1
|
Mante N, Undale V, Sanap A, Bhonde R, Tambe P, Bansode M, Gupta RK. Disease microenvironment preconditioning: An evolving approach to improve therapeutic efficacy of human mesenchymal stromal cells. Int Immunopharmacol 2025; 157:114701. [PMID: 40300358 DOI: 10.1016/j.intimp.2025.114701] [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: 01/14/2025] [Revised: 04/10/2025] [Accepted: 04/18/2025] [Indexed: 05/01/2025]
Abstract
Despite the tremendous success in preclinical models, the translation of human mesenchymal stromal cells (hMSCs) as a therapy in the clinic is not up to the expectation. Intrinsic factors (age, sex, health status, life style of the donor, source, cellular senescence, and oxidative stress in hMSCs), extrinsic factors (culture system, batch-to-batch variations, choice of biomaterials, cell processing and preservation protocols), and host microenvironment (inflammatory milieu, oxidative stress, and hypoxia in the recipient) compromise the overall therapeutic efficacy of the transplanted hMSCs. In recent times, the approach of 'Disease Microenvironment Preconditioning (DMP)' has garnered attention to overcome the host-associated attributes involved in compromised hMSCs therapeutic potential. In this review, we discuss various approaches of DMP of hMSCs by employing serum and other body fluids obtained from diseased patients/animals and small molecules, including cytokines such as IFN-γ, IL-6, IL-10, IL- β, TGF-β1, IL-1α, IL-1β, TNF-α, HMGB1, IL-17 A, and IL-8 which are associated with disease conditions. DMP strengthens hMSCs ability to adapt/acclimatize and respond more efficiently to the hostile microenvironment they encounter upon transplantation. DMP modulate hMSCs to withstand inflammation, survive under hypoxic and nutrient-deprived conditions, and resist oxidative stress. Evidence from various disease models ranging from cardiovascular and neurodegenerative disorders to autoimmune diseases and tissue injuries supports the role of DMP in improving hMSC survival, integration, and functional efficacy. While the potential of DMP to revolutionize MSC-based therapies is evident, challenges such as standardizing/optimizing protocols for preconditioning is essential. This review synthesizes current advancements in the approach of DMP aiming to propel the area of regenerative medicine.
Collapse
Affiliation(s)
- Nishant Mante
- Department of Pharmacology, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune 411018, India; Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pune 411018, India; Department of Pharmacology, School of Pharmacy and Research, Dr. D. Y. Patil Dnyan Prasad University, Pimpri, Pune 411018, India
| | - Vaishali Undale
- Department of Pharmacology, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune 411018, India; Department of Pharmacology, School of Pharmacy and Research, Dr. D. Y. Patil Dnyan Prasad University, Pimpri, Pune 411018, India.
| | - Avinash Sanap
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pune 411018, India.
| | - Ramesh Bhonde
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pune 411018, India
| | - Pratima Tambe
- Department of Pharmacology, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune 411018, India; Department of Pharmacology, School of Pharmacy and Research, Dr. D. Y. Patil Dnyan Prasad University, Pimpri, Pune 411018, India
| | - Manoj Bansode
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, Maharashtra, India
| | - Rajesh Kumar Gupta
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune 411033, Maharashtra, India
| |
Collapse
|
2
|
Yuan Z, Zhang Y, He X, Wang X, Wang X, Ren S, Su J, Shen J, Li X, Xiao Z. Engineering mesenchymal stem cells for premature ovarian failure: overcoming challenges and innovating therapeutic strategies. Theranostics 2024; 14:6487-6515. [PMID: 39479455 PMCID: PMC11519806 DOI: 10.7150/thno.102641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024] Open
Abstract
Premature ovarian failure (POF) is a leading cause of infertility in women, causing significant psychological and physical distress. Current therapeutic options are limited, necessitating the exploration of new treatments. Mesenchymal stem cells (MSCs), known for their remarkable homing and regenerative properties, have emerged as a promising intervention for POF. However, their clinical efficacy has been inconsistent. This paper aims to address these challenges by examining the cellular heterogeneity within MSC populations, which is crucial for identifying and selecting specific functional subpopulations for clinical applications. Understanding this heterogeneity can enhance therapeutic efficacy and ensure treatment stability. Additionally, this review comprehensively examines the literature on the effectiveness, safety, and ethical considerations of MSCs for ovarian regeneration, with a focus on preclinical and clinical trials. We also discuss potential strategies involving genetically and tissue-engineered MSCs. By integrating insights from these studies, we propose new directions for the design of targeted MSC treatments for POF and related disorders, potentially improving outcomes, addressing safety concerns, and expanding therapeutic options while ensuring ethical compliance.
