1
|
Li M, Tang Y, Zhou C, Geng Y, Zhang C, Hsu Y, Ma L, Guo W, Li M, Wang Y. The Application of Stem Cells and Exosomes in Promoting Nerve Conduits for Peripheral Nerve Repair. Biomater Res 2025; 29:0160. [PMID: 40231207 PMCID: PMC11994886 DOI: 10.34133/bmr.0160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 04/16/2025] Open
Abstract
The repair of peripheral nerve injury (PNI) presents a multifaceted and protracted challenge, with current therapeutic approaches failing to achieve optimal repair outcomes, thereby not satisfying the considerable clinical demand. The advent of tissue engineering has led to a growing body of experimental evidence indicating that the synergistic application of nerve conduits, which provide structural guidance, alongside the biological signals derived from exosomes and stem cells, yields superior therapeutic results for PNI compared to isolated interventions. This combined approach holds great promise for clinical application. In this review, we present the latest advancements in the treatment of PNI through the integration of stem cells or exosomes with nerve conduits. We have addressed the inadequate efficiency of exosomes or stem cells in conjunction with nerve conduits from 3 perspectives: enhancing stem cells or exosomes, improving nerve conduits, and incorporating physical stimulation.
Collapse
Affiliation(s)
- Mengen Li
- National Center for Trauma Medicine, Beijing 100044, China
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education,
Peking University, Beijing 100044, China
- Trauma Medicine Center,
Peking University People’s Hospital, Beijing 100044, China
- Department of Orthopedics and Trauma,
Peking University People’s Hospital, Beijing 100044, China
| | - Ye Tang
- National Center for Trauma Medicine, Beijing 100044, China
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education,
Peking University, Beijing 100044, China
- Trauma Medicine Center,
Peking University People’s Hospital, Beijing 100044, China
- Department of Orthopedics and Trauma,
Peking University People’s Hospital, Beijing 100044, China
| | - Chengkai Zhou
- National Center for Trauma Medicine, Beijing 100044, China
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education,
Peking University, Beijing 100044, China
- Trauma Medicine Center,
Peking University People’s Hospital, Beijing 100044, China
| | - Yan Geng
- National Center for Trauma Medicine, Beijing 100044, China
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education,
Peking University, Beijing 100044, China
- Trauma Medicine Center,
Peking University People’s Hospital, Beijing 100044, China
| | - Chenxi Zhang
- National Center for Trauma Medicine, Beijing 100044, China
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education,
Peking University, Beijing 100044, China
- Trauma Medicine Center,
Peking University People’s Hospital, Beijing 100044, China
| | - Yuwei Hsu
- National Center for Trauma Medicine, Beijing 100044, China
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education,
Peking University, Beijing 100044, China
- Trauma Medicine Center,
Peking University People’s Hospital, Beijing 100044, China
- Emergency Department,
Peking University People’s Hospital, Beijing 100044, China
| | - Le Ma
- National Center for Trauma Medicine, Beijing 100044, China
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education,
Peking University, Beijing 100044, China
- Trauma Medicine Center,
Peking University People’s Hospital, Beijing 100044, China
| | - Wei Guo
- Emergency Department,
Peking University People’s Hospital, Beijing 100044, China
| | - Ming Li
- National Center for Trauma Medicine, Beijing 100044, China
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education,
Peking University, Beijing 100044, China
- Trauma Medicine Center,
Peking University People’s Hospital, Beijing 100044, China
| | - Yanhua Wang
- National Center for Trauma Medicine, Beijing 100044, China
- Key Laboratory of Trauma and Neural Regeneration, Ministry of Education,
Peking University, Beijing 100044, China
- Department of Orthopedics and Trauma,
Peking University People’s Hospital, Beijing 100044, China
| |
Collapse
|
2
|
Wang JJ, Zheng Y, Li YL, Xiao Y, Ren YY, Tian YQ. Emerging role of mesenchymal stem cell-derived exosomes in the repair of acute kidney injury. World J Stem Cells 2025; 17:103360. [PMID: 40160687 PMCID: PMC11947899 DOI: 10.4252/wjsc.v17.i3.103360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/26/2024] [Accepted: 02/13/2025] [Indexed: 03/21/2025] Open
Abstract
Acute kidney injury (AKI) is a clinical syndrome characterized by a rapid deterioration in kidney function and has a significant impact on patient health and survival. Mesenchymal stem cells (MSCs) have the potential to enhance renal function by suppressing the expression of cell cycle inhibitors and reducing the expression of senescence markers and microRNAs via paracrine and endocrine mechanisms. MSC-derived exosomes can alleviate AKI symptoms by regulating DNA damage, apoptosis, and other related signaling pathways through the delivery of proteins, microRNAs, long-chain noncoding RNAs, and circular RNAs. This technique is both safe and effective. MSC-derived exosomes may have great application prospects in the treatment of AKI. Understanding the underlying mechanisms will foster the development of new and promising therapeutic strategies against AKI. This review focused on recent advancements in the role of MSCs in AKI repair as well as the mechanisms underlying the role of MSCs and their secreted exosomes. It is anticipated that novel and profound insights into the functionality of MSCs and their derived exosomes will emerge.
Collapse
Affiliation(s)
- Juan-Juan Wang
- Clinical Laboratory, The First People's Hospital of Yancheng, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng 224000, Jiangsu Province, China
| | - Yu Zheng
- Clinical Laboratory, The First People's Hospital of Yancheng, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng 224000, Jiangsu Province, China
| | - Yan-Lin Li
- Clinical Laboratory, The First People's Hospital of Yancheng, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng 224000, Jiangsu Province, China
| | - Yin Xiao
- Department of Medical Imaging, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou 221000, Jiangsu Province, China
| | - Yang-Yang Ren
- Clinical Laboratory, Xinyi People's Hospital, Xuzhou 221000, Jiangsu Province, China
| | - Yi-Qing Tian
- Clinical Laboratory, Xuzhou Central Hospital, Xuzhou 221000, Jiangsu Province, China.
| |
Collapse
|
3
|
Hao ZW, Zhang ZY, Wang ZP, Wang Y, Chen JY, Chen TH, Shi G, Li HK, Wang JW, Dong MC, Hong L, Li JF. Bioactive peptides and proteins for tissue repair: microenvironment modulation, rational delivery, and clinical potential. Mil Med Res 2024; 11:75. [PMID: 39639374 PMCID: PMC11619216 DOI: 10.1186/s40779-024-00576-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 10/25/2024] [Indexed: 12/07/2024] Open
Abstract
Bioactive peptides and proteins (BAPPs) are promising therapeutic agents for tissue repair with considerable advantages, including multifunctionality, specificity, biocompatibility, and biodegradability. However, the high complexity of tissue microenvironments and their inherent deficiencies such as short half-live and susceptibility to enzymatic degradation, adversely affect their therapeutic efficacy and clinical applications. Investigating the fundamental mechanisms by which BAPPs modulate the microenvironment and developing rational delivery strategies are essential for optimizing their administration in distinct tissue repairs and facilitating clinical translation. This review initially focuses on the mechanisms through which BAPPs influence the microenvironment for tissue repair via reactive oxygen species, blood and lymphatic vessels, immune cells, and repair cells. Then, a variety of delivery platforms, including scaffolds and hydrogels, electrospun fibers, surface coatings, assisted particles, nanotubes, two-dimensional nanomaterials, and nanoparticles engineered cells, are summarized to incorporate BAPPs for effective tissue repair, modification strategies aimed at enhancing loading efficiencies and release kinetics are also reviewed. Additionally, the delivery of BAPPs can be precisely regulated by endogenous stimuli (glucose, reactive oxygen species, enzymes, pH) or exogenous stimuli (ultrasound, heat, light, magnetic field, and electric field) to achieve on-demand release tailored for specific tissue repair needs. Furthermore, this review focuses on the clinical potential of BAPPs in facilitating tissue repair across various types, including bone, cartilage, intervertebral discs, muscle, tendons, periodontal tissues, skin, myocardium, nervous system (encompassing brain, spinal cord, and peripheral nerve), endometrium, as well as ear and ocular tissue. Finally, current challenges and prospects are discussed.
Collapse
Affiliation(s)
- Zhuo-Wen Hao
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zhe-Yuan Zhang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ze-Pu Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ying Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jia-Yao Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Tian-Hong Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Guang Shi
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Han-Ke Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jun-Wu Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Min-Chao Dong
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Li Hong
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Jing-Feng Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
4
|
Cai H, Han XJ, Luo ZR, Wang QL, Lu PP, Mou FF, Zhao ZN, Hu D, Guo HD. Pretreatment with Notoginsenoside R1 enhances the efficacy of neonatal rat mesenchymal stem cell transplantation in model of myocardial infarction through regulating PI3K/Akt/FoxO1 signaling pathways. Stem Cell Res Ther 2024; 15:419. [PMID: 39533348 PMCID: PMC11558819 DOI: 10.1186/s13287-024-04039-x] [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: 04/16/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Although stem cell transplantation is a promising approach for the treatment of myocardial infarction (MI), there are still some problems faced such as the low survival rate of stem cells. Here, we investigated the role of Notoginsenoside R1 (NGR1) pretreatment in improving the effects of neonatal rat bone marrow mesenchymal stem cell (MSC) transplantation for treatment of MI. METHODS Cardiac functions were detected by echocardiography and the myocardial infarct size was determined by Masson's trichrome staining in a rat model of MI. The cardioprotective effects of NGR1/LY294002 co-pretreated MSCs was evaluated to explore the underlying mechanism. The angiogenesis was determined by vWF and α-SMA immunofluorescence staining and cell apoptosis was detected by TUNEL. In vitro, the effects of NGR1 on stem cell proliferation was examined by CCK-8 and levels of P-Akt, P-CREB, P-FoxO1 were detected by western blot. Apoptosis, ROS content, and cytokine levels were examined by DAPI and TUNEL staining, a ROS assay kit, and ELISA, respectively. RESULTS NGR1 elevated the therapeutic effect of MSC transplantation on infarction by preserving cardiac function, increasing angiogenesis and expressions of IGF-1, VEGF, and SDF-1, and reducing cell apoptosis, whereas the addition of LY294002 prior to NGR1 treatment significantly counteracted the foregoing effects of NGR1. NGR1 pretreatment and SC79 pretreatment were similar in that both significantly increased P-Akt and P-FoxO1 levels in MSC and did not affect P-CREB levels. Besides, both NGR1 and SC79 promoted VEGF, SCF and bFGF levels in MSC cultures, and significantly reduced ROS accumulation and the attenuated cell apoptosis in MSC triggered by H2O2. Similarly, addition of LY294002 before NGR1 treatment significantly counteracted the aforementioned effects of NGR1 in vitro. CONCLUSIONS NGR1 pretreatment enhances the effect of MSC transplantation for treatment of MI through paracrine signaling, and the mechanism underlying this effect may be associated with PI3K/Akt/FoxO1 signaling pathways.
Collapse
Affiliation(s)
- Hao Cai
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiao-Jing Han
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhi-Rong Luo
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qiang-Li Wang
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ping-Ping Lu
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Fang-Fang Mou
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhi-Nan Zhao
- Development and Planning Division (Department of Discipline Development), Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Dan Hu
- Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China.
| | - Hai-Dong Guo
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
5
|
Garmany A, Arrell DK, Yamada S, Jeon R, Behfar A, Park S, Terzic A. Decoded cardiopoietic cell secretome linkage to heart repair biosignature. Stem Cells Transl Med 2024; 13:1144-1159. [PMID: 39259666 PMCID: PMC11555478 DOI: 10.1093/stcltm/szae067] [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: 04/09/2024] [Accepted: 08/03/2024] [Indexed: 09/13/2024] Open
Abstract
Cardiopoiesis-primed human stem cells exert sustained benefit in treating heart failure despite limited retention following myocardial delivery. To assess potential paracrine contribution, the secretome of cardiopoiesis conditioned versus naïve human mesenchymal stromal cells was decoded by directed proteomics augmented with machine learning and systems interrogation. Cardiopoiesis doubled cellular protein output generating a distinct secretome that segregated the conditioned state. Altering the expression of 1035 secreted proteins, cardiopoiesis reshaped the secretome across functional classes. The resolved differential cardiopoietic secretome was enriched in mesoderm development and cardiac progenitor signaling processes, yielding a cardiovasculogenic profile bolstered by upregulated cardiogenic proteins. In tandem, cardiopoiesis enhanced the secretion of immunomodulatory proteins associated with cytokine signaling, leukocyte migration, and chemotaxis. Network analysis integrated the differential secretome within an interactome of 1745 molecules featuring prioritized regenerative processes. Secretome contribution to the repair signature of cardiopoietic cell-treated infarcted hearts was assessed in a murine coronary ligation model. Intramyocardial delivery of cardiopoietic cells improved the performance of failing hearts, with undirected proteomics revealing 50 myocardial proteins responsive to cell therapy. Pathway analysis linked the secretome to cardiac proteome remodeling, pinpointing 17 cardiopoiesis-upregulated secretome proteins directly upstream of 44% of the cell therapy-responsive cardiac proteome. Knockout, in silico, of this 22-protein secretome-dependent myocardial ensemble eliminated indices of the repair signature. Accordingly, in vivo, cell therapy rendered the secretome-dependent myocardial proteome of an infarcted heart indiscernible from healthy counterparts. Thus, the secretagogue effect of cardiopoiesis transforms the human stem cell secretome, endows regenerative competency, and upregulates candidate paracrine effectors of cell therapy-mediated molecular restitution.
Collapse
Affiliation(s)
- Armin Garmany
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Alix School of Medicine, Regenerative Sciences Track, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - D Kent Arrell
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
| | - Satsuki Yamada
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
- Section of Geriatric Medicine & Gerontology, Department of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Ryounghoon Jeon
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
| | - Atta Behfar
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic, Rochester, MN, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Sungjo Park
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
| | - Andre Terzic
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
- Department of Medical Genetics, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
6
|
Sun S, Wang L, Tang Q, Yi J, Yu X, Cao Y, Jiang L, Liu J. Myocardial infarction in rats was alleviated by MSCs derived from the maternal segment of the human umbilical cord. Front Cell Dev Biol 2024; 12:1469541. [PMID: 39479514 PMCID: PMC11521943 DOI: 10.3389/fcell.2024.1469541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/25/2024] [Indexed: 11/02/2024] Open
Abstract
Background Mesenchymal stem cells (MSCs) are safe and effective in treating myocardial infarction (MI) and have broad application prospects. However, the heterogeneity of MSCs may affect their therapeutic effect on the disease. We recently found that MSCs derived from different segments of the same umbilical cord (UC) showed significant difference in the expression of genes that are related to heart development and injury repair. We therefore hypothesized that those MSCs with high expression of above genes are more effective to treat MI and tested it in this study. Methods MSCs were isolated from 3 cm-long segments of the maternal, middle and fetal segments of the UC (maternal-MSCs, middle-MSCs and fetal-MSCs, respectively). RNA-seq was used to analyze and compare the transcriptomes. We verified the effects of MSCs on oxygen-glucose deprivation (OGD)-induced cardiomyocyte apoptosis in vitro. In vivo, a rat MI model was established by ligating the left anterior descending coronary artery, and MSCs were injected into the myocardium surrounding the MI site. The therapeutic effects of MSCs derived from different segments of the UC were evaluated by examining cardiac function, histopathology, cardiomyocyte apoptosis, and angiogenesis. Results Compared to fetal-MSCs and middle-MSCs, maternal-MSCs exhibited significantly higher expression of genes that are associated with heart development, such as GATA-binding protein 4 (GATA4), and myocardin (MYOCD). Coculture with maternal-MSCs reduced OGD-induced cardiomyocyte apoptosis. In rats with MI, maternal-MSCs significantly restored cardiac contractile function and reduced the infarct size. Mechanistic experiments revealed that maternal-MSCs exerted cardioprotective effects by decreasing cardiomyocyte apoptosis, and promoting angiogenesis. Conclusion Our data demonstrated that maternal segment-derived MSCs were a superior cell source for regenerative repair after MI. Segmental localization of the entire UC when isolating hUCMSCs was necessary to improve the effectiveness of clinical applications.
