of a senescence-associated secretory phenotype, both of which accumulate as tissue pathology observed clinically in aged skin. This review details the influence of genetic and epigenetic factors that converge on normal cutaneous cellular processes to create the senescent state, thereby dictating the response of the skin to the forces of both intrinsic and extrinsic aging. From this work, it is clear that no single biomarker or process leads to senescence, but that it is a convergence of factors resulting in an overt aging phenotype.
Collapse
|
18
|
Ruiz-Aparicio PF, Vernot JP. Bone Marrow Aging and the Leukaemia-Induced Senescence of Mesenchymal Stem/Stromal Cells: Exploring Similarities. J Pers Med 2022; 12:jpm12050716. [PMID: 35629139 PMCID: PMC9147878 DOI: 10.3390/jpm12050716] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/20/2022] [Accepted: 04/27/2022] [Indexed: 12/17/2022] Open
Abstract
Bone marrow aging is associated with multiple cellular dysfunctions, including perturbed haematopoiesis, the propensity to haematological transformation, and the maintenance of leukaemia. It has been shown that instructive signals from different leukemic cells are delivered to stromal cells to remodel the bone marrow into a supportive leukemic niche. In particular, cellular senescence, a physiological program with both beneficial and deleterious effects on the health of the organisms, may be responsible for the increased incidence of haematological malignancies in the elderly and for the survival of diverse leukemic cells. Here, we will review the connection between BM aging and cellular senescence and the role that these processes play in leukaemia progression. Specifically, we discuss the role of mesenchymal stem cells as a central component of the supportive niche. Due to the specificity of the genetic defects present in leukaemia, one would think that bone marrow alterations would also have particular changes, making it difficult to envisage a shared therapeutic use. We have tried to summarize the coincident features present in BM stromal cells during aging and senescence and in two different leukaemias, acute myeloid leukaemia, with high frequency in the elderly, and B-acute lymphoblastic leukaemia, mainly a childhood disease. We propose that mesenchymal stem cells are similarly affected in these different leukaemias, and that the changes that we observed in terms of cellular function, redox balance, genetics and epigenetics, soluble factor repertoire and stemness are equivalent to those occurring during BM aging and cellular senescence. These coincident features may be used to explore strategies useful to treat various haematological malignancies.
Collapse
Affiliation(s)
- Paola Fernanda Ruiz-Aparicio
- Grupo de Investigación Fisiología Celular y Molecular, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 111321, Colombia;
| | - Jean-Paul Vernot
- Grupo de Investigación Fisiología Celular y Molecular, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 111321, Colombia;
- Instituto de Investigaciones Biomédicas, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá 111321, Colombia
- Correspondence:
| |
Collapse
|
19
|
Kukolj T, Lazarević J, Borojević A, Ralević U, Vujić D, Jauković A, Lazarević N, Bugarski D. A Single-Cell Raman Spectroscopy Analysis of Bone Marrow Mesenchymal Stem/Stromal Cells to Identify Inter-Individual Diversity. Int J Mol Sci 2022; 23:4915. [PMID: 35563306 PMCID: PMC9103070 DOI: 10.3390/ijms23094915] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 12/15/2022] Open
Abstract
The heterogeneity of stem cells represents the main challenge in regenerative medicine development. This issue is particularly pronounced when it comes to the use of primary mesenchymal stem/stromal cells (MSCs) due to a lack of identification markers. Considering the need for additional approaches in MSCs characterization, we applied Raman spectroscopy to investigate inter-individual differences between bone marrow MSCs (BM-MSCs). Based on standard biological tests, BM-MSCs of analyzed donors fulfill all conditions for their characterization, while no donor-related specifics were observed in terms of BM-MSCs morphology, phenotype, multilineage differentiation potential, colony-forming capacity, expression of pluripotency-associated markers or proliferative capacity. However, examination of BM-MSCs at a single-cell level by Raman spectroscopy revealed that despite similar biochemical background, fine differences in the Raman spectra of BM-MSCs of each donor can be detected. After extensive principal component analysis (PCA) of Raman spectra, our study revealed the possibility of this method to diversify BM-MSCs populations, whereby the grouping of cell populations was most prominent when cell populations were analyzed in pairs. These results indicate that Raman spectroscopy, as a label-free assay, could have a huge potential in understanding stem cell heterogeneity and sorting cell populations with a similar biochemical background that can be significant for the development of personalized therapy approaches.
Collapse
Affiliation(s)
- Tamara Kukolj
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia; (A.J.); (D.B.)
| | - Jasmina Lazarević
- Center for Solid State Physics and New Materials, Institute of Physics Belgrade, University of Belgrade, Pregrevica 118, 11080 Belgrade, Serbia; (J.L.); (U.R.); (N.L.)
| | - Ana Borojević
- Mother and Child Health Care Institute of Serbia ‘’Dr Vukan Čupić’’, 11000 Belgrade, Serbia; (A.B.); (D.V.)
| | - Uroš Ralević
- Center for Solid State Physics and New Materials, Institute of Physics Belgrade, University of Belgrade, Pregrevica 118, 11080 Belgrade, Serbia; (J.L.); (U.R.); (N.L.)
| | - Dragana Vujić
- Mother and Child Health Care Institute of Serbia ‘’Dr Vukan Čupić’’, 11000 Belgrade, Serbia; (A.B.); (D.V.)