Collapse
Affiliation(s)
- Zijun Yuan
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yinping Zhang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xinyu He
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiang Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xingyue Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Siqi Ren
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jiahong Su
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
| | - Xiang Li
- Sichuan College of Traditional Chinese Medicine, Sichuan Mianyang 621000, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Department of Pharmacology, School of Pharmacy, Sichuan College of Traditional Chinese Medicine, Sichuan Mianyang 621000, China
- Luzhou People's Hospital, Luzhou, Sichuan, China
| |
Collapse
|
3
|
Ghasemzadeh N, Pourrajab F, Dehghani Firoozabadi A, Rahnama M. Liposome-Mediated MicroRNA Delivery: An Additional Layer of Gene Network Regulation and Nuclear Reprogramming. IRANIAN BIOMEDICAL JOURNAL 2024; 28:245-54. [PMID: 39891467 PMCID: PMC11829158 DOI: 10.61186/ibj.4271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 10/29/2024] [Indexed: 02/10/2025]
Abstract
Background Developing miRNA-mediated cell engineering introduces a novel technology for cell reprogramming and generating patient-specific tissues for therapeutic use, facilitating basic research on human adult stem cells. Furthermore, optimizing a reprogramming method without transduction minimizes the risk of tumorigenesis, especially for reprogrammed cells. This study aimed to explore the use of liposomes as vehicles for delivering miRNAs to cells, focusing on their role in regulating gene networks and facilitating nuclear reprogramming. Methods This study utilized cationic liposomal nanoparticles preserved under different conditions to introduce miRNAs into hMSCs. Using qPCR, the effective induction of pluripotency factors (OCT4, SOX2, and NANOG) was examined. Results Results indicated that miR-302a and miR-34a regulate pluripotency by interacting with key transcription factors, including OCT4, SOX2, and NANOG. Notably, the expression pattern of OCT4 showed that lipoplexes containing miR-302a increased the expression of this gene, while in the case of miR-34a, it decreased. Additionally, the study found that pluripotency precursors can be induced by delivering liposomal microRNA (LP-miRs). Conclusion LP-miRs, as small-molecule therapeutics, can influence reprogramming/engineering and the conversion of cells into other lineages. These findings have significant implications for our understanding of the mechanisms underlying the regulation of pluripotency and may have potential applications in regenerative medicine.
Collapse
Affiliation(s)
- Navid Ghasemzadeh
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Fatemeh Pourrajab
- Department of Clinical Biochemistry and Molecular Biology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ali Dehghani Firoozabadi
- Yazd Cardiovascular Research Center, Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Maryam Rahnama
- Department of Applied Cell Science, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
4
|
Yu S, Yu S, Liu H, Liao N, Liu X. Enhancing mesenchymal stem cell survival and homing capability to improve cell engraftment efficacy for liver diseases. Stem Cell Res Ther 2023; 14:235. [PMID: 37667383 PMCID: PMC10478247 DOI: 10.1186/s13287-023-03476-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/25/2023] [Indexed: 09/06/2023] Open
Abstract
Although mesenchymal stem cell (MSC) transplantation provides an alternative strategy for end-stage liver disease (ESLD), further widespread application of MSC therapy is limited owing to low cell engraftment efficiency. Improving cell engraftment efficiency plays a critical role in enhancing MSC therapy for liver diseases. In this review, we summarize the current status and challenges of MSC transplantation for ESLD. We also outline the complicated cell-homing process and highlight how low cell engraftment efficiency is closely related to huge differences in extracellular conditions involved in MSC homing journeys ranging from constant, controlled conditions in vitro to variable and challenging conditions in vivo. Improving cell survival and homing capabilities enhances MSC engraftment efficacy. Therefore, we summarize the current strategies, including hypoxic priming, drug pretreatment, gene modification, and cytokine pretreatment, as well as splenectomy and local irradiation, used to improve MSC survival and homing capability, and enhance cell engraftment and therapeutic efficiency of MSC therapy. We hope that this review will provide new insights into enhancing the efficiency of MSC engraftment in liver diseases.
Collapse
Affiliation(s)
- Shaoxiong Yu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, People's Republic of China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, People's Republic of China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, People's Republic of China
| | - Saihua Yu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, People's Republic of China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, People's Republic of China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, People's Republic of China
| | - Haiyan Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, People's Republic of China
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, People's Republic of China
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, People's Republic of China
| | - Naishun Liao
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, People's Republic of China.
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, People's Republic of China.
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, People's Republic of China.
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025, People's Republic of China.
