151
|
Song Y, Peng C, Lv S, Cheng J, Liu S, Wen Q, Guan G, Liu G. Adipose-derived stem cells ameliorate renal interstitial fibrosis through inhibition of EMT and inflammatory response via TGF-β1 signaling pathway. Int Immunopharmacol 2017; 44:115-122. [PMID: 28092863 DOI: 10.1016/j.intimp.2017.01.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 12/15/2016] [Accepted: 01/06/2017] [Indexed: 12/31/2022]
Abstract
Adipose-derived stem cells (ADSCs) have been successfully used to treat acute kidney injury or acute renal failure. However, the effect of ADSCs on treating renal interstitial fibrosis remains unknown. Here, we assessed the therapeutic efficacy of ADSCs on renal interstitial fibrosis induced by unilateral ureter obstruction (UUO) and explored the potential mechanisms. After 7days of UUO, rats were injected with ADSCs (5×106) or vehicle via tail vein. We found that ADSCs administration significantly ameliorated renal interstitial fibrosis, the occurrence of epithelial-mesenchymal transition (EMT) and inflammatory response. Furthermore, ADSCs administration could inhibit the activation of transforming growth factor-β1 (TGF-β1) signaling pathway, which might play a crucial role in renal interstitial fibrosis of the UUO model rats. These results suggested that ADSCs treatment attenuates renal interstitial fibrosis possibly through inhibition of EMT and inflammatory response via TGF-β1 signaling pathway. Therefore, ADSCs may be an effective therapeutic strategy for the treatment of renal interstitial fibrosis.
Collapse
Affiliation(s)
- Yan Song
- Department of Nephrology, The Second Hospital of Shandong University, Shandong University, Jinan, China
| | - Changliang Peng
- Department of Spinal Surgery, The Second Hospital of Shandong University, Shandong University, Jinan, China
| | - Shasha Lv
- Department of Nephrology, The Second Hospital of Shandong University, Shandong University, Jinan, China
| | - Jing Cheng
- Department of Nephrology, The Second Hospital of Shandong University, Shandong University, Jinan, China
| | - Shanshan Liu
- Department of Nephrology, The Second Hospital of Shandong University, Shandong University, Jinan, China
| | - Qing Wen
- Department of Nephrology, The Second Hospital of Shandong University, Shandong University, Jinan, China
| | - Guangju Guan
- Department of Nephrology, The Second Hospital of Shandong University, Shandong University, Jinan, China.
| | - Gang Liu
- Department of Nephrology, The Second Hospital of Shandong University, Shandong University, Jinan, China.
| |
Collapse
|
152
|
Herzmann N, Salamon A, Fiedler T, Peters K. Lipopolysaccharide induces proliferation and osteogenic differentiation of adipose-derived mesenchymal stromal cells in vitro via TLR4 activation. Exp Cell Res 2017; 350:115-122. [DOI: 10.1016/j.yexcr.2016.11.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 11/04/2016] [Accepted: 11/15/2016] [Indexed: 12/22/2022]
|
153
|
Álvaro-Gracia JM, Jover JA, García-Vicuña R, Carreño L, Alonso A, Marsal S, Blanco F, Martínez-Taboada VM, Taylor P, Martín-Martín C, DelaRosa O, Tagarro I, Díaz-González F. Intravenous administration of expanded allogeneic adipose-derived mesenchymal stem cells in refractory rheumatoid arthritis (Cx611): results of a multicentre, dose escalation, randomised, single-blind, placebo-controlled phase Ib/IIa clinical trial. Ann Rheum Dis 2017; 76:196-202. [PMID: 27269294 DOI: 10.1136/annrheumdis-2015-208918] [Citation(s) in RCA: 183] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 05/17/2016] [Indexed: 12/13/2022]
Abstract
OBJECTIVES To evaluate the safety and tolerability of the intravenous administration of Cx611, a preparation of allogeneic expanded adipose-derived stem cells (eASCs), in patients with refractory rheumatoid arthritis (RA), as well as to obtain preliminary clinical efficacy data in this population. METHODS It is a multicentre, dose escalation, randomised, single-blind (double-blind for efficacy), placebo-controlled, phase Ib/IIa clinical trial. Patients with active refractory RA (failure to at least two biologicals) were randomised to receive three intravenous infusions of Cx611: 1 million/kg (cohort A), 2 million/kg (cohort B), 4 million/kg (cohort C) or placebo, on days 1, 8 and 15, and they were followed for therapy assessment for 24 weeks. RESULTS Fifty-three patients were treated (20 in cohort A, 20 in cohort B, 6 in cohort C and 7 in placebo group). A total of 141 adverse events (AEs) were reported. Seventeen patients from the group A (85%), 15 from the group B (75%), 6 from the group C (100%) and 4 from the placebo group (57%) experienced at least one AE.Eight AEs from 6 patients were grade 3 in intensity (severe), 5 in cohort A (lacunar infarction, diarrhoea, tendon rupture, rheumatoid nodule and arthritis), 2 in cohort B (sciatica and RA) and 1 in the placebo group (asthenia). Only one of the grade 3 AEs was serious (the lacunar infarction). American College of Rheumatology 20 responses for cohorts A, B, C and placebo were 45%, 20%, 33% and 29%, respectively, at month 1, and 25%, 15%, 17% and 0%, respectively, at month 3. CONCLUSIONS The intravenous infusion of Cx611 was in general well tolerated, without evidence of dose-related toxicity at the dose range and time period studied. In addition, a trend for clinical efficacy was observed. These data, in our opinion, justify further investigation of this innovative therapy in patients with RA. TRIAL REGISTRATION NUMBERS EudraCT: 2010-021602-37; NCT01663116; Results.
Collapse
Affiliation(s)
| | - Juan A Jover
- Hospital Universitario Clínico San Carlos de Madrid, Madrid, Spain
| | | | - Luis Carreño
- Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | | | | | - Francisco Blanco
- INIBIC-Complejo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - Victor M Martínez-Taboada
- Hospital Universitario Marqués de Valdecilla, Santander, Spain
- Facultad de Medicina, Universidad de Cantabria, Santander, Spain
| | - Peter Taylor
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | | | | | | | - Federico Díaz-González
- Department of Medicine, Universidad de La Laguna, La Laguna, Spain
- Complejo Hospitalario Universitario de Canarias, Tenerife, Spain
| |
Collapse
|
154
|
Yu L, Xu Y, Wang F, Yang C, Liu G, Song X. Functional Roles of Pattern Recognition Receptors That Recognize Virus Nucleic Acids in Human Adipose-Derived Mesenchymal Stem Cells. BIOMED RESEARCH INTERNATIONAL 2016; 2016:9872138. [PMID: 28105439 PMCID: PMC5220457 DOI: 10.1155/2016/9872138] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 11/01/2016] [Indexed: 12/24/2022]
Abstract
Human adipose-derived mesenchymal stem cells (hAD-MSCs) are mesenchymal stem cells with the capability to modulate immune responses. Evidence showing that hAD-MSCs could mediate innate immune responses through pattern recognition receptors (PRRs) is increasing. However, the roles of PRRs in regulating the innate sensing of virus nucleic acids (RNA and DNA) in hAD-MSCs have not yet been investigated. This study focused on the abundant expression of PRRs, including Toll-like receptor 3 (TLR3) and retinoic acid-inducible gene I (RIG-I), which recognize viral RNA, and gamma-interferon inducible protein 16 (IFI16), which recognizes viral DNA in hAD-MSCs. Poly(I:C), a synthetic dsRNA analogy, activated TLR3 and RIG-I and induced the expression of type I interferons (IFN-α/β) and antivirus proteins, including IFN-stimulating gene 15, 2'5'-oligoadenylate synthetase, and Mx GTPase 1 in hAD-MSCs, which were attenuated by the knockdown of each TLR3 or RIG-I. Synthetic herpes simplex viral DNA (HSV60) activated IFI16 and induced the expression of IFN-α/β and antivirus proteins in hAD-MSCs, which were inhibited by the knockdown of IFI16. Both poly(I:C) and HSV60 induced the expression of IFN-α/β through the phosphorylation of IFN-regulatory factor 3. All these results indicated that PRRs recognizing virus nucleic acids were expressed and can mediate antivirus responses in hAD-MSCs.
Collapse
Affiliation(s)
- Lili Yu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang, Henan 453003, China
| | - Yongtao Xu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China
- School of Biomedical Engineering, Xinxiang Medical University, Xinxiang, Henan 453003, China
| | - Fangchao Wang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang, Henan 453003, China
| | - Can Yang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang, Henan 453003, China
| | - Guoyan Liu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang, Henan 453003, China
| | - Xiangfeng Song
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang, Henan 453003, China
| |
Collapse
|
155
|
Immunomodulatory effects of adipose tissue-derived stem cells on elastin scaffold remodeling in diabetes. Tissue Eng Regen Med 2016; 13:701-712. [PMID: 30603451 DOI: 10.1007/s13770-016-0018-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/02/2016] [Accepted: 03/04/2016] [Indexed: 01/11/2023] Open
Abstract
Diabetes is a major risk factor for the progression of vascular disease, contributing to elevated levels of glycoxidation, chronic inflammation and calcification. Tissue engineering emerges as a potential solution for the treatment of vascular diseases however there is a considerable gap in the understanding of how scaffolds and stem cells will perform in patients with diabetes. We hypothesized that adipose tissue-derived stem cells (ASCs) by virtue of their immunosuppressive potential would moderate the diabetes-intensified inflammatory reactions and induce positive construct remodeling. To test this hypothesis, we prepared arterial elastin scaffolds seeded with autologous ASCs and implanted them subdermally in diabetic rats and compared inflammatory markers, macrophage polarization, matrix remodeling, calcification and bone protein expression to control scaffolds implanted with and without cells in nondiabetic rats. ASC-seeded scaffolds exhibited lower levels of CD8+ T-cells and CD68+ pan-macrophages and higher numbers of M2 macrophages, smooth muscle cell-like and fibroblast-like cells. Calcification and osteogenic markers were reduced in ASCseeded scaffolds implanted in non-diabetic rats but remained unchanged in diabetes, unless the scaffolds were first pre-treated with penta-galloyl glucose (PGG), a known anti-oxidative elastin-binding polyphenol. In conclusion, autologous ASC seeding in elastin scaffolds is effective in combating diabetes-related complications. To prevent calcification, the oxidative milieu needs to be reduced by elastin-binding antioxidants such as PGG.
Collapse
|
156
|
Shi R, Jin Y, Cao C, Han S, Shao X, Meng L, Cheng J, Zhang M, Zheng J, Xu J, Li M. Localization of human adipose-derived stem cells and their effect in repair of diabetic foot ulcers in rats. Stem Cell Res Ther 2016; 7:155. [PMID: 27770835 PMCID: PMC5075186 DOI: 10.1186/s13287-016-0412-2] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/12/2016] [Accepted: 09/15/2016] [Indexed: 12/15/2022] Open
Abstract
Background Diabetic foot ulcer (DFU) is an intractable diabetic complication. Patients suffering from diabetes mellitus (DM) frequently present with infected DFUs. In this study, a wound healing model on diabetic rat foot was established to mimic the pathophysiology of clinical patients who suffer from DFUs. Our study aimed to explore the localization of human adipose-derived stem cells (hADSCs) and the role of these cells in the repair of foot ulcerated tissue in diabetic rats, and thus to estimate the possibilities of adipose-derived stem cells for diabetic wound therapy. Method Sprague–Dawley rats were used to establish diabetic models by streptozotocin injection. A full-thickness foot dorsal skin wound was created by a 5 mm skin biopsy punch and a Westcott scissor. These rats were randomly divided into two groups: the hADSC-treated group and the phosphate-buffered saline (PBS) control group. The hADSC or PBS treatment was delivered through the left femoral vein of rats. We evaluated the localization of hADSCs with fluorescence immunohistochemistry and the ulcer area and ulcerative histology were detected dynamically. Result The hADSCs had a positive effect on the full-thickness foot dorsal skin wound in diabetic rats with a significantly reduced ulcer area at day 15. More granulation tissue formation, angiogenesis, cellular proliferation, and higher levels of growth factors expression were also detected in wound beds. Conclusions Our data suggest that hADSC transplantation has the potential to promote foot wound healing in diabetic rats, and transplantation of exogenous stem cells may be suitable for clinical application in the treatment of DFU. Electronic supplementary material The online version of this article (doi:10.1186/s13287-016-0412-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rongfeng Shi
- Department of Interventional & Vascular Surgery, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, 301 Yanchang Road, Shanghai, 200072, People's Republic of China
| | - Yinpeng Jin
- Shanghai Liver Diseases Research Center, The Nanjing Military Command, Shanghai, 200235, People's Republic of China
| | - Chuanwu Cao
- Department of Interventional & Vascular Surgery, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, 301 Yanchang Road, Shanghai, 200072, People's Republic of China.,Institute of Medical Intervention Engineering, Tongji University, 301 Yanchang Road, Shanghai, 200072, People's Republic of China
| | - Shilong Han
- Department of Interventional & Vascular Surgery, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, 301 Yanchang Road, Shanghai, 200072, People's Republic of China.,Institute of Medical Intervention Engineering, Tongji University, 301 Yanchang Road, Shanghai, 200072, People's Republic of China
| | - Xiaowen Shao
- Department of Obstetrics & Gynecology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, 200072, People's Republic of China
| | - Lingyu Meng
- Shanghai Liver Diseases Research Center, The Nanjing Military Command, Shanghai, 200235, People's Republic of China
| | - Jie Cheng
- Department of Interventional & Vascular Surgery, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, 301 Yanchang Road, Shanghai, 200072, People's Republic of China
| | - Meiling Zhang
- Department of Interventional & Vascular Surgery, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, 301 Yanchang Road, Shanghai, 200072, People's Republic of China
| | - Jiayi Zheng
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, 200072, People's Republic of China
| | - Jun Xu
- Advanced Institute of Translational Medicine, Tongji University, 1239 Siping Road, Shanghai, 200092, People's Republic of China. .,East Hospital, Tongji University, School of Medicine, Shanghai, 200092, People's Republic of China.
| | - Maoquan Li
- Department of Interventional & Vascular Surgery, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, 301 Yanchang Road, Shanghai, 200072, People's Republic of China. .,Institute of Medical Intervention Engineering, Tongji University, 301 Yanchang Road, Shanghai, 200072, People's Republic of China.
| |
Collapse
|
157
|
Potočar U, Hudoklin S, Kreft ME, Završnik J, Božikov K, Fröhlich M. Adipose-Derived Stem Cells Respond to Increased Osmolarities. PLoS One 2016; 11:e0163870. [PMID: 27706209 PMCID: PMC5051864 DOI: 10.1371/journal.pone.0163870] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 09/15/2016] [Indexed: 12/16/2022] Open
Abstract
Cell therapies present a feasible option for the treatment of degenerated cartilaginous and intervertebral disc (IVD) tissues. Microenvironments of these tissues are specific and often differ from the microenvironment of cells that, could be potentially used for therapy, e.g. human adipose-derived stem cells (hASC). To ensure safe and efficient implantation of hASC, it is important to evaluate how microenvironmental conditions at the site of implantation affect the implanted cells. This study has demonstrated that cartilaginous tissue-specific osmolarities ranging from 400-600 mOsm/L affected hASC in a dose- and time-dependent fashion in comparison to 300 mOsm/L. Increased osmolarities resulted in transient (nuclear DNA and actin reorganisation) and non-transient, long-term morphological changes (vesicle formation, increase in cell area, and culture morphology), as well as reduced proliferation in monolayer cultures. Increased osmolarities diminished acid proteoglycan production and compactness of chondrogenically induced pellet cultures, indicating decreased chondrogenic potential. Viability of hASC was strongly dependent on the type of culture, with hASC in monolayer culture being more tolerant to increased osmolarity compared to hASC in suspension, alginate-agarose hydrogel, and pellet cultures, thus emphasizing the importance of choosing relevant in vitro conditions according to the specifics of clinical application.
