51
|
Silveira GDP, Ishimura ME, Teixeira D, Galindo LT, Sardinha AA, Porcionatto M, Longo-Maugéri IM. Improvement of Mesenchymal Stem Cell Immunomodulatory Properties by Heat-Killed Propionibacterium acnes via TLR2. Front Mol Neurosci 2019; 11:489. [PMID: 30687005 PMCID: PMC6336115 DOI: 10.3389/fnmol.2018.00489] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 12/14/2018] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are an essential tool for regenerative medicine, which aims to develop new technologies to improve their effects to obtain useful transplantation results. MSC immunomodulatory role has been just demonstrated; however, how they react when they are stimulated by an adjuvant is poorly understood. Our group showed the adjuvant effect of killed Propionibacterium acnes (P. acnes) on hematopoietic stem cells. As these cells share the same MSCs bone marrow (BM) site and interact with each other, here we evaluated the P. acnes and its soluble polysaccharide (PS) effect on MSCs and their immunomodulatory role in a murine model of traumatic brain injury (TBI). The bacteria increased the absolute number of MSCs, including MSC subpopulations, and maintained MSC plasticity. P. acnes and PS enhanced MSC proliferation and improved their immunomodulatory effect. P. acnes-MSC and PS-MSC transplantation increased anti-inflammatory cytokine expression and diminished pro-inflammatory cytokine expression after injury. This effect seemed to be mediated via TLR2 since P. acnes-KOTLR2-MSC transplantation decreased TGF-β and IL-10 expression. Increasing in neural stem cells and neuroblasts after PS-MSC transplantation was also observed. The adjuvant effect of P. acnes is an alternative means of expanding MSCs and important to identify their subpopulations to know better their role under exogenous stimuli including inflammation resolution in an experimental model.
Collapse
Affiliation(s)
- Gabriela da Paz Silveira
- Division of Immunology, Department of Microbiology, Immunology and Parasitology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Mayari Eika Ishimura
- Division of Immunology, Department of Microbiology, Immunology and Parasitology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Daniela Teixeira
- Division of Immunology, Department of Microbiology, Immunology and Parasitology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Layla Tesla Galindo
- Division of Molecular Biology, Department of Biochemistry, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Agnes Araujo Sardinha
- Division of Molecular Biology, Department of Biochemistry, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Marimelia Porcionatto
- Division of Molecular Biology, Department of Biochemistry, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Ieda Maria Longo-Maugéri
- Division of Immunology, Department of Microbiology, Immunology and Parasitology, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
52
|
Li J, Chen T, Huang X, Zhao Y, Wang B, Yin Y, Cui Y, Zhao Y, Zhang R, Wang X, Wang Y, Dai J. Substrate-independent immunomodulatory characteristics of mesenchymal stem cells in three-dimensional culture. PLoS One 2018; 13:e0206811. [PMID: 30408051 PMCID: PMC6224081 DOI: 10.1371/journal.pone.0206811] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 10/21/2018] [Indexed: 01/02/2023] Open
Abstract
Mesenchymal stem cells (MSCs) play important roles in tissue regeneration, and multi-lineage differentiation and immunomodulation are two major characteristics of MSCs that are utilized in stem cell therapy. MSCs in vivo have a markedly different three-dimensional (3D) niche compared to the traditional two-dimensional (2D) culture in vitro. A 3D scaffold is predicted to provide an artificial 3D environment similar to the in vivo environment. Significant changes in MSC differentiation are shown to be occurred when under 3D culture. However, the immunomodulatory characteristics of MSCs under 3D culture remain unknown. In this study, 3D culture systems were constructed using different substrates to evaluate the common immunomodulatory characteristics of MSCs. Compared to the MSCs under 2D culture, the MSCs under 3D culture, which had higher stemness and maintained cell phenotype, showed altered immunophenotypic pattern. Gene expression profile analysis at mRNA and protein level detected by gene chip and protein chip, respectively, further revealed the difference between 3D cultured MSCs and 2D cultured MSCs, which was mainly concentrated in the immunoregulation related aspects. Moreover, the immunoregulatory role of 3D culture was confirmed by our immunosuppressive experiments. These findings demonstrated that the immunomodulatory capacities of MSCs were enhanced by the 3D geometry of substrates.
Collapse
Affiliation(s)
- Jing Li
- Laboratory of Translational Medicine, Chinese PLA General Hospital, Beijing, China
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Tong Chen
- University of Chinese Academy of Sciences, Beijing, China
- The State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Xiahe Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Yunshan Zhao
- Institute of General Surgery, Chinese PLA General Hospital, Beijing, China
| | - Bin Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Yanyun Yin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Yi Cui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Ruiping Zhang
- Department of Radiology, First Clinical Medical School of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiujie Wang
- The State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Yingchun Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- * E-mail:
| |
Collapse
|
53
|
Chen Y, Yang XQ, Tseng BY, Tsai YH, Tseng SH, Lee CH, Yao CL. Deferoxamine preconditioning activated hypoxia-inducible factor-1α and MyD88-dependent Toll-like receptor 4 signaling in intestinal stem cells. J Pediatr Surg 2018; 53:2349-2356. [PMID: 29475626 DOI: 10.1016/j.jpedsurg.2018.01.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 01/06/2018] [Accepted: 01/30/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND/PURPOSE Toll-like receptors (TLRs) are important regulators of innate immunity, and TLR4 pathway can regulate the survival, migration, and differentiation of stem cells, including intestinal stem cells (ISCs). Deferoxamine (DFO), a hypoxia-mimic compound, can activate the proliferation of ISCs. In this study, we investigated the response of TLR4 signaling to DFO-induced hypoxia in cultured ISCs in vitro. METHODS After DFO treatment, the crypt organoid number was counted, and the expression levels of Lgr5, Hsp70, HMGB1, HIF-1α, TLR4, MyD88, TRIF, and TRAM in ISCs were examined using QPCR and Western blotting. The chemical inhibitors of different signaling molecules were then used to determine their role in DFO-induced change in ISCs. RESULTS The expression levels of Lgr5, HIF-1α, TLR4, MyD88, and TRIF in ISCs increased after DFO treatment, with peak expression of these molecules 6h after DFO treatment. In addition, DFO-induced gene expression of Lgr5 and HIF-1α was partially reversed by pretreatment with the inhibitor of TLR4 or MyD88, but not TRIF inhibitor. Inhibition of HIF-1α also resulted in partial downregulation of DFO-induced elevation of Lgr5 and TLR4. CONCLUSIONS These results demonstrated that DFO treatment activated HIF-1α and the TLR4-MyD88 signaling pathway, which might mediate the activation of ISCs.
Collapse
Affiliation(s)
- Yun Chen
- Department of Surgery, Far Eastern Memorial Hospital, Pan-Chiao, New Taipei, Taiwan; Department of Chemical Engineering and Materials Science, Yuan Ze University, Chung-Li, Taoyuan, Taiwan
| | - Xiang-Qin Yang
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Chung-Li, Taoyuan, Taiwan
| | - Bor-Yuan Tseng
- Department of Surgery, Far Eastern Memorial Hospital, Pan-Chiao, New Taipei, Taiwan
| | - Ya-Hui Tsai
- Department of Surgery, Far Eastern Memorial Hospital, Pan-Chiao, New Taipei, Taiwan; Department of Chemical Engineering and Materials Science, Yuan Ze University, Chung-Li, Taoyuan, Taiwan; Department of Materials and Textiles, Oriental Institute of Technology, Pan-Chiao, New Taipei, Taiwan
| | - Sheng-Hong Tseng
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Cheng-Hung Lee
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Chung-Li, Taoyuan, Taiwan; Department of General Surgery, Buddhist Dalin Tzu Chi Hospital, Chia-Yi, Taiwan; School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Chao-Ling Yao
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Chung-Li, Taoyuan, Taiwan; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Chung-Li, Taoyuan, Taiwan.
| |
Collapse
|
54
|
Golovynska I, Kalmukova O, Svitina HM, Kyryk VM, Shablii VA, Senchylo NV, Ostrovska GV, Dzerzhinskyi M, Stepanov YV, Golovynskyi S, Ohulchanskyy TY, Liu L, Garmanchuk LV, Qu J. Morpho-Functional Characteristics of Bone Marrow Multipotent Mesenchymal Stromal Cells after Activation or Inhibition of Epidermal Growth Factor and Toll-Like Receptors or Treatment with DNA Intercalator Cisplatin. Cytometry A 2018; 95:24-33. [PMID: 30240134 DOI: 10.1002/cyto.a.23593] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 07/25/2018] [Accepted: 07/30/2018] [Indexed: 12/11/2022]
Abstract
This study is aimed to reveal morphological and functional changes in multipotent mesenchymal stromal cells (MSCs) isolated from the rat bone marrow after: (i) activation of Toll-like receptors (TLRs) with teichoic acid (TA), (ii) impact on epidermal growth factor (EGF) receptors with activator EGF or inhibitor Herceptin, and (iii) treatment with DNA intercalator Cisplatin. According to our results, TA and EGF cause an increase in the synthesis of glycosaminoglycans, c-Myc content, and protein in the MSC cytoplasm. It was observed that the cell population in G0 phase decreased and the cell population in G1 phase increased, when compared with control. At the same time, the cell population with a higher nuclear-cytoplasmic ratio (NCR) in S and G2 phases also increased. This indicates the manifestation of the MSC mesenchymal phenotype, exhibiting indirect metabolic signs of the regenerative potential increase. In other experiments, Herceptin was shown to suppress only the stemness signs of MSCs, while Cisplatin seriously affected cell viability in general, reducing synthetic and proliferative activities and causing cell morphology disturbances. © 2018 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Iuliia Golovynska
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, 518060, Shenzhen, People's Republic of China
| | - Olesia Kalmukova
- ESC Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, 01601, Kyiv, Ukraine.,State Institute of Genetic and Regenerative Medicine, National Academy of Medical Sciences of Ukraine, 04114, Kyiv, Ukraine
| | - Hanna M Svitina
- Pharmacen, Centre of Excellence for Pharmaceutical Sciences, North-West University, Private Bag X6001, Potchefstroom, 2520, South Africa
| | - Vitaliy M Kyryk
- State Institute of Genetic and Regenerative Medicine, National Academy of Medical Sciences of Ukraine, 04114, Kyiv, Ukraine
| | | | - Nataliya V Senchylo
- ESC Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, 01601, Kyiv, Ukraine
| | - Galyna V Ostrovska
- ESC Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, 01601, Kyiv, Ukraine
| | - Mykola Dzerzhinskyi
- ESC Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, 01601, Kyiv, Ukraine
| | - Yurii V Stepanov
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, 518060, Shenzhen, People's Republic of China
| | - Sergii Golovynskyi
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, 518060, Shenzhen, People's Republic of China
| | - Tymish Y Ohulchanskyy
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, 518060, Shenzhen, People's Republic of China
| | - Liwei Liu
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, 518060, Shenzhen, People's Republic of China
| | - Liudmila V Garmanchuk
- ESC Institute of Biology and Medicine, Taras Shevchenko National University of Kyiv, 01601, Kyiv, Ukraine
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, 518060, Shenzhen, People's Republic of China
| |
Collapse
|
55
|
Ruhl T, Kim BS, Beier JP. Cannabidiol restores differentiation capacity of LPS exposed adipose tissue mesenchymal stromal cells. Exp Cell Res 2018; 370:653-662. [DOI: 10.1016/j.yexcr.2018.07.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 07/17/2018] [Accepted: 07/19/2018] [Indexed: 02/01/2023]
|
56
|
Hashemzadeh MR, Eslaminejad MB, Salman Yazdi R, Aflatoonian R. Evaluation of toll-like receptor 4 expression in human bone marrow mesenchymal stem cells by lipopolysaccharides from Shigella. Biologicals 2018; 55:53-58. [PMID: 30042006 DOI: 10.1016/j.biologicals.2018.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 06/27/2018] [Indexed: 10/28/2022] Open
Abstract
Lipopolysaccharides (LPS) from gram negative bacteria stimulate toll-like receptor 4 (TLR4) expression in immune cells. Recent reports state that bone marrow-derived cells such as mesenchymal stem cells (MSCs) also express TLR proteins. Numerous researches have studied the effect of a number of LPSs on TLR4 expression, but no data exists on the effect of LPSs from different strains of one bacterial genus on TLR4 expression. In this study, we investigate the effects of various concentrations of LPS from different Shigella strains on TLR4 expression in human bone marrow (hBM)-MSCs. At the mRNA level, we have found that untreated hBM-MSCs (control) did not express TLR4 compared to the experimental groups. Cells treated with LPS from Shigella flexneri had the highest expression of TLR4, whereas cells treated with LPS from Shigella sonnei had the lowest expression. We observed that LPSs had a dose-dependent effect on TLR4 expression in all of the treatment groups. ELISA findings for interleukin-6 secretion have confirmed mRNA expression results for all treatment groups. Hence, LPS from S. flexneri can be considered as an optimum LPS to stimulate the immune system for vaccine production against shigellosis. Also, TLR activation in hBM-MSCs can modulate their function such as homing.
Collapse
Affiliation(s)
- Mohammad Reza Hashemzadeh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Reza Salman Yazdi
- Department of Andrology, Reproductidve Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Reza Aflatoonian
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.
| |
Collapse
|
57
|
Alimoradi E, Sisakhtnezhad S, Akrami H. Thymoquinone influences the expression of genes involved in self-renewal and immunomodulatory potential of mouse bone marrow-derived mesenchymal stem cells in vitro. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 60:216-224. [PMID: 29763882 DOI: 10.1016/j.etap.2018.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 05/04/2018] [Accepted: 05/06/2018] [Indexed: 06/08/2023]
Abstract
Thymoquinone (TQ) is an active ingredient of some medicinal herbs. Despite extensive studies on the biological and pharmacological properties of TQ, its effect on the characteristics of stem cells remains to be clarified. Therefore, this study was aimed to investigate the effect of TQ on viability, proliferation and immunomodulatory potential of mouse bone marrow-derived mesenchymal stem cells (BM-MSCs) in vitro. The BM-MSCs were isolated from young NMRI mice. The cytotoxic effect of TQ on the BM-MSCs was evaluated using MTT assay. Then, the effect of TQ on the proliferation of BM-MSCs and the mRNA expression of genes involved in self-renewal and immunomodulatory potential of MSCs was assessed by the cell counting and real-time PCR assays. Results showed that TQ reduces the number of BM-MSCs in a dose- and time-dependent manner. In addition, the half-maximal inhibitory concentration values of TQ on the BM-MSCs were 8 μg/ml at 24h and 4 μg/ml at 48 and 72h after treatment. Furthermore, about 90% of the BM-MSCs were alive after treatment with concentrations ≤2 μg/ml of TQ for 24h. The results of cell counting assay indicated that TQ at concentrations of 1-2 μg/ml significantly enhanced the proliferation of BM-MSCs (P < 0.05). The gene expression analysis also showed that Tlr3, Tlr4, Ccl2, Ccl3, Sox2, and Rex1 are overexpressed (Fold change ≥1.5) in the TQ-treated BM-MSCs compared with the untreated samples. In conclusion, these findings propose that TQ may regulate self-renewal and immunomodulatory potential of MSCs. However, the exact mechanisms and the roles of this regulation are required to be elucidated in further study.