- Mengchao Med-X Center, Fuzhou University, Fuzhou, 350116, People's Republic of China.
- The Liver Center of Fujian Province, Fujian Medical University, Fuzhou, 350025, People's Republic of China.
| |
Collapse
|
5
|
Xu X, Ye T, Wang Y, Pan L, Ye Y, Ding Z, Bao D. MicroRNA-139-5p inhibits inflammatory and oxidative stress responses of Salmonella-infected macrophages through modulating TRAF6. Pathog Dis 2021; 79:6209124. [PMID: 33822016 DOI: 10.1093/femspd/ftab018] [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: 02/17/2021] [Accepted: 03/19/2021] [Indexed: 11/14/2022] Open
Abstract
Evidence indicates that macrophages play an important role in the immune system. Therefore, research involving inflammatory and oxidative stress responses in macrophages is of great significance. Many factors contribute to inflammation and oxidative stress, including Salmonella. We investigated the effect of the miR-139-5p/TRAF6 axis on the inflammatory and oxidative stress responses of Salmonella -infected macrophages. Our findings revealed that miR-139-5p decreased IL-1β and TNF-α levels to inhibit Salmonella-induced inflammatory responses in the RAW264.7 macrophage cell line. Furthermore, miR-139-5p inhibited Salmonella-induced oxidative stress by strengthening SOD, CAT and GSH-PX activity, as well as lowering the malondialdehyde level in the RAW264.7 macrophages cell line. Subsequently, it was verified that TRAF6 was a downstream target of miR-139-5p in RAW264.7 cells. Rescue assays indicated that the over-expression of miR-139-5p inhibits the effects of TRAF6 on inflammatory and oxidative stress responses including Salmonella infection in RAW264.7 cells. To our knowledge, this study is the first to verify that miR-139-5p inhibits inflammatory and oxidative stress responses of Salmonella-infected macrophages through regulating TRAF6. This discovery may offer new insights on inflammatory and oxidative stress responses in macrophages.
Collapse
Affiliation(s)
- Xiaohong Xu
- Department of Laboratory Medicine, Sanmen People's Hospital, 15 Taihe road, Binhai new town, Sanmen county, Taizhou, Zhejiang 317100, China
| | - Tingyun Ye
- Department of Laboratory Medicine, Sanmen People's Hospital, 15 Taihe road, Binhai new town, Sanmen county, Taizhou, Zhejiang 317100, China
| | - Yizhang Wang
- Department of Laboratory Medicine, Sanmen People's Hospital, 15 Taihe road, Binhai new town, Sanmen county, Taizhou, Zhejiang 317100, China
| | - Lianlian Pan
- Department of Laboratory Medicine, Sanmen Hospital of Traditional Chinese Medicine, 287 Xinxing Street, Haiyou Town, Sanmen County, Taizhou, Zhejiang 317100, China
| | - Yali Ye
- Department of Laboratory Medicine, Sanmen People's Hospital, 15 Taihe road, Binhai new town, Sanmen county, Taizhou, Zhejiang 317100, China
| | - Zhengxiang Ding
- Department of Laboratory Medicine, Sanmen People's Hospital, 15 Taihe road, Binhai new town, Sanmen county, Taizhou, Zhejiang 317100, China
| | - Danni Bao
- Department of Laboratory Medicine, Sanmen People's Hospital, 15 Taihe road, Binhai new town, Sanmen county, Taizhou, Zhejiang 317100, China
| |
Collapse
|
6
|
Cai J, Qi H, Yao K, Yao Y, Jing D, Liao W, Zhao Z. Non-Coding RNAs Steering the Senescence-Related Progress, Properties, and Application of Mesenchymal Stem Cells. Front Cell Dev Biol 2021; 9:650431. [PMID: 33816501 PMCID: PMC8017203 DOI: 10.3389/fcell.2021.650431] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 02/12/2021] [Indexed: 02/05/2023] Open
Abstract
The thirst to postpone and even reverse aging progress has never been quenched after all these decades. Unequivocally, mesenchymal stem cells (MSCs), with extraordinary abilities such as self-renewal and multi-directional differentiation, deserve the limelight in this topic. Though having several affable clinical traits, MSCs going through senescence would, on one hand, contribute to age-related diseases and, on the other hand, lead to compromised or even counterproductive therapeutical outcomes. Notably, increasing evidence suggests that non-coding RNAs (ncRNAs) could invigorate various regulatory processes. With even a slight dip or an uptick of expression, ncRNAs would make a dent in or even overturn cellular fate. Thereby, a systematic illustration of ncRNAs identified so far to steer MSCs during senescence is axiomatically an urgent need. In this review, we introduce the general properties and mechanisms of senescence and its relationship with MSCs and illustrate the ncRNAs playing a role in the cellular senescence of MSCs. It is then followed by the elucidation of ncRNAs embodied in extracellular vesicles connecting senescent MSCs with other cells and diversified processes in and beyond the skeletal system. Last, we provide a glimpse into the clinical methodologies of ncRNA-based therapies in MSC-related fields. Hopefully, the intricate relationship between senescence and MSCs will be revealed one day and our work could be a crucial stepping-stone toward that future.
