751
|
Su J, Xie C, Fan Y, Cheng W, Hu Y, Huang Q, Shi H, Wang L, Ren J. Interleukin-25 enhances the capacity of mesenchymal stem cells to induce intestinal epithelial cell regeneration. Am J Transl Res 2017; 9:5320-5331. [PMID: 29312486 PMCID: PMC5752884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 11/16/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND This study aimed to investigate the influence of IL-25 on the capacity of mesenchymal stem cells (MSCs) to induce intestinal epithelial cell regeneration. METHODS The CD4+IL-25R+ cells and LGR5+IL-25R+ cells in colonic mucosa of Crohn's disease (CD) patients, ulcerative colitis (UC) patients and healthy controls were detected by immunofluorescence staining, and the CD4+IL-25R+ cells in peripheral blood were detected by flow cytometry. Rat MSCs were separated and stimulated with IL-25. Then, MSCs were further incubated in IL-25-free DMEM for 24 h, and this DMEM was collected as conditioned medium (CM). IEC-6 cells were divided into 3 groups: experimental group (CM and TNF-α), control group (DMEM and TNF-α) and negative control group (DMEM). RESULTS The CD4+IL-25R+ cells and LGR5+IL-25R+ cells significantly increased in the colonic mucosa of active CD patients and UC patients compared with IBD patients in remission and healthy controls. The CD4+IL-25R+ cells reduced in peripheral blood of IBD patients, which was inversely correlated with inflammatory markers (ESR and CRP). CM facilitated the migration and proliferation of IEC-6 cells in the presence of TNF-α. The protein expression of AKT, p38 and ERK increased in IEC-6 cells after treatment with CM and TNF-α. CONCLUSION IL-25R is involved in Th-related mucosal inflammation and proliferation of intestinal stem cells in IBD. IL-25 enhances the capacity of MSC to induce intestinal epithelial cell regeneration, and MSC therapy with IL-25 may be a new direction for IBD treatment.
Collapse
Affiliation(s)
- Jingling Su
- Department of Gastroenterology, Affiliated Zhongshan Hospital of Xiamen UniversityXiamen 361004, Fujian, China
| | - Chenxi Xie
- Department of Gastroenterology, Affiliated Zhongshan Hospital of Xiamen UniversityXiamen 361004, Fujian, China
| | - Yanyun Fan
- Department of Gastroenterology, Affiliated Zhongshan Hospital of Xiamen UniversityXiamen 361004, Fujian, China
| | - Weizi Cheng
- Department of Gastroenterology, Affiliated Zhongshan Hospital of Xiamen UniversityXiamen 361004, Fujian, China
| | - Yiqun Hu
- Department of Gastroenterology, Affiliated Zhongshan Hospital of Xiamen UniversityXiamen 361004, Fujian, China
| | - Qingwen Huang
- Department of Gastroenterology, Affiliated Zhongshan Hospital of Xiamen UniversityXiamen 361004, Fujian, China
| | - Huaxiu Shi
- Department of Gastroenterology, Affiliated Zhongshan Hospital of Xiamen UniversityXiamen 361004, Fujian, China
| | - Lin Wang
- Department of Gastroenterology, Affiliated Zhongshan Hospital of Xiamen UniversityXiamen 361004, Fujian, China
| | - Jianlin Ren
- Department of Gastroenterology, Affiliated Zhongshan Hospital of Xiamen UniversityXiamen 361004, Fujian, China
| |
Collapse
|
752
|
Arango JC, Puerta-Arias JD, Pino-Tamayo PA, Arboleda-Toro D, González Á. Bone marrow–derived mesenchymal stem cells transplantation alters the course of experimental paracoccidioidomycosis by exacerbating the chronic pulmonary inflammatory response. Med Mycol 2017; 56:884-895. [DOI: 10.1093/mmy/myx128] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 10/24/2017] [Indexed: 12/15/2022] Open
Affiliation(s)
- Julián Camilo Arango
- Medical and Experimental Mycology Group, Corporación para Investigaciones Biológicas (CIB), Universidad de Antioquia, Medellin, Colombia
- Microbiology School, Universidad de Antioquia, Medellin Colombia
| | - Juan David Puerta-Arias
- Medical and Experimental Mycology Group, Corporación para Investigaciones Biológicas (CIB), Universidad de Antioquia, Medellin, Colombia
| | - Paula Andrea Pino-Tamayo
- Medical and Experimental Mycology Group, Corporación para Investigaciones Biológicas (CIB), Universidad de Antioquia, Medellin, Colombia
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, USA
| | | | - Ángel González
- Microbiology School, Universidad de Antioquia, Medellin Colombia
- Basic and Applied Microbiology Research Group (MICROBA), Universidad de Antioquia, Medellin, Colombia
| |
Collapse
|
753
|
Mesenchymal Stem Cell Benefits Observed in Bone Marrow Failure and Acquired Aplastic Anemia. Stem Cells Int 2017; 2017:8076529. [PMID: 29333168 PMCID: PMC5733198 DOI: 10.1155/2017/8076529] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 08/15/2017] [Accepted: 09/10/2017] [Indexed: 12/16/2022] Open
Abstract
Acquired aplastic anemia (AA) is a type of bone marrow failure (BMF) syndrome characterized by partial or total bone marrow (BM) destruction resulting in peripheral blood (PB) pancytopenia, which is the reduction in the number of red blood cells (RBC) and white blood cells (WBC), as well as platelets (PLT). The first-line treatment option of AA is given by hematopoietic stem cell (HSCs) transplant and/or immunosuppressive (IS) drug administration. Some patients did not respond to the treatment and remain pancytopenic following IS drugs. The studies are in progress to test the efficacy of adoptive cellular therapies as mesenchymal stem cells (MSCs), which confer low immunogenicity and are reliable allogeneic transplants in refractory severe aplastic anemia (SAA) cases. Moreover, bone marrow stromal cells (BMSC) constitute an essential component of the hematopoietic niche, responsible for stimulating and enhancing the proliferation of HSCs by secreting regulatory molecules and cytokines, providing stimulus to natural BM microenvironment for hematopoiesis. This review summarizes scientific evidences of the hematopoiesis improvements after MSC transplant, observed in acquired AA/BMF animal models as well as in patients with acquired AA. Additionally, we discuss the direct and indirect contribution of MSCs to the pathogenesis of acquired AA.
Collapse
|
754
|
Kim D, Cho GS, Han C, Park DH, Park HK, Woo DH, Kim JH. Current Understanding of Stem Cell and Secretome Therapies in Liver Diseases. Tissue Eng Regen Med 2017; 14:653-665. [PMID: 30603518 PMCID: PMC6171672 DOI: 10.1007/s13770-017-0093-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 10/23/2017] [Accepted: 10/29/2017] [Indexed: 12/14/2022] Open
Abstract
Liver failure is one of the main risks of death worldwide, and it originates from repetitive injuries and inflammations of liver tissues, which finally leads to the liver cirrhosis or cancer. Currently, liver transplantation is the only effective treatment for the liver diseases although it has a limitation due to donor scarcity. Alternatively, cell therapy to regenerate and reconstruct the damaged liver has been suggested to overcome the current limitation of liver disease cures. Several transplantable cell types could be utilized for recovering liver functions in injured liver, including bone marrow cells, mesenchymal stem cells, hematopoietic stem cells, macrophages, and stem cell-derived hepatocytes. Furthermore, paracrine effects of transplanted cells have been suggested as a new paradigm for liver disease cures, and this application would be a new strategy to cure liver failures. Therefore, here we reviewed the current status and challenges of therapy using stem cells for liver disease treatments.
Collapse
Affiliation(s)
- Dongkyu Kim
- Laboratory of Stem Cells, NEXEL Co., Ltd., 9th Floor, 21 Wangsan-ro, Dongdaemun-gu, Seoul, 02580 Korea
| | - Gun-Sik Cho
- Laboratory of Stem Cells, NEXEL Co., Ltd., 9th Floor, 21 Wangsan-ro, Dongdaemun-gu, Seoul, 02580 Korea
| | - Choongseong Han
- Laboratory of Stem Cells, NEXEL Co., Ltd., 9th Floor, 21 Wangsan-ro, Dongdaemun-gu, Seoul, 02580 Korea
- Department of Oral Medicine and Oral Diagnosis, School of Dentistry and Dental Research Institute, Seoul National University, #101 Daehak-ro, Jongro-gu, Seoul, 03080 Korea
| | - Dong-Hyuk Park
- Department of Neurosurgery, Korea University Medical Center, Anam Hospital, Korea University College of Medicine, 73 Inchonro, Sungbuk-gu, Seoul, 02841 Korea
| | - Hee-Kyung Park
- Department of Oral Medicine and Oral Diagnosis, School of Dentistry and Dental Research Institute, Seoul National University, #101 Daehak-ro, Jongro-gu, Seoul, 03080 Korea
| | - Dong-Hun Woo
- Laboratory of Stem Cells, NEXEL Co., Ltd., 9th Floor, 21 Wangsan-ro, Dongdaemun-gu, Seoul, 02580 Korea
| | - Jong-Hoon Kim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Science Campus, Korea University, 145 Anam-ro, Seongbu-gu, Seoul, 02841 Korea
| |
Collapse
|
755
|
Bishop ES, Mostafa S, Pakvasa M, Luu HH, Lee MJ, Wolf JM, Ameer GA, He TC, Reid RR. 3-D bioprinting technologies in tissue engineering and regenerative medicine: Current and future trends. Genes Dis 2017; 4:185-195. [PMID: 29911158 PMCID: PMC6003668 DOI: 10.1016/j.gendis.2017.10.002] [Citation(s) in RCA: 355] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Advances in three-dimensional (3D) printing have increased feasibility towards the synthesis of living tissues. Known as 3D bioprinting, this technology involves the precise layering of cells, biologic scaffolds, and growth factors with the goal of creating bioidentical tissue for a variety of uses. Early successes have demonstrated distinct advantages over conventional tissue engineering strategies. Not surprisingly, there are current challenges to address before 3D bioprinting becomes clinically relevant. Here we provide an overview of 3D bioprinting technology and discuss key advances, clinical applications, and current limitations. While 3D bioprinting is a relatively novel tissue engineering strategy, it holds great potential to play a key role in personalized medicine.
Collapse
Affiliation(s)
- Elliot S Bishop
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medicine, Chicago, IL 60637, USA.,Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Sami Mostafa
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
| | - Mikhail Pakvasa
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
| | - Hue H Luu
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Michael J Lee
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jennifer Moriatis Wolf
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Guillermo A Ameer
- Biomedical Engineering Department, Northwestern University, Evanston, IL 60208, USA.,Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60616, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Russell R Reid
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medicine, Chicago, IL 60637, USA
| |
Collapse
|
756
|
Janczewski AM, Wojtkiewicz J, Malinowska E, Doboszyńska A. Can Youthful Mesenchymal Stem Cells from Wharton's Jelly Bring a Breath of Fresh Air for COPD? Int J Mol Sci 2017; 18:ijms18112449. [PMID: 29156550 PMCID: PMC5713416 DOI: 10.3390/ijms18112449] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/13/2017] [Accepted: 11/14/2017] [Indexed: 12/13/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a major global cause of morbidity and mortality, projected to become the 3rd cause of disease mortality worldwide by 2020. COPD is characterized by persistent and not fully reversible airflow limitation that is usually progressive and is associated with an abnormal chronic inflammatory response of the lung to noxious agents including cigarette smoke. Currently available therapeutic strategies aim to ease COPD symptoms but cannot prevent its progress or regenerate physiological lung structure or function. The urgently needed new approaches for the treatment of COPD include stem cell therapies among which transplantation of mesenchymal stem cells derived from Wharton’s jelly (WJ-MSCs) emerges as a promising therapeutic strategy because of the unique properties of these cells. The present review discusses the main biological properties of WJ-MSCs pertinent to their potential application for the treatment of COPD in the context of COPD pathomechanisms with emphasis on chronic immune inflammatory processes that play key roles in the development and progression of COPD.