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Aleksandra Jauković
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia; (A.J.); (D.B.)
| | - Nenad Lazarević
- Center for Solid State Physics and New Materials, Institute of Physics Belgrade, University of Belgrade, Pregrevica 118, 11080 Belgrade, Serbia; (J.L.); (U.R.); (N.L.)
| | - Diana Bugarski
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, 11129 Belgrade, Serbia; (A.J.); (D.B.)
| |
Collapse
|
20
|
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
|
21
|
Samoilova EM, Belopasov VV, Ekusheva EV, Zhang C, Troitskiy AV, Baklaushev VP. Epigenetic Clock and Circadian Rhythms in Stem Cell Aging and Rejuvenation. J Pers Med 2021; 11:1050. [PMID: 34834402 PMCID: PMC8620936 DOI: 10.3390/jpm11111050] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/12/2021] [Accepted: 10/14/2021] [Indexed: 12/12/2022] Open
Abstract
This review summarizes the current understanding of the interaction between circadian rhythms of gene expression and epigenetic clocks characterized by the specific profile of DNA methylation in CpG-islands which mirror the senescence of all somatic cells and stem cells in particular. Basic mechanisms of regulation for circadian genes CLOCK-BMAL1 as well as downstream clock-controlled genes (ССG) are also discussed here. It has been shown that circadian rhythms operate by the finely tuned regulation of transcription and rely on various epigenetic mechanisms including the activation of enhancers/suppressors, acetylation/deacetylation of histones and other proteins as well as DNA methylation. Overall, up to 20% of all genes expressed by the cell are subject to expression oscillations associated with circadian rhythms. Additionally included in the review is a brief list of genes involved in the regulation of circadian rhythms, along with genes important for cell aging, and oncogenesis. Eliminating some of them (for example, Sirt1) accelerates the aging process, while the overexpression of Sirt1, on the contrary, protects against age-related changes. Circadian regulators control a number of genes that activate the cell cycle (Wee1, c-Myc, p20, p21, and Cyclin D1) and regulate histone modification and DNA methylation. Approaches for determining the epigenetic age from methylation profiles across CpG islands in individual cells are described. DNA methylation, which characterizes the function of the epigenetic clock, appears to link together such key biological processes as regeneration and functioning of stem cells, aging and malignant transformation. Finally, the main features of adult stem cell aging in stem cell niches and current possibilities for modulating the epigenetic clock and stem cells rejuvenation as part of antiaging therapy are discussed.
Collapse
Affiliation(s)
- Ekaterina M. Samoilova
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA of Russia, 115682 Moscow, Russia; (A.V.T.); (V.P.B.)
| | | | - Evgenia V. Ekusheva
- Academy of Postgraduate Education of the Federal Scientific and Clinical Center for Specialized Types of Medical Care and Medical Technologies, FMBA of Russia, 125371 Moscow, Russia;
| | - Chao Zhang
- Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China;
| | - Alexander V. Troitskiy
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA of Russia, 115682 Moscow, Russia; (A.V.T.); (V.P.B.)
| | - Vladimir P. Baklaushev
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, FMBA of Russia, 115682 Moscow, Russia; (A.V.T.); (V.P.B.)
| |
Collapse
|
22
|
van Vliet T, Casciaro F, Demaria M. To breathe or not to breathe: Understanding how oxygen sensing contributes to age-related phenotypes. Ageing Res Rev 2021; 67:101267. [PMID: 33556549 DOI: 10.1016/j.arr.2021.101267] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/21/2021] [Accepted: 02/02/2021] [Indexed: 02/08/2023]
Abstract
Aging is characterized by a progressive loss of tissue integrity and functionality due to disrupted homeostasis. Molecular oxygen is pivotal to maintain tissue functions, and aerobic species have evolved a sophisticated sensing system to ensure proper oxygen supply and demand. It is not surprising that aberrations in oxygen and oxygen-associated pathways subvert health and promote different aspects of aging. In this review, we discuss emerging findings on how oxygen-sensing mechanisms regulate different cellular and molecular processes during normal physiology, and how dysregulation of oxygen availability lead to disease and aging. We describe various clinical manifestations associated with deregulation of oxygen balance, and how oxygen-modulating therapies and natural oxygen oscillations influence longevity. We conclude by discussing how a better understanding of oxygen-related mechanisms that orchestrate aging processes may lead to the development of new therapeutic strategies to extend healthy aging.
Collapse
|
23
|
REGγ regulates hair cycle by activating Lgr5 positive hair follicle stem cells. J Dermatol Sci 2021; 102:101-108. [PMID: 33933312 DOI: 10.1016/j.jdermsci.2021.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 03/27/2021] [Accepted: 04/12/2021] [Indexed: 01/14/2023]
Abstract
BACKGROUND REGγ acts as a proteasome activating factor mediating proteasome degradation of substrate proteins in an ATP and ubiquitination independent manner and also as an important regulator of cell cycle, proliferation and apoptosis. Hair cycle involves dynamic, continuous morphological changes of three stages (anagen, catagen and telogen). OBJECTIVE The function of REGγ in hair cycling is still unclear. METHODS Here, we used REGγ knockout 293 T cells, inducible 293WT and 293N151Y cell, REGγ knockout mice to identify the novel molecular mechanism of REGγ in regulating hair follicle stem cells. RESULTS In the present study, we found that REGγ deletion markedly delayed the transition of hair follicles from telogen to anagen and hair regeneration in mice. We also observed significant decrease of hair follicle stem cell number, stem-like property and proliferation ability. Interestingly, the results from real-time PCR, FACS, Western Blot and immunofluorescent analysis showed that REGγ deletion could greatly downregulate Lgr5 expression in the hair follicles. Meanwhile, REGγ was demonstrated to directly interact with LHX2 and promotes its degradation. Importantly, REGγ specific deletion in Lgr5+ stem cells induced the marked delay of hair regeneration after depilation. CONCLUSION These data together indicate that REGγ was a new mediator of Lgr5 expression in hair follicle at least partly by promoting the degradation of its suppressive transcription factor LHX2. It seemed that REGγ regulated hair anagen entry and hair regrowth by activating Lgr5 positive hair follicle stem cells.