Collapse
Affiliation(s)
- Jingyi Cai
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hexu Qi
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ke Yao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yang Yao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Dian Jing
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wen Liao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, Osaka Dental University, Hirakata, Japan
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Fan H, Hu Z, Wang S, Wu W, Liu X, Geng H. 5-aminolevulinic-acid-mediated sonodynamic therapy improves the prognosis of melanoma by inhibiting survivin expression. Cancer Biomark 2021; 28:301-308. [PMID: 32390599 DOI: 10.3233/cbm-190681] [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] [Indexed: 11/15/2022]
Abstract
BACKGROUND This study aimed to evaluate the relationship between survivin expression and melanoma after 5-aminolevulinic acid (5-ALA)-mediated sonodynamic therapy. METHODS Immunohistochemistry was used to detect survivin protein expression in human melanoma clinical samples. Subsequently, the effects of 5-ALA-mediated sonodynamic therapy were determined by measuring the volume of melanoma xenografts and the bodyweights of melanoma-bearing nude mice. The MTT assay was used to detect the viability of melanoma B16-F10 cells under the action of 5-ALA-mediated sonodynamic therapy, and Western blotting and PCR were used to detect survivin expression in melanoma cells and in the melanoma-xenograft model. RESULTS Survivin expression was significantly upregulated in human melanoma tissues compared with that of non-melanoma tissues. In the in vivo case, 5-ALA-mediated sonodynamic therapy significantly delayed tumor growth, prolonged the survival of mice, and inhibited the expression of survivin. In the in vitro case, 5-ALA-mediated sonodynamic therapy inhibited B16-F10 cell proliferation and decreased survivin expression at both protein and mRNA levels. CONCLUSION Our results suggest that 5-ALA-mediated sonodynamic therapy inhibited B16-F10 cell proliferation and melanoma-xenograft growth and prolonged survival of melanoma-bearing nude mice, which might be through downregulation of survivin expression.
Collapse
Affiliation(s)
- Haixia Fan
- Department of Oral Medicine, Jining Medical College, Shandong, China.,Department of Oral Medicine, Jining Medical College, Shandong, China
| | - Zheng Hu
- Laboratory of Sono- and Photo-theranostic Technologies, Harbin Institute of Technology, Harbin, Heilongjiang, China.,Department of Oral Medicine, Jining Medical College, Shandong, China
| | - Shan Wang
- Department of Oral Pathology, Hospital of Stomatology, The First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Wen Wu
- Department of Oral Medicine, Jining Medical College, Shandong, China
| | - Xue Liu
- Department of Oral Medicine, Jining Medical College, Shandong, China
| | - Haixia Geng
- Department of Oral Medicine, Jining Medical College, Shandong, China
| |
Collapse
|
8
|
Li T, Pang Q, Liu Y, Bai M, Peng Y, Zhang Z. Sulforaphane protects human umbilical vein endothelial cells from oxidative stress via the miR-34a/SIRT1 axis by upregulating nuclear factor erythroid-2-related factor 2. Exp Ther Med 2021; 21:186. [PMID: 33488795 PMCID: PMC7812584 DOI: 10.3892/etm.2021.9617] [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: 10/22/2019] [Accepted: 08/19/2020] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress-induced vascular endothelial cell dysfunction serves an essential role in the initiation and development of atherosclerosis. Sulforaphane (SFN), a naturally occurring antioxidant, has previously demonstrated to exert protective effects on the endothelium against oxidative stress. However, further studies are required to determine its underlying molecular mechanism prior to clinical application. Accumulating evidence suggests that alterations in the microRNA (miRNA/miR)-34a/sirtuin-1 (SIRT1) axis occur with oxidative stress. Therefore, the present study aimed to investigate if SFN exerts a protective role against oxidative stress in vascular endothelial cells through regulation of the miR-34a/SIRT1 axis. Human umbilical vein endothelial cells (HUVECs) were treated with H2O2 in the presence or absence of SFN pretreatment. Cell viability and apoptosis were analyzed using CellTiter-Blue and flow cytometry, respectively. Reverse transcription-quantitative PCR and western blot analyses were performed to determine changes in the expression levels of miR-34a and SIRT1. The expression levels of miR-34a and SIRT1 were artificially regulated following transfection with miR-34a mimic and inhibitor or SIRT1expression plasmid and small interfering RNA, respectively. Subsequently, the effect of the expression changes of miR-34 and SIRT1 on oxidative stress-induced cell injury was investigated. Dual-luciferase reporter assay was used to confirm the targeted binding of miR-34a to SIRT1. SFN was found to ameliorate cellular damage caused by H2O2 and inhibited intracellular reactive oxygen species production. In addition, miR-34a upregulation was accompanied with reduced SIRT1 expression in HUVECs, following H2O2 treatment. miR-34a was revealed to directly target SIRT1 by binding to its 3'-untranslated region. Down-regulation of miR-34a and up-regulation of SIRT1 increased the survival of HUVECs under oxidative stress. Taken together, the results of the present study suggest that SFN may protect HUVECs from oxidative stress by inducing changes in the miR-34a/SIRT1 axis via upregulation of nuclear factor erythroid-2-related factor 2 expression.