Collapse
Affiliation(s)
- Andrzej M Janczewski
- Department of Pulmonology, Faculty of Heath Sciences, University of Warmia and Mazury in Olsztyn, Jagiellońska 78, 10-357 Olsztyn, Poland.
| | - Joanna Wojtkiewicz
- Department of Pathophysiology, Faculty of Medicine, University of Warmia and Mazury in Olsztyn, Warszawska 30, 10-082 Olsztyn, Poland.
- Laboratory for Regenerative Medicine, Faculty of Medicine, University of Warmia and Mazury in Olsztyn, Warszawska 30, 10-082 Olsztyn, Poland.
- Foundation for the Nerve Cells Regeneration, Warszawska 30, 10-082 Olsztyn, Poland.
| | - Ewa Malinowska
- Department of Pulmonology, Faculty of Heath Sciences, University of Warmia and Mazury in Olsztyn, Jagiellońska 78, 10-357 Olsztyn, Poland.
| | - Anna Doboszyńska
- Department of Pulmonology, Faculty of Heath Sciences, University of Warmia and Mazury in Olsztyn, Jagiellońska 78, 10-357 Olsztyn, Poland.
| |
Collapse
|
757
|
Chow L, Johnson V, Regan D, Wheat W, Webb S, Koch P, Dow S. Safety and immune regulatory properties of canine induced pluripotent stem cell-derived mesenchymal stem cells. Stem Cell Res 2017; 25:221-232. [PMID: 29172152 DOI: 10.1016/j.scr.2017.11.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 11/08/2017] [Accepted: 11/10/2017] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) exhibit broad immune modulatory activity in vivo and can suppress T cell proliferation and dendritic cell activation in vitro. Currently, most MSC for clinical usage are derived from younger donors, due to ease of procurement and to the superior immune modulatory activity. However, the use of MSC from multiple unrelated donors makes it difficult to standardize study results and compare outcomes between different clinical trials. One solution is the use of MSC derived from induced pluripotent stem cells (iPSC); as iPSC-derived MSC have nearly unlimited proliferative potential and exhibit in vitro phenotypic stability. Given the value of dogs as a spontaneous disease model for pre-clinical evaluation of stem cell therapeutics, we investigated the functional properties of canine iPSC-derived MSC (iMSC), including immune modulatory properties and potential for teratoma formation. We found that canine iMSC downregulated expression of pluripotency genes and appeared morphologically similar to conventional MSC. Importantly, iMSC retained a stable phenotype after multiple passages, did not form teratomas in immune deficient mice, and did not induce tumor formation in dogs following systemic injection. We concluded therefore that iMSC were phenotypically stable, immunologically potent, safe with respect to tumor formation, and represented an important new source of cells for therapeutic modulation of inflammatory disorders.
Collapse
Affiliation(s)
- Lyndah Chow
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine, Colorado State University, Ft. Collins, CO, United States
| | - Valerie Johnson
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine, Colorado State University, Ft. Collins, CO, United States; Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine, Colorado State University, Ft. Collins, CO, United States
| | - Dan Regan
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine, Colorado State University, Ft. Collins, CO, United States; Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine, Colorado State University, Ft. Collins, CO, United States
| | - William Wheat
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine, Colorado State University, Ft. Collins, CO, United States
| | - Saiphone Webb
- Gates Center for Regenerative Medicine, Department of Dermatology, University of Colorado Denver, Aurora, CO, United States
| | - Peter Koch
- Gates Center for Regenerative Medicine, Department of Dermatology, University of Colorado Denver, Aurora, CO, United States
| | - Steven Dow
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine, Colorado State University, Ft. Collins, CO, United States.
| |
Collapse
|
758
|
Khezri S, Abtahi Froushani SM, Shahmoradi M. Nicotine Augments the Beneficial Effects of Mesenchymal Stem Cell-based Therapy in Rat Model of Multiple Sclerosis. Immunol Invest 2017; 47:113-124. [DOI: 10.1080/08820139.2017.1391841] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Shiva Khezri
- Department of biology, Faculty of science, Urmia University, Urmia, Iran
| | | | - Mozhgan Shahmoradi
- Department of biology, Faculty of science, Urmia University, Urmia, Iran
| |
Collapse
|
759
|
Lopez-Mendez TB, Santos-Vizcaino E, Blanco FJ, Pedraz JL, Hernandez RM, Orive G. Improved control over MSCs behavior within 3D matrices by using different cell loads in both in vitro and in vivo environments. Int J Pharm 2017; 533:62-72. [DOI: 10.1016/j.ijpharm.2017.09.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/05/2017] [Accepted: 09/07/2017] [Indexed: 12/19/2022]
|
760
|
Arango JC, Puerta-Arias JD, Pino-Tamayo PA, Salazar-Peláez LM, Rojas M, González Á. Impaired anti-fibrotic effect of bone marrow-derived mesenchymal stem cell in a mouse model of pulmonary paracoccidioidomycosis. PLoS Negl Trop Dis 2017; 11:e0006006. [PMID: 29040281 PMCID: PMC5659794 DOI: 10.1371/journal.pntd.0006006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 10/27/2017] [Accepted: 10/02/2017] [Indexed: 02/06/2023] Open
Abstract
Bone marrow-derived mesenchymal stem cells (BMMSCs) have been consider as a promising therapy in fibrotic diseases. Experimental models suggest that BMMSCs may be used as an alternative therapy to treat chemical- or physical-induced pulmonary fibrosis. We investigated the anti-fibrotic potential of BMMSCs in an experimental model of lung fibrosis by infection with Paracoccidioides brasiliensis. BMMSCs were isolated and purified from BALB/c mice using standardized methods. BALB/c male mice were inoculated by intranasal infection of 1.5x106P. brasiliensis yeasts. Then, 1x106 BMMSCs were administered intra venous at 8th week post-infection (p.i.). An additional group of mice was treated with itraconazole (ITC) two weeks before BMMSCs administration. Animals were sacrificed at 12th week p.i. Histopathological examination, fibrocytes counts, soluble collagen and fibrosis-related genes expression in lungs were evaluated. Additionally, human fibroblasts were treated with homogenized lung supernatants (HLS) to determine induction of collagen expression. Histological analysis showed an increase of granulomatous inflammatory areas in BMMSCs-treated mice. A significant increase of fibrocytes count, soluble collagen and collagen-3α1, TGF-β3, MMP-8 and MMP-15 genes expression were also observed in those mice. Interestingly, when combined therapy BMMSCs/ITC was used there is a decrease of TIMP-1 and MMP-13 gene expression in infected mice. Finally, human fibroblasts stimulated with HLS from infected and BMMSCs-transplanted mice showed a higher expression of collagen I. In conclusion, our findings indicate that late infusion of BMMSCs into mice infected with P. brasiliensis does not have any anti-fibrotic effect; possibly because their interaction with the fungus promotes collagen expression and tissue remodeling. This is the first study that evaluates the effect of BMMSCs therapy for lung fibrosis induced by the fungal pathogen Paracoccidioides brasiliensis, the causative agent of paracoccidioidomycosis, one of the most important systemic endemic mycosis diagnosed in South America and Central America. Our findings showed an impaired anti-fibrotic effect of BMMSCs transplantation. This effect could be triggered by either the chronic inflammatory microenvironment induced by P. brasiliensis or by a direct interaction between BMMSCs and the fungus, resulting in an exacerbation of the pulmonary fibrosis. In fact, the pro-fibrotic effect exerted by BMMSCs was toned-down by the usage of the antifungal ITC.
Collapse
Affiliation(s)
- Julián Camilo Arango
- Medical and Experimental Mycology Group, Corporación para Investigaciones Biológicas (CIB)–Universidad de Antioquia, Medellín, Colombia
- School of Microbiology, Universidad de Antioquia, Medellín, Colombia
| | - Juan David Puerta-Arias
- Medical and Experimental Mycology Group, Corporación para Investigaciones Biológicas (CIB)–Universidad de Antioquia, Medellín, Colombia
| | - Paula Andrea Pino-Tamayo
- Medical and Experimental Mycology Group, Corporación para Investigaciones Biológicas (CIB)–Universidad de Antioquia, Medellín, Colombia
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, New York, Unites States of America
| | | | - Mauricio Rojas
- Dorothy P. & Richard P. Simmons Center for Interstitial Lung Disease, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, Unites States of America
| | - Ángel González
- School of Microbiology, Universidad de Antioquia, Medellín, Colombia
- Basic and Applied Microbiology Research Group (MICROBA), Universidad de Antioquia, Medellín, Colombia
- * E-mail:
| |
Collapse
|
761
|
Wiegner R, Rudhart NE, Barth E, Gebhard F, Lampl L, Huber-Lang MS, Brenner RE. Mesenchymal stem cells in peripheral blood of severely injured patients. Eur J Trauma Emerg Surg 2017; 44:627-636. [PMID: 28986662 DOI: 10.1007/s00068-017-0849-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 09/25/2017] [Indexed: 12/15/2022]
Abstract
PURPOSE Mesenchymal stem cells (MSCs) are primarily stromal cells present in bone marrow and other tissues that are crucial for tissue regeneration and can be mobilized into peripheral blood after different types of organ damage. However, little is known about MSC appearance in blood in the setting of polytrauma. METHODS We conducted a monocentered and longitudinal observational clinical study in 11 polytraumatized patients with an injury severity score (ISS) ≥ 24 to determine the numbers of MSCs in peripheral blood. Blood was collected from healthy volunteers and patients after polytrauma in the emergency room and 4, 12, 24, 48 h, 5 and 10 day later, and cells carrying MSC-surface markers (negative for CD45, positive for CD29, CD73, CD90, CD105, and CD166 in different combinations also employing the more stringent markers STRO1 and MSCA1) were detected and characterized using flow cytometry. Relative numbers of MSC-like cells were correlated with clinical parameters to evaluate if specific injury patterns had an influence on their presence in the blood cell pool. RESULTS We were able to detect MSC marker-positive cells in both cohorts; however, the percentage of those cells present in the blood of patients during the first 10 day after injury was mostly similar to healthy volunteers, and significantly lowers starting at 4 h post trauma for one marker combination when compared to controls. Furthermore, the presence of a pelvis fracture was partly correlated with reduced relative numbers of MSC-like cells detectable in blood. CONCLUSIONS Polytrauma in humans was associated with partly reduced relative numbers of MSC-like cells detected in peripheral blood in the time course after injury. Further studies need to define if this reduction was due to lower mobilization from the bone marrow or to active migration to the sites of injury.