Collapse
|
24
|
Kim HN, Shin JY, Kim DY, Lee JE, Lee PH. Priming mesenchymal stem cells with uric acid enhances neuroprotective properties in parkinsonian models. J Tissue Eng 2021; 12:20417314211004816. [PMID: 33854750 PMCID: PMC8013923 DOI: 10.1177/20417314211004816] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 03/05/2021] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are a potential source of cell-based disease-modifying therapy in Parkinsonian disorders. A promising approach to develop in vitro culture methods that mimic natural MSC niche is cell priming. Uric acid (UA), a powerful antioxidant, scavenges reactive oxygen species, which has a vital role in maintaining self-renewal and differentiation potential of MSCs. Here, we demonstrated that UA treatment in naïve MSCs stimulated glycolysis and upregulated transcriptional factors responsible for regulation of stemness, leading to increase in the expression levels of osteogenesis-, adipogenesis-, and chondrogenesis-related genes. UA-primed MSCs had more enhanced neuroprotective properties in cellular and parkinsonian animal models compared to naïve MSCs by inhibiting apoptotic signaling pathways. Additionally, expression of miR-137 and miR-145 was decreased in UA-treated MSCs. Our data demonstrated that priming MSCs with UA augment neuroprotective properties through enhanced self-renewal and differentiation potential, suggesting a practical strategy for improving the application of MSCs in parkinsonian disorders.
Collapse
Affiliation(s)
- Ha Na Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Jin Young Shin
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea.,Severance Biomedical Science Institute, Yonsei University, Seoul, Korea
| | - Dong Yeol Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Eun Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Phil Hyu Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea.,Severance Biomedical Science Institute, Yonsei University, Seoul, Korea
| |
Collapse
|
25
|
Deng Z, Wang W, Xu X, Nie Y, Liu Y, Gould OEC, Ma N, Lendlein A. Biofunction of Polydopamine Coating in Stem Cell Culture. ACS APPLIED MATERIALS & INTERFACES 2021; 13:10748-10759. [PMID: 33594879 DOI: 10.1021/acsami.0c22565] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
High levels of reactive oxygen species (ROS) during stem cell expansion often lead to replicative senescence. Here, a polydopamine (PDA)-coated substrate was used to scavenge extracellular ROS for mesenchymal stem cell (MSC) expansion. The PDA-coated substrate could reduce the oxidative stress and mitochondrial damage in replicative senescent MSCs. The expression of senescence-associated β-galactosidase of MSCs from three human donors (both bone marrow- and adipose tissue-derived) was suppressed on PDA. The MSCs on the PDA-coated substrate showed a lower level of interleukin 6 (IL-6), one of the senescence-associated inflammatory components. Cellular senescence-specific genes, such as p53 and p21, were downregulated on the PDA-coated substrate, while the stemness-related gene, OCT4, was upregulated. The PDA-coated substrate strongly promoted the proliferation rate of MSCs, while the stem cell character and differentiation potential were retained. Large-scale expansion of stem cells would greatly benefit from the PDA-coated substrate.
Collapse
Affiliation(s)
- Zijun Deng
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, 14513 Teltow, Germany
- Institute of Chemistry and Biochemistry, Free University of Berlin, 14195 Berlin, Germany
| | - Weiwei Wang
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, 14513 Teltow, Germany
| | - Xun Xu
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, 14513 Teltow, Germany
| | - Yan Nie
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, 14513 Teltow, Germany
- Institute of Chemistry, University of Potsdam, 14476 Potsdam, Germany
| | - Yue Liu
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, 14513 Teltow, Germany
- Institute of Chemistry, University of Potsdam, 14476 Potsdam, Germany
| | - Oliver E C Gould
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, 14513 Teltow, Germany
| | - Nan Ma
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, 14513 Teltow, Germany
- Institute of Chemistry and Biochemistry, Free University of Berlin, 14195 Berlin, Germany
| | - Andreas Lendlein
- Institute of Active Polymers and Berlin-Brandenburg Centre for Regenerative Therapies, Helmholtz-Zentrum Geesthacht, 14513 Teltow, Germany
- Institute of Chemistry and Biochemistry, Free University of Berlin, 14195 Berlin, Germany
- Institute of Chemistry, University of Potsdam, 14476 Potsdam, Germany
| |
Collapse
|
26
|
Wen L, Miao Y, Fan Z, Zhang J, Guo Y, Dai D, Huang J, Liu Z, Chen R, Hu Z. Establishment of an Efficient Primary Culture System for Human Hair Follicle Stem Cells Using the Rho-Associated Protein Kinase Inhibitor Y-27632. Front Cell Dev Biol 2021; 9:632882. [PMID: 33748117 PMCID: PMC7973216 DOI: 10.3389/fcell.2021.632882] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/15/2021] [Indexed: 12/11/2022] Open
Abstract
Background Hair follicle tissue engineering is a promising strategy for treating hair loss. Human hair follicle stem cells (hHFSCs), which play a key role in the hair cycle, have potential applications in regenerative medicine. However, previous studies did not achieve efficient hHFSC expansion in vitro using feeder cells. Therefore, there is a need to develop an efficient primary culture system for the expansion and maintenance of hHFSCs. Methods The hHFSCs were obtained by two-step proteolytic digestion combined with microscopy. The cell culture dishes were coated with human fibronectin and inoculated with hHFSCs. The hHFSCs were harvested using a differential enrichment procedure. The effect of Rho-associated protein kinase (ROCK) inhibitor Y-27632, supplemented in keratinocyte serum-free medium (K-SFM), on adhesion, proliferation, and stemness of hHFSCs and the underlying molecular mechanisms were evaluated. Results The hHFSCs cultured in K-SFM, supplemented with Y-27632, exhibited enhanced adhesion and proliferation. Additionally, Y-27632 treatment maintained the stemness of hHFSCs and promoted the ability of hHFSCs to regenerate hair follicles in vivo. However, Y-27632-induced proliferation and stemness in hHFSCs were conditional and reversible. Furthermore, Y-27632 maintained propagation and stemness of hHFSCs through the ERK/MAPK pathway. Conclusion An efficient short-term culture system for primary hHFSCs was successfully established using human fibronectin and the ROCK inhibitor Y-27632, which promoted the proliferation, maintained the stemness of hHFSCs and promoted the ability to regenerate hair follicles in vivo. The xenofree culturing method used in this study provided a large number of high-quality seed cells, which have applications in hair follicle tissue engineering and stem cell therapy.