Collapse
Affiliation(s)
- Tao Li
- Department of Cardiology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Qi Pang
- Department of Traditional Chinese Medicine, The Gansu Gem Flower Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Yongbin Liu
- Department of Cardiology, The Gansu Gem Flower Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Ming Bai
- Department of Cardiology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Yu Peng
- Department of Cardiology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Zheng Zhang
- Department of Cardiology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
9
|
Nan LP, Wang F, Ran D, Zhou SF, Liu Y, Zhang Z, Huang ZN, Wang ZY, Wang JC, Feng XM, Zhang L. Naringin alleviates H 2O 2-induced apoptosis via the PI3K/Akt pathway in rat nucleus pulposus-derived mesenchymal stem cells. Connect Tissue Res 2020; 61:554-567. [PMID: 31294637 DOI: 10.1080/03008207.2019.1631299] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Purpose: To investigate the protective effect of naringin (Nar) on H2O2-induced apoptosis of nucleus pulposus-derived mesenchymal stem cells (NPMSC) and the potential mechanism in this process. Methods: Rat NPMSC were cultured in MSC culture medium or culture medium with different concentrations of H2O2. Nar or the combination of Nar and LY294002 was added into the culture medium to investigate the effects of Nar. Cell viability was evaluated by cell counting kit-8 (CCK-8) assay. The apoptosis rate was determined using Annexin V/PI dual staining and terminal deoxynucleotide transferase-mediated dUTP nick end labeling (TUNEL) assays. Additionally, the levels of reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) were analyzed by flow cytometry. ATP level in NPMSC was analyzed via ATP detection kit. Mitochondrial ultrastructure change was observed through transmission electron microscope (TEM). Levels of apoptosis-associated molecules (cleaved caspase-3, Bax and Bcl-2) were evaluated via RT-PCR and western blot, respectively. Results: The cells isolated from NP met the criteria for MSC. H2O2 significantly promoted NPMSC apoptosis in a dose and time-dependent manner. Nar showed no cytotoxicity effect on NPMSC up to a concentration of 100 μM for 24 h. Nar exhibited protective effects against H2O2-induced NPMSC apoptosis including apoptosis rate, expressions of proapoptosis and antiapoptosis related genes and protein. Nar could also alleviate H2O2-induced mitochondrial dysfunction of increased mitochondrial ROS production, reduced MMP, decreased intracellular ATP and mitochondrial ultrastructure change. However, these protected effects were inhibited after LY294002 treatment. Conclusions: Our results demonstrated that Nar efficiently attenuated H2O2-induced NPMSC apoptosis and mitochondrial dysfunction. The activation of ROS-mediated PI3K/Akt pathway may be the potential mechanism in this process.