Collapse
Affiliation(s)
| | - Samo Hudoklin
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Mateja Erdani Kreft
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Janja Završnik
- Department of biochemistry and molecular biology, Jozef Stefan Institute, Ljubljana, Slovenia
| | - Krešimir Božikov
- Department of Plastic Surgery and Burns, Division of Surgery, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Mirjam Fröhlich
- Educell Ltd., Trzin, Slovenia
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of biochemistry and molecular biology, Jozef Stefan Institute, Ljubljana, Slovenia
- * E-mail:
| |
Collapse
|
158
|
Rathinasabapathy A, Bruce E, Espejo A, Horowitz A, Sudhan DR, Nair A, Guzzo D, Francis J, Raizada MK, Shenoy V, Katovich MJ. Therapeutic potential of adipose stem cell-derived conditioned medium against pulmonary hypertension and lung fibrosis. Br J Pharmacol 2016; 173:2859-79. [PMID: 27448286 PMCID: PMC5275771 DOI: 10.1111/bph.13562] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 07/07/2016] [Accepted: 07/10/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND PURPOSE Pulmonary hypertension (PH) and pulmonary fibrosis (PF) are life threatening cardiopulmonary diseases. Existing pharmacological interventions have failed to improve clinical outcomes or reduce disease-associated mortality. Emerging evidence suggests that stem cells offer an effective treatment approach against various pathological conditions. It has been proposed that their beneficial actions may be mediated via secretion of paracrine factors. Herein, we evaluated the therapeutic potential of conditioned media (CM) from adipose stem cells (ASCs) against experimental models of PH and PF. EXPERIMENTAL APPROACH Monocrotaline (MCT) or bleomycin (Bleo) was injected into male Sprague-Dawley rats to induce PH or PF respectively. A subset of MCT and Bleo animals were treated with ASCs or CM. Echocardiographic and haemodynamic measurements were performed at the end of the study. Lung and heart tissues were harvested for RNA, protein and histological measurements. KEY RESULTS CM treatment attenuated MCT-induced PH by improving pulmonary blood flow and inhibiting cardiac remodelling. Further, histological studies revealed that right ventricular fibrosis, pulmonary vessel wall thickness and pericyte distribution were significantly decreased by CM administration. Likewise, CM therapy arrested the progression of PF in the Bleo model by reducing collagen deposition. Elevated expression of markers associated with tissue remodelling and inflammation were significantly reduced in both PF and PH lungs. Similar results were obtained with ASCs administration. CONCLUSIONS AND IMPLICATIONS Our study indicates that CM treatment is as effective as ASCs in treating PH and PF. These beneficial effects of CM may provide an innovative approach to treat cardiopulmonary disorders.
Collapse
Affiliation(s)
- Anandharajan Rathinasabapathy
- Pharmacodynamics, University of Florida, Gainesville, FL, USA
- Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Erin Bruce
- Pharmacodynamics, University of Florida, Gainesville, FL, USA
| | - Andrew Espejo
- Pharmacodynamics, University of Florida, Gainesville, FL, USA
| | - Alana Horowitz
- Pharmacodynamics, University of Florida, Gainesville, FL, USA
| | - Dhivya R Sudhan
- Radiation Oncology, University of Florida, Gainesville, FL, USA
| | - Anand Nair
- Comparative Biomedical Sciences, Louisiana State University, Baton Rouge, LA, USA
- Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Dominic Guzzo
- Pharmacodynamics, University of Florida, Gainesville, FL, USA
| | - Joseph Francis
- Comparative Biomedical Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - Mohan K Raizada
- Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA
| | - Vinayak Shenoy
- Pharmacodynamics, University of Florida, Gainesville, FL, USA.
- Pharmaceutical and Biomedical Sciences, California Health Sciences University, Clovis, CA, USA.
| | | |
Collapse
|
159
|
Zeng J, Wang F, Mao M. Co‑culture of fibroblast‑like synoviocytes with umbilical cord‑mesenchymal stem cells inhibits expression of pro‑inflammatory proteins, induces apoptosis and promotes chondrogenesis. Mol Med Rep 2016; 14:3887-93. [PMID: 27599675 DOI: 10.3892/mmr.2016.5721] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 08/09/2016] [Indexed: 11/05/2022] Open
Abstract
The present study aimed to investigate the effect of co‑culture of fibroblast‑like synoviocytes (FLS) with human umbilical cord‑mesenchymal stem cells (UC‑MSCs) on rheumatoid arthritis (RA) and to understand the mechanisms that mediate the induced changes. FLS and UC‑MSCs were isolated and cultured individually, FLS were then cultured with or without UC‑MSCs. The phenotype of UC‑MSCs was analyzed prior to co‑culture. The UC‑MSCs were successfully isolated and expanded, and exhibited a fibroblast‑like morphology. Enzyme‑linked immunosorbent assay (ELISA) and reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) were performed to determine the expression levels of interleukin (IL)‑1β, IL‑6, and chemokine (C‑C motif) ligand (CCL)‑2. The cell apoptosis rate was determined by flow cytometry. Furthermore, the RNAs of aggrecan and collagen type II were isolated and assessed in a chondrogenesis assay following co‑culture for 7, 14, 21 and 28 days. Protein expression levels of apoptosis‑related proteins, including B‑cell lymphoma (Bcl‑2), Bcl‑2‑associated X protein, p53 and phospho (p)‑AKT, and growth differentiation factor‑5 were analyzed by western blotting. ELISA and qRT‑PCR demonstrated that compared with FLS cultured alone, co‑culture with UC‑MSCs significantly downregulates the expression levels of IL‑1β, IL‑6 and CCL‑2. Additionally, the percentage of apoptotic cells was significantly increased in the co‑cultured cells (P<0.05), and the relative RNAs levels of aggrecan and collagen type II were increased compared with FLS alone. Furthermore, the expression levels of Bcl‑2 (P<0.05) and p‑AKT (P<0.05) were significantly decreased, whereas, p53 (P=0.001), Bax (P<0.01) and GDF‑5 (P<0.01) were increased by co‑culture of FLS with UC‑MSCs compared with FLS alone. In conclusion, co‑culture of FLS with UC‑MSCs may be important and clinically useful for the treatment of RA by inhibiting the expression of pro‑inflammatory mediators, inducing apoptosis and promoting chondrogenesis.
Collapse
Affiliation(s)
- Jingqi Zeng
- Department of Orthopedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410005, P.R. China
| | - Fan Wang
- Department of Orthopedics, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410005, P.R. China
| | - Minzhi Mao
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
160
|
Muir P, Hans EC, Racette M, Volstad N, Sample SJ, Heaton C, Holzman G, Schaefer SL, Bloom DD, Bleedorn JA, Hao Z, Amene E, Suresh M, Hematti P. Autologous Bone Marrow-Derived Mesenchymal Stem Cells Modulate Molecular Markers of Inflammation in Dogs with Cruciate Ligament Rupture. PLoS One 2016; 11:e0159095. [PMID: 27575050 PMCID: PMC5005014 DOI: 10.1371/journal.pone.0159095] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 06/27/2016] [Indexed: 01/22/2023] Open
Abstract
Mid-substance rupture of the canine cranial cruciate ligament rupture (CR) and associated stifle osteoarthritis (OA) is an important veterinary health problem. CR causes stifle joint instability and contralateral CR often develops. The dog is an important model for human anterior cruciate ligament (ACL) rupture, where rupture of graft repair or the contralateral ACL is also common. This suggests that both genetic and environmental factors may increase ligament rupture risk. We investigated use of bone marrow-derived mesenchymal stem cells (BM-MSCs) to reduce systemic and stifle joint inflammatory responses in dogs with CR. Twelve dogs with unilateral CR and contralateral stable partial CR were enrolled prospectively. BM-MSCs were collected during surgical treatment of the unstable CR stifle and culture-expanded. BM-MSCs were subsequently injected at a dose of 2x106 BM-MSCs/kg intravenously and 5x106 BM-MSCs by intra-articular injection of the partial CR stifle. Blood (entry, 4 and 8 weeks) and stifle synovial fluid (entry and 8 weeks) were obtained after BM-MSC injection. No adverse events after BM-MSC treatment were detected. Circulating CD8+ T lymphocytes were lower after BM-MSC injection. Serum C-reactive protein (CRP) was decreased at 4 weeks and serum CXCL8 was increased at 8 weeks. Synovial CRP in the complete CR stifle was decreased at 8 weeks. Synovial IFNγ was also lower in both stifles after BM-MSC injection. Synovial/serum CRP ratio at diagnosis in the partial CR stifle was significantly correlated with development of a second CR. Systemic and intra-articular injection of autologous BM-MSCs in dogs with partial CR suppresses systemic and stifle joint inflammation, including CRP concentrations. Intra-articular injection of autologous BM-MSCs had profound effects on the correlation and conditional dependencies of cytokines using causal networks. Such treatment effects could ameliorate risk of a second CR by modifying the stifle joint inflammatory response associated with cranial cruciate ligament matrix degeneration or damage.
Collapse
Affiliation(s)
- Peter Muir
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
- * E-mail:
| | - Eric C. Hans
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
| | - Molly Racette
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
| | - Nicola Volstad
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
| | - Susannah J. Sample
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
| | - Caitlin Heaton
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
| | - Gerianne Holzman
- UW Veterinary Care Hospital, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Susan L. Schaefer
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
| | - Debra D. Bloom
- Department of Medicine, School of Medicine & Public Health, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States of America
| | - Jason A. Bleedorn
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
| | - Zhengling Hao
- Comparative Orthopaedic Research Laboratory, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
| | - Ermias Amene
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
| | - M. Suresh
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
| | - Peiman Hematti
- Department of Medicine, School of Medicine & Public Health, University of Wisconsin-Madison, Madison, Wisconsin, 53705, United States of America
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, 53705, United States of America
| |
Collapse
|
161
|
Regmi S, Jeong JH. Superiority of three-dimensional stem cell clusters over monolayer culture: An archetype to biological application. Macromol Res 2016. [DOI: 10.1007/s13233-016-4107-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
162
|
Mesenchymal Stem Cells Enhance Nerve Regeneration in a Rat Sciatic Nerve Repair and Hindlimb Transplant Model. Sci Rep 2016; 6:31306. [PMID: 27510321 PMCID: PMC4980673 DOI: 10.1038/srep31306] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 07/18/2016] [Indexed: 01/16/2023] Open
Abstract
This study investigates the efficacy of local and intravenous mesenchymal stem cell (MSC) administration to augment neuroregeneration in both a sciatic nerve cut-and-repair and rat hindlimb transplant model. Bone marrow-derived MSCs were harvested and purified from Brown-Norway (BN) rats. Sciatic nerve transections and repairs were performed in three groups of Lewis (LEW) rats: negative controls (n = 4), local MSCs (epineural) injection (n = 4), and systemic MSCs (intravenous) injection (n = 4). Syngeneic (LEW-LEW) (n = 4) and allogeneic (BN-LEW) (n = 4) hindlimb transplants were performed and assessed for neuroregeneration after local or systemic MSC treatment. Rats undergoing sciatic nerve cut-and-repair and treated with either local or systemic injection of MSCs had significant improvement in the speed of recovery of compound muscle action potential amplitudes and axon counts when compared with negative controls. Similarly, rats undergoing allogeneic hindlimb transplants treated with local injection of MSCs exhibited significantly increased axon counts. Similarly, systemic MSC treatment resulted in improved nerve regeneration following allogeneic hindlimb transplants. Systemic administration had a more pronounced effect on electromotor recovery while local injection was more effective at increasing fiber counts, suggesting different targets of action. Local and systemic MSC injections significantly improve the pace and degree of nerve regeneration after nerve injury and hindlimb transplantation.
Collapse
|
163
|
Juhl AA, Karlsson P, Damsgaard TE. Fat grafting for alleviating persistent pain after breast cancer treatment: A randomized controlled trial. J Plast Reconstr Aesthet Surg 2016; 69:1192-202. [PMID: 27470295 DOI: 10.1016/j.bjps.2016.07.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 06/13/2016] [Accepted: 07/03/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Persistent pain is a common side effect of breast cancer treatment, affecting 24-52% of women after mastectomy. Recent studies have described analgesic effects of fat grafting in various settings. We aimed to investigate whether fat grafting had an analgesic effect on persistent pain after mastectomy and whether fat grafting had a remodeling effect on the mastectomy scar. METHODS This study was conducted as a randomized controlled trial. Patients were randomized to either receive fat grafting to the pain-afflicted area around the missing breast or a control group without any intervention. A total of 18 unilaterally mastectomized women with persistent pain ≥3 on the numerical rating scale were enrolled. Patients were examined at the baseline and at 3 and 6 months by using the DoloTest(®), visual analog scale (VAS) pain score, neuropathic pain symptom inventory, and patient and observer scar assessment scale. RESULTS A total of 15 patients were analyzed (fat grafted n = 8, control n = 7). The average amount of grafted fat was 71 ± 24.6 mL. Fat grafting showed a significant improvement in the pain as measured on the VAS pain scale (p = 0.001) with an average reduction of 54.9% and as measured on the neuropathic pain symptom inventory (p = 0.002). Furthermore, a significant improvement was observed in health-related quality of life (p = 0.007) and the quality of the scar (p < 0.001). CONCLUSION This is the first randomized controlled trial evaluating the analgesic effect of fat grafting. Fat grafting is a safe and effective technique for alleviating persistent pain after mastectomy.