Collapse
Affiliation(s)
- Lihong Wen
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yong Miao
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhexiang Fan
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiarui Zhang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yixuan Guo
- Department of Plastic and Burn Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Damao Dai
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junfei Huang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhen Liu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ruosi Chen
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiqi Hu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
27
|
Sun M, Tan J, Wang M, Wen W, He Y. Inorganic arsenic-mediated upregulation of AS3MT promotes proliferation of nonsmall cell lung cancer cells by regulating cell cycle genes. ENVIRONMENTAL TOXICOLOGY 2021; 36:204-212. [PMID: 32930475 DOI: 10.1002/tox.23026] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 06/11/2023]
Abstract
Long-term arsenic exposure can promote cancer through epigenetic mechanisms, and arsenite methyltransferase (AS3MT) plays an important role in this process. However, the expression patterns and mechanisms of AS3MT in arsenic carcinogenesis remain unclear. In this study, we found that the AS3MT was overexpressed in arsenic exposed population, non-small cell lung cancer (NSCLC) tissues, and A549 cells with sodium arsenite (NaAsO2 ) treatment for 48 hours. Besides, the level of AS3MT expression was positively correlated with the concentrations of urinary total arsenic (tAs), inorganic arsenic (iAs), methanearsonic acid (MMA), and dimethylarsinic acid (DMA) in all subjects. Functional experiments demonstrated that siRNA-mediated knockdown of AS3MT significantly inhibited proliferation of A549 cells. Mechanism investigation revealed that silencing of AS3MT inhibited proliferation by increasing mRNA expression levels of p21 and E2F1, and inhibiting CDK1, CDK2, CDK4, CDK6, Cyclin A2, Cyclin E1, Cyclin E2, and PCNA mRNA expression. Therefore, arsenic increased AS3MT expression in vivo and in vitro, which could directly act on the cell and affect the progression of NSCLC by regulating cell cycle genes.
Collapse
Affiliation(s)
- Mingjun Sun
- School of Public Health, Dali University, Dali, China
| | - Jingwen Tan
- School of Public Health, Kunming Medical University, Kunming, China
| | - Mengjie Wang
- School of Public Health, Kunming Medical University, Kunming, China
| | - Weihua Wen
- Yunnan Center for Disease Control and Prevention, Kunming, China
| | - Yuefeng He
- School of Public Health, Kunming Medical University, Kunming, China
| |
Collapse
|
28
|
Wang J, Qu J, Li Y, Feng Y, Ma J, Zhang L, Chu C, Hu H, Wang Y, Ji D. miR-149-5p Regulates Goat Hair Follicle Stem Cell Proliferation and Apoptosis by Targeting the CMTM3/AR Axis During Superior-Quality Brush Hair Formation. Front Genet 2020; 11:529757. [PMID: 33262781 PMCID: PMC7686784 DOI: 10.3389/fgene.2020.529757] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 09/25/2020] [Indexed: 12/21/2022] Open
Abstract
The Yangtze River Delta white goat is a unique goat species that can produce superior quality brush hair. CKLF-like MARVEL transmembrane domain-containing 3 (CMTM3), which influences the transcriptional activity of androgen receptor (AR), was identified as a candidate gene related to superior-quality brush hair formation. CMTM3 is generally expressed at low levels, but miR-149-5p is highly expressed in the skin tissues of these goats. The mechanism by which CMTM3 regulates the proliferation and apoptosis of goat hair follicle stem cells has not been elucidated. Here, RT-qPCR, western blotting, 5-ethynyl-2′-deoxyuridine (EdU), cell cycle, apoptosis, and dual-luciferase assays were used to investigate the role and regulatory mechanism of CMTM3 and miR-149-5p. Functional studies showed that CMTM3 overexpression inhibited proliferation and induced apoptosis in cultured hair follicle stem cells, whereas silencing CMTM3 markedly facilitated cell proliferation and deterred apoptosis in cultured hair follicle stem cells. Then, using bioinformatic predictions and the aforementioned assays, including dual-luciferase assays, RT-qPCR, and western blotting, we confirmed that miR-149-5p targets CMTM3 and preliminarily investigated the interaction between CMTM3 and AR in goat hair follicle stem cells. Furthermore, miR-149-5p overexpression significantly accelerated the proliferation and attenuated the apoptosis of hair follicle stem cells. Conversely, miR-149-5p inhibition suppressed the proliferation and induced the apoptosis of hair follicle stem cells. These results reveal a miR-149-5p-related regulatory framework for the miR-149-5p/CMTM3/AR axis during superior quality brush hair formation, in which CMTM3 plays a negative role.
