1
|
Wang Y, Liu N, Zhang X, Dai M, Zhang N, Huang G. Study on the repair function of radiation-induced salivary gland injury using human amniotic mesenchymal stem cells pre treated with hypoxia. JOURNAL OF STOMATOLOGY, ORAL AND MAXILLOFACIAL SURGERY 2025; 126:102223. [PMID: 39800062 DOI: 10.1016/j.jormas.2025.102223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/03/2025] [Indexed: 01/15/2025]
Abstract
OBJECTIVE To investigate the reparative effect of hypoxia pretreated hAMSCs on radiation-induced damage to salivary gland function in mice. METHODS hAMSCs were separated from human amniotic tissues by mechanical and enzymatic digestion methods and a 15 Gy electron beam was used to locally irradiate the neck of mouse to create a salivary gland injury model. The mouse models were randomly divided into four groups: control group, IR+PBS group, IR+Nor group and IR+HP group. RESULT Two months after hAMSCs injection, the saliva flow of mice in the IR+PBS group was significantly lower than that of the control group (P < 0.05). The saliva flow of mice in the IR+Nor group and IR+HP group were significantly increased compared to the IR+PBS group (P < 0.05). The cell apoptosis rate of the IR+PBS group was sensibly higher than that of the blank control group (P < 0.05). The cell apoptosis rates of the IR+Nor group and the IR+HP group were lower than that of the IR+PBS group. In addition, the apoptosis rate of the IR+HP group was lower than that of the IR+Nor group (P < 0.05). The changes of IOD of α-Amy in each group showed that the expression of α - Amy in the IR+PBS group was significantly lower than that in the blank control group (P < 0.05). Compared with the IR+PBS group, the IR+Nor group and the IR+HP group showed an obvious increase in the expression of α-Amy (P < 0.05). CONCLUSION Low oxygen pretreatment of hAMSCs could more effectively repair the function of radiation-induced salivary gland compared to normoxic cultivation.
Collapse
Affiliation(s)
- Yingxin Wang
- Department of Oral Maxillofacial Surgery, Suzhou Stomatological Hospital, Suzhou 215004, PR China
| | - Nana Liu
- Department of Periodontology, Suzhou Stomatological Hospital, Suzhou 215004, PR China
| | - Xin Zhang
- Department of Oral Maxillofacial Surgery, Suzhou Stomatological Hospital, Suzhou 215004, PR China
| | - Min Dai
- Department of Oral Maxillofacial Surgery, School and Hospital of Stomatology, Zunyi Medical University, Zunyi City, Guizhou Province 563099, PR China
| | - Nini Zhang
- Department of Oral Maxillofacial Surgery, School and Hospital of Stomatology, Zunyi Medical University, Zunyi City, Guizhou Province 563099, PR China.
| | - Guilin Huang
- Department of Stomatology, the Fifth Affiliated Hospital of Zunyi Medical University, Zhuhai 519090, PR China.
| |
Collapse
|
2
|
Kang D, Kim T, Choi GE, Park A, Yoon J, Yu J, Suh N. miR-29a-3p orchestrates key signaling pathways for enhanced migration of human mesenchymal stem cells. Cell Commun Signal 2024; 22:365. [PMID: 39020373 PMCID: PMC11256664 DOI: 10.1186/s12964-024-01737-0] [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: 07/04/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND The homing of human mesenchymal stem cells (hMSCs) is crucial for their therapeutic efficacy and is characterized by the orchestrated regulation of multiple signaling modules. However, the principal upstream regulators that synchronize these signaling pathways and their mechanisms during cellular migration remain largely unexplored. METHODS miR-29a-3p was exogenously expressed in either wild-type or DiGeorge syndrome critical region 8 (DGCR8) knockdown hMSCs. Multiple pathway components were analyzed using Western blotting, immunohistochemistry, and real-time quantitative PCR. hMSC migration was assessed both in vitro and in vivo through wound healing, Transwell, contraction, and in vivo migration assays. Extensive bioinformatic analyses using gene set enrichment analysis and Ingenuity pathway analysis identified enriched pathways, upstream regulators, and downstream targets. RESULTS The global depletion of microRNAs (miRNAs) due to DGCR8 gene silencing, a critical component of miRNA biogenesis, significantly impaired hMSC migration. The bioinformatics analysis identified miR-29a-3p as a pivotal upstream regulator. Its overexpression in DGCR8-knockdown hMSCs markedly improved their migration capabilities. Our data demonstrate that miR-29a-3p enhances cell migration by directly inhibiting two key phosphatases: protein tyrosine phosphatase receptor type kappa (PTPRK) and phosphatase and tensin homolog (PTEN). The ectopic expression of miR-29a-3p stabilized the polarization of the Golgi apparatus and actin cytoskeleton during wound healing. It also altered actomyosin contractility and cellular traction forces by changing the distribution and phosphorylation of myosin light chain 2. Additionally, it regulated focal adhesions by modulating the levels of PTPRK and paxillin. In immunocompromised mice, the migration of hMSCs overexpressing miR-29a-3p toward a chemoattractant significantly increased. CONCLUSIONS Our findings identify miR-29a-3p as a key upstream regulator that governs hMSC migration. Specifically, it was found to modulate principal signaling pathways, including polarization, actin cytoskeleton, contractility, and adhesion, both in vitro and in vivo, thereby reinforcing migration regulatory circuits.
Collapse
Affiliation(s)
- Dayeon Kang
- Department of Medical Sciences, General Graduate School, Soon Chun Hyang University, Asan, 31538, Republic of Korea
- Department of Pharmaceutical Engineering, College of Medical Sciences, Soon Chun Hyang University, Asan, 31538, Republic of Korea
| | - Taehwan Kim
- Department of Medical Sciences, General Graduate School, Soon Chun Hyang University, Asan, 31538, Republic of Korea
| | - Ga-Eun Choi
- Department of Medical Sciences, General Graduate School, Soon Chun Hyang University, Asan, 31538, Republic of Korea
| | - Arum Park
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Jin Yoon
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Jinho Yu
- Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Nayoung Suh
- Department of Medical Sciences, General Graduate School, Soon Chun Hyang University, Asan, 31538, Republic of Korea.
- Department of Pharmaceutical Engineering, College of Medical Sciences, Soon Chun Hyang University, Asan, 31538, Republic of Korea.
| |
Collapse
|
3
|
Moghimi V, Rahvarian J, Esmaeilzadeh Z, Mohammad-Pour N, Babaki D, Sadeghifar F, Esfehani RJ, Bidkhori HR, Roshan NM, Momeni-Moghaddam M, Naderi-Meshkin H. Adipose-derived human mesenchymal stem cells seeded on denuded or stromal sides of the amniotic membrane improve angiogenesis and collagen remodeling and accelerate healing of the full-thickness wound. Acta Histochem 2023; 125:152027. [PMID: 37062121 DOI: 10.1016/j.acthis.2023.152027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 04/06/2023] [Accepted: 04/08/2023] [Indexed: 04/18/2023]
Abstract
Several strategies have been proposed to enhance wound healing results. Along with other forms of wound dressing, the human amniotic membrane (HAM) has long been regarded as a biological wound dressing that decreases infection and enhances healing. This study investigates the feasibility and effectiveness of wound healing using decellularized HAM (dAM) and stromal HAM (sAM) in combination with adipose-derived human mesenchymal stem cells (AdMSCs). The dAM and sAM sides of HAM were employed as wound dressing scaffolds, and AdMSCs were seeded on top of either dAM or sAM. Sixty healthy Wistar rats were randomly divided into three groups: untreated wound, dAM/AdMSCs group, and sAM/AdMSCs group. The gene expression of VEGF and COL-I was measured in vitro. Wound healing was examined after wounding on days 3, 7, 14, and 21. The expression level of VEGF was significantly higher in sAM/AdMSCs than dAM/AdMSCs (P ≤ 0.05), but there was no significant difference in COL-I expression (P ≥ 0.05). In vivo research revealed that on day 14, wounds treated with sAM/AdMSCs had more vascularization than wounds treated with dAM/AdMSCs (P ≤ 0.01) and untreated wound groups on days 7 (P ≤ 0.05) and 14 (P ≤ 0.0001), respectively. On days 14 (P < 0.05 for sAM/AdMSCs, P < 0.01 for dAM/AdMSCs), and 21 (P < 0.05 for sAM/AdMSCs, P < 0.01 for dAM/AdMSCs), the collagen deposition in the wound bed was significantly thicker in the sAM/AdMSCs and dAM/AdMSCs groups compared to untreated wounds. The study demonstrated that the combination of sAM and AdMSCs promotes wound healing by enhancing angiogenesis and collagen remodeling.
Collapse
Affiliation(s)
- Vahid Moghimi
- Stem Cells and Regenerative Medicine Department, Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran; Department of Biology, Faculty of Science, Hakim Sabzevari University, Sabzevar, Iran
| | - Jeiran Rahvarian
- Stem Cells and Regenerative Medicine Department, Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran; Department of Clinical Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Zohreh Esmaeilzadeh
- Stem Cells and Regenerative Medicine Department, Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Najmeh Mohammad-Pour
- Stem Cells and Regenerative Medicine Department, Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran; Department of Biology, Faculty of Science, Hakim Sabzevari University, Sabzevar, Iran
| | - Danial Babaki
- Curriculum in Genetics and Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Fatemeh Sadeghifar
- Department of Biology, Faculty of Science, Hakim Sabzevari University, Sabzevar, Iran
| | - Reza Jafarzadeh Esfehani
- Stem Cells and Regenerative Medicine Department, Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran; 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 Department, Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran; Blood Borne Infections Research Center, Academic Center for Education, Culture and Research (ACECR)- Khorasan Razavi, Mashhad, Iran
| | | | | | - Hojjat Naderi-Meshkin
- Stem Cells and Regenerative Medicine Department, Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran; Wellcome-Wolfson Institute for Experimental Medicine, Belfast, UK.
| |
Collapse
|
4
|
To Explore the Stem Cells Homing to GBM: The Rise to the Occasion. Biomedicines 2022; 10:biomedicines10050986. [PMID: 35625723 PMCID: PMC9138893 DOI: 10.3390/biomedicines10050986] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/20/2022] [Accepted: 04/20/2022] [Indexed: 12/13/2022] Open
Abstract
Multiple efforts are currently underway to develop targeted therapeutic deliveries to the site of glioblastoma progression. The use of carriers represents advancement in the delivery of various therapeutic agents as a new approach in neuro-oncology. Mesenchymal stem cells (MSCs) and neural stem cells (NSCs) are used because of their capability in migrating and delivering therapeutic payloads to tumors. Two of the main properties that carrier cells should possess are their ability to specifically migrate from the bloodstream and low immunogenicity. In this article, we also compared the morphological and molecular features of each type of stem cell that underlie their migration capacity to glioblastoma. Thus, the major focus of the current review is on proteins and lipid molecules that are released by GBM to attract stem cells.