Collapse
Affiliation(s)
- Alexander A Juhl
- Plastic Surgery Research Unit, Department of Plastic and Breast Surgery, Aarhus University Hospital, Aarhus, Denmark.
| | - Páll Karlsson
- Danish Pain Research Center, Aarhus University Hospital, Aarhus, Denmark; Stereology and Electron Microscopy Laboratory, Aarhus University Hospital, 44 Nørrebrogade, 8000, Aarhus C, Denmark
| | - Tine E Damsgaard
- Plastic Surgery Research Unit, Department of Plastic and Breast Surgery, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
164
|
Demirel S, Yalvac ME, Tapsin S, Akyuz S, Ak E, Cetinel S, Yarat A, Sahin F. Tooth replantation with adipose tissue stem cells and fibrin sealant: microscopic analysis of rat's teeth. SPRINGERPLUS 2016; 5:656. [PMID: 28443212 PMCID: PMC5395512 DOI: 10.1186/s40064-016-2263-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 05/03/2016] [Indexed: 12/11/2022]
Abstract
Treatment for dental avulsion cases is early or late replantation of the traumatized teeth. Prognosis of the replanted tooth depends on the level of periodontal injury. Adipose tissue stem cells (ATSCs) were reported to improve periodontal ligament tissue (PDL) regeneration. Fibrin sealant (FS) contains thrombin and fibrinogen to form an adhesive fibrin clot routinely used in surgical procedures. Here, we aimed to investigate the effects of ATSCs + FS treatment on healing of PDL after tooth replantation in a rat model. After 60 min of extraction, maxillary central incisor teeth were replanted with ATSCs + FS. Two months later, the rats were sacrificed and hemimaxilla blocks were dissected out for histological analysis. The results showed that there was a significant improvement in histological findings of ATSCs + FS treated group compared to only FS treated and non-treated groups corresponding to reduced inflammatory resorption and increased new PDL formation. Furthermore, the ankylosis levels were lowered after ATSCs + FS treatment. Singular use of FS improved PDL healing moderately. Our results indicated that ATSCs + FS treatment improves PDL healing after tooth replantation suggesting a new therapeutic potential in the treatment of dental avulsion cases.
Collapse
Affiliation(s)
- Sezin Demirel
- Department of Pediatric Dentistry, Faculty of Dentistry, Marmara University, Istanbul, Turkey
| | - Mehmet Emir Yalvac
- Center for Gene Therapy, Research Institute at Nationwide Children's Hospital, Columbus, OH USA
| | - Sidika Tapsin
- Department of Genetics and Bioengineering, Yeditepe University, Istanbul, Turkey
| | - Serap Akyuz
- Department of Pediatric Dentistry, Faculty of Dentistry, Marmara University, Istanbul, Turkey
| | - Esin Ak
- Department of Histology and Embryology, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Sule Cetinel
- Department of Histology and Embryology, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Aysen Yarat
- Department of Basic Medical Sciences, Biochemistry, Faculty of Dentistry, Marmara University, Istanbul, Turkey
| | - Fikrettin Sahin
- Department of Genetics and Bioengineering, Yeditepe University, Istanbul, Turkey
| |
Collapse
|
165
|
Bajek A, Gurtowska N, Olkowska J, Kazmierski L, Maj M, Drewa T. Adipose-Derived Stem Cells as a Tool in Cell-Based Therapies. Arch Immunol Ther Exp (Warsz) 2016; 64:443-454. [PMID: 27178663 PMCID: PMC5085986 DOI: 10.1007/s00005-016-0394-x] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 01/20/2016] [Indexed: 02/06/2023]
Abstract
Recent development in stem cell isolation methods and expansion under laboratory conditions create an opportunity to use those aforementioned cells in tissue engineering and regenerative medicine. Particular attention is drawn towards mesenchymal stem cells (MSCs) being multipotent progenitors exhibiting several unique characteristics, including high proliferation potential, self-renewal abilities and multilineage differentiation into cells of mesodermal and non-mesodermal origin. High abundance of MSCs found in adipose tissue makes it a very attractive source of adult stem cells for further use in regenerative medicine applications. Despite immunomodulating properties of adipose-derived stem cells (ASCs) and a secretion of a wide variety of paracrine factors that facilitate tissue regeneration, effectiveness of stem cell therapy was not supported by the results of clinical trials. Lack of a single, universal stem cell marker, patient-to-patient variability, heterogeneity of ASC population combined with multiple widely different protocols of cell isolation and expansion hinder the ability to precisely identify and analyze biological properties of stem cells. The above issues contribute to conflicting data reported in literature. We will review the comprehensive information concerning characteristic features of ASCs. We will also review the regenerative potential and clinical application based on various clinical trials.
Collapse
Affiliation(s)
- Anna Bajek
- Department of Tissue Engineering, Nicolaus Copernicus University, Karłowicza 24, 85-092, Bydgoszcz, Poland.
| | - Natalia Gurtowska
- Department of Tissue Engineering, Nicolaus Copernicus University, Karłowicza 24, 85-092, Bydgoszcz, Poland
| | - Joanna Olkowska
- Department of Tissue Engineering, Nicolaus Copernicus University, Karłowicza 24, 85-092, Bydgoszcz, Poland
| | - Lukasz Kazmierski
- Department of Tissue Engineering, Nicolaus Copernicus University, Karłowicza 24, 85-092, Bydgoszcz, Poland
| | - Malgorzata Maj
- Department of Tissue Engineering, Nicolaus Copernicus University, Karłowicza 24, 85-092, Bydgoszcz, Poland
| | - Tomasz Drewa
- Department of Tissue Engineering, Nicolaus Copernicus University, Karłowicza 24, 85-092, Bydgoszcz, Poland.,Department of Urology, Nicolaus Copernicus Hospital, Torun, Poland
| |
Collapse
|
166
|
Lopez-Santalla M, Menta R, Mancheño-Corvo P, Lopez-Belmonte J, DelaRosa O, Bueren JA, Dalemans W, Lombardo E, Garin MI. Adipose-derived mesenchymal stromal cells modulate experimental autoimmune arthritis by inducing an early regulatory innate cell signature. IMMUNITY INFLAMMATION AND DISEASE 2016; 4:213-224. [PMID: 27957329 PMCID: PMC4879467 DOI: 10.1002/iid3.106] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 03/02/2016] [Accepted: 03/04/2016] [Indexed: 12/22/2022]
Abstract
Modulation of innate immune responses in rheumatoid arthritis and other immune-mediated disorders is of critical importance in the clinic since a growing body of information has shown the key contribution of dysregulated innate responses in the progression of the disease. Mesenchymal stromal cells (MSCs) are the focus of intensive efforts worldwide due to their key role in tissue regeneration and modulation of inflammation. In this study, we define innate immune responses occurring during the early course of treatment with a single dose of expanded adipose-derived MSCs (eASCs) in established collagen-induced arthritis. eASCs delay the progression of the disease during the early phase of the disease. This is accompanied by a transient induction of Ly6C+ monocytes that differentiate into IL10+F4/80+ cells in arthritic mice. Strikingly, the induced IL10+F4/80+ myeloid cells preferentially accumulated in the draining lymph nodes. This effect was accompanied with a concomitant declining of their frequencies in the spleens. Our results show that eASCs attenuate the arthritic process by inducing an early innate cell signature that involves a transient induction of Ly6C+ monocytes in periphery that differentiate into IL10+F4/80+ macrophages. Our findings demonstrate that early regulatory innate cell responses, involving the monocyte compartment, are targeted by the eASCs during the onset of collagen-induced inflammation.
Collapse
Affiliation(s)
- Mercedes Lopez-Santalla
- Hematopoietic Innovative Therapies DivisionCentro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER)MadridSpain; Advanced Therapy UnitCentro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM)MadridSpain
| | | | | | | | | | - Juan A Bueren
- Hematopoietic Innovative Therapies DivisionCentro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER)MadridSpain; Advanced Therapy UnitCentro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM)MadridSpain
| | | | | | - Marina I Garin
- Hematopoietic Innovative Therapies DivisionCentro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER)MadridSpain; Advanced Therapy UnitCentro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM)MadridSpain
| |
Collapse
|
167
|
López M, Bollag RJ, Yu JC, Isales CM, Eroglu A. Chemically Defined and Xeno-Free Cryopreservation of Human Adipose-Derived Stem Cells. PLoS One 2016; 11:e0152161. [PMID: 27010403 PMCID: PMC4806986 DOI: 10.1371/journal.pone.0152161] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 03/09/2016] [Indexed: 12/18/2022] Open
Abstract
The stromal compartment of adipose tissue harbors multipotent cells known as adipose-derived stem cells (ASCs). These cells can differentiate into various lineages including osteogenic, chrondrogenic, adipogenic, and neurogenic; this cellular fraction may be easily obtained in large quantities through a clinically safe liposuction procedure. Therefore, ASCs offer exceptional opportunities for tissue engineering and regenerative medicine. However, current practices involving ASCs typically use fetal bovine serum (FBS)-based cryopreservation solutions that are associated with risks of immunological reactions and of transmitting infectious diseases and prions. To realize clinical applications of ASCs, serum- and xeno-free defined cryopreservation methods are needed. To this end, an animal product-free chemically defined cryopreservation medium was formulated by adding two antioxidants (reduced glutathione and ascorbic acid 2-phosphate), two polymers (PVA and ficoll), two permeating cryoprotectants (ethylene glycol and dimethylsulfoxide), a disaccharide (trehalose), and a calcium chelator (EGTA) to HEPES-buffered DMEM/F12. To limit the number of experimental groups, the concentration of trehalose, both polymers, and EGTA was fixed while the presence of the permeating CPAs and antioxidants was varied. ASCs suspended either in different versions of the defined medium or in the conventional undefined cryopreservation medium (10% dimethylsulfoxide+10% DMEM/F12+80% serum) were cooled to -70°C at 1°C/min before being plunged into liquid nitrogen. Samples were thawed either in air or in a water bath at 37°C. The presence of antioxidants along with 3.5% concentration of each penetrating cryoprotectant improved the freezing outcome to the level of the undefined cryopreservation medium, but the plating efficiency was still lower than that of unfrozen controls. Subsequently, increasing the concentration of both permeating cryoprotectants to 5% further improved the plating efficiency to the level of unfrozen controls. Moreover, ASCs cryopreserved in this defined medium retained their multipotency and chromosomal normality. These results are of significance for tissue engineering and clinical applications of stem cells.
Collapse
Affiliation(s)
- Melany López
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Roni J. Bollag
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Jack C. Yu
- Department of Surgery, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Carlos M. Isales
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
- Orthopaedic Surgery, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Ali Eroglu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
- Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
168
|
Pianta S, Bonassi Signoroni P, Muradore I, Rodrigues MF, Rossi D, Silini A, Parolini O. Amniotic membrane mesenchymal cells-derived factors skew T cell polarization toward Treg and downregulate Th1 and Th17 cells subsets. Stem Cell Rev Rep 2016; 11:394-407. [PMID: 25348066 PMCID: PMC4451472 DOI: 10.1007/s12015-014-9558-4] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
We previously demonstrated that cells derived from the mesenchymal layer of the human amniotic membrane (hAMSC) and their conditioned medium (CM-hAMSC) modulate lymphocyte proliferation in a dose-dependent manner. In order to understand the mechanisms involved in immune regulation exerted by hAMSC, we analyzed the effects of CM-hAMSC on T-cell polarization towards Th1, Th2, Th17, and T-regulatory (Treg) subsets. We show that CM-hAMSC equally suppresses the proliferation of both CD4+ T-helper (Th) and CD8+ cytotoxic T-lymphocytes. Moreover, we prove that the CM-hAMSC inhibitory ability affects both central (CD45RO+CD62L+) and effector memory (CD45RO+CD62L−) subsets. We evaluated the phenotype of CD4+ cells in the MLR setting and showed that CM-hAMSC significantly reduced the expression of markers associated to the Th1 (T-bet+CD119+) and Th17 (RORγt+CD161+) populations, while having no effect on the Th2 population (GATA3+CD193+/GATA3+CD294+cells). T-cell subset modulation was substantiated through the analysis of cytokine release for 6 days during co-culture with alloreactive T-cells, whereby we observed a decrease in specific subset-related cytokines, such as a decrease in pro-inflammatory, Th1-related (TNFα, IFNγ, IL-1β), Th2 (IL-5, IL-6), Th9 (IL-9), and Th17 (IL-17A, IL-22). Furthermore, CM-hAMSC significantly induced the Treg compartment, as shown by an induction of proliferating CD4+FoxP3+ cells, and an increase of CD25+FoxP3+ and CD39+FoxP3+ Treg in the CD4+ population. Induction of Treg cells was corroborated by the increased secretion of TGF-β. Taken together, these data strengthen the findings regarding the immunomodulatory properties of CM-hAMSC derived from human amniotic membrane MSC, and in particular provide insights into their effect on regulation of T cell polarization.
