1
|
Waack N, Guirao T, Maquigussa E, Nishi E, Ormanji M, Ykuta O, Boim M. Stem cells prevent long-term deterioration of renal function after renal artery revascularization in a renovascular hypertension model in rats. Sci Rep 2025; 15:3397. [PMID: 39870783 PMCID: PMC11772754 DOI: 10.1038/s41598-025-87451-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 01/20/2025] [Indexed: 01/29/2025] Open
Abstract
Partial stenosis of the renal artery causes renovascular hypertension (RVH) and is accompanied by chronic renal ischemia, resulting in irreversible kidney damage. Revascularization constitutes the most efficient therapy for normalizing blood pressure (BP) and has significant benefits for renal function; however, the tissue damage caused by chronic hypoxia is not fully reversed. Mesenchymal stem cells (MSCs) have produced discrete results in minimizing RVH and renal tissue and functional improvements since the obstruction persists. This study aimed to evaluate the effects of administration of MSCs in combination with renal artery revascularization in rats subjected to RVH. The following groups were evaluated: control (SHAM), hypertensive (2K-1C), hypertensive treated with MSCs (MSC), hypertensive subjected to revascularization (REV), and hypertensive subjected to revascularization and treatment with MSCs (REV + MSC). The animals were followed up for 10 weeks. The animals in the MSC group received cell infusions at the 3rd, 5th, 7th and 9th weeks. In the REV and REV + MSC groups, the clip was removed by the 6th week (revascularization), and in the REV + MSC group, MSCs infusion was performed at the 6th and 8th weeks. Tail systolic blood pressure (SBP) was measured weekly, and histological parameters and renal function were evaluated at the end of the protocol. The clipped animals developed RVH, deterioration of total renal function (50% decrease in creatinine clearance), and significant proteinuria (15x increase). Treatment with MSCs had no detectable beneficial effects on kidney function or SBP. REV resulted in normalization of BP and a significant but partial reduction in proteinuria (80% vs. 2K-1C), but areas with renal fibrosis persisted. The combination of the two treatments was effective at normalizing all renal parameters as well as reversing proteinuria, reducing the number of ischemic glomeruli and atrophic tubules, indicating an improvement of the renal parenchyma. The results suggest that therapy with MSCs associated with revascularization can potentially help in the full recovery of renal function in the long term in patients with RVH.
Collapse
Affiliation(s)
- Nikolas Waack
- Renal Division, Department of Medicine, Universidade Federal de São Paulo, Rua Pedro de Toledo, 781, São Paulo, SP, 04039-032, Brazil
| | - Tatiana Guirao
- Renal Division, Department of Medicine, Universidade Federal de São Paulo, Rua Pedro de Toledo, 781, São Paulo, SP, 04039-032, Brazil
| | - Edgar Maquigussa
- Renal Division, Department of Medicine, Universidade Federal de São Paulo, Rua Pedro de Toledo, 781, São Paulo, SP, 04039-032, Brazil
| | - Erika Nishi
- Cardiovascular Physiology Division, Department of Medicine, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Milene Ormanji
- Renal Division, Department of Medicine, Universidade Federal de São Paulo, Rua Pedro de Toledo, 781, São Paulo, SP, 04039-032, Brazil
| | - Olinda Ykuta
- Renal Division, Department of Medicine, Universidade Federal de São Paulo, Rua Pedro de Toledo, 781, São Paulo, SP, 04039-032, Brazil
| | - Mirian Boim
- Renal Division, Department of Medicine, Universidade Federal de São Paulo, Rua Pedro de Toledo, 781, São Paulo, SP, 04039-032, Brazil.
| |
Collapse
|
2
|
Lim D, Kim I, Song Q, Kim JH, Atala A, Jackson JD, Yoo JJ. Development and intra-renal delivery of renal progenitor organoids for effective integration in vivo. Stem Cells Transl Med 2025; 14:szae078. [PMID: 39468757 PMCID: PMC11832275 DOI: 10.1093/stcltm/szae078] [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: 06/03/2022] [Accepted: 09/23/2024] [Indexed: 10/30/2024] Open
Abstract
Renal progenitor organoids have been proposed as a source of tissue for kidney regeneration; however, their clinical translatability has not been demonstrated due to an inability to mass-produce comprehensive renal progenitor organoids and the lack of an effective intra-renal delivery platform that facilitates rapid integration into functionally meaningful sites. This study addresses these shortcomings. Human-induced pluripotent stem cells were differentiated into renal progenitor cells using an established protocol and aggregated using a novel assembly method to produce high yields of organoids. Organoids were encapsulated in collagen-based scaffolds for in vitro study and in vivo implantation into mouse renal cortex. In vitro, the organoids demonstrated sustained cell viability and renal structure maturation over time. In vivo delivered organoids showed rapid integration into host renal parenchyma while showing tubular and glomerular-like structure development and maturity markers. This proof-of-concept study presents many promising results, providing a system of renal organoid formation and delivery that may support the development of clinically translatable therapies and the advancement of in vitro renal organoid studies.
Collapse
Affiliation(s)
- Diana Lim
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, United States
| | - Ickhee Kim
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, United States
| | - Qianqian Song
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, United States
| | - Ji Hyun Kim
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, United States
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, United States
| | - John D Jackson
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, United States
| | - James J Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, United States
| |
Collapse
|
3
|
Dos Santos LG, Ferreira PI, Krause A. Mesenchymal stem cell transplantation: Systematic review, meta-analysis and clinical applications for acute kidney injury and chronic kidney disease in dogs and cats. Res Vet Sci 2024; 175:105313. [PMID: 38851051 DOI: 10.1016/j.rvsc.2024.105313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/08/2024] [Accepted: 05/25/2024] [Indexed: 06/10/2024]
Abstract
Chronic kidney disease (CKD) and acute kidney injury (AKI) are diseases which affect the urinary tract characterized by the loss of renal function. Their therapy requires different therapeutic goals. Mesenchymal stem cells (MSC) transplantation has spread over the years as a treatment for many diseases. In the urinary tract, studies report anti-inflammatory, antiapoptotic, antifibrotic, antioxidant and angiogenic effects. This work reports the results of a meta-analysis about the effects of the MSC application in serum levels of creatinine in dogs and cats with AKI and CKD. The work followed PRISMA guidelines. Data were screened, selected, and extracted with characteristics about the studies. The kinds of injury were classified according to their identification and the risk of bias was calculated by the system SYRCLE. The results of each group were combined by the inverse variance method. The heterogeneity was evaluated by the I2 test. For the mean of creatinine, a meta-analysis was performed according to the study group and number of applications and separately for the control and treatment groups according to the kind of injury, dose, application route, and moment. At all, 4742 articles were found. Of these, 40 were selected for eligibility, 16 underwent qualitative analysis and 9 to the quantitative. The results denote advantage to the group treated with MSC over placebo. A statistical difference was observed both in combined analysis and in the subgroups division. However, a high heterogeneity was found, which indicates considerable variation between the studies, which indicates caution in generalize the results.
Collapse
Affiliation(s)
- Leonardo Gaspareto Dos Santos
- Federal University of Santa Maria, Veterinary Hospital, Department of Small Animal Clinic, Santa Maria, Rio Grande do Sul, Brazil
| | - Priscila Inês Ferreira
- Federal University of Santa Maria, Veterinary Hospital, Department of Small Animal Clinic, Santa Maria, Rio Grande do Sul, Brazil
| | - Alexandre Krause
- Federal University of Santa Maria, Veterinary Hospital, Department of Small Animal Clinic, Santa Maria, Rio Grande do Sul, Brazil.
| |
Collapse
|
4
|
Kim Y, Kang D, Choi GE, Kim SD, Yang SJ, Kim H, You D, Kim CS, Suh N. Therapeutic potential of BMSC-conditioned medium in an in vitro model of renal fibrosis using the RPTEC/TERT1 cell line. BMB Rep 2024; 57:116-121. [PMID: 38303564 PMCID: PMC10910087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 12/27/2023] [Accepted: 01/15/2024] [Indexed: 02/03/2024] Open
Abstract
We investigated the therapeutic potential of bone marrow-derived mesenchymal stem cell-conditioned medium (BMSC-CM) on immortalized renal proximal tubule epithelial cells (RPTEC/ TERT1) in a fibrotic environment. To replicate the increased stiffness characteristic of kidneys in chronic kidney disease, we utilized polyacrylamide gel platforms. A stiff matrix was shown to increase α-smooth muscle actin (α-SMA) levels, indicating fibrogenic activation in RPTEC/TERT1 cells. Interestingly, treatment with BMSC-CM resulted in significant reductions in the levels of fibrotic markers (α-SMA and vimentin) and increases in the levels of the epithelial marker E-cadherin and aquaporin 7, particularly under stiff conditions. Furthermore, BMSC-CM modified microRNA (miRNA) expression and reduced oxidative stress levels in these cells. Our findings suggest that BMSC-CM can modulate cellular morphology, miRNA expression, and oxidative stress in RPTEC/TERT1 cells, highlighting its therapeutic potential in fibrotic kidney disease. [BMB Reports 2024; 57(2): 116-121].
Collapse
Affiliation(s)
- Yunji Kim
- Department of Medical Sciences, General Graduate School, Soonchunhyang University, Asan 31538, Korea
| | - Dayeon Kang
- Department of Medical Sciences, General Graduate School, Soonchunhyang University, Asan 31538, Korea
- Department of Pharmaceutical Engineering, College of Medical Sciences, Soonchunhyang University, Asan 31538, Korea
| | - Ga-eun Choi
- Department of Medical Sciences, General Graduate School, Soonchunhyang University, Asan 31538, Korea
| | - Sang Dae Kim
- Department of Medical Sciences, General Graduate School, Soonchunhyang University, Asan 31538, Korea
| | | | - Hyosang Kim
- Division of Nephrology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Dalsan You
- Department of Urology, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Choung Soo Kim
- Urology Institute, Ewha Womans University Mokdong Hospital, Seoul 07985, Korea
| | - Nayoung Suh
- Department of Medical Sciences, General Graduate School, Soonchunhyang University, Asan 31538, Korea
- Department of Pharmaceutical Engineering, College of Medical Sciences, Soonchunhyang University, Asan 31538, Korea
| |
Collapse
|
5
|
Wang K, Liu T, Zhang Y, Lv H, Yao H, Zhao Y, Li J, Li X. Combined Placental Mesenchymal Stem Cells with Guided Nanoparticles Effective Against Diabetic Nephropathy in Mouse Model. Int J Nanomedicine 2024; 19:901-915. [PMID: 38293609 PMCID: PMC10826715 DOI: 10.2147/ijn.s446733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/13/2024] [Indexed: 02/01/2024] Open
Abstract
Background Diabetic nephropathy (DN) is a prevalent complication of diabetes mellitus and constitutes the primary cause of mortality in affected patients. Previous studies have shown that placental mesenchymal stem cells (PL-MSCs) can alleviate kidney dysfunction in animal models of DN. However, the limited ability of mesenchymal stem cells (MSCs) to home to damaged sites restricts their therapeutic potential. Enhancing the precision of PL-MSCs' homing to target tissues is therefore vital for the success of cell therapies in treating DN. Methods We developed Fe3O4 coated polydopamine nanoparticle (NP)-internalized MSCs and evaluated their therapeutic effectiveness in a mouse model of streptozotocin- and high-fat diet-induced DN, using an external magnetic field. Results Our study confirmed that NPs were effectively internalized into PL-MSCs without compromising their intrinsic stem cell properties. The magnetic targeting of PL-MSCs notably improved their homing to the kidney tissues in mice with DN, resulting in enhanced kidney function compared to the transplantation of PL-MSCs alone. Furthermore, the anti-inflammatory and antifibrotic attributes of PL-MSCs played a role in the recovery of kidney function and structure. Conclusion These results demonstrate that magnetically targeted therapy using PL-MSCs is a promising approach for treating diabetic nephropathy.
Collapse
Affiliation(s)
- Ke Wang
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
- Gynecology and Obstetrics Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Te Liu
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Yucheng Zhang
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Huiying Lv
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Hua Yao
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Ye Zhao
- Dermatological Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Jing Li
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Xiuying Li
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| |
Collapse
|
6
|
He L, Zheng Y, Liu M, Dong X, Shen L, He Y, An J, Zhang Y. Nd:YAG-photobiomodulation enhanced ADSCs multilineage differentiation and immunomodulation potentials. Lasers Med Sci 2023; 38:190. [PMID: 37608016 PMCID: PMC10444653 DOI: 10.1007/s10103-023-03818-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 06/17/2023] [Indexed: 08/24/2023]
Abstract
To investigate the effects of Nd: YAG (1064 nm) photobiomodulation on multilineage differentiation and immunomodulation potentials of adipose tissue-derived stem cells (ADSCs) in vitro and in vivo. For in vitro experiments, cells were divided into the control group (non-irradiated control ADSCs) and photobiomodulation groups. 0.5 J/cm2, 1 J/cm2, 2 J/cm2, and 4 J/cm2 were used for proliferation assays; for ADSCs adipogenic differentiation assays, 0.5 J/cm2, 1 J/cm2 were applied; 1 J/cm2 was used for migration and immunomodulation assays. The differentiation abilities were assessed by qPCR, Oil Red O staining, and Alizarin Red staining. The immunomodulation potential was assessed by qPCR and human cytokine array. DSS-induced colitis model. was used to test the effect of photobiomodulation on ADSCs immunomodulation potentials in vivo. Nd:YAG-based photobiomodulation dose-dependently promoted ADSCs proliferation and migration; 1 J/cm2 showed the best promotion effect on proliferation. Moreover, Nd:YAG photobiomodulation promoted ADSCs osteogenic differentiation and brown adipose adipogenic differentiation. The potential immunomodulation assays showed Nd:YAG photobiomodulation improved Anti-inflammation capacity of ADSCs and photobiomodulation irradiated ADSCs effectively alleviated DSS-induced colitis severity in vivo. Our study suggests Nd:YAG photobiomodulation might enhance the ADSCs multilineage differentiation and immunomodulation potentials. These results might help to enhance ADSCs therapeutic effects for clinical application. However, further studies are needed to explore the mechanisms of Nd:YAG photobiomodulation promoting multilineage differentiation and immunomodulation potentials of ADSCs.