Collapse
Affiliation(s)
- Elham Alimoradi
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | | | - Hassan Akrami
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| |
Collapse
|
58
|
Wu X, Xu L, Shen Y, Yu N, Zhang Y, Guo T. MALP-2, an agonist of TLR6, promotes the immune status without affecting the differentiation capacity of umbilical cord mesenchymal stem cells. Exp Ther Med 2017; 14:5540-5546. [PMID: 29285089 DOI: 10.3892/etm.2017.5262] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 04/07/2017] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are increasingly used in cell-based therapy due to their multiple differentiation capacity, low expression of co-stimulatory factors and immunosuppressive effect. However, accumulating studies reported the recognition and rejection of engrafted MSCs, which eventually led to the fail of clinical trials. Toll-like receptors (TLRs) are important in mediating the immune response. In the present study, macrophage-activated lipopeptide-2 (MALP-2) was introduced to activate the TLR6 pathway in umbilical cord MSCs (UCMSCs). PBLs isolated from healthy volunteers were co-cultured with UCMSCs to measure whether activation of TLR6 of UCMSCs could stimulate immune responses. Reverse transcription-quantitative polymerase chain reaction and immunohistochemistry were performed to detect pro-inflammatory molecules and differentiation status of UCMSCs, respectively. The results indicated that activation of TLR6 in UCMSCs increased the proliferation of peripheral blood leukocytes (PBLs) and enhanced the release of lactate dehydrogenase in damaged UCMSCs, which confirmed the role of TLR6 in promoting the immunogenicity of UCMSCs. Furthermore, quantitative polymerase chain reaction demonstrated that the expression of proinflammatory molecules (including IL-1β, IL-6, IL-8, IL-10, CCL1 and CCL4) was induced, whereas the expression of stem cell markers (Klf4 and Nanog) was inhibited. The differentiation results indicated that activation of TLR6 had no effect on the differentiation capacity of UCMSCs. All these findings suggest that stimulation of TLR6 pathway may increase the immunogenicity of UCMSCs in in vitro detections. In conclusion, the results of the current study indicated a new role of TLR6 in regulating the biological function of UCMSCs.
Collapse
Affiliation(s)
- Xiuli Wu
- Department of Laboratory Medicine, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Beijing 100816, P.R. China
| | - Lian Xu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Beijing 100816, P.R. China.,Department of Pathology, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yangmei Shen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Beijing 100816, P.R. China.,Department of Pathology, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Na Yu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Beijing 100816, P.R. China.,Department of Pathology, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yan Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Beijing 100816, P.R. China.,Department of Pathology, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Tao Guo
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Beijing 100816, P.R. China.,Department of Gynecology and Obstetrics, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
59
|
Casiraghi F, Perico N, Remuzzi G. Mesenchymal stromal cells for tolerance induction in organ transplantation. Hum Immunol 2017; 79:304-313. [PMID: 29288697 DOI: 10.1016/j.humimm.2017.12.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 12/06/2017] [Accepted: 12/18/2017] [Indexed: 12/20/2022]
Abstract
The primary challenge in organ transplantation continues to be the need to suppress the host immune system long-term to ensure prolonged allograft survival. Long-term non-specific immunosuppression can, however, result in life-threatening complications. Thus, efforts have been pursued to explore novel strategies that would allow minimization of maintenance immunosuppression, eventually leading to transplant tolerance. In this scenario, bone marrow-derived mesenchymal stromal cells (MSC), given their unique immunomodulatory properties to skew the balance between regulatory and memory T cells, have emerged as potential candidates for cell-based therapy to promote immune tolerance. Here, we review our initial clinical experience with bone marrow-derived MSC in living-donor kidney transplant recipients and provide an overview of the available results of other clinical programs with MSC in kidney and liver transplantation, highlighting hurdles and success of this innovative cell-based therapy.
Collapse
Affiliation(s)
| | - Norberto Perico
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy
| | - Giuseppe Remuzzi
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy; Unit of Nephrology and Dialysis, Azienda Socio Sanitaria Territoriale (ASST), Papa Giovanni XXIII, Bergamo, Italy; L. Sacco Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy.
| |
Collapse
|
60
|
Puddighinu G, D'Amario D, Foglio E, Manchi M, Siracusano A, Pontemezzo E, Cordella M, Facchiano F, Pellegrini L, Mangoni A, Tafani M, Crea F, Germani A, Russo MA, Limana F. Molecular mechanisms of cardioprotective effects mediated by transplanted cardiac ckit + cells through the activation of an inflammatory hypoxia-dependent reparative response. Oncotarget 2017; 9:937-957. [PMID: 29416668 PMCID: PMC5787525 DOI: 10.18632/oncotarget.22946] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 11/12/2017] [Indexed: 12/16/2022] Open
Abstract
The regenerative effects of cardiac ckit+ stem cells (ckit+CSCs) in acute myocardial infarction (MI) have been studied extensively, but how these cells exert a protective effect on cardiomyocytes is not well known. Growing evidences suggest that in adult stem cells injury triggers inflammatory signaling pathways which control tissue repair and regeneration. Aim of the present study was to determine the mechanisms underlying the cardioprotective effects of ckit+CSCs following transplantation in a murine model of MI. Following isolation and in vitro expansion, cardiac ckit+CSCs were subjected to normoxic and hypoxic conditions and assessed at different time points. These cells adapted to hypoxia as showed by the activation of HIF-1α and the expression of a number of genes, such as VEGF, GLUT1, EPO, HKII and, importantly, of alarmin receptors, such as RAGE, P2X7R, TLR2 and TLR4. Activation of these receptors determined an NFkB-dependent inflammatory and reparative gene response (IRR). Importantly, hypoxic ckit+CSCs increased the secretion of the survival growth factors IGF-1 and HGF. To verify whether activation of the IRR in a hypoxic microenvironment could exert a beneficial effect in vivo, autologous ckit+CSCs were transplanted into mouse heart following MI. Interestingly, transplantation of ckit+CSCs lowered apoptotic rates and induced autophagy in the peri-infarct area; further, it reduced hypertrophy and fibrosis and, most importantly, improved cardiac function. ckit+CSCs are able to adapt to a hypoxic environment and activate an inflammatory and reparative response that could account, at least in part, for a protective effect on stressed cardiomyocytes following transplantation in the infarcted heart.
Collapse
Affiliation(s)
- Giovanni Puddighinu
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Domenico D'Amario
- Department of Cardiovascular Sciences, Catholic University of The Sacred Heart, Rome, Italy
| | - Eleonora Foglio
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Melissa Manchi
- Department of Cardiovascular Sciences, Catholic University of The Sacred Heart, Rome, Italy
| | - Andrea Siracusano
- Department of Cardiovascular Sciences, Catholic University of The Sacred Heart, Rome, Italy
| | - Elena Pontemezzo
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Martina Cordella
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Francesco Facchiano
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Laura Pellegrini
- Department of Neurorehabilitation Sciences, Casa Cura Policlinico (CCP), Milan, Italy
| | - Antonella Mangoni
- Department of Pathological Anatomy, Catholic University of The Sacred Heart, Rome, Italy
| | - Marco Tafani
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Filippo Crea
- Department of Cardiovascular Sciences, Catholic University of The Sacred Heart, Rome, Italy
| | - Antonia Germani
- Laboratory of Vascular Pathology, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Fondazione Luigi Maria Monti, Rome, Italy
| | - Matteo Antonio Russo
- IRCCS San Raffaele Pisana, Rome, Italy.,MEBIC Consortium, San Raffaele Roma Open University, Rome, Italy
| | - Federica Limana
- IRCCS San Raffaele Pisana, Rome, Italy.,San Raffaele Roma Open University, Rome, Italy
| |
Collapse
|
61
|
Zhang B, Zhang J, Shi H, Mao F, Wang J, Yan Y, Zhang X, Qian H, Xu W. A novel method to isolate mesenchymal stem cells from mouse umbilical cord. Mol Med Rep 2017; 17:861-869. [PMID: 29115623 PMCID: PMC5780165 DOI: 10.3892/mmr.2017.7950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 05/11/2017] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stem cells (MSCs), derived from various tissues, are considered an ideal cell source for clinical use, among which MSCs from the umbilical cord exhibit advantages over those from adult tissues. In preclinical studies, mouse models and xenogeneic MSC treatment are most commonly used to imitate diseases and clinical practice, respectively. However, the efficiency of cross-species therapy remains controversial, making it difficult to elucidate the underlying mechanisms. Thus, allogeneic therapy may be more instructive and meaningful in clinical use. To confirm this hypothesis, the present study established a novel method for the isolation and expansion of MSCs from mouse umbilical cords (mUC-MSCs) to support in vivo experiments in mice. MSCs were isolated from mUCs and mouse bone marrow (mBM), and then identified by flow cytometry. The differences in mUC-MSCs and mBM-MSCs were analyzed using a growth curve and their differentiation ability. The results showed that the harvested cells exhibited general characteristics of MSCs and possessed the capacity for long-term culture. Despite having similar morphology and surface antigens to MSCs derived from mouse bone marrow, the mUC-MSCs showed differences in purification, proliferation, stem cell markers and differentiation. In addition to detailed characterization, the present study verified the presence of Toll-like receptor 3 (TLR3), an important component of immune responses, in mUC-MSCs. It was found that the activation of TLR3 upregulated the levels of stemness-related proteins, and enhanced the secretion and mRNA levels of inflammatory cytokines in the pre-treated mUC-MSCs. Collectively, the results of the present study provide further insight into the features of newly established mUC-MSCs, providing novel evidence for the selection of murine MSCs and their responses to TLR3 priming.
Collapse
Affiliation(s)
- Bin Zhang
- Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Jie Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Hui Shi
- Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Juanjuan Wang
- Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yongmin Yan
- Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Xu Zhang
- Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Hui Qian
- Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Wenrong Xu
- Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
62
|
The Immunomodulatory Effects of Mesenchymal Stem Cell Polarization within the Tumor Microenvironment Niche. Stem Cells Int 2017; 2017:4015039. [PMID: 29181035 PMCID: PMC5664329 DOI: 10.1155/2017/4015039] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 07/11/2017] [Accepted: 07/16/2017] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) represent a promising tool for cell therapy, particularly for their antitumor effects. This cell population can be isolated from multiple tissue sources and also display an innate ability to home to areas of inflammation, such as tumors. Upon entry into the tumor microenvironment niche, MSCs promote or inhibit tumor progression by various mechanisms, largely through the release of soluble factors. These factors can be immunomodulatory by activating or inhibiting both the adaptive and innate immune responses. The mechanisms by which MSCs modulate the immune response are not well understood. Because of this, the relationship between MSCs and immune cells within the tumor microenvironment niche continues to be an active area of research in order to help explain the apparent contradictory findings currently available in the literature. The ongoing research aims to enhance the potential of MSCs in future therapeutic applications.
Collapse
|
63
|
Sangiorgi B, De Freitas HT, Schiavinato JLDS, Leão V, Haddad R, Orellana MD, Faça VM, Ferreira GA, Covas DT, Zago MA, Panepucci RA. DSP30 enhances the immunosuppressive properties of mesenchymal stromal cells and protects their suppressive potential from lipopolysaccharide effects: A potential role of adenosine. Cytotherapy 2017; 18:846-59. [PMID: 27260206 DOI: 10.1016/j.jcyt.2016.04.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 04/06/2016] [Accepted: 04/13/2016] [Indexed: 01/09/2023]
Abstract
Multipotent mesenchymal stromal cells (MSC) are imbued with an immunosuppressive phenotype that extends to several immune system cells. In this study, we evaluated how distinct Toll-like receptor (TLR) agonists impact immunosuppressive properties of bone marrow (BM)-MSC and explored the potential mechanisms involved. We show that TLR4 stimulation by lipopolysaccharide (LPS) restricted the ability of MSC to suppress the proliferation of T lymphocytes, increasing the gene expression of interleukin (IL)-1β and IL-6. In contrast, stimulation of TLR9 by DSP30 induced proliferation and the suppressive potential of BM-MSC, coinciding with reducing tumor necrosis factor (TNF)-α expression, increased expression of transforming growth factor (TGF)-β1, increased percentages of BM-MSC double positive for the ectonucleotidases CD39+CD73+ and adenosine levels. Importantly, following simultaneous stimulation with LPS and DSP30, BM-MSC's ability to suppress T lymphocyte proliferation was comparable with that of non-stimulated BM-MSC levels. Moreover, stimulation of BM-MSC with LPS reduced significantly the gene expression levels, on co-cultured T lymphocyte, of IL-10 and interferon (IFN)γ, a cytokine with potential to enhance the immunosuppression mediated by MSC and ameliorate the clinical outcome of patients with graft-versus-host disease (GVHD). Altogether, our findings reiterate the harmful effects of LPS on MSC immunosuppression, besides indicating that DSP30 could provide a protective effect against LPS circulating in the blood of GVHD patients who receive BM-MSC infusions, ensuring a more predictable immunosuppressive effect. The novel effects and potential mechanisms following the stimulation of BM-MSC by DSP30 might impact their clinical use, by allowing the derivation of optimal "licensing" protocols for obtaining therapeutically efficient MSC.