Collapse
Affiliation(s)
- Stefano Pianta
- Centro di Ricerca E. Menni, Fondazione Poliambulanza-Istituto Ospedaliero, Via Bissolati, 57, I-25124, Brescia, Italy
| | | | | | | | | | | | | |
Collapse
|
169
|
Adipose-Derived Stem Cells for Tissue Engineering and Regenerative Medicine Applications. Stem Cells Int 2016; 2016:6737345. [PMID: 27057174 PMCID: PMC4761677 DOI: 10.1155/2016/6737345] [Citation(s) in RCA: 183] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 01/02/2016] [Accepted: 01/03/2016] [Indexed: 02/05/2023] Open
Abstract
Adipose-derived stem cells (ASCs) are a mesenchymal stem cell source with properties of self-renewal and multipotential differentiation. Compared to bone marrow-derived stem cells (BMSCs), ASCs can be derived from more sources and are harvested more easily. Three-dimensional (3D) tissue engineering scaffolds are better able to mimic the in vivo cellular microenvironment, which benefits the localization, attachment, proliferation, and differentiation of ASCs. Therefore, tissue-engineered ASCs are recognized as an attractive substitute for tissue and organ transplantation. In this paper, we review the characteristics of ASCs, as well as the biomaterials and tissue engineering methods used to proliferate and differentiate ASCs in a 3D environment. Clinical applications of tissue-engineered ASCs are also discussed to reveal the potential and feasibility of using tissue-engineered ASCs in regenerative medicine.
Collapse
|
170
|
Strong AL, Bowles AC, Wise RM, Morand JP, Dutreil MF, Gimble JM, Bunnell BA. Human Adipose Stromal/Stem Cells from Obese Donors Show Reduced Efficacy in Halting Disease Progression in the Experimental Autoimmune Encephalomyelitis Model of Multiple Sclerosis. Stem Cells 2016; 34:614-26. [PMID: 26700612 DOI: 10.1002/stem.2272] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 10/11/2015] [Accepted: 10/30/2015] [Indexed: 12/18/2022]
Abstract
Multiple sclerosis is an autoimmune disease that affects the white matter of the central nervous system and involves inflammation and demyelination. The recent advances in our understanding of adipose-derived stromal/stem cells (ASCs) and the utilization of these cells in clinical settings to treat diseases have made it essential to identify the most effective ASCs for therapy. Studies have not yet investigated the impact of obesity on the therapeutic efficacy of ASCs. Obesity is characterized by adipocyte hyperplasia and hypertrophy and can extend to metabolic and endocrine dysfunction. Investigating the impact obesity has on ASC biology will determine whether these cells are suitable for use in regenerative medicine. The therapeutic efficacy of ASCs isolated from lean subjects (body mass index [BMI] < 25; lnASCs) and obese subjects (BMI > 30; obASCs) were determined in murine experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis. Compared with the EAE disease-modifying effects of lnASCs, obASCs consistently failed to alleviate clinical symptoms or inhibit inflammation in the central nervous system. When activated, obASCs expressed higher mRNA levels of several pro-inflammatory cytokines compared with lnASCs. Additionally, conditioned media (CM) collected from the obASCs markedly enhanced the proliferation and differentiation of T cells; whereas, CM from lnASC did not. These results indicate that obesity reduces, or eliminates, the anti-inflammatory effects of human ASCs such that they may not be a suitable cell source for the treatment of autoimmune diseases. The data suggest that donor demographics may be particularly important when identifying suitable stem cells for treatment.
Collapse
Affiliation(s)
- Amy L Strong
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Annie C Bowles
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Rachel M Wise
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Joseph P Morand
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Maria F Dutreil
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Jeffrey M Gimble
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA.,Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA.,Department of Surgery, Tulane University School of Medicine, New Orleans, Louisiana, USA.,Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA.,LaCell LLC, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Bruce A Bunnell
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA.,Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
171
|
Wu H, Yan S, Chen J, Luo X, Li P, Jia X, Dai X, Wang C, Huang Q, Liu L, Zhang Y, Zhou A, Chang Y, Zhang L, Wei W. JAK1-STAT3 blockade by JAK inhibitor SHR0302 attenuates inflammatory responses of adjuvant-induced arthritis rats and decreases Th17 and total B cells. Joint Bone Spine 2016; 83:525-32. [PMID: 26832189 DOI: 10.1016/j.jbspin.2015.09.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 09/21/2015] [Indexed: 02/09/2023]
Abstract
OBJECTIVE To investigate the effects of JAK inhibitor (SHR0302) on adjuvant-induced arthritis (AA) rats and the partial mechanisms focused on T, B lymphocyte subsets through JAK1-STAT3 pathway, including Th17, Treg, total B cells and memory B cells. METHODS Animals were divided randomly into normal control, AA, SHR0302 (0.3,1.0, 3.0mg/kg) and MTX. The effects of SHR0302 on AA rats by evaluating arthritis index, arthritis global assessment and paw swelling degree, histopathology of joint and spleen. We examined the proliferation of T, B and FLS. Th17, Treg, total B and memory B cell proportion was measured by flow cytometry. Cytokines TNF-α, IL-1β, IL-10, IL-17 and antibody IgG1, IgG2a levels in serum were measured by Elisa. The expression of p-JAK1 and p-STAT3 was measured by western blot. RESULTS SHR0302 suppressed the severity of AA rats by attenuating the arthritis index, arthritis global assessment and paw swelling degree, and alleviated histopathology of spleen and joint of AA rats. SHR0302 can inhibit the proliferation of T, B and FLS, and down-regulated cytokines TNF-α, IL-1β, IL-17 and antibody IgG1, IgG2a levels, and suppressed the proportion of Th17 and total B, and inhibited JAK1-STAT3 phosphorylation. There was no significant effect on Treg function and memory B cell proportion. CONCLUSION SHR0302 may attenuate the severity of AA rats, partially through reducing Th17 function and total B cell proportion by inhibiting JAK1-STAT3 phosphorylation.
Collapse
Affiliation(s)
- Huaxun Wu
- Institute of clinical pharmacology of Anhui medical university, 81, meishan road, 230032 Hefei, China; Key laboratory of anti-inflammatory and immune medicine, ministry of Education, 230032 Hefei, China; Anhui collaborative innovation center of anti-inflammatory and immune medicine, 230032 Hefei, China
| | - Shangxue Yan
- Institute of clinical pharmacology of Anhui medical university, 81, meishan road, 230032 Hefei, China; Key laboratory of anti-inflammatory and immune medicine, ministry of Education, 230032 Hefei, China; Anhui collaborative innovation center of anti-inflammatory and immune medicine, 230032 Hefei, China
| | - Jingyu Chen
- Institute of clinical pharmacology of Anhui medical university, 81, meishan road, 230032 Hefei, China; Key laboratory of anti-inflammatory and immune medicine, ministry of Education, 230032 Hefei, China; Anhui collaborative innovation center of anti-inflammatory and immune medicine, 230032 Hefei, China
| | - Xuexia Luo
- Institute of clinical pharmacology of Anhui medical university, 81, meishan road, 230032 Hefei, China; Key laboratory of anti-inflammatory and immune medicine, ministry of Education, 230032 Hefei, China; Anhui collaborative innovation center of anti-inflammatory and immune medicine, 230032 Hefei, China
| | - Peipei Li
- Institute of clinical pharmacology of Anhui medical university, 81, meishan road, 230032 Hefei, China; Key laboratory of anti-inflammatory and immune medicine, ministry of Education, 230032 Hefei, China; Anhui collaborative innovation center of anti-inflammatory and immune medicine, 230032 Hefei, China
| | - Xiaoyi Jia
- Institute of clinical pharmacology of Anhui medical university, 81, meishan road, 230032 Hefei, China; Key laboratory of anti-inflammatory and immune medicine, ministry of Education, 230032 Hefei, China; Anhui collaborative innovation center of anti-inflammatory and immune medicine, 230032 Hefei, China
| | - Xing Dai
- Institute of clinical pharmacology of Anhui medical university, 81, meishan road, 230032 Hefei, China; Key laboratory of anti-inflammatory and immune medicine, ministry of Education, 230032 Hefei, China; Anhui collaborative innovation center of anti-inflammatory and immune medicine, 230032 Hefei, China
| | - Chun Wang
- Institute of clinical pharmacology of Anhui medical university, 81, meishan road, 230032 Hefei, China; Key laboratory of anti-inflammatory and immune medicine, ministry of Education, 230032 Hefei, China; Anhui collaborative innovation center of anti-inflammatory and immune medicine, 230032 Hefei, China
| | - Qiong Huang
- Institute of clinical pharmacology of Anhui medical university, 81, meishan road, 230032 Hefei, China; Key laboratory of anti-inflammatory and immune medicine, ministry of Education, 230032 Hefei, China; Anhui collaborative innovation center of anti-inflammatory and immune medicine, 230032 Hefei, China
| | - Lihua Liu
- Institute of clinical pharmacology of Anhui medical university, 81, meishan road, 230032 Hefei, China; Key laboratory of anti-inflammatory and immune medicine, ministry of Education, 230032 Hefei, China; Anhui collaborative innovation center of anti-inflammatory and immune medicine, 230032 Hefei, China
| | - Yunfang Zhang
- Institute of clinical pharmacology of Anhui medical university, 81, meishan road, 230032 Hefei, China; Key laboratory of anti-inflammatory and immune medicine, ministry of Education, 230032 Hefei, China; Anhui collaborative innovation center of anti-inflammatory and immune medicine, 230032 Hefei, China
| | - Aiwu Zhou
- Institute of clinical pharmacology of Anhui medical university, 81, meishan road, 230032 Hefei, China; Key laboratory of anti-inflammatory and immune medicine, ministry of Education, 230032 Hefei, China; Anhui collaborative innovation center of anti-inflammatory and immune medicine, 230032 Hefei, China
| | - Yan Chang
- Institute of clinical pharmacology of Anhui medical university, 81, meishan road, 230032 Hefei, China; Key laboratory of anti-inflammatory and immune medicine, ministry of Education, 230032 Hefei, China; Anhui collaborative innovation center of anti-inflammatory and immune medicine, 230032 Hefei, China
| | - LingLing Zhang
- Institute of clinical pharmacology of Anhui medical university, 81, meishan road, 230032 Hefei, China; Key laboratory of anti-inflammatory and immune medicine, ministry of Education, 230032 Hefei, China; Anhui collaborative innovation center of anti-inflammatory and immune medicine, 230032 Hefei, China
| | - Wei Wei
- Institute of clinical pharmacology of Anhui medical university, 81, meishan road, 230032 Hefei, China; Key laboratory of anti-inflammatory and immune medicine, ministry of Education, 230032 Hefei, China; Anhui collaborative innovation center of anti-inflammatory and immune medicine, 230032 Hefei, China.
| |
Collapse
|
172
|
Huang SH, Wu SH, Chang KP, Lin CH, Chang CH, Wu YC, Lee SS, Lin SD, Lai CS. Alleviation of neuropathic scar pain using autologous fat grafting. Ann Plast Surg 2016; 74 Suppl 2:S99-104. [PMID: 25695456 DOI: 10.1097/sap.0000000000000462] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Traumatic wounds inflict small- and large-fiber sensory nerve damage, causing neuropathic pain in scar tissue, thus impairing patients' quality of life and leading to the development of psychological disorders. Autologous fat grafting has been clinically shown to improve scar quality, but few studies have explored the effects of this technique on pain. The purpose of this study was to assess the effect of fat grafting on treating neuropathic scar pain. From February 2008 to January 2013, 13 patients who were identified using the Douleur Neuropathique 4 Questions (scores>4/10) were enrolled in this study. The Visual Analog Scale (VAS) and Neuropathic Pain Symptom Inventory (NPSI) were used to evaluate pain preoperatively and 1 week, 4 weeks, and 24 weeks postoperatively. The mechanism of trauma, scar location and size, duration of allodynia, fat graft volume, pharmacologic therapy duration, and total follow-up time were recorded. Thirteen patients experiencing neuropathic pain were enrolled in this study. The mean±SD age was 33.08±16.35 years. The mean duration of pain was 4.29±2.85 months. The mean VAS score before treatment was 7.54±1.05. The mean VAS scores decreased by 4.38±1.66 after 1 week of treatment (P=0.009), 5.38±2.06 after 4 weeks of treatment, and 5.62±2.18 after 24 weeks of treatment. The mean NPSI scores were 49.38±13.25 before treatment, 25±14.4 after 1 week of treatment (P=0.004), 21±17.78 after 4 weeks of treatment, and 14.62±16.88 after 24 weeks of treatment. The 13 patients followed a mean of 24 weeks; 10 (77%) of the patients had improvement of 5 or greater on the VAS score. The mean follow-up period was 19.3±12.26 months (range, 6-38 months). No surgical complications were noted in this series. In our study, both VAS and NPSI scores decreased significantly, revealing that the autologous fat grafting can alleviate neuropathic scar pain 1 week after operation and in the long term.