Collapse
Affiliation(s)
- Linhai He
- First Clinical Division, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, People's Republic of China
| | - Yi Zheng
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun Nandajie, Haidian District, Beijing, 100081, People's Republic of China
| | - Meng Liu
- Laser and Cosmetic Surgery Division, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, People's Republic of China
| | - Xian Dong
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun Nandajie, Haidian District, Beijing, 100081, People's Republic of China
| | - Lihang Shen
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun Nandajie, Haidian District, Beijing, 100081, People's Republic of China
| | - Yang He
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun Nandajie, Haidian District, Beijing, 100081, People's Republic of China
| | - Jingang An
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun Nandajie, Haidian District, Beijing, 100081, People's Republic of China
| | - Yi Zhang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 22 Zhongguancun Nandajie, Haidian District, Beijing, 100081, People's Republic of China.
| |
Collapse
|
7
|
Kameishi S, Dunn CM, Oka M, Kim K, Cho YK, Song SU, Grainger DW, Okano T. Rapid and effective preparation of clonal bone marrow-derived mesenchymal stem/stromal cell sheets to reduce renal fibrosis. Sci Rep 2023; 13:4421. [PMID: 36932137 PMCID: PMC10023793 DOI: 10.1038/s41598-023-31437-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 03/11/2023] [Indexed: 03/19/2023] Open
Abstract
Allogeneic "off-the-shelf" mesenchymal stem/stromal cell (MSC) therapy requires scalable, quality-controlled cell manufacturing and distribution systems to provide clinical-grade products using cryogenic cell banking. However, previous studies report impaired cell function associated with administering freeze-thawed MSCs as single cell suspensions, potentially compromising reliable therapeutic efficacy. Using long-term culture-adapted clinical-grade clonal human bone marrow MSCs (cBMSCs) in this study, we engineered cBMSC sheets in 24 h to provide rapid preparation. We then sought to determine the influence of cBMSC freeze-thawing on both in vitro production of pro-regenerative factors and in vivo ability to reduce renal fibrosis in a rat model compared to freshly harvested cBMSCs. Sheets from freeze-thawed cBMSCs sheets exhibited comparable in vitro protein production and gene expression of pro-regenerative factors [e.g., hepatocyte growth factor (HGF), vascular endothelial growth factor (VEGF), and interleukin 10 (IL-10)] to freshly harvested cBMSC sheets. Additionally, freeze-thawed cBMSC sheets successfully suppressed renal fibrosis in vivo in an established rat ischemia-reperfusion injury model. Despite previous studies reporting that freeze-thawed MSCs exhibit impaired cell functions compared to fresh MSC single cell suspensions, cell sheets engineered from freeze-thawed cBMSCs do not exhibit impaired cell functions, supporting critical steps toward future clinical translation of cBMSC-based kidney disease treatment.
Collapse
Affiliation(s)
- Sumako Kameishi
- Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA.
- Department of Molecular Pharmaceutics, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, Utah, 84112, USA.
| | - Celia M Dunn
- Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Masatoshi Oka
- Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA
- Department of Molecular Pharmaceutics, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, Utah, 84112, USA
- Department of Nephrology, Tokyo Women's Medical University, Tokyo, Japan
| | - Kyungsook Kim
- Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA
- Department of Molecular Pharmaceutics, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, Utah, 84112, USA
| | | | - Sun U Song
- SCM Lifescience Co., Ltd., Incheon, Republic of Korea
| | - David W Grainger
- Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA
- Department of Molecular Pharmaceutics, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, Utah, 84112, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Teruo Okano
- Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA.
- Department of Molecular Pharmaceutics, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, Utah, 84112, USA.
- Institute for Advanced Biomedical Sciences, Tokyo Women's Medical University, Tokyo, Japan.
| |
Collapse
|
8
|
Mahmoud EA, Baghdadi HM, Hassan R, Ghazy SE. Reparative potential of mesenchymal stem cells and platelet-rich plasma on irradiated submandibular glands of male albino rats. Arch Oral Biol 2023; 150:105674. [PMID: 36907047 DOI: 10.1016/j.archoralbio.2023.105674] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/11/2023] [Accepted: 03/06/2023] [Indexed: 03/14/2023]
Abstract
OBJECTIVE To appraise and compare the reparative role of bone marrow-mesenchymal stem cells (BM-MSCs) and platelet-rich plasma (PRP) against irradiation damage on albino rats' submandibular gland. DESIGN Seventy four male albino rats were used, one for BM-MSCs harvesting, 10 for PRP preparation, seven as control group (Group 1). The remaining 56 rats were subjected to single dose (6 Gy) gamma irradiation and were divided into equal four groups; (Group 2): received no treatment, (Group 3): each rat was injected with 1 × 105 BM-MSCs, (Group 4): each rat was injected with 0.5 ml/kg PRP, and (Group 5): each rat was injected with 1 × 105 BM-MSCs and 0.5 ml/kg PRP. Each group was further subdivided into two subgroups in which rats sacrificed after one and two weeks from irradiation. Any structural changes were examined histopathologically, immunohistochemically using proliferating cell nuclear antigen (PCNA) and CD31 primary antibodies and histochemically using picrosirius red (PSR) stain, then analyzed statistically. RESULTS Histopathological examination of Group 2 showed atrophied acini, with nuclear changes and signs of degeneration in duct systems. Treated groups revealed signs of regeneration in form of uniform acini and regenerated duct systems especially in Group 5 and in a time depended manner. Immunohistochemical examination revealed increased immunoexpression of PCNA and CD31, while histochemical examination showed decreased PSR in all treated groups in relation to the irradiated group and this was proved statistically. CONCLUSIONS BM-MSCs and PRP are effective as treatment for irradiation-induced submandibular gland damage. However, the combined therapy is recommended over each one separately.
Collapse
Affiliation(s)
- Esraa Ali Mahmoud
- Assistant Lecturer of Oral Pathology, Faculty of Dentistry, Ain Shams University, Cairo, Egypt.
| | | | - Rabab Hassan
- Associate Professor of Oral Biology, Faculty of Dentistry, Ain Shams University, Cairo, Egypt
| | - Shaimaa Eliwa Ghazy
- Assistant Professor of Oral Pathology, Faculty of Dentistry, Ain Shams University, Cairo, Egypt
| |
Collapse
|
9
|
Oka M, Kameishi S, Cho YK, Song SU, Grainger DW, Okano T. Clinically Relevant Mesenchymal Stem/Stromal Cell Sheet Transplantation Method for Kidney Disease. Tissue Eng Part C Methods 2023; 29:54-62. [PMID: 36719774 DOI: 10.1089/ten.tec.2022.0200] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Chronic kidney disease (CKD) is the irreversible loss of nephron function, leading to a build-up of toxins, prolonged inflammation, and ultimately fibrosis. Currently, no effective therapies exist to treat CKD due to its complex pathophysiology. Mesenchymal stem/stromal cell (MSC) transplantation is a promising strategy to treat kidney diseases, and multiple clinical trials are currently ongoing. We previously demonstrated that rat bone marrow-derived MSC (BMSC) sheets transplanted onto surgically decapsulated kidney exert therapeutic effects that suppressed renal fibrosis progression based on enhanced vascularization. However, there are clinical concerns about kidney decapsulation such as impaired glomerular filtration rate and Na+ ion and H2O excretion, leading to kidney dysfunction. Therefore, for transitioning from basic research to translational research using cell sheet therapy for kidney disease, it is essential to develop a new cell sheet transplantation strategy without kidney decapsulation. Significantly, we employed cell sheets engineered from clinical-grade human clonal BMSC (cBMSC) and transplanted these onto intact renal capsule to evaluate their therapeutic ability in the rat ischemia-reperfusion injury (IRI) model. Histological analysis 1-day postsurgery showed that cBMSC sheets engrafted well onto intact renal capsule. Interestingly, some grafted cBMSCs migrated into the renal parenchyma. At 1-3 days postsurgery (acute stage), grafted cBMSC sheets prevented tubular epithelial cell injury. At 28 days postsurgery (chronic phase), we observed that grafted cBMSC sheets suppressed renal fibrosis in the rat IRI model. Taken together, engineered cBMSC sheet transplantation onto intact renal capsule suppresses tubular epithelial cell injury and renal fibrosis, supporting further development as a possible clinically relevant strategy. Impact statement Chronic kidney disease (CKD) produces irreversible loss of nephron function, leading to toxemia, prolonged inflammation, and ultimately kidney fibrosis. Currently, no therapies exist to effectively treat CKD due to its complex pathophysiology. Mesenchymal stem/stromal cells (MSCs) are widely known to secret therapeutic paracrine factors, which is expected to provide a new effective therapy for unmet medical needs. However, unsatisfied MSC quality and administration methods to patients limit their therapeutic effects. In this study, we engineered clonal bone marrow-derived MSC sheets and established clinically relevant cell sheet transplantation strategy to treat renal fibrosis, which would improve MSC treatment for kidney disease.
Collapse
Affiliation(s)
- Masatoshi Oka
- Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA.,Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah, USA.,Department of Nephrology, Tokyo Women's Medical University, Tokyo, Japan
| | - Sumako Kameishi
- Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA.,Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah, USA
| | - Yun-Kyoung Cho
- Department of Nephrology, Tokyo Women's Medical University, Tokyo, Japan
| | - Sun U Song
- Department of Nephrology, Tokyo Women's Medical University, Tokyo, Japan
| | - David W Grainger
- Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA.,SCM Lifescience Co., Ltd., Republic of Korea
| | - Teruo Okano
- Cell Sheet Tissue Engineering Center (CSTEC), University of Utah, Salt Lake City, Utah, USA.,Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah, USA.,Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
10
|
Siddiqi S, Klomjit N, Jiang K, Conley SM, Zhu X, Saadiq IM, Ferguson CM, Tang H, Lerman A, Lerman LO. Efficacy of Human Embryonic Stem Cells Compared to Adipose Tissue-Derived Human Mesenchymal Stem/Stromal Cells for Repair of Murine Post-Stenotic Kidneys. Stem Cell Rev Rep 2023; 19:491-502. [PMID: 36048327 PMCID: PMC9905277 DOI: 10.1007/s12015-022-10443-8] [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] [Accepted: 08/08/2022] [Indexed: 02/07/2023]
Abstract
Clinical translation of mesenchymal stem/stromal cell (MSC) therapy has been impeded by the heterogenous nature and limited replicative potential of adult-derived MSCs. Human embryonic stem cell-derived MSCs (hESC-MSCs) that differentiate from immortal cell lines are phenotypically uniform and have shown promise in-vitro and in many disease models. Similarly, adipose tissue-derived MSCs (MSC(AT)) possess potent reparative properties. How these two cell types compare in efficacy, however, remains unknown. We randomly assigned mice to six groups (n = 7-8 each) that underwent unilateral RAS or a sham procedure (3 groups each). Two weeks post-operation, each mouse was administered either vehicle, MSC(AT)s, or hESC-MSCs (5 × 105 cells) into the aorta. Mice were scanned with micro-MRI to determine renal hemodynamics two weeks later and kidneys then harvested. hESC-MSCs and MSC(AT)s were similarly effective at lowering systolic blood pressure. However, MSC(AT)s more robustly increased renal perfusion, oxygenation, and glomerular filtration rate in the post-stenotic kidney, and more effectively mitigated tubular injury, fibrosis, and vascular remodeling. These observations suggest that MSC(AT) are more effective than hESC-MSC in ameliorating kidney dysfunction and tissue injury distal to RAS. Our findings highlight the importance of tissue source in selection of MSCs for therapeutic purposes and underscore the utility of cell-based therapy for kidney disease.
Collapse
Affiliation(s)
- Sarosh Siddiqi
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, 55905, Rochester, MN, USA
| | - Nattawat Klomjit
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, 55905, Rochester, MN, USA
- Division of Nephrology and Hypertension, University of Minnesota, Minneapolis, MN, USA
| | - Kai Jiang
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, 55905, Rochester, MN, USA
| | - Sabena M Conley
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, 55905, Rochester, MN, USA
| | - Xianyang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, 55905, Rochester, MN, USA
| | - Ishran M Saadiq
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, 55905, Rochester, MN, USA
| | - Christopher M Ferguson
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, 55905, Rochester, MN, USA
| | - Hui Tang
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, 55905, Rochester, MN, USA
| | - Amir Lerman
- Department of Cardiovascular Disease, Mayo Clinic, Rochester, MN, USA
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, 55905, Rochester, MN, USA.
| |
Collapse
|
11
|
Carstens MH, García N, Mandayam S, Workeneh B, Pastora I, Calderón C, Bertram KA, Correa D. Safety of Stromal Vascular Fraction Cell Therapy for Chronic Kidney Disease of Unknown Cause (Mesoamerican Nephropathy). Stem Cells Transl Med 2022; 12:7-16. [PMID: 36545894 PMCID: PMC9887091 DOI: 10.1093/stcltm/szac080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 10/29/2022] [Indexed: 12/24/2022] Open
Abstract
Chronic kidney disease of unknown cause (CKDu), also known as Mesoamerican nephropathy, typically presents as an ischemic nephropathy with chronic tubulointerstitial fibrosis in normotensive patients, rapidly progressing to kidney failure. In this first-in-human, open-label, safety study, we followed 18 patients with CKDu (stages 3-5) for 36 months after receiving a single infusion of angiogenic/anti-fibrotic autologous adipose-derived stromal vascular fraction (SVF) cells into their kidneys bilaterally via renal artery catheterization. SVF therapy was safe and well tolerated. There were no SVF-related serious adverse events and no procedural complications. Color Doppler evaluation at 2 months demonstrated increased perfusion to the interlobar and/or arcuate artery levels in each kidney evaluated (36/36) with a reduction in resistance index at the hilar artery (35/36) kidneys. Beyond 12 months, patients with initial eGFR <30 mL/minute/1.73 m2 deteriorated, whereas those ≥30 mL/minute/1.73 m2 further sustained their renal function, suggesting a possible renal protective effect in that group.
Collapse
Affiliation(s)
- Michael H Carstens
- Corresponding author: Michael H. Carstens, MD, FACS, Wake Forest Institute of Regenerative Medicine, 971 Technology Way, Winston-Salem, NC 27101, USA. Tel: +1 571 228 9940;
| | - Nelson García
- Department of Medicine, Universidad Nacional Autónoma de Nicaragua, León, Nicaragua,Nephrology section, Ministerio de Salud, República de Nicaragua
| | - Sreedhar Mandayam
- Department of Nephrology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Biruh Workeneh
- Department of Nephrology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Indiana Pastora
- Department of Medicine, Universidad Nacional Autónoma de Nicaragua, León, Nicaragua
| | - Carlos Calderón
- Department of Cardiology, Hospital San Juan de Dios, San José, Costa Rica
| | - Kenneth A Bertram
- Wake Forest University Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | | |
Collapse
|
12
|
Ahrabi B, Abbaszadeh HA, Piryaei A, Shekari F, Ahmady Roozbahany N, Rouhollahi M, Azam Sayahpour F, Ahrabi M, Azimi H, Moghadasali R. Autophagy-induced mesenchymal stem cell-derived extracellular vesicles ameliorated renal fibrosis in an in vitro model. BIOIMPACTS : BI 2022; 13:359-372. [PMID: 37736337 PMCID: PMC10509741 DOI: 10.34172/bi.2022.24256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 06/20/2022] [Accepted: 06/28/2022] [Indexed: 09/23/2023]
Abstract
Introduction Chronic and progressive damage to the kidney by inflammatory processes, may lead to an increase in the extracellular matrix production, a condition known as renal fibrosis. The current study aims to evaluate if the extracellular vesicles (EVs) derived from autophagic adipose-derived mesenchymal stem cells (ADMSCs) can reduce the inflammation and extracellular matrix accumulation in damaged kidney tissue. Methods Autophagy was induced in ADMSCs using 2µM concentration curcumin and was confirmed by evaluating LC3B, ATG7, and Beclin1 using real-time polymerase chain reaction (PCR) and Western blot. An in vitro renal fibrotic model was established in HEK-293 cells exposed to H2O2 (0.8mM) for 24 and 72 hours. The fibrotic model was confirmed through evaluation of collagen I, transforming growth factor-beta 1 (TGF-β1), E-cadherin, and vimentin genes expression using real-time PCR, collagen I protein by ELISA. After induction of fibrosis for 24 and 72 hours, the HEK cells were treated with NEVs (non-autophagy EVs) (50µM) or AEVs (autophagy EVs) (50µM) at 48, 96, and 124 hours, and then the samples were collected at 72 and 148 hours. Expression of collagen I, TGF-β1, E-cadherin, and vimentin Genes was evaluated via RT-PCR, and protein levels of IL1, TNF-α, IL4, IL10 using ELISA. Results Induction of autophagy using curcumin (2µM) for 24 hours significantly increased LC3B, Beclin1, and ATG7 in the ADMSCs. Upregulation in anti-fibrotic (E-cadherin) and anti-inflammatory (IL4, IL10) gene expression was significantly different in the fibrotic model treated by AEVs compared to NEVs. Also, the downregulation of fibrotic (TGF-β1, vimentin, collagen I) and pro-inflammatory (IL1, TNFα) gene expression was significantly different in AEVs compared with those treated by NEVs. Conclusion Our findings suggest that AEVs can be considered as a therapeutic modality for renal fibrosis in the future.