Collapse
Affiliation(s)
- Bruno Sangiorgi
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, São Paulo, Brazil; Centro de Terapia Celular, Fundação Hemocentro de Ribeirão Preto, São Paulo, Brazil
| | | | - Josiane Lilian Dos Santos Schiavinato
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, São Paulo, Brazil; Centro de Terapia Celular, Fundação Hemocentro de Ribeirão Preto, São Paulo, Brazil
| | - Vitor Leão
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, São Paulo, Brazil; Centro de Terapia Celular, Fundação Hemocentro de Ribeirão Preto, São Paulo, Brazil
| | - Rodrigo Haddad
- Faculdade de Ceilândia, Universidade de Brasília, Brasília, Brazil
| | | | - Vitor Marcel Faça
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, São Paulo, Brazil
| | | | - Dimas Tadeu Covas
- Centro de Terapia Celular, Fundação Hemocentro de Ribeirão Preto, São Paulo, Brazil
| | - Marco Antônio Zago
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, São Paulo, Brazil; Centro de Terapia Celular, Fundação Hemocentro de Ribeirão Preto, São Paulo, Brazil
| | - Rodrigo Alexandre Panepucci
- Universidade de São Paulo, Faculdade de Medicina de Ribeirão Preto, São Paulo, Brazil; Centro de Terapia Celular, Fundação Hemocentro de Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
64
|
Gorskaya YF, Tukhvatulin AI, Nesterenko VG. NLR2 and TLR3, TLR4, TLR5 Ligands, Injected In Vivo, Improve after 1 h the Efficiency of Cloning and Proliferative Activity of Bone Marrow Multipotent Stromal Cells and Reduce the Content of Osteogenic Multipotent Stromal Cells in CBA Mice. Bull Exp Biol Med 2017; 163:356-360. [PMID: 28744636 DOI: 10.1007/s10517-017-3803-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Indexed: 11/26/2022]
Abstract
Ligands NLR2 (muramyldipeptide) and TLR (bacterial LPS, flagellin, CpG-dinucleotide, and Poly I:C) and S. typhimurium antigenic complex by 1.5-3-fold increase the efficiency of cloning and content of multipotent stromal cells (MSC) in the bone marrow of CBA mice as soon as 1 h postinjection. The counts of large colonies (150-500 cells) increased by 2.5-3.3 times in comparison with intact bone marrow cultures at the expense of a decrease in the number of smaller colonies, which attests to enhanced proliferation of stromal cells in the colonies. The efficiency of cloning and hence, MSC content in the femoral bone decreased by 1.2-1.9 times after 3 h and increased again after 24 h to the level 1.3-1.5 times higher than the level 1 h postinjection (LPS, Poly I:C, and S. typhimurium antigenic complex). The dynamics of bone marrow MSC cloning efficiency after 1-3 h corresponded to the dynamics of serum cytokine concentrations during the same period. However, the levels of serum cytokines after 24 h in general were similar to those in intact mice or were lower. The concentrations of osteogenic multipotent stromal cells in the bone marrow decreased 2-3-fold after 3 h and thus persisted by 24 h postinjection. Twofold (at 24-h interval) and a single injection of S. typhimurium antigenic complex to mice led to a significant increase of cloning efficiency, observed as early as just 1 h postinjection (1.9 and 1.5 times, respectively). The same picture was observed for serum cytokines. On the whole, injections of TLR and NLR ligands and of S. typhimurium antigenic complex led to stromal tissue activation within 1 h postinjection, this activation consisting in a significant increase of the efficiency of cloning and of MSC count in the bone marrow, and also in an increase in their proliferative activity and a decrease (after 3 h) of osteogenic MSC concentration.
Collapse
Affiliation(s)
- Yu F Gorskaya
- Laboratory of Immunity Regulation and Immunological Tolerance, Moscow, Russia.
| | - A I Tukhvatulin
- Laboratory of Cellular Microbiology, N. F. Gamaleya Federal Research Centre of Epidemiology and Microbiology, the Ministry of Health of the Russian Federation, Moscow, Russia
| | - V G Nesterenko
- Laboratory of Immunity Regulation and Immunological Tolerance, Moscow, Russia
| |
Collapse
|
65
|
Guo S, Kang J, Ji B, Guo W, Ding Y, Wu Y, Tian W. Periodontal-Derived Mesenchymal Cell Sheets Promote Periodontal Regeneration in Inflammatory Microenvironment. Tissue Eng Part A 2017; 23:585-596. [PMID: 28437177 DOI: 10.1089/ten.tea.2016.0334] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- Shujuan Guo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Engineering Laboratory for Oral Regenerative Medicine, Sichuan University, Chengdu, P.R. China
- Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Jian Kang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Engineering Laboratory for Oral Regenerative Medicine, Sichuan University, Chengdu, P.R. China
- Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
- Department of Periodontics, Tianjin Stomatological Hospital, Tianjin, P.R. China
| | - Baohui Ji
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Engineering Laboratory for Oral Regenerative Medicine, Sichuan University, Chengdu, P.R. China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Weihua Guo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Engineering Laboratory for Oral Regenerative Medicine, Sichuan University, Chengdu, P.R. China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Yi Ding
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Yafei Wu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| | - Weidong Tian
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Engineering Laboratory for Oral Regenerative Medicine, Sichuan University, Chengdu, P.R. China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R. China
| |
Collapse
|
66
|
Yang Y, Wang Y, Li L, Chen F, Zhang P. Activation of the Toll-like receptor 8 pathway increases the immunogenicity of mesenchymal stem cells from umbilical cord. Mol Med Rep 2017; 16:2061-2068. [PMID: 28656222 DOI: 10.3892/mmr.2017.6806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 03/21/2017] [Indexed: 02/05/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are now widely used in clinical cell‑based therapy due to their characteristics of low immunogenicity, multiple differentiation potency and the capability to modulate immune responses. However, accumulated research has indicated the absence of engrafted MSCs because of the increased immunogenicity of MSCs. Toll‑like receptors (TLRs) are essential for the innate immune response and regulating the biological function of MSCs. The present study used human umbilical cord‑derived MSCs (UCMSCs) and activated the TLR8 pathway of UCMSCs to study the role of TLR8 in mediating the immune status of UCMSCs. The results demonstrated that the activation of TLR8 increased both the proliferation of peripheral blood mononuclear cells (PBMCs) isolated from healthy human volunteers and the release of lactate dehydrogenase (LDH) in supernatant from the PBMC‑UCMSCs co‑culture system. Reverse transcription-quantitative polymerase chain reaction indicated that the TLR8 agonist increased the expression of many co‑stimulatory molecules and pro‑inflammatory genes, and flow cytometry indicated that activation of the TLR8 agonist increased co‑stimulation protein levels but reduced specific surface markers, as confirmed by the part loss of stemness of UCMSCs. Finally, TLR8 increased osteocyte differentiation but had no effect on chondrocyte and adipocyte differentiation. The current study indicated the implication to TLR8 as regulators of the immunogenicity of UCMSCs.
Collapse
Affiliation(s)
- Yu Yang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yanwen Wang
- Laboratory of Pathology, Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Li Li
- Laboratory of Pathology, Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Fei Chen
- Laboratory of Pathology, Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Peng Zhang
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
67
|
Carbon Monoxide Improves Efficacy of Mesenchymal Stromal Cells During Sepsis by Production of Specialized Proresolving Lipid Mediators. Crit Care Med 2017; 44:e1236-e1245. [PMID: 27513357 PMCID: PMC5113254 DOI: 10.1097/ccm.0000000000001999] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Supplemental Digital Content is available in the text. Objectives: Mesenchymal stromal cells are being investigated as a cell-based therapy for a number of disease processes, with promising results in animal models of systemic inflammation and sepsis. Studies are ongoing to determine ways to further improve the therapeutic potential of mesenchymal stromal cells. A gas molecule that improves outcome in experimental sepsis is carbon monoxide. We hypothesized that preconditioning of mesenchymal stromal cells with carbon monoxide ex vivo would promote further therapeutic benefit when cells are administered in vivo after the onset of polymicrobial sepsis in mice. Design: Animal study and primary cell culture. Setting: Laboratory investigation. Subjects: BALB/c mice. Interventions: Polymicrobial sepsis was induced by cecal ligation and puncture. Mesenchymal stromal cells, mesenchymal stromal cells-conditioned with carbon monoxide, fibroblasts, or fibroblasts-conditioned with carbon monoxide were delivered by tail vein injections to septic mice. The mice were assessed for survival, bacterial clearance, and the inflammatory response during sepsis in each of the groups. Mesenchymal stromal cells were also assessed for their ability to promote bacterial phagocytosis by neutrophils, the production of specialized proresolving lipid mediators, and their importance for mesenchymal stromal cells function using gene silencing. Measurements and Main Results: Ex vivo preconditioning with carbon monoxide allowed mesenchymal stromal cells to be administered later after the onset of sepsis (6 hr), and yet maintain their therapeutic effect with increased survival. Carbon monoxide preconditioned mesenchymal stromal cells were also able to alleviate organ injury, improve bacterial clearance, and promote the resolution of inflammation. Mesenchymal stromal cells exposed to carbon monoxide, with docosahexaenoic acid substrate, produced specialized proresolving lipid mediators, particularly D-series resolvins, which promoted survival. Silencing of lipoxygenase pathways (5-lipoxygenase and 12/15-lipoxygenase), which are important enzymes for specialized proresolving lipid mediator biosynthesis, resulted in a loss of therapeutic benefit bestowed on mesenchymal stromal cells by carbon monoxide. Conclusions: Taken together, these data suggest that production of specialized proresolving lipid mediators contribute to improved mesenchymal stromal cell efficacy when exposed to carbon monoxide, resulting in an improved therapeutic response during sepsis.
Collapse
|
68
|
Najar M, Krayem M, Meuleman N, Bron D, Lagneaux L. Mesenchymal Stromal Cells and Toll-Like Receptor Priming: A Critical Review. Immune Netw 2017; 17:89-102. [PMID: 28458620 PMCID: PMC5407987 DOI: 10.4110/in.2017.17.2.89] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 02/24/2017] [Accepted: 02/25/2017] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal Stromal Cells (MSCs) are potential cellular candidates for several immunotherapy purposes. Their multilineage potential and immunomodulatory properties make them interesting tools for the treatment of various immunological diseases. However, depending on the local microenvironment, diverse biological functions of MSCs can be modulated. Indeed, during infections such as obtained following TLR-agonist engagement (called as TLR priming), the phenotype, multilineage potential, hematopoietic support and immunomodulatory capacity of MSCs can present critical changes, which could further affect their therapeutic potential. Thus, for appropriate clinical application of MSCs, it is important to well know and understand these effects in particular during infectious episodes and to find the suitable experimental settings to study that. Pre-stimulation of MSCs with a specific TLR ligand may serve as an effective priming step to modulate one of its function to achieve a desired therapeutic issue.
Collapse
Affiliation(s)
- Mehdi Najar
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Belgium
| | - Mohammad Krayem
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, Brussels 1000, Belgium
| | - Nathalie Meuleman
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Belgium
| | - Dominique Bron
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Belgium
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Belgium
| |
Collapse
|
69
|
Naftali-Shani N, Levin-Kotler LP, Palevski D, Amit U, Kain D, Landa N, Hochhauser E, Leor J. Left Ventricular Dysfunction Switches Mesenchymal Stromal Cells Toward an Inflammatory Phenotype and Impairs Their Reparative Properties Via Toll-Like Receptor-4. Circulation 2017; 135:2271-2287. [PMID: 28356441 DOI: 10.1161/circulationaha.116.023527] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 03/17/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Little is known about the potentially unfavorable effects of mesenchymal stromal cell (MSC) activation on the heart. MSCs can respond to tissue injury by anti- or proinflammatory activation. We aimed to study the potential negative interaction between left ventricular dysfunction (LVD) and MSC activation. METHODS We isolated MSCs from cardiac and subcutaneous fat tissues of mice with LVD 28 days after myocardial infarction or sham operation. To evaluate the effect of LVD on MSCs, we characterized cardiac MSCs and subcutaneous MSCs in vitro. Subsequently, we injected MSCs or saline into the infarcted myocardium of mice and evaluated LV remodeling and function 28 days after myocardial infarction. To test the hypothesis that toll-like receptor 4 (TLR4) mediates proinflammatory polarization of MSCs, we characterized cardiac MSCs from TLR4-/- and wild-type (WT) mice after inflammatory stimulation in vitro. Next, we transplanted cardiac MSCs from TLR4-/- and WT male mice into the infarcted myocardium of female WT mice and evaluated infarct size, MSC retention, inflammation, remodeling, and function after 7 days. RESULTS LVD switched cardiac MSCs toward an inflammatory phenotype, with increased secretion of inflammatory cytokines as well as chemokines. The effect of LVD on subcutaneous MSCs was less remarkable. Although transplantation of cardiac MSCs and subcutaneous MSCs from LVD and sham hearts did not improve LV remodeling and function, cardiac MSCs from LVD exacerbated anterior wall thinning 28 days after myocardial infarction. The inflammatory polarization of cardiac MSCs by LVD was mediated by TLR4, as we found less secretion of inflammatory cytokines and higher secretion of anti-inflammatory cytokines from activated cardiac MSCs of TLR4-deficient mice, compared with WT cardiac MSCs. Significantly, TLR4 deficiency preserved the expression of CD47 (don't eat me signal) on cardiac MSCs after both TLR4 stimulation in vitro and transplantation into the infarcted heart. Compared with WT cardiac MSCs and saline, TLR4-/- cardiac MSCs survived in the cardiac tissue and maintained their reparative properties, reduced infarct size, increased scar thickness, and attenuated LV dilatation 7 days after myocardial infarction. CONCLUSIONS The environment of the failing and infarcted myocardium drives resident and transplanted MSCs toward a proinflammatory phenotype and restricts their survival and reparative effects in a mechanism mediated by TLR4.