Collapse
Affiliation(s)
- Jian Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Key Laboratory of Animal Genetics and Molecular Breeding of Jiangsu Province, Yangzhou University, Yangzhou, China
| | - Jingwen Qu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yongjun Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Key Laboratory of Animal Genetics and Molecular Breeding of Jiangsu Province, Yangzhou University, Yangzhou, China
| | - Yunkui Feng
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Jinliang Ma
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Liuming Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Changjiang Chu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Huiru Hu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yanhu Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Dejun Ji
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| |
Collapse
|
29
|
Casciaro F, Borghesan M, Beretti F, Zavatti M, Bertucci E, Follo MY, Maraldi T, Demaria M. Prolonged hypoxia delays aging and preserves functionality of human amniotic fluid stem cells. Mech Ageing Dev 2020; 191:111328. [DOI: 10.1016/j.mad.2020.111328] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 08/07/2020] [Accepted: 08/09/2020] [Indexed: 01/10/2023]
|
30
|
Wang B, Liu F, Liu Z, Han X, Lian A, Zhang Y, Zuo K, Wang Y, Liu M, Zou F, Jiang Y, Jin M, Liu X, Liu J. Internalization of the TAT-PBX1 fusion protein significantly enhances the proliferation of human hair follicle-derived mesenchymal stem cells and delays their senescence. Biotechnol Lett 2020; 42:1877-1885. [PMID: 32436118 DOI: 10.1007/s10529-020-02909-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 05/07/2020] [Indexed: 01/20/2023]
Abstract
OBJECTIVES To express a TAT-PBX1 fusion protein using a prokaryotic expression system and to explore potential effects of TAT-PBX1 in the proliferation and senescence of human hair follicle-derived mesenchymal stem cells. RESULTS The TAT-PBX1 fusion was produced in inclusion bodies and heterogenously expressed in Rosetta (DE3) cells. Immunofluorescence staining showed that TAT-PBX1 fusion proteins were internalized by human hair follicle-derived mesenchymal stem cells. The growth rate of cells was increased after treatment with more than 5.0 μg/mL of TAT-PBX1. The rate of senescence-associated β-galactosidase positive cells was reduced in the 10.0 μg/mL TAT-PBX1 group (28%) than the 0 μg/mL control group (60%). Cells treated with the TAT-PBX1 fusion protein showed higher expression of p-AKT (1.22-fold that of the control), which indicates that TAT-PBX1 activated AKT pathway after cellular uptake. CONCLUSIONS The TAT-PBX1 fusion protein increased the proliferation of hair follicle mesenchymal stem cells and delayed their senescence by activating the AKT pathway following internalization by cells.
Collapse
Affiliation(s)
- Bo Wang
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021, China
| | - Feilin Liu
- Department of Ophthalmology, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, 130021, China
| | - Zinan Liu
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021, China
| | - Xing Han
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021, China
| | - Aobo Lian
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021, China
| | - Yuying Zhang
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021, China
| | - Kuiyang Zuo
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021, China
| | - Yuan Wang
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021, China
| | - Mingsheng Liu
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021, China
| | - Fei Zou
- Department of Pediatrics, The First Hospital of Jilin University, 71 Xinmin Avenue, Changchun, 130021, China
| | - Yixu Jiang
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021, China
| | - Minghua Jin
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021, China
| | - Xiaomei Liu
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021, China.
| | - Jinyu Liu
- Department of Toxicology, School of Public Health, Jilin University, 1163 Xinmin Avenue, Changchun, 130021, China.
| |
Collapse
|
31
|
Epigenetic Clock: DNA Methylation in Aging. Stem Cells Int 2020; 2020:1047896. [PMID: 32724310 PMCID: PMC7366189 DOI: 10.1155/2020/1047896] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/11/2020] [Accepted: 06/20/2020] [Indexed: 02/07/2023] Open
Abstract
Aging, which is accompanied by decreased organ function and increased disease incidence, limits human lifespan and has attracted investigators for thousands of years. In recent decades, with the rapid development of biology, scientists have shown that epigenetic modifications, especially DNA methylation, are key regulators involved in this process. Regular fluctuations in global DNA methylation levels have been shown to accurately estimate biological age and disease prognosis. In this review, we discuss recent findings regarding the relationship between variations in DNA methylation level patterns and aging. In addition, we introduce the known mechanisms by which DNA methylation regulators affect aging and related diseases. As more studies uncover the mechanisms by which DNA methylation regulates aging, antiaging interventions and treatments for related diseases may be developed that enable human life extension.
Collapse
|
32
|
Wang B, Liu XM, Liu ZN, Wang Y, Han X, Lian AB, Mu Y, Jin MH, Liu JY. Human hair follicle-derived mesenchymal stem cells: Isolation, expansion, and differentiation. World J Stem Cells 2020; 12:462-470. [PMID: 32742563 PMCID: PMC7360986 DOI: 10.4252/wjsc.v12.i6.462] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/18/2020] [Accepted: 05/29/2020] [Indexed: 02/06/2023] Open
Abstract
Hair follicles are easily accessible skin appendages that protect against cold and potential injuries. Hair follicles contain various pools of stem cells, such as epithelial, melanocyte, and mesenchymal stem cells (MSCs) that continuously self-renew, differentiate, regulate hair growth, and maintain skin homeostasis. Recently, MSCs derived from the dermal papilla or dermal sheath of the human hair follicle have received attention because of their accessibility and broad differentiation potential. In this review, we describe the applications of human hair follicle-derived MSCs (hHF-MSCs) in tissue engineering and regenerative medicine. We have described protocols for isolating hHF-MSCs from human hair follicles and their culture condition in detail. We also summarize strategies for maintaining hHF-MSCs in a highly proliferative but undifferentiated state after repeated in vitro passages, including supplementation of growth factors, 3D suspension culture technology, and 3D aggregates of MSCs. In addition, we report the potential of hHF-MSCs in obtaining induced smooth muscle cells and tissue-engineered blood vessels, regenerated hair follicles, induced red blood cells, and induced pluripotent stem cells. In summary, the abundance, convenient accessibility, and broad differentiation potential make hHF-MSCs an ideal seed cell source of regenerative medical and cell therapy.