Collapse
Affiliation(s)
- Shu-Hung Huang
- From the *Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, †Division of Plastic Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan, ‡Department of Surgery, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, §Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan, and ∥Department of Anesthesia, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
173
|
Yaochite JNU, de Lima KWA, Caliari-Oliveira C, Palma PVB, Couri CEB, Simões BP, Covas DT, Voltarelli JC, Oliveira MC, Donadi EA, Malmegrim KCR. Multipotent mesenchymal stromal cells from patients with newly diagnosed type 1 diabetes mellitus exhibit preserved in vitro and in vivo immunomodulatory properties. Stem Cell Res Ther 2016; 7:14. [PMID: 26781648 PMCID: PMC4861132 DOI: 10.1186/s13287-015-0261-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 11/16/2015] [Accepted: 12/07/2015] [Indexed: 12/25/2022] Open
Abstract
Background Type 1 diabetes mellitus (T1D) is characterized by autoimmune responses resulting in destruction of insulin-producing pancreatic beta cells. Multipotent mesenchymal stromal cells (MSCs) exhibit immunomodulatory potential, migratory capacity to injured areas and may contribute to tissue regeneration by the secretion of bioactive factors. Therefore, MSCs are considered as a promising approach to treat patients with different autoimmune diseases (AID), including T1D patients. Phenotypical and functional alterations have been reported in MSCs derived from patients with different AID. However, little is known about the properties of MSCs derived from patients with T1D. Since autoimmunity and the diabetic microenvironment may affect the biology of MSCs, it becomes important to investigate whether these cells are suitable for autologous transplantation. Thus, the aim of the present study was to evaluate the in vitro properties and the in vivo therapeutic efficacy of MSCs isolated from bone marrow of newly diagnosed T1D patients (T1D-MSCs) and to compare them with MSCs from healthy individuals (C-MSCs). Methods T1D-MSCs and C-MSCs were isolated and cultured until third passage. Then, morphology, cell diameter, expression of surface markers, differentiation potential, global microarray analyses and immunosuppressive capacity were in vitro analyzed. T1D-MSCs and C-MSCs therapeutic potential were evaluated using a murine experimental model of streptozotocin (STZ)-induced diabetes. Results T1D-MSCs and C-MSCs presented similar morphology, immunophenotype, differentiation potential, gene expression of immunomodulatory molecules and in vitro immunosuppressive capacity. When administered into diabetic mice, both T1D-MSCs and C-MSCs were able to reverse hyperglycemia, improve beta cell function and modulate pancreatic cytokine levels. Conclusions Thus, bone marrow MSCs isolated from T1D patients recently after diagnosis are not phenotypically or functionally impaired by harmful inflammatory and metabolic diabetic conditions. Our results provide support for the use of autologous MSCs for treatment of newly diagnosed T1D patients. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0261-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Juliana Navarro Ueda Yaochite
- Department of Biochemistry and Immunology, Basic and Applied Immunology Program, School of Medicine of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, Monte Alegre, 14049-900, Ribeirão Preto, São Paulo, Brazil. .,Department of Clinical and Toxicological Analysis, Federal University of Ceará, Alexandre Baraúna 949, Rodolfo Teófilo, 60430-160, Fortaleza, Ceará, Brazil. .,Regional Blood Center of Ribeirão Preto, University of São Paulo, Tenente Catão Roxo, 2501, Monte Alegre, 14051-140, Ribeirão Preto, São Paulo, Brazil.
| | - Kalil Willian Alves de Lima
- Department of Biochemistry and Immunology, Basic and Applied Immunology Program, School of Medicine of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, Monte Alegre, 14049-900, Ribeirão Preto, São Paulo, Brazil.
| | - Carolina Caliari-Oliveira
- Department of Biochemistry and Immunology, Basic and Applied Immunology Program, School of Medicine of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, Monte Alegre, 14049-900, Ribeirão Preto, São Paulo, Brazil. .,Regional Blood Center of Ribeirão Preto, University of São Paulo, Tenente Catão Roxo, 2501, Monte Alegre, 14051-140, Ribeirão Preto, São Paulo, Brazil.
| | - Patricia Vianna Bonini Palma
- Regional Blood Center of Ribeirão Preto, University of São Paulo, Tenente Catão Roxo, 2501, Monte Alegre, 14051-140, Ribeirão Preto, São Paulo, Brazil.
| | - Carlos Eduardo Barra Couri
- Department of Clinical Medicine, School of Medicine of Ribeirão Preto, University of São Paulo, Tenente Catão Roxo, 2501, Monte Alegre, 14051-140, Ribeirão Preto, São Paulo, Brazil.
| | - Belinda Pinto Simões
- Department of Clinical Medicine, School of Medicine of Ribeirão Preto, University of São Paulo, Tenente Catão Roxo, 2501, Monte Alegre, 14051-140, Ribeirão Preto, São Paulo, Brazil.
| | - Dimas Tadeu Covas
- Regional Blood Center of Ribeirão Preto, University of São Paulo, Tenente Catão Roxo, 2501, Monte Alegre, 14051-140, Ribeirão Preto, São Paulo, Brazil. .,Department of Clinical Medicine, School of Medicine of Ribeirão Preto, University of São Paulo, Tenente Catão Roxo, 2501, Monte Alegre, 14051-140, Ribeirão Preto, São Paulo, Brazil.
| | | | - Maria Carolina Oliveira
- Department of Clinical Medicine, School of Medicine of Ribeirão Preto, University of São Paulo, Tenente Catão Roxo, 2501, Monte Alegre, 14051-140, Ribeirão Preto, São Paulo, Brazil.
| | - Eduardo Antônio Donadi
- Department of Biochemistry and Immunology, Basic and Applied Immunology Program, School of Medicine of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, Monte Alegre, 14049-900, Ribeirão Preto, São Paulo, Brazil. .,Department of Clinical Medicine, School of Medicine of Ribeirão Preto, University of São Paulo, Tenente Catão Roxo, 2501, Monte Alegre, 14051-140, Ribeirão Preto, São Paulo, Brazil.
| | - Kelen Cristina Ribeiro Malmegrim
- Regional Blood Center of Ribeirão Preto, University of São Paulo, Tenente Catão Roxo, 2501, Monte Alegre, 14051-140, Ribeirão Preto, São Paulo, Brazil. .,Department of Clinical, Toxicological and Bromatological Analysis, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café, Monte Alegre, 14040-903, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
174
|
Zwezdaryk KJ, Ferris MB, Strong AL, Morris CA, Bunnell BA, Dhurandhar NV, Gimble JM, Sullivan DE. Human cytomegalovirus infection of human adipose-derived stromal/stem cells restricts differentiation along the adipogenic lineage. Adipocyte 2016; 5:53-64. [PMID: 27144097 DOI: 10.1080/21623945.2015.1119957] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 11/03/2015] [Accepted: 11/05/2015] [Indexed: 12/27/2022] Open
Abstract
Human adipose-derived stromal/stem cells (ASCs) display potential to be used in regenerative stem cell therapies and as treatments for inflammatory and autoimmune disorders. Despite promising use of ASCs as therapeutics, little is known about their susceptibility to infectious agents. In this study, we demonstrate that ASCs are highly susceptible to human cytomegalovirus (HCMV) infection and permissive for replication leading to release of infectious virions. Additionally, many basic ASC functions are inhibited during HCMV infection, such as differentiation and immunomodulatory potential. To our knowledge this is the first study examining potential adverse effects of HCMV infection on ASC biology. Our results suggest, that an active HCMV infection during ASC therapy may result in a poor clinical outcome due to interference by the virus.
Collapse
|
175
|
Luz-Crawford P, Djouad F, Toupet K, Bony C, Franquesa M, Hoogduijn MJ, Jorgensen C, Noël D. Mesenchymal Stem Cell-Derived Interleukin 1 Receptor Antagonist Promotes Macrophage Polarization and Inhibits B Cell Differentiation. Stem Cells 2015; 34:483-92. [PMID: 26661518 DOI: 10.1002/stem.2254] [Citation(s) in RCA: 199] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Revised: 09/03/2015] [Accepted: 09/22/2015] [Indexed: 12/13/2022]
Abstract
The role of interleukin 1 receptor antagonist (IL1RA) in mediating the immunosuppressive effect of mesenchymal stem/stromal cells (MSCs) has been reported in several studies. However, how MSC-derived IL1RA influences the host response has not been clearly investigated. We therefore derived MSCs from the bone marrow of IL1RA knockout mice and evaluated their immunosuppressive effect on different immune cell subsets. IL1RA deficient (IL1RA(-/-) ) or wild type (wt) MSCs inhibited to the same extend the proliferation of T lymphocytes. On the contrary, IL1RA(-/-) MSCs were less effective than wt MSCs to induce in vitro the macrophage polarization from M1 to M2 phenotype secreting IL10 and exerting a suppressive effect on CD4(+) T cells. Moreover compared with wt MSCs, IL1RA(-/-) MSCs did not efficiently support the survival of quiescent B lymphocytes and block their differentiation toward CD19(+) CD138(+) plasmablasts secreting IgG antibodies. The effectiveness of IL1RA secreted by MSCs in controlling inflammation was further shown in vivo using the collagen-induced arthritis murine model. MSCs lacking IL1RA expression were unable to protect mice from arthritic progression and even worsened clinical signs, as shown by higher arthritic score and incidence than control arthritic mice. IL1RA(-/-) MSCs were not able to decrease the percentage of Th17 lymphocytes and increase the percentage of Treg cells as well as decreasing the differentiation of B cells toward plasmablasts. Altogether, our results provide evidence of the key role of IL1RA secreted by MSCs to both control the polarization of macrophages toward a M2 phenotype and inhibit B cell differentiation in vivo.
Collapse
Affiliation(s)
- Patricia Luz-Crawford
- Inserm, u1183, Hôpital Saint-Eloi, Montpellier, France.,Université Montpellier, UFR de Médecine, Montpellier, France
| | - Farida Djouad
- Inserm, u1183, Hôpital Saint-Eloi, Montpellier, France.,Université Montpellier, UFR de Médecine, Montpellier, France
| | - Karine Toupet
- Inserm, u1183, Hôpital Saint-Eloi, Montpellier, France.,Université Montpellier, UFR de Médecine, Montpellier, France
| | - Claire Bony
- Inserm, u1183, Hôpital Saint-Eloi, Montpellier, France.,Université Montpellier, UFR de Médecine, Montpellier, France
| | - Marcella Franquesa
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Martin J Hoogduijn
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Christian Jorgensen
- Inserm, u1183, Hôpital Saint-Eloi, Montpellier, France.,Université Montpellier, UFR de Médecine, Montpellier, France.,Service d'Immuno-Rhumatologie Thérapeutique, Hôpital Lapeyronie, Montpellier, France
| | - Danièle Noël
- Inserm, u1183, Hôpital Saint-Eloi, Montpellier, France.,Université Montpellier, UFR de Médecine, Montpellier, France.,Service d'Immuno-Rhumatologie Thérapeutique, Hôpital Lapeyronie, Montpellier, France
| |
Collapse
|
176
|
Skalska U, Kontny E. Adipose-derived mesenchymal stem cells from infrapatellar fat pad of patients with rheumatoid arthritis and osteoarthritis have comparable immunomodulatory properties. Autoimmunity 2015; 49:124-31. [PMID: 26711868 DOI: 10.3109/08916934.2015.1113267] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Adipose-derived mesenchymal stem cells (ASCs) possess immunosuppressive properties, but their activity is dependent on stimuli provided by local environment. It is possible that proinflammatory milieu of rheumatoid joint affects ASCs function. To verify this hypothesis, rheumatoid ASCs (RA-ASCs) and osteoarthritic ASCs (OA-ASCs) derived from infrapatellar fat pad (IPFP) of the knee joint have been compared. RA- and OA-ASCs isolated from patients were cultured in vitro. Their secretory and proliferative activity was measured. Peripheral blood mononuclear cells (PBMCs) from healthy donors were co-cultured with ASCs. Then, PBMCs proliferation was measured by (3)H-thymidine incorporation method, cytokines secretion by immunoassays, T cells activation and regulatory T cells (Tregs) percentage - by flow cytometry. RA- and OA-ASCs properties in vitro were comparable, however, some differences in secretory activity occurred. RA- and OA-ASCs inhibited PBMCs proliferation and induced interleukin 10 production but up-regulated interleukin 17 A secretion and failed to limit release of other proinflammatory mediators (tumor necrosis factor [TNF], interferon γ [IFNγ], CCL5) by PBMCs. RA- and OA-ASCs did not suppress activation markers expression on T cells and did not trigger Tregs expansion. The present study shows that IPFP-ASCs from RA and OA patients have comparable functions in vitro. Their immunosuppressive activity seems to be impaired comparing to available data.
Collapse
Affiliation(s)
- Urszula Skalska
- a Department of Pathophysiology and Immunology , Institute of Rheumatology , Warsaw , Poland
| | - Ewa Kontny
- a Department of Pathophysiology and Immunology , Institute of Rheumatology , Warsaw , Poland
| |
Collapse
|
177
|
Nagata H, Ii M, Kohbayashi E, Hoshiga M, Hanafusa T, Asahi M. Cardiac Adipose-Derived Stem Cells Exhibit High Differentiation Potential to Cardiovascular Cells in C57BL/6 Mice. Stem Cells Transl Med 2015; 5:141-51. [PMID: 26683873 DOI: 10.5966/sctm.2015-0083] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Accepted: 10/15/2015] [Indexed: 12/17/2022] Open
Abstract
Adipose-derived stem cells (AdSCs) have recently been shown to differentiate into cardiovascular lineage cells. However, little is known about the fat tissue origin-dependent differences in AdSC function and differentiation potential. AdSC-rich cells were isolated from subcutaneous, visceral, cardiac (CA), and subscapular adipose tissue from mice and their characteristics analyzed. After four different AdSC types were cultured with specific differentiation medium, immunocytochemical analysis was performed for the assessment of differentiation into cardiovascular cells. We then examined the in vitro differentiation capacity and therapeutic potential of AdSCs in ischemic myocardium using a mouse myocardial infarction model. The cell density and proliferation activity of CA-derived AdSCs were significantly increased compared with the other adipose tissue-derived AdSCs. Immunocytochemistry showed that CA-derived AdSCs had the highest appearance rates of markers for endothelial cells, vascular smooth muscle cells, and cardiomyocytes among the AdSCs. Systemic transfusion of CA-derived AdSCs exhibited the highest cardiac functional recovery after myocardial infarction and the high frequency of the recruitment to ischemic myocardium. Moreover, long-term follow-up of the recruited CA-derived AdSCs frequently expressed cardiovascular cell markers compared with the other adipose tissue-derived AdSCs. Cardiac adipose tissue could be an ideal source for isolation of therapeutically effective AdSCs for cardiac regeneration in ischemic heart diseases. Significance: The present study found that cardiac adipose-derived stem cells have a high potential to differentiate into cardiovascular lineage cells (i.e., cardiomyocytes, endothelial cells, and vascular smooth muscle cells) compared with stem cells derived from other adipose tissue such as subcutaneous, visceral, and subscapular adipose tissue. Notably, only a small number of supracardiac adipose-derived stem cells that were systemically transplanted sufficiently improved cardiac functional recovery after myocardial infarction, differentiating into cardiovascular cells in the ischemic myocardium. These findings suggest a new autologous stem cell therapy for patients with myocardial ischemia, especially those with secondary myocardial ischemia after cardiovascular open chest surgery.