Collapse
Affiliation(s)
- Nili Naftali-Shani
- From Neufeld Cardiac Research Institute, Sackler Faculty of Medicine, Tel-Aviv University, Israel (N.N.-S., L.-P.L.-K., D.P., U.A., D.K., N.L., J.L.); Tamman Cardiovascular Research Institute, Leviev Heart Center, Sheba Medical Center, Tel-Hashomer, Israel (N.N.-S., L.-P.L.-K., D.P., U.A., D.K., N.L., J.L.); Sheba Center for Regenerative Medicine, Stem Cell and Tissue Engineering, Tel-Hashomer, Israel (N.N.-S., L.-P.L.-K., D.P., U.A., D.K., N.L., J.L.); and Cardiac Research Laboratory, Department of Cardiothoracic Surgery, Felsenstein Medical Research Center, Rabin Medical Center, Tel-Aviv University, Petah Tikva, Israel (E.H.)
| | - La-Paz Levin-Kotler
- From Neufeld Cardiac Research Institute, Sackler Faculty of Medicine, Tel-Aviv University, Israel (N.N.-S., L.-P.L.-K., D.P., U.A., D.K., N.L., J.L.); Tamman Cardiovascular Research Institute, Leviev Heart Center, Sheba Medical Center, Tel-Hashomer, Israel (N.N.-S., L.-P.L.-K., D.P., U.A., D.K., N.L., J.L.); Sheba Center for Regenerative Medicine, Stem Cell and Tissue Engineering, Tel-Hashomer, Israel (N.N.-S., L.-P.L.-K., D.P., U.A., D.K., N.L., J.L.); and Cardiac Research Laboratory, Department of Cardiothoracic Surgery, Felsenstein Medical Research Center, Rabin Medical Center, Tel-Aviv University, Petah Tikva, Israel (E.H.)
| | - Dahlia Palevski
- From Neufeld Cardiac Research Institute, Sackler Faculty of Medicine, Tel-Aviv University, Israel (N.N.-S., L.-P.L.-K., D.P., U.A., D.K., N.L., J.L.); Tamman Cardiovascular Research Institute, Leviev Heart Center, Sheba Medical Center, Tel-Hashomer, Israel (N.N.-S., L.-P.L.-K., D.P., U.A., D.K., N.L., J.L.); Sheba Center for Regenerative Medicine, Stem Cell and Tissue Engineering, Tel-Hashomer, Israel (N.N.-S., L.-P.L.-K., D.P., U.A., D.K., N.L., J.L.); and Cardiac Research Laboratory, Department of Cardiothoracic Surgery, Felsenstein Medical Research Center, Rabin Medical Center, Tel-Aviv University, Petah Tikva, Israel (E.H.)
| | - Uri Amit
- From Neufeld Cardiac Research Institute, Sackler Faculty of Medicine, Tel-Aviv University, Israel (N.N.-S., L.-P.L.-K., D.P., U.A., D.K., N.L., J.L.); Tamman Cardiovascular Research Institute, Leviev Heart Center, Sheba Medical Center, Tel-Hashomer, Israel (N.N.-S., L.-P.L.-K., D.P., U.A., D.K., N.L., J.L.); Sheba Center for Regenerative Medicine, Stem Cell and Tissue Engineering, Tel-Hashomer, Israel (N.N.-S., L.-P.L.-K., D.P., U.A., D.K., N.L., J.L.); and Cardiac Research Laboratory, Department of Cardiothoracic Surgery, Felsenstein Medical Research Center, Rabin Medical Center, Tel-Aviv University, Petah Tikva, Israel (E.H.)
| | - David Kain
- From Neufeld Cardiac Research Institute, Sackler Faculty of Medicine, Tel-Aviv University, Israel (N.N.-S., L.-P.L.-K., D.P., U.A., D.K., N.L., J.L.); Tamman Cardiovascular Research Institute, Leviev Heart Center, Sheba Medical Center, Tel-Hashomer, Israel (N.N.-S., L.-P.L.-K., D.P., U.A., D.K., N.L., J.L.); Sheba Center for Regenerative Medicine, Stem Cell and Tissue Engineering, Tel-Hashomer, Israel (N.N.-S., L.-P.L.-K., D.P., U.A., D.K., N.L., J.L.); and Cardiac Research Laboratory, Department of Cardiothoracic Surgery, Felsenstein Medical Research Center, Rabin Medical Center, Tel-Aviv University, Petah Tikva, Israel (E.H.)
| | - Natalie Landa
- From Neufeld Cardiac Research Institute, Sackler Faculty of Medicine, Tel-Aviv University, Israel (N.N.-S., L.-P.L.-K., D.P., U.A., D.K., N.L., J.L.); Tamman Cardiovascular Research Institute, Leviev Heart Center, Sheba Medical Center, Tel-Hashomer, Israel (N.N.-S., L.-P.L.-K., D.P., U.A., D.K., N.L., J.L.); Sheba Center for Regenerative Medicine, Stem Cell and Tissue Engineering, Tel-Hashomer, Israel (N.N.-S., L.-P.L.-K., D.P., U.A., D.K., N.L., J.L.); and Cardiac Research Laboratory, Department of Cardiothoracic Surgery, Felsenstein Medical Research Center, Rabin Medical Center, Tel-Aviv University, Petah Tikva, Israel (E.H.)
| | - Edith Hochhauser
- From Neufeld Cardiac Research Institute, Sackler Faculty of Medicine, Tel-Aviv University, Israel (N.N.-S., L.-P.L.-K., D.P., U.A., D.K., N.L., J.L.); Tamman Cardiovascular Research Institute, Leviev Heart Center, Sheba Medical Center, Tel-Hashomer, Israel (N.N.-S., L.-P.L.-K., D.P., U.A., D.K., N.L., J.L.); Sheba Center for Regenerative Medicine, Stem Cell and Tissue Engineering, Tel-Hashomer, Israel (N.N.-S., L.-P.L.-K., D.P., U.A., D.K., N.L., J.L.); and Cardiac Research Laboratory, Department of Cardiothoracic Surgery, Felsenstein Medical Research Center, Rabin Medical Center, Tel-Aviv University, Petah Tikva, Israel (E.H.)
| | - Jonathan Leor
- From Neufeld Cardiac Research Institute, Sackler Faculty of Medicine, Tel-Aviv University, Israel (N.N.-S., L.-P.L.-K., D.P., U.A., D.K., N.L., J.L.); Tamman Cardiovascular Research Institute, Leviev Heart Center, Sheba Medical Center, Tel-Hashomer, Israel (N.N.-S., L.-P.L.-K., D.P., U.A., D.K., N.L., J.L.); Sheba Center for Regenerative Medicine, Stem Cell and Tissue Engineering, Tel-Hashomer, Israel (N.N.-S., L.-P.L.-K., D.P., U.A., D.K., N.L., J.L.); and Cardiac Research Laboratory, Department of Cardiothoracic Surgery, Felsenstein Medical Research Center, Rabin Medical Center, Tel-Aviv University, Petah Tikva, Israel (E.H.).
| |
Collapse
|
70
|
Rubessa M, Polkoff K, Bionaz M, Monaco E, Milner DJ, Holllister SJ, Goldwasser MS, Wheeler MB. Use of Pig as a Model for Mesenchymal Stem Cell Therapies for Bone Regeneration. Anim Biotechnol 2017; 28:275-287. [PMID: 28267421 DOI: 10.1080/10495398.2017.1279169] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Bone is a plastic tissue with a large healing capability. However, extensive bone loss due to disease or trauma requires extreme therapy such as bone grafting or tissue-engineering applications. Presently, bone grafting is the gold standard for bone repair, but presents serious limitations including donor site morbidity, rejection, and limited tissue regeneration. The use of stem cells appears to be a means to overcome such limitations. Bone marrow mesenchymal stem cells (BMSC) have been the choice thus far for stem cell therapy for bone regeneration. However, adipose-derived stem cells (ASC) have similar immunophenotype, morphology, multilineage potential, and transcriptome compared to BMSC, and both types have demonstrated extensive osteogenic capacity both in vitro and in vivo in several species. The use of scaffolds in combination with stem cells and growth factors provides a valuable tool for guided bone regeneration, especially for complex anatomic defects. Before translation to human medicine, regenerative strategies must be developed in animal models to improve effectiveness and efficiency. The pig presents as a useful model due to similar macro- and microanatomy and favorable logistics of use. This review examines data that provides strong support for the clinical translation of the pig model for bone regeneration.
Collapse
Key Words
- ASC, adipose-derived stem cells
- BMP, bone morphogenetic protein
- BMSC, bone marrow mesenchymal stem cells
- Bone
- DEG, differentially expressed genes
- FDR, false-discovery rate
- HA, hydroxyapatite
- HA/TCP, hydroxyapatite/tricalcium phosphate
- MRI, magnetic resonance imaging
- MSC, mesenchymal stem cells
- ONFH, osteonecrosis of the femoral head
- PCL, Poly (ϵ-caprolactone)
- PEG, polyethylene glycol
- PLGA, polylactic-coglycolic acid
- TCP, beta tri-calcium phosphate
- USSC, unrestricted somatic stem cell
- scaffolds
- stem cells
- swine
- tissue engineering
Collapse
Affiliation(s)
- Marcello Rubessa
- a University of Illinois at Urbana-Champaign , Urbana , Illinois , USA
| | - Kathryn Polkoff
- a University of Illinois at Urbana-Champaign , Urbana , Illinois , USA
| | | | - Elisa Monaco
- b Oregon State University , Corvallis , Oregon , USA
| | - Derek J Milner
- a University of Illinois at Urbana-Champaign , Urbana , Illinois , USA
| | | | - Michael S Goldwasser
- a University of Illinois at Urbana-Champaign , Urbana , Illinois , USA.,d New Hanover Regional Medical Center , Wilmington , North Carolina , USA
| | - Matthew B Wheeler
- a University of Illinois at Urbana-Champaign , Urbana , Illinois , USA
| |
Collapse
|
71
|
Pajarinen J, Lin TH, Nabeshima A, Jämsen E, Lu L, Nathan K, Yao Z, Goodman SB. Mesenchymal stem cells in the aseptic loosening of total joint replacements. J Biomed Mater Res A 2017; 105:1195-1207. [PMID: 27977880 DOI: 10.1002/jbm.a.35978] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 12/06/2016] [Indexed: 02/06/2023]
Abstract
Peri-prosthetic osteolysis remains as the main long-term complication of total joint replacement surgery. Research over four decades has established implant wear as the main culprit for chronic inflammation in the peri-implant tissues and macrophages as the key cells mediating the host reaction to implant-derived wear particles. Wear debris activated macrophages secrete inflammatory mediators that stimulate bone resorbing osteoclasts; thus bone loss in the peri-implant tissues is increased. However, the balance of bone turnover is not only dictated by osteoclast-mediated bone resorption but also by the formation of new bone by osteoblasts; under physiological conditions these two processes are tightly coupled. Increasing interest has been placed on the effects of wear debris on the cells of the bone-forming lineage. These cells are derived primarily from multipotent mesenchymal stem cells (MSCs) residing in bone marrow and the walls of the microvasculature. Accumulating evidence indicates that wear debris significantly impairs MSC-to-osteoblast differentiation and subsequent bone formation. In this review, we summarize the current understanding of the effects of biomaterial implant wear debris on MSCs. Emerging treatment options to improve initial implant integration and treat developing osteolytic lesions by utilizing or targeting MSCs are also discussed. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 1195-1207, 2017.
Collapse
Affiliation(s)
- Jukka Pajarinen
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| | - Tzu-Hua Lin
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| | - Akira Nabeshima
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| | - Eemeli Jämsen
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California.,Department of Medicine, Clinicum, University of Helsinki, and Helsinki University Hospital, Helsinki, Finland
| | - Laura Lu
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| | - Karthik Nathan
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| | - Zhenyu Yao
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| | - Stuart B Goodman
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
72
|
Shirjang S, Mansoori B, Solali S, Hagh MF, Shamsasenjan K. Toll-like receptors as a key regulator of mesenchymal stem cell function: An up-to-date review. Cell Immunol 2016; 315:1-10. [PMID: 28284487 DOI: 10.1016/j.cellimm.2016.12.005] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 12/07/2016] [Accepted: 12/24/2016] [Indexed: 02/07/2023]
Abstract
Understanding the role of toll-like receptors (TLRs) in the immunomodulation potential, differentiation, migration, and survival of mesenchymal stem cells (MSCs) is absolutely vital to fully exploiting their MSC-based therapeutic potential. Furthermore, through recognition of exogenous or endogenous ligands produced upon injury, TLRs have been linked to allograft rejection and maintenance of chronic inflammatory diseases, including Crohn's disease, rheumatoid arthritis. Characterizing the effect of TLRs in biological control of MSCs fate and function could improve our knowledge about the MSC-based cell therapy and immunotherapy. In this paper, we outline the impacts of TLR activation and mechanisms on MSCs immunomodulatory functions, differentiation, migration, and survivability. Moreover, we indicate that the expression patterns of TLRs in MSCs from different sources.
Collapse
Affiliation(s)
- Solmaz Shirjang
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Solali
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Karim Shamsasenjan
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
73
|
Ryu DB, Lim JY, Lee SE, Park G, Min CK. Induction of Indoleamine 2,3-dioxygenase by Pre-treatment with Poly(I:C) May Enhance the Efficacy of MSC Treatment in DSS-induced Colitis. Immune Netw 2016; 16:358-365. [PMID: 28035211 PMCID: PMC5195845 DOI: 10.4110/in.2016.16.6.358] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 12/05/2016] [Accepted: 12/08/2016] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been used experimentally for treating inflammatory disorders, partly owing to their immunosuppressive properties. The goal of the study was to determine whether TLR ligands can enhance the therapeutic efficacy of bone marrow-derived MSCs for the treatment of inflammatory bowel disease. Mice (C57BL6) were administered with 4% dextran sulfate sodium (DSS) in drinking water for 7 days and injected with MSCs on days 1 and 3 following DSS ingestion. Our results demonstrated that among various TLR ligands, MSCs treated with polyinosinic-polycytidylic acid [poly(I:C)], which is a TLR3 ligand, more profoundly induced IDO, which is a therapeutically relevant immunosuppressive factor, without any observable phenotype change in vitro. The poly(I:C)-treated MSCs attenuated the pathologic severity of DSS-induced murine colitis when injected i.p. but not i.v. In summary, preconditioning MSCs with poly(I:C) might improve their efficacy in treating DSS-induced colitis, and this effect at least partly depends on the enhancement of their immunosuppressive activity through increasing their production of IDO.
Collapse
Affiliation(s)
- Da-Bin Ryu
- Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Ji-Young Lim
- Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Sung-Eun Lee
- Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Gyeongsin Park
- Department of Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Chang-Ki Min
- Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
74
|
Fuenzalida P, Kurte M, Fernández-O'ryan C, Ibañez C, Gauthier-Abeliuk M, Vega-Letter AM, Gonzalez P, Irarrázabal C, Quezada N, Figueroa F, Carrión F. Toll-like receptor 3 pre-conditioning increases the therapeutic efficacy of umbilical cord mesenchymal stromal cells in a dextran sulfate sodium-induced colitis model. Cytotherapy 2016; 18:630-41. [PMID: 27059200 DOI: 10.1016/j.jcyt.2016.02.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 02/01/2016] [Accepted: 02/04/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND AIMS Immunomodulatory properties of human umbilical cord-derived mesenchymal stromal cells (UCMSCs) can be differentially modulated by toll-like receptors (TLR) agonists. Here, the therapeutic efficacy of short TLR3 and TLR4 pre-conditioning of UCMSCs was evaluated in a dextran sulfate sodium (DSS)-induced colitis in mice. The novelty of this study is that although modulation of human MSCs activity by TLRs is not a new concept, this is the first time that short TLR pre-conditioning has been carried out in a murine inflammatory model of acute colitis. METHODS C57BL/6 mice were exposed to 2.5% dextran sulfate sodium (DSS) in drinking water ad libitum for 7 days. At days 1 and 3, mice were injected intraperitoneally with 1 × 10(6) UCMSCs untreated or TLR3 and TLR4 pre-conditioned UCMSCs. UCMSCs were pre-conditioned with poly(I:C) for TLR3 and LPS for TLR4 for 1 h at 37°C and 5% CO2. We evaluated clinical signs of disease and body weights daily. At the end of the experiment, colon length and histological changes were assessed. RESULTS poly(I:C) pre-conditioned UCMSCs significantly ameliorated the clinical and histopathological severity of DSS-induced colitis compared with UCMSCs or LPS pre-conditioned UCMSCs. In contrast, infusion of LPS pre-conditioned UCMSCs significantly increased clinical signs of disease, colon shortening and histological disease index in DSS-induced colitis. CONCLUSIONS These results show that short in vitro TLR3 pre-conditioning with poly(I:C) enhances the therapeutic efficacy of UCMSCs, which is a major breakthrough for developing improved treatments to patients with inflammatory bowel disease.
