1
|
Lee H, Lee TW, Chandrasekharan A, Sung SE, Yim SG, Kim S, Seong KY, Seo MS, Yang SY. Injectable Self-Crosslinkable Thiolated Hyaluronic Acid for Stem Cell Therapy of Atopic Dermatitis. ACS Biomater Sci Eng 2022; 8:1613-1622. [PMID: 35245045 DOI: 10.1021/acsbiomaterials.1c01374] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Stem cell therapies offer great promise in regenerative medicine to reinstate the normal function of diseased tissue, thereby avoiding the need for replacement. In stem cell therapies, damaged cells are replaced or restored by regulating inflammation and the immune system. However, the low survival rate and local retention of transplanted cells pose a significant challenge. In this study, injectable self-crosslinkable hydrogels using thiol-functionalized hyaluronic acid (HA-SH) were developed to improve the efficacy of mesenchymal stem cells (MSCs) for treating atopic dermatitis (AD)-related inflammatory lesions. The gelation kinetics and mechanical properties of HA-SH hydrogels were easily tuned by varying the concentration of the polymer in the precursor solution before injection. The MSC-laden HA-SH hydrogels exhibited high cell viability (>80%) for 1 week and good in vivo biocompatibility after implantation beneath the mouse skin. Moreover, the MSC-laden HA-SH hydrogel showed increased expression of anti-inflammatory cytokines, which can alleviate the immune response. In an AD animal model, a reduction in epidermal thickness and mast cell infiltration was achieved by applying a self-crosslinkable HA-SH solution including MSCs. This HA-based injectable hydrogel represents a potential carrier of stem cells, and its strong immunomodulation capabilities can be utilized for treating inflammation-related diseases.
Collapse
Affiliation(s)
- Hyeseon Lee
- Department of Biomaterials Science (BK21 Four Program), Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Korea
| | - Tae Wook Lee
- Department of Biomaterials Science (BK21 Four Program), Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Korea
| | - Ajeesh Chandrasekharan
- Department of Biomaterials Science (BK21 Four Program), Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Korea
| | - Soo-Eun Sung
- Department of Biomaterials Science (BK21 Four Program), Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Korea.,Department of Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea
| | - Sang-Gu Yim
- Department of Biomaterials Science (BK21 Four Program), Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Korea
| | - Sodam Kim
- Department of Biomaterials Science (BK21 Four Program), Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Korea
| | - Keum-Yong Seong
- Department of Biomaterials Science (BK21 Four Program), Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Korea
| | - Min-Soo Seo
- Department of Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea
| | - Seung Yun Yang
- Department of Biomaterials Science (BK21 Four Program), Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Korea
| |
Collapse
|
2
|
Placental Mesenchymal Stromal Cells (PMSCs) and PMSC-Derived Extracellular Vesicles (PMSC-EVs) Attenuated Renal Fibrosis in Rats with Unilateral Ureteral Obstruction (UUO) by Regulating CD4 + T Cell Polarization. Stem Cells Int 2020; 2020:2685820. [PMID: 32774389 PMCID: PMC7396053 DOI: 10.1155/2020/2685820] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 04/22/2020] [Accepted: 06/09/2020] [Indexed: 12/12/2022] Open
Abstract
Purpose Recent evidence has shown that CD4+ T helper (Th) cells are involved in renal inflammation and fibrosis. However, whether renal fibrosis can be alleviated by intervening in the polarization of CD4+ T cells remains unknown. Our research investigated the effects of intravenously administered placenta mesenchymal stromal cells (PMSCs) or treatment with extracellular EVs (EVs) derived from PMSCs (PMSC-EVs) on the polarization of CD4+ T cells in rats with unilateral ureteral obstruction (UUO). We further verified how PMSCs affect inflammatory factor secretion and the levels of regulatory T (Treg) and Th17 CD4+ T cells in vitro. Materials and Methods We evaluated renal interstitial inflammation and fibrosis by pathological section staining, tested the polarization of CD4+ T cells (Th17 and Treg phenotypes) by flow cytometry (FCM) and immunohistochemistry, and detected the cytokines secreted by CD4+ T cells by enzyme-linked immunosorbent assay (ELISA). Results Compared with that of control rats, the renal tissue of PMSC-treated rats exhibited lower renal Masson scores and more Foxp3+ cell infiltration, with a significantly decreased IL17A+CD4+ T cell/CD4+ T cell ratio and a significantly elevated anti-inflammatory cytokine (IL-10) level. When CD4+ T cells were cocultured with PMSCs, CD4+IL17A+ cell percentages were decreased in a UUO model after 7 days of coculture with PMSCs. The secretion of TGF-β and IL-10 was significantly increased (P < 0.05), while the secretion of IFN-γ, IL-17, and IL-6 was significantly decreased (P < 0.05) in the PMSC coculture group. Moreover, after treatment with PMSC-EVs, tubulointerstitial fibrosis was alleviated, and Foxp3+/IL-17+ cell infiltration was increased in the kidneys of UUO model animals on day 7. Conclusions PMSCs can convert the inflammatory environment into an anti-inflammatory environment by affecting the polarization of CD4+ T cells and macrophages, inhibiting the inflammatory factors IFN-γ and IL-17, and upregulating the expression of the anti-inflammatory factors TGF-β and IL-10, ultimately leading to renal protection. Such functions may be mediated by the paracrine activity of PMSC-EVs.