Collapse
Affiliation(s)
- Behnaz Ahrabi
- Department of Biology and Anatomical Sciences, school of medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Laser Applications in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hojjat Allah Abbaszadeh
- Department of Biology and Anatomical Sciences, school of medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Laser Applications in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Piryaei
- Department of Biology and Anatomical Sciences, school of medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Advanced Therapy Medicinal Product Technology Development Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research, Tehran, Iran
| | | | - Mahya Rouhollahi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Forough Azam Sayahpour
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahnaz Ahrabi
- Department of Endodontics, Dental Faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Azimi
- Department of English Language Teaching, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Moghadasali
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
13
|
The therapeutic potential of Camel Wharton jelly mesenchymal stem cells (CWJ-MSCs) in canine chronic kidney disease model. Stem Cell Res Ther 2022; 13:387. [PMID: 35908006 PMCID: PMC9338563 DOI: 10.1186/s13287-022-03076-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/14/2022] [Indexed: 11/10/2022] Open
Abstract
Background Chronic kidney disease (CKD) is a worldwide health problem that its incidence increases nowadays with the increase in the risk of environmental pollution. CKD can progress to end-stage renal disease (ESRD) which usually ends fatally. This study aimed to examine the therapeutic potential of Camel Wharton jelly-mesenchymal stem cells (CWJ-MSCs) in chronic kidney disease model induced in dogs. Methods CWJ-MSCs were injected directed to the kidney with ultrasonographic guidance in dogs with 5/6 nephrectomy to evaluate its therapeutic potency in such cases. Analysis of variance was applied in normally distributed quantitative variables while a non-parametric Mann–Whitney test was used for non-normally distributed quantitative variables. Results The serum urea and creatinine in the treated group were significantly decreased transferring dogs in the treated group from stage 3 to stage 2 CKD according to the IRIS staging system. Histopathology of renal tissue revealed improving CKD lesions by increasing regeneration of degenerated tubules, maintaining the integrity of glomeruli. New vascularization with blood vessels remodeling were common findings. Periodic acid Schiff stain of renal tissue showed the integrity of renal tubules and thickness of the glomerular basement membrane. Fibrosis of cortex and medulla was lower in the treated group than in the CKD model as monitored by Mallory’s trichrome stain (MTC). NGAL and KIM-1 genes expression were decreased while VEGF and EGF genes expression were increased indicating renal tissue repair. Conclusions CWJ-MSCs have a therapeutic potential in the CKD model induced in dogs.
Collapse
|
14
|
Sharun K, Jambagi K, Kumar R, Gugjoo MB, Pawde AM, Tuli HS, Dhama K, Amarpal. Clinical applications of adipose-derived stromal vascular fraction in veterinary practice. Vet Q 2022; 42:151-166. [PMID: 35841195 PMCID: PMC9364732 DOI: 10.1080/01652176.2022.2102688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Adipose tissue-derived stromal vascular fraction (AdSVF) comprises a heterogeneous cell population, including the multipotent mesenchymal stem cells, hematopoietic stem cells, immune cells, endothelial cells, fibroblasts, and pericytes. As such, multipotent adipose tissue-derived mesenchymal stem cells (AdMSCs), are one of the important components of AdSVF. Commonly used techniques to harvest AdSVF involve enzymatic or non-enzymatic methods. The enzymatic method is considered to be the gold standard technique due to its higher yield. The cellular components of AdSVF can be resuspended in normal saline, platelet-rich plasma, or phosphate-buffered saline to produce a ready-to-use solution. Freshly isolated AdSVF has exhibited promising osteogenic and vasculogenic capacity. AdSVF has already been proven to possess therapeutic potential for osteoarthritis management. It is also an attractive therapeutic option for enhancing wound healing. In addition, the combined use of AdSVF and platelet-rich plasma has an additive stimulatory effect in accelerating wound healing and can be considered an alternative to AdMSC treatment. It is also widely used for managing various orthopaedic conditions in clinical settings and has the potential for regenerating bone, cartilage, and tendons. Autologous AdSVF cells are used along with bone substitutes and other biological factors as an alternative to conventional bone grafting techniques owing to their promising osteogenic and vasculogenic capacity. It can also be used for treating osteonecrosis, meniscus tear, chondromalacia, and tendon injuries in veterinary practice. It has several advantages over in vitro expanded AdMSC, including precluding the need for culturing, reduced risk of cell contamination, and cost-effectiveness, making it ideal for clinical use.
Collapse
Affiliation(s)
- Khan Sharun
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh-243122, India
| | - Kaveri Jambagi
- Division of Medicine, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh-243122, India
| | - Rohit Kumar
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh-243122, India
| | - Mudasir Bashir Gugjoo
- Division of Veterinary Clinical Complex, Faculty of Veterinary Sciences & Animal Husbandry, SKUAST-Kashmir, Shuhama, Srinagar, Jammu and Kashmir-190006, India
| | - Abhijit M Pawde
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh-243122, India
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, Haryana, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh-243122, India
| | - Amarpal
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh-243122, India
| |
Collapse
|
15
|
Søndergaard RH, Højgaard LD, Reese-Petersen AL, Hoeeg C, Mathiasen AB, Haack-Sørensen M, Follin B, Genovese F, Kastrup J, Juhl M, Ekblond A. Adipose-derived stromal cells increase the formation of collagens through paracrine and juxtacrine mechanisms in a fibroblast co-culture model utilizing macromolecular crowding. Stem Cell Res Ther 2022; 13:250. [PMID: 35690799 PMCID: PMC9188050 DOI: 10.1186/s13287-022-02923-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 05/29/2022] [Indexed: 11/24/2022] Open
Abstract
Background Adipose-derived stromal cells (ASCs) possess a multitude of regenerative capabilities, which include immunomodulation, angiogenesis, and stimulation of extracellular matrix (ECM) remodeling. However, the underlying mechanisms leading to ECM remodeling remain largely elusive and highlight the need for functional in vitro models for mode of action studies. Therefore, the purpose of this study was to develop an in vitro co-culture model to investigate the capabilities of ASCs to modulate fibroblasts and ECM. Methods An ECM in vitro model with ASCs and normal human dermal fibroblasts (NHDFs) was established utilizing macromolecular crowding, ascorbic acid, and TGF-β stimulation. Paracrine and juxtacrine co-cultures were created using transwell inserts and cell cultures with direct cell–cell contacts. The cultures were screened using RT2 PCR Profiler Arrays; the protein levels of myofibroblast differentiation marker alpha smooth muscle actin (αSMA) and ECM remodeling enzymes were analyzed using western blot on cell lysates; the formation of collagen type I, III, VI, and fibronectin was investigated using ELISA on culture supernatants; and the deposition of collagens was analyzed using immunocytochemistry. Results TGF-β stimulation of NHDF monocultures increased the expression of 18 transcripts relevant for ECM formation and remodeling, the protein levels of αSMA and matrix metalloproteinase-2 (MMP-2), the formation of collagen type I, III, VI, and fibronectin, and the deposition of collagen type I and VI and decreased the protein levels of MMP-14. Inclusion of ASCs in the ECM co-culture model increased the formation of collagen type I and III through paracrine mechanisms and the formation of collagen type VI through juxtacrine mechanisms. Conclusions The co-culture model provides effective stimulation of NHDF monocultures by TGF-β for enhanced formation and deposition of ECM. In the model, ASCs induce changes in ECM by increasing formation of collagen type I, III and VI. The obtained results could guide further investigations of ASCs’ capabilities and underlying mechanisms related to ECM formation and remodeling. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02923-y.
Collapse
Affiliation(s)
- Rebekka Harary Søndergaard
- Cardiology Stem Cell Centre, The Heart Centre, Copenhagen University Hospital Rigshospitalet, Henrik Harpestrengs vej 4C, Dept. 9302, 2100, Copenhagen, Denmark.
| | - Lisbeth Drozd Højgaard
- Cardiology Stem Cell Centre, The Heart Centre, Copenhagen University Hospital Rigshospitalet, Henrik Harpestrengs vej 4C, Dept. 9302, 2100, Copenhagen, Denmark
| | | | - Cecilie Hoeeg
- Cardiology Stem Cell Centre, The Heart Centre, Copenhagen University Hospital Rigshospitalet, Henrik Harpestrengs vej 4C, Dept. 9302, 2100, Copenhagen, Denmark
| | - Anders Bruun Mathiasen
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Mandana Haack-Sørensen
- Cardiology Stem Cell Centre, The Heart Centre, Copenhagen University Hospital Rigshospitalet, Henrik Harpestrengs vej 4C, Dept. 9302, 2100, Copenhagen, Denmark
| | - Bjarke Follin
- Cardiology Stem Cell Centre, The Heart Centre, Copenhagen University Hospital Rigshospitalet, Henrik Harpestrengs vej 4C, Dept. 9302, 2100, Copenhagen, Denmark
| | - Federica Genovese
- Nordic Bioscience A/S, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| | - Jens Kastrup
- Cardiology Stem Cell Centre, The Heart Centre, Copenhagen University Hospital Rigshospitalet, Henrik Harpestrengs vej 4C, Dept. 9302, 2100, Copenhagen, Denmark
| | - Morten Juhl
- Cardiology Stem Cell Centre, The Heart Centre, Copenhagen University Hospital Rigshospitalet, Henrik Harpestrengs vej 4C, Dept. 9302, 2100, Copenhagen, Denmark
| | - Annette Ekblond
- Cardiology Stem Cell Centre, The Heart Centre, Copenhagen University Hospital Rigshospitalet, Henrik Harpestrengs vej 4C, Dept. 9302, 2100, Copenhagen, Denmark
| |
Collapse
|
16
|
Li Y, Ricardo SD, Samuel CS. Enhancing the Therapeutic Potential of Mesenchymal Stromal Cell-Based Therapies with an Anti-Fibrotic Agent for the Treatment of Chronic Kidney Disease. Int J Mol Sci 2022; 23:ijms23116035. [PMID: 35682717 PMCID: PMC9181689 DOI: 10.3390/ijms23116035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/24/2022] [Accepted: 05/26/2022] [Indexed: 01/02/2023] Open
Abstract
Chronic kidney disease (CKD) affects 1 in 10 members of the general population, placing these patients at an increasingly high risk of kidney failure. Despite the significant burden of CKD on various healthcare systems, there are no effective cures that reverse or even halt its progression. In recent years, human bone-marrow-derived mesenchymal stromal cells (BM-MSCs) have been recognised as a novel therapy for CKDs, owing to their well-established immunomodulatory and tissue-reparative properties in preclinical settings, and their promising safety profile that has been demonstrated in patients with CKDs from several clinical trials. However, renal fibrosis (scarring), a hallmark of CKD, has been shown to impair the viability and functionality of BM-MSCs post-transplantation. This has suggested that BM-MSCs might require a pre-treatment or adjunct therapy that can enhance the viability and therapeutic efficacy of these stromal cells in chronic disease settings. To address this, recent studies that have combined BM-MSCs with the anti-fibrotic drug serelaxin (RLX), have demonstrated the enhanced therapeutic potential of this combination therapy in normotensive and hypertensive preclinical models of CKD. In this review, a critical appraisal of the preclinical data available on the anti-fibrotic and renoprotective actions of BM-MSCs or RLX alone and when combined, as a treatment option for normotensive vs. hypertensive CKD, is discussed.
Collapse
Affiliation(s)
- Yifang Li
- Cardiovascular Disease Program, Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia;
| | - Sharon D. Ricardo
- Development and Stem Cells Program, Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
- Correspondence: (S.D.R.); (C.S.S.)
| | - Chrishan S. Samuel
- Cardiovascular Disease Program, Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia;
- Development and Stem Cells Program, Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Melbourne, VIC 3010, Australia
- Correspondence: (S.D.R.); (C.S.S.)
| |
Collapse
|
17
|
Zhang L, Ma XJN, Fei YY, Han HT, Xu J, Cheng L, Li X. Stem cell therapy in liver regeneration: Focus on mesenchymal stem cells and induced pluripotent stem cells. Pharmacol Ther 2022; 232:108004. [PMID: 34597754 DOI: 10.1016/j.pharmthera.2021.108004] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/11/2021] [Accepted: 09/23/2021] [Indexed: 02/07/2023]
Abstract
The liver has the ability to repair itself after injury; however, a variety of pathological changes in the liver can affect its ability to regenerate, and this could lead to liver failure. Mesenchymal stem cells (MSCs) are considered a good source of cells for regenerative medicine, as they regulate liver regeneration through different mechanisms, and their efficacy has been demonstrated by many animal experiments and clinical studies. Induced pluripotent stem cells, another good source of MSCs, have also made great progress in the establishment of organoids, such as liver disease models, and in drug screening. Owing to the recent developments in MSCs and induced pluripotent stem cells, combined with emerging technologies including graphene, nano-biomaterials, and gene editing, precision medicine and individualized clinical treatment may be realized in the near future.