Collapse
Affiliation(s)
- Bo Wang
- Department of Toxicology, School of Public Health, Jilin University, Changchun 130021, Jilin Province, China
| | - Xiao-Mei Liu
- Department of Toxicology, School of Public Health, Jilin University, Changchun 130021, Jilin Province, China
| | - Zi-Nan Liu
- Department of Toxicology, School of Public Health, Jilin University, Changchun 130021, Jilin Province, China
| | - Yuan Wang
- Department of Toxicology, School of Public Health, Jilin University, Changchun 130021, Jilin Province, China
| | - Xing Han
- Department of Toxicology, School of Public Health, Jilin University, Changchun 130021, Jilin Province, China
| | - Ao-Bo Lian
- Department of Toxicology, School of Public Health, Jilin University, Changchun 130021, Jilin Province, China
| | - Ying Mu
- Research Center for Analytical Instrumentation, Institute of Cyber-Systems and Control, State Key Laboratory of Industrial Control Technology, Zhejiang University, Hangzhou 310000, Zhejiang Province, China
| | - Ming-Hua Jin
- Department of Toxicology, School of Public Health, Jilin University, Changchun 130021, Jilin Province, China
| | - Jin-Yu Liu
- Department of Toxicology, School of Public Health, Jilin University, Changchun 130021, Jilin Province, China
| |
Collapse
|
33
|
Zhou X, Hong Y, Zhang H, Li X. Mesenchymal Stem Cell Senescence and Rejuvenation: Current Status and Challenges. Front Cell Dev Biol 2020; 8:364. [PMID: 32582691 PMCID: PMC7283395 DOI: 10.3389/fcell.2020.00364] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 04/23/2020] [Indexed: 12/14/2022] Open
Abstract
Over the past decades, mesenchymal stem cell (MSC)-based therapy has been intensively investigated and shown promising results in the treatment of various diseases due to their easy isolation, multiple lineage differentiation potential and immunomodulatory effects. To date, hundreds of phase I and II clinical trials using MSCs have been completed and many are ongoing. Accumulating evidence has shown that transplanted allogeneic MSCs lose their beneficial effects due to immunorejection. Nevertheless, the function of autologous MSCs is adversely affected by age, a process termed senescence, thus limiting their therapeutic potential. Despite great advances in knowledge, the potential mechanisms underlying MSC senescence are not entirely clear. Understanding the molecular mechanisms that contribute to MSC senescence is crucial when exploring novel strategies to rejuvenate senescent MSCs. In this review, we aim to provide an overview of the biological features of senescent MSCs and the recent progress made regarding the underlying mechanisms including epigenetic changes, autophagy, mitochondrial dysfunction and telomere shortening. We also summarize the current approaches to rejuvenate senescent MSCs including gene modification and pretreatment strategies. Collectively, rejuvenation of senescent MSCs is a promising strategy to enhance the efficacy of autologous MSC-based therapy, especially in elderly patients.
Collapse
Affiliation(s)
- Xueke Zhou
- Department of Emergency Medicine, Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| | - Yimei Hong
- Department of Emergency Medicine, Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hao Zhang
- School of Pharmacy, Bengbu Medical College, Bengbu, China
| | - Xin Li
- Department of Emergency Medicine, Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
34
|
Clemente-Periván SI, Gómez-Gómez Y, Leyva-Vázquez MA, Lagunas-Martínez A, Organista-Nava J, Illades-Aguiar B. Role of Oct3/4 in Cervical Cancer Tumorigenesis. Front Oncol 2020; 10:247. [PMID: 32219062 PMCID: PMC7079573 DOI: 10.3389/fonc.2020.00247] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 02/13/2020] [Indexed: 12/16/2022] Open
Abstract
Cervical cancer (CC) is the fourth most common type of cancer that affects women. Compared to other types of cancer, CC has a high mortality rate in women worldwide. Several factors contribute to the development of CC, but persistent high-risk human papillomavirus infection is the main etiologic agent associated with the development of CC. Moreover, several studies reported that alterations in the expression of transcription factors present in a small subpopulation of cells within tumors called cancer stem cells (CSCs), which contribute to the development of CC by promoting tumorigenicity and metastasis. These transcription factors affect self-renewal and maintenance of pluripotency and differentiation in stem cells. OCT3/4 belongs to the family of transcription factors with the POU domain. It consists of five exons and can be edited by alternative splicing into three main transcripts: OCT3/4A, OCT3/4B, and OCT3/4B1. The OCT3/4 expression in CSCs promotes carcinogenesis and the development of malignant tumors, and the loss of expression leads to the loss of self-renewal and proliferation and favors apoptosis. This review describes the main roles of OCT3/4 in CC and its importance in several biological processes that contribute to the development of CC and may serve as molecular targets to improve prognosis of CC.
Collapse
Affiliation(s)
- Sayuri Itzel Clemente-Periván
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Mexico
| | - Yazmín Gómez-Gómez
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Mexico
| | - Marco Antonio Leyva-Vázquez
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Mexico
| | - Alfredo Lagunas-Martínez
- Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | - Jorge Organista-Nava
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Mexico
| | - Berenice Illades-Aguiar
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Mexico
| |
Collapse
|
35
|
Neri S, Borzì RM. Molecular Mechanisms Contributing to Mesenchymal Stromal Cell Aging. Biomolecules 2020; 10:E340. [PMID: 32098040 PMCID: PMC7072652 DOI: 10.3390/biom10020340] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/13/2020] [Accepted: 02/19/2020] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are a reservoir for tissue homeostasis and repair that age during organismal aging. Beside the fundamental in vivo role of MSCs, they have also emerged in the last years as extremely promising therapeutic agents for a wide variety of clinical conditions. MSC use frequently requires in vitro expansion, thus exposing cells to replicative senescence. Aging of MSCs (both in vivo and in vitro) can affect not only their replicative potential, but also their properties, like immunomodulation and secretory profile, thus possibly compromising their therapeutic effect. It is therefore of critical importance to unveil the underlying mechanisms of MSC senescence and to define shared methods to assess MSC aging status. The present review will focus on current scientific knowledge about MSC aging mechanisms, control and effects, including possible anti-aging treatments.