Collapse
Affiliation(s)
- R Wiegner
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081, Ulm, Germany
| | - N-E Rudhart
- Department of Orthopedics, Division for Biochemistry of Joint and Connective Tissue Diseases, University of Ulm, Oberer Eselsberg 45, 89081, Ulm, Germany
| | - E Barth
- Department of Anesthesiology, University Hospital of Ulm, 89081, Ulm, Germany
| | - F Gebhard
- Department of Orthopedic Trauma, Hand-, Plastic- and Reconstructive Surgery, University Hospital of Ulm, 89081, Ulm, Germany
| | - L Lampl
- Department of Anesthesiology, Military Hospital Ulm, 89081, Ulm, Germany
| | - M S Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081, Ulm, Germany
| | - R E Brenner
- Department of Orthopedics, Division for Biochemistry of Joint and Connective Tissue Diseases, University of Ulm, Oberer Eselsberg 45, 89081, Ulm, Germany.
| |
Collapse
|
762
|
Zhou Y, Tsai TL, Li WJ. Strategies to retain properties of bone marrow-derived mesenchymal stem cells ex vivo. Ann N Y Acad Sci 2017; 1409:3-17. [PMID: 28984359 DOI: 10.1111/nyas.13451] [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: 04/14/2017] [Revised: 07/05/2017] [Accepted: 07/18/2017] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSCs) have been extensively used for cell therapies and tissue engineering. The current MSC strategy requires a large quantity of cells for such applications, which can be achieved through cell expansion in culture. In the body, stem cell fate is largely determined by their microenvironment, known as the niche. The complex and dynamic stem cell niche provides physical, mechanical, and chemical cues to collaboratively regulate cell activities. It remains a great challenge to maintain the properties of MSCs in culture. Constructing a microenvironment as an engineered stem cell niche in culture to maintain MSC phenotypes, properties, and functions is a viable strategy to address the issue. Here, we review the current understanding of MSC behavior in the bone marrow niche, describe different strategies to engineer an in vitro microenvironment for maintaining MSC properties and functions, and discuss previous findings on environmental factors critical to the modulation of MSC activities in engineered microenvironments.
Collapse
Affiliation(s)
- Yaxian Zhou
- Laboratory of Musculoskeletal Biology and Regenerative Medicine, Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin
| | - Tsung-Lin Tsai
- Laboratory of Musculoskeletal Biology and Regenerative Medicine, Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin
| | - Wan-Ju Li
- Laboratory of Musculoskeletal Biology and Regenerative Medicine, Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
763
|
Baldari S, Di Rocco G, Piccoli M, Pozzobon M, Muraca M, Toietta G. Challenges and Strategies for Improving the Regenerative Effects of Mesenchymal Stromal Cell-Based Therapies. Int J Mol Sci 2017; 18:2087. [PMID: 28974046 PMCID: PMC5666769 DOI: 10.3390/ijms18102087] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 09/18/2017] [Accepted: 09/28/2017] [Indexed: 12/11/2022] Open
Abstract
Cell-based therapies have the potential to revolutionize current treatments for diseases with high prevalence and related economic and social burden. Unfortunately, clinical trials have made only modest improvements in restoring normal function to degenerating tissues. This limitation is due, at least in part, to the death of transplanted cells within a few hours after transplant due to a combination of mechanical, cellular, and host factors. In particular, mechanical stress during implantation, extracellular matrix loss upon delivery, nutrient and oxygen deprivation at the recipient site, and host inflammatory response are detrimental factors limiting long-term transplanted cell survival. The beneficial effect of cell therapy for regenerative medicine ultimately depends on the number of administered cells reaching the target tissue, their viability, and their promotion of tissue regeneration. Therefore, strategies aiming at improving viable cell engraftment are crucial for regenerative medicine. Here we review the major factors that hamper successful cell engraftment and the strategies that have been studied to enhance the beneficial effects of cell therapy. Moreover, we provide a perspective on whether mesenchymal stromal cell-derived extracellular vesicle delivery, as a cell-free regenerative approach, may circumvent current cell therapy limitations.
Collapse
Affiliation(s)
- Silvia Baldari
- Department of Research, Advanced Diagnostic, and Technological Innovation, Regina Elena National Cancer Institute, via E. Chianesi 53, Rome 00144, Italy.
| | - Giuliana Di Rocco
- Department of Research, Advanced Diagnostic, and Technological Innovation, Regina Elena National Cancer Institute, via E. Chianesi 53, Rome 00144, Italy.
| | - Martina Piccoli
- Stem Cells and Regenerative Medicine Laboratory, Foundation Institute of Pediatric Research "Città della Speranza", corso Stati Uniti 4, Padova 35127, Italy.
| | - Michela Pozzobon
- Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, Padova 35128, Italy.
| | - Maurizio Muraca
- Department of Women's and Children's Health, University of Padova, Via Giustiniani 3, Padova 35128, Italy.
| | - Gabriele Toietta
- Department of Research, Advanced Diagnostic, and Technological Innovation, Regina Elena National Cancer Institute, via E. Chianesi 53, Rome 00144, Italy.
| |
Collapse
|
764
|
Sonic hedgehog (SHH) signaling improves the angiogenic potential of Wharton's jelly-derived mesenchymal stem cells (WJ-MSC). Stem Cell Res Ther 2017; 8:203. [PMID: 28962669 PMCID: PMC5622478 DOI: 10.1186/s13287-017-0653-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 08/24/2017] [Accepted: 08/29/2017] [Indexed: 02/07/2023] Open
Abstract
Background Wharton’s jelly-derived mesenchymal stem cells (WJ-MSC) show remarkable therapeutic potential to repair tissue upon injury via paracrine signaling by secreting diverse trophic factors that promote angiogenesis. However, the mechanisms and signaling pathways that regulate the induction of these specific factors are still mostly unknown. Emerging evidence suggests that Sonic hedgehog (SHH) plays a central role in angiogenesis and tissue maintenance. However, its contribution to the angiogenic potential of MSC has not been fully addressed. The aim of this work was to characterize the expression of the SHH pathway components in WJ-MSC primary cultures and to evaluate their angiogenic responsiveness to SHH signaling. Methods Primary cell cultures obtained from human umbilical cords were treated with pharmacological modulators of the SHH pathway. We evaluated the modulation of diverse trophic factors in cell lysates, conditioned medium, and functional in vitro assays. In addition, we determined the angiogenic potential of the SHH pathway in the chicken chorioallantoic membrane, an in vivo model. Results Our results show that WJ-MSC express components of the canonical SHH pathway and are activated by its signaling. In fact, we provide evidence of basal autocrine/paracrine SHH signaling in WJ-MSC. SHH pathway stimulation promotes the secretion of angiogenic factors such as activin A, angiogenin, angiopoietin 1, granulocyte-macrophage colony-stimulating factor, matrix metallometallopeptidase -9, and urokinase-type plasminogen activator, enhancing the pro-angiogenic capabilities of WJ-MSC both in vitro and in vivo. Conclusion WJ-MSC are a cell population responsive to SHH pathway stimulation. Basal SHH signaling is in part responsible for the angiogenic inductive properties of WJ-MSC. Overall, exogenous activation of the SHH pathway enhances the angiogenic properties of WJ-MSC, making this cell population an ideal target for treating tissue injury. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0653-8) contains supplementary material, which is available to authorized users.
Collapse
|
765
|
Muslimov AR, Timin AS, Petrova AV, Epifanovskaya OS, Shakirova AI, Lepik KV, Gorshkov A, Il'inskaja EV, Vasin AV, Afanasyev BV, Fehse B, Sukhorukov GB. Mesenchymal Stem Cells Engineering: Microcapsules-Assisted Gene Transfection and Magnetic Cell Separation. ACS Biomater Sci Eng 2017; 3:2314-2324. [PMID: 33445290 DOI: 10.1021/acsbiomaterials.7b00482] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Stem cell engineering-the manipulation and functionalization of stem cells involving genetic modification-can significantly expand their applicability for cell therapy in humans. Toward this aim, reliable, standardized, and cost-effective methods for cell manipulation are required. Here we explore the potential of magnetic multilayer capsules to serve as a universal platform for nonviral gene transfer, stem cell magnetization, and magnetic cell separation to improve gene transfer efficiency. In particular, the following experiments were performed: (i) a study of the process of internalization of magnetic capsules into stem cells, including capsule co-localization with established markers of endo-lysosomal pathway; (ii) characterization and quantification of capsule uptake with confocal microscopy, electron microscopy, and flow cytometry; (iii) intracellular delivery of messenger RNA and separation of gene-modified cells by magnetic cell sorting (MACS); and (iv) analysis of the influence of capsules on cell proliferation potential. Importantly, based on the internalization of magnetic capsules, transfected cells became susceptible to external magnetic fields, which made it easy to enrich gene-modified cells using MACS (purity ∼95%), and also to influence their migration behavior. In summary, our results underline the high potential of magnetic capsules in stem cell functionalization, namely (i) to increase gene-transfer efficiency and (ii) to facilitate enrichment and targeting of transfected cells. Finally, we did not observe a negative impact of the capsules used on the proliferative capacity of stem cells, proving their high biocompatibility.
Collapse
Affiliation(s)
- Albert R Muslimov
- First Pavlov State Medical University of St. Petersburg, Lev Tolstoy str., 6/8, 197022 St. Petersburg, Russian Federation.,Research Institute of Influenza, Popova str., 15/17, 197376 Saint-Petersburg, Russian Federation.,RASA center in St. Petersburg, Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya, 29, 195251 St. Petersburg, Russian Federation
| | - Alexander S Timin
- First Pavlov State Medical University of St. Petersburg, Lev Tolstoy str., 6/8, 197022 St. Petersburg, Russian Federation.,RASA Center in Tomsk, Tomsk Polytechnic University, pros. Lenina, 30, 634050 Tomsk, Russian Federation
| | - Aleksandra V Petrova
- Research Institute of Influenza, Popova str., 15/17, 197376 Saint-Petersburg, Russian Federation.,Department of Molecular Biology, Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya, 29, 195251 St. Petersburg, Russian Federation
| | - Olga S Epifanovskaya
- First Pavlov State Medical University of St. Petersburg, Lev Tolstoy str., 6/8, 197022 St. Petersburg, Russian Federation
| | - Alena I Shakirova
- First Pavlov State Medical University of St. Petersburg, Lev Tolstoy str., 6/8, 197022 St. Petersburg, Russian Federation
| | - Kirill V Lepik
- First Pavlov State Medical University of St. Petersburg, Lev Tolstoy str., 6/8, 197022 St. Petersburg, Russian Federation.,RASA center in St. Petersburg, Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya, 29, 195251 St. Petersburg, Russian Federation
| | - Andrey Gorshkov
- Research Institute of Influenza, Popova str., 15/17, 197376 Saint-Petersburg, Russian Federation
| | - Eugenia V Il'inskaja
- Research Institute of Influenza, Popova str., 15/17, 197376 Saint-Petersburg, Russian Federation
| | - Andrey V Vasin
- Research Institute of Influenza, Popova str., 15/17, 197376 Saint-Petersburg, Russian Federation.,Department of Molecular Biology, Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya, 29, 195251 St. Petersburg, Russian Federation
| | - Boris V Afanasyev
- First Pavlov State Medical University of St. Petersburg, Lev Tolstoy str., 6/8, 197022 St. Petersburg, Russian Federation
| | - Boris Fehse
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, 20246, Martinistraße 52, 20251 Hamburg, Germany
| | - Gleb B Sukhorukov
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, United Kingdom
| |
Collapse
|
766
|
Cheng W, Su J, Hu Y, Huang Q, Shi H, Wang L, Ren J. Interleukin-25 primed mesenchymal stem cells achieve better therapeutic effects on dextran sulfate sodium-induced colitis via inhibiting Th17 immune response and inducing T regulatory cell phenotype. Am J Transl Res 2017; 9:4149-4160. [PMID: 28979689 PMCID: PMC5622258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Accepted: 08/17/2017] [Indexed: 06/07/2023]
Abstract
AIM This study aimed to investigate the anti-inflammatory mechanism of IL-25 mediated mesenchymal stem cells (MSC) treatment for inflammatory bowel disease (IBD) in a DSS-induced rat colitis model. METHODS Rats with DSS-induced colitis were divided into control and treatment groups: normal control group (rats fed with water), DSS group (rats fed with DSS solution), MSC group (DSS-treated rats injected intravenously with GFP-MSCs), IL-25-MSC group (DSS-treated rats injected intravenously with IL-25 primed GFP-MSCs), and mesalazine group (DSS-treated rats fed with mesalazine). RESULTS In IL-25-MSC group, therapeutic efficacy (clinical symptoms) was better than in MSC group, but comparable to mesalazine group. In IL-25-MSC group and mesalazine group, fewer infiltrating inflammatory cells and lower pathological score were observed in the intestine. The FOXP3+ cells and IL-4+ cells decreased, but IL-17A+ cells and IFN-γ+ cells increased in the peripheral blood and colonic mucosa after DSS induced colitis, and these phenomena were reversed by MSC or mesalazine treatment. IL-17A+ cells reduced and FOXP3+ cells increased in IL-25-MSC group as compared with MSC group. The expressions of Ki67 and LGR5 were significantly elevated in MSC treatment groups as compared with normal control group, DSS group, and mesalazine group. Definite GFP positive cells were not observed in the intestine of MSC-treated rats. CONCLUSION IL-25 primed MSCs exert improved therapeutic effects on the intestinal inflammation of IBD rats which may be related to the inhibition of Th17 immune response and induction of T Regulatory cell phenotype. Thus, IL-25 may be an attractive candidate for MSC-based therapy of IBD.