Collapse
Affiliation(s)
- Lu Zhang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, PR China; Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, PR China; The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China
| | - Xiao-Jing-Nan Ma
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China
| | - Yuan-Yuan Fei
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, PR China; Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, PR China
| | - Heng-Tong Han
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China
| | - Jun Xu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China
| | - Lu Cheng
- Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, PR China
| | - Xun Li
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, PR China; Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, PR China; Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, Lanzhou 730000, PR China; Hepatopancreatobiliary Surgery Institute of Gansu Province, Lanzhou 730000, PR China; The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China.
| |
Collapse
|
18
|
Treatment of Chronic Kidney Disease with Extracellular Vesicles from Mesenchymal Stem Cells and CD133 + Expanded Cells: A Comparative Preclinical Analysis. Int J Mol Sci 2022; 23:ijms23052521. [PMID: 35269664 PMCID: PMC8910174 DOI: 10.3390/ijms23052521] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 01/06/2023] Open
Abstract
Chronic kidney disease (CKD) is characterized by structural abnormalities and the progressive loss of kidney function. Extracellular vesicles (EVs) from human umbilical cord tissue (hUCT)-derived mesenchymal stem cells (MSCs) and expanded human umbilical cord blood (hUCB)-derived CD133+ cells (eCD133+) maintain the characteristics of the parent cells, providing a new form of cell-free treatment. We evaluated the effects of EVs from hUCT-derived MSCs and hUCB-derived CD133+ cells on rats with CDK induced by an adenine-enriched diet. EVs were isolated by ultracentrifugation and characterized by nanoparticle tracking analysis (NTA) and electron microscopy. The animals were randomized and divided into the MSC-EV group, eEPC-EV group and control group. Infusions occurred on the seventh and 14th days after CKD induction. Evaluations of kidney function were carried out by biochemical and histological analyses. Intense labeling of the α-SMA protein was observed when comparing the control with MSC-EVs. In both groups treated with EVs, a significant increase in serum albumin was observed, and the increase in cystatin C was inhibited. The results indicated improvements in renal function in CKD, demonstrating the therapeutic potential of EVs derived from MSCs and eCD133+ cells and suggesting the possibility that in the future, more than one type of EV will be used concurrently.
Collapse
|
19
|
Caneparo C, Sorroza-Martinez L, Chabaud S, Fradette J, Bolduc S. Considerations for the clinical use of stem cells in genitourinary regenerative medicine. World J Stem Cells 2021; 13:1480-1512. [PMID: 34786154 PMCID: PMC8567446 DOI: 10.4252/wjsc.v13.i10.1480] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/12/2021] [Accepted: 09/17/2021] [Indexed: 02/06/2023] Open
Abstract
The genitourinary tract can be affected by several pathologies which require repair or replacement to recover biological functions. Current therapeutic strategies are challenged by a growing shortage of adequate tissues. Therefore, new options must be considered for the treatment of patients, with the use of stem cells (SCs) being attractive. Two different strategies can be derived from stem cell use: Cell therapy and tissue therapy, mainly through tissue engineering. The recent advances using these approaches are described in this review, with a focus on stromal/mesenchymal cells found in adipose tissue. Indeed, the accessibility, high yield at harvest as well as anti-fibrotic, immunomodulatory and proangiogenic properties make adipose-derived stromal/SCs promising alternatives to the therapies currently offered to patients. Finally, an innovative technique allowing tissue reconstruction without exogenous material, the self-assembly approach, will be presented. Despite advances, more studies are needed to translate such approaches from the bench to clinics in urology. For the 21st century, cell and tissue therapies based on SCs are certainly the future of genitourinary regenerative medicine.
Collapse
Affiliation(s)
- Christophe Caneparo
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Quebec G1J1Z4, Canada
| | - Luis Sorroza-Martinez
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Quebec G1J1Z4, Canada
| | - Stéphane Chabaud
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Quebec G1J1Z4, Canada
| | - Julie Fradette
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Quebec G1J1Z4, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Quebec G1V0A6, Canada
| | - Stéphane Bolduc
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Quebec G1J1Z4, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Quebec G1V0A6, Canada
| |
Collapse
|
20
|
Impact of allogeneic feline uterine-derived mesenchymal stromal cell intravenous treatment on renal function of nephrectomized cats with chronic kidney disease. Res Vet Sci 2021; 141:33-41. [PMID: 34653723 DOI: 10.1016/j.rvsc.2021.09.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 08/27/2021] [Accepted: 09/26/2021] [Indexed: 12/20/2022]
Abstract
Chronic kidney disease (CKD) is a common condition and leading cause of mortality in cats. Mesenchymal stromal cells (MSCs) may have a therapeutic effect on CKD. The aim of this pilot study was to determine efficacy of systemically-administered allogeneic uterine tissue-derived MSCs (UMSCs) in cats with CKD. Eighteen renal-compromised, unilaterally nephrectomized cats received two doses of 3 × 107 allogeneic UMSCs given intravenously (IV) with a 2-week dose interval. The primary endpoint was renal function, with treatment success defined by a 20% increase in glomerular filtration rate (GFR; iohexol clearance) and/or a 20% decrease in plasma creatinine in 50% of the cats. Secondary endpoints included diet and water consumption, body weight, urine characteristics, and adverse events. Treatment was well tolerated and associated with a statistically meaningful increase in GFR on Days 13, 28, 57, 99, 121 and 182, compared with baseline (P < 0.0001 for Days 13 to 99 inclusive; P = 0.0029 and P = 0.0225 for Days 121 and 182, respectively). Greater than 50% of the cats demonstrated a 20% increase in GFR on all days except Day 150, at which point GFR measurements were consistently above baseline. Statistically meaningful increases in diet and water consumption were observed. Substantial improvements in GFR were observed throughout the six-month evaluation period (excluding Day 150) in more than 50% of cats, thereby meeting the primary endpoint. Therefore, this IV-administered, allogeneic cellular therapy may support both renal function and clinical status of cats with CKD.
Collapse
|
21
|
Wiśniewska J, Sadowska A, Wójtowicz A, Słyszewska M, Szóstek-Mioduchowska A. Perspective on Stem Cell Therapy in Organ Fibrosis: Animal Models and Human Studies. Life (Basel) 2021; 11:life11101068. [PMID: 34685439 PMCID: PMC8538998 DOI: 10.3390/life11101068] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 12/17/2022] Open
Abstract
Tissue fibrosis is characterized by excessive deposition of extracellular matrix (ECM) components that result from the disruption of regulatory processes responsible for ECM synthesis, deposition, and remodeling. Fibrosis develops in response to a trigger or injury and can occur in nearly all organs of the body. Thus, fibrosis leads to severe pathological conditions that disrupt organ architecture and cause loss of function. It has been estimated that severe fibrotic disorders are responsible for up to one-third of deaths worldwide. Although intensive research on the development of new strategies for fibrosis treatment has been carried out, therapeutic approaches remain limited. Since stem cells, especially mesenchymal stem cells (MSCs), show remarkable self-renewal, differentiation, and immunomodulatory capacity, they have been intensively tested in preclinical studies and clinical trials as a potential tool to slow down the progression of fibrosis and improve the quality of life of patients with fibrotic disorders. In this review, we summarize in vitro studies, preclinical studies performed on animal models of human fibrotic diseases, and recent clinical trials on the efficacy of allogeneic and autologous stem cell applications in severe types of fibrosis that develop in lungs, liver, heart, kidney, uterus, and skin. Although the results of the studies seem to be encouraging, there are many aspects of cell-based therapy, including the cell source, dose, administration route and frequency, timing of delivery, and long-term safety, that remain open areas for future investigation. We also discuss the contemporary status, challenges, and future perspectives of stem cell transplantation for therapeutic options in fibrotic diseases as well as we present recent patents for stem cell-based therapies in organ fibrosis.
Collapse
|
22
|
Lipid Disorders in NAFLD and Chronic Kidney Disease. Biomedicines 2021; 9:biomedicines9101405. [PMID: 34680522 PMCID: PMC8533451 DOI: 10.3390/biomedicines9101405] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/19/2021] [Accepted: 09/30/2021] [Indexed: 12/19/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver dysfunction and is characterized by exaggerated lipid accumulation, inflammation and even fibrosis. It has been shown that NAFLD increases the risk of other chronic diseases, particularly chronic kidney disease (CKD). Lipid in excess could lead to liver and kidney lesions and even end-stage disease through diverse pathways. Dysregulation of lipid uptake, oxidation or de novo lipogenesis contributes to the toxic effects of ectopic lipids which promotes the development and progression of NAFLD and CKD via triggering oxidative stress, apoptosis, pro-inflammatory and profibrotic responses. Importantly, dyslipidemia and release of pro-inflammatory cytokines caused by NAFLD (specifically, nonalcoholic steatohepatitis) are considered to play important roles in the pathological progression of CKD. Growing evidence of similarities between the pathogenic mechanisms of NAFLD and those of CKD has attracted attention and urged researchers to discover their common therapeutic targets. Here, we summarize the current understanding of molecular aberrations underlying the lipid metabolism of NAFLD and CKD and clinical evidence that suggests the relevance of these pathways in humans. This review also highlights the orchestrated inter-organ cross-talk in lipid disorders, as well as therapeutic options and opportunities to counteract NAFLD and CKD.
Collapse
|
23
|
Exosomes derived from mesenchymal stem cells ameliorate renal fibrosis via delivery of miR-186-5p. Hum Cell 2021; 35:83-97. [PMID: 34585365 DOI: 10.1007/s13577-021-00617-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/16/2021] [Indexed: 01/08/2023]
Abstract
Evidence has shown that mesenchymal stem cells' (MSCs) therapy has potential application in treating chronic kidney disease (CKD). In addition, MSCs-derived exosomes can improve the renal function and prevent the progression of CKD. However, the mechanisms by which MSCs-derived exosomes (MSCs-Exo) ameliorate renal fibrosis in CKD remain largely unclear. To mimic an in vitro model of renal fibrosis, rat kidney tubular epithelial cells (NRK52E) were stimulated with transforming growth factor (TGF)-β1. In addition, we established an in vivo model of unilateral ureteric obstruction (UUO)-induced renal fibrosis. Meanwhile, we exploited exosomes derived from MSCs for delivering miR-186-5p agomir into NRK52E cells or kidneys in vitro and in vivo. In this study, we found that level of miR-186-5p was significantly downregulated in TGF-β1-stimulated NRK52E cells and the obstructed kidneys of UUO mice. In addition, miR-186-5p can be transferred from MSCs to NRK52E cells via exosomes. MSCs-delivered miR-186-5p markedly reduced the accumulation of extracellular matrix (ECM) protein, and inhibited epithelial-to-mesenchymal transition (EMT) and apoptosis in TGF-β1-stimulated NRK52E cells. Moreover, exosomal miR-186-5p from MSCs attenuated kidney injury and fibrosis in a UUO mouse model via inhibition of the ECM protein accumulation and EMT process. Meanwhile, dual-luciferase assay showed that miR-186-5p downregulated Smad5 expression via direct binding with the 3'-UTR of Smad5. Collectively then, these findings indicated that exosomal miR-186-5p derived from MSCs could attenuate renal fibrosis in vitro and in vivo by downregulation of Smad5. These findings may help to understand the role of MSCs' exosomes in alleviating renal fibrosis in CKD.
Collapse
|
24
|
Calcat-i-Cervera S, Sanz-Nogués C, O'Brien T. When Origin Matters: Properties of Mesenchymal Stromal Cells From Different Sources for Clinical Translation in Kidney Disease. Front Med (Lausanne) 2021; 8:728496. [PMID: 34616756 PMCID: PMC8488400 DOI: 10.3389/fmed.2021.728496] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/19/2021] [Indexed: 12/14/2022] Open
Abstract
Advanced therapy medicinal products (ATMPs) offer new prospects to improve the treatment of conditions with unmet medical needs. Kidney diseases are a current major health concern with an increasing global prevalence. Chronic renal failure appears after many years of impairment, which opens a temporary window to apply novel therapeutic approaches to delay or halt disease progression. The immunomodulatory, anti-inflammatory, and pro-regenerative properties of mesenchymal stromal cells (MSCs) have sparked interest for their use in cell-based regenerative therapies. Currently, several early-phase clinical trials have been completed and many are ongoing to explore MSC safety and efficacy in a wide range of nephropathies. However, one of the current roadblocks to the clinical translation of MSC therapies relates to the lack of standardization and harmonization of MSC manufacturing protocols, which currently hinders inter-study comparability. Studies have shown that cell culture processing variables can have significant effects on MSC phenotype and functionality, and these are highly variable across laboratories. In addition, heterogeneity within MSC populations is another obstacle. Furthermore, MSCs may be isolated from several sources which adds another variable to the comparative assessment of outcomes. There is now a growing body of literature highlighting unique and distinctive properties of MSCs according to the tissue origin, and that characteristics such as donor, age, sex and underlying medical conditions may alter the therapeutic effect of MSCs. These variables must be taken into consideration when developing a cell therapy product. Having an optimal scale-up strategy for MSC manufacturing is critical for ensuring product quality while minimizing costs and time of production, as well as avoiding potential risks. Ideally, optimal scale-up strategies must be carefully considered and identified during the early stages of development, as making changes later in the bioprocess workflow will require re-optimization and validation, which may have a significant long-term impact on the cost of the therapy. This article provides a summary of important cell culture processing variables to consider in the scale-up of MSC manufacturing as well as giving a comprehensive review of tissue of origin-specific biological characteristics of MSCs and their use in current clinical trials in a range of renal pathologies.
Collapse
Affiliation(s)
| | | | - Timothy O'Brien
- Regenerative Medicine Institute (REMEDI), CÚRAM, Biomedical Science Building, National University of Ireland, Galway, Ireland
| |
Collapse
|
25
|
Corrêa RR, Juncosa EM, Masereeuw R, Lindoso RS. Extracellular Vesicles as a Therapeutic Tool for Kidney Disease: Current Advances and Perspectives. Int J Mol Sci 2021; 22:ijms22115787. [PMID: 34071399 PMCID: PMC8198688 DOI: 10.3390/ijms22115787] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/18/2021] [Accepted: 05/18/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) have been described as important mediators of cell communication, regulating several physiological processes, including tissue recovery and regeneration. In the kidneys, EVs derived from stem cells have been shown to support tissue recovery in diverse disease models and have been considered an interesting alternative to cell therapy. For this purpose, however, several challenges remain to be overcome, such as the requirement of a high number of EVs for human therapy and the need for optimization of techniques for their isolation and characterization. Moreover, the kidney’s complexity and the pathological process to be treated require that EVs present a heterogeneous group of molecules to be delivered. In this review, we discuss the recent advances in the use of EVs as a therapeutic tool for kidney diseases. Moreover, we give an overview of the new technologies applied to improve EVs’ efficacy, such as novel methods of EV production and isolation by means of bioreactors and microfluidics, bioengineering the EV content and the use of alternative cell sources, including kidney organoids, to support their transfer to clinical applications.