Collapse
Affiliation(s)
- Shuifen Sun
- Regenerative Medicine Research Center, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China, The First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Cell Therapy Engineering Research Center for Cardiovascular Diseases in Yunnan Province, Kunming, Yunnan, China
- Key Laboratory of Innovative Application for Traditional Chinese Medicine in Yunnan Province, Kunming, Yunnan, China
| | - Linping Wang
- Regenerative Medicine Research Center, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China, The First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Cell Therapy Engineering Research Center for Cardiovascular Diseases in Yunnan Province, Kunming, Yunnan, China
- Key Laboratory of Innovative Application for Traditional Chinese Medicine in Yunnan Province, Kunming, Yunnan, China
| | - Qisheng Tang
- Regenerative Medicine Research Center, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China, The First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Cell Therapy Engineering Research Center for Cardiovascular Diseases in Yunnan Province, Kunming, Yunnan, China
- Key Laboratory of Innovative Application for Traditional Chinese Medicine in Yunnan Province, Kunming, Yunnan, China
| | - Jialian Yi
- Regenerative Medicine Research Center, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China, The First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Cell Therapy Engineering Research Center for Cardiovascular Diseases in Yunnan Province, Kunming, Yunnan, China
- Key Laboratory of Innovative Application for Traditional Chinese Medicine in Yunnan Province, Kunming, Yunnan, China
| | - Xin Yu
- Medicine School, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yu Cao
- Cell Therapy Engineering Research Center for Cardiovascular Diseases in Yunnan Province, Kunming, Yunnan, China
- Key Laboratory of Innovative Application for Traditional Chinese Medicine in Yunnan Province, Kunming, Yunnan, China
- Department of Cardiovascular Surgery, The First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Lihong Jiang
- Regenerative Medicine Research Center, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China, The First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Cell Therapy Engineering Research Center for Cardiovascular Diseases in Yunnan Province, Kunming, Yunnan, China
- Key Laboratory of Innovative Application for Traditional Chinese Medicine in Yunnan Province, Kunming, Yunnan, China
- Department of Cardiovascular Surgery, The First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jie Liu
- Regenerative Medicine Research Center, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China, The First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Cell Therapy Engineering Research Center for Cardiovascular Diseases in Yunnan Province, Kunming, Yunnan, China
- Key Laboratory of Innovative Application for Traditional Chinese Medicine in Yunnan Province, Kunming, Yunnan, China
| |
Collapse
|
7
|
Lam J, Yu J, Lee B, Campagna C, Yoo S, Baek K, Jeon NL, Sung KE. Characterizing On-Chip Angiogenesis Induction in a Microphysiological System as a Functional Measure of Mesenchymal Stromal Cell Bioactivity. Adv Biol (Weinh) 2024; 8:e2300094. [PMID: 37409400 DOI: 10.1002/adbi.202300094] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 06/12/2023] [Indexed: 07/07/2023]
Abstract
Mesenchymal stromal cells (MSCs) continue to be proposed for clinical investigation to treat myriad diseases given their purported potential to stimulate endogenous regenerative processes, such as angiogenesis. However, MSC functional heterogeneity has hindered clinical success and still poses a substantial manufacturing challenge from a product quality control perspective. Here, a quantitative bioassay based on an enhanced-throughput is described, microphysiological system (MPS) to measure the specific bioactivity of MSCs to stimulate angiogenesis as a potential measure of MSC potency. Using this novel bioassay, MSCs derived from multiple donors at different passages are co-cultured with human umbilical vein endothelial cells and exhibit significant heterogeneity in angiogenic potency between donors and cell passage. Depending on donor source and cellular passage number, MSCs varied in their ability to stimulate tip cell dominant or stalk cell dominant phenotypes in angiogenic sprout morphology which correlated with expression levels of hepatocyte growth factor (HGF). These findings suggest that MSC angiogenic bioactivity may be considered as a possible potency attribute in MSC quality control strategies. Development of a reliable and functionally relevant potency assay for measuring clinically relevant potency attributes of MSCs will help to improve consistency in quality and thereby, accelerate clinical development of these cell-based products.
Collapse
Affiliation(s)
- Johnny Lam
- Office of Therapeutic Product, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| | - James Yu
- Office of Therapeutic Product, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
- Department of Mechanical Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Byungjun Lee
- Qureator, Inc., 7094 Miratech Drive, Suite 110, San Diego, CA, 92121, USA
| | - Courtney Campagna
- Office of Therapeutic Product, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| | - Sanghee Yoo
- Qureator, Inc., 7094 Miratech Drive, Suite 110, San Diego, CA, 92121, USA
| | - Kyusuk Baek
- Qureator, Inc., 7094 Miratech Drive, Suite 110, San Diego, CA, 92121, USA
| | - Noo Li Jeon
- Department of Mechanical Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
- Institute of Advanced Machines and Design, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Kyung E Sung
- Office of Therapeutic Product, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| |
Collapse
|
8
|
Annicchiarico A, Barile B, Buccoliero C, Nicchia GP, Brunetti G. Alternative therapeutic strategies in diabetes management. World J Diabetes 2024; 15:1142-1161. [PMID: 38983831 PMCID: PMC11229975 DOI: 10.4239/wjd.v15.i6.1142] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/17/2024] [Accepted: 04/12/2024] [Indexed: 06/11/2024] Open
Abstract
Diabetes is a heterogeneous metabolic disease characterized by elevated blood glucose levels resulting from the destruction or malfunction of pancreatic β cells, insulin resistance in peripheral tissues, or both, and results in a non-sufficient production of insulin. To adjust blood glucose levels, diabetic patients need exogenous insulin administration together with medical nutrition therapy and physical activity. With the aim of improving insulin availability in diabetic patients as well as ameliorating diabetes comorbidities, different strategies have been investigated. The first approaches included enhancing endogenous β cell activity or transplanting new islets. The protocol for this kind of intervention has recently been optimized, leading to standardized procedures. It is indicated for diabetic patients with severe hypoglycemia, complicated by impaired hypoglycemia awareness or exacerbated glycemic lability. Transplantation has been associated with improvement in all comorbidities associated with diabetes, quality of life, and survival. However, different trials are ongoing to further improve the beneficial effects of transplantation. Furthermore, to overcome some limitations associated with the availability of islets/pancreas, alternative therapeutic strategies are under evaluation, such as the use of mesenchymal stem cells (MSCs) or induced pluripotent stem cells for transplantation. The cotransplantation of MSCs with islets has been successful, thus providing protection against proinflammatory cytokines and hypoxia through different mechanisms, including exosome release. The use of induced pluripotent stem cells is recent and requires further investigation. The advantages of MSC implantation have also included the improvement of diabetes-related comorbidities, such as wound healing. Despite the number of advantages of the direct injection of MSCs, new strategies involving biomaterials and scaffolds have been developed to improve the efficacy of mesenchymal cell delivery with promising results. In conclusion, this paper offered an overview of new alternative strategies for diabetes management while highlighting some limitations that will need to be overcome by future approaches.
Collapse
Affiliation(s)
- Alessia Annicchiarico
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Bari 70125, Italy
| | - Barbara Barile
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Bari 70125, Italy
| | - Cinzia Buccoliero
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Bari 70125, Italy
| | - Grazia Paola Nicchia
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Bari 70125, Italy
| | - Giacomina Brunetti
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Bari 70125, Italy
| |
Collapse
|
9
|
Rajendran AK, Anthraper MSJ, Hwang NS, Rangasamy J. Osteogenesis and angiogenesis promoting bioactive ceramics. MATERIALS SCIENCE AND ENGINEERING: R: REPORTS 2024; 159:100801. [DOI: 10.1016/j.mser.2024.100801] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
10
|
Benjamin ESB, Vinod E, Illangeswaran RSS, Rajamani BM, Vidhyadharan RT, Bagchi A, Maity A, Mohan A, Parasuraman G, Amirtham SM, Abraham A, Velayudhan SR, Balasubramanian P. Immortalised chronic myeloid leukemia (CML) derived mesenchymal stromal cells (MSCs) line retains the immunomodulatory and chemoprotective properties of CML patient-derived MSCs. Cell Signal 2024; 116:111067. [PMID: 38281615 DOI: 10.1016/j.cellsig.2024.111067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 01/30/2024]
Abstract
Despite the success of Tyrosine kinase inhibitors (TKIs) in treating chronic myeloid leukemia (CML), leukemic stem cells (LSCs) persist, contributing to relapse and resistance. CML Mesenchymal Stromal Cells (MSCs) help in LSC maintenance and protection from TKIs. However, the limited passage and self-differentiation abilities of primary CML MSCs hinder extensive research. To overcome this, we generated and characterized an immortalised CML patient-derived MSC (iCML MSC) line and assessed its role in LSC maintenance. We also compared the immunophenotype and differentiation potential between primary CML MSCs at diagnosis, post-treatment, and with normal bone marrow MSCs. Notably, CML MSCs exhibited enhanced chondrogenic differentiation potential compared to normal MSCs. The iCML MSC line retained the trilineage differentiation potential and was genetically stable, enabling long-term investigations. Functional studies demonstrated that iCML MSCs protected CML CD34+ cells from imatinib-induced apoptosis, recapitulating the bone marrow microenvironment-mediated resistance observed in patients. iCML MSC-conditioned media enabled CML CD34+ and AML blast cells to proliferate rapidly, with no impact on healthy donor CD34+ cells. Gene expression profiling revealed dysregulated genes associated with calcium metabolism in CML CD34+ cells cocultured with iCML MSCs, providing insights into potential therapeutic targets. Further, cytokine profiling revealed that the primary CML MSC lines abundantly secreted 25 cytokines involved in immune regulation, supporting the hypothesis that CML MSCs create an immune modulatory microenvironment that promotes growth and protects against TKIs. Our study establishes the utility of iCML MSCs as a valuable model to investigate leukemic-stromal interactions and study candidate genes involved in mediating TKI resistance in CML LSCs.
Collapse
Affiliation(s)
- Esther Sathya Bama Benjamin
- Department of Haematology, Christian Medical College, Ranipet campus, India; Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, India
| | - Elizabeth Vinod
- Department of Physiology, Christain Medical College, Vellore, India; Centre for Stem Cell Research (A Unit of inStem, Bengaluru), CMC Campus, Vellore, India
| | | | | | | | - Abhirup Bagchi
- Centre for Stem Cell Research (A Unit of inStem, Bengaluru), CMC Campus, Vellore, India
| | - Arnab Maity
- Department of Haematology, Christian Medical College, Ranipet campus, India
| | - Ajith Mohan
- Department of Haematology, Christian Medical College, Ranipet campus, India
| | | | | | - Aby Abraham
- Department of Haematology, Christian Medical College, Ranipet campus, India
| | - Shaji R Velayudhan
- Department of Haematology, Christian Medical College, Ranipet campus, India; Centre for Stem Cell Research (A Unit of inStem, Bengaluru), CMC Campus, Vellore, India
| | | |
Collapse
|
11
|
Zhu D, Hu Y, Kong X, Luo Y, Zhang Y, Wu Y, Tan J, Chen J, Xu T, Zhu L. Enhanced burn wound healing by controlled-release 3D ADMSC-derived exosome-loaded hyaluronan hydrogel. Regen Biomater 2024; 11:rbae035. [PMID: 38628545 PMCID: PMC11018541 DOI: 10.1093/rb/rbae035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 02/23/2024] [Accepted: 03/17/2024] [Indexed: 04/19/2024] Open
Abstract
Adipose mesenchymal stem cell (ADMSC)-derived exosomes (ADMSC-Exos) have shown great potential in regenerative medicine and been evidenced benefiting wound repair such as burns. However, the low yield, easy loss after direct coating, and no suitable loading system to improve their availability and efficacy hinder their clinical application for wound healing. And few studies focused on the comparison of biological functions between exosomes derived from different culture techniques, especially in exosome-releasing hydrogel system. Therefore, we designed a high-performance exosome controllable releasing hydrogel system for burn wound healing, namely loading 3D-printed microfiber culture-derived exosomes in a highly biocompatible hyaluronic acid (HA). In this project, we compared the biological functions in vitro and in a burn model among exosomes derived from the conventional two-dimensional (2D) plate culture (2D-Exos), microcarrier culture (2.5D-Exos), and 3D-printed microfiber culture (3D-Exos). Results showed that compared with 2D-Exos and 2.5D-Exos, 3D-Exos promoted HACATs and HUVECs cell proliferation and migration more significantly. Additionally, 3D-Exos had stronger angiogenesis-promoting effects in tube formation of (HUVECs) cells. Moreover, we found HA-loaded 3D-Exos showed better burn wound healing promotion compared to 2D-Exos and 2.5D-Exos, including accelerated burn wound healing rate and better collagen remodeling. The study findings reveal that the HA-loaded, controllable-release 3D-Exos repair system distinctly augments therapeutic efficacy in terms of wound healing, while concurrently introducing a facile application approach. This system markedly bolsters the exosomal loading efficiency, provides a robust protective milieu, and potentiates the inherent biological functionalities of the exosomes. Our findings provide a rationale for more efficient utilization of high-quality and high-yield 3D exosomes in the future, and a novel strategy for healing severe burns.
Collapse
Affiliation(s)
- Delong Zhu
- Department of Dermatology & Plastic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Ying Hu
- Department of Dermatology & Plastic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Xiangkai Kong
- Department of Dermatology & Plastic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Yuansen Luo
- Department of the Second Plastic Surgery, The First People’s Hospital of Foshan, Foshan 528000, China
| | - Yi Zhang
- Department of Research and Development, Huaqing Zhimei (Shenzhen) Biotechnology Co., Ltd, Shenzhen 518107, People’s Republic of China
| | - Yu Wu
- Department of Dermatology & Plastic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Jiameng Tan
- Department of Dermatology & Plastic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| | - Jianwei Chen
- Center for Bio-Intelligent Manufacturing and Living Matter Bioprinting, Research Institute of Tsinghua University in Shenzhen, Tsinghua University, Shenzhen 518057, People’s Republic of China
| | - Tao Xu
- Center for Bio-Intelligent Manufacturing and Living Matter Bioprinting, Research Institute of Tsinghua University in Shenzhen, Tsinghua University, Shenzhen 518057, People’s Republic of China
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, People’s Republic of China
| | - Lei Zhu
- Department of Dermatology & Plastic Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| |
Collapse
|
12
|
Maličev E, Jazbec K. An Overview of Mesenchymal Stem Cell Heterogeneity and Concentration. Pharmaceuticals (Basel) 2024; 17:350. [PMID: 38543135 PMCID: PMC10975472 DOI: 10.3390/ph17030350] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/22/2024] [Accepted: 03/05/2024] [Indexed: 01/06/2025] Open
Abstract
Mesenchymal stem cells (MSCs) are of great interest in cell therapies due to the immunomodulatory and other effects they have after autologous or allogeneic transplantation. In most clinical applications, a high number of MSCs is required; therefore, the isolated MSC population must be expanded in the cell culture until the desired number is reached. Analysing freshly isolated MSCs is challenging due to their rareness and heterogeneity, which is noticeable among donors, tissues, and cell subpopulations. Although the phenotype of MSCs in tissue can differ from those of cultured cells, phenotyping and counting are usually performed only after MSC proliferation. As MSC applicability is a developing and growing field, there is a need to implement phenotyping and counting methods for freshly isolated MSCs, especially in new one-step procedures where isolated cells are implanted immediately without cell culturing. Only by analysing harvested cells can we correctly evaluate such studies. This review describes multilevel heterogeneity and concentrations of MSCs and different strategies for phenotype determination and enumeration of freshly isolated MSCs.