Collapse
|
767
|
Targeting Inflammation in Rotator Cuff Tendon Degeneration and Repair. TECHNIQUES IN SHOULDER AND ELBOW SURGERY 2017; 18:84-90. [PMID: 28947893 DOI: 10.1097/bte.0000000000000124] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Rotator cuff degeneration is a common affliction that results in pain and disability. Tendinopathy was historically classified with or without the involvement of the immune system. However, technological advancements in screening have shown that the immune system is both present and active in all forms of tendinopathy. During injury and healing, the coordinated effort of numerous immune cell populations work with the resident stromal cells to break down damaged tissues and stimulate remodeling. These cells deploy a wide array of tools, including phagocytosis, enzyme secretion, and chemotactic gradients to direct these processes. Yet, there remains a knowledge gap in our understanding of the sequence of critical events and regulatory factors that mediate this is process in injury and healing. Furthermore, current treatments do not specifically target inflammation at the molecular level. Typical regimens include non-steroidal anti-inflammatory drugs or corticosteroids; however, researchers have found irrevocable functional deficits following treatment, and have disputed their long-term efficacy. Therefore, developing therapeutics that specifically consider the nuances of the immune system are necessary to improve patient outcomes.
Collapse
|
768
|
Abstract
BACKGROUND Allogeneic mesenchymal stem cells (MSCs) show great potential for the treatment of military and civilian trauma based on their reduced immunogenicity and ability to modulate inflammation and immune function in the recipient. Although generally considered to be safe, MSCs express tissue factor (TF), a potent activator of coagulation. In the current study, we evaluated multiple MSC populations for tissue factor expression and procoagulant activity to characterize safety considerations for systemic use of MSCs in trauma patients who may have altered coagulation homeostasis. METHODS Multiple MSC populations derived from either human adipose tissue or bone marrow were expanded in the recommended stem cell media. Stem cell identity was confirmed using a well-characterized panel of positive and negative markers. Tissue factor expression on the cell surface was evaluated by flow cytometry with anti-CD142 antibody. Effects on blood coagulation were determined by thromboelastography and calibrated automated thrombogram assays using platelet-poor plasma or whole blood. RESULTS Mesenchymal stem cells express tissue factor on their surfaces and are procoagulant in the presence of blood or plasma. The adipose-derived MSCs (Ad-MSC) evaluated were more procoagulant and expressed more tissue factor than bone marrow MSCs (BM-MSCs), which showed a greater variability in TF expression. Bone marrow MSCs were identified that exhibited low procoagulant activity, whereas all Ad-MSCs examined exhibited high procoagulant activity. The percentage of cells in a given population expressing surface tissue factor correlates roughly with functional procoagulant activity. Mesenchymal stem cell tissue factor expression and procoagulant activity change over time in culture. CONCLUSIONS All MSC populations are not equivalent; care should be taken to select cells for clinical use that minimize potential safety problems and maximize chance of patient benefit. Adipose-derived MSCs seem more consistently procoagulant than BM-MSCs, presenting a potential safety concern for systemic administration in coagulopathic patients. Donor variation exists between different cell populations, and culture handling conditions may also determine coagulation activity. Cells must be routinely monitored during preparation to ensure that they retain the desired characteristics before patient administration.
Collapse
|
769
|
Luk F, Carreras-Planella L, Korevaar SS, de Witte SFH, Borràs FE, Betjes MGH, Baan CC, Hoogduijn MJ, Franquesa M. Inflammatory Conditions Dictate the Effect of Mesenchymal Stem or Stromal Cells on B Cell Function. Front Immunol 2017; 8:1042. [PMID: 28894451 PMCID: PMC5581385 DOI: 10.3389/fimmu.2017.01042] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 08/11/2017] [Indexed: 12/16/2022] Open
Abstract
The immunomodulatory capacity of mesenchymal stem or stromal cells (MSC) makes them a promising tool for treatment of immune disease and organ transplantation. The effects of MSC on B cells are characterized by an abrogation of plasmablast formation and induction of regulatory B cells (Bregs). It is, however, unknown how MSC interact with B cells under inflammatory conditions. In this study, adipose tissue-derived MSC were pretreated with 50 ng/ml IFN-γ for 96 h (MSC–IFN-γ) to simulate inflammatory conditions. Mature B cells were obtained from spleens by CD43− selection. B cells were co-cultured with MSC and stimulated with anti-IgM, anti-CD40, and IL-2; and after 7 days, B cell proliferation, phenotype, Immunoglobulin-G (IgG), and IL-10 production were analyzed. MSC did not inhibit B cell proliferation but increased the percentage of CD38high CD24high B cells (Bregs) and IL-10 production, while MSC–IFN-γ significantly reduced B cell proliferation and inhibited IgG production by B cells in a more potent fashion but did not induce Bregs or IL-10 production. Both MSC and MSC–IFN-γ required proximity to target cells and being metabolically active to exert their effects. Indoleamine 2,3 dioxygenase expression was highly induced in MSC–IFN-γ and was responsible of the anti-proliferative and Breg reduction since addition of tryptophan (TRP) restored MSC properties. Immunological conditions dictate the effect of MSC on B cell function. Under immunological quiescent conditions, MSC stimulate Breg induction; whereas, under inflammatory conditions, MSC inhibit B cell proliferation and maturation through depletion of TRP. This knowledge is useful for customizing MSC therapy for specific purposes by appropriate pretreatment of MSC.
Collapse
Affiliation(s)
- Franka Luk
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Laura Carreras-Planella
- REMAR Group and Nephrology Service, Germans Trias i Pujol Health Science Institute & University Hospital, Badalona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sander S Korevaar
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Samantha F H de Witte
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Francesc E Borràs
- REMAR Group and Nephrology Service, Germans Trias i Pujol Health Science Institute & University Hospital, Badalona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Michiel G H Betjes
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Carla C Baan
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Martin J Hoogduijn
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Marcella Franquesa
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, Netherlands.,REMAR Group and Nephrology Service, Germans Trias i Pujol Health Science Institute & University Hospital, Badalona, Spain
| |
Collapse
|
770
|
Mesenchymal Stem Cells in Restoration of Fertility at Experimental Pelvic Inflammatory Disease. Stem Cells Int 2017; 2017:2014132. [PMID: 28928773 PMCID: PMC5591961 DOI: 10.1155/2017/2014132] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 06/15/2017] [Accepted: 07/17/2017] [Indexed: 12/17/2022] Open
Abstract
Inflammatory disorders account for a significant percentage of gynecologic diseases, particularly in women of reproductive age. It is known that stem cells have anti-inflammatory and regenerative properties. Based on this, we investigated the effect of intravenous administration of cryopreserved mesenchymal stem cells (cMSCs) of bone marrow on experimental chronic inflammation of the ovaries. The paper shows that on the 21st day after cMSC therapy, leukocyte infiltration of ovaries was slightly relative to the control group without treatment, and the ratio of developing and atretic follicles in the animals with cMSC injection dramatically increased, while in the control, it still remained on the side of atretic forms. The number of apoptotic oocytes after stimulation of superovulation in the control group was significantly higher (85.3 ± 5.2%) than that in the animals with therapy (5.7 ± 0.8%). Relative number of fertilized eggs in the group with cMSC therapy was higher by 40% compare to that in the control. Pregnancy rate in natural estrous cycle after cell administration increased by 20%, and average number of litters in this group was two times significantly higher than that in the control. So the intravenous injection of cMSCs has the restorative effect on the fertility at experimental pelvic inflammatory disease.
Collapse
|
771
|
Deriving vascular smooth muscle cells from mesenchymal stromal cells: Evolving differentiation strategies and current understanding of their mechanisms. Biomaterials 2017; 145:9-22. [PMID: 28843066 DOI: 10.1016/j.biomaterials.2017.08.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/07/2017] [Accepted: 08/14/2017] [Indexed: 12/11/2022]
Abstract
Vascular smooth muscle cells (VSMCs) play essential roles in regulating blood vessel form and function. Regeneration of functional vascular smooth muscle tissue to repair vascular diseases is an area of intense research in tissue engineering and regenerative medicine. For functional vascular smooth muscle tissue regeneration to become a practical therapy over the next decade, the field will need to have access to VSMC sources that are effective, robust and safe. While pluripotent stem cells hold good future promise to this end, more immediate translation is expected to come from approaches that generate functional VSMCs from adult sources of multipotent adipose-derived and bone marrow-derived mesenchymal stromal cells (ASCs and BMSCs). The research to this end is extensive and is dominated by studies relating to classical biochemical signalling molecules used to induce differentiation of ASCs and BMSCs. However, prolonged use of the biochemical induction factors is costly and can cause potential endotoxin contamination in the culture. Over recent years several non-traditional differentiation approaches have been devised to mimic defined aspects of the native micro-environment in which VSMCs reside to contribute to the differentiation of VSMC-like cells from ASCs and BMSCs. In this review, the promises and limitations of several non-traditional culture approaches (e.g., co-culture, biomechanical, and biomaterial stimuli) targeting VSMC differentiation are discussed. The extensive crosstalk between the underlying signalling cascades are delineated and put into a translational context. It is expected that this review will not only provide significant insight into VSMC differentiation strategies for vascular smooth muscle tissue engineering applications, but will also highlight the fundamental importance of engineering the cellular microenvironment on multiple scales (with consideration of different combinatorial pathways) in order to direct cell differentiation fate and obtain cells of a desired and stable phenotype. These strategies may ultimately be applied to different sources of stem cells in the future for a range of biomaterial and tissue engineering disciplines.