Collapse
Affiliation(s)
- Li-Ping Nan
- Department of Orthopedics, Dalian Medical University , Dalian, Liaoning, China.,Department of Orthopedics, Clinical Medical College of Yangzhou University , Yangzhou, Jiangsu, China
| | - Feng Wang
- Department of Orthopedics, Dalian Medical University , Dalian, Liaoning, China.,Department of Orthopedics, Clinical Medical College of Yangzhou University , Yangzhou, Jiangsu, China
| | - Di Ran
- College of Veterinary Medicine, Yangzhou University , Yangzhou, China
| | - Shi-Feng Zhou
- Department of Orthopedics, Clinical Medical College of Yangzhou University , Yangzhou, Jiangsu, China
| | - Yang Liu
- Department of Orthopedics, Dalian Medical University , Dalian, Liaoning, China.,Department of Orthopedics, Clinical Medical College of Yangzhou University , Yangzhou, Jiangsu, China
| | - Zhen Zhang
- Department of Orthopedics, Dalian Medical University , Dalian, Liaoning, China.,Department of Orthopedics, Clinical Medical College of Yangzhou University , Yangzhou, Jiangsu, China
| | - Ze-Nan Huang
- Department of Orthopedics, Clinical Medical College of Yangzhou University , Yangzhou, Jiangsu, China
| | - Ze-Yu Wang
- Department of Orthopedics, Clinical Medical College of Yangzhou University , Yangzhou, Jiangsu, China
| | - Jing-Cheng Wang
- Department of Orthopedics, Clinical Medical College of Yangzhou University , Yangzhou, Jiangsu, China
| | - Xin-Min Feng
- Department of Orthopedics, Clinical Medical College of Yangzhou University , Yangzhou, Jiangsu, China
| | - Liang Zhang
- Department of Orthopedics, Clinical Medical College of Yangzhou University , Yangzhou, Jiangsu, China
| |
Collapse
|
10
|
Zhang F, Peng W, Zhang J, Wang L, Dong W, Zheng Y, Wang Z, Xie Z, Wang T, Wang C, Yan Y. PARK7 enhances antioxidative-stress processes of BMSCs via the ERK1/2 pathway. J Cell Biochem 2020; 122:222-234. [PMID: 32918333 PMCID: PMC7820948 DOI: 10.1002/jcb.29845] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 08/15/2020] [Accepted: 08/31/2020] [Indexed: 12/15/2022]
Abstract
Oxidative stresss in the microenvironment surrounding lesions induces apoptosis of transplanted bone‐marrow‐derived mesenchymal stem cells (BMSCs). Hence, there is an urgent need for improving antioxidative‐stress processes of transplanted BMSCs to further promote their survival. The present study reports the role and mechanism of Parkinson's disease protein 7 (PARK7) in enhancing antioxidative activity in BMSCs. We used a PARK7 lentivirus to transfect BMSCs to up‐ or downregulate PARK7, and then used H2O2 to simulate oxidative stress in BMSCs in vitro. Overexpression of PARK7 effectively reduced reactive oxygen species and malondialdehyde, protected mitochondrial membrane potential, and resisted oxidative‐stress‐induced apoptosis of BMSCs, but the expression of PARK7 was downregulated, these results were reversed. At the same time, we also found that overexpression of PARK7 increased extracellular‐regulated protein kinase 1/2 (ERK1/2) phosphorylation and nuclear translocation, as well as upregulated Elk1 phosphorylation and superoxide dismutase (SOD) expression. In contrast, when U0126 was used to block the ERK1/2 pathway, ERK1/2 and Elk1 phosphorylation levels were downregulated, ERK1/2 nuclear translocation and SOD content were significantly reduced, and PARK7‐overexperssion‐induced antioxidative activity was completely blocked. Collectively, our results suggest that PARK7 overexpression increased antioxidative‐stress processes and survival of BMSCs subjected to H2O2 via activating the ERK1/2 signaling pathway. Our findings may guide the development of a PARK7‐specific strategy for improving the transplantation efficacy of BMSCs.
Collapse
Affiliation(s)
- Fei Zhang
- Department of Traumatologic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China.,School of clinical medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Wuxun Peng
- Department of Traumatologic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China.,School of clinical medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Jian Zhang
- Department of Traumatologic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China.,School of clinical medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Lei Wang
- Department of Statistics, Guizhou Maternal and Child Health Hospital, Guiyang, Guizhou, China
| | - Wentao Dong
- Department of Traumatologic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China.,School of clinical medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yinggang Zheng
- Department of Orthopedics, The Affiliated Wudang Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhenwen Wang
- Department of Orthopedics, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhihong Xie
- School of clinical medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Tao Wang
- School of clinical medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Chuan Wang
- School of clinical medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yanglin Yan
- School of clinical medicine, Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
11
|
Improved therapeutics of modified mesenchymal stem cells: an update. J Transl Med 2020; 18:42. [PMID: 32000804 PMCID: PMC6993499 DOI: 10.1186/s12967-020-02234-x] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 01/22/2020] [Indexed: 12/15/2022] Open
Abstract
Background Mesenchymal stromal cells (MSCs) have attracted intense interest due to their powerful intrinsic properties of self-regeneration, immunomodulation and multi-potency, as well as being readily available and easy to isolate and culture. Notwithstanding, MSC based therapy suffers reduced efficacy due to several challenges which include unfavorable microenvironmental factors in vitro and in vivo. Body In the quest to circumvent these challenges, several modification techniques have been applied to the naïve MSC to improve its inherent therapeutic properties. These modification approaches can be broadly divided into two groups to include genetic modification and preconditioning modification (using drugs, growth factors and other molecules). This field has witnessed great progress and continues to gather interest and novelty. We review these innovative approaches in not only maintaining, but also enhancing the inherent biological activities and therapeutics of MSCs with respect to migration, homing to target site, adhesion, survival and reduced premature senescence. We discuss the application of the improved modified MSC in some selected human diseases. Possible ways of yet better enhancing the therapeutic outcome and overcoming challenges of MSC modification in the future are also elaborated. Conclusion The importance of prosurvival and promigratory abilities of MSCs in their therapeutic applications can never be overemphasized. These abilities are maintained and even further enhanced via MSC modifications against the inhospitable microenvironment during culture and transplantation. This is a turning point in MSC-based therapy with promising preclinical studies and higher future prospect.