Collapse
|
3
|
Beegle JR. A Preview of Selected Articles. Stem Cells 2018. [DOI: 10.1002/stem.2946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Julie R. Beegle
- Institute for Regenerative Cures, University of California, Davis, Sacramento, California, USA
| |
Collapse
|
4
|
Yoshida K, Nakashima A, Doi S, Ueno T, Okubo T, Kawano KI, Kanawa M, Kato Y, Higashi Y, Masaki T. Serum-Free Medium Enhances the Immunosuppressive and Antifibrotic Abilities of Mesenchymal Stem Cells Utilized in Experimental Renal Fibrosis. Stem Cells Transl Med 2018; 7:893-905. [PMID: 30269426 PMCID: PMC6265641 DOI: 10.1002/sctm.17-0284] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 08/25/2018] [Indexed: 12/15/2022] Open
Abstract
Serum used in culture medium brings risks of immune reactions or infections and thus may hinder using ex vivo expanded mesenchymal stem cells (MSCs) for medical treatment. Here, we cultured MSCs in a serum-free medium (SF-MSCs) and in a medium containing 10% fetal bovine serum (10%MSCs) and investigated their effects on inflammation and fibrosis. MSC-conditioned medium suppressed transforming growth factor-β1-induced phosphorylation of Smad2 in HK-2 cells, with no significant difference between the two MSCs. This finding suggests that the direct antifibrotic effect of SF-MSCs is similar to that of 10%MSCs. However, immunohistochemistry revealed that renal fibrosis induced by unilateral ureteral obstruction in rats was more significantly ameliorated by the administration of SF-MSCs than by that of 10%MSCs. Coculture of MSCs and monocytic THP-1 cell-derived macrophages using a Transwell system showed that SF-MSCs significantly induced polarization from the proinflammatory M1 to the immunosuppressive M2 phenotype macrophages, suggesting that SF-MSCs strongly suppress the persistence of inflammation. Furthermore, the gene expression of tumor necrosis factor-α-induced protein 6 (TSG-6), which inhibits the recruitment of inflammatory cells, was higher in SF-MSCs than in 10%MSCs, and TSG-6 knockdown in SF-MSCs attenuated the anti-inflammatory responses in unilateral ureteral obstruction rats. These findings imply that SF culture conditions can enhance the immunosuppressive and antifibrotic abilities of MSCs and the administration of ex vivo expanded SF-MSCs has the potential to be a useful therapy for preventing the progression of renal fibrosis. Stem Cells Translational Medicine 2018;7:893-905.