Collapse
Affiliation(s)
- Patricia Fuenzalida
- Cellular and Molecular Immunology Laboratory, Faculty of Medicine, University of the Andes, Santiago, Chile
| | - Mónica Kurte
- Cellular and Molecular Immunology Laboratory, Faculty of Medicine, University of the Andes, Santiago, Chile
| | - Catalina Fernández-O'ryan
- Cellular and Molecular Immunology Laboratory, Faculty of Medicine, University of the Andes, Santiago, Chile
| | - Cristina Ibañez
- Cellular and Molecular Immunology Laboratory, Faculty of Medicine, University of the Andes, Santiago, Chile
| | - Melanie Gauthier-Abeliuk
- Cellular and Molecular Immunology Laboratory, Faculty of Medicine, University of the Andes, Santiago, Chile
| | - Ana María Vega-Letter
- Cellular and Molecular Immunology Laboratory, Faculty of Medicine, University of the Andes, Santiago, Chile
| | - Paz Gonzalez
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, University of the Andes, Santiago, Chile; Cells for Cells, Santiago, Chile, Faculty of Medicine, University of the Andes, Santiago, Chile
| | - Carlos Irarrázabal
- Laboratory of Integrative and Molecular Physiology, Faculty of Medicine, University of the Andes, Santiago, Chile
| | - Nataly Quezada
- Cellular and Molecular Immunology Laboratory, Faculty of Medicine, University of the Andes, Santiago, Chile
| | - Fernando Figueroa
- Cellular and Molecular Immunology Laboratory, Faculty of Medicine, University of the Andes, Santiago, Chile
| | - Flavio Carrión
- Cellular and Molecular Immunology Laboratory, Faculty of Medicine, University of the Andes, Santiago, Chile.
| |
Collapse
|
75
|
Lin TH, Pajarinen J, Lu L, Nabeshima A, Cordova LA, Yao Z, Goodman SB. NF-κB as a Therapeutic Target in Inflammatory-Associated Bone Diseases. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2016; 107:117-154. [PMID: 28215222 DOI: 10.1016/bs.apcsb.2016.11.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Inflammation is a defensive mechanism for pathogen clearance and maintaining tissue homeostasis. In the skeletal system, inflammation is closely associated with many bone disorders including fractures, nonunions, periprosthetic osteolysis (bone loss around orthopedic implants), and osteoporosis. Acute inflammation is a critical step for proper bone-healing and bone-remodeling processes. On the other hand, chronic inflammation with excessive proinflammatory cytokines disrupts the balance of skeletal homeostasis involving osteoblastic (bone formation) and osteoclastic (bone resorption) activities. NF-κB is a transcriptional factor that regulates the inflammatory response and bone-remodeling processes in both bone-forming and bone-resorption cells. In vitro and in vivo evidences suggest that NF-κB is an important potential therapeutic target for inflammation-associated bone disorders by modulating inflammation and bone-remodeling process simultaneously. The challenges of NF-κB-targeting therapy in bone disorders include: (1) the complexity of canonical and noncanonical NF-κB pathways; (2) the fundamental roles of NF-κB-mediated signaling for bone regeneration at earlier phases of tissue damage and acute inflammation; and (3) the potential toxic effects on nontargeted cells such as lymphocytes. Recent developments of novel inhibitors with differential approaches to modulate NF-κB activity, and the controlled release (local) or bone-targeting drug delivery (systemic) strategies, have largely increased the translational application of NF-κB therapy in bone disorders. Taken together, temporal modulation of NF-κB pathways with the combination of recent advanced bone-targeting drug delivery techniques is a highly translational strategy to reestablish homeostasis in the skeletal system.
Collapse
Affiliation(s)
- T-H Lin
- Stanford University, Stanford, CA, United States
| | - J Pajarinen
- Stanford University, Stanford, CA, United States
| | - L Lu
- Stanford University, Stanford, CA, United States
| | - A Nabeshima
- Stanford University, Stanford, CA, United States
| | - L A Cordova
- Stanford University, Stanford, CA, United States; Faculty of Dentistry, University of Chile, Santiago, Chile
| | - Z Yao
- Stanford University, Stanford, CA, United States
| | - S B Goodman
- Stanford University, Stanford, CA, United States.
| |
Collapse
|
76
|
Mesenchymal Stem Cells Regulate the Innate and Adaptive Immune Responses Dampening Arthritis Progression. Stem Cells Int 2016; 2016:3162743. [PMID: 27847522 PMCID: PMC5101398 DOI: 10.1155/2016/3162743] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 10/03/2016] [Indexed: 12/22/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent stem cells that are able to immunomodulate cells from both the innate and the adaptive immune systems promoting an anti-inflammatory environment. During the last decade, MSCs have been intensively studied in vitro and in vivo in experimental animal model of autoimmune and inflammatory disorders. Based on these studies, MSCs are currently widely used for the treatment of autoimmune diseases such as rheumatoid arthritis (RA) characterized by complex deregulation of the immune systems. However, the therapeutic properties of MSCs in arthritis are still controverted. These controversies might be due to the diversity of MSC sources and isolation protocols used, the time, the route and dose of MSC administration, the variety of the mechanisms involved in the MSCs suppressive effects, and the complexity of arthritis pathogenesis. In this review, we discuss the role of the interactions between MSCs and the different immune cells associated with arthritis pathogenesis and the possible means described in the literature that could enhance MSCs therapeutic potential counteracting arthritis development and progression.
Collapse
|
77
|
Urrutia M, Fernández S, González M, Vilches R, Rojas P, Vásquez M, Kurte M, Vega-Letter AM, Carrión F, Figueroa F, Rojas P, Irarrázabal C, Fuentealba RA. Overexpression of Glutamate Decarboxylase in Mesenchymal Stem Cells Enhances Their Immunosuppressive Properties and Increases GABA and Nitric Oxide Levels. PLoS One 2016; 11:e0163735. [PMID: 27662193 PMCID: PMC5035029 DOI: 10.1371/journal.pone.0163735] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 09/13/2016] [Indexed: 12/12/2022] Open
Abstract
The neurotransmitter GABA has been recently identified as a potent immunosuppressive agent that targets both innate and adaptive immune systems and prevents disease progression of several autoimmunity models. Mesenchymal stem cells (MSCs) are self-renewing progenitor cells that differentiate into various cell types under specific conditions, including neurons. In addition, MSC possess strong immunosuppressive capabilities. Upon cytokine priming, undifferentiated MSC suppress T-cell proliferation via cell-to-cell contact mechanisms and the secretion of soluble factors like nitric oxide, prostaglandin E2 and IDO. Although MSC and MSC-derived neuron-like cells express some GABAergic markers in vitro, the role for GABAergic signaling in MSC-mediated immunosuppression remains completely unexplored. Here, we demonstrate that pro-inflammatory cytokines selectively regulate GAD-67 expression in murine bone marrow-MSC. However, expression of GAD-65 is required for maximal GABA release by MSC. Gain of function experiments using GAD-67 and GAD-65 co-expression demonstrates that GAD increases immunosuppressive function in the absence of pro-inflammatory licensing. Moreover, GAD expression in MSC evokes an increase in both GABA and NO levels in the supernatants of co-cultured MSC with activated splenocytes. Notably, the increase in NO levels by GAD expression was not observed in cultures of isolated MSC expressing GAD, suggesting crosstalk between these two pathways in the setting of immunosuppression. These results indicate that GAD expression increases MSC-mediated immunosuppression via secretion of immunosuppressive agents. Our findings may help reconsider GABAergic activation in MSC for immunological disorders.
Collapse
Affiliation(s)
- Mariana Urrutia
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Sebastián Fernández
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Marisol González
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Rodrigo Vilches
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Pablo Rojas
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Manuel Vásquez
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Mónica Kurte
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Ana María Vega-Letter
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Flavio Carrión
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Fernando Figueroa
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Patricio Rojas
- Departamento de Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Carlos Irarrázabal
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Rodrigo A. Fuentealba
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
- * E-mail:
| |
Collapse
|
78
|
Su X, Yao X, Sun Z, Han Q, Zhao RC. Optimization of Reference Genes for Normalization of Reverse Transcription Quantitative Real-Time Polymerase Chain Reaction Results in Senescence Study of Mesenchymal Stem Cells. Stem Cells Dev 2016; 25:1355-65. [PMID: 27484587 DOI: 10.1089/scd.2016.0031] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Recently, it has been suggested that cellular senescence is associated with stem cell exhaustion, which reduces the regenerative potential of tissues and contributes to aging and age-related diseases. Mesenchymal stem cells (MSCs) attract a large amount of attention in stem cell research and regeneration medicine because they possess multiple advantages and senescent MSCs could be one of the most useful stem cell models in aging studies. It is important to quantitatively evaluate senescence markers to both identify and study the mechanisms involved in MSC senescence. Reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR) is currently the most widely used tool to quantify the mRNA levels of markers. However, no report has demonstrated the optimal reference genes that should be used to normalize RT-qPCR in senescence studies of MSCs. In this study, we compared 16 commonly used reference genes (GAPDH, ACTB, RPL13A, TBP, B2M, GUSB, RPLPO, YWHAZ, RPS18, EEF1A1, ATP5F1, HPRT1, PGK1, TFRC, UBC, and PPIA) in proliferating or replicative-senescent human adipose-derived MSCs (hAD-MSCs) that were isolated from seven healthy donors aged 29-59 years old. Three algorithms (geNorm, NormFinder, and BestKeeper) were used to determine the most optimal reference gene. The results showed that PPIA exhibited the most stable expression during senescence, while the widely used ACTB exhibited the lowest stability. We also confirmed that different reference genes lead to different evaluations of senescence markers. Our work ensures that results obtained from senescence studies of hAD-MSCs will be appropriately evaluated in both basic research and clinical trials.
Collapse
Affiliation(s)
- Xiaodong Su
- 1 Institute of Basic Medical Sciences, School of Basic Medicine, Peking Union Medical College, Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences , Beijing, People's Republic of China
| | - Xinglei Yao
- 1 Institute of Basic Medical Sciences, School of Basic Medicine, Peking Union Medical College, Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences , Beijing, People's Republic of China .,2 State Key Laboratory of Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences , Beijing, People's Republic of China
| | - Zhao Sun
- 3 Department of Oncology, School of Basic Medicine, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences , Beijing, People's Republic of China
| | - Qin Han
- 1 Institute of Basic Medical Sciences, School of Basic Medicine, Peking Union Medical College, Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences , Beijing, People's Republic of China
| | - Robert Chunhua Zhao
- 1 Institute of Basic Medical Sciences, School of Basic Medicine, Peking Union Medical College, Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences , Beijing, People's Republic of China
| |
Collapse
|
79
|
Weinstock A, Pevsner-Fischer M, Porat Z, Selitrennik M, Zipori D. Cultured Mesenchymal Stem Cells Stimulate an Immune Response by Providing Immune Cells with Toll-Like Receptor 2 Ligand. Stem Cell Rev Rep 2016; 11:826-40. [PMID: 26250539 DOI: 10.1007/s12015-015-9614-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) serve as supporting and regulatory cells, by providing tissues with multiple factors and are also known for their immunosuppressive capabilities. Our laboratory had previously shown that MSCs expressed toll-like receptor (TLR) 2 and are activated by its ligand Pam3Cys. TLR2 is an important component of the innate immune system, as it recognizes bacterial lipopeptides, thus priming a pro-inflammatory immune response. This study showed that Pam3Cys attached extensively to cells of both wild-type and TLR2 deficient cultured MSCs, thus, independently of TLR2. The TLR2 independent binding occurred through the adsorption of the palmitoyl moieties of Pam3Cys. It was further showed that Pam3Cys was transferred from cultured MSCs to immune cells. Moreover, Pam3Cys provided to the immune cells induced a pro-inflammatory response in vitro and in vivo. Overall, it is demonstrated herein that a TLR2 ligand bound to MSCs also through a TLR2 independent mechanism. Furthermore, the ligand incorporated by MSCs is subsequently released to stimulate an immune response both in vitro and in vivo. It is thus suggested that during bacterial infection, stromal cells may retain a reservoir of the TLR2 ligands, in a long-term manner, and release them slowly to maintain an immune response.
Collapse
Affiliation(s)
- Ada Weinstock
- Department of Molecular Cell Biology, Weizmann Institute of Science, 234 Herzl Street, Rehovot, 76100, Israel.
| | - Meirav Pevsner-Fischer
- Department of Molecular Cell Biology, Weizmann Institute of Science, 234 Herzl Street, Rehovot, 76100, Israel.
| | - Ziv Porat
- FACS Unit, Biological Services, Weizmann Institute of Science, 234 Herzl Street, Rehovot, 76100, Israel.
| | - Michael Selitrennik
- Department of Molecular Cell Biology, Weizmann Institute of Science, 234 Herzl Street, Rehovot, 76100, Israel.
| | - Dov Zipori
- Department of Molecular Cell Biology, Weizmann Institute of Science, 234 Herzl Street, Rehovot, 76100, Israel.
| |
Collapse
|
80
|
Cho KA, Park M, Kim YH, Ryu KH, Woo SY. Poly I:C primes the suppressive function of human palatine tonsil-derived MSCs against Th17 differentiation by increasing PD-L1 expression. Immunobiology 2016; 222:394-398. [PMID: 27594385 DOI: 10.1016/j.imbio.2016.08.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 08/26/2016] [Accepted: 08/29/2016] [Indexed: 12/22/2022]
Abstract
It has been established that mesenchymal stem cells (MSCs) can have a suppressive effect on T cells, yet much remains unknown about the underlying mechanisms that support this effect. The T cell co-stimulatory pathway involving the programmed death-1 (PD-1) receptor and its ligand PD-L1 regulates T cell activation, tolerance, and subsequent immune-mediated tissue damage. In this study, human palatine tonsil-derived MSCs (T-MSCs) constitutively expressed PD-L1 and exhibited a suppressive activity that specifically targeted murine Th17 differentiation. Additionally, polyinosinic-polycytidylic acid (poly I:C), a Toll-like receptor 3 (TLR3) ligand, increased PD-L1 expression on T-MSCs. The elevated PD-L1 levels enhanced the suppressive functions of T-MSCs on Th17 differentiation. Therefore, pre-stimulation of T-MSCs with poly I:C may serve as an effective therapeutic priming step for modulating Th17-dominant immune responses.