Collapse
Affiliation(s)
- Hiroki Nagata
- Department of Pharmacology, Faculty of Medicine, Osaka Medical College, Osaka, Japan Department of Internal Medicine (I), Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Masaaki Ii
- Department of Pharmacology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Eiko Kohbayashi
- Department of Internal Medicine (I), Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Masaaki Hoshiga
- Department of Internal Medicine (III), Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Toshiaki Hanafusa
- Department of Internal Medicine (I), Faculty of Medicine, Osaka Medical College, Osaka, Japan
| | - Michio Asahi
- Department of Pharmacology, Faculty of Medicine, Osaka Medical College, Osaka, Japan
| |
Collapse
|
178
|
Hunter RK, Nevitt CD, Gaskins JT, Keller BB, Bohler HCL, LeBlanc AJ. Adipose-Derived Stromal Vascular Fraction Cell Effects on a Rodent Model of Thin Endometrium. PLoS One 2015; 10:e0144823. [PMID: 26657744 PMCID: PMC4684382 DOI: 10.1371/journal.pone.0144823] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 11/24/2015] [Indexed: 12/26/2022] Open
Abstract
Endometrial dysfunction affects approximately 1% of infertile women, and there is currently no standard therapy for improving fertility treatment outcomes in these patients. In our study, we utilized a rodent model of thin endometrium to test whether intrauterine application of adipose-derived stromal vascular fraction cells (SVF) could improve morphological and physiological markers of endometrial receptivity. Using anhydrous ethanol, endometrial area and gland density were significantly reduced in our model of thin endometrium. Application of SVF was associated with a 29% reduction in endometrial vascular endothelial growth factor (VEGF) expression and significant increases in uterine artery systolic/diastolic velocity ratios and resistance index values, suggesting reduced diastolic microvascular tone. However, no significant improvements in endometrial area or gland density were observed following SVF treatment. 3D confocal imaging demonstrated poor engraftment of SVF cells into recipient tissue, which likely contributed to the negative results of this study. We suspect modified treatment protocols utilizing adjuvant estrogen and/or tail vein cell delivery may improve SVF retention and therapeutic response in subsequent studies. SVF is an easily-obtainable cell product with regenerative capability that may have a future role in the treatment of infertile women with endometrial dysfunction.
Collapse
Affiliation(s)
- Robert K. Hunter
- Department of Obstetrics, Gynecology and Women’s Health, Division of Reproductive Endocrinology and Infertility, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Chris D. Nevitt
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Jeremy T. Gaskins
- Department of Bioinformatics and Biostatistics, University of Louisville School of Public Health and Information Sciences, Louisville, Kentucky, United States of America
| | - Bradley B. Keller
- Cardiovascular Innovation Institute, Louisville, Kentucky, United States of America
| | - Henry C. L. Bohler
- Department of Obstetrics, Gynecology and Women’s Health, Division of Reproductive Endocrinology and Infertility, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Amanda J. LeBlanc
- Cardiovascular Innovation Institute, Louisville, Kentucky, United States of America
- Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
179
|
Kasir R, Vernekar VN, Laurencin CT. Regenerative Engineering of Cartilage Using Adipose-Derived Stem Cells. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2015; 1:42-49. [PMID: 26998511 PMCID: PMC4795960 DOI: 10.1007/s40883-015-0005-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 11/19/2015] [Indexed: 12/13/2022]
Abstract
Injury to the articular cartilage occurs commonly in the general population and undergoes minimal spontaneous healing. Traditional methods of cartilage repair provide no long-term cure and are significant causes of morbidity. For this reason, stem cell therapies have recently been investigated for their ability to regenerate cartilage, and the results have been promising. Since the discovery that adipose tissue is a major source of mesenchymal stem cells in 2001, scientists have been studying the use of adipose-derived stem cells (ASCs) for the treatment of various disorders including lesions of the articular cartilage. ASCs hold several advantages over autologous chondrocytes for cartilage repair, including but not limited to their anti-inflammatory effects, their multi-lineage differentiation potential, and their ability to form new cartilage in a defect. Whereas several investigations have been made in in vitro and animal models, there have been surprisingly little clinical studies on the intra-articular use of adipose-derived stem cells, despite their first isolation about a decade and a half ago. The few studies that have been conducted are encouraging. With approval for various stem cell therapies on the horizon, this review seeks to update the clinician and the researcher on the current state-of-the-art use of adipose-derived stem cells for the treatment of cartilage disorders and the regenerative engineering of cartilaginous tissue.
Collapse
Affiliation(s)
- Rafid Kasir
- Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
- School of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Varadraj N. Vernekar
- Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
- School of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Cato T. Laurencin
- Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
- School of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
180
|
Adipose- and Bone Marrow-Derived Mesenchymal Stem Cells Prolong Graft Survival in Vascularized Composite Allotransplantation. Transplantation 2015; 99:1765-73. [PMID: 26102613 DOI: 10.1097/tp.0000000000000731] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Strategies aiming at minimization or elimination of systemic immunosuppression are key immediate goals for clinical expansion of vascularized composite allotransplantation (VCA). We compared the in vitro and in vivo immunomodulatory efficacy of adipose-derived mesenchymal stem cells (AD-MSCs) and bone marrow (BM)-derived MSCs in a rat VCA model. METHODS Both cell types were tested in vitro for suppressor function using mixed lymphocyte reactivity assays. AD-MSCs or BM-MSCs were administered intravenously (1 × 10 or 5 × 10 cells/animal) to Lewis rat recipients of mismatched Brown Norway hindlimb transplants. Short course tacrolimus (FK-506) monotherapy was withdrawn at postoperative day 21. In vivo regulatory T-cell induction, peripheral blood chimerism, and microchimerism in lymphatic organs were analyzed. RESULTS AD-MSCs and BM-MSCs exhibited strong dose-dependent suppressor function in vitro, which was significantly more pronounced for AD cells. In vivo, all animals revealed peripheral multi-lineage chimerism at four weeks (P < 0.01) independent of cell type and dosage. Regulatory T-cell levels were increased with both cell types, the most in AD-MSC groups. These immunomodulatory effects were only transient. MSC treatment resulted in long-term (>120 day) allograft survival in 47% of the animals, which correlated with durable microchimerism in BM and spleen. CONCLUSIONS AD-MSCs and BM-MSCs exert immunomodulatory effects that prolong survival of immunogenic skin-bearing VCA grafts with short course (21 day) tacrolimus induction therapy. The in vivo findings in terms of allograft survival did not reflect superior immunomodulatory characteristics of AD-MSCs found in vitro.
Collapse
|
181
|
Feisst V, Meidinger S, Locke MB. From bench to bedside: use of human adipose-derived stem cells. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2015; 8:149-62. [PMID: 26586955 PMCID: PMC4636091 DOI: 10.2147/sccaa.s64373] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Since the discovery of adipose-derived stem cells (ASC) in human adipose tissue nearly 15 years ago, significant advances have been made in progressing this promising cell therapy tool from the laboratory bench to bedside usage. Standardization of nomenclature around the different cell types used is finally being adopted, which facilitates comparison of results between research groups. In vitro studies have assessed the ability of ASC to undergo mesenchymal differentiation as well as differentiation along alternate lineages (transdifferentiation). Recently, focus has shifted to the immune modulatory and paracrine effects of transplanted ASC, with growing interest in the ASC secretome as a source of clinical effect. Bedside use of ASC is advancing alongside basic research. An increasing number of safety-focused Phase I and Phase IIb trials have been published without identifying any significant risks or adverse events in the short term. Phase III trials to assess efficacy are currently underway. In many countries, regulatory frameworks are being developed to monitor their use and assure their safety. As many trials rely on ASC injected at a distant site from the area of clinical need, strategies to improve the homing and efficacy of transplanted cells are also being explored. This review highlights each of these aspects of the bench-to-bedside use of ASC and summarizes their clinical utility across a variety of medical specialties.
Collapse
Affiliation(s)
- Vaughan Feisst
- Dunbar Laboratory, School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Sarah Meidinger
- Dunbar Laboratory, School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Michelle B Locke
- Department of Surgery, Faculty of Medicine and Health Sciences, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
182
|
Alawad A, Altuwaijri S, Aljarbu A, Kryczek I, Niu Y, Al-sobayil FA, Chang C, Bayoumi A, Zou W, Rudat V, Hammad M. Depletion of androgen receptor (AR) in mesenchymal stem cells (MSCs) inhibits induction of CD4+CD25+FOX3+ regulatory T (Treg) cells via androgen TGF-β interaction. J Appl Biomed 2015. [DOI: 10.1016/j.jab.2015.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
183
|
Tomé M, Sepúlveda JC, Delgado M, Andrades JA, Campisi J, González MA, Bernad A. miR-335 correlates with senescence/aging in human mesenchymal stem cells and inhibits their therapeutic actions through inhibition of AP-1 activity. Stem Cells 2015; 32:2229-44. [PMID: 24648336 DOI: 10.1002/stem.1699] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 02/25/2014] [Accepted: 03/02/2014] [Indexed: 12/13/2022]
Abstract
MicroRNAs, small noncoding RNAs, regulate gene expression primarily at the posttranscriptional level. We previously found that miR-335 is critically involved in the regulation and differentiation capacity of human mesenchymal stem cells (hMSCs) in vitro. In this study, we investigated the significance of miR-335 for the therapeutic potential of hMSCs. Analysis of hMSCs in ex vivo culture demonstrated a significant and progressive increase in miR-335 that is prevented by telomerase. Expression levels of miR-335 were also positively correlated with donor age of hMSCs, and were increased by stimuli that induce cell senescence, such as γ-irradiation and standard O2 concentration. Forced expression of miR-335 resulted in early senescence-like alterations in hMSCs, including: increased SA-β-gal activity and cell size, reduced cell proliferation capacity, augmented levels of p16 protein, and the development of a senescence-associated secretory phenotype. Furthermore, overexpression of miR-335 abolished the in vivo chondro-osseous potential of hMSCs, and disabled their immunomodulatory capacity in a murine experimental model of lethal endotoxemia. These effects were accompanied by a severely reduced capacity for cell migration in response to proinflammatory signals and a marked reduction in Protein Kinase D1 phosphorylation, resulting in a pronounced decrease of AP-1 activity. Our results demonstrate that miR-335 plays a key role in the regulation of reparative activities of hMSCs and suggests that it might be considered a marker for the therapeutic potency of these cells in clinical applications.
Collapse
Affiliation(s)
- María Tomé
- Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
184
|
Strong AL, Burow ME, Gimble JM, Bunnell BA. Concise review: The obesity cancer paradigm: exploration of the interactions and crosstalk with adipose stem cells. Stem Cells 2015; 33:318-26. [PMID: 25267443 DOI: 10.1002/stem.1857] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 07/28/2014] [Accepted: 08/06/2014] [Indexed: 01/04/2023]
Abstract
With the recognition of obesity as a global health crisis, researchers have devoted greater effort to defining and understanding the pathophysiological molecular pathways regulating the biology of adipose tissue and obesity. Obesity, the excessive accumulation of adipose tissue due to hyperplasia and hypertrophy, has been linked to an increased incidence and aggressiveness of colon, hematological, prostate, and postmenopausal breast cancers. The increased morbidity and mortality of obesity-associated cancers have been attributed to higher levels of hormones, adipokines, and cytokines secreted by the adipose tissue. The increased amount of adipose tissue also results in higher numbers of adipose stromal/stem cells (ASCs). These ASCs have been shown to impact cancer progression directly through several mechanisms, including the increased recruitment of ASCs to the tumor site and increased production of cytokines and growth factors by ASCs and other cells within the tumor stroma. Emerging evidence indicates that obesity induces alterations in the biologic properties of ASCs, subsequently leading to enhanced tumorigenesis and metastasis of cancer cells. This review will discuss the links between obesity and cancer tumor progression, including obesity-associated changes in adipose tissue, inflammation, adipokines, and chemokines. Novel topics will include a discussion of the contribution of ASCs to this complex system with an emphasis on their role in the tumor stroma. The reciprocal and circular feedback loop between obesity and ASCs as well as the mechanisms by which ASCs from obese patients alter the biology of cancer cells and enhance tumorigenesis will be discussed.
Collapse
Affiliation(s)
- Amy L Strong
- Center for Stem Cell Research and Regenerative Medicine
| | | | | | | |
Collapse
|
185
|
Han TTY, Toutounji S, Amsden BG, Flynn LE. Adipose-derived stromal cells mediate in vivo adipogenesis, angiogenesis and inflammation in decellularized adipose tissue bioscaffolds. Biomaterials 2015; 72:125-37. [PMID: 26360790 DOI: 10.1016/j.biomaterials.2015.08.053] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 08/25/2015] [Accepted: 08/28/2015] [Indexed: 12/20/2022]
Abstract
Decellularized adipose tissue (DAT) has shown promise as an adipogenic bioscaffold for soft tissue augmentation and reconstruction. The objective of the current study was to investigate the effects of allogeneic adipose-derived stem/stromal cells (ASCs) on in vivo fat regeneration in DAT bioscaffolds using an immunocompetent rat model. ASC seeding significantly enhanced angiogenesis and adipogenesis, with cell tracking studies indicating that the newly-forming tissues were host-derived. Incorporating ASCs also mediated the inflammatory response and promoted a more constructive macrophage phenotype. A fraction of the CD163(+) macrophages in the implants expressed adipogenic markers, with higher levels of this "adipocyte-like" phenotype in proximity to the developing adipose tissues. Our results indicate that the combination of ASCs and adipose extracellular matrix (ECM) provides an inductive microenvironment for adipose regeneration mediated by infiltrating host cell populations. The DAT scaffolds are a useful tissue-specific model system for investigating the mechanisms of in vivo adipogenesis that may help to develop a better understanding of this complex process in the context of both regeneration and disease. Overall, combining adipose-derived matrices with ASCs is a highly promising approach for the in situ regeneration of host-derived adipose tissue.