Collapse
Affiliation(s)
- Ken Yoshida
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Ayumu Nakashima
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan.,Department of Stem Cell Biology and Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shigehiro Doi
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Toshinori Ueno
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Tomoe Okubo
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| | - Ki-Ichiro Kawano
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Masami Kanawa
- Natural Science Center for Basic Research and Development, Hiroshima University, Hiroshima, Japan
| | - Yukio Kato
- Department of Stem Cell Biology and Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan.,TWOCELLS Company, Limited, Hiroshima, Japan
| | - Yukihito Higashi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan.,Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | - Takao Masaki
- Department of Nephrology, Hiroshima University Hospital, Hiroshima, Japan
| |
Collapse
|
5
|
Song IH, Jung KJ, Lee TJ, Kim JY, Sung EG, Bae YC, Park YH. Mesenchymal stem cells attenuate adriamycin-induced nephropathy by diminishing oxidative stress and inflammation via downregulation of the NF-kB. Nephrology (Carlton) 2018; 23:483-492. [PMID: 28326639 DOI: 10.1111/nep.13047] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 03/05/2017] [Accepted: 03/15/2017] [Indexed: 01/17/2023]
Abstract
AIM This study aimed to evaluate the molecular mechanism mitigating progress of chronic nephropathy by mesenchymal stem cells (MSCs). METHODS Rats were divided into normal control (Normal), adriamycin (ADR)+vehicle (CON), and ADR+MSC (MSC) groups. Nephropathy was induced by ADR (4 mg/kg) and MSCs (2 × 106 ) were injected. Rats were euthanized 1 or 6 weeks after ADR injection. NF-kB, MAPKs, inflammation, oxidative stress, profibrotic molecules, and nephrin expression were evaluated. Electron and light microscopy were used for structural analysis. MSCs were co-cultured with renal tubular epithelial cells or splenocytes to evaluate relation with oxidative stress and inflammatory molecules RESULTS: Adriamycin treatment upregulated inflammation, oxidative stress, and profibrotic molecules; this was mitigated by MSCs. Glomerulosclerosis and interstitial fibrosis were observed in ADR-treated groups, and were more prominent in the CON group than in the MSC group. Fusion of foot processes and loss of slit diaphragms were also more prominent in the CON group than in the MSC group. In vitro, MSCs reduced oxidative stress related molecules, inflammatory cytokines, and NF-kB transcription. MSC- or ADR-induced regulation of NF-kB transcriptional activity was confirmed by a luciferase reporter assay. CONCLUSIONS Mesenchymal stem cells attenuate ADR-induced nephropathy by diminishing oxidative stress and inflammation via downregulation of NF-kB.
Collapse
Affiliation(s)
- In-Hwan Song
- Department of Anatomy, Yeungnam University College of Medicine, Daegu, South Korea
| | - Kyong-Jin Jung
- Department of Anatomy, Yeungnam University College of Medicine, Daegu, South Korea
| | - Tae-Jin Lee
- Department of Anatomy, Yeungnam University College of Medicine, Daegu, South Korea
| | - Joo-Young Kim
- Department of Anatomy, Yeungnam University College of Medicine, Daegu, South Korea
| | - Eon-Gi Sung
- Department of Anatomy, Yeungnam University College of Medicine, Daegu, South Korea
| | - Young Chul Bae
- Pediatrics, Yeungnam University College of Medicine, Daegu, South Korea
| | - Yong Hoon Park
- Department of Anatomy, Kyungpook National University School of Dentistry, Daegu, South Korea
| |
Collapse
|
6
|
Guan Q, Ezzati P, Spicer V, Krokhin O, Wall D, Wilkins JA. Interferon γ induced compositional changes in human bone marrow derived mesenchymal stem/stromal cells. Clin Proteomics 2017; 14:26. [PMID: 28694743 PMCID: PMC5501357 DOI: 10.1186/s12014-017-9161-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 06/28/2017] [Indexed: 12/18/2022] Open
Abstract