Collapse
Affiliation(s)
- Kyung-Ah Cho
- Department of Microbiology, School of Medicine, Ewha Womans University, Seoul 07985, Republic of Korea
| | - Minhwa Park
- Department of Microbiology, School of Medicine, Ewha Womans University, Seoul 07985, Republic of Korea
| | - Yu-Hee Kim
- Department of Microbiology, School of Medicine, Ewha Womans University, Seoul 07985, Republic of Korea
| | - Kyung-Ha Ryu
- Department of Pediatrics, School of Medicine, Ewha Womans University, Seoul 07985, Republic of Korea
| | - So-Youn Woo
- Department of Microbiology, School of Medicine, Ewha Womans University, Seoul 07985, Republic of Korea.
| |
Collapse
|
81
|
Wang P, Zeng F, He L, Wang J, Zhang T, Zhang D. Alteration of the immune status of umbilical cord mesenchymal stem cells stimulated by TLR1/2 agonist, Pam3Csk. Mol Med Rep 2016; 14:2206-12. [PMID: 27431529 DOI: 10.3892/mmr.2016.5520] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 05/27/2016] [Indexed: 11/05/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been widely used in clinical trials due to their multiple differentiation ability, low immunogenicity and immunosuppressant effects on immune response. However, accumulating evidence has indicated that MSCs may stimulate in vivo immune responses and result in the disappearance of MSCs following engrafting. Toll‑like receptors (TLRs) are important in immune response induction against invaded pathogens, however, the function of TLRs in regulating the immune status of MSCs has been seldom reported. The present stimulated umbilical cord (UC) MSCs by treatment with the TLR1/2 agonist, Pam3Csk, the to determine whether activation of TLR1/2 signaling alters the immune status of UCMSCs. The results indicated that activation of TLR1/2 increased the proliferation of peripheral blood mononuclear cells (PBMCs) and the production of lactate dehydrogenase in a PBMC‑MSC co‑culture system. The study also demonstrated that Pam3Csk induced the secretion of pro‑inflammatory molecules, and increased the expression levels of cytokine and chemokines in UCMSCs. Flow cytometry analysis indicated that the levels of surface co‑stimulators, CD80 and CD86, were increased on UCMSCs in the presence of Pam3Csk, whereas activation of TLR1/2 exerted no observable effect on the differentiation abilities of UCMSCs. The results of the current study indicated that activation of TLR1/2 signaling may alter the immune status of UCMSCs, however, further mechanistic research is required in future studies.
Collapse
Affiliation(s)
- Pingxi Wang
- Department of Orthopedic Surgery, The Central Hospital of Dazhou, Dazhou, Sichuan 635000, P.R. China
| | - Fanwei Zeng
- Department of Orthopedic Surgery, The Central Hospital of Dazhou, Dazhou, Sichuan 635000, P.R. China
| | - Lina He
- Department of Pharmacy, The Central Hospital of Dazhou, Dazhou, Sichuan 635000, P.R. China
| | - Jin Wang
- Department of Orthopedic Surgery, The Central Hospital of Dazhou, Dazhou, Sichuan 635000, P.R. China
| | - Tingjiu Zhang
- Department of Orthopedic Surgery, The Central Hospital of Dazhou, Dazhou, Sichuan 635000, P.R. China
| | - Dong Zhang
- Department of Orthopedic Surgery, The Central Hospital of Dazhou, Dazhou, Sichuan 635000, P.R. China
| |
Collapse
|
82
|
Karimi MH, Barzkar Z, Babaee M, Naghdi M. Evaluating Effect of Mesenchymal Stem Cells on Expression of TLR2 and TLR4 in Mouse DCs. Adv Pharm Bull 2016; 6:179-86. [PMID: 27478779 DOI: 10.15171/apb.2016.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 03/20/2016] [Accepted: 04/02/2016] [Indexed: 12/29/2022] Open
Abstract
PURPOSE Mesenchymal stem cells (MSCs) are multipotent cells and recent findings suggest immunomodulatory effect of them on immune cells including T cells and dendritic cells (DCs). DCs are the most potent antigen presenting cells. It seems because of immunoregulatory properties of MSCs, they can affect the maturation and differentiation of DCs. DCs express a kind of surface receptors called toll-like receptors (TLRs) and play a key role in maturation process and activation of DCs. The aim of this study was to evaluate expression of TLR2 and TLR4 on DCs after exposure to mesenchymal stem cell's supernatant in culture media containing LPS and devoid of it. METHODS In this experimental study, MSCs and DCs were extracted from adult Balb/c mouse bone marrow and spleen, respectively. MSCs supernatant were collected 24 and 48 h after 5(th) passage, and in adjusted with DCs culture. Isolated DCs were co-cultured with MSCs supernatant, incubation time were 24 and 48 hours. mRNA levels of TLR2 and TLR4 were evaluated using real time PCR technique. RESULTS The results demonstrated that although, expressions of these two receptors were up-regulated in culture media lacking LPS in comparison with the control group but the increase was not significant. There were no significant associations between LPS stimulated DCs with and without MSCs supernatants. CONCLUSION According to the results presented here, it appears that TLR2 and TLR4 gene expressions on the DCs are not affected by MSCs supernatant.
Collapse
Affiliation(s)
| | - Zahra Barzkar
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Maryam Babaee
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Majid Naghdi
- Department of Anatomical Science, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
83
|
Pleyer L, Valent P, Greil R. Mesenchymal Stem and Progenitor Cells in Normal and Dysplastic Hematopoiesis-Masters of Survival and Clonality? Int J Mol Sci 2016; 17:ijms17071009. [PMID: 27355944 PMCID: PMC4964385 DOI: 10.3390/ijms17071009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 05/20/2016] [Accepted: 06/08/2016] [Indexed: 02/07/2023] Open
Abstract
Myelodysplastic syndromes (MDS) are malignant hematopoietic stem cell disorders that have the capacity to progress to acute myeloid leukemia (AML). Accumulating evidence suggests that the altered bone marrow (BM) microenvironment in general, and in particular the components of the stem cell niche, including mesenchymal stem cells (MSCs) and their progeny, play a pivotal role in the evolution and propagation of MDS. We here present an overview of the role of MSCs in the pathogenesis of MDS, with emphasis on cellular interactions in the BM microenvironment and related stem cell niche concepts. MSCs have potent immunomodulatory capacities and communicate with diverse immune cells, but also interact with various other cellular components of the microenvironment as well as with normal and leukemic stem and progenitor cells. Moreover, compared to normal MSCs, MSCs in MDS and AML often exhibit altered gene expression profiles, an aberrant phenotype, and abnormal functional properties. These alterations supposedly contribute to the “reprogramming” of the stem cell niche into a disease-permissive microenvironment where an altered immune system, abnormal stem cell niche interactions, and an impaired growth control lead to disease progression. The current article also reviews molecular targets that play a role in such cellular interactions and possibilities to interfere with abnormal stem cell niche interactions by using specific targeted drugs.
Collapse
Affiliation(s)
- Lisa Pleyer
- 3rd Medical Department with Hematology and Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Laboratory for Immunological and Molecular Cancer Research, Oncologic Center, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria.
- Center for Clinical Cancer and Immunology Trials at Salzburg Cancer Research Institute, 5020 Salzburg, Austria.
- 3rd Medical Department, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology & Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, 1090 Vienna, Austria.
| | - Richard Greil
- 3rd Medical Department with Hematology and Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Laboratory for Immunological and Molecular Cancer Research, Oncologic Center, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria.
- Center for Clinical Cancer and Immunology Trials at Salzburg Cancer Research Institute, 5020 Salzburg, Austria.
- 3rd Medical Department, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| |
Collapse
|
84
|
Gingival Mesenchymal Stem/Progenitor Cells: A Unique Tissue Engineering Gem. Stem Cells Int 2016; 2016:7154327. [PMID: 27313628 PMCID: PMC4903147 DOI: 10.1155/2016/7154327] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/28/2016] [Accepted: 04/28/2016] [Indexed: 12/27/2022] Open
Abstract
The human gingiva, characterized by its outstanding scarless wound healing properties, is a unique tissue and a pivotal component of the periodontal apparatus, investing and surrounding the teeth in their sockets in the alveolar bone. In the last years gingival mesenchymal stem/progenitor cells (G-MSCs), with promising regenerative and immunomodulatory properties, have been isolated and characterized from the gingival lamina propria. These cells, in contrast to other mesenchymal stem/progenitor cell sources, are abundant, readily accessible, and easily obtainable via minimally invasive cell isolation techniques. The present review summarizes the current scientific evidence on G-MSCs' isolation, their characterization, the investigated subpopulations, the generated induced pluripotent stem cells- (iPSC-) like G-MSCs, their regenerative properties, and current approaches for G-MSCs' delivery. The review further demonstrates their immunomodulatory properties, the transplantation preconditioning attempts via multiple biomolecules to enhance their attributes, and the experimental therapeutic applications conducted to treat multiple diseases in experimental animal models in vivo. G-MSCs show remarkable tissue reparative/regenerative potential, noteworthy immunomodulatory properties, and primary experimental therapeutic applications of G-MSCs are very promising, pointing at future biologically based therapeutic techniques, being potentially superior to conventional clinical treatment modalities.
Collapse
|
85
|
Wang X, Zhu Y, Xu B, Wang J, Liu X. Identification of TLR2 and TLR4‑induced microRNAs in human mesenchymal stem cells and their possible roles in regulating TLR signals. Mol Med Rep 2016; 13:4969-80. [PMID: 27121537 PMCID: PMC4878556 DOI: 10.3892/mmr.2016.5197] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 01/14/2016] [Indexed: 02/06/2023] Open
Abstract
Toll-like receptors (TLRs) are expressed in human bone marrow-derived mesenchymal stromal cells (BM-MSCs), and the activation of TLRs is important in proliferation, differentiation, migration and hematopoiesis-supporting functions of BM-MSCs. However, the molecular mechanisms underlying these processes remain to be elucidated. MicroRNAs (miRNAs) are involved in various biological functions by mediating mRNA degradation or inhibiting translation of target genes. The present study aimed to identify whether TLRs regulate the expression of miRNAs in BM-MSCs and elucidate the regulatory roles of miRNAs. Illumina high-throughput sequencing was used to profile miRNAs expressed in BM-MSCs stimulated with TLR2 agonist, PAM3CSK4 (PM) or TLR4 agonist, lipopolysaccharides (LPS). A marked expression change upon PM or LPS treatment was observed for 164 known miRNAs and six novel miRNAs that were identified. The expression of six novel miRNAs and 40 randomly selected known miRNAs was further validated by reverse transcription-quantitative polymerase chain reaction. In addition, bioinformatic methods were used to predict the potential target genes of the abundant known miRNAs. The gene ontology analysis demonstrated that predicted targets were enriched in the regulation of signal transduction, cellular processes and macromolecule metabolic processes. Kyoto Encyclopedia of Genes and Genomes pathway analysis suggested that these potential targets were involved in numerous important pathways, predominantly including mitogen-activated protein kinase, phosphati-dylinositol-4,5-bisphosphate 3-kinase-Akt, neurotrophin and cancer-associated signaling pathways. The present study aimed to identify the global expression change of miRNAs in BM-MSCs stimulated with LPS and PM, providing the opportunity to further elucidate the roles of miRNAs in mediating TLR signals to regulate the functions of BM-MSCs.
Collapse
Affiliation(s)
- Xingbing Wang
- Department of Hematology, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui 230001, P.R. China
| | - Yunxia Zhu
- Department of Hematology, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui 230001, P.R. China
| | - Bo Xu
- Reproductive Medicine Center, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui 230001, P.R. China
| | - Jing Wang
- Department of Hematology, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui 230001, P.R. China
| | - Xin Liu
- Department of Hematology, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui 230001, P.R. China
| |
Collapse
|
86
|
Lindsay SL, Johnstone SA, McGrath MA, Mallinson D, Barnett SC. Comparative miRNA-Based Fingerprinting Reveals Biological Differences in Human Olfactory Mucosa- and Bone-Marrow-Derived Mesenchymal Stromal Cells. Stem Cell Reports 2016; 6:729-742. [PMID: 27117785 PMCID: PMC4940454 DOI: 10.1016/j.stemcr.2016.03.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 03/23/2016] [Accepted: 03/24/2016] [Indexed: 12/22/2022] Open
Abstract
Previously we reported that nestin-positive human mesenchymal stromal cells (MSCs) derived from the olfactory mucosa (OM) enhanced CNS myelination in vitro to a greater extent than bone-marrow-derived MSCs (BM-MSCs). miRNA-based fingerprinting revealed the two MSCs were 64% homologous, with 26 miRNAs differentially expressed. We focused on miR-146a-5p and miR-140-5p due to their reported role in the regulation of chemokine production and myelination. The lower expression of miR-140-5p in OM-MSCs correlated with higher secretion of CXCL12 compared with BM-MSCs. Addition of CXCL12 and its pharmacological inhibitors to neural co-cultures supported these data. Studies on related miR-146a-5p targets demonstrated that OM-MSCs had lower levels of Toll-like receptors and secreted less pro-inflammatory cytokines, IL-6, IL-8, and CCL2. OM-MSCs polarized microglia to an anti-inflammatory phenotype, illustrating potential differences in their inflammatory response. Nestin-positive OM-MSCs could therefore offer a cell transplantation alternative for CNS repair, should these biological behaviors be translated in vivo. OM-MSCs share 64% miRNA homology to BM-MSCs and differentially express 26 miRNAs CXCL12 promotes CNS myelination and is negatively regulated by miR-140-5p in BM-MSCs miR-146a-5p negatively regulates IL-6, IL-8, TLR2, and TLR4 on OM-MSCs These properties make OM-MSCs a suitable candidate for transplant-mediated CNS repair
Collapse
Affiliation(s)
- Susan Louise Lindsay
- Institute of Infection, Inflammation and Immunity, Glasgow Biomedical Research Centre, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK
| | - Steven Andrew Johnstone
- Institute of Infection, Inflammation and Immunity, Glasgow Biomedical Research Centre, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK
| | - Michael Anthony McGrath
- Institute of Infection, Inflammation and Immunity, Glasgow Biomedical Research Centre, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK
| | - David Mallinson
- Institute of Infection, Inflammation and Immunity, Glasgow Biomedical Research Centre, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK; Sistemic UK, Kelvin Campus, Maryhill Road, Glasgow G20 0SP, UK
| | - Susan Carol Barnett
- Institute of Infection, Inflammation and Immunity, Glasgow Biomedical Research Centre, University of Glasgow, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK.
| |
Collapse
|
87
|
Abstract
Adult or somatic stem cells are tissue-resident cells with the ability to proliferate, exhibit self-maintenance as well as to generate new cells with the principal phenotypes of the tissue in response to injury or disease. Due to their easy accessibility and their potential use in regenerative medicine, adult stem cells raise the hope for future personalisable therapies. After infection or during injury, they are exposed to broad range of pathogen or damage-associated molecules leading to changes in their proliferation, migration and differentiation. The sensing of such damage and infection signals is mostly achieved by Toll-Like Receptors (TLRs) with Toll-like receptor 4 being responsible for recognition of bacterial lipopolysaccharides (LPS) and endogenous danger-associated molecular patterns (DAMPs). In this review, we examine the current state of knowledge on the TLR4-mediated signalling in different adult stem cell populations. Specifically, we elaborate on the role of TLR4 and its ligands on proliferation, differentiation and migration of mesenchymal stem cells, hematopoietic stem cells as well as neural stem cells. Finally, we discuss conceptual and technical pitfalls in investigation of TLR4 signalling in stem cells.