Collapse
Affiliation(s)
- Simona Neri
- IRCCS Istituto Ortopedico Rizzoli, Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, 40136 Bologna, Italy;
| | | |
Collapse
|
36
|
Balzano F, Campesi I, Cruciani S, Garroni G, Bellu E, Dei Giudici S, Angius A, Oggiano A, Rallo V, Capobianco G, Dessole S, Ventura C, Montella A, Maioli M. Epigenetics, Stem Cells, and Autophagy: Exploring a Path Involving miRNA. Int J Mol Sci 2019; 20:5091. [PMID: 31615086 PMCID: PMC6834298 DOI: 10.3390/ijms20205091] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/07/2019] [Accepted: 10/10/2019] [Indexed: 02/07/2023] Open
Abstract
MiRNAs, a small family of non-coding RNA, are now emerging as regulators of stem cell pluripotency, differentiation, and autophagy, thus controlling stem cell behavior. Stem cells are undifferentiated elements capable to acquire specific phenotype under different kind of stimuli, being a main tool for regenerative medicine. Within this context, we have previously shown that stem cells isolated from Wharton jelly multipotent stem cells (WJ-MSCs) exhibit gender differences in the expression of the stemness related gene OCT4 and the epigenetic modulator gene DNA-Methyltransferase (DNMT1). Here, we further analyze this gender difference, evaluating adipogenic and osteogenic differentiation potential, autophagic process, and expression of miR-145, miR-148a, and miR-185 in WJ-MSCs derived from males and females. These miRNAs were selected since they are involved in OCT4 and DNMT1 gene expression, and in stem cell differentiation. Our results indicate a difference in the regulatory circuit involving miR-148a/DNMT1/OCT4 autophagy in male WJ-MSCs as compared to female cells. Moreover, no difference was detected in the expression of the two-differentiation regulating miRNA (miR-145 and miR-185). Taken together, our results highlight a different behavior of WJ-MSCs from males and females, disclosing the chance to better understand cellular processes as autophagy and stemness, usable for future clinical applications.
Collapse
Affiliation(s)
- Francesca Balzano
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
| | - Ilaria Campesi
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
| | - Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
| | - Giuseppe Garroni
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
| | - Emanuela Bellu
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
| | - Silvia Dei Giudici
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, 07100 Sassari, Italy.
| | - Andrea Angius
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato, 09042 Cagliari, Italy.
| | - Annalisa Oggiano
- Istituto Zooprofilattico Sperimentale della Sardegna, Via Vienna 2, 07100 Sassari, Italy.
| | - Vincenzo Rallo
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato, 09042 Cagliari, Italy.
| | - Giampiero Capobianco
- Department of Medical, Surgical and experimental Sciences, Gynecologic and Obstetric Clinic, University of Sassari, 07100 Sassari, Italy.
| | - Salvatore Dessole
- Department of Medical, Surgical and experimental Sciences, Gynecologic and Obstetric Clinic, University of Sassari, 07100 Sassari, Italy.
| | - Carlo Ventura
- National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB)-Eldor Lab, at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
| | - Andrea Montella
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
- Operative Unit of Clinical Genetics and Developmental Biology, Viale San Pietro 43/B, 07100 Sassari, Italy.
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato, 09042 Cagliari, Italy.
- Center for developmental biology and reprogramming-CEDEBIOR, Department of Biomedical Sciences, University of Sassari Viale San Pietro 43/B, 07100 Sassari, Italy.
| |
Collapse
|
37
|
Liu Y, Zhao S, Luo L, Wang J, Zhu Z, Xiang Q, Deng Y, Zhao Z. Mesenchymal stem cell-derived exosomes ameliorate erection by reducing oxidative stress damage of corpus cavernosum in a rat model of artery injury. J Cell Mol Med 2019; 23:7462-7473. [PMID: 31512385 PMCID: PMC6815831 DOI: 10.1111/jcmm.14615] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/01/2019] [Accepted: 08/09/2019] [Indexed: 12/13/2022] Open
Abstract
Erectile dysfunction (ED) is a common ageing male's disease, and vascular ED accounts for the largest proportion of all types of ED. One of the mechanisms of vascular ED in the clinic is arterial insufficiency, which mainly caused by atherosclerosis, trauma and surgical. Moreover, oxidative stress damage after tissue ischemia usually aggravated the progress of ED. As a new way of acellular therapy, mesenchymal stem cell-derived exosomes (MSC-Exos) have great potential in ED treatment. In the current study, we have explored the mechanism of MSC-Exos therapy in a rat model of internal iliac artery injury-induced ED. Compared with intracavernous (IC) injection of phosphate-buffered saline after artery injury, of note, we observed that both mesenchymal stem cells (MSCs) and MSC-Exos through IC injection could improve the erectile function to varying degrees. More specifically, IC injection MSC-Exos could promote cavernous sinus endothelial formation, reduce the organization oxidative stress damage, and improve the nitric oxide synthase and smooth muscle content in the corpus cavernosum. With similar potency compared with the stem cell therapy and other unique advantages, IC injection of MSC- Exos could be an effective treatment to ameliorate erectile function in a rat model of arterial injury.
Collapse
Affiliation(s)
- Yangzhou Liu
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shankun Zhao
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Urology, Zhejiang Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Lianmin Luo
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiamin Wang
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhiguo Zhu
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qian Xiang
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yihan Deng
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhigang Zhao
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
38
|
Yonemura H, Futakuchi A, Inoue-Mochita M, Fujimoto T, Takahashi E, Tanihara H, Inoue T. DNA methyltransferase inhibitor suppresses fibrogenetic changes in human conjunctival fibroblasts. Mol Vis 2019; 25:382-390. [PMID: 31523116 PMCID: PMC6707755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 07/19/2019] [Indexed: 11/11/2022] Open
Abstract
Purpose This study aimed to clarify the effects of a DNA methyltransferase inhibitor on fibrogenetic changes in human conjunctival fibroblasts (HConF). Methods HConF were pretreated with the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine (5-Aza-dC) for 48 h. After one passage, the cells were treated with 5 ng/ml of transforming growth factor (TGF)-β2 for 48 h, and the expression levels of α-smooth muscle actin (α-SMA), extracellular matrix proteins, and phosphorylated Smad3 were evaluated with western blotting. A fusion construct between the COL1A2 promoter and the luciferase gene was introduced into the HConF after the first passage, and the construct's activity was detected via a luciferase reporter gene assay. Results TGF-β2-induced upregulation of α-SMA was suppressed by pretreatment with 5-Aza-dC (0.1, 1.0, and 10 μM) in a dose-dependent manner. Upregulation of type I collagen was also suppressed by 10 μM 5-Aza-dC pretreatment. In contrast, 5-Aza-dC had no inhibitory effect on the expression of fibronectin or phosphorylated Smad3. However, COL1A2 promoter activity was suppressed with 5-Aza-dC pretreatment. Conclusions In HConF, fibrogenetic changes were partly suppressed with a DNA methyltransferase inhibitor, suggesting an indirect inhibitory effect of the inhibitor on the COL1A2 promoter in HConF.