Background Mesenchymal stem/stromal cells (MSC) display a range of immunoregulatory properties which can be enhanced by the exposure to cytokines such interferon γ (IFN-γ). However the compositional changes associated with the ‘licensing’ of these cells have not been clearly defined. The present study was undertaken to provide a detailed comparative proteomic analysis of the compositional changes that occur in human bone marrow derived MSC following 20 h treatment with IFN-γ. Methods 2D LC MSMS analysis of control and IFN-γ treated cells from 5 different healthy donors provided confident identification of more than 8400 proteins. Results In total 210 proteins were shown to be significantly altered in their expression levels (≥|2SD|) following IFN-γ treatment. The changes for several of these proteins were confirmed by flow cytometry. STRING analysis determined that approximately 30% of the altered proteins physically interacted in described interferon mediated processes. Comparison of the list of proteins that were identified as changed in the proteomic analysis with data for the same proteins in the Interferome DB indicated that ~35% of these proteins have not been reported to be IFN-γ responsive in a range of cell types. Conclusions This data provides an in depth analysis of the proteome of basal and IFN-γ treated human mesenchymal stem cells and it identifies a number of novel proteins that may contribute to the immunoregulatory capacity if IFN-γ licensed cells. Electronic supplementary material The online version of this article (doi:10.1186/s12014-017-9161-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qingdong Guan
- Manitoba Centre for Advanced Cell and Tissue Therapy, Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB Canada.,Cellular Therapy Laboratory, CancerCare Manitoba, Winnipeg, MB Canada
| | - Peyman Ezzati
- Manitoba Centre for Proteomics and Systems Biology, Section of Biomedical Proteomics, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba and Health Sciences Centre, 799 John Buhler Research Centre, 715 McDermot Ave, Winnipeg, MB R3E 3P4 Canada
| | - Victor Spicer
- Manitoba Centre for Proteomics and Systems Biology, Section of Biomedical Proteomics, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba and Health Sciences Centre, 799 John Buhler Research Centre, 715 McDermot Ave, Winnipeg, MB R3E 3P4 Canada
| | - Oleg Krokhin
- Manitoba Centre for Proteomics and Systems Biology, Section of Biomedical Proteomics, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba and Health Sciences Centre, 799 John Buhler Research Centre, 715 McDermot Ave, Winnipeg, MB R3E 3P4 Canada
| | - Donna Wall
- Division of Haematology/Oncology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON Canada
| | - John A Wilkins
- Manitoba Centre for Proteomics and Systems Biology, Section of Biomedical Proteomics, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba and Health Sciences Centre, 799 John Buhler Research Centre, 715 McDermot Ave, Winnipeg, MB R3E 3P4 Canada
| |
Collapse
|
7
|
Wang B, Yao K, Huuskes BM, Shen HH, Zhuang J, Godson C, Brennan EP, Wilkinson-Berka JL, Wise AF, Ricardo SD. Mesenchymal Stem Cells Deliver Exogenous MicroRNA-let7c via Exosomes to Attenuate Renal Fibrosis. Mol Ther 2016; 24:1290-301. [PMID: 27203438 DOI: 10.1038/mt.2016.90] [Citation(s) in RCA: 286] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 04/20/2016] [Indexed: 02/07/2023] Open
Abstract
The advancement of microRNA (miRNA) therapies has been hampered by difficulties in delivering miRNA to the injured kidney in a robust and sustainable manner. Using bioluminescence imaging in mice with unilateral ureteral obstruction (UUO), we report that mesenchymal stem cells (MSCs), engineered to overexpress miRNA-let7c (miR-let7c-MSCs), selectively homed to damaged kidneys and upregulated miR-let7c gene expression, compared with nontargeting control (NTC)-MSCs. miR-let7c-MSC therapy attenuated kidney injury and significantly downregulated collagen IVα1, metalloproteinase-9, transforming growth factor (TGF)-β1, and TGF-β type 1 receptor (TGF-βR1) in UUO kidneys, compared with controls. In vitro analysis confirmed that the transfer of miR-let7c from miR-let7c-MSCs occurred via secreted exosomal uptake, visualized in NRK52E cells using cyc3-labeled pre-miRNA-transfected MSCs with/without the exosomal inhibitor, GW4869. The upregulated expression of fibrotic genes in NRK52E cells induced by TGF-β1 was repressed following the addition of isolated exosomes or indirect coculture of miR-let7c-MSCs, compared with NTC-MSCs. Furthermore, the cotransfection of NRK52E cells using the 3'UTR of TGF-βR1 confirmed that miR-let7c attenuates TGF-β1-driven TGF-βR1 gene expression. Taken together, the effective antifibrotic function of engineered MSCs is able to selectively transfer miR-let7c to damaged kidney cells and will pave the way for the use of MSCs for therapeutic delivery of miRNA targeted at kidney disease.