Collapse
|
5
|
Modifying strategies for SDF-1/CXCR4 interaction during mesenchymal stem cell transplantation. Gen Thorac Cardiovasc Surg 2021; 70:1-10. [PMID: 34510332 PMCID: PMC8732940 DOI: 10.1007/s11748-021-01696-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 09/04/2021] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cell (MSC) transplantation is regarded as a promising candidate for the treatment of ischaemic heart disease. The major hurdles for successful clinical translation of MSC therapy are poor survival, retention, and engraftment in the infarcted heart. Stromal cell-derived factor-1/chemokine receptor 4 (SDF-1/CXCR4) constitutes one of the most efficient chemokine/chemokine receptor pairs regarding cell homing. In this review, we mainly focused on previous studies on how to regulate the SDF-1/CXCR4 interaction through various priming strategies to maximize the efficacy of mesenchymal stem cell transplantation on ischaemic hearts or to facilitate the required effects. The strengthened measures for enhancing the therapeutic efficacy of the SDF-1/CXCR4 interaction for mesenchymal stem cell transplantation included the combination of chemokines and cytokines, hormones and drugs, biomaterials, gene engineering, and hypoxia. The priming strategies on recipients for stem cell transplantation included ischaemic conditioning and device techniques.
Collapse
|
6
|
Zhu GQ, Jeon SH, Lee KW, Cho HJ, Ha US, Hong SH, Lee JY, Kwon EB, Kim HJ, Lee SM, Kim HY, Kim SW, Bae WJ. Engineered Stem Cells Improve Neurogenic Bladder by Overexpressing SDF-1 in a Pelvic Nerve Injury Rat Model. Cell Transplant 2021; 29:963689720902466. [PMID: 32067480 PMCID: PMC7444235 DOI: 10.1177/0963689720902466] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
There is still a lack of sufficient research on the mechanism behind neurogenic
bladder (NB) treatment. The aim of this study was to explore the effect of
overexpressed stromal cell-derived factor-1 (SDF-1) secreted by engineered
immortalized mesenchymal stem cells (imMSCs) on the NB. In this study, primary
bone marrow mesenchymal stem cells (BM-MSCs) were transfected into immortalized
upregulated SDF-1-engineered BM-MSCs (imMSCs/eSDF-1+) or immortalized normal SDF-1-engineered BM-MSCs
(imMSCs/eSDF-1−). NB rats induced by bilateral pelvic nerve (PN)
transection were treated with imMSCs/eSDF-1+, imMSCs/eSDF-1−, or sham. After a 4-week treatment, the bladder function was assessed by
cystometry and voiding pattern analysis. The PN and bladder tissues were
evaluated via immunostaining and western blotting analysis. We found that imMSCs/eSDF-1+ expressed higher levels of SDF-1 in vitro and in vivo. The treatment of imMSCs/eSDF-1+ improved NB and evidently stimulated the recovery of bladder wall in NB
rats. The recovery of injured nerve was more effective in the NB+imMSCs/eSDF-1+ group than in other groups. High SDF-1 expression improved the levels of
vascular endothelial growth factor and basic fibroblast growth factor. Apoptosis
was decreased after imMSCs injection, and was detected rarely in the NB+imMSCs/eSDF-1+ group. Injection of imMSCs boosted the expression of neuronal nitric
oxide synthase, p-AKT, and p-ERK in the NB+imMSCs/eSDF-1+ group than in other groups. Our findings demonstrated that overexpression
of SDF-1 induced additional MSC homing to the injured tissue, which improved the
NB by accelerating the restoration of injured nerve in a rat model.
Collapse
Affiliation(s)
- Guan Qun Zhu
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Catholic Integrative Medicine Research Institute, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Hwan Jeon
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Catholic Integrative Medicine Research Institute, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kyu Won Lee
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyuk Jin Cho
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - U-Syn Ha
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sung-Hoo Hong
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ji Youl Lee
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Bi Kwon
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Catholic Integrative Medicine Research Institute, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyo-Jin Kim
- Department of Stem Cell Therapy, SL BIGEN, Seongnam, Republic of Korea
| | - Soon Min Lee
- Department of Stem Cell Therapy, SL BIGEN, Seongnam, Republic of Korea
| | - Hey-Yon Kim
- Department of Stem Cell Therapy, SL BIGEN, Seongnam, Republic of Korea
| | - Sea Woong Kim
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Catholic Integrative Medicine Research Institute, The Catholic University of Korea, Seoul, Republic of Korea
| | - Woong Jin Bae
- Department of Urology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Catholic Integrative Medicine Research Institute, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
7
|
Fadera S, Cheng NC, Young TH, Lee IC. In vitro study of SDF-1α-loaded injectable and thermally responsive hydrogels for adipose stem cell therapy by SDF-1/CXCR4 axis. J Mater Chem B 2020; 8:10360-10372. [PMID: 33108417 DOI: 10.1039/d0tb01961e] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Stem cell-based approaches have become a promising therapeutic strategy for treating ischemic diseases. The aim of this study was to develop injectable hydrogel systems for the local release of stromal cell-derived factor-1α (SDF-1α) to recruit adipose stem cells (ASCs) that express CXCR4 to achieve stem cell therapy and therapeutic angiogenesis. Thermoresponsive and injectable chitosan (CS)/β-glycerophosphate disodium salt pentahydrate (βGP) hydrogels with different concentrations of hyaluronic acid (HA) were designed and fabricated to achieve local and sustained release of SDF-1α for ASC recruitment. Herein, the material structures, physical properties, gelation temperature, and gelation time of hydrogels with different compositions were determined. The incorporation of 0.9% HA in CS-based hydrogels not only enhanced the gelation time but also increased the strength of the hydrogels. In addition, the results revealed that the thermoresponsive and injectable CS/βGP/HA hydrogels showed good biocompatibility. In addition, the in vitro release profiles showed that the hydrogels achieved sustained release of SDF-1α over 7 days and enhanced ASC migration. The results revealed that the hydrogels with HA enhanced the sustained release effect compared with the hydrogel without HA, indicating that the HA content regulated the physical and release properties of the injectable hydrogels. Therefore, thermoresponsive and injectable CS/βGP/HA hydrogels may provide an alternative for treating ischemic diseases via SDF-1/CXCR4 axis for ASC recruitment and retention.
Collapse
Affiliation(s)
- Siaka Fadera
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| | - Nai-Chen Cheng
- Department of Surgery, National Taiwan University Hospital and College of Medicine, 7 Chung-Shan S Rd, Taipei 100, Taiwan
| | - Tai-Horng Young
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, 1 Jen-Ai Rd, Taipei 100, Taiwan.
| | - I-Chi Lee
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
8
|
Heirani-Tabasi A, Mirahmadi M, Mishan MA, Naderi-Meshkin H, Toosi S, Matin MM, Bidkhori HR, Bahrami AR. Comparison the effects of hypoxia-mimicking agents on migration-related signaling pathways in mesenchymal stem cells. Cell Tissue Bank 2020; 21:643-653. [PMID: 32815062 DOI: 10.1007/s10561-020-09851-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/10/2020] [Indexed: 12/17/2022]
Abstract
Adipose-derived mesenchymal stem cells (Ad-MSCs) have been designated as the promising agents for clinical applications for easy accessibility, multi-linage differentiation and immunomodulation capacity. Despite this, optimal cell delivery conditions have remained as a clinical challenge and improvement of stem cell homing to the target organs is being considered as a major strategy in cell therapy systemic injection. It has been shown that homing of mesenchymal stem cells are increased when treated with physical or chemical hypoxia-mimicking factors, however, efficiency of different agents remained to be determined. In this study, hypoxia-mimicking agents, including valproic acid (VPA), cobalt chloride (CoCl2) and deferoxamine (DFX) were examined to determine whether they are able to activate signaling molecules involved in migration of Ad-MSCs in vitro. We report that Ad-MSCs treated by DFX resulted in a significantly enhanced mRNA expression of MAPK4 (associated with MAPK signaling pathway), INPP4B (associated with Inositol polyphosphate pathway), VEGF-A and VEGF-C (associated with cytokine-cytokine receptor pathways), IL-8 and its receptor, CXCR2 (associated with IL-8 signaling pathway). While the cells treated with VPA did not show such effects and CoCl2 only upregulated VEGF-A and VEGF-C gene expression. Furthermore, results of wound-healing assays showed migration capacity of Ad-MSCs treated with DFX significantly increased 8 and 24 h of the treatment. This study provides credible evidence around DFX, which might be an effective drug for pharmacological preconditioning of Ad-MSCs to boost their homing capacity and regeneration of damaged tissues though, activation of the migration-related signaling pathways.
Collapse
Affiliation(s)
- Asieh Heirani-Tabasi
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Mahdi Mirahmadi
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Mohammad Amir Mishan
- Ocular Tissue Engineering Research Center, Research Institute for Ophthalmology and Vision Science, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hojjat Naderi-Meshkin
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Shirin Toosi
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Maryam M Matin
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran.,Cell and Molecular Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.,Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hamid Reza Bidkhori
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Ahmad Reza Bahrami
- Stem Cells and Regenerative Medicine Research Group, Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran. .,Cell and Molecular Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran. .,Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
9
|
Babajani A, Soltani P, Jamshidi E, Farjoo MH, Niknejad H. Recent Advances on Drug-Loaded Mesenchymal Stem Cells With Anti-neoplastic Agents for Targeted Treatment of Cancer. Front Bioeng Biotechnol 2020; 8:748. [PMID: 32793565 PMCID: PMC7390947 DOI: 10.3389/fbioe.2020.00748] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 06/11/2020] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs), as an undifferentiated group of adult multipotent cells, have remarkable antitumor features that bring them up as a novel choice to treat cancers. MSCs are capable of altering the behavior of cells in the tumor microenvironment, inducing an anti-inflammatory effect in tumor cells, inhibiting tumor angiogenesis, and preventing metastasis. Besides, MSCs can induce apoptosis and inhibit the proliferation of tumor cells. The ability of MSCs to be loaded with chemotherapeutic drugs and release them in the site of primary and metastatic neoplasms makes them a preferable choice as targeted drug delivery procedure. Targeted drug delivery minimizes unexpected side effects of chemotherapeutic drugs and improves clinical outcomes. This review focuses on recent advances on innate antineoplastic features of MSCs and the effect of chemotherapeutic drugs on viability, proliferation, and the regenerative capacity of various kinds of MSCs. It also discusses the efficacy and mechanisms of drug loading and releasing procedures along with in vivo and in vitro preclinical outcomes of antineoplastic effects of primed MSCs for clinical prospection.