Collapse
|
772
|
Effects of a defined xeno-free medium on the growth and neurotrophic and angiogenic properties of human adult stem cells. Cytotherapy 2017; 19:629-639. [PMID: 28366194 DOI: 10.1016/j.jcyt.2017.02.360] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 02/22/2017] [Accepted: 02/24/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND The growth properties and neurotrophic and angiogenic effects of human mesenchymal stromal cells (MSCs) cultured in a defined xeno-free, serum-free medium (MesenCult-XF) were investigated. METHODS Human MSCs from adipose tissue (ASCs) and bone marrow (BMSCs) were cultured in Minimum Essential Medium-alpha (α-MEM) containing fetal calf serum or in MesenCult-XF. Proliferation was measured over 10 passages and the colony-forming unit (CFU) assay and expression of cluster of differentiation (CD) surface markers were determined. Neurite outgrowth and angiogenic activity of the MSCs were determined. RESULTS At early passage, both ASCs and BMSCs showed better proliferation in MesenCult-XF compared with standard α-MEM-containing serum. However, CFUs were significantly lower in MesenCult-XF. ASCs cultured in MesenCult-XF continued to expand at faster rates than cells grown in serum. BMSCs showed morphological changes at late passage in MesenCult-XF and stained positive for senescence β-galactosidase activity. Expression levels of CD73 and CD90 were similar in both cell types under the various culture conditions but CD105 was significantly reduced at passage 10 in MesenCult-XF. In vitro stimulation of the cells enhanced the expression of brain-derived neurotrophic factor (BDNF), vascular endothelial growth factor (VEGF-A) and angiopoietin-1. Stimulated ASCs grown in MesenCult-XF evoked the longest neurite outgrowth in a neuron co-culture model. Stimulated BMSCs grown in MesenCult-XF produced the most extensive network of capillary-like tube structures in an in vitro angiogenesis assay. CONCLUSIONS ASCs and BMSCs exhibit high levels of neurotrophic and angiogenic activity when grown in the defined serum-free medium indicating their suitability for treatment of various neurological conditions. However, long-term expansion in MesenCult-XF might be restricted to ASCs.
Collapse
|
773
|
Cellular Reparative Mechanisms of Mesenchymal Stem Cells for Retinal Diseases. Int J Mol Sci 2017; 18:ijms18081406. [PMID: 28788088 PMCID: PMC5577990 DOI: 10.3390/ijms18081406] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 06/09/2017] [Accepted: 06/12/2017] [Indexed: 12/22/2022] Open
Abstract
The use of multipotent mesenchymal stem cells (MSCs) has been reported as promising for the treatment of numerous degenerative disorders including the eye. In retinal degenerative diseases, MSCs exhibit the potential to regenerate into retinal neurons and retinal pigmented epithelial cells in both in vitro and in vivo studies. Delivery of MSCs was found to improve retinal morphology and function and delay retinal degeneration. In this review, we revisit the therapeutic role of MSCs in the diseased eye. Furthermore, we reveal the possible cellular mechanisms and identify the associated signaling pathways of MSCs in reversing the pathological conditions of various ocular disorders such as age-related macular degeneration (AMD), retinitis pigmentosa, diabetic retinopathy, and glaucoma. Current stem cell treatment can be dispensed as an independent cell treatment format or with the combination of other approaches. Hence, the improvement of the treatment strategy is largely subjected by our understanding of MSCs mechanism of action.
Collapse
|
774
|
Neuron-Specific Fluorescence Reporter-Based Live Cell Tracing for Transdifferentiation of Mesenchymal Stem Cells into Neurons by Chemical Compound. Stem Cells Int 2017; 2017:8452830. [PMID: 28808446 PMCID: PMC5541830 DOI: 10.1155/2017/8452830] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 02/13/2017] [Accepted: 03/01/2017] [Indexed: 12/12/2022] Open
Abstract
Although transdifferentiation of mesenchymal stem cells (MSCs) into neurons increases the possibility of therapeutic use of MSCs for neurodevelopmental disorders, the use of MSCs has the limitation on differentiation efficiency to neuronal lineage and lack of an easy method to monitor the transdifferentiation. In this study, using time-lapse live cell imaging, we assessed the neuronal differentiation of MSCs induced by a small molecule “NHPDQC (N-hydroxy-2-oxo-3-(3-phenylprophyl)-1,2-dihydroquinoxaline-6-carboxamide, C18H17N3O3).” Plasmid vector containing red fluorescence reporter genes under the control of the tubulin α1 (Tα1) promoter (pTα1-DsRed2) traced the neuronal differentiation of MSCs. Two days after NHPDQC treatment, MSCs showed neuron-like phenotype with neurite outgrowth and high expression of neuron-specific markers in more than 95% cells. The fluorescence signals increased in the cytoplasm of pTα1-DsRed2-transfected MSCs after NHPDQC treatment. In vitro monitoring of MSCs along the time courses showed progressive increase of fluorescence till 30 h after treatment, corresponding with the increase in neurite length. We examined an efficient neuronal differentiation of MSCs by NHPDQC alone and monitored the temporal changes of neuronal differentiation by neuron-specific fluorescence reporter along time. This method would help further our understanding of the differentiation of MSCs to produce neurons by simple treatment of small molecule.
Collapse
|
775
|
Parga JA, García-Garrote M, Martínez S, Raya Á, Labandeira-García JL, Rodríguez-Pallares J. Prostaglandin EP2 Receptors Mediate Mesenchymal Stromal Cell-Neuroprotective Effects on Dopaminergic Neurons. Mol Neurobiol 2017; 55:4763-4776. [DOI: 10.1007/s12035-017-0681-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 06/29/2017] [Indexed: 12/20/2022]
|
776
|
Chen X, Wang S, Cao W. Mesenchymal stem cell-mediated immunomodulation in cell therapy of neurodegenerative diseases. Cell Immunol 2017; 326:8-14. [PMID: 28778534 DOI: 10.1016/j.cellimm.2017.06.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 06/21/2017] [Accepted: 06/23/2017] [Indexed: 12/19/2022]
Abstract
Dysfunction of immune responses has been identified to involve in the pathogenesis of various neurodegenerative diseases. Abnormal activation of glia cells and/or infiltration of peripheral adaptive immune cells always sustains neuroinflammation and the disease progression. Obviously, the regulation of neuroinflammation has become a potential therapeutic strategy against neurodegenerative diseases. Mesenchymal stem cells (MSCs) exhibit complex interactions with various immune cells including T cells, macrophages and especially resident glia cells in the central nervous system. In response to tissue injury signals, MSCs adopt specific phenotype to suppress or promote immune responses depending on the inflammatory microenvironment they reside. Therefore, manipulation of MSCs may hold great potentials to improve MSC-based therapy on neurodegenerative diseases. Here we review MSC-mediated immunomodulation in cell therapy of neurodegenerative diseases, providing fundamental information for guiding appropriate applications of MSCs in clinical settings.
Collapse
Affiliation(s)
- Xiaodong Chen
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiaotong University School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| | - Shijia Wang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiaotong University School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| | - Wei Cao
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiaotong University School of Medicine, 320 Yueyang Road, Shanghai 200031, China.
| |
Collapse
|
777
|
Galectin-3 Knockdown Impairs Survival, Migration, and Immunomodulatory Actions of Mesenchymal Stromal Cells in a Mouse Model of Chagas Disease Cardiomyopathy. Stem Cells Int 2017; 2017:3282656. [PMID: 28769980 PMCID: PMC5523546 DOI: 10.1155/2017/3282656] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 05/27/2017] [Accepted: 06/05/2017] [Indexed: 01/13/2023] Open
Abstract
Therapies based on transplantation of mesenchymal stromal cells (MSC) hold promise for the management of inflammatory disorders. In chronic Chagas disease cardiomyopathy (CCC), caused by chronic infection with Trypanosoma cruzi, the exacerbated immune response plays a critical pathophysiological role and can be modulated by MSC. Here, we investigated the role of galectin-3 (Gal-3), a beta-galactoside-binding lectin with several actions on immune responses and repair process, on the immunomodulatory potential of MSC. Gal-3 knockdown in MSC did not affect the immunophenotype or differentiation potential. However, Gal-3 knockdown MSC showed decreased proliferation, survival, and migration. Additionally, when injected intraperitoneally into mice with CCC, Gal-3 knockdown MSC showed impaired migration in vivo. Transplantation of control MSC into mice with CCC caused a suppression of cardiac inflammation and fibrosis, reducing expression levels of CD45, TNFα, IL-1β, IL-6, IFNγ, and type I collagen. In contrast, Gal-3 knockdown MSC were unable to suppress the immune response or collagen synthesis in the hearts of mice with CCC. Finally, infection with T. cruzi demonstrated parasite survival in wild-type but not in Gal-3 knockdown MSC. These findings demonstrate that Gal-3 plays a critical role in MSC survival, proliferation, migration, and therapeutic potential in CCC.
Collapse
|
778
|
Franceschetti T, De Bari C. The potential role of adult stem cells in the management of the rheumatic diseases. Ther Adv Musculoskelet Dis 2017; 9:165-179. [PMID: 28717403 PMCID: PMC5502944 DOI: 10.1177/1759720x17704639] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 02/28/2017] [Indexed: 12/27/2022] Open
Abstract
Adult stem cells are considered as appealing therapeutic candidates for inflammatory and degenerative musculoskeletal diseases. A large body of preclinical research has contributed to describing their immune-modulating properties and regenerative potential. Additionally, increasing evidence suggests that stem cell differentiation and function are disrupted in the pathogenesis of rheumatic diseases. Clinical studies have been limited, for the most part, to the application of adult stem cell-based treatments on small numbers of patients or as a 'salvage' therapy in life-threatening disease cases. Nevertheless, these preliminary studies indicate that adult stem cells are promising tools for the long-term treatment of rheumatic diseases. This review highlights recent knowledge acquired in the fields of hematopoietic and mesenchymal stem cell therapy for the management of systemic sclerosis (SSc), systemic lupus erythematosus (SLE), rheumatoid arthritis (RA) and osteoarthritis (OA) and the potential mechanisms mediating their function.
Collapse
Affiliation(s)
- Tiziana Franceschetti
- Arthritis & Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Cosimo De Bari
- Arthritis & Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
779
|
Prockop DJ, Oh JY, Lee RH. Data against a Common Assumption: Xenogeneic Mouse Models Can Be Used to Assay Suppression of Immunity by Human MSCs. Mol Ther 2017. [PMID: 28647464 DOI: 10.1016/j.ymthe.2017.06.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Much of what we know about immunology suggests that little is to be gained from experiments in which human cells are administered to immunocompetent mice. Multiple reports have demonstrated that this common assumption does not hold for experiments with human mesenchymal stem/stromal cells (hMSCs). The data demonstrate that hMSCs can suppress immune responses to a variety of stimuli in immunocompetent mice by a range of different mechanisms that are similar to those employed by mouse MSCs. Therefore, further experiments with hMSCs in mice will make it possible to generate preclinical data that will improve both the efficacy and safety of the clinical trials with the cells that are now in progress.