Collapse
Affiliation(s)
- Tim Tian Y Han
- Department of Chemical Engineering, Queen's University, 19 Division Street, Kingston, Ontario, Canada, K7L 3N6; Human Mobility Research Centre, Kingston General Hospital, 76 Stuart Street, Kingston, Ontario, Canada, K7L 2V7
| | - Sandra Toutounji
- Department of Chemical Engineering, Queen's University, 19 Division Street, Kingston, Ontario, Canada, K7L 3N6
| | - Brian G Amsden
- Department of Chemical Engineering, Queen's University, 19 Division Street, Kingston, Ontario, Canada, K7L 3N6; Human Mobility Research Centre, Kingston General Hospital, 76 Stuart Street, Kingston, Ontario, Canada, K7L 2V7
| | - Lauren E Flynn
- Department of Chemical Engineering, Queen's University, 19 Division Street, Kingston, Ontario, Canada, K7L 3N6; Human Mobility Research Centre, Kingston General Hospital, 76 Stuart Street, Kingston, Ontario, Canada, K7L 2V7; Department of Chemical and Biochemical Engineering, Thompson Engineering Building, The University of Western Ontario, London, Ontario, Canada, N6A 5B9; Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada, N6A 5C1.
| |
Collapse
|
186
|
Human Adipose-Derived Mesenchymal Stem Cells Modulate Experimental Autoimmune Arthritis by Modifying Early Adaptive T Cell Responses. Stem Cells 2015. [DOI: 10.1002/stem.2113] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
187
|
Abstract
PURPOSE OF REVIEW The prognosis of patients with respiratory failure in the ICU remains poor, while current therapeutic approaches are aimed at minimizing ventilator-induced lung injury. Stem cell-based therapies have the potential to transform respiratory failure treatment by achieving lung repair. The purpose of this article is to critically review the large body of clinical and experimental work performed with respect to the use of stem/progenitor cells in respiratory failure, and to discuss current challenges and future directions. RECENT FINDINGS Since the initial report of cell therapy for lung injury in 2005, numerous preclinical and clinical studies have been performed that support the ability of various stem cell populations to improve physiologic lung function and reduce inflammation in both infective and sterile acute respiratory distress syndrome. Nevertheless, many important issues (e.g., mechanism of action, long-term engraftment, optimal cell type, dose, route of administration) remain to be resolved. SUMMARY Cell-based therapeutics hold promise, particularly for acute respiratory distress syndrome, and early preclinical testing has been encouraging. To advance clinical testing of cell therapies in respiratory failure, and to help ensure that this approach will facilitate bench-to-bedside and bedside-to-bench discoveries, parallel paths of basic and clinical research are needed, including measures of cell therapy effectiveness in vivo and in vitro.
Collapse
|
188
|
Human adipose-derived mesenchymal stem cells attenuate collagen antibody-induced autoimmune arthritis by inducing expression of FCGIIB receptors. BMC Musculoskelet Disord 2015. [PMID: 26210906 PMCID: PMC4515315 DOI: 10.1186/s12891-015-0634-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background Adipose-derived stem cells (ASCs) are mesenchymal stem cells (MSCs) derived from adipose tissue. MSCs have multiple properties including anti-inflammatory and immunomodulatory effects in various disease models and human diseases. However, the mechanisms underlying this wide range of effects need to be explored. Methods Collagen antibody-induced arthritis (CAIA) is a unique model in which arthritis is rapidly and strongly induced. ASCs were intraperitoneally infused into CAIA mice before or after arthritis induction. The serum levels of various cytokines, adipokines, and chemokines were measured. The expression of FC gamma receptors (FCGRs) was investigated in peritoneal macrophages ex vivo. RAW264.7 cells and ASCs were co-cultured to elucidate the direct and indirect role of ASCs on FCGR expression. Results ASCs attenuated arthritis in CAIA mice. Serum levels of tumor necrosis factor α, interleukin (IL)-15, resistin, and leptin were reduced in ASC-treated CAIA mice, whereas serum levels of IL-6 and adiponectin were not affected. In peritoneal macrophages isolated from ASC-treated mice, expression of FCGRIIB, which is immunoinhibitory, was higher than that of FCGRI. Co-culture of ASCs with RAW264.7 cells modulated the expression of FCGRs. The expression patterns and timings of peak expression differed among FCGRs. Expression of FCGRIIB was higher and peaked earlier than that of FCGRI. FCGRIII expression was not affected by this co-culture. Conclusions This is a study to show that ASCs have anti-arthritic effects in CAIA mice. Modulation of FCGRs by ASCs might be a therapeutic mechanism in this antibody-associated arthritis model.
Collapse
|
189
|
Research Progress on Regulatory T Cells in Acute Kidney Injury. J Immunol Res 2015; 2015:174164. [PMID: 26273681 PMCID: PMC4529954 DOI: 10.1155/2015/174164] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 07/02/2015] [Indexed: 02/06/2023] Open
Abstract
Immune inflammation is crucial in mediating acute kidney injury (AKI). Immune cells of both the innate and adaptive immune systems substantially contribute to overall renal damage in AKI. Regulatory T cells (Tregs) are key regulator of immunological function and have been demonstrated to ameliorate injury in several murine experimental models of renal inflammation. Recent studies have illuminated the renal-protective function of Tregs in AKI. Tregs appear to exert beneficial effects in both the acute injury phase and the recovery phase of AKI. Additionally, Tregs-based immunotherapy may represent a promising approach to ameliorate AKI and promote recovery from AKI. This review will highlight the recent insights into the role of Tregs and their therapeutic potential in AKI.
Collapse
|
190
|
Xie ZH, Liu Z, Zhang XR, Yang H, Wei LF, Wang Y, Xu SL, Sun L, Lai C, Bi JZ, Wang XY. Wharton's Jelly-derived mesenchymal stem cells alleviate memory deficits and reduce amyloid-β deposition in an APP/PS1 transgenic mouse model. Clin Exp Med 2015; 16:89-98. [PMID: 26188488 DOI: 10.1007/s10238-015-0375-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 07/06/2015] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in the elderly and is characterized by amyloid plaques, neurofibrillary tangles, and neuronal loss. Cumulative evidence supports that neuroinflammation is an important factor for the pathogenesis of AD and contributes to amyloid beta (Aβ) generation. However, there has been no effective treatment for AD. Wharton's Jelly-derived mesenchymal stem cells (WJ-MSCs) have a potential therapeutic effect in the treatment for neurological diseases. In the present study, we evaluated the therapeutic effect of WJ-MSC transplantation on the neuropathology and memory deficits in amyloid precursor protein (APP) and presenilin-1 (PS1) double-transgenic mice and discussed the mechanism. WJ-MSCs were intravenously transplanted into the APP/PS1 mice. Four weeks after treatment, WJ-MSCs significantly improved the spatial learning and alleviated the memory decline in the APP/PS1 mice. Aβ deposition and soluble Aβ levels were significantly reduced after WJ-MSC treatment. Furthermore, WJ-MSCs significantly increased the expression of the anti-inflammatory cytokine, IL-10. Meanwhile, pro-inflammatory microglial activation and the expressions of pro-inflammatory cytokines, IL-1β and TNFα, were significantly down-regulated by WJ-MSC treatment. Thus, our findings suggest that WJ-MSCs might produce beneficial effects on the prevention and treatment for AD through modulation of neuroinflammation.
Collapse
Affiliation(s)
- Zhao-Hong Xie
- Department of Neurology, The Second Hospital of Shandong University, 247#, Beiyuan Dajie, Jinan, 250033, China
| | - Zhen Liu
- Department of Neurology, The Second Hospital of Shandong University, 247#, Beiyuan Dajie, Jinan, 250033, China
| | - Xiao-Ran Zhang
- Department of Pharmacy, Jinan Central Hospital, 105#, Jiefang Road, Jinan, 250013, China
| | - Hui Yang
- Department of Neurology, The Second Hospital of Shandong University, 247#, Beiyuan Dajie, Jinan, 250033, China
| | - Li-Fei Wei
- Department of Neurology, The Second Hospital of Shandong University, 247#, Beiyuan Dajie, Jinan, 250033, China
| | - Yun Wang
- Department of Neurology, The Second Hospital of Shandong University, 247#, Beiyuan Dajie, Jinan, 250033, China
| | - Shun-Liang Xu
- Department of Neurology, The Second Hospital of Shandong University, 247#, Beiyuan Dajie, Jinan, 250033, China
| | - Lin Sun
- Department of Neurology, The Second Hospital of Shandong University, 247#, Beiyuan Dajie, Jinan, 250033, China
| | - Chao Lai
- Department of Neurology, The Second Hospital of Shandong University, 247#, Beiyuan Dajie, Jinan, 250033, China
| | - Jian-Zhong Bi
- Department of Neurology, The Second Hospital of Shandong University, 247#, Beiyuan Dajie, Jinan, 250033, China
| | - Xiao-Yun Wang
- Department of Neurology, The Second Hospital of Shandong University, 247#, Beiyuan Dajie, Jinan, 250033, China.
| |
Collapse
|
191
|
In Vitro Effects of Strontium on Proliferation and Osteoinduction of Human Preadipocytes. Stem Cells Int 2015; 2015:871863. [PMID: 26240575 PMCID: PMC4512617 DOI: 10.1155/2015/871863] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 03/10/2015] [Indexed: 12/15/2022] Open
Abstract
Development of tools to be used for in vivo bone tissue regeneration focuses on cellular models and differentiation processes. In searching for all the optimal sources, adipose tissue-derived mesenchymal stem cells (hADSCs or preadipocytes) are able to differentiate into osteoblasts with analogous characteristics to bone marrow mesenchymal stem cells, producing alkaline phosphatase (ALP), collagen, osteocalcin, and calcified nodules, mainly composed of hydroxyapatite (HA). The possibility to influence bone differentiation of stem cells encompasses local and systemic methods, including the use of drugs administered systemically. Among the latter, strontium ranelate (SR) represents an interesting compound, acting as an uncoupling factor that stimulates bone formation and inhibits bone resorption. The aim of our study was to evaluate the in vitro effects of a wide range of strontium (Sr(2+)) concentrations on proliferation, ALP activity, and mineralization of a novel finite clonal hADSCs cell line, named PA20-h5. Sr(2+) promoted PA20-h5 cell proliferation while inducing the increase of ALP activity and gene expression as well as HA production during in vitro osteoinduction. These findings indicate a role for Sr(2+) in supporting bone regeneration during the process of skeletal repair in general, and, more specifically, when cell therapies are applied.
Collapse
|
192
|
Parvizi M, Harmsen MC. Therapeutic Prospect of Adipose-Derived Stromal Cells for the Treatment of Abdominal Aortic Aneurysm. Stem Cells Dev 2015; 24:1493-505. [DOI: 10.1089/scd.2014.0517] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Mojtaba Parvizi
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Martin C. Harmsen
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
193
|
Choi EW, Shin IS, Song JW, Lee M, Yun TW, Yang J, Choi KS, Kim SJ. Effects of Transplantation of CTLA4Ig-Overexpressing Adipose Tissue-Derived Mesenchymal Stem Cells in Mice With Sustained Severe Rheumatoid Arthritis. Cell Transplant 2015; 25:243-59. [PMID: 26087385 DOI: 10.3727/096368915x688470] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
CTLA4Ig has therapeutic potential for rheumatoid arthritis patients unresponsive to methotrexate (MTX) or TNF-α blockers. However, recombinant CTLA4Ig proteins are short acting and expensive. Adipose tissue-derived mesenchymal stem cells (ASCs) present an ideal stem cell source for practical regenerative medicine due to their abundant availability and their beneficial properties including immunomodulation, homing activity, paracrine effects, and differentiation ability. Therefore, we aimed to determine whether CTLA4Ig and human ASCs show synergistic effects on immunomodulation and clinical improvement of sustained severe rheumatoid arthritis in a mouse model. hASCs overexpressing CTLA4Ig (CTLA4Ig-hASC) were serially transplanted into mice with collagen-induced arthritis. Arthritic mice were subjected to four treatments based on their arthritis score on day 62 postimmunization: control (C group), hASC (H group), CTLA4Ig-hASC (CT group), and MTX (MTX group). A group of healthy mice was used as a normal control (N). Mice in the N and C groups were infused with 150 µl saline, and 2 × 10(6) hASCs or CTLA4Ig-hASCs in 150 µl of saline were intravenously administered to those in the H and CT groups, respectively, on days 63, 70, 77, and 84 after CII immunization. About 1 mg/kg of methotrexate was intraperitoneally administered to the MTX group three times a week for 4 weeks. Serial hASC and CTLA4Ig-hASC transplantation modulated various cytokines and chemokines related to the development of rheumatoid arthritis. Both treatments protected against destruction of cartilage, with CTLA4Ig-hASCs being most effective. Serum levels of CII autoantibodies and C-telopeptide of type II collagen were significantly low in the group transplanted with CTLA4Ig-hASCs. In vitro, ASC and CTLA4Ig-hASC treatment significantly decreased T-bet and GATA-3 expression in splenocytes from arthritic mice, and CTLA4Ig-hASC treatment significantly increased the ratio of Treg/Th17 (CD4(+)CD25(+)FoxP3(+)/CD4(+)CD25(+)RORγt) cells. Serial hASC and CTLA4Ig-hASC transplantation offers promising treatment for rheumatoid arthritis, and CTLA4Ig-hASCs showed stronger therapeutic effects than nontransduced hASCs.
Collapse
Affiliation(s)
- Eun Wha Choi
- Laboratory Animal Research Center, Samsung Biomedical Research Institute, Gangnam-gu, Seoul, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
194
|
Zeng SL, Wang LH, Li P, Wang W, Yang J. Mesenchymal stem cells abrogate experimental asthma by altering dendritic cell function. Mol Med Rep 2015; 12:2511-20. [PMID: 25936350 PMCID: PMC4464445 DOI: 10.3892/mmr.2015.3706] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 03/26/2015] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been investigated in the treatment of numerous autoimmune diseases. However, the immune properties of MSCs on the development of asthma have remained to be fully elucidated. Airway dendritic cells (DCs) have an important role in the pathogenesis of allergic asthma, and disrupting their function may be a novel therapeutic approach. The present study used a mouse model of asthma to demonstrate that transplantation of MSCs suppressed features of asthma by targeting the function of lung myeloid DCs. MSCs suppressed the maturation and migration of lung DCs to the mediastinal lymph nodes, and thereby reducing the allergen-specific T helper type 2 (Th2) response in the nodes. In addition, MSC-treated DCs were less potent in activating naive and effector Th2 cells and the capacity of producing chemokine (C-C motif) ligand 17 (CCL17) and CCL22, which are chemokines attracting Th2 cells, to the airways was reduced. These results supported that MSCs may be used as a potential treatment for asthma.