Collapse
Affiliation(s)
- Elvira Maličev
- Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, 1000 Ljubljana, Slovenia;
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva ulica 101, 1000 Ljubljana, Slovenia
| | - Katerina Jazbec
- Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, 1000 Ljubljana, Slovenia;
| |
Collapse
|
13
|
Kulovic-Sissawo A, Tocantins C, Diniz MS, Weiss E, Steiner A, Tokic S, Madreiter-Sokolowski CT, Pereira SP, Hiden U. Mitochondrial Dysfunction in Endothelial Progenitor Cells: Unraveling Insights from Vascular Endothelial Cells. BIOLOGY 2024; 13:70. [PMID: 38392289 PMCID: PMC10886154 DOI: 10.3390/biology13020070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 02/24/2024]
Abstract
Endothelial dysfunction is associated with several lifestyle-related diseases, including cardiovascular and neurodegenerative diseases, and it contributes significantly to the global health burden. Recent research indicates a link between cardiovascular risk factors (CVRFs), excessive production of reactive oxygen species (ROS), mitochondrial impairment, and endothelial dysfunction. Circulating endothelial progenitor cells (EPCs) are recruited into the vessel wall to maintain appropriate endothelial function, repair, and angiogenesis. After attachment, EPCs differentiate into mature endothelial cells (ECs). Like ECs, EPCs are also susceptible to CVRFs, including metabolic dysfunction and chronic inflammation. Therefore, mitochondrial dysfunction of EPCs may have long-term effects on the function of the mature ECs into which EPCs differentiate, particularly in the presence of endothelial damage. However, a link between CVRFs and impaired mitochondrial function in EPCs has hardly been investigated. In this review, we aim to consolidate existing knowledge on the development of mitochondrial and endothelial dysfunction in the vascular endothelium, place it in the context of recent studies investigating the consequences of CVRFs on EPCs, and discuss the role of mitochondrial dysfunction. Thus, we aim to gain a comprehensive understanding of mechanisms involved in EPC deterioration in relation to CVRFs and address potential therapeutic interventions targeting mitochondrial health to promote endothelial function.
Collapse
Affiliation(s)
- Azra Kulovic-Sissawo
- Perinatal Research Laboratory, Department of Obstetrics and Gynaecology, Medical University of Graz, Auenbruggerplatz 14, 8036 Graz, Austria
- Research Unit Early Life Determinants (ELiD), Medical University of Graz, Auenbruggerplatz 14, 8036 Graz, Austria
| | - Carolina Tocantins
- Perinatal Research Laboratory, Department of Obstetrics and Gynaecology, Medical University of Graz, Auenbruggerplatz 14, 8036 Graz, Austria
- Research Unit Early Life Determinants (ELiD), Medical University of Graz, Auenbruggerplatz 14, 8036 Graz, Austria
- CNC-UC-Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-504 Coimbra, Portugal
- Doctoral Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3004-531 Coimbra, Portugal
| | - Mariana S Diniz
- Perinatal Research Laboratory, Department of Obstetrics and Gynaecology, Medical University of Graz, Auenbruggerplatz 14, 8036 Graz, Austria
- Research Unit Early Life Determinants (ELiD), Medical University of Graz, Auenbruggerplatz 14, 8036 Graz, Austria
- CNC-UC-Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-504 Coimbra, Portugal
- Doctoral Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3004-531 Coimbra, Portugal
| | - Elisa Weiss
- Perinatal Research Laboratory, Department of Obstetrics and Gynaecology, Medical University of Graz, Auenbruggerplatz 14, 8036 Graz, Austria
- Research Unit Early Life Determinants (ELiD), Medical University of Graz, Auenbruggerplatz 14, 8036 Graz, Austria
| | - Andreas Steiner
- Perinatal Research Laboratory, Department of Obstetrics and Gynaecology, Medical University of Graz, Auenbruggerplatz 14, 8036 Graz, Austria
- Research Unit Early Life Determinants (ELiD), Medical University of Graz, Auenbruggerplatz 14, 8036 Graz, Austria
| | - Silvija Tokic
- Research Unit of Analytical Mass Spectrometry, Cell Biology and Biochemistry of Inborn Errors of Metabolism, Department of Paediatrics and Adolescent Medicine, Medical University of Graz, Auenbruggerplatz 34, 8036 Graz, Austria
| | - Corina T Madreiter-Sokolowski
- Division of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria
| | - Susana P Pereira
- CNC-UC-Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, 3004-504 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-504 Coimbra, Portugal
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, 4200-450 Porto, Portugal
| | - Ursula Hiden
- Perinatal Research Laboratory, Department of Obstetrics and Gynaecology, Medical University of Graz, Auenbruggerplatz 14, 8036 Graz, Austria
- Research Unit Early Life Determinants (ELiD), Medical University of Graz, Auenbruggerplatz 14, 8036 Graz, Austria
| |
Collapse
|
14
|
Borzone FR, Giorello MB, Sanmartin MC, Yannarelli G, Martinez LM, Chasseing NA. Mesenchymal stem cells and cancer-associated fibroblasts as a therapeutic strategy for breast cancer. Br J Pharmacol 2024; 181:238-256. [PMID: 35485850 DOI: 10.1111/bph.15861] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/21/2022] [Accepted: 04/22/2022] [Indexed: 11/26/2022] Open
Abstract
Breast cancer is the most common type of cancer and the leading cause of death among women. Recent evidence suggests that mesenchymal stromal/stem cells and cancer-associated fibroblasts (CAFs) have an essential role in cancer progression, invasion and therapy resistance. Therefore, they are considered as highly promising future therapeutic targets against breast cancer. The intrinsic tumour tropism and immunomodulatory capacities of mesenchymal stromal/stem cells are of special relevance for developing mesenchymal stromal/stem cells-based anti-tumour therapies that suppress primary tumour growth and metastasis. In addition, the utilization of therapies that target the stromal components of the tumour microenvironment in combination with standard drugs is an innovative tool that could improve patients' response to therapies and their survival. In this review, we discuss the currently available information regarding the possible use of mesenchymal stromal/stem cells-derived anti-tumour therapies, as well as the utilization of therapies that target CAFs in breast cancer microenvironment. Finally, these data can serve as a guide map for future research in this field, ultimately aiding the effective transition of these results into the clinic. LINKED ARTICLES: This article is part of a themed issue on Cancer Microenvironment and Pharmacological Interventions. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.2/issuetoc.
Collapse
Affiliation(s)
- Francisco Raúl Borzone
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María Belén Giorello
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María Cecilia Sanmartin
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Laboratorio de Regulación Génica y Células Madre, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMeTTyB), Universidad Favaloro-CONICET, Buenos Aires, Argentina
| | - Gustavo Yannarelli
- Laboratorio de Regulación Génica y Células Madre, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMeTTyB), Universidad Favaloro-CONICET, Buenos Aires, Argentina
| | - Leandro Marcelo Martinez
- Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Norma Alejandra Chasseing
- Laboratorio de Inmunohematología, Instituto de Biología y Medicina Experimental (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
15
|
Skovronova R, Scaccia E, Calcat-I-Cervera S, Bussolati B, O'Brien T, Bieback K. Adipose stromal cells bioproducts as cell-free therapies: manufacturing and therapeutic dose determine in vitro functionality. J Transl Med 2023; 21:723. [PMID: 37840135 PMCID: PMC10577984 DOI: 10.1186/s12967-023-04602-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/06/2023] [Indexed: 10/17/2023] Open
Abstract
BACKGROUND Extracellular vesicles (EV) are considered a cell-free alternative to mesenchymal stromal cell (MSC) therapy. Numerous reports describe the efficacy of EV in conferring immunomodulation and promoting angiogenesis, yet others report these activities to be conveyed in EV-free bioproducts. We hypothesized that this discrepancy may depend either on the method of isolation or rather the relative impact of the individual bioactive components within the MSC secretome. METHODS To answer this question, we performed an inter-laboratory study evaluating EV generated from adipose stromal cells (ASC) by either sequential ultracentrifugation (UC) or size-exclusion chromatography (SEC). The effect of both EV preparations on immunomodulation and angiogenesis in vitro was compared to that of the whole secretome and of the EV-free protein fraction after SEC isolation. RESULTS In the current study, neither the EV preparations, the secretome or the protein fraction were efficacious in inhibiting mitogen-driven T cell proliferation. However, EV generated by SEC stimulated macrophage phagocytic activity to a similar extent as the secretome. In turn, tube formation and wound healing were strongly promoted by the ASC secretome and protein fraction, but not by EV. Within the secretome/protein fraction, VEGF was identified as a potential driver of angiogenesis, and was absent in both EV preparations. CONCLUSIONS Our data indicate that the effects of ASC on immunomodulation and angiogenesis are EV-independent. Specific ASC-EV effects need to be dissected for their use as cell-free therapeutics.
Collapse
Affiliation(s)
- Renata Skovronova
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Eleonora Scaccia
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service, Baden-Württemberg-Hessen, Friedrich-Ebert-Str.107, 68167, Mannheim, Germany
| | - Sandra Calcat-I-Cervera
- College of Medicine, Nursing and Health Science, School of Medicine, Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Timothy O'Brien
- College of Medicine, Nursing and Health Science, School of Medicine, Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service, Baden-Württemberg-Hessen, Friedrich-Ebert-Str.107, 68167, Mannheim, Germany.
- Mannheim Institute of Innate Immunoscience, Medical Faculty of Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
16
|
Mahjoor M, Fakouri A, Farokhi S, Nazari H, Afkhami H, Heidari F. Regenerative potential of mesenchymal stromal cells in wound healing: unveiling the influence of normoxic and hypoxic environments. Front Cell Dev Biol 2023; 11:1245872. [PMID: 37900276 PMCID: PMC10603205 DOI: 10.3389/fcell.2023.1245872] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 08/11/2023] [Indexed: 10/31/2023] Open
Abstract
The innate and adaptive immune systems rely on the skin for various purposes, serving as the primary defense against harmful environmental elements. However, skin lesions may lead to undesirable consequences such as scarring, accelerated skin aging, functional impairment, and psychological effects over time. The rising popularity of mesenchymal stromal cells (MSCs) for skin wound treatment is due to their potential as a promising therapeutic option. MSCs offer advantages in terms of differentiation capacity, accessibility, low immunogenicity, and their central role in natural wound-healing processes. To accelerate the healing process, MSCs promote cell migration, angiogenesis, epithelialization, and granulation tissue development. Oxygen plays a critical role in the formation and expansion of mammalian cells. The term "normoxia" refers to the usual oxygen levels, defined at 20.21 percent oxygen (160 mm of mercury), while "hypoxia" denotes oxygen levels of 2.91 percent or less. Notably, the ambient O2 content (20%) in the lab significantly differs from the 2%-9% O2 concentration in their natural habitat. Oxygen regulation of hypoxia-inducible factor-1 (HIF-1) mediated expression of multiple genes plays a crucial role in sustaining stem cell destiny concerning proliferation and differentiation. This study aims to elucidate the impact of normoxia and hypoxia on MSC biology and draw comparisons between the two. The findings suggest that expanding MSC-based regenerative treatments in a hypoxic environment can enhance their growth kinetics, genetic stability, and expression of chemokine receptors, ultimately increasing their effectiveness.
Collapse
Affiliation(s)
- Mohamad Mahjoor
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Arshia Fakouri
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Simin Farokhi
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Hojjatollah Nazari
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW, Australia
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Fatemeh Heidari
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Anatomy, Faculty of Medicine, Qom University of Medical Sciences, Qom, Iran
| |
Collapse
|
17
|
Sionov RV, Ahdut-HaCohen R. A Supportive Role of Mesenchymal Stem Cells on Insulin-Producing Langerhans Islets with a Specific Emphasis on The Secretome. Biomedicines 2023; 11:2558. [PMID: 37761001 PMCID: PMC10527322 DOI: 10.3390/biomedicines11092558] [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: 08/15/2023] [Revised: 09/06/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Type 1 Diabetes (T1D) is a chronic autoimmune disease characterized by a gradual destruction of insulin-producing β-cells in the endocrine pancreas due to innate and specific immune responses, leading to impaired glucose homeostasis. T1D patients usually require regular insulin injections after meals to maintain normal serum glucose levels. In severe cases, pancreas or Langerhans islet transplantation can assist in reaching a sufficient β-mass to normalize glucose homeostasis. The latter procedure is limited because of low donor availability, high islet loss, and immune rejection. There is still a need to develop new technologies to improve islet survival and implantation and to keep the islets functional. Mesenchymal stem cells (MSCs) are multipotent non-hematopoietic progenitor cells with high plasticity that can support human pancreatic islet function both in vitro and in vivo and islet co-transplantation with MSCs is more effective than islet transplantation alone in attenuating diabetes progression. The beneficial effect of MSCs on islet function is due to a combined effect on angiogenesis, suppression of immune responses, and secretion of growth factors essential for islet survival and function. In this review, various aspects of MSCs related to islet function and diabetes are described.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- The Institute of Biomedical and Oral Research (IBOR), Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Ronit Ahdut-HaCohen
- Department of Medical Neurobiology, Institute of Medical Research, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel;
- Department of Science, The David Yellin Academic College of Education, Jerusalem 9103501, Israel
| |
Collapse
|
18
|
Yang Y, Wu Y, Yang D, Neo SH, Kadir ND, Goh D, Tan JX, Denslin V, Lee EH, Yang Z. Secretive derived from hypoxia preconditioned mesenchymal stem cells promote cartilage regeneration and mitigate joint inflammation via extracellular vesicles. Bioact Mater 2023; 27:98-112. [PMID: 37006826 PMCID: PMC10063382 DOI: 10.1016/j.bioactmat.2023.03.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 02/23/2023] [Accepted: 03/22/2023] [Indexed: 03/31/2023] Open
Abstract
Secretome derived from mesenchymal stem cells (MSCs) have profound effects on tissue regeneration, which could become the basis of future MSCs therapies. Hypoxia, as the physiologic environment of MSCs, has great potential to enhance MSCs paracrine therapeutic effect. In our study, the paracrine effects of secretome derived from MSCs preconditioned in normoxia and hypoxia was compared through both in vitro functional assays and an in vivo rat osteochondral defect model. Specifically, the paracrine effect of total EVs were compared to that of soluble factors to characterize the predominant active components in the hypoxic secretome. We demonstrated that hypoxia conditioned medium, as well as the corresponding EVs, at a relatively low dosage, were efficient in promoting the repair of critical-sized osteochondral defects and mitigated the joint inflammation in a rat osteochondral defect model, relative to their normoxia counterpart. In vitro functional test shows enhancement through chondrocyte proliferation, migration, and matrix deposition, while inhibit IL-1β-induced chondrocytes senescence, inflammation, matrix degradation, and pro-inflammatory macrophage activity. Multiple functional proteins, as well as a change in EVs' size profile, with enrichment of specific EV-miRNAs were detected with hypoxia preconditioning, implicating complex molecular pathways involved in hypoxia pre-conditioned MSCs secretome generated cartilage regeneration.