Collapse
Affiliation(s)
- Bo Wang
- Department of Anatomy and Developmental Biology, Monash University, Victoria, Australia
| | - Kevin Yao
- Department of Anatomy and Developmental Biology, Monash University, Victoria, Australia
| | - Brooke M Huuskes
- Department of Anatomy and Developmental Biology, Monash University, Victoria, Australia
| | - Hsin-Hui Shen
- Department of Microbiology, Monash University, Victoria, Australia
| | - Junli Zhuang
- Department of Anatomy and Developmental Biology, Monash University, Victoria, Australia
| | - Catherine Godson
- Diabetes Complications Research Centre, Conway Institute and School of Medicine, University College Dublin, Dublin, Ireland
| | - Eoin P Brennan
- Diabetes Complications Research Centre, Conway Institute and School of Medicine, University College Dublin, Dublin, Ireland
| | | | - Andrea F Wise
- Department of Anatomy and Developmental Biology, Monash University, Victoria, Australia
| | - Sharon D Ricardo
- Department of Anatomy and Developmental Biology, Monash University, Victoria, Australia
| |
Collapse
|
8
|
Yang N, Baban B, Isales CM, Shi XM. Crosstalk between bone marrow-derived mesenchymal stem cells and regulatory T cells through a glucocorticoid-induced leucine zipper/developmental endothelial locus-1-dependent mechanism. FASEB J 2015; 29:3954-63. [PMID: 26038125 DOI: 10.1096/fj.15-273664] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 05/26/2015] [Indexed: 12/31/2022]
Abstract
Bone marrow is a reservoir for regulatory T (T(reg)) cells, but how T(reg) cells are regulated in that environment remains poorly understood. We show that expression of glucocorticoid (GC)-induced leucine zipper (GILZ) in bone marrow mesenchymal lineage cells or bone marrow-derived mesenchymal stem cells (BMSCs) increases the production of T(reg) cells via a mechanism involving the up-regulation of developmental endothelial locus-1 (Del-1), an endogenous leukocyte-endothelial adhesion inhibitor. We found that the expression of Del-1 is increased ∼4-fold in the bone tissues of GILZ transgenic (Tg) mice, and this increase is coupled with a significant increase in the production of IL-10 (2.80 vs. 0.83) and decrease in the production of IL-6 (0.80 vs. 2.33) and IL-12 (0.25 vs. 1.67). We also show that GILZ-expressing BMSCs present antigen in a way that favors T(reg) cells. These results indicate that GILZ plays a critical role mediating the crosstalk between BMSCs and T(reg) in the bone marrow microenvironment. These data, together with our previous findings that overexpression of GILZ in BMSCs antagonizes TNF-α-elicited inflammatory responses, suggest that GILZ plays important roles in bone-immune cell communication and BMSC immune suppressive functions.
Collapse
Affiliation(s)
- Nianlan Yang
- *Department of Neuroscience and Regenerative Medicine, Department of Oral Biology, and Department of Orthopaedic Surgery, Georgia Regents University, Augusta, Georgia, USA
| | - Babak Baban
- *Department of Neuroscience and Regenerative Medicine, Department of Oral Biology, and Department of Orthopaedic Surgery, Georgia Regents University, Augusta, Georgia, USA
| | - Carlos M Isales
- *Department of Neuroscience and Regenerative Medicine, Department of Oral Biology, and Department of Orthopaedic Surgery, Georgia Regents University, Augusta, Georgia, USA
| | - Xing-Ming Shi
- *Department of Neuroscience and Regenerative Medicine, Department of Oral Biology, and Department of Orthopaedic Surgery, Georgia Regents University, Augusta, Georgia, USA
| |
Collapse
|
9
|
AlAhmari LS, AlShenaifi JY, AlAnazi RA, Abdo AA. Autologous bone marrow-derived cells in the treatment of liver disease patients. Saudi J Gastroenterol 2015; 21:5-10. [PMID: 25672232 PMCID: PMC4355864 DOI: 10.4103/1319-3767.151211] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Liver transplantation is universally accepted as a "cure" procedure, and yet is not universally applicable for the treatment of end-stage liver diseases (ESLD) because of the shortage of donors, operative complications, risk of rejection, and high cost. Bioartificial liver device is an option to temporarily improve the liver function and to bridge the patients to liver transplantation. However, bioartificial liver device has many problems in clinical application, such as hepatocyte allograft rejection and maintenance of hepatocyte viability and function. Another therapeutic option is stem cell transplantation. There are two broad types of stem cells: Embryonic stem cells and adult stem cells. The latter are sourced from bone marrow (BM), adipose tissue, and blood. This review will concentrate on BM-derived cells. BM-derived cell transplantation, although not ideal, is theoretically an optimal modality for the treatment of ESLD. Autologous BM-derived cells have no graft rejection, have the capability of regeneration and self-renewal, and are multipotent stem cells that can differentiate into a variety of cell types which include hepatocytes. The pathway from BM-derived cell to hepatocyte is well documented. The present review summarizes the delivery routes of BM-derived cells to the liver, the evidences of engraftment of BM-derived cells in the liver, and the possible mechanisms of BM-derived cells in liver repair and regeneration, and finally, updates the clinical applications.