Collapse
Affiliation(s)
- Raphael Rodrigues Corrêa
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Estela Mancheño Juncosa
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands;
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands;
- Correspondence: (R.M.); (R.S.L.); Tel.: +31-30-253-3529 (R.M.); Tel.: +55-21-3938-6520 (R.S.L.)
| | - Rafael Soares Lindoso
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands;
- National Institute of Science and Technology for Regenerative Medicine-REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
- Correspondence: (R.M.); (R.S.L.); Tel.: +31-30-253-3529 (R.M.); Tel.: +55-21-3938-6520 (R.S.L.)
| |
Collapse
|
26
|
Mansourabadi AH, Mohamed Khosroshahi L, Noorbakhsh F, Amirzargar A. Cell therapy in transplantation: A comprehensive review of the current applications of cell therapy in transplant patients with the focus on Tregs, CAR Tregs, and Mesenchymal stem cells. Int Immunopharmacol 2021; 97:107669. [PMID: 33965760 DOI: 10.1016/j.intimp.2021.107669] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 02/07/2023]
Abstract
Organ transplantation is a practical treatment for patients with end-stage organ failure. Despite the advances in short-term graft survival, long-term graft survival remains the main challenge considering the increased mortality and morbidity associated with chronic rejection and the toxicity of immunosuppressive drugs. Since a novel therapeutic strategy to induce allograft tolerance seems urgent, focusing on developing novel and safe approaches to prolong graft survival is one of the main goals of transplant investigators. Researchers in the field of organ transplantation are interested in suppressing or optimizing the immune responses by focusing on immune cells including mesenchymal stem cells (MSCs), polyclonal regulatory Tcells (Tregs), and antigen-specific Tregs engineered with chimeric antigen receptors (CAR Tregs). We review the mechanistic pathways, phenotypic and functional characteristics of these cells, and their promising application in organ transplantation.
Collapse
Affiliation(s)
- Amir Hossein Mansourabadi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, 009821 Tehran, Iran; Network of Immunity in Infection, Malignancy, and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), 009821 Tehran, Iran; Systematic Review and Meta-Analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), 009821 Tehran, Iran
| | - Leila Mohamed Khosroshahi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, 009821 Tehran, Iran
| | - Farshid Noorbakhsh
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, 009821 Tehran, Iran.
| | - Aliakbar Amirzargar
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, 009821 Tehran, Iran.
| |
Collapse
|
27
|
Kang SH, Kim MY, Eom YW, Baik SK. Mesenchymal Stem Cells for the Treatment of Liver Disease: Present and Perspectives. Gut Liver 2021; 14:306-315. [PMID: 31581387 PMCID: PMC7234888 DOI: 10.5009/gnl18412] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 12/14/2018] [Accepted: 12/23/2018] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cell transplantation is an emerging therapy for treating chronic liver diseases. The potential of this treatment has been evaluated in preclinical and clinical studies. Although the mechanisms of mesenchymal stem cell transplantation are still not completely understood, accumulating evidence has revealed that their immunomodulation, differentiation, and antifibrotic properties play a crucial role in liver regeneration. The safety and therapeutic effects of mesenchymal stem cells in patients with chronic liver disease have been observed in many clinical studies. However, only modest improvements have been seen, partly because of the limited feasibility of transplanted cells at present. Here, we discuss several strategies targeted at improving viable cell engraftment and the potential challenges in the use of extracellular vesicle-based therapies for liver disease in the future.
Collapse
Affiliation(s)
- Seong Hee Kang
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea.,Cell Therapy and Tissue Engineering Center, Yonsei University Wonju College of Medicine, Wonju, Korea.,Institute of Evidence Based Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Moon Young Kim
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea.,Cell Therapy and Tissue Engineering Center, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Young Woo Eom
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea.,Cell Therapy and Tissue Engineering Center, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Soon Koo Baik
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea.,Cell Therapy and Tissue Engineering Center, Yonsei University Wonju College of Medicine, Wonju, Korea.,Institute of Evidence Based Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| |
Collapse
|
28
|
Elowsson Rendin L, Löfdahl A, Kadefors M, Söderlund Z, Tykesson E, Rolandsson Enes S, Wigén J, Westergren-Thorsson G. Harnessing the ECM Microenvironment to Ameliorate Mesenchymal Stromal Cell-Based Therapy in Chronic Lung Diseases. Front Pharmacol 2021; 12:645558. [PMID: 34040521 PMCID: PMC8142268 DOI: 10.3389/fphar.2021.645558] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/17/2021] [Indexed: 12/21/2022] Open
Abstract
It is known that the cell environment such as biomechanical properties and extracellular matrix (ECM) composition dictate cell behaviour including migration, proliferation, and differentiation. Important constituents of the microenvironment, including ECM molecules such as proteoglycans and glycosaminoglycans (GAGs), determine events in both embryogenesis and repair of the adult lung. Mesenchymal stromal/stem cells (MSC) have been shown to have immunomodulatory properties and may be potent actors regulating tissue remodelling and regenerative cell responses upon lung injury. Using MSC in cell-based therapy holds promise for treatment of chronic lung diseases such as idiopathic pulmonary fibrosis (IPF) and chronic obstructive pulmonary disease (COPD). However, so far clinical trials with MSCs in COPD have not had a significant impact on disease amelioration nor on IPF, where low cell survival rate and pulmonary retention time are major hurdles to overcome. Research shows that the microenvironment has a profound impact on transplanted MSCs. In our studies on acellular lung tissue slices (lung scaffolds) from IPF patients versus healthy individuals, we see a profound effect on cellular activity, where healthy cells cultured in diseased lung scaffolds adapt and produce proteins further promoting a diseased environment, whereas cells on healthy scaffolds sustain a healthy proteomic profile. Therefore, modulating the environmental context for cell-based therapy may be a potent way to improve treatment using MSCs. In this review, we will describe the importance of the microenvironment for cell-based therapy in chronic lung diseases, how MSC-ECM interactions can affect therapeutic output and describe current progress in the field of cell-based therapy.
Collapse
Affiliation(s)
- Linda Elowsson Rendin
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Sotoodehnejadnematalahi F, Moghadasali R, Hajinasrollah M, Ehsani E, Hajizadeh-Saffar E, Sodeifi N, Saidi R, Zarrabi M, Farzanehkhah M, Sadeghi B, Baharvand H, Aghdami N. Immunomodulatory Activity of Human Bone Marrow and Adipose-Derived Mesenchymal Stem Cells Prolongs Allogenic Skin Graft Survival in Nonhuman Primates. CELL JOURNAL 2021; 23:1-13. [PMID: 33650815 PMCID: PMC7944119 DOI: 10.22074/cellj.2021.6895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 11/09/2019] [Indexed: 11/04/2022]
Abstract
Objective In the present study, we examined the tolerance-inducing effects of human adipose-derived mesenchymal stem cells (hAD-MSCs) and bone marrow-derived MSCs (hBM-MSCs) on a nonhuman primate model of skin transplantation. Materials and Methods In this experimental study, allogenic and xenogeneic of immunomodulatory properties of human AD-MSCs and BM-MSCs were evaluated by mixed lymphocyte reaction (MLR) assays. Human MSCs were obtained from BM or AD tissues (from individuals of either sex with an age range of 35 to 65 years) and intravenously injected (2×106 MSCs/kg) after allogeneic skin grafting in a nonhuman primate model. The skin sections were evaluated by H and E staining for histopathological evaluations, particularly inflammation and rejection reaction of grafts after 96 hours of cell injection. At the mRNA and protein levels, cellular mediators of inflammation, such as CD4+IL-17+ (T helper 17; Th17) and CD4+INF-γ+ (T helper 1, Th1) cells, along with CD4+FoxP3+ cells (Treg), as the mediators of immunomodulation, were measured by RT-PCR and flow cytometry analyses. Results A significant Treg cells expansion was observed in MSCs-treated animals which reached the zenith at 24 hours and remained at a high concentration for 96 hours; however, Th1 and Th17 cells were significantly decreased. Our results showed that human MSCs significantly decrease Th1 and Th17 cell proliferation by decreasing interleukin-17 (IL-17) and interferon-γ (INF-γ) production and significantly increase Treg cell proliferation by increasing FoxP3 production. They also extend the allogenic skin graft survival in nonhuman primates. Histological evaluations showed no obvious presence of inflammatory cells or skin redness or even bulging after MSCs injection up to 96 hours, compared to the group without MSCs. There were no significant differences between hBM-MSCs and hAD-MSCs in terms of histopathological scores and inflammatory responses (P<0.05). Conclusion It seems that MSCs could be regarded as a valuable immunomodulatory tool to reduce the use of immunosuppressive agents.
Collapse
Affiliation(s)
| | - Reza Moghadasali
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran. .,Department of Regenerative Medicine, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Mostafa Hajinasrollah
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ehsan Ehsani
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Biology, Roudehen Branch, Islamic Azad University, Roudehen, Iran
| | - Ensiyeh Hajizadeh-Saffar
- Department of Regenerative Medicine, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Niloofar Sodeifi
- Department of Andrology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Reza Saidi
- Department of Surgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Zarrabi
- Royan Stem Cell Technology Company, Cord Blood Bank, Tehran, Iran
| | | | - Bahareh Sadeghi
- Royan Stem Cell Technology Company, Cord Blood Bank, Tehran, Iran
| | - Hossein Baharvand
- Department of Biology, School of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Nasser Aghdami
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| |
Collapse
|
30
|
Wu L, Rong C, Zhou Q, Zhao X, Zhuansun XM, Wan S, Sun MM, Wang SL. Bone Marrow Mesenchymal Stem Cells Ameliorate Cisplatin-Induced Renal Fibrosis via miR-146a-5p/Tfdp2 Axis in Renal Tubular Epithelial Cells. Front Immunol 2021; 11:623693. [PMID: 33664736 PMCID: PMC7921314 DOI: 10.3389/fimmu.2020.623693] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/24/2020] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have regenerative properties in acute kidney injury (AKI). However, the potential function of MSCs in chronic kidney disease remains elusive. Renal fibrosis is the common endpoint of chronic progressive kidney diseases and causes a considerable health burden worldwide. In this study, the protective effects of bone marrow mesenchymal stem cells (BM-MSCs) were assessed in repeated administration of low-dose cisplatin-induced renal fibrosis mouse model in vivo as well as a TGF-β1-induced fibrotic model in vitro. Differentially expressed miRNAs in mouse renal tubular epithelial cells (mRTECs) regulated by BM-MSCs were screened by high-throughput sequencing. We found microRNA (miR)-146a-5p was the most significant up-regulated miRNA in mRTECs. In addition, the gene Tfdp2 was identified as one target gene of miR-146a-5p by bioinformatics analysis. The expression of Tfdp2 in the treatment of BM-MSCs on cisplatin-induced renal injury was evaluated by immunohistochemistry analysis. Our results indicate that BM-MSC attenuates cisplatin-induced renal fibrosis by regulating the miR-146a-5p/Tfdp2 axis in mRTECs.
Collapse
Affiliation(s)
- Lei Wu
- Department of Pathology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou, China.,Collaborative Innovation Center of Clinical Immunology, Sihong People's Hospital, Soochow University, Suzhou, China
| | - Chao Rong
- Department of Pathology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou, China
| | - Qing Zhou
- Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xin Zhao
- Department of Pathology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou, China
| | - Xue-Mei Zhuansun
- Laboratory Animal Research Center, Medical College of Soochow University, Suzhou, China
| | - Shan Wan
- Department of Pathology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou, China
| | - Mao-Min Sun
- Laboratory Animal Research Center, Medical College of Soochow University, Suzhou, China
| | - Shou-Li Wang
- Department of Pathology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou, China.,Collaborative Innovation Center of Clinical Immunology, Sihong People's Hospital, Soochow University, Suzhou, China
| |
Collapse
|
31
|
Akan E, Cetinkaya B, Kipmen-Korgun D, Ozmen A, Koksoy S, Mendilcioğlu İ, Sakinci M, Suleymanlar G, Korgun ET. Effects of amnion derived mesenchymal stem cells on fibrosis in a 5/6 nephrectomy model in rats. Biotech Histochem 2021; 96:594-607. [PMID: 33522283 DOI: 10.1080/10520295.2021.1875502] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Chronic kidney disease (CKD) is characterized by disruption of the glomerulus, tubule and vascular structures by renal fibrosis. Mesenchymal stem cells (MSC) ameliorate CKD. We investigated the effects of human amnion derived MSC (hAMSC) on fibrosis using expression of transforming growth factor beta (TGF-β), collagen type I (COL-1) and bone morphogenetic protein (BMP-7). We also investigated levels of urinary creatinine and nitrogen in CKD. We used a 5/6 nephrectomy (5/6 Nx) induced CKD model. We used 36 rats in six groups of six animals: sham group, 5/6 Nx group, 15 days after 5/6 Nx (5/6 Nx + 15) group, 30 days after 5/6 Nx (5/6 Nx + 30) group, transfer of hAMSC 15 days after 5/6 Nx (5/6 Nx + hAMSC + 15) group and transfer of hAMSC 30 days after 5/6 Nx (5/6 Nx + hAMSC + 30) group. We isolated 106 hAMSC from the amnion and transplanted them via the rat tail vein into the 5/6 Nx + hAMSC + 15 and 5/6 Nx + hAMSC + 30 groups. We measured the expression of BMP-7, COL-1 and TGF-β using western blot and immunohistochemistry, and their gene expressions were analyzed by quantitative real time PCR. TGF-β and COL-1 protein, and gene expressions were increased in the 5/6 Nx +30 group compared to the 5/6 Nx + hAMSC + 30 group. Conversely, both protein and gene expression of BMP-7 was increased in 5/6 Nx + hAMSC + 30 group compared to the 5/6 Nx groups. Increased TGF-β together with decreased BMP-7 expression may cause fibrosis by epithelial-mesenchymal transition due to chronic renal injury. Increased COL-1 levels cause accumulation of extracellular matrix in CKD. Levels of urea, creatinine and nitrogen were increased significantly in 5/6 Nx + 15 and 5/6 Nx + 30 groups compared to the hAMSC groups. We found that hAMSC ameliorate CKD.
Collapse
Affiliation(s)
- Ezgi Akan
- Department of Medical Biochemistry, Akdeniz University Medical School, Antalya, Turkey
| | - Busra Cetinkaya
- Department of Histology and Embryology, Akdeniz University, Medical School, Antalya, Turkey.,Department of Histology and Embryology, Medical Faculty, Bulent Ecevit University, Zonguldak, Turkey
| | - Dijle Kipmen-Korgun
- Department of Medical Biochemistry, Akdeniz University Medical School, Antalya, Turkey
| | - Aslı Ozmen
- Department of Histology and Embryology, Akdeniz University, Medical School, Antalya, Turkey
| | - Sadi Koksoy
- Department of Medical Microbiology and Immunology, Akdeniz University Medical School, Antalya, Turkey
| | - İnanc Mendilcioğlu
- Department of Obstetrics and Gynecology, Akdeniz University Medical School, Antalya, Turkey
| | - Mehmet Sakinci
- Department of Obstetrics and Gynecology, Akdeniz University Medical School, Antalya, Turkey
| | - Gultekin Suleymanlar
- Division of Nephrology, Department of Internal Medicine, Medical Faculty, Akdeniz University, Antalya, Turkey
| | - Emin Turkay Korgun
- Department of Histology and Embryology, Akdeniz University, Medical School, Antalya, Turkey
| |
Collapse
|
32
|
Liu D, Zheng W, Pan S, Liu Z. Concise review: current trends on applications of stem cells in diabetic nephropathy. Cell Death Dis 2020; 11:1000. [PMID: 33221823 PMCID: PMC7680458 DOI: 10.1038/s41419-020-03206-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 12/15/2022]
Abstract
Diabetic nephropathy, with high prevalence, is the main cause of renal failure in diabetic patients. The strategies for treating DN are limited with not only high cost but an unsatisfied effect. Therefore, the effective treatment of DN needs to be explored urgently. In recent years, due to their self-renewal ability and multi-directional differentiation potential, stem cells have exerted therapeutic effects in many diseases, such as graft-versus-host disease, autoimmune diseases, pancreatic diseases, and even acute kidney injury. With the development of stem cell technology, stem cell-based regenerative medicine has been tried to be applied to the treatment of DN. Related stem cells include embryonic stem cells, induced pluripotent stem cells, mesenchymal cells, and endothelial progenitor cells. Undoubtedly, stem cell transplantation has achieved certain results in the treatment of DN animal models. However, stem cell therapy still remains certain thorny issues during treatment. For instance, poor engraftment and limited differentiation of stem cells caused by the diabetic microenvironment, differentiation into unwanted cell lineages, and malignant transformation or genetic aberrations of stem cells. At present, various researches on the therapeutic effects of stem cells in DN with different opinions are reported and the specific mechanism of stem cells is still unclear. We review here the potential mechanism of stem cells as new therapeutic agents in the treatment of DN. Also, we review recent findings and updated information about not only the utilization of stem cells on DN in both preclinical and clinical trials but limitations and future expectations of stem cell-based therapy for DN.