Collapse
|
19
|
Jakl V, Popp T, Haupt J, Port M, Roesler R, Wiese S, Friemert B, Rojewski MT, Schrezenmeier H. Effect of Expansion Media on Functional Characteristics of Bone Marrow-Derived Mesenchymal Stromal Cells. Cells 2023; 12:2105. [PMID: 37626914 PMCID: PMC10453497 DOI: 10.3390/cells12162105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/07/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
The therapeutic efficacy of mesenchymal stromal cells (MSCs) has been shown to rely on their immunomodulatory and regenerative properties. In order to obtain sufficient numbers of cells for clinical applications, MSCs have to be expanded ex vivo. Expansion media with xenogeneic-free (XF) growth-promoting supplements like human platelet lysate (PL) or serum- and xenogeneic-free (SF/XF) formulations have been established as safe and efficient, and both groups provide different beneficial qualities. In this study, MSCs were expanded in XF or SF/XF media as well as in mixtures thereof. MSCs cultured in these media were analyzed for phenotypic and functional properties. MSC expansion was optimal with SF/XF conditions when PL was present. Metabolic patterns, consumption of growth factors, and secretome of MSCs differed depending on the type and concentration of supplement. The lactate per glucose yield increased along with a higher proportion of PL. Many factors in the supernatant of cultured MSCs showed distinct patterns depending on the supplement (e.g., FGF-2, TGFβ, and insulin only in PL-expanded MSC, and leptin, sCD40L PDGF-AA only in SF/XF-expanded MSC). This also resulted in changes in cell characteristics like migratory potential. These findings support current approaches where growth media may be utilized for priming MSCs for specific therapeutic applications.
Collapse
Affiliation(s)
- Viktoria Jakl
- Institute for Transfusion Medicine, University Hospital Ulm, 89081 Ulm, Germany; (V.J.)
| | - Tanja Popp
- Bundeswehr Institute of Radiobiology, 80937 Munich, Germany (J.H.); (M.P.)
| | - Julian Haupt
- Bundeswehr Institute of Radiobiology, 80937 Munich, Germany (J.H.); (M.P.)
- Clinic for Trauma Surgery and Orthopedics, Army Hospital Ulm, 89081 Ulm, Germany
| | - Matthias Port
- Bundeswehr Institute of Radiobiology, 80937 Munich, Germany (J.H.); (M.P.)
| | - Reinhild Roesler
- Core Unit of Mass Spectrometry and Proteomics, Ulm University Medical Center, 89081 Ulm, Germany; (R.R.); (S.W.)
| | - Sebastian Wiese
- Core Unit of Mass Spectrometry and Proteomics, Ulm University Medical Center, 89081 Ulm, Germany; (R.R.); (S.W.)
| | - Benedikt Friemert
- Clinic for Trauma Surgery and Orthopedics, Army Hospital Ulm, 89081 Ulm, Germany
| | - Markus T. Rojewski
- Institute for Transfusion Medicine, University Hospital Ulm, 89081 Ulm, Germany; (V.J.)
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service Baden-Württemberg—Hessia and University Hospital Ulm, 89081 Ulm, Germany
| | - Hubert Schrezenmeier
- Institute for Transfusion Medicine, University Hospital Ulm, 89081 Ulm, Germany; (V.J.)
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Donation Service Baden-Württemberg—Hessia and University Hospital Ulm, 89081 Ulm, Germany
| |
Collapse
|
20
|
Khan S, Mahgoub S, Fallatah N, Lalor PF, Newsome PN. Liver Disease and Cell Therapy: Advances Made and Remaining Challenges. Stem Cells 2023; 41:739-761. [PMID: 37052348 PMCID: PMC10809282 DOI: 10.1093/stmcls/sxad029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 02/27/2023] [Indexed: 04/14/2023]
Abstract
The limited availability of organs for liver transplantation, the ultimate curative treatment for end stage liver disease, has resulted in a growing and unmet need for alternative therapies. Mesenchymal stromal cells (MSCs) with their broad ranging anti-inflammatory and immunomodulatory properties have therefore emerged as a promising therapeutic agent in treating inflammatory liver disease. Significant strides have been made in exploring their biological activity. Clinical application of MSC has shifted the paradigm from using their regenerative potential to one which harnesses their immunomodulatory properties. Reassuringly, MSCs have been extensively investigated for over 30 years with encouraging efficacy and safety data from translational and early phase clinical studies, but questions remain about their utility. Therefore, in this review, we examine the translational and clinical studies using MSCs in various liver diseases and their impact on dampening immune-mediated liver damage. Our key observations include progress made thus far with use of MSCs for clinical use, inconsistency in the literature to allow meaningful comparison between different studies and need for standardized protocols for MSC manufacture and administration. In addition, the emerging role of MSC-derived extracellular vesicles as an alternative to MSC has been reviewed. We have also highlighted some of the remaining clinical challenges that should be addressed before MSC can progress to be considered as therapy for patients with liver disease.
Collapse
Affiliation(s)
- Sheeba Khan
- National Institute for Health Research, Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, West Midlands, UK
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
- Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, Birmingham, West Midlands, UK
| | - Sara Mahgoub
- National Institute for Health Research, Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, West Midlands, UK
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
- Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, Birmingham, West Midlands, UK
| | - Nada Fallatah
- National Institute for Health Research, Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, West Midlands, UK
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Patricia F Lalor
- National Institute for Health Research, Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, West Midlands, UK
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
| | - Philip N Newsome
- National Institute for Health Research, Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, West Midlands, UK
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
- Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, Birmingham, West Midlands, UK
| |
Collapse
|
21
|
Lin SC, Panthi S, Hsuuw YH, Chen SH, Huang MJ, Sieber M, Hsuuw YD. Regenerative Effect of Mesenchymal Stem Cell on Cartilage Damage in a Porcine Model. Biomedicines 2023; 11:1810. [PMID: 37509451 PMCID: PMC10376751 DOI: 10.3390/biomedicines11071810] [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: 05/17/2023] [Revised: 06/10/2023] [Accepted: 06/16/2023] [Indexed: 07/30/2023] Open
Abstract
Osteoarthritis (OA) is a major public and animal health challenge with significant economic consequences. Cartilage degradation plays a critical role in the initiation and progression of degenerative joint diseases, such as OA. Mesenchymal stem cells (MSCs) have become increasingly popular in the field of cartilage regeneration due to their promising results. The objective of this preclinical study was to evaluate the regenerative effects of mesenchymal stem cells (MSCs) in the repair of knee cartilage defects using a porcine model. Seven healthy LYD breed white pigs, aged 9-10 weeks and weighing approximately 20 ± 3 kg, were used in the experimental protocol. Full-thickness defects measuring 8 mm in diameter and 5 mm in depth were induced in the lateral femoral condyle of the posterior limbs in both knee joints using a sterile puncture technique while the knee was maximally flexed. Following a 1-week induction phase, the pig treatment groups received a 0.3 million/kg MSC transplant into the damaged knee region, while the placebo group received a control solution as a treatment. Magnetic resonance imaging (MRI), computerized tomography (CT), visual macroscopic examination, histological analysis, and cytokine concentration analysis were used to assess cartilage regeneration. The findings revealed that human adipose-derived mesenchymal stem cells (hADSCs) were more effective in repairing cartilage than pig umbilical cord-derived mesenchymal stem cells (pUCMSCs). These results suggest that MSC-based treatments hold promise as a treatment option for cartilage repair, which aid in the treatment of OA. However, further studies with larger sample sizes and longer follow-up periods are required to fully demonstrate the safety and efficacy of these therapies in both animals and humans.
Collapse
Affiliation(s)
- Sheng-Chuan Lin
- Department of Tropical Agriculture and International Cooperation, National Pingtung University of Science and Technology, No. 1, Xuefu Rd, Neipu Township, Pingtung 91201, Taiwan
- Deng Chuan Animal Hospital, Kaohsiung 81361, Taiwan
| | - Sankar Panthi
- Department of Tropical Agriculture and International Cooperation, National Pingtung University of Science and Technology, No. 1, Xuefu Rd, Neipu Township, Pingtung 91201, Taiwan
| | - Yu-Her Hsuuw
- Department of Tropical Agriculture and International Cooperation, National Pingtung University of Science and Technology, No. 1, Xuefu Rd, Neipu Township, Pingtung 91201, Taiwan
- Deng Chuan Animal Hospital, Kaohsiung 81361, Taiwan
| | | | | | | | - Yan-Der Hsuuw
- Department of Tropical Agriculture and International Cooperation, National Pingtung University of Science and Technology, No. 1, Xuefu Rd, Neipu Township, Pingtung 91201, Taiwan
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan
| |
Collapse
|
22
|
Chatchavalvanich S, Boomsma RA, Tietema JM, Geenen DL. Inhibition of Gap Junction Formation Prior to Implantation of Bone Marrow-Derived Mesenchymal Cells Improves Function in the Ischemic Myocardium. Int J Mol Sci 2023; 24:9653. [PMID: 37298612 PMCID: PMC10253678 DOI: 10.3390/ijms24119653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/22/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Bone marrow-derived mesenchymal stem cells (BM-MSC) are reported to induce beneficial effects in the heart following ischemia, but a loss of these cells within hours of implantation could significantly diminish their long-term effect. We hypothesized that early coupling between BM-MSC and ischemic cardiomyocytes through gap junctions (GJ) may play an important role in stem cell survival and retention in the acute phase of myocardial ischemia. To determine the effect of GJ inhibition on murine BM-MSC in vivo, we induced ischemia in mice using 90 min left anterior descending coronary artery (LAD) occlusion followed by BM-MSC implantation and reperfusion. The inhibition of GJ coupling prior to BM-MSC implantation led to early improvement in cardiac function compared to mice in which GJ coupling was not inhibited. Our results with in vitro studies also demonstrated increased survival in BM-MSCs subjected to hypoxia after inhibition of GJ. While functional GJ are critical for the long-term integration of stem cells within the myocardium, early GJ communication may represent a novel paradigm whereby ischemic cardiomyocytes induce a "bystander effect" when coupled to newly transplanted BM-MSC and thus impair cell retention and survival.
Collapse
Affiliation(s)
- Santipongse Chatchavalvanich
- Department of Basic Biomedical Sciences, Dr. William M. Scholl College of Podiatric Medicine, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA;
| | - Robert A. Boomsma
- Biology Department, Trinity Christian College, Palos Heights, IL 60463, USA;
| | - Jack M. Tietema
- Michigan State University College of Human Medicine, Grand Rapids, MI 49503, USA;
| | - David L. Geenen
- Physician Assistant Studies Department, College of Health Professions, Grand Valley State University, Grand Rapids, MI 49503, USA
| |
Collapse
|
23
|
Disruption of CCL2 in Mesenchymal Stem Cells as an Anti-Tumor Approach against Prostate Cancer. Cancers (Basel) 2023; 15:cancers15020441. [PMID: 36672395 PMCID: PMC9857005 DOI: 10.3390/cancers15020441] [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: 12/08/2022] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND MSCs are known to secrete abundant CCL2, which plays a crucial role in recruiting TAMs, promoting tumor progression. It is important to know whether disrupting MSC-derived CCL2 affects tumor growth. METHODS Murine bone marrow-derived MSCs were characterized by their surface markers and differentiation abilities. Proliferation and migration assays were performed in order to evaluate the functions of MSCs on cancer cells. CCL2 expression in MSCs was reduced by small interfering RNA (siRNA) or completely disrupted by CRISPR/Cas9 knockout (KO) approaches. An immune-competent syngeneic murine model of prostate cancer was applied in order to assess the role of tumor cell- and MSC-derived CCL2. The tumor microenvironment was analyzed to monitor the immune profile. RESULTS We confirmed that tumor cell-derived CCL2 was crucial for tumor growth and MSCs migration. CCL2 KO MSCs inhibited the migration of the monocyte/macrophage but not the proliferation of tumor cells in vitro. However, the mice co-injected with tumor cells and CCL2 KO MSCs exhibited anti-tumor effects when compared with those given tumor cell alone and with control MSCs, partly due to increased infiltration of CD45+CD11b+Ly6G- mononuclear myeloid cells. CONCLUSIONS Disruption of MSC-derived CCL2 enhances anti-tumor functions in an immune-competent syngeneic mouse model for prostate cancer.
Collapse
|
24
|
Wang D, Xie Y, Peng HQ, Wen ZM, Ying ZY, Geng C, Wu J, Lv HY, Xu B. LPS preconditioning of MSC-CM improves protection against hypoxia/reoxygenation-induced damage in H9c2 cells partly via HMGB1/Bach1 signalling. Clin Exp Pharmacol Physiol 2022; 49:1319-1333. [PMID: 36052438 DOI: 10.1111/1440-1681.13714] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/14/2022] [Accepted: 08/28/2022] [Indexed: 01/31/2023]
Abstract
Mesenchymal stem cell-derived conditioned medium (MSC-CM) improves cardiac function after myocardial infarction; however, this cardioprotective effect is moderate and transient. Lipopolysaccharide (LPS) pretreatment partially improves MSC-CM-mediated cardioprotective effects owing to the presence of paracrine factors. However, the mechanism underlying these improved effects remains unknown. To study the effect of LPS-pretreated MSC-CM on hypoxia/reoxygenation (H/R)-induced injury, MSCs were treated with or without LPS (400 ng/mL) for 48 h, and the supernatant was collected (MSC-CM). Subsequently, H9c2 cells were co-cultured with Nor-CM (CM derived from LPS-untreated MSCs) and LPS-CM (CM derived from LPS-pretreated MSCs) for 24 h and subjected to H/R. MSC-CM inhibited the progression of H/R-induced injury in H9c2 cells, and this protective effect was enhanced via LPS pretreatment as evidenced by the improved apoptosis assessment index (i.e. caspase-3 and B-cell lymphoma-2 [Bcl-2] expression) and decreased levels of lactic dehydrogenase (LDH) and cardiac troponin (cTn). In addition, the results of haematoxylin-eosin staining (H&E), transmission electron microscopy (TEM) and TdT-mediated dUTP nick-end labelling (TUNEL) validated that MSC-CM inhibited H/R-induced injury in H9c2 cardiomyocytes. LPS pretreatment downregulated the expression of high mobility group box-1 (HMGB1) and BTB and CNC homology-1 (Bach1) proteins in MSCs but upregulated the expression of vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF) and insulin-like growth factor (IGF). HMGB1 knockdown (MSC/siHMGB1-CM) significantly decreased the expression of Bach1 and increased the expression of VEGF, HGF and IGF. Bach1 knockdown (MSC/siBach1-CM) did not alter the production of HMGB1 but increased the expression of VEGF and IGF. LPS pretreatment did not alter the expression of the paracrine factors VEGF and HGF in the MSC/siHMGB1 group but increased their expression in the MSC/siBach1 group. The myocyte anti-apoptotic effects of MSCs/siBach1-CM were similar to those of untreated MSCs, which were not enhanced by LPS. LPS-pretreated MSC-CM protects H9c2 cells against H/R-induced injury partly through the HMGB1/Bach1 signalling pathway.