Collapse
Affiliation(s)
- Darwin J Prockop
- Institute for Regenerative Medicine, College of Medicine, Texas A&M University, 1114 TAMU, 206 Olsen Boulevard, College Station, TX 77845, USA.
| | - Joo Youn Oh
- Department of Ophthalmology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 110-744, Korea; Laboratory of Ocular Regenerative Medicine and Immunology, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 110-744, Korea
| | - Ryang Hwa Lee
- Institute for Regenerative Medicine, College of Medicine, Texas A&M University, 1114 TAMU, 206 Olsen Boulevard, College Station, TX 77845, USA
| |
Collapse
|
780
|
Royce SG, Rele S, Broughton BRS, Kelly K, Samuel CS. Intranasal administration of mesenchymoangioblast-derived mesenchymal stem cells abrogates airway fibrosis and airway hyperresponsiveness associated with chronic allergic airways disease. FASEB J 2017. [PMID: 28626025 DOI: 10.1096/fj.201700178r] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Structural changes known as airway remodeling (AWR) characterize chronic/severe asthma and contribute to lung dysfunction. Thus, we assessed the in vivo efficacy of induced pluripotent stem cell and mesenchymoangioblast-derived mesenchymal stem cells (MCA-MSCs) on AWR in a murine model of chronic allergic airways disease (AAD)/asthma. Female Balb/c mice were subjected to a 9-wk model of ovalbumin (Ova)-induced chronic AAD and treated intravenously or intranasally with MCA-MSCs from weeks 9 to 11. Changes in airway inflammation (AI), AWR, and airway hyperresponsiveness (AHR) were assessed. Ova-injured mice presented with AI, goblet cell metaplasia, epithelial thickening, increased airway TGF-β1 levels, subepithelial myofibroblast and collagen accumulation, total lung collagen concentration, and AHR (all P < 0.001 vs. uninjured control group). Apart from epithelial thickness, all other parameters measured were significantly, although not totally, decreased by intravenous delivery of MCA-MSCs to Ova-injured mice. In comparison, intranasal delivery of MCA-MSCs to Ova-injured mice significantly decreased all parameters measured (all P < 0.05 vs. Ova group) and, most notably, normalized aberrant airway TGF-β1 levels, airway/lung fibrosis, and AHR to values measured in uninjured animals. MCA-MSCs also increased collagen-degrading gelatinase levels. Hence, direct delivery of MCA-MSCs offers great therapeutic benefit for the AWR and AHR associated with chronic AAD.-Royce, S. G., Rele, S., Broughton, B. R. S., Kelly, K., Samuel, C. S. Intranasal administration of mesenchymoangioblast-derived mesenchymal stem cells abrogates airway fibrosis and airway hyperresponsiveness associated with chronic allergic airways disease.
Collapse
Affiliation(s)
- Simon G Royce
- Fibrosis Laboratory, Monash University, Clayton, Victoria, Australia; .,Department of Medicine, Central Clinical School, Monash University, Prahran, Victoria, Australia; and
| | - Siddharth Rele
- Fibrosis Laboratory, Monash University, Clayton, Victoria, Australia
| | - Brad R S Broughton
- Cardiovascular and Pulmonary Pharmacology Group, Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Kilian Kelly
- Cynata Therapeutics, Armadale, Victoria, Australia
| | - Chrishan S Samuel
- Fibrosis Laboratory, Monash University, Clayton, Victoria, Australia;
| |
Collapse
|
781
|
The effect of thymic mesenchymal stromal cells on arginase activity and nitric oxide produced by mouse macrophages. UKRAINIAN BIOCHEMICAL JOURNAL 2017. [DOI: 10.15407/ubj89.03.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
782
|
Arisi MF, Chirico EN, Sebeny R, Muthukumaran G, Mu A, De Jonghe BC, Margulies KB, Libonati JR. Myocardial apoptosis and mesenchymal stem cells with acute exercise. Physiol Rep 2017; 5:5/11/e13297. [PMID: 28576853 PMCID: PMC5471436 DOI: 10.14814/phy2.13297] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 05/02/2017] [Indexed: 01/01/2023] Open
Abstract
Aerobic exercise confers many health benefits. However, numerous reports have shown that acute aerobic exercise can injure the heart. We tested the general hypothesis that acute moderate‐intensity exercise in rodents induces cardiomyocyte damage and stimulates mesenchymal stem cells (MSCs) to increase paracrine‐mediated protective effects on cardiomyocytes. A single session of treadmill running (13 m/min, 0% grade, for 45 min) in untrained C57BL/6 male mice (n = 18) increased cleaved poly ADP‐ribose polymerase (PARP), a marker of apoptosis, in the myocardium 24 h postexercise. Microarray analysis of mouse myocardium identified 11 relevant apoptotic genes and several shifts in matrix remodeling transcripts over the postexercise window. Postexercise cardiomyocyte death was recapitulated in neonatal rat cardiomyocytes (NRCMs) by culturing cells in 2% plasma harvested from exercised rats. The increased cell death observed in exercise‐treated NRCMs was attenuated by β‐adrenergic blockade, but not antioxidant treatment. MSC survival, proliferation, and chemotaxis showed no significant differences between sedentary and exercise plasma conditions, despite increased IL‐6, TNF‐α, IL‐1β, and IFN‐γ secretions from MSCs treated with exercise plasma. NRCM survival was increased nearly 500% when cocultured with MSCs, but this effect was not altered under exercise plasma culture conditions. Our results suggest acute moderate‐intensity aerobic treadmill running in exercise‐naïve rodents induces temporal cardiomyocyte death due to plasma‐borne factors, namely, catecholaminergic stress. Even though exercise conditions prompt an inflammatory response in MSCs, the exercise milieu does not alter the MSC‐protective phenotype on cardiomyocytes.
Collapse
Affiliation(s)
- Maria F Arisi
- School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Roxanne Sebeny
- School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Anbin Mu
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Bart C De Jonghe
- School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Joseph R Libonati
- School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
783
|
Tozetti PA, Caruso SR, Mizukami A, Fernandes TR, da Silva FB, Traina F, Covas DT, Orellana MD, Swiech K. Expansion strategies for human mesenchymal stromal cells culture under xeno-free conditions. Biotechnol Prog 2017; 33:1358-1367. [DOI: 10.1002/btpr.2494] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 04/12/2017] [Indexed: 01/16/2023]
Affiliation(s)
- Patrícia Aparecida Tozetti
- Hemotherapy Center of Ribeirão Preto; Ribeirão Preto Medical School, University of São Paulo; Ribeirão Preto SP Brazil
| | - Samia Rigotto Caruso
- Hemotherapy Center of Ribeirão Preto; Ribeirão Preto Medical School, University of São Paulo; Ribeirão Preto SP Brazil
| | - Amanda Mizukami
- Hemotherapy Center of Ribeirão Preto; Ribeirão Preto Medical School, University of São Paulo; Ribeirão Preto SP Brazil
| | - Taisa Risque Fernandes
- Hemotherapy Center of Ribeirão Preto; Ribeirão Preto Medical School, University of São Paulo; Ribeirão Preto SP Brazil
| | - Fernanda Borges da Silva
- Dept. of Internal Medicine; Ribeirão Preto Medical School, University of São Paulo; Ribeirão Preto SP Brazil
| | - Fabiola Traina
- Dept. of Internal Medicine; Ribeirão Preto Medical School, University of São Paulo; Ribeirão Preto SP Brazil
| | - Dimas Tadeu Covas
- Hemotherapy Center of Ribeirão Preto; Ribeirão Preto Medical School, University of São Paulo; Ribeirão Preto SP Brazil
- Dept. of Internal Medicine; Ribeirão Preto Medical School, University of São Paulo; Ribeirão Preto SP Brazil
| | - Maristela Delgado Orellana
- Hemotherapy Center of Ribeirão Preto; Ribeirão Preto Medical School, University of São Paulo; Ribeirão Preto SP Brazil
| | - Kamilla Swiech
- Dept. of Pharmaceutical Sciences, School of Pharmaceutical Sciences of Ribeirão Preto; University of São Paulo; Ribeirão Preto SP Brazil
- Hemotherapy Center of Ribeirão Preto; Ribeirão Preto Medical School, University of São Paulo; Ribeirão Preto SP Brazil
| |
Collapse
|
784
|
Simkin J, Gawriluk TR, Gensel JC, Seifert AW. Macrophages are necessary for epimorphic regeneration in African spiny mice. eLife 2017; 6:e24623. [PMID: 28508748 PMCID: PMC5433844 DOI: 10.7554/elife.24623] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 04/11/2017] [Indexed: 02/06/2023] Open
Abstract
How the immune system affects tissue regeneration is not well understood. In this study, we used an emerging mammalian model of epimorphic regeneration, the African spiny mouse, to examine cell-based inflammation and tested the hypothesis that macrophages are necessary for regeneration. By directly comparing inflammatory cell activation in a 4 mm ear injury during regeneration (Acomys cahirinus) and scarring (Mus musculus), we found that both species exhibited an acute inflammatory response, with scarring characterized by stronger myeloperoxidase activity. In contrast, ROS production was stronger and more persistent during regeneration. By depleting macrophages during injury, we demonstrate a functional requirement for these cells to stimulate regeneration. Importantly, the spatial distribution of activated macrophage subtypes was unique during regeneration with pro-inflammatory macrophages failing to infiltrate the regeneration blastema. Together, our results demonstrate an essential role for inflammatory cells to regulate a regenerative response.
Collapse
Affiliation(s)
- Jennifer Simkin
- Department of Biology, University of Kentucky, Lexington, United States
- Department of Physiology, University of Kentucky, Lexington, United States
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, United States
| | - Thomas R Gawriluk
- Department of Biology, University of Kentucky, Lexington, United States
| | - John C Gensel
- Department of Physiology, University of Kentucky, Lexington, United States
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, United States
| | - Ashley W Seifert
- Department of Biology, University of Kentucky, Lexington, United States
| |
Collapse
|
785
|
Diverging Concepts and Novel Perspectives in Regenerative Medicine. Int J Mol Sci 2017; 18:ijms18051021. [PMID: 28486410 PMCID: PMC5454934 DOI: 10.3390/ijms18051021] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 04/28/2017] [Accepted: 05/03/2017] [Indexed: 12/31/2022] Open
Abstract
Regenerative medicine has rapidly evolved, due to progress in cell and molecular biology allowing the isolation, characterization, expansion, and engineering of cells as therapeutic tools. Despite past limited success in the clinical translation of several promising preclinical results, this novel field is now entering a phase of renewed confidence and productivity, marked by the commercialization of the first cell therapy products. Ongoing issues in the field include the use of pluripotent vs. somatic and of allogenic vs. autologous stem cells. Moreover, the recognition that several of the observed beneficial effects of cell therapy are not due to integration of the transplanted cells, but rather to paracrine signals released by the exogenous cells, is generating new therapeutic perspectives in the field. Somatic stem cells are outperforming embryonic and induced pluripotent stem cells in clinical applications, mainly because of their more favorable safety profile. Presently, both autologous and allogeneic somatic stem cells seem to be equally safe and effective under several different conditions. Recognition that a number of therapeutic effects of transplanted cells are mediated by paracrine signals, and that such signals can be found in extracellular vesicles isolated from culture media, opens novel therapeutic perspectives in the field of regenerative medicine.
Collapse
|
786
|
De Luca L, Trino S, Laurenzana I, Lamorte D, Caivano A, Del Vecchio L, Musto P. Mesenchymal Stem Cell Derived Extracellular Vesicles: A Role in Hematopoietic Transplantation? Int J Mol Sci 2017; 18:ijms18051022. [PMID: 28486431 PMCID: PMC5454935 DOI: 10.3390/ijms18051022] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 05/04/2017] [Accepted: 05/04/2017] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are a heterogeneous cellular population containing different progenitors able to repair tissues, support hematopoiesis, and modulate immune and inflammatory responses. Several clinical trials have used MSCs in allogeneic hematopoietic stem cell transplantation (allo-HSCT) to prevent hematopoietic stem cell (HSC) engraftment failure, reduce aplasia post chemotherapy, and to control graft versus host disease (GvHD). The efficacy of MSCs is linked to their immune suppressive and anti-inflammatory properties primarily due to the release of soluble factors. Recent studies indicate that most of these effects are mediated by extracellular vesicles (EVs). MSC-EVs have therefore therapeutic effects in regenerative medicine, tumor inhibition, and immune-regulation. MSC-EVs may offer specific advantages for patient safety, such as lower propensity to trigger innate and adaptive immune responses. It has been also shown that MSC-EVs can prevent or treat acute-GvHD by modulating the immune-response and, combined with HSCs, may contribute to the hematopoietic microenvironment reconstitution. Finally, MSC-EVs may provide a new potential therapeutic option (e.g., transplantation, gene therapy) for different diseases, particularly hematological malignancies. In this review, we will describe MSC and MSC-EVs role in improving allo-HSCT procedures and in treating GvHD.