Collapse
|
12
|
Peng Y, Fan JY, Xiong J, Lou Y, Zhu Y. miR-34a Enhances the Susceptibility of Gastric Cancer to Platycodin D by Targeting Survivin. Pathobiology 2019; 86:296-305. [PMID: 31711057 DOI: 10.1159/000502913] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 08/25/2019] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Platycodin D (PD), a triterpenoid saponin isolated from Platycodon grandiflorum, has a well-known anti-tumor effect in multiple human cancers, including gastric cancer (GC). miR-34a plays an important role in the progression of GC. However, the relationship between miR-34a and the susceptibility of GC cells to PD is still unclear. The aim of our research was to investigate the functions of miR-34a in mediating the susceptibility of GC to PD. METHODS qPCR was performed to detect the expression level of miR-34a and survivin in GC cells. The expression of survivin, Bcl-2, Bax, and cleaved caspase-3 was analyzed using Western blot. Cell viability was detected by MTT assay, and apoptosis was analyzed via Annexin V-FITC/PI staining followed by flow cy-tometry. The colony formation and scratch-wound assays were applied to assess cell proliferation and migration. Caspase-3/7 activity was detected by a Caspase-Glo®3/7 detection kit. The relationship between miR-34a and survivin was determined by dual luciferase reporter gene assay. Finally, a GC xenograft mouse model was used to confirm our findings in vivo. RESULTS The expression of miR-34a decreased but survivin increased inversely in human GC cells. Survivin is a direct target of miR-34a and may be negatively regulated by miR-34a. PD could inhibit GC cell proliferation and induce apoptosis. Importantly, overexpression miR-34a or suppressing survivin was shown to enhance the susceptibility of GC to PD both in vitro and in vivo. CONCLUSIONS miR-34a could modulate the susceptibility of GC to PD via targeting survivin, suggesting miR-34a overexpression may serve as a novel strategy to sensitize GC to anti-cancer drugs.
Collapse
Affiliation(s)
- Yao Peng
- Intensive Care Unit, Second Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, China
| | - Jing-Ying Fan
- College of Integrated Chinese and Western Medicine, Hunan University of Traditional Chinese Medicine, Changsha, China
| | - Jian Xiong
- Graduate School of Hunan University of Traditional Chinese Medicine, Changsha, China
| | - Yu Lou
- Graduate School of Hunan University of Traditional Chinese Medicine, Changsha, China
| | - Ying Zhu
- Department of Gastroenterology, First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, China,
| |
Collapse
|
13
|
Fulzele S, Mendhe B, Khayrullin A, Johnson M, Kaiser H, Liu Y, Isales CM, Hamrick MW. Muscle-derived miR-34a increases with age in circulating extracellular vesicles and induces senescence of bone marrow stem cells. Aging (Albany NY) 2019; 11:1791-1803. [PMID: 30910993 PMCID: PMC6461183 DOI: 10.18632/aging.101874] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 03/10/2019] [Indexed: 12/12/2022]
Abstract
Extracellular vesicles (EVs) are known to play important roles in cell-cell communication. Here we investigated the role of muscle-derived EVs and their microRNAs in the loss of bone stem cell populations with age. Aging in male and female C57BL6 mice was associated with a significant increase in expression of the senescence-associated microRNA miR-34a-5p (miR-34a) in skeletal muscle and in serum -derived EVs. Muscle-derived, alpha-sarcoglycan positive, EVs isolated from serum samples also showed a significant increase in miR-34a with age. EVs were isolated from conditioned medium of C2C12 mouse myoblasts and primary human myotubes after cells were treated with hydrogen peroxide to simulate oxidative stress. These EVs were shown to have elevated levels of miR-34a, and these EVs decreased viability of bone marrow mesenchymal (stromal) cells (BMSCs) and increased BMSC senescence. A lentiviral vector system was used to overexpress miR-34a in C2C12 cells, and EVs isolated from these transfected cells were observed to home to bone in vivo and to induce senescence and decrease Sirt1 expression of primary bone marrow cells ex vivo. These findings suggest that aged skeletal muscle is a potential source of circulating, senescence-associated EVs that may directly impact stem cell populations in tissues such as bone via their microRNA cargo.