Collapse
Affiliation(s)
- Leenah S. AlAhmari
- Liver Disease Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Jumanah Y. AlShenaifi
- Liver Disease Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Reema A. AlAnazi
- Liver Disease Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Ayman A. Abdo
- Liver Disease Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia,Address for correspondence: Prof. Ayman A. Abdo, Department of Medicine, College of Medicine, King Saud University, Saudi Arabia. E-mail:
| |
Collapse
|
10
|
Ozbek E, Adas G, Otunctemur A, Duruksu G, Koc B, Polat EC, Kemik Sarvan A, Okcu A, Kamali G, Subasi C, Karaoz E. Role of Mesenchymal Stem Cells Transfected With Vascular Endothelial Growth Factor in Maintaining Renal Structure and Function in Rats with Unilateral Ureteral Obstruction. EXP CLIN TRANSPLANT 2014; 13:262-72. [PMID: 25542189 DOI: 10.6002/ect.2014.0080] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVES Mesenchymal stem cells hold promise for renal disease treatment. Vascular endothelial growth factor may heal tubule-interstitial fibrosis in unilateral ureteral obstruction by inhibiting epithelial-mesenchymal transition. We investigated the protective effect of vascular endothelial growth factor in transfected mesenchymal stem cells in unilateral ureteral obstruction-induced renal injury in rats. MATERIALS AND METHODS Male Wistar Albino rats (32 rats; weight, 250-300 g) were divided into 4 equal groups: group 1, control; group 2, unilateral ureteral obstruction; group 3, unilateral ureteral obstruction and mesenchymal stem cells; and group 4, unilateral ureteral obstruction and vascular endothelial growth factor-transfected mesenchymal stem cells. Vascular endothelial growth factor-transfected mesenchymal stem cells were administered intravenously before onset of unilateral ureteral obstruction. On day 14, the rats were killed and kidneys were retrieved. Tubular necrosis, mononuclear cell infiltration, and interstitial fibrosis were evaluated in paraffin blocks. We evaluated green fluorescent protein-positive and vascular endothelial growth factor-positive cells; anti-inflammatory (Prostaglandin E2 receptor) and interleukin 1 receptor antagonist), proinflammatory/anti-inflammatory (interleukin 6), and proinflammatory (MPO) cytokine expression levels; and levels of nitric oxide; transforming growth factor β1, E-cadherin, and hydroxyproline. RESULTS Green fluorescent protein-positive cells were negative in the renal parenchyma in groups 1 and 2 and positive in groups 3 and 4. Vascular endothelial growth factor levels were significantly higher in group 4. Transforming growth factor β1, nitric oxide, and E-cadherin levels were significantly higher in the unilateral ureteral obstruction than control group; however, in the study groups, these values were not significantly different from the unilateral ureteral obstruction group. In stem cell-transplanted tissue samples, EP3, interleukin 1 receptor antagonist, and interleukin 6 levels were elevated, but MPO expression levels were low. Although there were significant differences for tubular necrosis and fibrosis in group 2, there were significant reductions in tubular injury and fibrosis in groups 3 and 4. CONCLUSIONS Systemic stem cells transplanted into the kidney protected against unilateral ureteral obstruction-induced renal epithelial-mesenchymal transition and renal fibrosis.
Collapse
Affiliation(s)
- Emin Ozbek
- From the Okmeydani Training and Research Hospital, Department of Urology, Istanbul, Turkey
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Duffy MM, McNicholas BA, Monaghan DA, Hanley SA, McMahon JM, Pindjakova J, Alagesan S, Fearnhead HO, Griffin MD. Mesenchymal stem cells and a vitamin D receptor agonist additively suppress T helper 17 cells and the related inflammatory response in the kidney. Am J Physiol Renal Physiol 2014; 307:F1412-26. [DOI: 10.1152/ajprenal.00024.2014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) suppress T helper (Th)17 cell differentiation and are being clinically pursued for conditions associated with aberrant Th17 responses. Whether such immunomodulatory effects are enhanced by coadministration of MSCs with other agents is not well known. In the present study, individual and combined effects of MSCs and the vitamin D receptor (VDR) agonist paricalcitol on Th17 induction were investigated in vitro and in a mouse model of sterile kidney inflammation (unilateral ureteral obstruction). In vitro, MSCs and paricalcitol additively suppressed Th17 differentiation, although only MSCs suppressed expression of Th17-associated transcriptions factors. Combined administration of MSCs and paricalcitol resulted in an early ( day 3) reduction of intrarenal CD4+ and CD8+ T cells, CD11b+/lymphocyte antigen 6G+ neutrophils, and inflammatory (lymphocyte antigen 6Chi) monocytes as well as reduced transcript for IL-17 compared with untreated animals. Later ( day 8), obstructed kidneys of MSC/paricalcitol double-treated mice, but not mice treated with either intervention alone, had reduced tubular injury and interstitial fibrosis as well as lower numbers of neutrophils and inflammatory monocytes and an increase in the ratio between M2 (CD206+) and M1 (CD206−) macrophages compared with control mice. Adjunctive therapy with VDR agonists may enhance the immunosuppressive properties of MSCs in the setting of pathogenic Th17-type immune responses and related inflammatory responses.