Collapse
Affiliation(s)
- Amirhesam Babajani
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Pegah Soltani
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elham Jamshidi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Student Research Committee, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hadi Farjoo
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Liao X, Yang X, Deng H, Hao Y, Mao L, Zhang R, Liao W, Yuan M. Injectable Hydrogel-Based Nanocomposites for Cardiovascular Diseases. Front Bioeng Biotechnol 2020; 8:251. [PMID: 32296694 PMCID: PMC7136457 DOI: 10.3389/fbioe.2020.00251] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 03/11/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs), including a series of pathological disorders, severely affect millions of people all over the world. To address this issue, several potential therapies have been developed for treating CVDs, including injectable hydrogels as a minimally invasive method. However, the utilization of injectable hydrogel is a bit restricted recently owing to some limitations, such as transporting the therapeutic agent more accurately to the target site and prolonging their retention locally. This review focuses on the advances in injectable hydrogels for CVD, detailing the types of injectable hydrogels (natural or synthetic), especially that complexed with stem cells, cytokines, nano-chemical particles, exosomes, genetic material including DNA or RNA, etc. Moreover, we summarized the mainly prominent mechanism, based on which injectable hydrogel present excellent treating effect of cardiovascular repair. All in all, it is hopefully that injectable hydrogel-based nanocomposites would be a potential candidate through cardiac repair in CVDs treatment.
Collapse
Affiliation(s)
- Xiaoshan Liao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xushan Yang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Hong Deng
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yuting Hao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Lianzhi Mao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Rongjun Zhang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Wenzhen Liao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Miaomiao Yuan
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
11
|
Thyparambil NJ, Gutgesell LC, Bromet BA, Flowers LE, Greaney S, Day DE, Semon JA. Bioactive borate glass triggers phenotypic changes in adipose stem cells. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2020; 31:35. [PMID: 32206916 DOI: 10.1007/s10856-020-06366-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 02/17/2020] [Indexed: 06/10/2023]
Abstract
A bioactive borate glass, 13-93B3 (B3), has been used successfully in the clinic to treat chronic, nonhealing wounds without scarring. However, the mechanism by which B3 stimulates wound healing is poorly understood. Because adipose stem cells (ASCs) have been shown to have multiple roles in wound repair, we hypothesized that B3 triggers ASCs. In this study, we evaluate the effects of B3 on ASC survival, migration, differentiation, and protein secretion in vitro. In concentrations ≤10 mg/ml, B3 did not affect ASC viability under static conditions. B3 promoted the migration of ASCs but did not increase differentiation into bone or fat. B3 also decreased ASCs secretion of collagen I, PAI-1, MCP-1, DR6, DKK-1, angiogenin, IL-1, IGFBP-6, VEGF, and TIMP-2; increased expression of IL-1R and E-selectin; had a transient decrease in IL-6 secretion; and had a transient increase in bFGF secretion. Together, these results show that B3 alters the protein secretion of ASCs.
Collapse
Affiliation(s)
- Nathan J Thyparambil
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA
| | - Lisa C Gutgesell
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA
| | - Bradley A Bromet
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA
| | - Lauren E Flowers
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA
| | - Samantha Greaney
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA
| | - Delbert E Day
- Department of Material Science and Engineering, Missouri University of Science and Technology, Rolla, MO, USA
- Center for Biomedical Science and Engineering, Missouri University of Science and Technology, Rolla, MO, USA
| | - Julie A Semon
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA.
- Center for Biomedical Science and Engineering, Missouri University of Science and Technology, Rolla, MO, USA.
| |
Collapse
|
12
|
Thyparambil NJ, Gutgesell LC, Hurley CC, Flowers LE, Day DE, Semon JA. Adult stem cell response to doped bioactive borate glass. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2020; 31:13. [PMID: 31965357 DOI: 10.1007/s10856-019-6353-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 12/21/2019] [Indexed: 06/10/2023]
Abstract
Bioactive glasses have transformed healthcare due to their versatility. Bioactive borate glass, in particular, has shown remarkable healing properties for both hard and soft tissues. Incorporating dopants into the composition of bioactive glass helps to control mechanical properties, and it increases their usefulness for clinical applications. Using a bioactive borate glass, 13-93B3 (B3), we investigated eleven dopants on the viability and migration potential of adipose stem cells (ASCs), a therapeutic source of cells used in tissue engineering and cell therapy. Our results show that under standard cell culture conditions, only Cu-doped B3 decreased cell viability, while only Y-doped B3 attracted ASCs as it dissolved in cell culture media. Using a transwell invasion assay, priming ASCs with Co, Fe, Ga, I, Sr, or Zn-doped B3 increased their homing capacity. Because there is widespread interest in optimizing and enhancing the homing efficiency of ASCs and other therapeutic cells, we then tested if priming bone marrow mesenchymal stem cells (BMSCs) with dopants also increased their homing capacity. In the case of BMSCs, there was a significant increase in invasion when cells were primed with any of the doped-B3 glasses. This work shows that incorporating dopants into borate glasses can provide a platform for a safe and efficient method that stimulates endogenous cells and healing mechanisms.
Collapse
Affiliation(s)
- Nathan J Thyparambil
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA
| | - Lisa C Gutgesell
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA
| | - Cassandra C Hurley
- Department of Material Science and Engineering, Missouri University of Science and Technology, Rolla, MO, USA
| | - Lauren E Flowers
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA
| | - Delbert E Day
- Department of Material Science and Engineering, Missouri University of Science and Technology, Rolla, MO, USA
- Center for Biomedical Science and Engineering, Missouri University of Science and Technology, Rolla, MO, USA
| | - Julie A Semon
- Department of Biological Sciences, Missouri University of Science and Technology, Rolla, MO, USA.
- Center for Biomedical Science and Engineering, Missouri University of Science and Technology, Rolla, MO, USA.
| |
Collapse
|
13
|
Mollazadeh S, Fazly Bazzaz BS, Neshati V, de Vries AAF, Naderi-Meshkin H, Mojarad M, Neshati Z, Kerachian MA. T- Box20 inhibits osteogenic differentiation in adipose-derived human mesenchymal stem cells: the role of T- Box20 on osteogenesis. ACTA ACUST UNITED AC 2019; 26:8. [PMID: 31548928 PMCID: PMC6751895 DOI: 10.1186/s40709-019-0099-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 08/29/2019] [Indexed: 12/12/2022]
Abstract
Background Skeletal development and its cellular function are regulated by various transcription factors. The T-box (Tbx) family of transcription factors have critical roles in cellular differentiation as well as heart and limbs organogenesis. These factors possess activator and/or repressor domains to modify the expression of target genes. Despite the obvious effects of Tbx20 on heart development, its impact on bone development is still unknown. Methods To investigate the consequence by forced Tbx20 expression in the osteogenic differentiation of human mesenchymal stem cells derived from adipose tissue (Ad-MSCs), these cells were transduced with a bicistronic lentiviral vector encoding Tbx20 and an enhanced green fluorescent protein. Results Tbx20 gene delivery system suppressed the osteogenic differentiation of Ad-MSCs, as indicated by reduction in alkaline phosphatase activity and Alizarin Red S staining. Consistently, reverse transcription-polymerase chain reaction analyses showed that Tbx20 gain-of-function reduced the expression levels of osteoblast marker genes in osteo-inductive Ad-MSCs cultures. Accordingly, Tbx20 negatively affected osteogenesis through modulating expression of key factors involved in this process. Conclusion The present study suggests that Tbx20 could inhibit osteogenic differentiation in adipose-derived human mesenchymal stem cells.
Collapse
Affiliation(s)
- Samaneh Mollazadeh
- 1Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran.,2Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bibi Sedigheh Fazly Bazzaz
- 2Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,3Department of Food and Drug Control, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,4School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vajiheh Neshati
- 2Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Antoine A F de Vries
- 5Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hojjat Naderi-Meshkin
- 6Stem Cell and Regenerative Medicine Research Group, Academic Center for Education, Culture Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Majid Mojarad
- 7Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,8Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zeinab Neshati
- 9Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Mohammad Amin Kerachian
- 7Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,8Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
14
|
Mollazadeh S, Fazly Bazzaz BS, Neshati V, de Vries AAF, Naderi-Meshkin H, Mojarad M, Mirahmadi M, Neshati Z, Kerachian MA. Overexpression of MicroRNA-148b-3p stimulates osteogenesis of human bone marrow-derived mesenchymal stem cells: the role of MicroRNA-148b-3p in osteogenesis. BMC MEDICAL GENETICS 2019; 20:117. [PMID: 31262253 PMCID: PMC6604430 DOI: 10.1186/s12881-019-0854-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 06/24/2019] [Indexed: 12/23/2022]
Abstract
Background Mesenchymal stem cells (MSCs) are attractive choices in regenerative medicine and can be genetically modified to obtain better results in therapeutics. Bone development and metabolism are controlled by various factors including microRNAs (miRs) interference, which are small non-coding endogenous RNAs. Methods In the current study, the effects of forced miR-148b expression was evaluated on osteogenic activity. Human bone marrow-derived mesenchymal stem cells (BM-MSCs) were transduced with bicistronic lentiviral vector encoding hsa-miR-148b-3p or -5p and the enhanced green fluorescent protein. Fourteen days post-transduction, immunostaining as well as Western blotting were used to analyze osteogenesis. Results Overexpression of miR-148b-3p increased the osteogenic differentiation of human BM-MSCs as demonstrated by anenhancement of mineralized nodular formation and an increase in the levels of osteoblastic differentiation biomarkers, alkaline phosphatase and collagen type I. Conclusions Since lentivirally overexpressed miR-148b-3p increased osteogenic differentiation capability of BM-MSCs, this miR could be applied as a therapeutic modulator to optimize bone function. Electronic supplementary material The online version of this article (10.1186/s12881-019-0854-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Samaneh Mollazadeh
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bibi Sedigheh Fazly Bazzaz
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Food and Drug Control, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vajiheh Neshati
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Antoine A F de Vries
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Hojjat Naderi-Meshkin
- Stem Cell and Regenerative Medicine Research Group, Academic Center for Education, Culture Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Majid Mojarad
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahdi Mirahmadi
- Stem Cell and Regenerative Medicine Research Group, Academic Center for Education, Culture Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Zeinab Neshati
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Mohammad Amin Kerachian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran. .,Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
15
|
Adipose Tissue-Derived Stem Cells Have the Ability to Differentiate into Alveolar Epithelial Cells and Ameliorate Lung Injury Caused by Elastase-Induced Emphysema in Mice. Stem Cells Int 2019; 2019:5179172. [PMID: 31281377 PMCID: PMC6590553 DOI: 10.1155/2019/5179172] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/20/2019] [Accepted: 05/06/2019] [Indexed: 12/17/2022] Open
Abstract
Chronic obstructive pulmonary disease is a leading cause of mortality globally, with no effective therapy yet established. Adipose tissue-derived stem cells (ADSCs) are useful for ameliorating lung injury in animal models. However, whether ADSCs differentiate into functional cells remains uncertain, and no study has reported on the mechanism by which ADSCs improve lung functionality. Thus, in this study, we examined whether ADSCs differentiate into lung alveolar cells and are able to ameliorate lung injury caused by elastase-induced emphysema in model mice. Here, we induced ADSCs to differentiate into type 2 alveolar epithelial cells in vitro. We demonstrated that ADSCs can differentiate into type 2 alveolar epithelial cells in an elastase-induced emphysematous lung and that ADSCs improve pulmonary function of emphysema model mice, as determined with spirometry and 129Xe MRI. These data revealed a novel function for ADSCs in promoting repair of the damaged lung by direct differentiation into alveolar epithelial cells.