Collapse
Affiliation(s)
- Shao-Lin Zeng
- Department of Respiratory Medicine, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Li-Hui Wang
- Department of Respiratory Medicine, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Ping Li
- Department of Respiratory Medicine, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Wei Wang
- Department of Respiratory Medicine, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Jiong Yang
- Department of Respiratory Medicine, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
195
|
Wada N, Gronthos S, Bartold PM. Immunomodulatory effects of stem cells. Periodontol 2000 2015; 63:198-216. [PMID: 23931061 DOI: 10.1111/prd.12024] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2012] [Indexed: 02/06/2023]
Abstract
Adult-derived mesenchymal stem cells have received considerable attention over the past two decades for their potential use in tissue engineering, principally because of their potential to differentiate into multiple stromal-cell lineages. Recently, the immunomodulatory properties of mesenchymal stem cells have attracted interest as a unique property of these cells that may be harnessed for novel therapeutic approaches in immune-mediated diseases. Mesenchymal stem cells have been shown to inhibit the proliferation of activated T-cells both in vitro and in vivo but to stimulate T-regulatory cell proliferation. Mesenchymal stem cells are also known to be weakly immunogenic and to exert immunosuppressive effects on B-cells, natural killer cells, dendritic cells and neutrophils through various mechanisms. Furthermore, intravenous administration of allogeneic mesenchymal stem cells has shown a marked suppression of host immune reactions in preclinical animal models of large-organ transplant rejection and in various autoimmune- and inflammatory-based diseases. Some clinical trials utilizing human mesenchymal stem cells have also produced promising outcomes in patients with graft-vs.-host disease and autoimmune diseases. Mesenchymal stem cells identified from various dental tissues, including periodontal ligament stem cells, also possess multipotent and immunomodulatory properties. Hence, dental mesenchymal stem cells may represent an alternate cell source, not only for tissue regeneration but also as therapies for autoimmune- and inflammatory-mediated diseases. These findings have elicited interest in dental tissue mesenchymal stem cells as alternative cell sources for modulating alloreactivity during tissue regeneration following transplantation into human leukocyte antigen-mismatched donors. To examine this potential in periodontal regeneration, future work will need to assess the capacity of allogeneic periodontal ligament stem cells to regenerate periodontal ligament in animal models of periodontal disease. The present review describes the immunosuppressive effects of mesenchymal stem cells on various types of immune cells, the potential mechanisms through which they exert their mode of action and the preclinical animal studies and human clinical trials that have utilized mesenchymal stem cells, including those populations originating from dental structures.
Collapse
|
196
|
Minteer DM, Marra KG, Rubin JP. Adipose stem cells: biology, safety, regulation, and regenerative potential. Clin Plast Surg 2015; 42:169-79. [PMID: 25827561 DOI: 10.1016/j.cps.2014.12.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This article discusses adipose-derived stem cell (ASC) biology, describes the current knowledge in the literature for the safety and regulation of ASCs, and provides a brief overview of the regenerative potential of ASCs. It is not an exhaustive listing of all available clinical studies or every study applying ASCs in tissue engineering and regenerative medicine, but is an objective commentary of these topics.
Collapse
Affiliation(s)
- Danielle M Minteer
- Department of Bioengineering, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Kacey G Marra
- Department of Bioengineering, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA; Department of Plastic Surgery, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA 15213, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15213, USA
| | - J Peter Rubin
- Department of Bioengineering, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA; Department of Plastic Surgery, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA 15213, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15213, USA.
| |
Collapse
|
197
|
Cho YB, Park KJ, Yoon SN, Song KH, Kim DS, Jung SH, Kim M, Jeong HY, Yu CS. Long-term results of adipose-derived stem cell therapy for the treatment of Crohn's fistula. Stem Cells Transl Med 2015; 4:532-7. [PMID: 25829404 DOI: 10.5966/sctm.2014-0199] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 02/09/2015] [Indexed: 12/16/2022] Open
Abstract
A previous phase II clinical trial of adipose-derived stem cell (ASC) therapy for fistulae associated with Crohn's disease, a devastating condition with a high recurrence rate, demonstrated safety and therapeutic potential with a 1-year sustained response. In the present study, 41 of the 43 phase II trial patients were followed for an additional year, regardless of response in the initial year. At 24 months, complete healing was observed in 21 of 26 patients (80.8%) in modified per protocol analysis and 27 of 36 patients (75.0%) in modified intention-to-treat analysis. No adverse events related to ASC administration were observed. Furthermore, complete closure after initial treatment was well-sustained. These results strongly suggest that autologous ASCs may be a novel treatment option for Crohn's fistulae.
Collapse
Affiliation(s)
- Yong Beom Cho
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea; Department of Surgery, Seoul National University College of Medicine, Seoul, Korea; Department of Colon & Rectal Surgery, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea; Department of Surgery, Daehang Hospital, Seoul, Korea; Department of Surgery, Yeungnam University Medical Center, Daegu, Korea; Anterogen Co., Ltd., Seoul, Korea
| | - Kyu Joo Park
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea; Department of Surgery, Seoul National University College of Medicine, Seoul, Korea; Department of Colon & Rectal Surgery, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea; Department of Surgery, Daehang Hospital, Seoul, Korea; Department of Surgery, Yeungnam University Medical Center, Daegu, Korea; Anterogen Co., Ltd., Seoul, Korea
| | - Sang Nam Yoon
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea; Department of Surgery, Seoul National University College of Medicine, Seoul, Korea; Department of Colon & Rectal Surgery, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea; Department of Surgery, Daehang Hospital, Seoul, Korea; Department of Surgery, Yeungnam University Medical Center, Daegu, Korea; Anterogen Co., Ltd., Seoul, Korea
| | - Kee Ho Song
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea; Department of Surgery, Seoul National University College of Medicine, Seoul, Korea; Department of Colon & Rectal Surgery, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea; Department of Surgery, Daehang Hospital, Seoul, Korea; Department of Surgery, Yeungnam University Medical Center, Daegu, Korea; Anterogen Co., Ltd., Seoul, Korea
| | - Do Sun Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea; Department of Surgery, Seoul National University College of Medicine, Seoul, Korea; Department of Colon & Rectal Surgery, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea; Department of Surgery, Daehang Hospital, Seoul, Korea; Department of Surgery, Yeungnam University Medical Center, Daegu, Korea; Anterogen Co., Ltd., Seoul, Korea
| | - Sang Hun Jung
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea; Department of Surgery, Seoul National University College of Medicine, Seoul, Korea; Department of Colon & Rectal Surgery, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea; Department of Surgery, Daehang Hospital, Seoul, Korea; Department of Surgery, Yeungnam University Medical Center, Daegu, Korea; Anterogen Co., Ltd., Seoul, Korea
| | - Mihyung Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea; Department of Surgery, Seoul National University College of Medicine, Seoul, Korea; Department of Colon & Rectal Surgery, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea; Department of Surgery, Daehang Hospital, Seoul, Korea; Department of Surgery, Yeungnam University Medical Center, Daegu, Korea; Anterogen Co., Ltd., Seoul, Korea
| | - Hee Young Jeong
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea; Department of Surgery, Seoul National University College of Medicine, Seoul, Korea; Department of Colon & Rectal Surgery, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea; Department of Surgery, Daehang Hospital, Seoul, Korea; Department of Surgery, Yeungnam University Medical Center, Daegu, Korea; Anterogen Co., Ltd., Seoul, Korea
| | - Chang Sik Yu
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea; Department of Surgery, Seoul National University College of Medicine, Seoul, Korea; Department of Colon & Rectal Surgery, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea; Department of Surgery, Daehang Hospital, Seoul, Korea; Department of Surgery, Yeungnam University Medical Center, Daegu, Korea; Anterogen Co., Ltd., Seoul, Korea
| |
Collapse
|
198
|
Lombardo E, Poll TVD, DelaRosa O, Dalemans W. Mesenchymal stem cells as a therapeutic tool to treat sepsis. World J Stem Cells 2015; 7:368-379. [PMID: 25815121 PMCID: PMC4369493 DOI: 10.4252/wjsc.v7.i2.368] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 11/13/2014] [Accepted: 12/17/2014] [Indexed: 02/06/2023] Open
Abstract
Sepsis is a clinical syndrome caused by a deregulated host response to an infection. Sepsis is the most frequent cause of death in hospitalized patients. Although knowledge of the pathogenesis of sepsis has increased substantially during the last decades, attempts to design effective and specific therapies targeting components of the derailed host response have failed. Therefore, there is a dramatic need for new and mechanistically alternative therapies to treat this syndrome. Based on their immunomodulatory properties, adult mesenchymal stem or stromal cells (MSCs) can be a novel therapeutic tool to treat sepsis. Indeed, MSCs reduce mortality in experimental models of sepsis by modulating the deregulated inflammatory response against bacteria through the regulation of multiple inflammatory networks, the reprogramming of macrophages and neutrophils towards a more anti-inflammatory phenotype and the release of anti-microbial peptides. This report will review the current knowledge on the effects of MSC treatment in preclinical experimental small animal models of sepsis.
Collapse
|
199
|
Chen G, Jin Y, Shi X, Qiu Y, Zhang Y, Cheng M, Wang X, Chen C, Wu Y, Jiang F, Li L, Zhou H, Fu Q, Liu X. Adipose-derived stem cell-based treatment for acute liver failure. Stem Cell Res Ther 2015; 6:40. [PMID: 25890008 PMCID: PMC4425851 DOI: 10.1186/s13287-015-0040-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 11/14/2014] [Accepted: 03/05/2015] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Acute liver failure (ALF) is a highly lethal disease, for which effective therapeutic methods are limited. Although allogeneic liver transplantation is a viable treatment method for ALF, there is a serious shortage of liver donors. Recent studies suggest that stem cell transplantation is a more promising alternative. Hence, we investigate whether human adipose-derived stem cells (ASCs) have the therapeutic potential for ALF in this study based on the studies of rat models. METHODS Sprague Dawley rats were used to establish ALF models by D-galactosamine injection. These rats were randomly divided into a human ASC-treated group and a phosphate-buffered saline (PBS) control group. The human ASCs or PBS was transplanted through the spleen of rats. The indices of hepatic function and hepatic histology were dynamically detected, and the survival rates of rats were also counted. Double-fluorescence immunohistochemistry was employed to detect the ASC fate after transplantation. Moreover, both concentrated ASC conditional media and ASC lysates were transplanted through the femoral vain of rats to investigate the therapeutic potential for ALF. RESULTS The ASC transplantation group showed improved viability in comparison with the sham control. Histological and biochemical analysis suggested that liver morphology and function were improved in terms of cell proliferation and apoptosis. Although a plethora of ASCs persist in the spleen, the improvement in liver function was obvious. However, ASCs did not differentiate into hepatocytes after engrafting to livers within 3 days. In addition, both concentrated serum-free ASC conditional media and ASC lysates, characterized by high levels of hepatocyte growth factor and vascular endothelial growth factor, demonstrated obvious improvement in terms of high survival rates of ALF rats. CONCLUSION Our data suggest that ASC transplantation has the potential for ALF treatment partly by the mechanism of secreting growth factors contributing to liver regeneration.
Collapse
Affiliation(s)
- Guangfeng Chen
- Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchangzhong Road, Shanghai, 200072, P.R. China.
| | - Yinpeng Jin
- Shanghai Liver Diseases Research Center, The Nanjing Military Command, 9585 Humin Road, Shanghai, 200235, P.R. China.
| | - Xiujuan Shi
- Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchangzhong Road, Shanghai, 200072, P.R. China.
| | - Yu Qiu
- Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchangzhong Road, Shanghai, 200072, P.R. China.
| | - Yushan Zhang
- Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchangzhong Road, Shanghai, 200072, P.R. China.
| | - Mingliang Cheng
- Department of Infectious Diseases, Affiliated Hospital, Guiyang Medical College, 9 Beijing Road, Guiyang, 550004, P.R. China.
| | - Xiaojin Wang
- Shanghai Liver Diseases Research Center, The Nanjing Military Command, 9585 Humin Road, Shanghai, 200235, P.R. China.
| | - Chengwei Chen
- Shanghai Liver Diseases Research Center, The Nanjing Military Command, 9585 Humin Road, Shanghai, 200235, P.R. China.
| | - Yinxia Wu
- Shanghai Liver Diseases Research Center, The Nanjing Military Command, 9585 Humin Road, Shanghai, 200235, P.R. China.
| | - Fuzhu Jiang
- Shanghai Liver Diseases Research Center, The Nanjing Military Command, 9585 Humin Road, Shanghai, 200235, P.R. China.
| | - Li Li
- Shanghai Liver Diseases Research Center, The Nanjing Military Command, 9585 Humin Road, Shanghai, 200235, P.R. China.
| | - Heng Zhou
- Shanghai Liver Diseases Research Center, The Nanjing Military Command, 9585 Humin Road, Shanghai, 200235, P.R. China.
| | - Qingchun Fu
- Shanghai Liver Diseases Research Center, The Nanjing Military Command, 9585 Humin Road, Shanghai, 200235, P.R. China.
| | - Xiaoqing Liu
- Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchangzhong Road, Shanghai, 200072, P.R. China.
| |
Collapse
|
200
|
Remission of collagen-induced arthritis through combination therapy of microfracture and transplantation of thermogel-encapsulated bone marrow mesenchymal stem cells. PLoS One 2015; 10:e0120596. [PMID: 25774788 PMCID: PMC4361318 DOI: 10.1371/journal.pone.0120596] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 01/24/2015] [Indexed: 12/11/2022] Open
Abstract
The persistent inflammation of rheumatoid arthritis (RA) always leads to partial synovial hyperplasia and the destruction of articular cartilage. Bone marrow mesenchymal stem cells (BMMSCs) have been proven to possess immunosuppressive effects, and widely explored in the treatment of autoimmune diseases. However, poor inhibitory effect on local inflammatory state and limited capacity of preventing destruction of articular cartilage by systemic BMMSCs transplantation were observed. Herein, toward the classical type II collagen-induced arthritis in rats, the combination treatment of microfracture and in situ transplantation of thermogel-encapsulated BMMSCs was verified to obviously down-regulate the ratio of CD4+ to CD8+ T lymphocytes in peripheral blood. In addition, it resulted in the decreased levels of inflammatory cytokines, such as interleukin-1β, tumor necrosis factor-α and anti-collagen type II antibody, in the serum. Simultaneously, the combination therapy also could inhibit the proliferation of antigen specific lymphocytes and local joint inflammatory condition, and prevent the articular cartilage damage. The results indicated that the treatment programs could effectively stimulate the endogenous and exogenous BMMSCs to exhibit the immunosuppression and cartilage protection capability. This study provided a new therapeutic strategy for autoimmune inflammatory diseases, such as RA.
Collapse
|