Collapse
Affiliation(s)
- Luciana De Luca
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| | - Stefania Trino
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| | - Ilaria Laurenzana
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| | - Daniela Lamorte
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| | - Antonella Caivano
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| | - Luigi Del Vecchio
- CEINGE Biotecnologie Avanzate s.c.a r.l., 80147 Naples, Italy.
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80138 Napoli, Italy.
| | - Pellegrino Musto
- Scientific Direction, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| |
Collapse
|
787
|
Elbadry MI, Espinoza JL, Nakao S. Induced pluripotent stem cell technology: A window for studying the pathogenesis of acquired aplastic anemia and possible applications. Exp Hematol 2017; 49:9-18. [DOI: 10.1016/j.exphem.2016.12.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/09/2016] [Accepted: 12/25/2016] [Indexed: 01/08/2023]
|
788
|
Tesauro M, Mauriello A, Rovella V, Annicchiarico-Petruzzelli M, Cardillo C, Melino G, Di Daniele N. Arterial ageing: from endothelial dysfunction to vascular calcification. J Intern Med 2017; 281:471-482. [PMID: 28345303 DOI: 10.1111/joim.12605] [Citation(s) in RCA: 224] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Complex structural and functional changes occur in the arterial system with advancing age. The aged artery is characterized by changes in microRNA expression patterns, autophagy, smooth muscle cell migration and proliferation, and arterial calcification with progressively increased mechanical vessel rigidity and stiffness. With age the vascular smooth muscle cells modify their phenotype from contractile to 'synthetic' determining the development of intimal thickening as early as the second decade of life as an adaptive response to forces acting on the arterial wall. The increased permeability observed in intimal thickening could represent the substrate on which low-level atherosclerotic stimuli can promote the development of advanced atherosclerotic lesions. In elderly patients the atherosclerotic plaques tend to be larger with increased vascular stenosis. In these plaques there is a progressive accumulation of both lipids and collagen and a decrease of inflammation. Similarly the plaques from elderly patients show more calcification as compared with those from younger patients. The coronary artery calcium score is a well-established marker of adverse cardiovascular outcomes. The presence of diffuse calcification in a severely stenotic segment probably induces changes in mechanical properties and shear stress of the arterial wall favouring the rupture of a vulnerable lesion in a less stenotic adjacent segment. Oxidative stress and inflammation appear to be the two primary pathological mechanisms of ageing-related endothelial dysfunction even in the absence of clinical disease. Arterial ageing is no longer considered an inexorable process. Only a better understanding of the link between ageing and vascular dysfunction can lead to significant advances in both preventative and therapeutic treatments with the aim that in the future vascular ageing may be halted or even reversed.
Collapse
Affiliation(s)
- M Tesauro
- Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - A Mauriello
- Department of Experimental Medicine and Surgery, University of Rome 'Tor Vergata', Rome, Italy
| | - V Rovella
- Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | | | - C Cardillo
- Department of Internal Medicine, Catholic University, Rome, Italy
| | - G Melino
- Department of Experimental Medicine and Surgery, University of Rome 'Tor Vergata', Rome, Italy.,Medical Research Council, Toxicology Unit, Leicester University, Leicester, UK
| | - N Di Daniele
- Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| |
Collapse
|
789
|
|
790
|
Abu-Elmagd M, Alghamdi MA, Shamy M, Khoder MI, Costa M, Assidi M, Kadam R, Alsehli H, Gari M, Pushparaj PN, Kalamegam G, Al-Qahtani MH. Evaluation of the Effects of Airborne Particulate Matter on Bone Marrow-Mesenchymal Stem Cells (BM-MSCs): Cellular, Molecular and Systems Biological Approaches. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2017; 14:ijerph14040440. [PMID: 28425934 PMCID: PMC5409640 DOI: 10.3390/ijerph14040440] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 04/07/2017] [Accepted: 04/14/2017] [Indexed: 12/13/2022]
Abstract
Particulate matter (PM) contains heavy metals that affect various cellular functions and gene expression associated with a range of acute and chronic diseases in humans. However, the specific effects they exert on the stem cells remain unclear. Here, we report the effects of PM collected from the city of Jeddah on proliferation, cell death, related gene expression and systems of biological analysis in bone marrow mesenchymal stem cells (BM-MSCs), with the aim of understanding the underlying mechanisms. PM2.5 and PM10 were tested in vitro at various concentrations (15 to 300 µg/mL) and durations (24 to 72 h). PMs induced cellular stress including membrane damage, shrinkage and death. Lower concentrations of PM2.5 increased proliferation of BM-MSCs, while higher concentrations served to decrease it. PM10 decreased BM-MSCs proliferation in a concentration-dependent manner. The X-ray fluorescence spectrometric analysis showed that PM contains high levels of heavy metals. Ingenuity Pathway Analysis (IPA) and hierarchical clustering analyses demonstrated that heavy metals were associated with signaling pathways involving cell stress/death, cancer and chronic diseases. qRT-PCR results showed differential expression of the apoptosis genes (BCL2, BAX); inflammation associated genes (TNF-α and IL-6) and the cell cycle regulation gene (p53). We conclude that PM causes inflammation and cell death, and thereby predisposes to chronic debilitating diseases.
Collapse
Affiliation(s)
- Muhammad Abu-Elmagd
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia.
| | - Mansour A Alghamdi
- Department of Environmental Sciences, Faculty of Meteorology, Environment and Arid Land Agriculture, King Abdulaziz University, P.O. Box 80208, Jeddah 21589, Saudi Arabia.
| | - Magdy Shamy
- Department of Environmental Sciences, Faculty of Meteorology, Environment and Arid Land Agriculture, King Abdulaziz University, P.O. Box 80208, Jeddah 21589, Saudi Arabia.
| | - Mamdouh I Khoder
- Department of Environmental Sciences, Faculty of Meteorology, Environment and Arid Land Agriculture, King Abdulaziz University, P.O. Box 80208, Jeddah 21589, Saudi Arabia.
| | - Max Costa
- New York University School of Medicine, Nelson Institute of Environmental Medicine, New York, NY 10987, USA.
| | - Mourad Assidi
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia.
| | - Roaa Kadam
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia.
| | - Haneen Alsehli
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia.
| | - Mamdooh Gari
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia.
| | - Peter Natesan Pushparaj
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia.
| | - Gauthaman Kalamegam
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia.
| | - Mohammed H Al-Qahtani
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
791
|
Robinson AM, Rahman AA, Miller S, Stavely R, Sakkal S, Nurgali K. The neuroprotective effects of human bone marrow mesenchymal stem cells are dose-dependent in TNBS colitis. Stem Cell Res Ther 2017; 8:87. [PMID: 28420434 PMCID: PMC5395912 DOI: 10.1186/s13287-017-0540-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/14/2017] [Accepted: 03/17/2017] [Indexed: 02/08/2023] Open
Abstract
Background The incidence of inflammatory bowel diseases (IBD) is increasing worldwide with patients experiencing severe impacts on their quality of life. It is well accepted that intestinal inflammation associates with extensive damage to the enteric nervous system (ENS), which intrinsically innervates the gastrointestinal tract and regulates all gut functions. Hence, treatments targeting the enteric neurons are plausible for alleviating IBD and associated complications. Mesenchymal stem cells (MSCs) are gaining wide recognition as a potential therapy for many diseases due to their immunomodulatory and neuroprotective qualities. However, there is a large discrepancy regarding appropriate cell doses used in both clinical trials and experimental models of disease. We have previously demonstrated that human bone marrow MSCs exhibit neuroprotective and anti-inflammatory effects in a guinea-pig model of 2,4,6-trinitrobenzene-sulfonate (TNBS)-induced colitis; but an investigation into whether this response is dose-dependent has not been conducted. Methods Hartley guinea-pigs were administered TNBS or sham treatment intra-rectally. Animals in the MSC treatment groups received either 1 × 105, 1 × 106 or 3 × 106 MSCs by enema 3 hours after induction of colitis. Colon tissues were collected 72 hours after TNBS administration to assess the effects of MSC treatments on the level of inflammation and damage to the ENS by immunohistochemical and histological analyses. Results MSCs administered at a low dose, 1 × 105 cells, had little or no effect on the level of immune cell infiltrate and damage to the colonic innervation was similar to the TNBS group. Treatment with 1 × 106 MSCs decreased the quantity of immune infiltrate and damage to nerve processes in the colonic wall, prevented myenteric neuronal loss and changes in neuronal subpopulations. Treatment with 3 × 106 MSCs had similar effects to 1 × 106 MSC treatments. Conclusions The neuroprotective effect of MSCs in TNBS colitis is dose-dependent. Increasing doses higher than 1 × 106 MSCs demonstrates no further therapeutic benefit than 1 × 106 MSCs in preventing enteric neuropathy associated with intestinal inflammation. Furthermore, we have established an optimal dose of MSCs for future studies investigating intestinal inflammation, the enteric neurons and stem cell therapy in this model.
Collapse
Affiliation(s)
- Ainsley M Robinson
- College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia
| | - Ahmed A Rahman
- College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia
| | - Sarah Miller
- College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia
| | - Rhian Stavely
- College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia
| | - Samy Sakkal
- College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia
| | - Kulmira Nurgali
- College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia.
| |
Collapse
|
792
|
Cagliani J, Grande D, Molmenti EP, Miller EJ, Rilo HL. Immunomodulation by Mesenchymal Stromal Cells and Their Clinical Applications. JOURNAL OF STEM CELL AND REGENERATIVE BIOLOGY 2017; 3:10.15436/2471-0598.17.022. [PMID: 29104965 PMCID: PMC5667922 DOI: 10.15436/2471-0598.17.022] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mesenchymal stromal cells (MSCs) are multipotent progenitor cells that can be isolated and expanded from various sources. MSCs modulate the function of immune cells, including T and B lymphocytes, dendritic cells, and natural killer cells. An understanding of the interaction between MSCs and the inflammatory microenvironment will provide critical information in revealing the precise in vivo mechanisms involved in MSCs-mediated therapeutic effects, and for designing more practical protocols for the clinical use of these cells. In this review we describe the current knowledge of the unique biological properties of MSCs, the immunosuppressive effects on immune-competent cells and the paracrine role of soluble factors. A summary of the participation of MSCs in preclinical and clinical studies in treating autoimmune diseases and other diseases is described. We also discuss the current challenges of their use and their potential roles in cell therapies.