Collapse
|
16
|
CXCR4-Overexpressing Umbilical Cord Mesenchymal Stem Cells Enhance Protection against Radiation-Induced Lung Injury. Stem Cells Int 2019; 2019:2457082. [PMID: 30867667 PMCID: PMC6379846 DOI: 10.1155/2019/2457082] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/31/2018] [Accepted: 11/07/2018] [Indexed: 12/13/2022] Open
Abstract
Less quantity of transplanted mesenchymal stem cells (MSCs) influences the therapeutic effects on radiation-induced lung injury (RILI). Previous studies have demonstrated that MSCs overexpressing Chemokine (C-X-C motif) receptor 4 (CXCR4) could increase the quantity of transplanted cells to local tissues. In the present study, we conducted overexpressing CXCR4 human umbilical cord mesenchymal stem cell (HUMSC) therapy for RILI. C57BL mice received single dose of thoracic irradiation with 13 Gy of X-rays and then were administered saline, control HUMSCs, or CXCR4-overexpressing HUMSCs via tail vein. Transfection with CXCR4 enhanced the quantity of transplanted HUMSCs in the radiation-induced injured lung tissues. CXCR4-overexpressing HUMSCs not only improved histopathological changes but also decreased the radiation-induced expression of SDF-1, TGF-β1, α-SMA, and collagen I and inhibited the radiation-induced decreased expression of E-cadherin. Transplanted CXCR4-overexpressing HUMSCs also could express pro-SP-C, indicated adopting the feature of ATII. These finding suggests that CXCR4-overexpressing HUMSCs enhance the protection against RILI and may be a promising strategy for RILI treatment.
Collapse
|
17
|
Zhao H, Xu K, Zhu P, Wang C, Chi Q. Smart hydrogels with high tunability of stiffness as a biomimetic cell carrier. Cell Biol Int 2019; 43:84-97. [DOI: 10.1002/cbin.11091] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 12/23/2018] [Indexed: 12/23/2022]
Affiliation(s)
- Han Zhao
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences; University of Science and Technology of China; Hefei China
| | - Kang Xu
- Department of Cardiovascular Surgery; Union Hospital; Tongji Medical College, Huazhong University of Science and Technology; Wuhan China
| | - Peng Zhu
- Department of Cardiovascular Surgery; Union Hospital; Tongji Medical College, Huazhong University of Science and Technology; Wuhan China
| | - Chunli Wang
- “111 ” Project Laboratory of Biomechanics and Tissue Repair; Bioengineering College; Chongqing University; Chongqing China
| | - Qingjia Chi
- Hubei Key Laboratory of Theory and Application of Advanced Materials Mechanics; Department of Mechanics and Engineering Structure; Wuhan University of Technology; Wuhan China
| |
Collapse
|
18
|
Jiao J, Huang J, Zhang Z. Hydrogels based on chitosan in tissue regeneration: How do they work? A mini review. J Appl Polym Sci 2018. [DOI: 10.1002/app.47235] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Jiao Jiao
- Neuropsychiatric Institute; Medical School of Southeast University; Nanjing Jiangsu 210009 China
- Department of Neurology; Affiliated ZhongDa Hospital; Nanjing Jiangsu 210009 China
| | - Jinjian Huang
- Lab for Trauma and Surgical Infections, Department of Surgery; Jinling Hospital; Nanjing Jiangsu 210002 China
| | - Zhijun Zhang
- Neuropsychiatric Institute; Medical School of Southeast University; Nanjing Jiangsu 210009 China
- Department of Neurology; Affiliated ZhongDa Hospital; Nanjing Jiangsu 210009 China
| |
Collapse
|
19
|
Enhancement of the efficacy of mesenchymal stem cells in the treatment of ischemic diseases. Biomed Pharmacother 2018; 109:2022-2034. [PMID: 30551458 DOI: 10.1016/j.biopha.2018.11.068] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/17/2018] [Accepted: 11/19/2018] [Indexed: 02/05/2023] Open
Abstract
Ischemic diseases refer to a wide range of diseases caused by reduced blood flow and a subsequently deficient oxygen and nutrient supply. The pathogenesis of ischemia is multifaceted and primarily involves inflammation, oxidative stress and an apoptotic response. Over the last decade, mesenchymal stem cells (MSCs) have been widely studied as potential cell therapy agents for ischemic diseases due to their multiple favourable functions. However, the low homing and survival rates of transplanted cells have been concerns limiting for their clinical application. Recently, increasing studies have attempted to enhance the efficacy of MSCs by various strategies including genetic modification, pretreatment, combined application and biomaterial application. The purpose of this review is to summarize these creative strategies and the progress in basic and preclinical studies.
Collapse
|
20
|
Rafaiee R, Ahmadiankia N. Bone Marrow Derived Mesenchymal Stem Cells in Addiction Related Hippocampal Damages. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2018; 7:69-79. [PMID: 30276162 PMCID: PMC6148505 DOI: 10.22088/ijmcm.bums.7.2.69] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/13/2018] [Indexed: 11/30/2022]
Abstract
The brain is an important organ that controls all sensory and motor actions, memory, and emotions. Each anatomical and physiological modulation in various brain centers, results in psychological, behavioral, and sensory-motor changes. Alcohol and addictive drugs such as opioids and amphetamines have been shown to exert a great impact on brain, specifically on the hippocampus. Emerging evidence has indicated that altered hippocampal neurogenesis is associated with the pathophysiology of neuropsychological disorders including addiction. The addictive drugs impair neurogenesis and undermine the function of neural stem/progenitor cells in hippocampus. This feature was claimed to be one of the underlying mechanisms of behavioral changes in patients with addiction. As the impairment of stem cells’ function has been proven to be the underlying cause of pathologic neuroadaptations in the brain, the administration of stem cell populations has shown promising results for re-modulating of neuronal status in the brain and especially in the hippocampus. Among the different types of stem cells, bone marrow derived mesenchymal stem cells are the most proper candidates for stem cell therapies. In this review article, the recent studies on the effects of addictive drugs on brain neurogenesis, and also the promising potential effects of stem cells in curing addiction related hippocampal damages are discussed.
Collapse
Affiliation(s)
- Raheleh Rafaiee
- Addiction Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | | |
Collapse
|
21
|
Neshati V, Mollazadeh S, Fazly Bazzaz BS, Iranshahi M, Mojarrad M, Naderi-Meshkin H, Kerachian MA. Cardiogenic effects of characterized Geum urbanum extracts on adipose-derived human mesenchymal stem cells. Biochem Cell Biol 2018; 96:610-618. [DOI: 10.1139/bcb-2017-0313] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Stem cell therapy is considered as a promising treatment for cardiovascular diseases. Adipose-derived mesenchymal stem cells (ADMSCs) have the ability to undergo cardiomyogenesis. Medicinal plants are effective and safe candidates for cell differentiation. Therefore, the aim of our study was to investigate cardiogenic effects of characterized (HPLC–UV) extracts of Geum urbanum on ADMSCs of adipose tissue. The methanolic extracts of the root and aerial parts of G. urbanum were obtained and MTT assay was used for studying their cytotoxic effects. Then, cells were treated with 50 or 100 μg/mL of the extracts from root and aerial parts of G. urbanum. MTT assay showed that the extracts of G. urbanum did not have any toxic effects on ADMSCs. Immunostaining results showed increase in the expression of α-actinin and cardiac troponin I (cTnI), and quantitative real-time reverse-transcription PCR data confirmed the upregulation of ACTN, ACTC1, and TNNI3 genes in ADMSCs after treatment. According to HPLC fingerprinting, some cardiogenic effects of G. urbanum extracts are probably due to ellagic and gallic acid derivatives. Our findings indicated that G. urbanum extracts effectively upregulated some essential cardiogenic markers, which confirmed the therapeutic role of this plant as a traditional cardiac medicine.