Collapse
|
88
|
Linard C, Strup-Perrot C, Lacave-Lapalun JV, Benderitter M. Flagellin preconditioning enhances the efficacy of mesenchymal stem cells in an irradiation-induced proctitis model. J Leukoc Biol 2016; 100:569-80. [PMID: 26992430 DOI: 10.1189/jlb.3a0915-393r] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 02/29/2016] [Indexed: 12/12/2022] Open
Abstract
The success of mesenchymal stem cell transplantation for proctitis depends not only on cell donors but also on host microenvironmental factors, which play a major role in conditioning mesenchymal stem cell immunosuppressive action and repair. This study sought to determine if flagellin, a TLR5 ligand, can enhance the mesenchymal stem cell treatment efficacy in radiation-induced proctitis. With the use of a colorectal model of 27 Gy irradiation in rats, we investigated and compared the effects on immune capacity and remodeling at 28 d after irradiation of the following: 1) systemic mesenchymal stem cell (5 × 10(6)) administration at d 7 after irradiation, 2) administration of flagellin at d 3 and systemic mesenchymal stem cell administration at d 7, and 3) in vitro preconditioning of mesenchymal stem cells with flagellin, 24 h before their administration on d 7. The mucosal CD8(+) T cell population was normalized after treatment with flagellin-preconditioned mesenchymal stem cells or flagellin plus mesenchymal stem cells, whereas mesenchymal stem cells alone did not alter the radiation-induced elevation of CD8(+) T cell frequency. Mesenchymal stem cell treatment returned the irradiation-elevated frequency of CD25(+) cells in the mucosa-to-control levels, whereas both flagellin-preconditioned mesenchymal stem cell and flagellin-plus-mesenchymal stem cell treatment each significantly increased not only CD25(+) cell frequency but also forkhead box p3 and IL-2Rα expression. Specifically, IL-10 was overexpressed after flagellin-preconditioned mesenchymal stem cell treatment. Analysis of collagen expression showed that the collagen type 1/collagen type 3 ratio, an indicator of wound-healing maturation, was low in the irradiated and mesenchymal stem cell-treated groups and returned to the normal level only after the flagellin-preconditioned mesenchymal stem cell treatment. This was associated with a reduction in myofibroblast accumulation. In a proctitis model, flagellin-preconditioned mesenchymal stem cells improved colonic immune capacity and enhanced tissue remodeling.
Collapse
Affiliation(s)
- Christine Linard
- Institut de Radioprotection et de Sûreté Nucléaire, Fontenay-aux-Roses, France
| | - Carine Strup-Perrot
- Institut de Radioprotection et de Sûreté Nucléaire, Fontenay-aux-Roses, France
| | | | - Marc Benderitter
- Institut de Radioprotection et de Sûreté Nucléaire, Fontenay-aux-Roses, France
| |
Collapse
|
89
|
Yu Y, Wu RX, Gao LN, Xia Y, Tang HN, Chen FM. Stromal cell-derived factor-1-directed bone marrow mesenchymal stem cell migration in response to inflammatory and/or hypoxic stimuli. Cell Adh Migr 2016; 10:342-59. [PMID: 26745021 DOI: 10.1080/19336918.2016.1139287] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Directing cell trafficking toward a target site of interest is critical for advancing stem cell therapy in clinical theranostic applications. In this study, we investigated the effects of inflammatory and/or hypoxic stimuli on the migration of bone marrow mesenchymal stem cells (BMMSCs) during in vitro culture and after in vivo implantation. Using tablet scratch experiments and observations from a transwell system, we found that both inflammatory and hypoxic stimuli significantly enhanced cell migration. However, the combination of inflammatory and hypoxic stimuli did not result in a synergistic effect. The presence of stromal cell-derived factor-1 (SDF-1) significantly enhanced cell migration irrespective of the incubation conditions, and these positive effects could be blocked by treatment with AMD3100. Based on a time course experiment, we found that preconditioning cells with either inflammatory or hypoxic stimuli for 24 h or with both stimuli for 12 h led to high levels of chemokine receptor type 4 (CXCR4) expression. In vivo studies further demonstrated that pretreatment of BMMSCs with inflammatory and/or hypoxic stimuli resulted in an increased number of systemically injected cells migrating toward skin injuries, and local SDF-1 administration significantly increased cell migration. These findings suggest that in vitro control of either inflammatory or hypoxic stimuli has significant potential to enhance SDF-1-directed BMMSC migration via the upregulation of CXCR4 expression. Although combining the stimuli did not necessarily lead to a synergistic effect, the potential to reduce the dose and time required for cell preconditioning indicates that combinations of various strategies warrant further exploration.
Collapse
Affiliation(s)
- Yang Yu
- a State Key Laboratory of Military Stomatology, Department of Periodontology, School of Stomatology, Fourth Military Medical University , Xi'an , P. R., China.,b Shaanxi Key Laboratory of Stomatology, Biomaterials Unit, School of Stomatology, Fourth Military Medical University , Xi'an , P. R., China
| | - Rui-Xin Wu
- a State Key Laboratory of Military Stomatology, Department of Periodontology, School of Stomatology, Fourth Military Medical University , Xi'an , P. R., China.,b Shaanxi Key Laboratory of Stomatology, Biomaterials Unit, School of Stomatology, Fourth Military Medical University , Xi'an , P. R., China
| | - Li-Na Gao
- a State Key Laboratory of Military Stomatology, Department of Periodontology, School of Stomatology, Fourth Military Medical University , Xi'an , P. R., China.,b Shaanxi Key Laboratory of Stomatology, Biomaterials Unit, School of Stomatology, Fourth Military Medical University , Xi'an , P. R., China
| | - Yu Xia
- a State Key Laboratory of Military Stomatology, Department of Periodontology, School of Stomatology, Fourth Military Medical University , Xi'an , P. R., China.,b Shaanxi Key Laboratory of Stomatology, Biomaterials Unit, School of Stomatology, Fourth Military Medical University , Xi'an , P. R., China
| | - Hao-Ning Tang
- a State Key Laboratory of Military Stomatology, Department of Periodontology, School of Stomatology, Fourth Military Medical University , Xi'an , P. R., China.,b Shaanxi Key Laboratory of Stomatology, Biomaterials Unit, School of Stomatology, Fourth Military Medical University , Xi'an , P. R., China
| | - Fa-Ming Chen
- a State Key Laboratory of Military Stomatology, Department of Periodontology, School of Stomatology, Fourth Military Medical University , Xi'an , P. R., China.,b Shaanxi Key Laboratory of Stomatology, Biomaterials Unit, School of Stomatology, Fourth Military Medical University , Xi'an , P. R., China
| |
Collapse
|
90
|
Fawzy El-Sayed KM, Klingebiel P, Dörfer CE. Toll-like Receptor Expression Profile of Human Dental Pulp Stem/Progenitor Cells. J Endod 2016; 42:413-7. [PMID: 26769027 DOI: 10.1016/j.joen.2015.11.014] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 11/12/2015] [Accepted: 11/17/2015] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Human dental pulp stem/progenitor cells (DPSCs) show remarkable regenerative potential in vivo. During regeneration, DPSCs may interact with their inflammatory environment via toll-like receptors (TLRs). The present study aimed to depict for the first time the TLR expression profile of DPSCs. METHODS Cells were isolated from human dental pulp, STRO-1-immunomagnetically sorted, and seeded out to obtain single colony-forming units. DPSCs were characterized for CD14, CD34, CD45, CD73, CD90, CD105, and CD146 expression and for their multilineage differentiation potential. After incubation of DPSCs in basic or inflammatory medium (interleukin-1β, interferon-γ, interferon-α, tumor necrosis factor-α), TLR expression profiles were generated (DPSCs and DPSCs-i). RESULTS DPSCs showed all characteristics of stem/progenitor cells. In basic medium DPSCs expressed TLRs 1-10 in different quantities. The inflammatory medium upregulated the expression of TLRs 2, 3, 4, 5, and 8, downregulated TLRs 1, 7, 9, and 10, and abolished TLR6. CONCLUSIONS The current study describes for the first time the distinctive TLR expression profile of DPSCs in uninflamed and inflamed conditions.
Collapse
Affiliation(s)
- Karim M Fawzy El-Sayed
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, Kiel, Germany; Oral Medicine and Periodontology Department, Faculty of Oral and Dental Medicine, Cairo University, Cairo, Egypt.
| | - Pauline Klingebiel
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, Kiel, Germany
| | - Christof E Dörfer
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, Kiel, Germany
| |
Collapse
|
91
|
Prieto P, Fernández-Velasco M, Fernández-Santos ME, Sánchez PL, Terrón V, Martín-Sanz P, Fernández-Avilés F, Boscá L. Cell Expansion-Dependent Inflammatory and Metabolic Profile of Human Bone Marrow Mesenchymal Stem Cells. Front Physiol 2016; 7:548. [PMID: 27899899 PMCID: PMC5110552 DOI: 10.3389/fphys.2016.00548] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 10/31/2016] [Indexed: 02/05/2023] Open
Abstract
Stem cell therapy has emerged as a promising new area in regenerative medicine allowing the recovery of viable tissues. Among the many sources of adult stem cells, bone marrow-derived are easy to expand in culture via plastic adherence and their multipotentiality for differentiation make them ideal for clinical applications. Interestingly, several studies have indicated that MSCs expansion in vitro may be limited mainly due to "cell aging" related to the number of cell divisions in culture. We have determined that MSCs exhibit a progressive decline across successive passages in the expression of stem cell markers, in plasticity and in the inflammatory response, presenting low immunogenicity. We have exposed human MSCs after several passages to TLRs ligands and analyzed their inflammatory response. These cells responded to pro-inflammatory stimuli (i.e., NOS-2 expression) and to anti-inflammatory cytokines (i.e., HO1 and Arg1) until two expansions, rapidly declining upon subculture. Moreover, in the first passages, MSCs were capable to release IL1β, IL6, and IL8, as well as to produce active MMPs allowing them to migrate. Interestingly enough, after two passages, anaerobic glycolysis was enhanced releasing high levels of lactate to the extracellular medium. All these results may have important implications for the safety and efficacy of MSCs-based cell therapies.
Collapse
Affiliation(s)
- Patricia Prieto
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM)Madrid, Spain
| | | | - María E. Fernández-Santos
- Servicio de Cardiología, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio MarañónMadrid, Spain
| | - Pedro L. Sánchez
- Servicio de Cardiología, Hospital Clínico de SalamancaSalamanca, Spain
| | - Verónica Terrón
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM)Madrid, Spain
| | - Paloma Martín-Sanz
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM)Madrid, Spain
| | - Francisco Fernández-Avilés
- Servicio de Cardiología, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio MarañónMadrid, Spain
- Facultad de Medicina, Universidad Complutense de Madrid, Ciudad UniversitariaMadrid, Spain
- *Correspondence: Francisco Fernández-Avilés
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM)Madrid, Spain
- Lisardo Boscá
| |
Collapse
|
92
|
Fawzy-El-Sayed K, Mekhemar M, Adam-Klages S, Kabelitz D, Dörfer C. TlR expression profile of human gingival margin-derived stem progenitor cells. Med Oral Patol Oral Cir Bucal 2016; 21:e30-8. [PMID: 26615501 PMCID: PMC4765758 DOI: 10.4317/medoral.20593] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 08/07/2015] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Gingival margin-derived stem/progenitor cells (G-MSCs) show remarkable periodontal regenerative potential in vivo. During regeneration, G-MSCs may interact with their inflammatory environment via toll-like-receptors (TLRs). The present study aimed to depict the G-MSCs TLRs expression profile. MATERIAL AND METHODS Cells were isolated from free gingival margins, STRO-1-immunomagnetically sorted and seeded to obtain single colony forming units (CFUs). G-MSCs were characterized for CD14, CD34, CD45, CD73, CD90, CD105, CD146 and STRO-1 expression, and for multilineage differentiation potential. Following G-MSCs' incubation in basic or inflammatory medium (IL-1β, IFN-γ, IFN-α, TNF-α) a TLR expression profile was generated. RESULTS G-MSCs showed all stem/progenitor cells' characteristics. In basic medium G-MSCs expressed TLRs 1, 2, 3, 4, 5, 6, 7, and 10. The inflammatory medium significantly up-regulated TLRs 1, 2, 4, 5, 7 and 10 and diminished TLR 6 (p≤0.05, Wilcoxon-Signed-Ranks-Test). CONCLUSIONS The current study describes for the first time the distinctive TLRs expression profile of G-MSCs under uninflamed and inflamed conditions.
Collapse
Affiliation(s)
- Karim Fawzy-El-Sayed
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts-Universität zu Kiel, Arnold-Heller-Str. 3, Haus 26, 24105 Kiel, Germany,
| | | | | | | | | |
Collapse
|
93
|
Chen G, Yue A, Ruan Z, Yin Y, Wang R, Ren Y, Zhu L. Comparison of biological characteristics of mesenchymal stem cells derived from maternal-origin placenta and Wharton's jelly. Stem Cell Res Ther 2015; 6:228. [PMID: 26607396 PMCID: PMC4660673 DOI: 10.1186/s13287-015-0219-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 10/21/2015] [Accepted: 10/29/2015] [Indexed: 12/04/2022] Open
Abstract
INTRODUCTION Although mesenchymal stem cells (MSCs) from different sources share many similar characteristics, they also exhibit individual properties. In this study, we compared MSCs derived from Wharton's jelly in the umbilical cord with those derived from the decidual basalis in the maternal part of the placenta to better understand the similarities and differences between these two cell types. METHOD The morphology, immunophenotype (as assessed using flow cytometry), and multi-lineage differentiation potential were analyzed. Karyotype analysis was carried out to determine the origin of the MSCs. Growth kinetics were evaluated using analysis of the population doubling time and cell cycle. Immunosuppressive function was analyzed using mixed lymphocyte culture. RESULTS MSCs from Wharton's jelly and the decidua basalis exhibited similar morphology, immunophenotype, and differentiation potential to osteogenesis and adipogenesis. The percentage of MSCs in the G0/G1 phase was higher in the case of Wharton's jelly than in the case of the decidua basalis (P < 0.05). Decidual MSCs displayed more remarkable immunosuppressive effects on phytohemagglutinin-stimulated T-cell proliferation (P < 0.05). CONCLUSION MSCs from both sources had similar basic biological properties, but decidual MSCs had slower proliferation and stronger immunosuppressive function.