Collapse
Affiliation(s)
- Michelle M. Duffy
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science and College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | - Bairbre A. McNicholas
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science and College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | - David A. Monaghan
- National Centre for Biomedical Engineering Science and College of Science, National University of Ireland, Galway, Galway, Ireland; and
| | - Shirley A. Hanley
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science and College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | - Jill M. McMahon
- National Centre for Biomedical Engineering Science and College of Science, National University of Ireland, Galway, Galway, Ireland; and
| | - Jana Pindjakova
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science and College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | - Senthilkumar Alagesan
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science and College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| | - Howard O. Fearnhead
- National Centre for Biomedical Engineering Science and College of Medicine, Nursing and Health Sciences, Discipline of Pharmacology and Therapeutics, National University of Ireland, Galway, Galway, Ireland
| | - Matthew D. Griffin
- Regenerative Medicine Institute, National Centre for Biomedical Engineering Science and College of Medicine, Nursing and Health Sciences, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
12
|
Huuskes BM, Wise AF, Cox AJ, Lim EX, Payne NL, Kelly DJ, Samuel CS, Ricardo SD. Combination therapy of mesenchymal stem cells and serelaxin effectively attenuates renal fibrosis in obstructive nephropathy. FASEB J 2014; 29:540-53. [PMID: 25395452 DOI: 10.1096/fj.14-254789] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Chronic kidney disease (CKD) results from the development of fibrosis, ultimately leading to end-stage renal disease (ESRD). Although human bone marrow-derived mesenchymal stem cells (MSCs) can accelerate renal repair following acute injury, the establishment of fibrosis during CKD may affect their potential to influence regeneration capacity. Here we tested the novel combination of MSCs with the antifibrotic serelaxin to repair and protect the kidney 7 d post-unilateral ureteral obstruction (UUO), when fibrosis is established. Male C57BL6 mice were sham-operated or UUO-inured (n = 4-6) and received vehicle, MSCs (1 × 10(6)), serelaxin (0.5 mg/kg per d), or the combination of both. In vivo tracing studies with luciferin/enhanced green fluorescent protein (eGFP)-tagged MSCs showed specific localization in the obstructed kidney where they remained for 36 h. Combination therapy conferred significant protection from UUO-induced fibrosis, as indicated by hydroxyproline analysis (P < 0.001 vs. vehicle, P < 0.05 vs. MSC or serelaxin alone). This was accompanied by preserved structural architecture, decreased tubular epithelial injury (P < 0.01 vs. MSCs alone), macrophage infiltration, and myofibroblast localization in the kidney (both P < 0.01 vs. vehicle). Combination therapy also stimulated matrix metalloproteinase (MMP)-2 activity over either treatment alone (P < 0.05 vs. either treatment alone). These results suggest that the presence of an antifibrotic in conjunction with MSCs ameliorates established kidney fibrosis and augments tissue repair to a greater extent than either treatment alone.