Collapse
Affiliation(s)
- Vajiheh Neshati
- Biotechnology Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samaneh Mollazadeh
- Biotechnology Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bibi Sedigheh Fazly Bazzaz
- Biotechnology Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrdad Iranshahi
- Biotechnology Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Mojarrad
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hojjat Naderi-Meshkin
- Stem Cell and Regenerative Medicine Research Group, Academic Center for Education, Culture, Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Mohammad Amin Kerachian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
22
|
Hassanzadeh H, Matin MM, Naderi-Meshkin H, Bidkhori HR, Mirahmadi M, Raeesolmohaddeseen M, Sanjar-Moussavi N, Bahrami AR. Using paracrine effects of Ad-MSCs on keratinocyte cultivation and fabrication of epidermal sheets for improving clinical applications. Cell Tissue Bank 2018; 19:531-547. [PMID: 30105667 DOI: 10.1007/s10561-018-9702-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 05/14/2018] [Indexed: 12/13/2022]
Abstract
Recent advances in wound healing have made cell therapy a potential approach for the treatment of various types of skin defects such as trauma, burns, scars and diabetic leg ulcers. Cultured keratinocytes have been applied to burn patients since 1981. Patients with acute and chronic wounds can be treated with autologous/allograft cultured keratinocytes. There are various methods for cultivation of epidermal keratinocytes used in cell therapy. One of the important properties of an efficient cell therapy is the preservation of epidermal stem cells. Mesenchymal Stem Cells (MSCs) are major regulatory cells involved in the acceleration of wound healing via induction of cell proliferation, angiogenesis and stimulating the release of paracrine signaling molecules. Considering the beneficial effects of MSCs on wound healing, the main aim of the present study is investigating paracrine effects of Adipose-derived Mesenchymal Stem Cell (Ad-MSCs) on cultivation of keratinocytes with focusing on preservation of stem cells and their differentiation process. We further introduced a new approach for culturing isolated keratinocytes in vitro in order to generate epidermal keratinocyte sheets without using a feeder layer. To do so, Ad-MSC conditioned medium was applied as an alternative to commercial media for keratinocyte cultivation. In this study, the expression of several stem/progenitor cell (P63, K19 and K14) and differentition (K10, IVL and FLG) markers was examined using real time PCR on days 7, 14 and 21 of culture in keratinocytes in Ad-MSC conditioned medium. P63 and α6 integrin expression was also evaluated via flow cytometry. The results were compared with control group including keratinocytes cultured in EpiLife medium and our data indicated that this Ad-MSC conditioned medium is a good alternative for keratinocyte cultivation and producing epidermal sheets for therapeutic and clinical purposes. The reasons are the expression of stem cell and differentiation markers and overcoming the requirement for feeder layer which leads to a xenograft-free transplantation. Besides, this approach has low cost and is easier to perform. However, more in vitro and in vivo experiments as well as safety evaluation required before clinical applications.
Collapse
Affiliation(s)
- Halimeh Hassanzadeh
- Stem Cell and Regenerative Medicine Research Group, Iranian Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran.,Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Maryam M Matin
- Stem Cell and Regenerative Medicine Research Group, Iranian Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran. .,Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran. .,Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Hojjat Naderi-Meshkin
- Stem Cell and Regenerative Medicine Research Group, Iranian Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Hamid Reza Bidkhori
- Stem Cell and Regenerative Medicine Research Group, Iranian Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Mahdi Mirahmadi
- Stem Cell and Regenerative Medicine Research Group, Iranian Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Mahmood Raeesolmohaddeseen
- Stem Cell and Regenerative Medicine Research Group, Iranian Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | | | - Ahmad Reza Bahrami
- Stem Cell and Regenerative Medicine Research Group, Iranian Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran. .,Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran. .,Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
23
|
Naderi-Meshkin H, Ahmadiankia N. Cancer metastasis versus stem cell homing: Role of platelets. J Cell Physiol 2018; 233:9167-9178. [PMID: 30105746 DOI: 10.1002/jcp.26937] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 06/12/2018] [Indexed: 12/12/2022]
Abstract
One of the major obstacles in achieving a successful stem cell therapy is insufficient homing of transplanted cells. To overcome this obstacle, understanding the underlying mechanisms of stem cell homing is of obvious importance. Central to this review is the concept that cancer metastasis can be viewed as a role model to build up a comprehensive concept of stem cell homing. In this novel perspective, the prosurvival choices of the cancerous cells in the bloodstream, their arrest, extravasation, and proliferation at the secondary site can be exploited in favor of targeted stem cell homing. To date, tumor cells have been found to employ a wide variety of strategies to promote metastasis. One of these strategies is through their ability to activate platelets and subsequently activated platelets serve cancer cell survival and metastasis. Accordingly, in the first part of this review the roles of platelets in cancer metastasis as well as stem cell homing are discussed. Next, we provide some lessons learned from cancer metastasis in favor of developing strategies for improvement of stem cell homing with emphasis on the role of platelets. Based on direct or indirect evidence from metastasis, strategies such as manipulation of stem cells to enhance interaction with platelets, preconditioning-pretreatment of stem cells with platelets in vitro, and coinjection of both stem cells and platelets are proposed to improve stem cell homing.
Collapse
Affiliation(s)
- Hojjat Naderi-Meshkin
- Stem Cells and Regenerative Medicine Research Group, Iranian Academic Center for Education, Culture Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Naghmeh Ahmadiankia
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.,Cancer Prevention Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| |
Collapse
|
24
|
Ranganath SH. Bioengineered cellular and cell membrane-derived vehicles for actively targeted drug delivery: So near and yet so far. Adv Drug Deliv Rev 2018; 132:57-80. [PMID: 29935987 DOI: 10.1016/j.addr.2018.06.012] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/31/2018] [Accepted: 06/18/2018] [Indexed: 12/16/2022]
Abstract
Cellular carriers for drug delivery are attractive alternatives to synthetic nanoparticles owing to their innate homing/targeting abilities. Here, we review molecular interactions involved in the homing of Mesenchymal stem cells (MSCs) and other cell types to understand the process of designing and engineering highly efficient, actively targeting cellular vehicles. In addition, we comprehensively discuss various genetic and non-genetic strategies and propose futuristic approaches of engineering MSC homing using micro/nanotechnology and high throughput small molecule screening. Most of the targeting abilities of a cell come from its plasma membrane, thus, efforts to harness cell membranes as drug delivery vehicles are gaining importance and are highlighted here. We also recognize and report the lack of detailed characterization of cell membranes in terms of safety, structural integrity, targeting functionality, and drug transport. Finally, we provide insights on future development of bioengineered cellular and cell membrane-derived vesicles for successful clinical translation.
Collapse
Affiliation(s)
- Sudhir H Ranganath
- Bio-INvENT Lab, Department of Chemical Engineering, Siddaganga Institute of Technology, B.H. Road, Tumakuru, 572103, Karnataka, India.
| |
Collapse
|
25
|
Zheng XB, He XW, Zhang LJ, Qin HB, Lin XT, Liu XH, Zhou C, Liu HS, Hu T, Cheng HC, He XS, Wu XR, Chen YF, Ke J, Wu XJ, Lan P. Bone marrow-derived CXCR4-overexpressing MSCs display increased homing to intestine and ameliorate colitis-associated tumorigenesis in mice. Gastroenterol Rep (Oxf) 2018; 7:127-138. [PMID: 30976426 PMCID: PMC6454852 DOI: 10.1093/gastro/goy017] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 01/28/2018] [Accepted: 02/08/2018] [Indexed: 12/13/2022] Open
Abstract
Background and Objective Increasing interest has developed in the therapeutic potential of bone marrow-derived mesenchymal stem cells (MSCs) for the treatment of inflammatory bowel disease (IBD) and IBD-induced cancer. However, whether MSCs have the ability to suppress or promote tumor development remains controversial. The stromal cell-derived factor 1 (SDF-1)/C-X-C chemokine receptor type 4 (CXCR4) axis is well known to play a critical role in the homing of MSCs. In this study, we aimed to evaluate the role of CXCR4-overexpressing MSCs on the tumorigenesis of IBD. Methods MSCs were transduced with lentiviral vector carrying either CXCR4 or green fluorescent protein (GFP). Chemotaxis and invasion assays were used to detect CXCR4 expression. A mouse model of colitis-associated tumorigenesis was established using azoxymethane and dextran sulfate sodium (DSS). The mice were divided into three groups and then injected with phosphate buffer saline (PBS), MSC-GFP or MSC-CXCR4. Results Compared with the mice injected with MSC-GFP, the mice injected with MSC-CXCR4 showed relieved weight loss, longer colons, lower tumor numbers and decreased tumor load; expression of pro-inflammatory cytokines decreased, and signal transducer and activator of transcription 3 (STAT3) phosphorylation level in colon tissue was down-regulated. Conclusion CXCR4-overexpressing MSCs exhibited effective anti-tumor function, which may be associated with enhanced homing to inflamed intestinal tissues.
Collapse
Affiliation(s)
- Xiao-Bin Zheng
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Xiao-Wen He
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Long-Juan Zhang
- Laboratory of Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Hua-Bo Qin
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Xu-Tao Lin
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Xuan-Hui Liu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Chi Zhou
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Hua-Shan Liu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Tuo Hu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Hai-Chun Cheng
- Department of Gastrointestinal Surgery, Shenzhen Baoan Shajing People's Hospital, Guangzhou Medical University, Shenzhen, Guangdong, P.R. China
| | - Xiao-Sheng He
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Xian-Rui Wu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Yu-Feng Chen
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Jia Ke
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Xiao-Jian Wu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Ping Lan
- Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| |
Collapse
|
26
|
Neshati V, Mollazadeh S, Fazly Bazzaz BS, de Vries AA, Mojarrad M, Naderi-Meshkin H, Neshati Z, Kerachian MA. Cardiomyogenic differentiation of human adipose-derived mesenchymal stem cells transduced with Tbx20-encoding lentiviral vectors. J Cell Biochem 2018; 119:6146-6153. [PMID: 29637615 DOI: 10.1002/jcb.26818] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 02/28/2018] [Indexed: 12/29/2022]
Abstract
Ischemic heart disease often results in myocardial infarction and is the leading cause of mortality and morbidity worldwide. Improvement in the function of infarcted myocardium is a main purpose of cardiac regenerative medicine. One possible way to reach this goal is via stem cell therapy. Mesenchymal stem cells (MSCs) are multipotent stromal cells that can differentiate into a variety of cell types but display limited cardiomyogenic differentiation potential. Members of the T-box family of transcription factors including Tbx20 play important roles in heart development and cardiomyocyte homeostasis. Therefore, in the current study, we investigated the potential of Tbx20 to enhance the cardiomyogenic differentiation of human adipose-derived MSCs (ADMSCs). Human ADMSCs were transduced with a bicistronic lentiviral vector encoding Tbx20 (murine) and the enhanced green fluorescent protein (eGFP) and analyzed 7 and 14 days post transduction. Transduction of human ADMSCs with this lentiviral vector increased the expression of the cardiomyogenic differentiation markers ACTN1, TNNI3, ACTC1, NKX2.5, TBX20 (human), and GATA4 as revealed by RT-qPCR. Consistently, immunocytological results showed elevated expression of α-actinin and cardiac troponin I in these cells in comparison to the cells transduced with control lentiviral particles coding for eGFP alone. Accordingly, forced expression of Tbx20 exerts cardiomyogenic effects on human ADMSCs by increasing the expression of cardiomyogenic differentiation markers at the RNA and protein level.