Collapse
Affiliation(s)
- Gecai Chen
- Department of Cardiology, Taizhou Renmin Hospital, Taizhou, Jiangsu Province, China.
| | - Aihuan Yue
- Stem Cell Research Center, Taizhou, Jiangsu Province, China.
| | - Zhongbao Ruan
- Department of Cardiology, Taizhou Renmin Hospital, Taizhou, Jiangsu Province, China.
| | - Yigang Yin
- Department of Cardiology, Taizhou Renmin Hospital, Taizhou, Jiangsu Province, China.
| | - Ruzhu Wang
- Department of Cardiology, Taizhou Renmin Hospital, Taizhou, Jiangsu Province, China.
| | - Yin Ren
- Department of Cardiology, Taizhou Renmin Hospital, Taizhou, Jiangsu Province, China.
| | - Li Zhu
- Department of Cardiology, Taizhou Renmin Hospital, Taizhou, Jiangsu Province, China.
| |
Collapse
|
94
|
Transcriptome Analysis of Long Noncoding RNAs in Toll-Like Receptor 3-Activated Mesenchymal Stem Cells. Stem Cells Int 2015; 2016:6205485. [PMID: 26681952 PMCID: PMC4670881 DOI: 10.1155/2016/6205485] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 05/21/2015] [Accepted: 06/11/2015] [Indexed: 11/25/2022] Open
Abstract
Mesenchymal stem cells (MSCs) possess great immunomodulatory capacity which lays the foundation for their therapeutic effects in a variety of diseases. Recently, toll-like receptors (TLR) have been shown to modulate MSC functions; however, the underlying molecular mechanisms are poorly understood. Emerging evidence suggests that long noncoding RNAs (lncRNAs) are an important class of regulators involved in a wide range of biological processes. To explore the potential involvement of lncRNAs in TLR stimulated MSCs, we performed a comprehensive lncRNA and mRNA profiling through microarray. 10.2% of lncRNAs (1733 out of 16967) and 15.1% of mRNA transcripts (1760 out of 11632) were significantly differentially expressed (absolute fold-change ≥5
, P value ≤0.05) in TLR3 stimulated MSCs. Furthermore, we characterized the differentially expressed lncRNAs through their classes and length distribution and correlated them with differentially expressed mRNA. Here, we are the first to determine genome-wide lncRNAs expression patterns in TLR3 stimulated MSCs by microarray and this work could provide a comprehensive framework of the transcriptome landscapes of TLR3 stimulated MSCs.
Collapse
|
95
|
Li Q, Ma Y, Li L, Bao J, Zhang L. Flagellin influences the expression of a variety of important cytokines and chemokines without affecting the immune status of umbilical cord mesenchymal stem cells. Mol Med Rep 2015; 12:6955-61. [PMID: 26330280 DOI: 10.3892/mmr.2015.4276] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 07/22/2015] [Indexed: 02/05/2023] Open
Abstract
Multipotent differentiation ability, low expression of co‑stimulatory molecules and immunosuppressive effects render mesenchymal stem cells (MSCs) the best candidate for use in cell‑based therapy. At present, MSCs are widely used in clinical trials, which have, however, failed to demonstrate any long‑term therapeutic benefits of MSCs. Therefore, to improve the therapeutic potential of MSCs, it is important to understand the underlying mechanisms of the rapid disappearance of MSCs after transplantation. Toll‑like receptors (TLRs) have important roles in the induction of the immune response and the regulation of biological functions. The present study employed TLR5 agonist flagellin in order to assess whether the activation of the TLR5 pathway alters the immune status and influences the differentiation ability of MSCs isolated from umbilical cords (UC). Assessment of the proliferation of peripheral blood mononuclear cells co‑cultured with UCMSCs as well as their lactate dehydrogenase release indicated that flagellin stimulation failed to increase the immunogenicity of UCMSCs. Flow cytometric and histological cell differentiation analyses also confirmed that TLR5 activation neither enhanced the expression of co‑stimulatory molecules (CD80, CD86 and human leukocyte antigen‑E), nor influenced the differentiation ability of UCMSCs. However, polymerase chain reaction analysis indicated the induction of pro‑inflammatory molecules by flagellin, which suggested that the induced immune‑associated molecules may mediate biological functions of UCMSCs other than those assessed by the present study. Future studies should focus on the role of TLR5 in regulating other functions of UCMSCs.
Collapse
Affiliation(s)
- Quansheng Li
- Department of Biliary Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yu Ma
- Department of Thyroid and Breast Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Li Li
- Laboratory of Pathology, Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ji Bao
- Laboratory of Pathology, Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Li Zhang
- Laboratory of Pathology, Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
96
|
Ebert R, Benisch P, Krug M, Zeck S, Meißner-Weigl J, Steinert A, Rauner M, Hofbauer L, Jakob F. Acute phase serum amyloid A induces proinflammatory cytokines and mineralization via toll-like receptor 4 in mesenchymal stem cells. Stem Cell Res 2015; 15:231-9. [PMID: 26135899 DOI: 10.1016/j.scr.2015.06.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 05/20/2015] [Accepted: 06/21/2015] [Indexed: 01/05/2023] Open
Abstract
The role of serum amyloid A (SAA) proteins, which are ligands for toll-like receptors, was analyzed in human bone marrow-derived mesenchymal stem cells (hMSCs) and their osteogenic offspring with a focus on senescence, differentiation and mineralization. In vitro aged hMSC developed a senescence-associated secretory phenotype (SASP), resulting in enhanced SAA1/2, TLR2/4 and proinflammatory cytokine (IL6, IL8, IL1β, CXCL1, CXCL2) expression before entering replicative senescence. Recombinant human SAA1 (rhSAA1) induced SASP-related genes and proteins in MSC, which could be abolished by cotreatment with the TLR4-inhibitor CLI-095. The same pattern of SASP-resembling genes was stimulated upon induction of osteogenic differentiation, which is accompanied by autocrine SAA1/2 expression. In this context additional rhSAA1 enhanced the SASP-like phenotype, accelerated the proinflammatory phase of osteogenic differentiation and enhanced mineralization. Autocrine/paracrine and rhSAA1 via TLR4 stimulate a proinflammatory phenotype that is both part of the early phase of osteogenic differentiation and the development of senescence. This signaling cascade is tightly involved in bone formation and mineralization, but may also propagate pathological extraosseous calcification conditions such as calcifying inflammation and atherosclerosis.
Collapse
Affiliation(s)
- Regina Ebert
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Brettreichstrasse 11, 97074 Würzburg, Germany.
| | - Peggy Benisch
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Brettreichstrasse 11, 97074 Würzburg, Germany
| | - Melanie Krug
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Brettreichstrasse 11, 97074 Würzburg, Germany
| | - Sabine Zeck
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Brettreichstrasse 11, 97074 Würzburg, Germany
| | - Jutta Meißner-Weigl
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Brettreichstrasse 11, 97074 Würzburg, Germany
| | - Andre Steinert
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Brettreichstrasse 11, 97074 Würzburg, Germany
| | - Martina Rauner
- Division of Endocrinology, Diabetes Bone Metabolism, Technical University of Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Lorenz Hofbauer
- Division of Endocrinology, Diabetes Bone Metabolism, Technical University of Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Franz Jakob
- Orthopedic Center for Musculoskeletal Research, Orthopedic Department, University of Würzburg, Brettreichstrasse 11, 97074 Würzburg, Germany.
| |
Collapse
|
97
|
Human Bone Marrow-Derived Mesenchymal Stromal Cells Differentially Inhibit Cytokine Production by Peripheral Blood Monocytes Subpopulations and Myeloid Dendritic Cells. Stem Cells Int 2015; 2015:819084. [PMID: 26060498 PMCID: PMC4427776 DOI: 10.1155/2015/819084] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 04/05/2015] [Indexed: 12/17/2022] Open
Abstract
The immunosuppressive properties of mesenchymal stromal/stem cells (MSC) rendered them an attractive therapeutic approach for immune disorders and an increasing body of evidence demonstrated their clinical value. However, the influence of MSC on the function of specific immune cell populations, namely, monocyte subpopulations, is not well elucidated. Here, we investigated the influence of human bone marrow MSC on the cytokine and chemokine expression by peripheral blood classical, intermediate and nonclassical monocytes, and myeloid dendritic cells (mDC), stimulated with lipopolysaccharide plus interferon (IFN)γ. We found that MSC effectively inhibit tumor necrosis factor- (TNF-) α and macrophage inflammatory protein- (MIP-) 1β protein expression in monocytes and mDC, without suppressing CCR7 and CD83 protein expression. Interestingly, mDC exhibited the highest degree of inhibition, for both TNF-α and MIP-1β, whereas the reduction of TNF-α expression was less marked for nonclassical monocytes. Similarly, MSC decreased mRNA levels of interleukin- (IL-) 1β and IL-6 in classical monocytes, CCL3, CCL5, CXCL9, and CXCL10 in classical and nonclassical monocytes, and IL-1β and CXCL10 in mDC. MSC do not impair the expression of maturation markers in monocytes and mDC under our experimental conditions; nevertheless, they hamper the proinflammatory function of monocytes and mDC, which may impede the development of inflammatory immune responses.
Collapse
|
98
|
Ryu JH, Park M, Kim BK, Ryu KH, Woo SY. Tonsil-derived mesenchymal stromal cells produce CXCR2-binding chemokines and acquire follicular dendritic cell-like phenotypes under TLR3 stimulation. Cytokine 2015; 73:225-35. [PMID: 25794662 DOI: 10.1016/j.cyto.2015.02.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 02/22/2015] [Accepted: 02/23/2015] [Indexed: 01/09/2023]
Abstract
We previously isolated mesenchymal stromal cells from human tonsils (T-MSCs) and showed the potential of these cells to differentiate into the mesodermal lineage and acquire a follicular dendritic cell (FDC) phenotype under cytokine stimulation. Because these T-MSCs were originally isolated from inflamed tonsillar tissues, we were curious about their activation status in response to innate immune stimuli, such as Toll-like receptors (TLRs). Therefore, we analyzed the expression profile of TLRs in T-MSCs and stimulated the T-MSCs with TLR agonists. TLR3 stimuli induced C-C chemokine receptor type 6 expression in T-MSCs after 24h. Furthermore, results from cytokine arrays showed increases in epithelial neutrophil-activating peptide-78/C-X-C motif chemokine (CXCL) 5, granulocyte chemotactic protein-2/CXCL6, growth-related oncogene-α/CXCL1, interleukin-8/CXCL8, and interferon gamma-induced protein-10/CXCL10. CD54 expression was also increased after TLR3 stimulation. However, co-culturing T-MSCs with human B cells did not induce B-cell proliferation. This suggests that TLR3 stimulates the differentiation of T-MSCs into FDC-like cells and induces chemokine secretion, possibly by recruiting C-X-C chemokine receptor 2-expressing immune cells. In addition, T-MSCs also appeared to exert immunomodulatory effects by inhibiting B-cell proliferation, possibly by down-regulating CD18.
Collapse
Affiliation(s)
- Jung-Hwa Ryu
- Department of Microbiology, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea
| | - Minhwa Park
- Department of Microbiology, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea
| | - Bo-Kyung Kim
- Department of Microbiology, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea
| | - Kyung-Ha Ryu
- Department of Pediatrics, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea
| | - So-Youn Woo
- Department of Microbiology, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea.
| |
Collapse
|
99
|
Zhang L, Liu D, Pu D, Wang Y, Li L, He Y, Li Y, Li L, Qiu Z, Zhao S, Li W. The role of Toll-like receptor 3 and 4 in regulating the function of mesenchymal stem cells isolated from umbilical cord. Int J Mol Med 2015; 35:1003-10. [PMID: 25695694 DOI: 10.3892/ijmm.2015.2106] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 02/04/2015] [Indexed: 02/05/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have been applied to cell-based therapy due to their multiple differentiation ability, the low expression of co-stimulatory molecules and immunosuppressive properties. Despite their immunomodulatory role, the issue of the survival and permanence of MSCs at the site of injury has not yet been fully resolved. Therefore, in order to improve the therapeutic potential of MSCs, it is important to study the mechanisms mediating the relative instability of MSCs in clinical trials. The Toll-like receptors (TLRs) are an important component of innate and adaptive immune responses. In this study, we demonstrate that the activation of two TLRs, namely TLR3 and TLR4, in human umbilical cord-derived MSCs (UCMSCs) induces the expression of inflammatory markers. In addition, as shown by our results, TLR3 upregulated the expression of stem cell markers, while TLR4 downregulated their expression. The upregulation in the expression of the inflammatory markers did not alter the immune status of the UCMSCs or mediate the immune attack of the MSCs by allogeneic immune cells. We found that the activation of TLR3 inhibited the differentiation of UCMSCs into osteocytes, while that of TLR4 increased this differentiation to a certain extent. Taken together, the results of this study provide a new role for TLR3 and TLR4 as regulators of the biological functions of UCMSCs.
Collapse
Affiliation(s)
- Li Zhang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Dan Liu
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Dan Pu
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yanwen Wang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Li Li
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yanqi He
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yalun Li
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Lei Li
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Zhixin Qiu
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Shuang Zhao
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Weimin Li
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
100
|
Zhang L, Liu D, Pu D, Wang Y, Li L, He Y, Li Y, Li L, Li W. The TLR7 agonist Imiquimod promote the immunogenicity of mesenchymal stem cells. Biol Res 2015; 48:6. [PMID: 25654296 PMCID: PMC4363195 DOI: 10.1186/0717-6287-48-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 01/07/2015] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are considered the best candidate in stem cells therapy due to their multipotent differentiation ability, low expression of co-stimulatory molecules (CD80, CD86, CD34 and HLA-II) and immunosuppression effects on in vivo immune responses. MSCs were now widely used in clinical trials but received no encourage results. The major problem was the fate of engrafted MSCs in vivo could not be defined. Some studies indicated that MSCs could induce immune response and result in the damage and rejection of MSCs. As toll like receptors (TLRs) are important in inducing of immune responses, in this study we study the role of TLR7 in mediating the immune status of MSCs isolated from umbilical cord. RESULTS Our results indicated that TLR7 agonist Imiquimod could increase the proliferation of PBMC isolated from healthy human volunteers and release of lactate dehydrogenase (LDH) in supernatant from PBMC-UCMSCs co-culture system. Flow cytometry and quantitative PCR also confirmed the regulated expression of surface co-stimulatory molecules and pro-inflammatory genes (IL-6, IL-8, IL-12, TGF-β and TNF-α). And the down-regulation expression of stem cell markers also confirmed the loss of stemness of UCMSCs. We also found that the osteo-differentiation ability of UCMSCs was enhanced in the presence of Imiquimod. CONCLUSION To our knowledge, this is the first report that activation of TLR7 pathway increases the immunogenicity of UCMSCs. Extensive researches have now been conducted to study whether the change of immune status will be help in tumor rejection based on the tumor-tropism of MSCs.
Collapse
|