Collapse
Affiliation(s)
- Dongwei Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P.R. China.,Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P.R. China.,Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P.R. China.,Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, 450052, P.R. China
| | - Wen Zheng
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P.R. China.,Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P.R. China.,Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P.R. China.,Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, 450052, P.R. China
| | - Shaokang Pan
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P.R. China.,Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P.R. China.,Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P.R. China.,Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, 450052, P.R. China
| | - Zhangsuo Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P.R. China. .,Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P.R. China. .,Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P.R. China. .,Core Unit of National Clinical Medical Research Center of Kidney Disease, Zhengzhou, 450052, P.R. China.
| |
Collapse
|
33
|
Hong HS, Kim S, Jin Y, Son Y. Substance P enhances the therapeutic effect of MSCs by modulating their angiogenic potential. J Cell Mol Med 2020; 24:12560-12571. [PMID: 32985796 PMCID: PMC7687016 DOI: 10.1111/jcmm.15804] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/30/2020] [Accepted: 08/02/2020] [Indexed: 01/01/2023] Open
Abstract
Bone marrow mesenchymal stem cell (MSC) therapy acts through multiple differentiations in damaged tissue or via secretion of paracrine factors, as demonstrated in various inflammatory and ischaemic diseases. However, long‐term ex vivo culture to obtain a sufficient number of cells in MSC transplantation leads to cellular senescence, deficiency of the paracrine potential, and loss of survival rate post‐transplantation. In this study, we evaluated whether supplementation of MSCs with substance P (SP) can improve their therapeutic potential. SP treatment elevated the secretion of paracrine/angiogenic factors, including VEGF, SDF‐1a and PDGF‐BB, from late passage MSCs in vitro. MSCs supplemented with SP accelerated epidermal/dermal regeneration and neovascularization and suppressed inflammation in vivo, compared to MSCs transplanted alone. Importantly, supplementation with SP enabled the incorporation of transplanted human MSCs into the host vasculature as pericytes via PDGF signalling, leading to the direct engagement of transplanted cells in compact vasculature formation. Our results showed that SP is capable of restoring the cellular potential of senescent stem cells, possibly by modulating the generation of paracrine factors from MSCs, which might accelerate MSC‐mediated tissue repair. Thus, SP is anticipated to be a potential beneficial agent in MSC therapy for inflammatory or ischaemic diseases and cutaneous wounds.
Collapse
Affiliation(s)
- Hyun Sook Hong
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul, Korea.,East-West Medical Research Institute, Kyung Hee University Hospital, Seoul, Korea.,Kyung Hee Institute of Regenerative Medicine (KIRM), Medical Science Research Institute, Kyung Hee University Medical Center, Seoul, Korea
| | - Suna Kim
- Department of Genetic Engineering, College of Life Science, Graduate School of Biotechnology, Kyung Hee University, Yong In, Korea
| | - Yinji Jin
- Department of Genetic Engineering, College of Life Science, Graduate School of Biotechnology, Kyung Hee University, Yong In, Korea
| | - Youngsook Son
- Kyung Hee Institute of Regenerative Medicine (KIRM), Medical Science Research Institute, Kyung Hee University Medical Center, Seoul, Korea.,Department of Genetic Engineering, College of Life Science, Graduate School of Biotechnology, Kyung Hee University, Yong In, Korea
| |
Collapse
|
34
|
Kheradmand A, Hashemitabar M, Kheradmand P, Valizadeh F, Kavosh A. Protective Effect of Wharton's Jelly-derived Mesenchymal Stem Cells on Renal Fibrosis in Rats with Unilateral Ureteral Obstruction. EUR UROL SUPPL 2020; 20:48-53. [PMID: 34337457 PMCID: PMC8317893 DOI: 10.1016/j.euros.2020.06.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2020] [Indexed: 12/11/2022] Open
Abstract
Background Renal failure is a global medical problem. The use of mesenchymal stem cells (MSCs) for preservation and regeneration of renal tissue in acute and chronic kidney diseases has recently been the focus of investigation. Objective To evaluate the protective effect of MSC injections in a rat model of kidney obstruction. Design setting and participants We assigned 15 male Wistar rats to three separate groups: the normal group underwent left nephrectomy; the control group underwent laparotomy and left ureter ligation followed by saline injection into the aorta; and the study group received MSCs injected into the aorta inferior to the left renal artery after ligation of the left ureter. Kidneys were harvested 4 wk later and renal parenchyma samples were used for trichrome staining and for expression analyses. Outcome measurements and statistical analysis The degree of kidney fibrosis was assessed on pathology. Real-time polymerase chain reaction was used to determine expression levels of VEGF, TNF-α, and E-cadherin, and ΔCT and ΔΔCT values were calculated. Data were analyzed using SPSS v19 with paired t tests and nonparametric independent-sample Kruskal-Wallis tests. Results and limitations Fibrosis in the study group decreased from grade 3 or 4 to grade 1. In the control group, TNF-α expression increased and E-cadherin expression decreased. After MSC injection into obstructed kidneys, TNF-α and E-cadherin expression levels decreased and increased respectively, reaching similar levels to those in the normal group. No correlation between tissue regeneration and VEGF levels was observed. More research is needed to focus on other angiogenic factors. Conclusions MSC injection could prevent fibrosis in obstructed rat kidney via alterations in TNF-α and E-cadherin expression. Patient summary We investigated the effect of stem cell injection in rats with kidney obstruction. The treatment led to changes in the levels of two biomarkers and reduced the amount of kidney fibrosis caused by kidney obstruction.
Collapse
Affiliation(s)
- Alireza Kheradmand
- Urology Department, Golestan Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahmood Hashemitabar
- Anatomy and Embryology Department, Ahwaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Parvin Kheradmand
- Pathology Department, Imam Khomaini Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Farzin Valizadeh
- Urology Department, Golestan Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Aryan Kavosh
- Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
35
|
Ramírez-Bajo MJ, Martín-Ramírez J, Bruno S, Pasquino C, Banon-Maneus E, Rovira J, Moya-Rull D, Lazo-Rodriguez M, Campistol JM, Camussi G, Diekmann F. Nephroprotective Potential of Mesenchymal Stromal Cells and Their Extracellular Vesicles in a Murine Model of Chronic Cyclosporine Nephrotoxicity. Front Cell Dev Biol 2020; 8:296. [PMID: 32432111 PMCID: PMC7214690 DOI: 10.3389/fcell.2020.00296] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 04/06/2020] [Indexed: 12/13/2022] Open
Abstract
Background Cell therapies and derived products have a high potential in aiding tissue and organ repairing and have therefore been considered as potential therapies for treating renal diseases. However, few studies have evaluated the impact of these therapies according to the stage of chronic kidney disease. The aim of this study was to evaluate the renoprotective effect of murine bone marrow mesenchymal stromal cells (BM-MSCs), their extracellular vesicles (EVs) and EVs-depleted conditioned medium (dCM) in an aggressive mouse model of chronic cyclosporine (CsA) nephrotoxicity in a preventive and curative manner. Methods After 4 weeks of CsA-treatment (75 mg/kg daily) mice developed severe nephrotoxicity associated with a poor survival rate of 25%, and characterized by tubular vacuolization, casts, and cysts in renal histology. BM-MSC, EVs and dCM groups were administered as prophylaxis or as treatment of CsA nephrotoxicity. The effect of the cell therapies was analyzed by assessing renal function, histological damage, apoptotic cell death, and gene expression of fibrotic mediators. Results Combined administration of CsA and BM-MSCs ameliorated the mice survival rates (6-15%), but significantly renal function, and histological parameters, translating into a reduction of apoptosis and fibrotic markers. On the other hand, EVs and dCM administration were only associated with a partial recovery of renal function or histological damage. Better results were obtained when used as treatment rather than as prophylactic regimen i.e., cell therapy was more effective once the damage was established. Conclusion In this study, we showed that BM-MSCs induce an improvement in renal outcomes in an animal model of CsA nephrotoxicity, particularly if the inflammatory microenvironment is already established. EVs and dCM treatment induce a partial recovery, indicating that further experiments are required to adjust timing and dose for better long-term outcomes.
Collapse
Affiliation(s)
- María José Ramírez-Bajo
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Red de Investigación Renal (REDINREN), Madrid, Spain
| | - Javier Martín-Ramírez
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Stefania Bruno
- Dipartimento di Scienze Mediche, Università degli Studi di Torino, Centro di Biotecnologie Molecolari, Turin, Italy
| | - Chiara Pasquino
- Dipartimento di Scienze Mediche, Università degli Studi di Torino, Centro di Biotecnologie Molecolari, Turin, Italy
| | - Elisenda Banon-Maneus
- Red de Investigación Renal (REDINREN), Madrid, Spain.,Laboratori Experimental de Nefrologia I Trasplantament (LENIT), Fundació Clínic per la Recerca Biomèdica (FCRB), Barcelona, Spain
| | - Jordi Rovira
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Red de Investigación Renal (REDINREN), Madrid, Spain
| | - Daniel Moya-Rull
- Red de Investigación Renal (REDINREN), Madrid, Spain.,Laboratori Experimental de Nefrologia I Trasplantament (LENIT), Fundació Clínic per la Recerca Biomèdica (FCRB), Barcelona, Spain
| | - Marta Lazo-Rodriguez
- Laboratori Experimental de Nefrologia I Trasplantament (LENIT), Fundació Clínic per la Recerca Biomèdica (FCRB), Barcelona, Spain
| | - Josep M Campistol
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Red de Investigación Renal (REDINREN), Madrid, Spain.,Laboratori Experimental de Nefrologia I Trasplantament (LENIT), Fundació Clínic per la Recerca Biomèdica (FCRB), Barcelona, Spain.,Departament de Nefrologia i Trasplantament Renal, ICNU, Hospital Clínic, Barcelona, Spain
| | - Giovanni Camussi
- Dipartimento di Scienze Mediche, Università degli Studi di Torino, Centro di Biotecnologie Molecolari, Turin, Italy
| | - Fritz Diekmann
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Red de Investigación Renal (REDINREN), Madrid, Spain.,Laboratori Experimental de Nefrologia I Trasplantament (LENIT), Fundació Clínic per la Recerca Biomèdica (FCRB), Barcelona, Spain.,Departament de Nefrologia i Trasplantament Renal, ICNU, Hospital Clínic, Barcelona, Spain
| |
Collapse
|
36
|
Ahmadi A, Rad NK, Ezzatizadeh V, Moghadasali R. Kidney Regeneration: Stem Cells as a New Trend. Curr Stem Cell Res Ther 2020; 15:263-283. [DOI: 10.2174/1574888x15666191218094513] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 12/23/2022]
Abstract
Renal disease is a major worldwide public health problem that affects one in ten people.
Renal failure is caused by the irreversible loss of the structural and functional units of kidney (nephrons)
due to acute and chronic injuries. In humans, new nephrons (nephrogenesis) are generated until
the 36th week of gestation and no new nephron develops after birth. However, in rodents, nephrogenesis
persists until the immediate postnatal period. The postnatal mammalian kidney can partly repair
their nephrons. The kidney uses intrarenal and extra-renal cell sources for maintenance and repair.
Currently, it is believed that dedifferentiation of surviving tubular epithelial cells and presence of resident
stem cells have important roles in kidney repair. Many studies have shown that stem cells obtained
from extra-renal sites such as the bone marrow, adipose and skeletal muscle tissues, in addition
to umbilical cord and amniotic fluid, have potential therapeutic benefits. This review discusses the
main mechanisms of renal regeneration by stem cells after a kidney injury.
Collapse
Affiliation(s)
- Amin Ahmadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Niloofar K. Rad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Vahid Ezzatizadeh
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Reza Moghadasali
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
37
|
Liu Q, Lv S, Liu J, Liu S, Wang Y, Liu G. Mesenchymal stem cells modified with angiotensin-converting enzyme 2 are superior for amelioration of glomerular fibrosis in diabetic nephropathy. Diabetes Res Clin Pract 2020; 162:108093. [PMID: 32109518 DOI: 10.1016/j.diabres.2020.108093] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 02/14/2020] [Accepted: 02/24/2020] [Indexed: 01/06/2023]
Abstract
AIMS This study aimed to detect the effect of angiotensin-converting enzyme (ACE) 2-modified mesenchymal stem cells (MSCs) on glomerular fibrosis in vitro and in vivo and investigate the underlying molecular mechanism. METHODS MSCs transduced with the ACE2 gene (MSCs-ACE2) were cocultured with glomerular mesangial cells (GMCs) following Ang II stimulation. MSCs-ACE2 were transplanted into streptozotocin-induced diabetic rats. Physical, biochemical and morphological parameters were measured, and fibrotic indicators and renin-angiotensin system (RAS) components in GMCs and kidney tissues were assessed. RESULTS The transduction efficiency of MSCs was as high as 85%. The modified MSCs secreted soluble ACE2 protein into the culture medium. After transplantation into rats with diabetes, MSCs-ACE2 targeted injured kidneys and enhanced local expression of ACE2. Compared with MSC treatment alone, MSC-ACE2 treatment was superior in reducing albuminuria and improving glomerulosclerosis. In vitro and in vivo, MSCs-ACE2 were more beneficial than MSCs alone in decreasing Ang II and increasing Ang1-7, thereby inhibiting the detrimental effects of Ang II accumulation by downregulating collagen I and fibronectin (FN) expression and inhibiting the transforming growth factor (TGF-β)/Smad pathway. CONCLUSIONS MSCs modified with ACE2 therapy have additional benefits to the progression of diabetic nephropathy (DN) by inhibiting renal RAS activation and reducing glomerular fibrosis.