Collapse
Affiliation(s)
- Vajiheh Neshati
- Biotechnology Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samaneh Mollazadeh
- Biotechnology Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Antoine Af de Vries
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Majid Mojarrad
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hojjat Naderi-Meshkin
- Stem Cell and Regenerative Medicine Research Department, Iranian Academic Center for Education, Culture Research (ACECR), Mashhad Branch, Mashhad, Iran
| | - Zeinab Neshati
- Department of Biology, Faculty of Sciences, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Mohammad Amin Kerachian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
27
|
Hajinejad M, Pasbakhsh P, Omidi A, Mortezaee K, Nekoonam S, Mahmoudi R, Kashani IR. Resveratrol pretreatment enhanced homing of SDF-1α-preconditioned bone marrow-derived mesenchymal stem cells in a rat model of liver cirrhosis. J Cell Biochem 2018; 119:2939-2950. [PMID: 29130552 DOI: 10.1002/jcb.26500] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/09/2017] [Indexed: 02/06/2023]
Abstract
Stromal cell-derived factor-1α (SDF-1α) has been known to implicate in homing of MSCs, and resveratrol has been reported to have a positive influence on SDF-1 level in the site of injury. In this study, a combined strategy was applied to evaluate bone marrow-derived MSCs (BMSCs) homing to the rat model of liver cirrhosis induced by common bile duct ligation (CBDL): (1) pretreatment delivery of resveratrol into the cirrhotic liver, and (2) transplantation of ex vivo BMSC preconditioning with SDF-1α. BMSCs were preconditioned with 10 ng/µL SDF-1α for 1 h and then labeled with the CM-Dil. Cirrhosis was induced by CBDL. Animals received intraperitoneal injection of resveratrol for 7 days, started on day 28 of CBDL post-operative. On day 36 post-operative, 1 × 106 of SDF-1α-preconditioned BMSCs was injected via caudal vein. Animals were sacrificed at 72 h post-cell transplantation. Immunofluorescence and flow cytometry assessments showed that the BMSC+SDF+RV group had an increased rate of homing into the liver, but it had a decreased rate of homing into the lung and spleen, as compared with the other groups (P < 0.05). The BMSC+SDF+RV group showed high protein expression of SIRT1, but low protein expression of p53 in the liver (P < 0.05 vs other groups). CXCR4 and matrix metalloproteinase (MMP)-9 highly expressed in SDF-1α-preconditioned BMSCs in vitro, and that AKTs and CXCL12 expressed in injured liver undergoing resveratrol injection. Our findings suggest that reseveratrol pretreatment prior to SDF-1α preconditioning could be a promising strategy for designing cell-based therapies for liver cirrhosis.
Collapse
Affiliation(s)
- Mehrdad Hajinejad
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parichehr Pasbakhsh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ameneh Omidi
- Department of Anatomy, Medical Sciences Faculty, Tarbiat Modares University, Tehran, Iran
| | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Saied Nekoonam
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Mahmoudi
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Iraj Ragerdi Kashani
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Wu T, Yang L, Chen Y, Ni Y, Jiang J, Zhang W, Zhou Q, Zheng X, Wang Q, Fu Z, Li H. Pilose antler polypeptides ameliorates hypoxic-ischemic encephalopathy by activated neurotrophic factors and SDF1/CXCR4 axis in rats. Acta Biochim Biophys Sin (Shanghai) 2018; 50:254-262. [PMID: 29385398 DOI: 10.1093/abbs/gmy005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Indexed: 12/16/2022] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a complex condition which is associated with high mortality and morbidity. However, few promising treatments for HIE exist. In the present study, the central objective was to identify the therapeutic effect of pilose antler polypeptides (PAP) on HIE in rats. Sprague-Dawley (SD) rats (14 days old) were used and divided into three groups, including control group, hypoxic-ischemia (HI) group and PAP group. After 21 days of treatment, locomotor activity was improved in PAP-treated rats, brain atrophy was decreased and cerebral edema was mitigated to some extent. Real-time quantitative polymerase chain reaction (RT-qPCR) analysis indicated that PAP administration decreased the expressions of inflammatory cytokines and apoptosis genes in hippocampus compared with HI group. Furthermore, the mRNA expressions of genes related to neurotrophic factors were significantly increased in the hippocampus. In addition, the expressions of oxidative stress markers were all down-regulated after PAP administration. Moreover, PAP up-regulated both the mRNA and protein levels of SDF1 and CXCR4, which may activate the SDF1/CXCR4 axis to moderate brain injury. These results suggest that PAP may be potentially used in the treatment of HIE.
Collapse
Affiliation(s)
- Tao Wu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Luna Yang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yan Chen
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yinhua Ni
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Jianguo Jiang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Wanjing Zhang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Qianchen Zhou
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Xiaojun Zheng
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Qi Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Haifeng Li
- Children’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
29
|
Heirani-Tabasi A, Naderi-Meshkin H, Matin MM, Mirahmadi M, Shahriyari M, Ahmadiankia N, Sanjar Moussavi N, Bidkhori HR, Raeesolmohaddeseen M, Bahrami AR. Augmented migration of mesenchymal stem cells correlates with the subsidiary CXCR4 variant. Cell Adh Migr 2018; 12:118-126. [PMID: 29466916 DOI: 10.1080/19336918.2016.1243643] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Use of mesenchymal stem cells (MSCs) has been introduced as a promising tool, for structural and functional recovery of damaged tissues/organs. Studies have indicated that interactions between chemokine receptors and their ligands have a critical role in homing of MSCs to the site of injury. Although CXCR4 variants have been characterized, the exact role of each transcript in homing has remained unclear. In this study, cells were pretreated with various hypoxia-mimicking compounds (valproic acid, cobalt-chloride, and deferoxamine mesylate). Results indicated that both variants of CXCR4 were overexpressed after 24 hours of treatments and their expression could cooperatively induce and promote the cell migration. Moreover, deferoxamine mesylate was more effective in overexpression of variant A (lo), which resulted in higher level of CXCR4 protein and the highest rate of migration of the cells. In conclusion, our findings may have important potential implications in clinical applications, reinforcing the concept that manipulating the expression of specific CXCR4 variants may increase migration of MSCs.
Collapse
Affiliation(s)
- Asieh Heirani-Tabasi
- a Department of Biology , Faculty of Science, Ferdowsi University of Mashhad , Mashhad , Iran.,b Stem Cells and Regenerative Medicine Research Group , Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch , Mashhad , Iran
| | - Hojjat Naderi-Meshkin
- b Stem Cells and Regenerative Medicine Research Group , Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch , Mashhad , Iran
| | - Maryam M Matin
- a Department of Biology , Faculty of Science, Ferdowsi University of Mashhad , Mashhad , Iran.,b Stem Cells and Regenerative Medicine Research Group , Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch , Mashhad , Iran.,c Cell and Molecular Biotechnology Research Group , Institute of Biotechnology, Ferdowsi University of Mashhad , Mashhad , Iran
| | - Mahdi Mirahmadi
- b Stem Cells and Regenerative Medicine Research Group , Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch , Mashhad , Iran
| | - Mina Shahriyari
- b Stem Cells and Regenerative Medicine Research Group , Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch , Mashhad , Iran
| | | | - Nasser Sanjar Moussavi
- e Department of Surgery , Faculty of Medicine, Islamic Azad University-Mashhad Branch , Iran
| | - Hamid Reza Bidkhori
- b Stem Cells and Regenerative Medicine Research Group , Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch , Mashhad , Iran
| | - Mahmood Raeesolmohaddeseen
- b Stem Cells and Regenerative Medicine Research Group , Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch , Mashhad , Iran
| | - Ahmad Reza Bahrami
- a Department of Biology , Faculty of Science, Ferdowsi University of Mashhad , Mashhad , Iran.,b Stem Cells and Regenerative Medicine Research Group , Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch , Mashhad , Iran.,c Cell and Molecular Biotechnology Research Group , Institute of Biotechnology, Ferdowsi University of Mashhad , Mashhad , Iran
| |
Collapse
|
30
|
Olaru AM, Marin L, Morariu S, Pricope G, Pinteala M, Tartau-Mititelu L. Biocompatible chitosan based hydrogels for potential application in local tumour therapy. Carbohydr Polym 2018; 179:59-70. [DOI: 10.1016/j.carbpol.2017.09.066] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/04/2017] [Accepted: 09/21/2017] [Indexed: 01/02/2023]
|
31
|
Ritz U, Gerke R, Götz H, Stein S, Rommens PM. A New Bone Substitute Developed from 3D-Prints of Polylactide (PLA) Loaded with Collagen I: An In Vitro Study. Int J Mol Sci 2017; 18:E2569. [PMID: 29186036 PMCID: PMC5751172 DOI: 10.3390/ijms18122569] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 11/20/2017] [Accepted: 11/27/2017] [Indexed: 01/04/2023] Open
Abstract
Although a lot of research has been performed, large segmental bone defects caused by trauma, infection, bone tumors or revision surgeries still represent big challenges for trauma surgeons. New and innovative bone substitutes are needed. Three-dimensional (3D) printing is a novel procedure to create 3D porous scaffolds that can be used for bone tissue engineering. In the present study, solid discs as well as porous cage-like 3D prints made of polylactide (PLA) are coated or filled with collagen, respectively, and tested for biocompatibility and endotoxin contamination. Microscopic analyses as well as proliferation assays were performed using various cell types on PLA discs. Stromal-derived factor (SDF-1) release from cages filled with collagen was analyzed and the effect on endothelial cells tested. This study confirms the biocompatibility of PLA and demonstrates an endotoxin contamination clearly below the FDA (Food and Drug Administration) limit. Cells of various cell types (osteoblasts, osteoblast-like cells, fibroblasts and endothelial cells) grow, spread and proliferate on PLA-printed discs. PLA cages loaded with SDF-1 collagen display a steady SDF-1 release, support cell growth of endothelial cells and induce neo-vessel formation. These results demonstrate the potential for PLA scaffolds printed with an inexpensive desktop printer in medical applications, for example, in bone tissue engineering.