Collapse
Affiliation(s)
- Brooke M Huuskes
- *Department of Anatomy and Developmental Biology and Department of Pharmacology, Monash University, Clayton, Victoria, Australia; Department of Medicine, University of Melbourne, St. Vincent's Hospital, Melbourne, Australia; and Australia Regenerative Medicine Institute (ARMI) and Monash University, Clayton, Victoria, Australia
| | - Andrea F Wise
- *Department of Anatomy and Developmental Biology and Department of Pharmacology, Monash University, Clayton, Victoria, Australia; Department of Medicine, University of Melbourne, St. Vincent's Hospital, Melbourne, Australia; and Australia Regenerative Medicine Institute (ARMI) and Monash University, Clayton, Victoria, Australia
| | - Alison J Cox
- *Department of Anatomy and Developmental Biology and Department of Pharmacology, Monash University, Clayton, Victoria, Australia; Department of Medicine, University of Melbourne, St. Vincent's Hospital, Melbourne, Australia; and Australia Regenerative Medicine Institute (ARMI) and Monash University, Clayton, Victoria, Australia
| | - Ee X Lim
- *Department of Anatomy and Developmental Biology and Department of Pharmacology, Monash University, Clayton, Victoria, Australia; Department of Medicine, University of Melbourne, St. Vincent's Hospital, Melbourne, Australia; and Australia Regenerative Medicine Institute (ARMI) and Monash University, Clayton, Victoria, Australia
| | - Natalie L Payne
- *Department of Anatomy and Developmental Biology and Department of Pharmacology, Monash University, Clayton, Victoria, Australia; Department of Medicine, University of Melbourne, St. Vincent's Hospital, Melbourne, Australia; and Australia Regenerative Medicine Institute (ARMI) and Monash University, Clayton, Victoria, Australia
| | - Darren J Kelly
- *Department of Anatomy and Developmental Biology and Department of Pharmacology, Monash University, Clayton, Victoria, Australia; Department of Medicine, University of Melbourne, St. Vincent's Hospital, Melbourne, Australia; and Australia Regenerative Medicine Institute (ARMI) and Monash University, Clayton, Victoria, Australia
| | - Chrishan S Samuel
- *Department of Anatomy and Developmental Biology and Department of Pharmacology, Monash University, Clayton, Victoria, Australia; Department of Medicine, University of Melbourne, St. Vincent's Hospital, Melbourne, Australia; and Australia Regenerative Medicine Institute (ARMI) and Monash University, Clayton, Victoria, Australia
| | - Sharon D Ricardo
- *Department of Anatomy and Developmental Biology and Department of Pharmacology, Monash University, Clayton, Victoria, Australia; Department of Medicine, University of Melbourne, St. Vincent's Hospital, Melbourne, Australia; and Australia Regenerative Medicine Institute (ARMI) and Monash University, Clayton, Victoria, Australia
| |
Collapse
|
13
|
Wise AF, Williams TM, Kiewiet MBG, Payne NL, Siatskas C, Samuel CS, Ricardo SD. Human mesenchymal stem cells alter macrophage phenotype and promote regeneration via homing to the kidney following ischemia-reperfusion injury. Am J Physiol Renal Physiol 2014; 306:F1222-35. [PMID: 24623144 DOI: 10.1152/ajprenal.00675.2013] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) ameliorate injury and accelerate repair in many organs, including the kidney, although the reparative mechanisms and interaction with macrophages have not been elucidated. This study investigated the reparative potential of human bone marrow-derived MSCs and traced their homing patterns following administration to mice with ischemia-reperfusion (IR) injury using whole body bioluminescence imaging. The effect of MSCs on macrophage phenotype following direct and indirect coculture was assessed using qPCR. Human cytokine production was measured using multiplex arrays. After IR, MSCs homed to injured kidneys where they afforded protection indicated by decreased proximal tubule kidney injury molecule-1 expression, blood urea nitrogen, and serum creatinine levels. SDS-PAGE and immunofluorescence labeling revealed MSCs reduced collagen α1(I) and IV by day 7 post-IR. Gelatin zymography confirmed that MSC treatment significantly increased matrix metalloproteinase-9 activity in IR kidneys, which contributed to a reduction in total collagen. Following direct and indirect coculture, macrophages expressed genes indicative of an anti-inflammatory "M2" phenotype. MSC-derived human GM-CSF, EGF, CXCL1, IL-6, IL-8, MCP-1, PDGF-AA, and CCL5 were identified in culture supernatants. In conclusion, MSCs home to injured kidneys and promote repair, which may be mediated by their ability to promote M2 macrophage polarization.
Collapse
Affiliation(s)
- Andrea F Wise
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Timothy M Williams
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Mensiena B G Kiewiet
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Natalie L Payne
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia; and
| | - Christopher Siatskas
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Chrishan S Samuel
- Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Sharon D Ricardo
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia;
| |
Collapse
|