Collapse
Affiliation(s)
- Qingzhen Liu
- Nephrology Research Institute of Shandong University, The Second Hospital of Shandong University, Shandong University, Jinan, Shandong, China
| | - Shasha Lv
- Nephrology Research Institute of Shandong University, The Second Hospital of Shandong University, Shandong University, Jinan, Shandong, China
| | - Jiaxi Liu
- College of Liberal Arts, University of Minnesota, USA
| | - Shanshan Liu
- Nephrology Research Institute of Shandong University, The Second Hospital of Shandong University, Shandong University, Jinan, Shandong, China
| | - Yinghui Wang
- Nephrology Research Institute of Shandong University, The Second Hospital of Shandong University, Shandong University, Jinan, Shandong, China
| | - Gang Liu
- Nephrology Research Institute of Shandong University, The Second Hospital of Shandong University, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
38
|
Abumoawad A, Saad A, Ferguson CM, Eirin A, Herrmann SM, Hickson LJ, Goksu BB, Bendel E, Misra S, Glockner J, Dietz AB, Lerman LO, Textor SC. In a Phase 1a escalating clinical trial, autologous mesenchymal stem cell infusion for renovascular disease increases blood flow and the glomerular filtration rate while reducing inflammatory biomarkers and blood pressure. Kidney Int 2020; 97:793-804. [PMID: 32093917 PMCID: PMC7284953 DOI: 10.1016/j.kint.2019.11.022] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 11/14/2019] [Accepted: 11/22/2019] [Indexed: 02/08/2023]
Abstract
Atherosclerotic renovascular disease (ARVD) reduces tissue perfusion and eventually leads to loss of kidney function with limited therapeutic options. Here we describe results of Phase 1a escalating dose clinical trial of autologous mesenchymal stem cell infusion for ARVD. Thirty-nine patients with ARVD were studied on two occasions separated by three months. Autologous adipose-derived mesenchymal stem cells were infused through the renal artery in 21 patients at three different dose levels (1, 2.5 and 5.0 × 105 cells/kg) in seven patients each. We measured renal blood flow, glomerular filtration rate (GFR) (iothalamate and estimated GFR), renal vein cytokine levels, blood pressure, and tissue oxygenation before and three months after stem cell delivery. These indices were compared to those of 18 patients with ARVD matched for age, kidney function and blood pressure receiving medical therapy alone that underwent an identical study protocol. Cultured mesenchymal stem cells were also studied in vitro. For the entire stem cell treated-cohort, mean renal blood flow in the treated stenotic kidney significantly increased after stem cell infusion from (164 to 190 ml/min). Hypoxia, renal vein inflammatory cytokines, and angiogenic biomarkers significantly decreased following stem cell infusion. Mean systolic blood pressure significantly fell (144 to 136 mmHg) and the mean two-kidney GFR (Iothalamate) modestly but significantly increased from (53 to 56 ml/min). Changes in GFR and blood pressure were largest in the high dose stem cell treated individuals. No such changes were observed in the cohort receiving medical treatment alone. Thus, our data demonstrate the potential for autologous mesenchymal stem cell to increase blood flow, GFR and attenuate inflammatory injury in post-stenotic kidneys. The observation that some effects are dose-dependent and related to in-vitro properties of mesenchymal stem cell may direct efforts to maximize potential therapeutic efficacy.
Collapse
Affiliation(s)
| | - Ahmed Saad
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA; Department of Family Medicine, Creighton University, Omaha, Nebraska, USA
| | | | - Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Sandra M Herrmann
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - LaTonya J Hickson
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Busra B Goksu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Emily Bendel
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sanjay Misra
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - James Glockner
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Allan B Dietz
- Transfusion Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Stephen C Textor
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA.
| |
Collapse
|
39
|
Sun DZ, Abelson B, Babbar P, Damaser MS. Harnessing the mesenchymal stem cell secretome for regenerative urology. Nat Rev Urol 2020; 16:363-375. [PMID: 30923338 DOI: 10.1038/s41585-019-0169-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The extensive arsenal of bioactive molecules secreted by mesenchymal stem cells (MSCs), known as the secretome, has demonstrated considerable therapeutic benefit in regenerative medicine. Investigation into the therapeutic potential of the secretome has enabled researchers to replicate the anti-inflammatory, pro-angiogenic and trophic effects of stem cells without the need for the cells themselves. Furthermore, treatment with the MSC secretome could circumvent hurdles associated with cellular therapy, including oncogenic transformation, immunoreactivity and cost. Thus, a clear rationale exists for investigating the therapeutic potential of the MSC secretome in regenerative urology. Indeed, preclinical studies have demonstrated the therapeutic benefits of the MSC secretome in models of stress urinary incontinence, renal disease, bladder dysfunction and erectile dysfunction. However, the specific mechanisms underpinning therapeutic activity are unclear and require further research before clinical translation. Improvements in current proteomic methods used to characterize the secretome will be necessary to provide further insight into stem cells and their secretome in regenerative urology.
Collapse
Affiliation(s)
- Daniel Z Sun
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA. .,Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, OH, USA. .,Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| | - Benjamin Abelson
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA.,Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, OH, USA.,Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Paurush Babbar
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA.,Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, OH, USA.,Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Margot S Damaser
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA.,Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, OH, USA.,Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.,Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| |
Collapse
|
40
|
Ogbadu J, Singh G, Aggarwal D. Factors affecting the transition of acute kidney injury to chronic kidney disease: Potential mechanisms and future perspectives. Eur J Pharmacol 2019; 865:172711. [DOI: 10.1016/j.ejphar.2019.172711] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 09/22/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022]
|
41
|
Kim HS, Lee JS, Lee HK, Park EJ, Jeon HW, Kang YJ, Lee TY, Kim KS, Bae SC, Park JH, Han SB. Mesenchymal Stem Cells Ameliorate Renal Inflammation in Adriamycin-induced Nephropathy. Immune Netw 2019; 19:e36. [PMID: 31720047 PMCID: PMC6829076 DOI: 10.4110/in.2019.19.e36] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 09/27/2019] [Accepted: 10/02/2019] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSCs) ameliorate the renal injury in Adriamycin (ADR)-induced nephropathy, but the mechanisms underlying their efficacy remain incompletely understood. In this study, we demonstrated that MSCs increased the survival, recovered body weight loss, and decreased proteinuria and serum creatinine levels in ADR-treated mice. MSCs also prevented podocyte damage and renal fibrosis by decreasing the expression of fibronectin, collagen 1α1, and α-smooth muscle actin. From a mechanistic perspective, MSCs inhibited renal inflammation by lowering the expression of CCL4, CCL7, CCL19, IFN-α/β, TGF-β, TNF-α, and chitinase 3-like 1. In summary, our data demonstrate that MSCs improve renal functions by inhibiting renal inflammation in ADR-induced nephropathy.
Collapse
Affiliation(s)
- Hyung Sook Kim
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Korea
| | - Jae Seob Lee
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Korea
| | - Hong Kyung Lee
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Korea
| | - Eun Jae Park
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Korea
| | - Hye Won Jeon
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Korea
| | - Yu Jeong Kang
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Korea
| | - Tae Yong Lee
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Korea
- Bioengineering Institute, Corestem Inc., Seoul 04763, Korea
| | - Kyung Suk Kim
- Bioengineering Institute, Corestem Inc., Seoul 04763, Korea
| | - Sang-Cheol Bae
- Hanyang University Hospital for Rheumatic Diseases, Seoul 04763, Korea
| | - Ji Hyun Park
- College of Pharmacy, Korea University, Sejong 30019, Korea
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Korea
| |
Collapse
|
42
|
Conrad S, Weber K, Walliser U, Geburek F, Skutella T. Stem Cell Therapy for Tendon Regeneration: Current Status and Future Directions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1084:61-93. [PMID: 30043235 DOI: 10.1007/5584_2018_194] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In adults the healing tendon generates fibrovascular scar tissue and recovers never histologically, mechanically, and functionally which leads to chronic and to degenerative diseases. In this review, the processes and mechanisms of tendon development and fetal regeneration in comparison to adult defect repair and degeneration are discussed in relation to regenerative therapeutic options. We focused on the application of stem cells, growth factors, transcription factors, and gene therapy in tendon injury therapies in order to intervene the scarring process and to induce functional regeneration of the lesioned tissue. Outlines for future therapeutic approaches for tendon injuries will be provided.
Collapse
Affiliation(s)
| | - Kathrin Weber
- Tierärztliches Zentrum für Pferde in Kirchheim Altano GmbH, Kirchheim unter Teck, Germany
| | - Ulrich Walliser
- Tierärztliches Zentrum für Pferde in Kirchheim Altano GmbH, Kirchheim unter Teck, Germany
| | - Florian Geburek
- Justus-Liebig-University Giessen, Faculty of Veterinary Medicine, Clinic for Horses - Department of Surgery, Giessen, Germany
| | - Thomas Skutella
- Institute for Anatomy and Cell Biology, Medical Faculty, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
43
|
Shi Y, Wang Y, Li Q, Liu K, Hou J, Shao C, Wang Y. Immunoregulatory mechanisms of mesenchymal stem and stromal cells in inflammatory diseases. Nat Rev Nephrol 2019; 14:493-507. [PMID: 29895977 DOI: 10.1038/s41581-018-0023-5] [Citation(s) in RCA: 779] [Impact Index Per Article: 129.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cells (MSCs; also referred to as mesenchymal stromal cells) have attracted much attention for their ability to regulate inflammatory processes. Their therapeutic potential is currently being investigated in various degenerative and inflammatory disorders such as Crohn's disease, graft-versus-host disease, diabetic nephropathy and organ fibrosis. The mechanisms by which MSCs exert their therapeutic effects are multifaceted, but in general, these cells are thought to enable damaged tissues to form a balanced inflammatory and regenerative microenvironment in the presence of vigorous inflammation. Studies over the past few years have demonstrated that when exposed to an inflammatory environment, MSCs can orchestrate local and systemic innate and adaptive immune responses through the release of various mediators, including immunosuppressive molecules, growth factors, exosomes, chemokines, complement components and various metabolites. Interestingly, even nonviable MSCs can exert beneficial effects, with apoptotic MSCs showing immunosuppressive functions in vivo. Because the immunomodulatory capabilities of MSCs are not constitutive but rather are licensed by inflammatory cytokines, the net outcomes of MSC activation might vary depending on the levels and the types of inflammation within the residing tissues. Here, we review current understanding of the immunomodulatory mechanisms of MSCs and the issues related to their therapeutic applications.
Collapse
Affiliation(s)
- Yufang Shi
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, China. .,CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences/Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Yu Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences/Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qing Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences/Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Keli Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences/Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jianquan Hou
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, China
| | - Changshun Shao
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, China
| | - Ying Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences/Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
44
|
Khalifa J, François S, Rancoule C, Riccobono D, Magné N, Drouet M, Chargari C. Gene therapy and cell therapy for the management of radiation damages to healthy tissues: Rationale and early results. Cancer Radiother 2019; 23:449-465. [PMID: 31400956 DOI: 10.1016/j.canrad.2019.06.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 06/06/2019] [Indexed: 12/14/2022]
Abstract
Nowadays, ionizing radiations have numerous applications, especially in medicine for diagnosis and therapy. Pharmacological radioprotection aims at increasing detoxification of free radicals. Radiomitigation aims at improving survival and proliferation of damaged cells. Both strategies are essential research area, as non-contained radiation can lead to harmful effects. Some advances allowing the comprehension of normal tissue injury mechanisms, and the discovery of related predictive biomarkers, have led to developing several highly promising radioprotector or radiomitigator drugs. Next to these drugs, a growing interest does exist for biotherapy in this field, including gene therapy and cell therapy through mesenchymal stem cells. In this review article, we provide an overview of the management of radiation damages to healthy tissues via gene or cell therapy in the context of radiotherapy. The early management aims at preventing the occurrence of these damages before exposure or just after exposure. The late management offers promises in the reversion of constituted late damages following irradiation.
Collapse
Affiliation(s)
- J Khalifa
- Départment de radiothérapie, institut Claudius-Regaud, institut universitaire du cancer de Toulouse - Oncopole, 1, avenue Irène-Joliot-Curie, 31100 Toulouse, France.
| | - S François
- Institut de recherche biomédicale des armées, BP73, 91223 Brétigny-sur-Orge cedex, France
| | - C Rancoule
- Département de radiothérapie, institut de cancérologie de la Loire Lucien-Neuwirth, 108 bis, avenue Albert-Raimond, 42270 Saint-Priest-en-Jarez, France; Laboratoire de radiobiologie cellulaire et moléculaire, UMR 5822, institut de physique nucléaire de Lyon (IPNL), 69622 Villeurbanne, France; UMR 5822, CNRS, domaine scientifique de la Doua, 4, rue Enrico-Fermi, 69622 Villeurbanne cedex, France; UMR 5822, université Lyon 1, domaine scientifique de la Doua, 4, rue Enrico-Fermi, 69622 Villeurbanne cedex, France; UMR 5822, université de Lyon, domaine scientifique de la Doua, 4, rue Enrico-Fermi, 69622 Villeurbanne cedex, France
| | - D Riccobono
- Institut de recherche biomédicale des armées, BP73, 91223 Brétigny-sur-Orge cedex, France
| | - N Magné
- Département de radiothérapie, institut de cancérologie de la Loire Lucien-Neuwirth, 108 bis, avenue Albert-Raimond, 42270 Saint-Priest-en-Jarez, France; Laboratoire de radiobiologie cellulaire et moléculaire, UMR 5822, institut de physique nucléaire de Lyon (IPNL), 69622 Villeurbanne, France; UMR 5822, CNRS, domaine scientifique de la Doua, 4, rue Enrico-Fermi, 69622 Villeurbanne cedex, France; UMR 5822, université Lyon 1, domaine scientifique de la Doua, 4, rue Enrico-Fermi, 69622 Villeurbanne cedex, France; UMR 5822, université de Lyon, domaine scientifique de la Doua, 4, rue Enrico-Fermi, 69622 Villeurbanne cedex, France
| | - M Drouet
- Institut de recherche biomédicale des armées, BP73, 91223 Brétigny-sur-Orge cedex, France
| | - C Chargari
- Institut de recherche biomédicale des armées, BP73, 91223 Brétigny-sur-Orge cedex, France; Service de santé des armées, école du Val-de-Grâce, 74, boulevard de Port-Royal, 75005 Paris, France; Département de radiothérapie, Gustave-Roussy Cancer Campus, 114, rue Édouard-Vailant, 94805 Villejuif, France
| |
Collapse
|
45
|
Liu B, Ding FX, Liu Y, Xiong G, Lin T, He DW, Zhang YY, Zhang DY, Wei GH. Human umbilical cord-derived mesenchymal stem cells conditioned medium attenuate interstitial fibrosis and stimulate the repair of tubular epithelial cells in an irreversible model of unilateral ureteral obstruction. Nephrology (Carlton) 2019; 23:728-736. [PMID: 28667820 DOI: 10.1111/nep.13099] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 05/11/2017] [Accepted: 06/25/2017] [Indexed: 01/06/2023]
Abstract
AIM The growing number of patients suffering from chronic renal disease (CKD) is a challenge for the development of innovative therapies. Researchers have studied the therapeutic effects of cell therapy in acute kidney injury (AKI). However, the therapeutic effect of conditional medium (CM) in the CKD models have been rarely reported. Here, we examined the effects of umbilical cord derived-mesenchymal stem cells (hUC-MSCs) CM on renal fibrosis in a rat model of unilateral ureteral obstruction (UUO). METHODS Animals were randomly divided into three groups: sham-operated, UUO, UUO + CM. CM was administered via the left renal artery after total ligation of the left ureter. Rats were killed after 14 days of obstruction. Histological changes and oxidative stress parameters were assessed. Western blotting and immunohistochemistry analysis were used to measure epithelial-mesenchymal transition (EMT) markers, including epithelial cadherin (E-cadherin), α-smooth muscle actin (α-SMA), tumour necrosis factor-α (TNF-α), Collagen-I, and transforming growth factor β1 (TGF-β1). Proliferation and apoptosis of renal tubular epithelial cells (RTEs) were also measured. RESULTS HucMSC-CM significantly reduced the levels of malondialdehyde (MDA) and reactive oxygen species (ROS), and increased the activity of glutathione (GSH) induced by UUO. Moreover, CM significantly reduced the expression of TGF-β1, α-SMA, TNF-α and Collagen-I in UUO kidney, promoted the proliferation of RTEs and inhibited its apoptosis. In addition, the increased expression of E-cadherin also reflects the effective improvement of renal interstitial fibrosis. CONCLUSION This study shows that CM protects UUO-induced kidney damage and therefore could be a potential tool to prevent CKD progression.