Collapse
Affiliation(s)
- Ulrike Ritz
- Department of Orthopaedics and Traumatology, BiomaTiCS, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany.
| | - Rebekka Gerke
- Department of Orthopaedics and Traumatology, BiomaTiCS, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany.
| | - Hermann Götz
- Platform for Biomaterial Research, University Medical Center, BiomaTiCS, Johannes Gutenberg University, 55131 Mainz, Germany.
| | - Stefan Stein
- Georg-Speyer-Haus-Institute for Tumor Biology and Experimental Therapy, 60659 Frankfurt, Germany.
| | - Pol Maria Rommens
- Department of Orthopaedics and Traumatology, BiomaTiCS, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany.
| |
Collapse
|
32
|
Jeong YM, Cheng XW, Lee S, Lee KH, Cho H, Kang JH, Kim W. Preconditioning with far-infrared irradiation enhances proliferation, cell survival, and migration of rat bone marrow-derived stem cells via CXCR4-ERK pathways. Sci Rep 2017; 7:13718. [PMID: 29057951 PMCID: PMC5651919 DOI: 10.1038/s41598-017-14219-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 10/06/2017] [Indexed: 01/06/2023] Open
Abstract
Far-infrared radiation (FIR) has been shown to exert positive effects on the cardiovascular system. However, the biological effects of FIR on bone marrow-derived stem cells (BMSCs) are not understood. In the present study, BMSCs were isolated from rat femur bone marrow and cultured in vitro. To investigate the effects of an FIR generator with an energy flux of 0.13 mW/cm2 on rat BMSCs, survival of BMSCs was measured by crystal violet staining, and cell proliferation was additionally measured using Ez-Cytox cell viability, EdU, and Brd U assays. FIR preconditioning was found to significantly increase BMSC proliferation and survival against H2O2. The scratch and transwell migration assays showed that FIR preconditioning resulted in an increase in BMSC migration. qRT-PCR and Western blot analyses demonstrated that FIR upregulated Nanog, Sox2, c-Kit, Nkx2.5, and CXCR4 at both the mRNA and protein levels. Consistent with these observations, PD98059 (an ERK inhibitor) and AMD3100 (a CXCR4 inhibitor) prevented the activation of CXCR4/ERK and blocked the cell proliferation and migration induced by FIR. Overall, these findings provide the first evidence that FIR confers a real and significant benefit on the preconditioning of BMSCs, and might lead to novel strategies for improving BMSC therapy for cardiac ischemia.
Collapse
Affiliation(s)
- Yun-Mi Jeong
- Division of Cardiology, Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University, Seoul, Republic of Korea
| | - Xian Wu Cheng
- Division of Cardiology, Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University, Seoul, Republic of Korea.,The Department of Cardiology, Yanbian University Hospital, Yanji, China
| | - Sora Lee
- Division of Cardiology, Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University, Seoul, Republic of Korea
| | - Kyung Hye Lee
- Division of Cardiology, Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University, Seoul, Republic of Korea
| | - Haneul Cho
- Division of Cardiology, Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University, Seoul, Republic of Korea
| | - Jung Hee Kang
- Division of Cardiology, Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University, Seoul, Republic of Korea
| | - Weon Kim
- Division of Cardiology, Department of Internal Medicine, Kyung Hee University Hospital, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
33
|
Heirani-Tabasi A, Toosi S, Mirahmadi M, Mishan MA, Bidkhori HR, Bahrami AR, Behravan J, Naderi-Meshkin H. Chemokine Receptors Expression in MSCs: Comparative Analysis in Different Sources and Passages. Tissue Eng Regen Med 2017; 14:605-615. [PMID: 30603514 DOI: 10.1007/s13770-017-0069-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 06/19/2017] [Accepted: 06/29/2017] [Indexed: 12/13/2022] Open
Abstract
MSC-based therapy is providing a cure for degenerative diseases with unmet medical need and usually iliac crest bone marrow (ICBM) are being applied in clinics. Alternative sources, including adipose tissue and reamer/irrigator/aspirator hold great potential for isolating MCSs. Here, we compared original MSCs features of adipose tissue (Ad-MSCs) and bone marrow of long-bone (RIA-MSCs) or iliac crest, and the expression of chemokine receptors (including CXCR4, CX3CR1, CXCR6, CXCR2, CCR1 and CCR7) in these three sources, which are important in the context of homing. We further investigated the role of SDF-1/CXCR4 axis as a key player in motility of different population of MSCs using Transwell migration assay. All cells exhibited typical MSCs characteristics. However, different MSCs sources expressed different levels of chemokine receptors. Generally, the expression of these chemokine receptors was decreased with increasing passage (P) number from 2 to 3. Interestingly, it was observed that the CXCR4 expression and migration capacity in Ad-MSCs is significantly higher than ICBM and RIA-MSCs in P2. Although our data showed that CXCR4 had highest expression in P2 Ad-MSCs, but it dramatically declined following sub-culturing in the P3. Hence, to improve homing of MSCs by means of chemokine/their receptors axis, the source of isolation and passage number should be considered for clinical applications.
Collapse
Affiliation(s)
- Asieh Heirani-Tabasi
- Department of stem Cells and Regenerative Medicine Research, ACECR-Khorasan Razavi Branch Institute, Mashhad, 917751436 Iran
| | - Shirin Toosi
- 2Department of Biotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, 917751436 Iran
| | - Mahdi Mirahmadi
- Department of stem Cells and Regenerative Medicine Research, ACECR-Khorasan Razavi Branch Institute, Mashhad, 917751436 Iran
| | - Mohammad Amir Mishan
- Department of stem Cells and Regenerative Medicine Research, ACECR-Khorasan Razavi Branch Institute, Mashhad, 917751436 Iran
| | - Hamid Reza Bidkhori
- Department of stem Cells and Regenerative Medicine Research, ACECR-Khorasan Razavi Branch Institute, Mashhad, 917751436 Iran
| | - Ahmad Reza Bahrami
- Department of stem Cells and Regenerative Medicine Research, ACECR-Khorasan Razavi Branch Institute, Mashhad, 917751436 Iran
- 3Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, 917751436 Iran
| | - Javad Behravan
- 2Department of Biotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, 917751436 Iran
| | - Hojjat Naderi-Meshkin
- Department of stem Cells and Regenerative Medicine Research, ACECR-Khorasan Razavi Branch Institute, Mashhad, 917751436 Iran
| |
Collapse
|
34
|
Barhanpurkar-Naik A, Mhaske ST, Pote ST, Singh K, Wani MR. Interleukin-3 enhances the migration of human mesenchymal stem cells by regulating expression of CXCR4. Stem Cell Res Ther 2017; 8:168. [PMID: 28705238 PMCID: PMC5512829 DOI: 10.1186/s13287-017-0618-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 06/06/2017] [Accepted: 06/23/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) represent an important source for cell therapy in regenerative medicine. MSCs have shown promising results for repair of damaged tissues in various degenerative diseases in animal models and also in human clinical trials. However, little is known about the factors that could enhance the migration and tissue-specific engraftment of exogenously infused MSCs for successful regenerative cell therapy. Previously, we have reported that interleukin-3 (IL-3) prevents bone and cartilage damage in animal models of rheumatoid arthritis and osteoarthritis. Also, IL-3 promotes the differentiation of human MSCs into functional osteoblasts and increases their in-vivo bone regenerative potential in immunocompromised mice. However, the role of IL-3 in migration of MSCs is not yet known. In the present study, we investigated the role of IL-3 in migration of human MSCs under both in-vitro and in-vivo conditions. METHODS MSCs isolated from human bone marrow, adipose and gingival tissues were used for in-vitro cell migration, motility and wound healing assays in the presence or absence of IL-3. The effect of IL-3 preconditioning on expression of chemokine receptors and integrins was examined by flow cytometry and real-time PCR. The in-vivo migration of IL-3-preconditioned MSCs was investigated using a subcutaneous matrigel-releasing stromal cell-derived factor-1 alpha (SDF-1α) model in immunocompromised mice. RESULTS We observed that human MSCs isolated from all three sources express IL-3 receptor-α (IL-3Rα) both at gene and protein levels. IL-3 significantly enhances in-vitro migration, motility and wound healing abilities of MSCs. Moreover, IL-3 preconditioning upregulates expression of chemokine (C-X-C motif) receptor 4 (CXCR4) on MSCs, which leads to increased migration of cells towards SDF-1α. Furthermore, CXCR4 antagonist AMD3100 decreases the migration of IL-3-treated MSCs towards SDF-1α. Importantly, IL-3 also induces in-vivo migration of MSCs towards subcutaneously implanted matrigel-releasing-SDF-1α in immunocompromised mice. CONCLUSIONS The present study demonstrates for the first time that IL-3 has an important role in enhancing the migration of human MSCs through regulation of the CXCR4/SDF-1α axis. These findings suggest a potential role of IL-3 in improving the efficacy of MSCs in regenerative cell therapy.
Collapse
Affiliation(s)
| | - Suhas T. Mhaske
- National Centre for Cell Science, S. P. Pune University Campus, Pune, 411 007 India
| | - Satish T. Pote
- National Centre for Cell Science, S. P. Pune University Campus, Pune, 411 007 India
| | - Kanupriya Singh
- National Centre for Cell Science, S. P. Pune University Campus, Pune, 411 007 India
| | - Mohan R. Wani
- National Centre for Cell Science, S. P. Pune University Campus, Pune, 411 007 India
| |
Collapse
|
35
|
A Hyper-Crosslinked Carbohydrate Polymer Scaffold Facilitates Lineage Commitment and Maintains a Reserve Pool of Proliferating Cardiovascular Progenitors. Transplant Direct 2017; 3:e153. [PMID: 28573188 PMCID: PMC5441984 DOI: 10.1097/txd.0000000000000667] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 02/12/2017] [Indexed: 12/17/2022] Open
Abstract
Background Cardiovascular progenitor cells (CPCs) have been cultured on various scaffolds to resolve the challenge of cell retention after transplantation and to improve functional outcome after cell-based cardiac therapy. Previous studies have reported successful culture of fully differentiated cardiomyocytes on scaffolds of various types, and ongoing efforts are focused on optimizing the mix of cardiomyocytes and endothelial cells as well as on the identification of a source of progenitors capable of reversing cardiovascular damage. A scaffold culture that fosters cell differentiation into cardiomyocytes and endothelial cells while maintaining a progenitor reserve would benefit allogeneic cell transplantation. Methods Isl-1 + c-Kit + CPCs were isolated as clonal populations from human and sheep heart tissue. After hyper-crosslinked carbohydrate polymer scaffold culture, cells were assessed for differentiation, intracellular signaling, cell cycling, and growth factor/chemokine expression using real time polymerase chain reaction, flow cytometry, immunohistochemistry, and calcium staining. Results Insulin-like growth factor 1, hepatocyte growth factor, and stromal cell derived factor 1α paracrine factors were induced, protein kinase B signaling was activated, extracellular signal-regulated kinase phosphorylation was reduced and differentiation into both cardiomyocytes and endothelial cells was induced by scaffold-based cell culture. Interestingly, movement of CPCs out of the G1 phase of the cell cycle and increased expression of pluripotency genes PLOU5F1 (Oct4) and T (Brachyury) within a portion of the cultured population occurred, which suggests the maintenance of a progenitor population. Two-color immunostaining and 3-color fluorescence-activated cell sorting analysis confirmed the presence of both Isl-1 expressing undifferentiated cells and differentiated cells identified by troponin T and von Willebrand factor expression. Ki-67 labeling verified the presence of proliferating cells that remained in situ alongside the differentiated functional derivatives. Conclusions Cloned Isl-1 + c-kit + CPCs maintained on a hyper-cross linked polymer scaffold retain dual potential for proliferation and differentiation, providing a scaffold-based stem cell source for transplantation of committed and proliferating cardiovascular progenitors for functional testing in preclinical models of cell-based repair.