Collapse
Affiliation(s)
- Dan Wang
- Department of Clinical Pharmacy, The Second Affiliated Hospital of Dalian Medical University, Dalian, China.,Department of Pharmacy, Ordos Central Hospital, Ordos, China
| | - Yu Xie
- Department of Clinical Pharmacy, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hui-Qian Peng
- Department of Clinical Pharmacy, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhi-Min Wen
- Department of Clinical Laboratory, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zi-Yue Ying
- Department of Clinical Laboratory, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Cong Geng
- Department of Clinical Laboratory, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jun Wu
- Department of Echocardiography, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hui-Yi Lv
- Department of Clinical Pharmacy, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Bing Xu
- Department of Clinical Pharmacy, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
25
|
Hajighasemlou S, Nikbakht M, Pakzad S, Azadbakht A, Muhammadnejad S, Mirmoghtadaei M, Gharibzadeh S, Seyhoun I, Verdi J. Anti-inflammatory effect of mesenchymal stem cells on hepatocellular carcinoma in the xenograft mice model. Vet Med Sci 2022; 8:2086-2091. [PMID: 35838746 PMCID: PMC9514506 DOI: 10.1002/vms3.886] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the fifth most diagnosed cancer and the second leading cause of cancer-related deaths worldwide. Sorafenib is the standard treatment used in the advanced stages of HCC. Cell therapy with mesenchymal stem cells (MSCs)-based cell therapy has proven effective in immune regulation and tumour growth inhibition. OBJECTIVES In this study, we investigated the anti-inflammatory effect of MSCs on HCC xenografts. METHODS Human HepG2 cell lines were subcutaneously implanted into the flank of 12 nude mice, divided into three groups: the control group, the IV group (intravenous MSCs injection) and the local group (local MSCs injection). Mice were sacrificed 6 weeks after tumour implantation, and tumours were resected entirety. Quantitative real-time polymerase chain reaction (qRT-PCR) measured the gene expression of inflammatory markers, including tumour necrosis factor-α (TNF-α), interleukin (IL)-1α and IL-10. Aspartate transaminase (AST), alanine transaminase (ALT) and urea levels were measured using spectrophotometry to ensure the safety of MSC therapy. RESULTS Gene expressions for all three inflammatory markers were reduced in both MSCs groups compared to the control group. AST, ALT and urea levels remained in normal ranges. CONCLUSIONS MSC therapy can reduce inflammation in HCC xenograft mouse models.
Collapse
Affiliation(s)
- Saieh Hajighasemlou
- Department of Tissue Engineering & Applied Cell SciencesTehran University of Medical Sciences (TUMS)TehranIran
- Food and Drug AdministrationMinistry of Health and Medical EducationTehranIran
| | - Mohsen Nikbakht
- Hematology, Oncology & Stem Cell Transplantation Research CenterTehran University of Medical Sciences (TUMS)TehranIran
| | - Saeedreza Pakzad
- Food and Drug AdministrationMinistry of Health and Medical EducationTehranIran
| | - Abdolnaser Azadbakht
- Department of Biomedical Engineering, Central Tehran BranchIslamic Azad universityTehranIran
- Stem cells Research Center, Tissue Engineering and Regenerative Medicine Institute, Central Tehran BranchIslamic Azad universityTehranIran
| | - Samad Muhammadnejad
- Gene Therapy Research Center, Digestive Diseases Research InstituteTehran University of Medical SciencesTehranIran
| | - Milad Mirmoghtadaei
- Immunology, Asthma and Allergy Research InstituteTehran University of Medical Sciences (TUMS)TehranIran
- Children's Medical Center, Pediatrics Center of ExcellenceTehran University of Medical Sciences (TUMS)TehranIran
| | - Safoora Gharibzadeh
- Department of Epidemiology and Biostatistics Research Centre for Emerging and Reemerging Infectious DiseasesPasteur instituteTehranIran
| | - Iman Seyhoun
- Department of Tissue Engineering & Applied Cell SciencesTehran University of Medical Sciences (TUMS)TehranIran
| | - Javad Verdi
- Department of Tissue Engineering & Applied Cell SciencesTehran University of Medical Sciences (TUMS)TehranIran
| |
Collapse
|
26
|
Ou X, Wen T, Ying J, He Q, Xuan A, Ruan D. MCP‑1/CCR2 axis inhibits the chondrogenic differentiation of human nucleus pulposus mesenchymal stem cells. Mol Med Rep 2022; 26:277. [PMID: 35856417 DOI: 10.3892/mmr.2022.12793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 11/15/2021] [Indexed: 11/06/2022] Open
Affiliation(s)
- Xuancheng Ou
- Department of Spine Surgery, The Central Hospital of Yongzhou, Yongzhou, Hunan 425000, P.R. China
| | - Tianyong Wen
- Department of Orthopedic Surgery, The Sixth Medical Centre of PLA General Hospital, Beijing 100053, P.R. China
| | - Jinwei Ying
- Department of Orthopedic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Qing He
- Department of Orthopedic Surgery, The Sixth Medical Centre of PLA General Hospital, Beijing 100053, P.R. China
| | - Anwu Xuan
- Department of Orthopedic Surgery, The Sixth Medical Centre of PLA General Hospital, Beijing 100053, P.R. China
| | - Dike Ruan
- Department of Orthopedic Surgery, The Sixth Medical Centre of PLA General Hospital, Beijing 100053, P.R. China
| |
Collapse
|
27
|
Neural Differentiation Potential of Mesenchymal Stem Cells Enhanced by Biocompatible Chitosan-Gold Nanocomposites. Cells 2022; 11:cells11121861. [PMID: 35740991 PMCID: PMC9221394 DOI: 10.3390/cells11121861] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/28/2022] [Accepted: 06/04/2022] [Indexed: 02/04/2023] Open
Abstract
Chitosan (Chi) is a natural polymer that has been demonstrated to have potential as a promoter of neural regeneration. In this study, Chi was prepared with various amounts (25, 50, and 100 ppm) of gold (Au) nanoparticles for use in in vitro and in vivo assessments. Each as-prepared material was first characterized by UV-visible spectroscopy (UV-Vis), Fourier-transform infrared spectroscopy (FTIR), atomic force microscopy (AFM), scanning electron microscopy (SEM), and Dynamic Light Scattering (DLS). Through the in vitro experiments, Chi combined with 50 ppm of Au nanoparticles demonstrated better biocompatibility. The platelet activation, monocyte conversion, and intracellular ROS generation was remarkably decreased by Chi–Au 50 pm treatment. Furthermore, Chi–Au 50 ppm could facilitate colony formation and strengthen matrix metalloproteinase (MMP) activation in mesenchymal stem cells (MSCs). The lower expression of CD44 in Chi–Au 50 ppm treatment demonstrated that the nanocomposites could enhance the MSCs undergoing differentiation. Chi–Au 50 ppm was discovered to significantly induce the expression of GFAP, β-Tubulin, and nestin protein in MSCs for neural differentiation, which was verified by real-time PCR analysis and immunostaining assays. Additionally, a rat model involving subcutaneous implantation was used to evaluate the superior anti-inflammatory and endothelialization abilities of a Chi–Au 50 ppm treatment. Capsule formation and collagen deposition were decreased. The CD86 expression (M1 macrophage polarization) and leukocyte filtration (CD45) were remarkably reduced as well. In summary, a Chi polymer combined with 50 ppm of Au nanoparticles was proven to enhance the neural differentiation of MSCs and showed potential as a biosafe nanomaterial for neural tissue engineering.
Collapse
|
28
|
Gerdesmeyer L, Zielhardt P, Klüter T, Gollwitzer H, Gerdesmeyer L, Hausdorf J, Ringeisen M, Knobloch K, Saxena A, Krath A. Stimulation of human bone marrow mesenchymal stem cells by electromagnetic transduction therapy - EMTT. Electromagn Biol Med 2022; 41:304-314. [DOI: 10.1080/15368378.2022.2079672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Ludger Gerdesmeyer
- Department of Sports Medicine, Palo Alto Medical Center, Palo Alto, USA
- Department of Orthopedic and Traumatology, Technical University Munich, Munich Germany
- Städtisches Krankenhaus Kiel, Kiel
| | - Paula Zielhardt
- Department of Orthopaedic Surgery and Traumatology, University Schleswig Holstein, Kiel, Germany
| | - Tim Klüter
- Department of Orthopaedic Surgery and Traumatology, University Schleswig Holstein, Kiel, Germany
| | - Hans Gollwitzer
- Department of Orthopedic and Traumatology, Technical University Munich, Munich Germany
| | - Lennart Gerdesmeyer
- Department of Orthopedic and Traumatology, Technical University Munich, Munich Germany
| | - Joerg Hausdorf
- Orthopedic Department, Physical Medicine and Rehabilitation, University Hospital of Munich, Germany
| | - Martin Ringeisen
- Department of Orthopedic Surgery and Traumatology, Orthopaedic Medical Center Dr. Ringeisen, Augsburg, Germany
| | - Karsten Knobloch
- Orthopedic Department, SportPraxis Prof. Dr. med. Karsten Knobloch, Hannover
| | - Amol Saxena
- Department of Sports Medicine, Palo Alto Medical Center, Palo Alto, USA
| | - André Krath
- Department of Orthopaedic Surgery and Traumatology, University Schleswig Holstein, Kiel, Germany
| |
Collapse
|
29
|
Maurya DK, Bandekar M, Sandur SK. Soluble factors secreted by human Wharton’s jelly mesenchymal stromal/stem cells exhibit therapeutic radioprotection: A mechanistic study with integrating network biology. World J Stem Cells 2022; 14:347-361. [PMID: 35722198 PMCID: PMC9157603 DOI: 10.4252/wjsc.v14.i5.347] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/25/2022] [Accepted: 05/08/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Human Wharton’s jelly-derived mesenchymal stromal/stem cells (hWJ-MSCs) have gained considerable attention in their applications in cell-based therapy due to several advantages offered by them. Recently, we reported that hWJ-MSCs and their conditioned medium have significant therapeutic radioprotective potential. This finding raised an obvious question to identify unique features of hWJ-MSCs over other sources of stem cells for a better understanding of its radioprotective mechanism.
AIM To understand the radioprotective mechanism of soluble factors secreted by hWJ-MSCs and identification of their unique genes.
METHODS Propidium iodide staining, endogenous spleen colony-forming assay, and survival study were carried out for radioprotection studies. Homeostasis-driven proliferation assay was performed for in vivo lymphocyte proliferation. Analysis of RNAseq data was performed to find the unique genes of WJ-MSCs by comparing them with bone marrow mesenchymal stem cells, embryonic stem cells, and human fibroblasts. Gene enrichment analysis and protein-protein interaction network were used for pathway analysis.
RESULTS Co-culture of irradiated murine splenic lymphocytes with WJ-MSCs offered significant radioprotection to lymphocytes. WJ-MSC transplantation increased the homeostasis-driven proliferation of the lymphocytes. Neutralization of WJ-MSC conditioned medium with granulocyte-colony stimulating factor antibody abolished therapeutic radioprotection. Transcriptome analysis showed that WJ-MSCs share several common genes with bone marrow MSCs and embryonic stem cells and express high levels of unique genes such as interleukin (IL)1-α, IL1-β, IL-6, CXCL3, CXCL5, CXCL8, CXCL2, CCL2, FLT-1, and IL-33. It was also observed that WJ-MSCs preferentially modulate several cellular pathways and processes that handle the repair and regeneration of damaged tissues compared to stem cells from other sources. Cytokine-based network analysis showed that most of the radiosensitive tissues have a more complex network for the elevated cytokines.
CONCLUSION Systemic infusion of WJ-MSC conditioned media will have significant potential for treating accidental radiation exposed victims.
Collapse
Affiliation(s)
- Dharmendra Kumar Maurya
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai 400085, Maharashtra, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Mayuri Bandekar
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai 400085, Maharashtra, India
- University of Mumbai, Kalina, Mumbai 400098, India
| | - Santosh Kumar Sandur
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai 400085, Maharashtra, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| |
Collapse
|
30
|
Bian D, Wu Y, Song G, Azizi R, Zamani A. The application of mesenchymal stromal cells (MSCs) and their derivative exosome in skin wound healing: a comprehensive review. Stem Cell Res Ther 2022; 13:24. [PMID: 35073970 PMCID: PMC8785459 DOI: 10.1186/s13287-021-02697-9] [Citation(s) in RCA: 165] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/22/2021] [Indexed: 12/15/2022] Open
Abstract
Recently, mesenchymal stromal cells (MSCs) and also their exosome has become a game-changing tool in the context of tissue engineering and regenerative medicine. MSCs due to their competencies to establish skin cells, such as fibroblast and keratinocyte, and also their unique attribute to suppress inflammation in wound site has attracted increasing attention among scholars. In addition, MSC's other capabilities to induce angiogenesis as a result of secretion of pro-angiogenic factors accompanied with marked anti-fibrotic activities, which mainly mediated by the releases matrix metalloproteinase (MMPs), make them a rational and effective strategy to accelerate wound healing with a small scar. Since the chief healing properties of the MSCs depend on their paracrine effects, it appears that MSCs-derived exosomes also can be an alternative option to support wound healing and skin regeneration as an innovative cell-free approach. Such exosomes convey functional cargos (e.g., growth factor, cytokine, miRNA, etc.) from MSCs to target cells, thereby affecting the recipient skin cells' biological events, such as migration, proliferation, and also secretion of ECM components (e.g., collagen). The main superiorities of exosome therapy over parental MSCs are the diminished risk of tumor formation and also lower immunogenicity. Herein, we deliver an overview of recent in vivo reports rendering the therapeutic benefits of the MSCs-based therapies to ease skin wound healing, and so improving quality of life among patients suffering from such conditions.
Collapse
Affiliation(s)
- Donghui Bian
- Department of Burns and Plastic Surgery, 960 Hospital of the People’s Liberation Army, Jinan, 250031 China
| | - Yan Wu
- Department of Burns and Plastic Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250013 China
| | - Guodong Song
- Department of Burns and Plastic Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250013 China
| | - Ramyar Azizi
- Department of Immunology, Medicine Faculty, Tabriz University of Medical Science, Tabriz, Iran
| | - Amir Zamani
- Shiraz Transplant Center, Abu Ali Sina Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
31
|
Culenova M, Nicodemou A, Novakova ZV, Debreova M, Smolinská V, Bernatova S, Ivanisova D, Novotna O, Vasicek J, Varga I, Ziaran S, Danisovic L. Isolation, Culture and Comprehensive Characterization of Biological Properties of Human Urine-Derived Stem Cells. Int J Mol Sci 2021; 22:12503. [PMID: 34830384 PMCID: PMC8624597 DOI: 10.3390/ijms222212503] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) represent an attractive source within the field of tissue engineering. However, their harvesting often requires invasive medical procedures. Urine-derived stem cells (UDSCs) display similar properties to MSCs, and their obtention and further processing is non-invasive for the donors as well as low cost. Here, we offer a comprehensive analysis of their biological properties. The goal of this study was to analyze their morphology, stemness, differentiation potential and cytokine profile. We have successfully isolated UDSCs from 25 urine samples. First colonies emerged up to 9 days after the initial seeding. Cell doubling time was 45 ± 0.24 SD, and when seeded at the density of 100 cells/cm2, they formed 42 ± 6.5 SD colonies within 10 days. Morphological analyzes revealed that two different types of the cell populations have been present. The first type had a rice-grain shape and the second one was characterized by a polyhedral shape. In several cell cultures, dome-shaped cells were observed as well. All examined UDSCs expressed typical MSC-like surface markers, CD73, CD90 and CD105. Moreover, conditioned media from UDSCs were harvested, and cytokine profile has been evaluated showing a significantly higher secretory rate of IL-8, IL-6 and chemokines MCP-1 and GM-CSF. We have also successfully induced human UDSCs into chondrogenic, osteogenic and myogenic cell lineages. Our findings indicate that UDSCs might have immense potential in the regeneration of the damaged tissues.
Collapse
Affiliation(s)
- Martina Culenova
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia; (M.C.); (A.N.); (Z.V.N.); (V.S.); (S.B.); (D.I.)
- Panara Ltd., Krskanska 21, 949 05 Nitra, Slovakia
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia; (M.D.); (I.V.); (S.Z.)
| | - Andreas Nicodemou
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia; (M.C.); (A.N.); (Z.V.N.); (V.S.); (S.B.); (D.I.)