Collapse
Affiliation(s)
- Hitomi Yonemura
- Department of Ophthalmology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Akiko Futakuchi
- Department of Ophthalmology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Miyuki Inoue-Mochita
- Department of Ophthalmology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Tomokazu Fujimoto
- Department of Ophthalmology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Eri Takahashi
- Department of Ophthalmology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | | | - Toshihiro Inoue
- Department of Ophthalmology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
39
|
Fafián-Labora JA, Morente-López M, Arufe MC. Effect of aging on behaviour of mesenchymal stem cells. World J Stem Cells 2019; 11:337-346. [PMID: 31293716 PMCID: PMC6600848 DOI: 10.4252/wjsc.v11.i6.337] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 03/29/2019] [Accepted: 05/06/2019] [Indexed: 02/06/2023] Open
Abstract
Organs whose source is the mesoderm lineage contain a subpopulation of stem cells that are able to differentiate among mesodermal derivatives (chondrocytes, osteocytes, adipocytes). This subpopulation of adult stem cells, called "mesenchymal stem cells" or "mesenchymal stromal cells (MSCs)", contributes directly to the homeostatic maintenance of their organs; hence, their senescence could be very deleterious for human bodily functions. MSCs are easily isolated and amenable their expansion in vitro because of the research demanding to test them in many diverse clinical indications. All of these works are shown by the rapidly expanding literature that includes many in vivo animal models. We do not have an in-depth understanding of mechanisms that induce cellular senescence, and to further clarify the consequences of the senescence process in MSCs, some hints may be derived from the study of cellular behaviour in vivo and in vitro, autophagy, mitochondrial stress and exosomal activity. In this particular work, we decided to review these biological features in the literature on MSC senescence over the last three years.
Collapse
Affiliation(s)
- Juan Antonio Fafián-Labora
- Grupo de Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Ciencias Biomédicas y Medicina, Universidade da Coruña, A Coruña 15006, Spain
| | - Miriam Morente-López
- Grupo de Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Ciencias Biomédicas y Medicina, Universidade da Coruña, A Coruña 15006, Spain
| | - María C Arufe
- Grupo de Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Ciencias Biomédicas y Medicina, Universidade da Coruña, A Coruña 15006, Spain.
| |
Collapse
|
40
|
Jin X, Li Y, Guo Y, Jia Y, Qu H, Lu Y, Song P, Zhang X, Shao Y, Qi D, Xu W, Quan C. ERα is required for suppressing OCT4-induced proliferation of breast cancer cells via DNMT1/ISL1/ERK axis. Cell Prolif 2019; 52:e12612. [PMID: 31012189 PMCID: PMC6668970 DOI: 10.1111/cpr.12612] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/04/2019] [Accepted: 03/18/2019] [Indexed: 12/12/2022] Open
Abstract
Objective POU5F1 (OCT4) is implicated in cancer stem cell self‐renewal. Currently, some studies have shown that OCT4 has a dual function in suppressing or promoting cancer progression. However, the precise molecular mechanism of OCT4 in breast cancer progression remains unclear. Materials and Methods RT‐PCR and Western blot were utilized to investigate OCT4 expression in breast cancer tissues and cells. Cell proliferation assays and mouse models were applied to determine the effects of OCT4 on breast cancer cell proliferation. DNMT1 inhibitors, ChIP, CoIP, IHC and ERα inhibitors were used to explore the molecular mechanism of OCT4 in breast cancer. Results OCT4 was down‐regulated in breast cancer tissues, and the overexpression of OCT4 promoted MDA‐MB‐231 cell proliferation and inhibited the proliferation of MCF‐7 cells in vitro and in vivo, respectively. Two DNMT1 inhibitors (5‐aza‐dC and zebularine) suppressed OCT4‐induced MDA‐MB‐231 cell proliferation through Ras/Raf1/ERK inactivation by targeting ISL1, which is the downstream of DNMT1. In contrast, OCT4 interacted with ERα, decreased DNMT1 expression and inactivated the Ras/Raf1/ERK signalling pathway in MCF‐7 cells. Moreover, ERα inhibitor (AZD9496) reversed the suppression of OCT4‐induced proliferation in MCF‐7 cells via the activation of ERK signalling pathway. Conclusions OCT4 is dependent on ERα to suppress the proliferation of breast cancer cells through DNMT1/ISL1/ERK axis.
Collapse
Affiliation(s)
- Xiangshu Jin
- The Key Laboratory of Pathology, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| | - Yanru Li
- The Key Laboratory of Pathology, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| | - Yantong Guo
- The Key Laboratory of Pathology, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| | - Yiyang Jia
- The Key Laboratory of Pathology, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| | - Huinan Qu
- The Key Laboratory of Pathology, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| | - Yan Lu
- The Key Laboratory of Pathology, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| | - Peiye Song
- The Key Laboratory of Pathology, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| | - Xiaoli Zhang
- The Key Laboratory of Pathology, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| | - Yijia Shao
- The Key Laboratory of Pathology, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| | - Da Qi
- The Key Laboratory of Pathology, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| | - Wenhong Xu
- The Key Laboratory of Pathology, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| | - Chengshi Quan
- The Key Laboratory of Pathology, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| |
Collapse
|