Collapse
Affiliation(s)
- Bo Liu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| | - Feng-Xia Ding
- Department of Respiratory Medicine, Chongqing Key Laboratory of Pediatrics, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| | - Yang Liu
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| | - Geng Xiong
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Tao Lin
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Da-Wei He
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yuan-Yuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - De-Ying Zhang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, China
| | - Guang-Hui Wei
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
46
|
Shi X, Liu J, Chen D, Zhu M, Yu J, Xie H, Zhou L, Li L, Zheng S. MSC-triggered metabolomic alterations in liver-resident immune cells isolated from CCl 4-induced mouse ALI model. Exp Cell Res 2019; 383:111511. [PMID: 31362001 DOI: 10.1016/j.yexcr.2019.111511] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 07/04/2019] [Accepted: 07/24/2019] [Indexed: 12/29/2022]
Abstract
Clinical trials testing mesenchymal stem cell (MSC) as a cellular remedy for acute liver injury (ALI) are underway, but its underlying mechanism has not been thoroughly scrutinized. We highlight that the metabolomic profile of the liver-resident immune cells is significantly altered after MSC administration; its potential correlation with ALI remission is discussed in this study. C57BL/6 mice are randomly divided into three groups: the sham group, MSC-treated ALI group and PBS-treated ALI group; acute liver injury is induced by intraperitoneal injection of carbon tetrachloride. A high-performance chemical isotope labeling liquid chromatography-mass spectrometry (CIL LC-MS) is exploited to profile amine, phenol and carbonyl submetabolome of the liver-resident immune cells in different treatments. 4295 peak pairs are quantified and 2461 peak pairs are further identified in zero-reaction and one-reaction libraries. Clear separation of the three groups is observed in the global PCA and OPLS-DA analyses. We identified 256 metabolites to be candidate biomarkers for ALI-activated immunity and 114 metabolites to be candidate biomarkers for MSC-modulated immunity. Ariginine, aspartate and glutamate metabolism are most affected in both cases, with eight significantly regulated metabolites as joints (glutamic-gamma-semialdehyde, aspartate acid, glutamate acid, gamma-Aminobutyric acidorinithine, 2-keto-glutaramic acid, N-acetylornithine, citrulline and ornithine). These findings shed new light on the therapeutic benefit of immune modulation during ALI rescue. It needs to be further investigated whether exogenous supply of certain metabolites will exert a profound impact on the metabolic network, crosstalking with immune responses and modulating ALI prognosis.
Collapse
Affiliation(s)
- Xiaowei Shi
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Rd., Hangzhou, 310003, China; NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, 310003, China.
| | - Jingqi Liu
- Department of Chemistry, University of Alberta, Edmonton, Alberta, T6G 2G2, Canada; State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Rd., Hangzhou, 310003, China.
| | - Deying Chen
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Rd., Hangzhou, 310003, China; State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Rd., Hangzhou, 310003, China.
| | - Minglei Zhu
- Department of Chemistry, University of Alberta, Edmonton, Alberta, T6G 2G2, Canada.
| | - Jiong Yu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Rd., Hangzhou, 310003, China; State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Rd., Hangzhou, 310003, China.
| | - Haiyang Xie
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Rd., Hangzhou, 310003, China; NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, 310003, China.
| | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Rd., Hangzhou, 310003, China; NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, 310003, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Rd., Hangzhou, 310003, China.
| | - Liang Li
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Rd., Hangzhou, 310003, China; Department of Chemistry, University of Alberta, Edmonton, Alberta, T6G 2G2, Canada.
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Rd., Hangzhou, 310003, China; NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, 310003, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Rd., Hangzhou, 310003, China.
| |
Collapse
|
47
|
Villanueva S, González F, Lorca E, Tapia A, Valentina López G, Strodthoff R, Fajre F, Carreño JE, Valjalo R, Vergara C, Lecanda M, Bartolucci J, Figueroa FE, Khoury M. Adipose tissue-derived mesenchymal stromal cells for treating chronic kidney disease: A pilot study assessing safety and clinical feasibility. Kidney Res Clin Pract 2019; 38:176-185. [PMID: 31189223 PMCID: PMC6577210 DOI: 10.23876/j.krcp.18.0139] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/25/2019] [Accepted: 02/18/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) is a growing public health concern, and available treatments are insufficient in limiting disease progression. New strategies, including regenerative cell-based therapies, have emerged as therapeutic alternatives. Results from several groups, including our own, have reported evidence of a supportive role for mesenchymal stromal cells (MSCs) in functional recovery and prevention of tissue damage in murine models of CKD. Prompted by these data, an open pilot study was conducted to assess the safety and efficacy of a single injection of autologous adipose tissue-derived MSCs (AT-MSCs) for treatment of CKD. METHODS AT-MSCs were infused intravenously into six CKD patients at a dose of 1 million cells/kg. Patients were stabilized and followed for one year prior to MSC infusion and one year following infusion. RESULTS No patients presented with adverse effects. Statistically significant improvement in urinary protein excretion was observed in AT-MSCs transplanted patients, from a median of 0.75 g/day (range, 0.15-9.57) at baseline to 0.54 g/day (range, 0.01-2.66) at month 12 (P = 0.046). The glomerular filtration rate was not significantly decreased post-infusion of AT-MSCs. CONCLUSION Findings from this pilot study demonstrate that intravenous infusion of autologous expanded AT-MSCs into CKD patients was not associated with adverse effects and could benefit patients already undergoing standard medical treatment.
Collapse
Affiliation(s)
- Sandra Villanueva
- Laboratory of Molecular and Integrative Physiology, Faculty of Medicine, Universidad de Los Andes, Santiago,
Chile
| | | | - Eduardo Lorca
- Department of Nephrology, Hospital Salvador, Santiago,
Chile
| | - Andrés Tapia
- Laboratory of Nano-regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago,
Chile
| | - G Valentina López
- Cells for Cells, Faculty of Medicine, Universidad de Los Andes, Santiago,
Chile
| | - Rocío Strodthoff
- Laboratory of Molecular and Integrative Physiology, Faculty of Medicine, Universidad de Los Andes, Santiago,
Chile
| | - Francisca Fajre
- Laboratory of Molecular and Integrative Physiology, Faculty of Medicine, Universidad de Los Andes, Santiago,
Chile
| | - Juan E. Carreño
- Laboratory of Molecular and Integrative Physiology, Faculty of Medicine, Universidad de Los Andes, Santiago,
Chile
| | - Ricardo Valjalo
- Department of Nephrology, Hospital Salvador, Santiago,
Chile
| | - César Vergara
- Laboratory of Molecular and Integrative Physiology, Faculty of Medicine, Universidad de Los Andes, Santiago,
Chile
| | - Manuel Lecanda
- Laboratory of Molecular and Integrative Physiology, Faculty of Medicine, Universidad de Los Andes, Santiago,
Chile
| | - Jorge Bartolucci
- Cells for Cells, Faculty of Medicine, Universidad de Los Andes, Santiago,
Chile
| | - Fernando E. Figueroa
- Laboratory of Nano-regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago,
Chile
- Program for Translational Research in Cell Therapy, Universidad de Los Andes, Santiago,
Chile
- Consorcio Regenero, the Chilean Consortium for Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago,
Chile
| | - Maroun Khoury
- Laboratory of Nano-regenerative Medicine, Faculty of Medicine, Universidad de Los Andes, Santiago,
Chile
- Cells for Cells, Faculty of Medicine, Universidad de Los Andes, Santiago,
Chile
- Program for Translational Research in Cell Therapy, Universidad de Los Andes, Santiago,
Chile
| |
Collapse
|
48
|
Cai X, Wang L, Wang X, Hou F. miR-124a enhances therapeutic effects of bone marrow stromal cells transplant on diabetic nephropathy-related epithelial-to-mesenchymal transition and fibrosis. J Cell Biochem 2019; 121:299-312. [PMID: 31190436 DOI: 10.1002/jcb.29170] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 05/20/2019] [Accepted: 05/23/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Epithelial-to-mesenchymal transition (EMT) has been gradually considered as one of the major pathways that causes the production of interstitial myofibroblasts in diseased kidneys. MATERIALS AND METHODS The study was done to investigate the effect of a bone marrow stromal cell (BMSCs) transplant on rat podocytes and diabetic nephropathy (DN) rats in high-glucose concentration, and to explore the effect of miR-124a on BMSC therapy. High glucose-injured podocytes and streptozotocin-induced DN rats have been respectively used as injury models in in vitro and in vivo studies. Podocyte viability was measured using the Cell Counting Kit-8 assay. Renal pathological examination was observed by HE staining and Masson staining. The messenger RNA and protein levels were determined via real-time polymerase chain reaction and Western blotting, respectively. RESULTS By mediating the activation of caveolin-1 (cav-1) and β-catenin and affecting the expression levels of EMT biomarkers including p-cadherin, synaptopodin, fibroblast-specific protein-1, α-smooth muscle actin and snail, our in vitro study confirmed that miR-124a played a significant role in the treatment of high glucose-induced podocyte injury by BMSCs. The therapeutic effects of the BMSC transplant on DN rats were also proved to be further enhanced by miR-124a overexpression in BMSCs, and such a phenomenon was accompanied by the improvement of renal fibrosis and mitigation of DN-related kidney impairment. Regulation of fibronectin, collagen1, and EMT-related proteins was closely implicated with the mechanism, and the activation of cav-1 and β-catenin was also possibly involved. CONCLUSION The study demonstrated the pivotal effect of miR-124a on BMSC therapy for DN rats via mitigating EMT and fibrosis. Our results provide a novel insight into how therapeutic effects of BMSCs can be improved at the posttranscriptional level.
Collapse
Affiliation(s)
- Xiaojun Cai
- Department of Endocrinology, Heilongjiang Provincial Academy of Chinese Medical Science, Harbin, Heilongjiang, China
| | - Lei Wang
- Department of Endocrinology, Heilongjiang Provincial Academy of Chinese Medical Science, Harbin, Heilongjiang, China
| | - Xuling Wang
- Department of Endocrinology, Heilongjiang Provincial Academy of Chinese Medical Science, Harbin, Heilongjiang, China
| | - Fengyan Hou
- Department of Endocrinology, Heilongjiang Provincial Academy of Chinese Medical Science, Harbin, Heilongjiang, China
| |
Collapse
|
49
|
Li X, Chen S, Yan L, Wang J, Pei M. Prospective application of stem cells to prevent post-operative skeletal fibrosis. J Orthop Res 2019; 37:1236-1245. [PMID: 30835890 PMCID: PMC9202416 DOI: 10.1002/jor.24266] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 02/17/2019] [Indexed: 02/04/2023]
Abstract
Post-operative skeletal fibrosis is considered one of the major complications causing dysfunction of the skeletal system and compromising the outcomes of clinical treatment. Limited success has been achieved using current therapies; more effective therapies to reduce post-operative skeletal fibrosis are needed. Stem cells possess the ability to repair and regenerate damaged tissue. Numerous studies show that stem cells serve as a promising therapeutic approach for fibrotic diseases in tissues other than the skeletal system by inhibiting the inflammatory response and secreting favorable cytokines through activating specific signaling pathways, acting as so-called medicinal signaling cells. In this review, current therapies are summarized for post-operative skeletal fibrosis. Given that stem cells are used as a promising therapeutic approach for fibrotic diseases, little effort has been undertaken to use stem cells to prevent post-operative skeletal fibrosis. This review aims at providing useful information for the potential application of stem cells in preventing post-operative skeletal fibrosis in the near future. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1236-1245, 2019.
Collapse
Affiliation(s)
- Xiaolei Li
- Department of Orthopaedics, Orthopaedics Institute, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu, 225001, China,Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, 26506, USA
| | - Song Chen
- Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan, 610083, China
| | - Lianqi Yan
- Department of Orthopaedics, Orthopaedics Institute, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu, 225001, China
| | - Jingcheng Wang
- Department of Orthopaedics, Orthopaedics Institute, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu, 225001, China,Co-Corresponding author: Jingcheng Wang, MD, Department of Orthopaedics, Subei People’s Hospital, 98 West Nantong Road, Yangzhou 225001, China;
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, 26506, USA,Exercise Physiology, West Virginia University, Morgantown, WV, 26506, USA,WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, 26506, USA,Corresponding author: Ming Pei MD, PhD, Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, PO Box 9196, 64 Medical Center Drive, Morgantown, WV 26506-9196, USA, Telephone: 304-293-1072; Fax: 304-293-7070;
| |
Collapse
|
50
|
Zhao L, Hu C, Zhang P, Jiang H, Chen J. Mesenchymal stem cell therapy targeting mitochondrial dysfunction in acute kidney injury. J Transl Med 2019; 17:142. [PMID: 31046805 PMCID: PMC6498508 DOI: 10.1186/s12967-019-1893-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 04/25/2019] [Indexed: 12/13/2022] Open
Abstract
Mitochondria take part in a network of cellular processes that regulate cell homeostasis. Defects in mitochondrial function are key pathophysiological changes during acute kidney injury (AKI). Mesenchymal stem cells (MSCs) have shown promising regenerative effects in experimental AKI models, but the specific mechanism is still unclear. Some studies have demonstrated that MSCs are able to target mitochondrial dysfunction during AKI. In this review, we summarize these articles, providing an integral and updated view of MSC therapy targeting mitochondrial dysfunction during AKI, which is aimed at promoting the therapeutic effect of MSCs in AKI patients.
Collapse
Affiliation(s)
- Lingfei Zhao
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang, People's Republic of China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Chenxia Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Ping Zhang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang, People's Republic of China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Hua Jiang
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang, People's Republic of China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China. .,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang, People's Republic of China. .,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|