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia; (M.D.); (I.V.); (S.Z.)
| | - Zuzana Varchulova Novakova
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia; (M.C.); (A.N.); (Z.V.N.); (V.S.); (S.B.); (D.I.)
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia; (M.D.); (I.V.); (S.Z.)
| | - Michaela Debreova
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia; (M.D.); (I.V.); (S.Z.)
| | - Veronika Smolinská
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia; (M.C.); (A.N.); (Z.V.N.); (V.S.); (S.B.); (D.I.)
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia; (M.D.); (I.V.); (S.Z.)
| | - Sona Bernatova
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia; (M.C.); (A.N.); (Z.V.N.); (V.S.); (S.B.); (D.I.)
| | - Dana Ivanisova
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia; (M.C.); (A.N.); (Z.V.N.); (V.S.); (S.B.); (D.I.)
| | - Olga Novotna
- Department of Pediatric Urology, Faculty of Medicine, Comenius University, Limbova 1, 831 01 Bratislava, Slovakia;
| | - Jaromir Vasicek
- Institute of Farm Animal Genetics and Reproduction, NPPC-Research Institute for Animal Production in Nitra, Hlohovecka 2, 951 41 Luzianky, Slovakia;
- Institute of Biotechnology, Faculty of Biotechnology and Food Science, Slovak University of Agriculture in Nitra, Tr. A. Hlinku 2, 949 76 Nitra, Slovakia
| | - Ivan Varga
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia; (M.D.); (I.V.); (S.Z.)
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Stanislav Ziaran
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia; (M.D.); (I.V.); (S.Z.)
- Department of Urology, Faculty of Medicine, Comenius University, Limbova 5, 833 05 Bratislava, Slovakia
| | - Lubos Danisovic
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia; (M.C.); (A.N.); (Z.V.N.); (V.S.); (S.B.); (D.I.)
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia; (M.D.); (I.V.); (S.Z.)
| |
Collapse
|
32
|
Shariati M, Esfahani RJ, Bidkhori HR, Sabouri E, Mehrzad S, Sadr-Nabavi A. Cell-based treatment of cerebral palsy: still a long way ahead. Curr Stem Cell Res Ther 2021; 17:741-749. [PMID: 34727864 DOI: 10.2174/1574888x16666211102090230] [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: 04/21/2021] [Revised: 07/28/2021] [Accepted: 08/19/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Cerebral palsy (CP) is a permanent neurodevelopmental disorder with considerable global disability. Various rehabilitation strategies are currently available. However, none represents a convincing curative result. Cellular therapy recently holds much promise as an alternative strategy to repair neurologic defects. METHOD In this narrative review, a comprehensive search of the MEDLINE and ClinicalTrials.gov was made, using the terms: "cell therapy" and "cerebral palsy", including published and registered clinical studies, respectively. RESULTS The early effects of these studies demonstrated that using cell therapy in CP patients is safe and improves the deficits for a variable duration. Despite such hopeful early bird results, the long-term outcomes are not conclusive. CONCLUSIONS Due to the heterogeneous nature of CP, personal factors seem essential to consider. Cell dosage, routes of administration, and repeated dosing are pivotal to establish optimal personalized treatments. Future clinical trials should consider employing other cell types, specific cell modifications before administration, and cell-free platforms.
Collapse
Affiliation(s)
- Mohammad Shariati
- Stem Cells and Regenerative Medicine Research Department, Academic Center for Education, Culture, and Research (ACECR)-Khorasan Razavi, Mashhad. Iran
| | - Reza Jafarzadeh Esfahani
- Blood Borne Infections Research Center, Academic Center for Education, Culture and Research (ACECR)- Khorasan Razavi, Mashhad. Iran
| | - Hamid Reza Bidkhori
- Stem Cells and Regenerative Medicine Research Department, Academic Center for Education, Culture, and Research (ACECR)-Khorasan Razavi, Mashhad. Iran
| | - Ehsan Sabouri
- Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad. Iran
| | - Shadi Mehrzad
- Stem Cells and Regenerative Medicine Research Department, Academic Center for Education, Culture, and Research (ACECR)-Khorasan Razavi, Mashhad. Iran
| | - Ariane Sadr-Nabavi
- Department of Neurology, School of Medicine, Mashhad University of Medical Sciences, Mashhad. Iran
| |
Collapse
|
33
|
2, 3, 7, 8-Tetrachlorodibenzo-p-dioxin potential impacts on peripheral blood mononuclear cells of endometriosis women. J Reprod Immunol 2021; 149:103439. [PMID: 34781065 DOI: 10.1016/j.jri.2021.103439] [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: 04/11/2021] [Revised: 08/22/2021] [Accepted: 10/27/2021] [Indexed: 11/22/2022]
Abstract
Endometriosis happens following the implantation of endometrial-derived tissues outside the uterine cavity. It has been suggested that 2, 3, 7, 8-Tetrachlorodibenzo-p-dioxin (TCDD) is involved in endometriosis development. Furthermore, aryl hydrocarbon receptor (AHR), as a TCDD receptor, has been demonstrated to regulate immune responses. Nonetheless, data regarding the mechanisms, through which TCDD influences the immune system in endometriosis, are still inconclusive. Therefore, frequency of regulatory T cells (Tregs) and the expression of FOXP3, AHR and indoleamine 2, 3-dioxygenase 1 (IDO1) from endometriosis and non-endometriosis individuals were investigated in the absence and presence of TCDD; also, the concentration of IL-6 and kynurenine in the supernatant of cultures was assessed. The impact of TCDD-treated PBMCs on the migration capacity of menstrual blood-derived stromal stem cells (MenSCs) and monocyte chemoattractant protein-1 (MCP-1) and IL-6 production was determined. Here, we found that AHR and IDO1 expression levels were lower in endometriosis PBMCs; however, TCDD treatment increased AHR, FOXP3, IDO1, IL-6, and Treg levels in the endometriosis group (P ≤ 0.05-0.0001). TCDD-treated PBMCs increased the migration capacity of MenSCs and up-regulated MCP-1 and IL-6 levels in the PBMCs/MenSCs co-culture (P ≤ 0.01-0.0001). In conclusion, these results shed light on the probable mechanisms, through which AHR activation by chemical toxicants can impact inflammatory immune mediators involved in the development of endometriosis; also, these data support the idea that TCDD could promote endometriosis progression.
Collapse
|
34
|
Hung HS, Kao WC, Shen CC, Chang KB, Tang CM, Yang MY, Yang YC, Yeh CA, Li JJ, Hsieh HH. Inflammatory Modulation of Polyethylene Glycol-AuNP for Regulation of the Neural Differentiation Capacity of Mesenchymal Stem Cells. Cells 2021; 10:2854. [PMID: 34831077 PMCID: PMC8616252 DOI: 10.3390/cells10112854] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/12/2021] [Accepted: 10/20/2021] [Indexed: 12/15/2022] Open
Abstract
A nanocomposite composed of polyethylene glycol (PEG) incorporated with various concentrations (~17.4, ~43.5, ~174 ppm) of gold nanoparticles (Au) was created to investigate its biocompatibility and biological performance in vitro and in vivo. First, surface topography and chemical composition was determined through UV-visible spectroscopy (UV-Vis), Fourier-transform infrared spectroscopy (FTIR), atomic force microscopy (AFM), scanning electron microscopy (SEM), free radical scavenging ability, and water contact angle measurement. Additionally, the diameters of the PEG-Au nanocomposites were also evaluated through dynamic light scattering (DLS) assay. According to the results, PEG containing 43.5 ppm of Au demonstrated superior biocompatibility and biological properties for mesenchymal stem cells (MSCs), as well as superior osteogenic differentiation, adipocyte differentiation, and, particularly, neuronal differentiation. Indeed, PEG-Au 43.5 ppm induced better cell adhesion, proliferation and migration in MSCs. The higher expression of the SDF-1α/CXCR4 axis may be associated with MMPs activation and may have also promoted the differentiation capacity of MSCs. Moreover, it also prevented MSCs from apoptosis and inhibited macrophage and platelet activation, as well as reactive oxygen species (ROS) generation. Furthermore, the anti-inflammatory, biocompatibility, and endothelialization capacity of PEG-Au was measured in a rat model. After implanting the nanocomposites into rats subcutaneously for 4 weeks, PEG-Au 43.5 ppm was able to enhance the anti-immune response through inhibiting CD86 expression (M1 polarization), while also reducing leukocyte infiltration (CD45). Moreover, PEG-Au 43.5 ppm facilitated CD31 expression and anti-fibrosis ability. Above all, the PEG-Au nanocomposite was evidenced to strengthen the differentiation of MSCs into various cells, including fat, vessel, and bone tissue and, particularly, nerve cells. This research has elucidated that PEG combined with the appropriate amount of Au nanoparticles could become a potential biomaterial able to cooperate with MSCs for tissue regeneration engineering.
Collapse
Affiliation(s)
- Huey-Shan Hung
- Graduate Institute of Biomedical Science, China Medical University, Taichung 40402, Taiwan; (W.-C.K.); (K.-B.C.); (C.-A.Y.); (J.-J.L.)
- Translational Medicine Research, China Medical University Hospital, Taichung 40402, Taiwan
| | - Wei-Chien Kao
- Graduate Institute of Biomedical Science, China Medical University, Taichung 40402, Taiwan; (W.-C.K.); (K.-B.C.); (C.-A.Y.); (J.-J.L.)
| | - Chiung-Chyi Shen
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung 407204, Taiwan; (C.-C.S.); (M.-Y.Y.); (Y.-C.Y.)
- Department of Physical Therapy, Hung Kuang University, Taichung 433304, Taiwan
- Basic Medical Education Center, Central Taiwan University of Science and Technology, Taichung 406053, Taiwan
| | - Kai-Bo Chang
- Graduate Institute of Biomedical Science, China Medical University, Taichung 40402, Taiwan; (W.-C.K.); (K.-B.C.); (C.-A.Y.); (J.-J.L.)
| | - Cheng-Ming Tang
- College of Oral Medicine, Chung Shan Medical University, Taichung 40201, Taiwan;
- Blood Bank, Taichung Veterans General Hospital, Taichung 407204, Taiwan;
| | - Meng-Yin Yang
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung 407204, Taiwan; (C.-C.S.); (M.-Y.Y.); (Y.-C.Y.)
| | - Yi-Chin Yang
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung 407204, Taiwan; (C.-C.S.); (M.-Y.Y.); (Y.-C.Y.)
| | - Chun-An Yeh
- Graduate Institute of Biomedical Science, China Medical University, Taichung 40402, Taiwan; (W.-C.K.); (K.-B.C.); (C.-A.Y.); (J.-J.L.)
| | - Jia-Jhan Li
- Graduate Institute of Biomedical Science, China Medical University, Taichung 40402, Taiwan; (W.-C.K.); (K.-B.C.); (C.-A.Y.); (J.-J.L.)
| | - Hsien-Hsu Hsieh
- Blood Bank, Taichung Veterans General Hospital, Taichung 407204, Taiwan;
| |
Collapse
|
35
|
Hassanshahi G, Roohi MA, Esmaeili SA, Pourghadamyari H, Nosratabadi R. Involvement of various chemokine/chemokine receptor axes in trafficking and oriented locomotion of mesenchymal stem cells in multiple sclerosis patients. Cytokine 2021; 148:155706. [PMID: 34583254 DOI: 10.1016/j.cyto.2021.155706] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 09/04/2021] [Accepted: 09/07/2021] [Indexed: 12/25/2022]
Abstract
Multiple sclerosis (MS) is a specific type of chronic immune-mediated disease in which the immune responses are almost run against the central nervous system (CNS). Despite intensive research, a known treatment for MS disease yet to be introduced. Thus, the development of novel and safe medications needs to be considered for the disease management. Application of mesenchymal stem cells (MSCs) as an emerging approach was recruited forthe treatment of MS. MSCs have several sources and they can be derived from the umbilical cord, adipose tissue, and bone marrow. Chemokines are low molecular weight proteins that their functional activities are achieved by binding to the cell surface G protein-coupled receptors (GPCRs). Chemokine and chemokine receptors are of the most important and effective molecules in MSC trafficking within the different tissues in hemostatic and non-hemostatic circumstances. Chemokine/chemokine receptor axes play a pivotal role in the recruitment and oriented trafficking of immune cells both towards and within the CNS and it appears that chemokine/chemokine receptor signaling may be the most important leading mechanisms in the pathogenesis of MS. In this article, we hypothesized that the chemokine/chemokine receptor axes network have crucial and efficacious impacts on behavior of the MSCs, nonetheless, the exact responsibility of these axes on the targeted tropism of MSCs to the CNS of MS patients yet remained to be fully elucidated. Therefore, we reviewed the ability of MSCs to migrate and home into the CNS of MS patients via expression of various chemokine receptors in response to chemokines expressed by cells of CNS tissue, to provide a great source of knowledge.
Collapse
Affiliation(s)
- Gholamhossein Hassanshahi
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Amin Roohi
- Department of Medical Immunology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Pourghadamyari
- Department of Clinical Biochemistry, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Reza Nosratabadi
- Department of Medical Immunology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
36
|
Ahani-Nahayati M, Niazi V, Moradi A, Pourjabbar B, Roozafzoon R, Baradaran-Rafii A, Keshel SH. Umbilical cord mesenchymal stem/stromal cells potential to treat organ disorders; an emerging strategy. Curr Stem Cell Res Ther 2021; 17:126-146. [PMID: 34493190 DOI: 10.2174/1574888x16666210907164046] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 06/04/2021] [Accepted: 06/11/2021] [Indexed: 11/22/2022]
Abstract
Currently, mesenchymal stem/stromal cells (MSCs) have attracted growing attention in the context of cell-based therapy in regenerative medicine. Following the first successful procurement of human MSCs from bone marrow (BM), these cells isolation has been conducted from various origins, in particular, the umbilical cord (UC). Umbilical cord-derived mesenchymal stem/stromal cells (UC-MSCs) can be acquired by a non-invasive plan and simply cultured, and thereby signifies their superiority over MSCs derived from other sources for medical purposes. Due to their unique attributes, including self-renewal, multipotency, and accessibility concomitant with their immunosuppressive competence and lower ethical concerns, UC-MSCs therapy is described as encouraging therapeutic options in cell-based therapies. Regardless of their unique aptitude to adjust inflammatory response during tissue recovery and delivering solid milieu for tissue restoration, UC-MSCs can be differentiated into a diverse spectrum of adult cells (e.g., osteoblast, chondrocyte, type II alveolar, hepatocyte, and cardiomyocyte). Interestingly, they demonstrate a prolonged survival and longer telomeres compared with MSCs derived from other sources, suggesting that UC-MSCs are desired source to use in regenerative medicine. In the present review, we deliver a brief review of UC-MSCs isolation, expansion concomitantly with immunosuppressive activities, and try to collect and discuss recent pre-clinical and clinical researches based on the use of UC-MSCs in regenerative medicine, focusing on with special focus on in vivo researches.