Collapse
Affiliation(s)
- Sadanand Fulzele
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Bharati Mendhe
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Andrew Khayrullin
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Maribeth Johnson
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Helen Kaiser
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Yutao Liu
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Carlos M. Isales
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Mark W. Hamrick
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
14
|
Horie S, Gonzalez HE, Laffey JG, Masterson CH. Cell therapy in acute respiratory distress syndrome. J Thorac Dis 2018; 10:5607-5620. [PMID: 30416812 DOI: 10.21037/jtd.2018.08.28] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is driven by a severe pro-inflammatory response resulting in lung damage, impaired gas exchange and severe respiratory failure. There is no specific treatment that effectively improves outcome in ARDS. However, in recent years, cell therapy has shown great promise in preclinical ARDS studies. A wide range of cells have been identified as potential candidates for use, among these are mesenchymal stromal cells (MSCs), which are adult multi-lineage cells that can modulate the immune response and enhance repair of damaged tissue. The therapeutic potential of MSC therapy for sepsis and ARDS has been demonstrated in multiple in vivo models. The therapeutic effect of these cells seems to be due to two different mechanisms; direct cellular interaction, and paracrine release of different soluble products such as extracellular vesicles (EVs)/exosomes. Different approaches have also been studied to enhance the therapeutic effect of these cells, such as the over-expression of anti-inflammatory or pro-reparative molecules. Several clinical trials (phase I and II) have already shown safety of MSCs in ARDS and other diseases. However, several translational issues still need to be addressed, such as the large-scale production of cells, and their potentiality and variability, before the therapeutic potential of stem cells therapies can be realized.
Collapse
Affiliation(s)
- Shahd Horie
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland
| | - Hector Esteban Gonzalez
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland
| | - John G Laffey
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland.,Department of Anesthesia and Intensive Care Medicine, Galway University Hospitals, SAOLTA Hospital Group, Ireland
| | - Claire H Masterson
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
15
|
Jiang J, Song Z, Zhang L. miR-155-5p Promotes Progression of Acute Respiratory Distress Syndrome by Inhibiting Differentiation of Bone Marrow Mesenchymal Stem Cells to Alveolar Type II Epithelial Cells. Med Sci Monit 2018; 24:4330-4338. [PMID: 29936517 PMCID: PMC6049011 DOI: 10.12659/msm.910316] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background We investigated whether microRNA-155-5p is involved in the differentiation of bone marrow mesenchymal stem cells (BMSCs) into alveolar type II epithelial (AT II) cells by regulating the Wnt signaling pathway, thus participating in the development of acute respiratory distress syndrome (ARDS). Material/Methods Serum levels of microRNA-155-5p in 50 ARDS patients and 50 normal controls were detected by quantitative real-time PCR (qRT-PCR). Marrow mesenchymal stem cells (MCSs) were isolated from mouse bone marrow and identified by flow cytometry. Subsequently, the effect of microRNA-155-5p on differentiation of BMSCs into AT II cells was evaluated by detecting the expression levels of AT II-specific genes. The expression levels of proteins in the Wnt signaling pathway after overexpression or knockdown of microRNA-155-5p were detected by Western blot. Results Serum levels of microRNA-155-5p in ARDS patients were significantly higher than that in normal controls. Expression levels of AT II-specific genes were enhanced after downregulating microRNA-155-5p in BMSCs. MicroRNA-155-5p overexpression showed the opposite result. Furthermore, microRNA-155-5p inhibited the expression levels of proteins in the Wnt signaling pathway. Conclusions MicroRNA-155-5p can attenuate the differentiation of BMSCs into AT II cells by inhibiting the Wnt signaling pathway, thus promoting the progression of ARDS.
Collapse
Affiliation(s)
- Jing Jiang
- Department of Respiratory Medicine, Yantai Yuhuangding Hospital, Yantai, Shandong, China (mainland)
| | - Zhifang Song
- Department of Pulmonary Disease, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong, China (mainland)
| | - Lanyue Zhang
- Department of Pulmonary Disease, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong, China (mainland)
| |
Collapse
|