Collapse
|
36
|
Mollazadeh S, Neshati V, Fazly Bazzaz BS, Iranshahi M, Mojarrad M, Naderi-Meshkin H, Kerachian MA. StandardizedSophora pachycarpaRoot Extract Enhances Osteogenic Differentiation in Adipose-derived Human Mesenchymal Stem Cells. Phytother Res 2017; 31:792-800. [DOI: 10.1002/ptr.5803] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 02/20/2017] [Accepted: 02/24/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Samaneh Mollazadeh
- Biotechnology Research Center; Mashhad University of Medical Sciences; Mashhad Iran
| | - Vajiheh Neshati
- Biotechnology Research Center; Mashhad University of Medical Sciences; Mashhad Iran
| | - Bibi Sedigheh Fazly Bazzaz
- Biotechnology Research Center; Mashhad University of Medical Sciences; Mashhad Iran
- School of Pharmacy; Mashhad University of Medical Sciences; Mashhad Iran
| | - Mehrdad Iranshahi
- Biotechnology Research Center; Mashhad University of Medical Sciences; Mashhad Iran
- School of Pharmacy; Mashhad University of Medical Sciences; Mashhad Iran
| | - Majid Mojarrad
- Medical Genetics Research Center, Faculty of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
- Department of Medical Genetics, Faculty of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
| | - Hojjat Naderi-Meshkin
- Stem Cells and Regenerative Medicine Research Group; Academic Center for Education, Culture and Research (ACECR), Khorasan-Razavi Branch; Mashhad Iran
| | - Mohammad Amin Kerachian
- Medical Genetics Research Center, Faculty of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
- Department of Medical Genetics, Faculty of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
| |
Collapse
|
37
|
Ahmed NEMB, Murakami M, Kaneko S, Nakashima M. The effects of hypoxia on the stemness properties of human dental pulp stem cells (DPSCs). Sci Rep 2016; 6:35476. [PMID: 27739509 PMCID: PMC5064411 DOI: 10.1038/srep35476] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 09/30/2016] [Indexed: 12/21/2022] Open
Abstract
Recent studies have demonstrated that culture under hypoxia has beneficial effects on mesenchymal stem cells (MSCs). However, there are limitations to achieving a stable condition in conventional hypoxic CO2 incubators. DPSCs are a unique type of MSCs which are promising in many regenerative therapies. In this study, we investigated the ideal hypoxic culture environment for DPSCs using a new system that can provide controlled O2 environment. The effects of hypoxia (3%, 5%) on the stemness properties of DPSCs. Their morphology, proliferation rate, expression of stem cell markers, migration ability, mRNA expression of angiogenic/neurotrophic factors and immunomodulatory genes were evaluated and compared. Additionally, the effect of the discrete secretome on proliferation, migration, and neurogenic induction was assessed. Hypoxic DPSCs were found to be smaller in size and exhibited larger nuclei. 5% O2 significantly increased the proliferation rate, migration ability, expression of stem cell markers (CXCR4 and G-CSFR), and expression of SOX2, VEGF, NGF, and BDNF genes of DPSCs. Moreover, secretome collected from 5%O2 cultures displayed higher stimulatory effects on proliferation and migration of NIH3T3 cells and on neuronal differentiation of SH-SY5Y cells. These results demonstrate that 5%O2 may be ideal for enhancing DPSCs growth, stem cell properties, and secretome trophic effect.
Collapse
Affiliation(s)
- Nermeen El-Moataz Bellah Ahmed
- Department of Stem Cell Biology and Regenerative Medicine, National Center for Geriatrics and Gerontology, Research Institute, Obu, Aichi, Japan.,Department of Oro-dental genetics, Division of Human Genetics and Human Genome, National research center, Cairo, Egypt
| | - Masashi Murakami
- Department of Stem Cell Biology and Regenerative Medicine, National Center for Geriatrics and Gerontology, Research Institute, Obu, Aichi, Japan
| | - Satoru Kaneko
- Reproduction Center, Gynecology, Ichikawa General Hospital, Tokyo Dental College, Sugano, Ichikawa, Chiba, Japan
| | - Misako Nakashima
- Department of Stem Cell Biology and Regenerative Medicine, National Center for Geriatrics and Gerontology, Research Institute, Obu, Aichi, Japan
| |
Collapse
|
38
|
Ezquer FE, Ezquer ME, Vicencio JM, Calligaris SD. Two complementary strategies to improve cell engraftment in mesenchymal stem cell-based therapy: Increasing transplanted cell resistance and increasing tissue receptivity. Cell Adh Migr 2016; 11:110-119. [PMID: 27294313 PMCID: PMC5308221 DOI: 10.1080/19336918.2016.1197480] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Over the past 2 decades, therapies based on mesenchymal stem cells (MSC) have been tested to treat several types of diseases in clinical studies, due to their potential for tissue repair and regeneration. Currently, MSC-based therapy is considered a biologically safe procedure, with the therapeutic results being very promising. However, the benefits of these therapies are not stable in the long term, and the final outcomes manifest with high inter-patient variability. The major cause of these therapeutic limitations results from the poor engraftment of the transplanted cells. Researchers have developed separate strategies to improve MSC engraftment. One strategy aims at increasing the survival of the transplanted MSCs in the recipient tissue, rendering them more resistant to the hostile microenvironment (cell-preconditioning). Another strategy aims at making the damaged tissue more receptive to the transplanted cells, favoring their interactions (tissue-preconditioning). In this review, we summarize several approaches using these strategies, providing an integral and updated view of the recent developments in MSC-based therapies. In addition, we propose that the combined use of these different conditioning strategies could accelerate the process to translate experimental evidences from pre-clinic studies to the daily clinical practice.
Collapse
Affiliation(s)
- Fernando E Ezquer
- a Centro de Medicina Regenerativa, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo , Santiago , Chile
| | - Marcelo E Ezquer
- a Centro de Medicina Regenerativa, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo , Santiago , Chile
| | | | - Sebastián D Calligaris
- a Centro de Medicina Regenerativa, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo , Santiago , Chile
| |
Collapse
|
39
|
Ritz U, Kögler P, Höfer I, Frank P, Klees S, Gebhard S, Brendel C, Kaufmann K, Hofmann A, Rommens PM, Jonas U. Photocrosslinkable polysaccharide hydrogel composites based on dextran or pullulan–amylose blends with cytokines for a human co-culture model of human osteoblasts and endothelial cells. J Mater Chem B 2016; 4:6552-6564. [DOI: 10.1039/c6tb00654j] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Polysaccharide hyrogel composites demonstrate fundamental potential as biomaterials for bone regeneration in vitro.
Collapse
Affiliation(s)
- Ulrike Ritz
- University Medical Center of the Johannes Gutenberg University Mainz
- Department of Orthopaedics and Traumatology
- Biomatics Group Mainz
- Germany
| | - Peter Kögler
- Macromolecular Chemistry
- University of Siegen
- Germany
| | - Isabel Höfer
- Macromolecular Chemistry
- University of Siegen
- Germany
- TU Hamburg-Harburg
- Umwelttechnik und Energiewirtschaft
| | - Petra Frank
- Macromolecular Chemistry
- University of Siegen
- Germany
| | - Sven Klees
- Macromolecular Chemistry
- University of Siegen
- Germany
| | | | | | | | - Alexander Hofmann
- University Medical Center of the Johannes Gutenberg University Mainz
- Department of Orthopaedics and Traumatology
- Biomatics Group Mainz
- Germany
| | - Pol Maria Rommens
- University Medical Center of the Johannes Gutenberg University Mainz
- Department of Orthopaedics and Traumatology
- Biomatics Group Mainz
- Germany
| | - Ulrich Jonas
- Macromolecular Chemistry
- University of Siegen
- Germany
| |
Collapse
|
40
|
Cao Y, Liu T, Yin ML, Zhang BY, Song HL. Role of CX3CR1 in repair of injured intestinal epithelial cells by bone marrow mesenchymal stem cells. Shijie Huaren Xiaohua Zazhi 2015; 23:3670-3682. [DOI: 10.11569/wcjd.v23.i23.3670] [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] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the role of chemokine receptor CX3CR1 in the repair of injured intestinal epithelial cells by bone marrow mesenchymal stem cells (BM-MSCs).
METHODS: BM-MSCs were cultured and identified in vitro. Caco-2 cells were exposed to tumor necrosis factor alpha (TNF-α) to establish a cell model of injured intestinal epithelium. Cells were divided into six groups: BM-MSCs, Caco-2 cells, Caco-2 cells treated with TNF-α, co-cultured BM-MSCs and Caco-2 cells, co-cultured BM-MSCs and Caco-2 cells treated with TNF-α, and co-cultured BM-MSCs and Caco-2 cells treated with anti-CX3CR1 and TNF-α. The expression of tight junction proteins and mRNAs in Caco-2 cells, and CX3CR1 protein and mRNA in BM-MSCs was detected by immunofluorescence, Western blot and RT-PCR.
RESULTS: We selected 100 ng/mL TNF-α for 48 h to establish the injured model, because the expression of zonula occluden 1(ZO-1) and Occludin was reduced significantly at this time point (P < 0.05). The protein and mRNA levels of ZO-1, Occludin and CX3CR1 had no significant changes when BM-MSCs were co-cultured with untreated Caco-2 cells, but increased when BM-MSCs were co-cultured with injured Caco-2 cells (P < 0.05). When CX3CR1 was blocked, the protein and mRNA levels of ZO-1 and Occludin decreased significantly.
CONCLUSION: CX3CR1 participates in the repair of injured intestinal epithelial cells by BM-MSCs.
Collapse
|