Collapse
Affiliation(s)
- Milad Ahani-Nahayati
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Science, Tehran. Iran
| | - Vahid Niazi
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Science, Tehran. Iran
| | - Alireza Moradi
- Department of Physiology, School of Medicine, Iran University of Medical Science, Tehran. Iran
| | - Bahareh Pourjabbar
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Science, Tehran. Iran
| | - Reza Roozafzoon
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Science, Tehran. Iran
| | | | - Saeed Heidari Keshel
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Science, Tehran. Iran
| |
Collapse
|
37
|
Anti-Inflammatory Fibronectin-AgNP for Regulation of Biological Performance and Endothelial Differentiation Ability of Mesenchymal Stem Cells. Int J Mol Sci 2021; 22:ijms22179262. [PMID: 34502171 PMCID: PMC8430779 DOI: 10.3390/ijms22179262] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/22/2021] [Accepted: 08/23/2021] [Indexed: 01/21/2023] Open
Abstract
The engineering of vascular regeneration still involves barriers that need to be conquered. In the current study, a novel nanocomposite comprising of fibronectin (denoted as FN) and a small amount of silver nanoparticles (AgNP, ~15.1, ~30.2 or ~75.5 ppm) was developed and its biological function and biocompatibility in Wharton's jelly-derived mesenchymal stem cells (MSCs) and rat models was investigated. The surface morphology as well as chemical composition for pure FN and the FN-AgNP nanocomposites incorporating various amounts of AgNP were firstly characterized by atomic force microscopy (AFM), UV-Visible spectroscopy (UV-Vis), and Fourier-transform infrared spectroscopy (FTIR). Among the nanocomposites, FN-AgNP with 30.2 ppm silver nanoparticles demonstrated the best biocompatibility as assessed through intracellular ROS production, proliferation of MSCs, and monocytes activation. The expression levels of pro-inflammatory cytokines, TNF-α, IL-1β, and IL-6, were also examined. FN-AgNP 30.2 ppm significantly inhibited pro-inflammatory cytokine expression compared to other materials, indicating superior performance of anti-immune response. Mechanistically, FN-AgNP 30.2 ppm significantly induced greater expression of vascular endothelial growth factor (VEGF) and stromal-cell derived factor-1 alpha (SDF-1α) and promoted the migration of MSCs through matrix metalloproteinase (MMP) signaling pathway. Besides, in vitro and in vivo studies indicated that FN-AgNP 30.2 ppm stimulated greater protein expressions of CD31 and von Willebrand Factor (vWF) as well as facilitated better endothelialization capacity than other materials. Furthermore, the histological tissue examination revealed the lowest capsule formation and collagen deposition in rat subcutaneous implantation of FN-AgNP 30.2 ppm. In conclusion, FN-AgNP nanocomposites may facilitate the migration and proliferation of MSCs, induce endothelial cell differentiation, and attenuate immune response. These finding also suggests that FN-AgNP may be a potential anti-inflammatory surface modification strategy for vascular biomaterials.
Collapse
|
38
|
Bornert F, Clauss F, Hua G, Idoux-Gillet Y, Keller L, Fernandez De Grado G, Offner D, Smaida R, Wagner Q, Fioretti F, Kuchler-Bopp S, Schulz G, Wenzel W, Gentile L, Risser L, Müller B, Huck O, Benkirane-Jessel N. Mechanistic Illustration: How Newly-Formed Blood Vessels Stopped by the Mineral Blocks of Bone Substitutes Can Be Avoided by Using Innovative Combined Therapeutics. Biomedicines 2021; 9:952. [PMID: 34440156 PMCID: PMC8394928 DOI: 10.3390/biomedicines9080952] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/16/2021] [Accepted: 08/01/2021] [Indexed: 12/30/2022] Open
Abstract
One major limitation for the vascularization of bone substitutes used for filling is the presence of mineral blocks. The newly-formed blood vessels are stopped or have to circumvent the mineral blocks, resulting in inefficient delivery of oxygen and nutrients to the implant. This leads to necrosis within the implant and to poor engraftment of the bone substitute. The aim of the present study is to provide a bone substitute currently used in the clinic with suitably guided vascularization properties. This therapeutic hybrid bone filling, containing a mineral and a polymeric component, is fortified with pro-angiogenic smart nano-therapeutics that allow the release of angiogenic molecules. Our data showed that the improved vasculature within the implant promoted new bone formation and that the newly-formed bone swapped the mineral blocks of the bone substitutes much more efficiently than in non-functionalized bone substitutes. Therefore, we demonstrated that our therapeutic bone substitute is an advanced therapeutical medicinal product, with great potential to recuperate and guide vascularization that is stopped by mineral blocks, and can improve the regeneration of critical-sized bone defects. We have also elucidated the mechanism to understand how the newly-formed vessels can no longer encounter mineral blocks and pursue their course of vasculature, giving our advanced therapeutical bone filling great potential to be used in many applications, by combining filling and nano-regenerative medicine that currently fall short because of problems related to the lack of oxygen and nutrients.
Collapse
Affiliation(s)
- Fabien Bornert
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - François Clauss
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Guoqiang Hua
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Ysia Idoux-Gillet
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Laetitia Keller
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Gabriel Fernandez De Grado
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Damien Offner
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Rana Smaida
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Quentin Wagner
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Florence Fioretti
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Sabine Kuchler-Bopp
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Georg Schulz
- Biomaterials Science Center, University of Basel, Gewerbestrasse 14, CH-4123 Allschwil, Switzerland; (G.S.); (B.M.)
| | - Wolfgang Wenzel
- Institute of Nanotechnology, Karlsruhe Institute of Technology, Campus North, Building 640, DE-76131 Karlsruhe, Germany;
| | - Luca Gentile
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
| | - Laurent Risser
- Toulouse Institute of Mathematics, UMR 5219 University of Toulouse, CNRS UPS IMT, 31062 Toulouse, France;
| | - Bert Müller
- Biomaterials Science Center, University of Basel, Gewerbestrasse 14, CH-4123 Allschwil, Switzerland; (G.S.); (B.M.)
| | - Olivier Huck
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| | - Nadia Benkirane-Jessel
- INSERM (French National Institute of Health and Medical Research) UMR 1260, Regenerative Nanomedicine, CRBS, 1 Rue Eugène Boeckel, 67000 Strasbourg, France; (F.B.); (F.C.); (G.H.); (Y.I.-G.); (L.K.); (G.F.D.G.); (D.O.); (R.S.); (Q.W.); (F.F.); (S.K.-B.); (L.G.); (O.H.)
- Faculty of Dental Surgery, University of Strasbourg, University Hospital Strasbourg (HUS), 8 Rue de Sainte Elisabeth, 67000 Strasbourg, France
- Department of Pediatric Dentistry, University Hospital Strasbourg (HUS), 1 Place de l’Hôpital, 67000 Strasbourg, France
| |
Collapse
|
39
|
Basnett P, Matharu RK, Taylor CS, Illangakoon U, Dawson JI, Kanczler JM, Behbehani M, Humphrey E, Majid Q, Lukasiewicz B, Nigmatullin R, Heseltine P, Oreffo ROC, Haycock JW, Terracciano C, Harding SE, Edirisinghe M, Roy I. Harnessing Polyhydroxyalkanoates and Pressurized Gyration for Hard and Soft Tissue Engineering. ACS APPLIED MATERIALS & INTERFACES 2021; 13:32624-32639. [PMID: 34228435 DOI: 10.1021/acsami.0c19689] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Organ dysfunction is a major cause of morbidity and mortality. Transplantation is typically the only definitive cure, challenged by the lack of sufficient donor organs. Tissue engineering encompasses the development of biomaterial scaffolds to support cell attachment, proliferation, and differentiation, leading to tissue regeneration. For efficient clinical translation, the forming technology utilized must be suitable for mass production. Herein, uniaxial polyhydroxyalkanoate scaffolds manufactured by pressurized gyration, a hybrid scalable spinning technique, are successfully used in bone, nerve, and cardiovascular applications. Chorioallantoic membrane and in vivo studies provided evidence of vascularization, collagen deposition, and cellular invasion for bone tissue engineering. Highly efficient axonal outgrowth was observed in dorsal root ganglion-based 3D ex vivo models. Human induced pluripotent stem cell derived cardiomyocytes exhibited a mature cardiomyocyte phenotype with optimal calcium handling. This study confirms that engineered polyhydroxyalkanoate-based gyrospun fibers provide an exciting and unique toolbox for the development of scalable scaffolds for both hard and soft tissue regeneration.
Collapse
Affiliation(s)
- Pooja Basnett
- School of Life Sciences, University of Westminster, London W1W 6UW, U.K
| | - Rupy K Matharu
- Department of Mechanical Engineering, University College London, London WC1E 7JE, U.K
| | - Caroline S Taylor
- Department of Materials Science and Engineering, University of Sheffield, Sheffield S1 3JD, U.K
| | - Upulitha Illangakoon
- Department of Mechanical Engineering, University College London, London WC1E 7JE, U.K
| | - Jonathan I Dawson
- Centre for Human Development, Stem Cells and Regeneration, University of Southampton, Southampton SO16 6YD, U.K
| | - Janos M Kanczler
- Centre for Human Development, Stem Cells and Regeneration, University of Southampton, Southampton SO16 6YD, U.K
| | - Mehrie Behbehani
- Department of Materials Science and Engineering, University of Sheffield, Sheffield S1 3JD, U.K
| | - Eleanor Humphrey
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, U.K
| | - Qasim Majid
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, U.K
| | | | - Rinat Nigmatullin
- School of Life Sciences, University of Westminster, London W1W 6UW, U.K
| | - Phoebe Heseltine
- Department of Mechanical Engineering, University College London, London WC1E 7JE, U.K
| | - Richard O C Oreffo
- Centre for Human Development, Stem Cells and Regeneration, University of Southampton, Southampton SO16 6YD, U.K
| | - John W Haycock
- Department of Materials Science and Engineering, University of Sheffield, Sheffield S1 3JD, U.K
| | - Cesare Terracciano
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, U.K
| | - Sian E Harding
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, U.K
| | - Mohan Edirisinghe
- Department of Mechanical Engineering, University College London, London WC1E 7JE, U.K
| | - Ipsita Roy
- Department of Materials Science and Engineering, University of Sheffield, Sheffield S1 3JD, U.K
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, U.K
| |
Collapse
|
40
|
Ahmed E, Saleh T, Xu M. Recellularization of Native Tissue Derived Acellular Scaffolds with Mesenchymal Stem Cells. Cells 2021; 10:cells10071787. [PMID: 34359955 PMCID: PMC8304639 DOI: 10.3390/cells10071787] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/03/2021] [Accepted: 07/12/2021] [Indexed: 12/22/2022] Open
Abstract
The functionalization of decellularized scaffolds is still challenging because of the recellularization-related limitations, including the finding of the most optimal kind of cell(s) and the best way to control their distribution within the scaffolds to generate native mimicking tissues. That is why researchers have been encouraged to study stem cells, in particular, mesenchymal stem cells (MSCs), as alternative cells to repopulate and functionalize the scaffolds properly. MSCs could be obtained from various sources and have therapeutic effects on a wide range of inflammatory/degenerative diseases. Therefore, in this mini-review, we will discuss the benefits using of MSCs for recellularization, the factors affecting their efficiency, and the drawbacks that may need to be overcome to generate bioengineered transplantable organs.
Collapse
Affiliation(s)
- Ebtehal Ahmed
- Department of Pathology, Faculty of Veterinary Medicine, Assiut University, Assiut 71515, Egypt;
| | - Tarek Saleh
- Department of Animal Surgery, Faculty of Veterinary Medicine, Assiut University, Assiut 71515, Egypt;
| | - Meifeng Xu
- Department of Pathology and Laboratory Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
- Correspondence: or ; Tel.: +1-513-558-4725; Fax: +1-513-558-2141
| |
Collapse
|
41
|
Wang Z, Zhu X, Cong X. Spatial micro-variation of 3D hydrogel stiffness regulates the biomechanical properties of hMSCs. Biofabrication 2021; 13. [PMID: 34107453 DOI: 10.1088/1758-5090/ac0982] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/09/2021] [Indexed: 02/06/2023]
Abstract
Human mesenchymal stem cells (hMSCs) are one of the most promising candidates for cell-based therapeutic products. Nonetheless, their biomechanical phenotype afterin vitroexpansion is still unsatisfactory, for example, restricting the efficiency of microcirculation of delivered hMSCs for further cell therapies. Here, we propose a scheme using maleimide-dextran hydrogel with locally varied stiffness in microscale to modify the biomechanical properties of hMSCs in three-dimensional (3D) niches. We show that spatial micro-variation of stiffness can be controllably generated in the hydrogel with heterogeneously cross-linking via atomic force microscopy measurements. The result of 3D cell culture experiment demonstrates the hydrogels trigger the formation of multicellular spheroids, and the derived hMSCs could be rationally softened via adjustment of the stiffness variation (SV) degree. Importantly,in vitro, the hMSCs modified with the higher SV degree can pass easier through capillary-shaped micro-channels. Further, we discuss the underlying mechanics of the increased cellular elasticity by focusing on the effect of rearranged actin networks, via the proposed microscopic model of biomechanically modified cells. Overall, this work highlights the effectiveness of SV-hydrogels in reprogramming and manufacturing hMSCs with designed biomechanical properties for improved therapeutic potential.
Collapse
Affiliation(s)
- Zheng Wang
- College of Mechanical and Electrical Engineering, Hohai University, Changzhou, Jiangsu 213022, People's Republic of China
| | - Xiaolu Zhu
- College of Mechanical and Electrical Engineering, Hohai University, Changzhou, Jiangsu 213022, People's Republic of China.,Changzhou Key Laboratory of Digital Manufacture Technology, Hohai University, Changzhou, Jiangsu 213022, People's Republic of China.,Jiangsu Key Laboratory of Special Robot Technology, Hohai University, Changzhou, Jiangsu 213022, People's Republic of China
| | - Xiuli Cong
- Department of Orthopaedics, Zhejiang Hospital, No. 12 Lingyin Road, Hangzhou, Zhejiang 310013, People's Republic of China
| |
Collapse
|
42
|
Kostadinova M, Mourdjeva M. Potential of Mesenchymal Stem Cells in Anti-Cancer Therapies. Curr Stem Cell Res Ther 2021; 15:482-491. [PMID: 32148199 DOI: 10.2174/1574888x15666200310171547] [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: 08/21/2019] [Revised: 09/27/2019] [Accepted: 11/12/2019] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem/stromal cells (MSCs) are localized throughout the adult body as a small population in the stroma of the tissue concerned. In injury, tissue damage, or tumor formation, they are activated and leave their niche to migrate to the site of injury, where they release a plethora of growth factors, cytokines, and other bioactive molecules. With the accumulation of data about the interaction between MSCs and tumor cells, the dualistic role of MSCs remains unclear. However, a large number of studies have demonstrated the natural anti-tumor properties inherent in MSCs, so this is the basis for intensive research for new methods using MSCs as a tool to suppress cancer cell development. This review focuses specifically on advanced approaches in modifying MSCs to become a powerful, precision- targeted tool for killing cancer cells, but not normal healthy cells. Suppression of tumor growth by MSCs can be accomplished by inducing apoptosis or cell cycle arrest, suppressing tumor angiogenesis, or blocking mechanisms mediating metastasis. In addition, the chemosensitivity of cancer cells may be increased so that the dose of the chemotherapeutic agent used could be significantly reduced.
Collapse
Affiliation(s)
- Milena Kostadinova
- Department of Molecular Immunology, Institute of Biology and Immunology of Reproduction "Acad. Kiril Bratanov", Bulgarian Academy of Sciences, 73 Tsarigradsko Shose, 1113 Sofia, Bulgaria
| | - Milena Mourdjeva
- Department of Molecular Immunology, Institute of Biology and Immunology of Reproduction "Acad. Kiril Bratanov", Bulgarian Academy of Sciences, 73 Tsarigradsko Shose, 1113 Sofia, Bulgaria
| |
Collapse
|
43
|
Eiro N, Fraile M, Fernández-Francos S, Sánchez R, Costa LA, Vizoso FJ. Importance of the origin of mesenchymal (stem) stromal cells in cancer biology: "alliance" or "war" in intercellular signals. Cell Biosci 2021; 11:109. [PMID: 34112253 PMCID: PMC8194017 DOI: 10.1186/s13578-021-00620-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 05/31/2021] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells (MSCs) play a central role in the intercellular signaling within the tumor microenvironment (TME), exchanging signals with cancer cells and tumor stromal cells, such as cancer-associated fibroblasts and inflammatory mononuclear cells. Research attributes both pro-tumor and anti-tumor actions to MSCs; however, evidence indicates that MSCs specific effect on the tumor depends on the source of the MSCs and the type of tumor. There are consistent data proving that MSCs from reproductive tissues, such as the uterus, umbilical cord or placenta, have potent anti-tumor effects and tropism towards tumor tissues. More interestingly, products derived from MSCs, such as secretome or extracellular vesicles, seem to reproduce the effects of their parental cells, showing a potential advantage for clinical treatments by avoiding the drawbacks associated with cell therapy. Given these perspectives, it appears necessary new research to optimize the production, safety and antitumor potency of the products derived from the MSCs suitable for oncological therapies.
Collapse
Affiliation(s)
- Noemi Eiro
- Unit Research, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33290, Gijón, Asturias, Spain.
| | - Maria Fraile
- Unit Research, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33290, Gijón, Asturias, Spain
| | - Silvia Fernández-Francos
- Unit Research, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33290, Gijón, Asturias, Spain
| | - Rosario Sánchez
- Department of Surgery, Fundación Hospital de Jove, 33290, Gijón, Asturias, Spain
| | - Luis A Costa
- Unit Research, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33290, Gijón, Asturias, Spain
| | - Francisco J Vizoso
- Unit Research, Fundación Hospital de Jove, Avda. Eduardo Castro 161, 33290, Gijón, Asturias, Spain. .,Department of Surgery, Fundación Hospital de Jove, 33290, Gijón, Asturias, Spain.
| |
Collapse
|
44
|
Moskaleva EY, Zhorova ES, Semochkina YP, Shuvatova VG, Rodina AV, Saprykin VP. Mechanisms of Stimulation of the Growth of Mouse Mammary Adenocarcinoma by Mesenchymal Stem Cells. Bull Exp Biol Med 2021; 171:141-149. [PMID: 34050834 DOI: 10.1007/s10517-021-05186-4] [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: 10/09/2020] [Indexed: 10/21/2022]
Abstract
Mesenchymal stem cells from the adipose tissue (AT MSC) and the bone marrow (BM MSC) stimulated migration of melanoma B16 cells, while mammary adenocarcinoma Ca755 cells stimulated migration of mesenchymal stem cells. Mesenchymal stem cells retained these properties at late terms after γ-irradiation in vitro. Tumors that developed after injection of Ca755 cells alone and in combinations with BM MSC or AT MSC had similar histological structure corresponding to breast adenocarcinoma. Only AT MSC stimulated tumor growth, which was determined by more intensive secretion of factors stimulating proliferation of tumor cells, including chemokine CCL2. The use of AT MSC in regenerative medicine requires careful monitoring of the absence of tumors in patients.
Collapse
Affiliation(s)
- E Yu Moskaleva
- Laboratory of Cell Biology and Molecular Medicine, National Research Centre "Kurchatov Institute", Moscow, Russia.
| | - E S Zhorova
- Laboratory of Radiation Carcinogenesis and Toxicology, A. I. Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency of Russia, Moscow, Russia
| | - Yu P Semochkina
- Laboratory of Cell Biology and Molecular Medicine, National Research Centre "Kurchatov Institute", Moscow, Russia
| | - V G Shuvatova
- Laboratory of Cell Biology and Molecular Medicine, National Research Centre "Kurchatov Institute", Moscow, Russia
| | - A V Rodina
- Laboratory of Cell Biology and Molecular Medicine, National Research Centre "Kurchatov Institute", Moscow, Russia
| | - V P Saprykin
- Department of Physiology, Ecology and Medical and Biological Knowledge, Faculty of Biology and Chemistry, Moscow State Regional University, Moscow, Russia
| |
Collapse
|
45
|
Noureddini M, Bagheri-Mohammadi S. Adult Hippocampal Neurogenesis and Alzheimer's Disease: Novel Application of Mesenchymal Stem Cells and their Role in Hippocampal Neurogenesis. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2021; 10:1-10. [PMID: 34268249 PMCID: PMC8256831 DOI: 10.22088/ijmcm.bums.10.1.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 02/07/2020] [Indexed: 10/31/2022]
Abstract
The neurogenesis can occur in two regions of the adult mammalian brain throughout the lifespan: the subgranular zone of the hippocampal dentate gyrus, and the subventricular zone of the lateral ventricle. The proliferation and maturation of neural progenitor cells are tightly regulated through intrinsic and extrinsic factors. The integration of maturated cells into the circuitry of the adult hippocampus emphasizes the importance of adult hippocampal neurogenesis in learning and memory. There is a large body of evidence demonstrating that alteration in the neurogenesis process in the adult hippocampus results in an early event in the course of Alzheimer's disease (AD). In AD condition, the number and maturation of neurons declines progressively in the hippocampus. Innovative therapies are required to modulate brain homeostasis. Mesenchymal stem cells (MSCs) hold an immense potential to regulate the neurogenesis process, and are currently tested in some brain-related disorders, such as AD. Therefore, the aim of this review is to discuss the use of MSCs to regulate endogenous adult neurogenesis and their significant impact on future strategies for the treatment of AD.
Collapse
Affiliation(s)
- Mahdi Noureddini
- Department of Physiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran.,Department of Applied Cell Sciences, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Saeid Bagheri-Mohammadi
- Department of Physiology and Neurophysiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
46
|
Zhang LL, Xiong YY, Yang YJ. The Vital Roles of Mesenchymal Stem Cells and the Derived Extracellular Vesicles in Promoting Angiogenesis After Acute Myocardial Infarction. Stem Cells Dev 2021; 30:561-577. [PMID: 33752473 DOI: 10.1089/scd.2021.0006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Acute myocardial infarction (AMI) is an event of ischemic myocardial necrosis caused by acute coronary artery occlusion, which ultimately leads to a large loss of cardiomyocytes. The prerequisite of salvaging ischemic myocardium and improving cardiac function of patients is to provide adequate blood perfusion in the infarcted area. Apart from reperfusion therapy, it is also urgent and imperative to promote angiogenesis. Recently, growing evidence based on promising preclinical data indicates that mesenchymal stem cells (MSCs) can provide therapeutic effects on AMI by promoting angiogenesis. Extracellular vesicles (EVs), membrane-encapsulated vesicles with complex cargoes, including proteins, nucleic acids, and lipids, can be derived from MSCs and represent part of their functions, so EVs also possess the ability to promote angiogenesis. However, poor control of the survival and localization of MSCs hindered clinical transformation and made scientists start looking for new approaches based on MSCs. Identifying the role of MSCs and their derived EVs in promoting angiogenesis can provide a theoretical basis for improved MSC-based methods, and ultimately promote the clinical treatment of AMI. This review highlights potential proangiogenic mechanisms of transplanted MSCs and the derived EVs after AMI and summarizes the latest literature concerning the novel methods based on MSCs to maximize the angiogenesis capability.
Collapse
Affiliation(s)
- Li-Li Zhang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yu-Yan Xiong
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yue-Jin Yang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
47
|
Semenova E, Grudniak MP, Machaj EK, Bocian K, Chroscinska-Krawczyk M, Trochonowicz M, Stepaniec IM, Murzyn M, Zagorska KE, Boruczkowski D, Kolanowski TJ, Oldak T, Rozwadowska N. Mesenchymal Stromal Cells from Different Parts of Umbilical Cord: Approach to Comparison & Characteristics. Stem Cell Rev Rep 2021; 17:1780-1795. [PMID: 33860454 PMCID: PMC8553697 DOI: 10.1007/s12015-021-10157-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2021] [Indexed: 02/06/2023]
Abstract
Mesenchymal stromal/stem cells (MSCs) are a unique population of cells that play an important role in the regeneration potential of the body. MSCs exhibit a characteristic phenotype and are capable of modulating the immune response. MSCs can be isolated from various tissues such as: bone marrow, adipose tissue, placenta, umbilical cord and others. The umbilical cord as a source of MSCs, has strong advantages, such as no-risk procedure of tissue retrieval after birth and easiness of the MSCs isolation. As the umbilical cord (UC) is a complex organ and we decided to evaluate, whether the cells derived from different regions of umbilical cord show similar or distinct properties. In this study we characterized and compared MSCs from three regions of the umbilical cord: Wharton's Jelly (WJ), the perivascular space (PRV) and the umbilical membrane (UCM). The analysis was carried out in terms of morphology, phenotype, immunomodulation potential and secretome. Based on the obtained results, we were able to conclude, that MSCs derived from distinct UC regions differ in their properties. According to our result WJ-MSCs have high and stabile proliferation potential and phenotype, when compare with other MSCs and can be treated as a preferable source of cells for medical application.
Collapse
Affiliation(s)
- Ekaterina Semenova
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland
| | - Mariusz P Grudniak
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland
| | - Eugeniusz K Machaj
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland
| | - Katarzyna Bocian
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland.,Faculty of Biology, Department of Immunology, University of Warsaw, Warsaw, Poland
| | | | - Marzena Trochonowicz
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland
| | - Igor M Stepaniec
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland
| | - Magdalena Murzyn
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland.,Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Karolina E Zagorska
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland
| | - Dariusz Boruczkowski
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland
| | - Tomasz J Kolanowski
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland.,Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Tomasz Oldak
- Research and Development Department, Polish Stem Cell Bank, FamiCord Group, Ul. Jana Pawla II 29, 00-867, Warsaw, Poland.
| | | |
Collapse
|
48
|
Xu L, Willumeit-Römer R, Luthringer-Feyerabend BJC. Mesenchymal Stem Cell and Oxygen Modulate the Cocultured Endothelial Cells in the Presence of Magnesium Degradation Products. ACS APPLIED BIO MATERIALS 2021; 4:2398-2407. [DOI: 10.1021/acsabm.0c01289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Lei Xu
- Institute of Materials Research, Division for Metallic Biomaterials, Helmholtz-Zentrum Geesthacht (HZG), Geesthacht 21502, Germany
| | - Regine Willumeit-Römer
- Institute of Materials Research, Division for Metallic Biomaterials, Helmholtz-Zentrum Geesthacht (HZG), Geesthacht 21502, Germany
| | | |
Collapse
|
49
|
Sridharan R, Kelly DJ, O'Brien FJ. Substrate Stiffness Modulates the Crosstalk Between Mesenchymal Stem Cells and Macrophages. J Biomech Eng 2021; 143:031001. [PMID: 33067618 DOI: 10.1115/1.4048809] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Indexed: 07/25/2024]
Abstract
Upon implantation of a biomaterial, mesenchymal stem cells (MSCs) and macrophages contribute to the wound healing response and the regeneration cascade. Although biomaterial properties are known to direct MSC differentiation and macrophage polarization, the role of biomaterial cues, specifically stiffness, in directing the crosstalk between the two cell types is still poorly understood. This study aimed to elucidate the role of substrate stiffness in modulating the immunomodulatory properties of MSCs and to shed light on their complex interactions with macrophages when presented with diverse biomaterial stiffness cues, a situation analogous to the implant environment where multiple cell types interact with an implanted biomaterial to determine regenerative outcomes. We show that MSCs do not play an immunomodulatory role in the absence of an inflammatory stimulus. Using collagen-coated polyacrylamide gels of varying stiffness values, we demonstrate that the immunomodulatory capability of MSCs in the presence of an inflammatory stimulus is not dependent on the stiffness of the underlying substrate. Moreover, using paracrine and direct contact culture models, we show that a bidirectional crosstalk between MSCs and macrophages is necessary for promoting anti-inflammatory responses and positive immunomodulation, which is dependent on the stiffness of the underlying substrate. We finally show that direct cell-cell contact is not essential for this effect, with paracrine interactions promoting immunomodulatory interactions between MSCs and macrophages. Together, these results demonstrate that biophysical cues such as stiffness that are presented by biomaterials can be tuned to promote positive interactions between MSCs and macrophages which can in turn direct the downstream regenerative response.
Collapse
Affiliation(s)
- Rukmani Sridharan
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin 2 D02 YN77, Ireland; Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 PN40, Ireland; Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, Dublin 2, D02 PN40, Ireland
| | - Daniel J Kelly
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin 2 D02 YN77, Ireland; Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 PN40, Ireland; Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, Dublin 2, D02 PN40, Ireland
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin 2 D02 YN77, Ireland; Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 PN40, Ireland; Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, Dublin 2, D02 PN40, Ireland
| |
Collapse
|
50
|
Peng H, Chelvarajan L, Donahue R, Gottipati A, Cahall CF, Davis KA, Tripathi H, Al-Darraji A, Elsawalhy E, Dobrozsi N, Srinivasan A, Levitan BM, Kong R, Gao E, Abdel-Latif A, Berron BJ. Polymer Cell Surface Coating Enhances Mesenchymal Stem Cell Retention and Cardiac Protection. ACS APPLIED BIO MATERIALS 2021; 4:1655-1667. [PMID: 35014513 DOI: 10.1021/acsabm.0c01473] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Mesenchymal stem cell (MSC) therapy has been widely tested in clinical trials to promote healing post-myocardial infarction. However, low cell retention and the need for a large donor cell number in human studies remain a key challenge for clinical translation. Natural biomaterials such as gelatin are ideally suited as scaffolds to deliver and enhance cell engraftment after transplantation. A potential drawback of MSC encapsulation in the hydrogel is that the bulky matrix may limit their biological function and interaction with the surrounding tissue microenvironment that conveys important injury signals. To overcome this limitation, we adopted a gelatin methacrylate (gelMA) cell-coating technique that photocross-links gelatin on the individual cell surface at the nanoscale. The present study investigated the cardiac protection of gelMA coated, hypoxia preconditioned MSCs (gelMA-MSCs) in a murine myocardial infarction (MI) model. We demonstrate that the direct injection of gelMA-MSC results in significantly higher myocardial engraftment 7 days after MI compared to uncoated MSCs. GelMA-MSC further amplified MSC benefits resulting in enhanced cardioprotection as measured by cardiac function, scar size, and angiogenesis. Improved MSC cardiac retention also led to a greater cardiac immunomodulatory function after injury. Taken together, this study demonstrated the efficacy of gelMA-MSCs in treating cardiac injury with a promising potential to reduce the need for donor MSCs through enhanced myocardial engraftment.
Collapse
Affiliation(s)
- Hsuan Peng
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, Kentucky 40508, United States
| | - Lakshman Chelvarajan
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, Kentucky 40508, United States
| | - Renee Donahue
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, Kentucky 40508, United States
| | - Anuhya Gottipati
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Calvin F Cahall
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Kara A Davis
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Himi Tripathi
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, Kentucky 40508, United States
| | - Ahmed Al-Darraji
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, Kentucky 40508, United States
| | - Eman Elsawalhy
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, Kentucky 40508, United States
| | - Nicholas Dobrozsi
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, Kentucky 40508, United States
| | - Amrita Srinivasan
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, Kentucky 40508, United States
| | - Bryana M Levitan
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, Kentucky 40508, United States.,Department of Physiology, University of Kentucky, Lexington, Kentucky 40508, United States
| | - Raymond Kong
- MilliporeSigma, Seattle, Washington 98119, United States
| | - Erhe Gao
- The Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, United States
| | - Ahmed Abdel-Latif
- Gill Heart and Vascular Institute and Division of Cardiovascular Medicine, University of Kentucky and the Lexington VA Medical Center, Lexington, Kentucky 40508, United States
| | - Brad J Berron
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky 40506, United States
| |
Collapse
|