Collapse
Affiliation(s)
- Joaquin Cagliani
- The Feinstein Institute for Medical Research, Center for Heart and Lungs, Northwell Health System, Manhasset, N Y, USA
- The Elmezzi Graduate School of Molecular Medicine, Northwell Health System, Manhasset, NY, USA
| | - Daniel Grande
- The Feinstein Institute for Medical Research, Orthopedic Research Laboratory, Northwell Health System, Manhasset, N Y, USA
| | - Ernesto P Molmenti
- Transplantation of Surgery, Department of Surgery, Northwell Health System, Manhasset, NY, USA
| | - Edmund J. Miller
- The Feinstein Institute for Medical Research, Center for Heart and Lungs, Northwell Health System, Manhasset, N Y, USA
| | - Horacio L.R. Rilo
- Pancreas Disease Center, Department of Surgery, Northwell Health System, Manhasset, NY, USA
| |
Collapse
|
793
|
Mesenchymal stem cells cannot affect mRNA expression of toll-like receptors in different tissues during sepsis. Inflamm Res 2017; 66:547-551. [PMID: 28391364 DOI: 10.1007/s00011-017-1042-6] [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: 03/08/2017] [Revised: 03/31/2017] [Accepted: 04/03/2017] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVE AND DESIGN Experimental animal models and human clinical studies support a crucial role for TLRs in infectious diseases. The aim of this study was to test the ability of MSCs, which have immunomodulatory effects, of altering the mRNA expression of toll-like receptors during a experimental model of sepsis in different tissues. MATERIALS AND METHODS Three experimental groups (male C57BL/6 mice) were formed for the test: control group, untreated septic group and septic group treated with MSCs (1 × 106 cells/animal). Lungs, cortex, kidney, liver and colon tissue were dissected after 12 h of sepsis induction and TLR2/3/4/9 mRNA were evaluated by RT-qPCR. RESULTS We observed a decrease of TLR2 and 9 mRNA expression in the liver of the sepsis group, while TLR3 was decreased in the lung and liver. No change was found between the sepsis group and the sepsis + MSC group. CONCLUSIONS In this model of experimental sepsis the MSCs were unable to modify the mRNA expression of the different toll-like receptors evaluated.
Collapse
|
794
|
Stemness Maintenance Properties in Human Oral Stem Cells after Long-Term Passage. Stem Cells Int 2017; 2017:5651287. [PMID: 28469672 PMCID: PMC5392399 DOI: 10.1155/2017/5651287] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 12/20/2016] [Accepted: 01/30/2017] [Indexed: 12/21/2022] Open
Abstract
Background. Neural crest-derived mesenchymal stem cells (MSCs) from human oral tissues possess immunomodulatory and regenerative properties and are emerging as a potential therapeutic tool to treat diverse diseases, such as multiple sclerosis, myocardial infarction, and connective tissue damages. In addition to cell-surface antigens, dental MSCs express embryonic stem cell markers as neural crest cells originate from the ectoderm layer. In vitro passages may eventually modify these embryonic marker expressions and other stemness properties, including proliferation. In the present study, we have investigated the expression of proteins involved in cell proliferation/senescence and embryonic stem cell markers during early (passage 2) and late passages (passage 15) in MSCs obtained from human gingiva, periodontal, and dental pulp tissues. Methods. Cell proliferation assay, beta galactosidase staining, immunocytochemistry, and real-time PCR techniques were applied. Results. Cell proliferation assay showed no difference between early and late passages while senescence markers p16 and p21 were considerably increased in late passage. Embryonic stem cell markers including SKIL, MEIS1, and JARID2 were differentially modulated between P2 and P15 cells. Discussion. Our results suggest that the presence of embryonic and proliferation markers even in late passage may potentially endorse the application of dental-derived MSCs in stem cell therapy-based clinical trials.
Collapse
|
795
|
Dykstra JA, Facile T, Patrick RJ, Francis KR, Milanovich S, Weimer JM, Kota DJ. Concise Review: Fat and Furious: Harnessing the Full Potential of Adipose-Derived Stromal Vascular Fraction. Stem Cells Transl Med 2017; 6:1096-1108. [PMID: 28186685 PMCID: PMC5388064 DOI: 10.1002/sctm.16-0337] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 10/14/2016] [Accepted: 11/07/2016] [Indexed: 12/28/2022] Open
Abstract
Due to their capacity to self-renew, proliferate and generate multi-lineage cells, adult-derived stem cells offer great potential for use in regenerative therapies to stop and/or reverse degenerative diseases such as diabetes, heart failure, Alzheimer's disease and others. However, these subsets of cells can be isolated from different niches, each with differing potential for therapeutic applications. The stromal vascular fraction (SVF), a stem cell enriched and adipose-derived cell population, has garnered interest as a therapeutic in regenerative medicine due to its ability to secrete paracrine factors that accelerate endogenous repair, ease of accessibility and lack of identified major adverse effects. Thus, one can easily understand the rush to employ adipose-derived SVF to treat human disease. Perhaps faster than any other cell preparation, SVF is making its way to clinics worldwide, while critical preclinical research needed to establish SVF safety, efficacy and optimal, standardized clinical procedures are underway. Here, we will provide an overview of the current knowledge driving this phenomenon, its regulatory issues and existing studies, and propose potential unmapped applications. Stem Cells Translational Medicine 2017;6:1096-1108.
Collapse
Affiliation(s)
- Jordan A. Dykstra
- Children's Health Research Center, Sanford Research, Sioux FallsSouth DakotaUSA
| | - Tiffany Facile
- Children's Health Research Center, Sanford Research, Sioux FallsSouth DakotaUSA
| | - Ryan J. Patrick
- Children's Health Research Center, Sanford Research, Sioux FallsSouth DakotaUSA
| | - Kevin R. Francis
- Children's Health Research Center, Sanford Research, Sioux FallsSouth DakotaUSA
- Department of PediatricsThe University of South Dakota Sanford School of MedicineVermillion, South DakotaUSA
| | - Samuel Milanovich
- Children's Health Research Center, Sanford Research, Sioux FallsSouth DakotaUSA
- Department of PediatricsThe University of South Dakota Sanford School of MedicineVermillion, South DakotaUSA
| | - Jill M. Weimer
- Children's Health Research Center, Sanford Research, Sioux FallsSouth DakotaUSA
- Department of PediatricsThe University of South Dakota Sanford School of MedicineVermillion, South DakotaUSA
| | - Daniel J. Kota
- Children's Health Research Center, Sanford Research, Sioux FallsSouth DakotaUSA
| |
Collapse
|
796
|
Lai PL, Lin H, Chen SF, Yang SC, Hung KH, Chang CF, Chang HY, Lu FL, Lee YH, Liu YC, Huang HC, Lu J. Efficient Generation of Chemically Induced Mesenchymal Stem Cells from Human Dermal Fibroblasts. Sci Rep 2017; 7:44534. [PMID: 28303927 PMCID: PMC5356011 DOI: 10.1038/srep44534] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 02/10/2017] [Indexed: 12/13/2022] Open
Abstract
Human mesenchymal stromal/stem cells (MSCs) are multipotent and currently undergoing hundreds of clinical trials for disease treatments. To date, no studies have generated induced MSCs from skin fibroblasts with chemicals or growth factors. Here, we established the first chemical method to convert primary human dermal fibroblasts into multipotent, induced MSC-like cells (iMSCs). The conversion method uses a defined cocktail of small molecules and growth factors, and it can achieve efficient conversion with an average rate of 38% in 6 days. The iMSCs have much higher clonogenicity than fibroblasts, and they can be maintained and expanded in regular MSC medium for at least 8 passages and further differentiated into osteoblasts, adipocytes, and chondrocytes. Moreover, the iMSCs can suppress LPS-mediated acute lung injury as effectively as bone marrow-derived mesenchymal stem cells. This finding may greatly benefit stem cell biology, cell therapy, and regenerative medicine.
Collapse
Affiliation(s)
- Pei-Lun Lai
- Genome and Systems Biology Degree Program, College of Life Science, National Taiwan University, Taipei, Taiwan.,Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Hsuan Lin
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University Medical College, Taipei, Taiwan
| | - Shang-Fu Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan.,Institute of Molecular and Cellular Biology and Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan
| | | | - Kuo-Hsuan Hung
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | | | - Hsiang-Yi Chang
- Genome and Systems Biology Degree Program, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Frank Leigh Lu
- Department of Pediatrics, National Taiwan University Hospital and National Taiwan University Medical College, Taipei, Taiwan
| | - Yi-Hsuan Lee
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yu-Chuan Liu
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Hsiao-Chun Huang
- Genome and Systems Biology Degree Program, College of Life Science, National Taiwan University, Taipei, Taiwan.,Institute of Molecular and Cellular Biology and Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Electronics Engineering, College of Electrical Engineering and Computer Science, National Taiwan University, Taipei, Taiwan
| | - Jean Lu
- Genome and Systems Biology Degree Program, College of Life Science, National Taiwan University, Taipei, Taiwan.,Genomics Research Center, Academia Sinica, Taipei, Taiwan.,National RNAi Platform/National Core Facility Program for Biotechnology, Taipei, Taiwan.,Department of Life Science, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
797
|
Espagnolle N, Balguerie A, Arnaud E, Sensebé L, Varin A. CD54-Mediated Interaction with Pro-inflammatory Macrophages Increases the Immunosuppressive Function of Human Mesenchymal Stromal Cells. Stem Cell Reports 2017; 8:961-976. [PMID: 28330617 PMCID: PMC5390105 DOI: 10.1016/j.stemcr.2017.02.008] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/09/2017] [Accepted: 02/10/2017] [Indexed: 02/08/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) sense and modulate inflammation and represent potential clinical treatment for immune disorders. However, many details of the bidirectional interaction of MSCs and the innate immune compartment are still unsolved. Here we describe an unconventional but functional interaction between pro-inflammatory classically activated macrophages (M1MΦ) and MSCs, with CD54 playing a central role. CD54 was upregulated and enriched specifically at the contact area between M1MФ and MSCs. Moreover, the specific interaction induced calcium signaling and increased the immunosuppressive capacities of MSCs dependent on CD54 mediation. Our data demonstrate that MSCs can detect an inflammatory microenvironment via a direct and physical interaction with innate immune cells. This finding opens different perspectives for MSC-based cell therapy. Unconventional but functional interaction between M1MФ and MSCs CD54-dependent M1MФ-MSC interaction increases MSC immunosuppressive properties First characterization of physical interaction between stromal cells and MΦ
Collapse
Affiliation(s)
- Nicolas Espagnolle
- STROMALab (Team 2), Université de Toulouse, EFS, INP-ENVT, INSERM U1031, UPS, 31042 Toulouse, France; EFS Pyrénées-Méditerranée, 31042 Toulouse, France
| | - Adélie Balguerie
- STROMALab (Team 2), Université de Toulouse, EFS, INP-ENVT, INSERM U1031, UPS, 31042 Toulouse, France; EFS Pyrénées-Méditerranée, 31042 Toulouse, France
| | - Emmanuelle Arnaud
- STROMALab (Team 1), Université de Toulouse, CNRS ERL5311, EFS, INP-ENVT, INSERM U1031, UPS, 31042 Toulouse, France
| | - Luc Sensebé
- STROMALab (Team 2), Université de Toulouse, EFS, INP-ENVT, INSERM U1031, UPS, 31042 Toulouse, France; EFS Pyrénées-Méditerranée, 31042 Toulouse, France.
| | - Audrey Varin
- STROMALab (Team 2), Université de Toulouse, EFS, INP-ENVT, INSERM U1031, UPS, 31042 Toulouse, France; EFS Pyrénées-Méditerranée, 31042 Toulouse, France.
| |
Collapse
|
798
|
Balbi C, Piccoli M, Barile L, Papait A, Armirotti A, Principi E, Reverberi D, Pascucci L, Becherini P, Varesio L, Mogni M, Coviello D, Bandiera T, Pozzobon M, Cancedda R, Bollini S. First Characterization of Human Amniotic Fluid Stem Cell Extracellular Vesicles as a Powerful Paracrine Tool Endowed with Regenerative Potential. Stem Cells Transl Med 2017; 6:1340-1355. [PMID: 28271621 PMCID: PMC5442724 DOI: 10.1002/sctm.16-0297] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 11/08/2016] [Accepted: 12/21/2016] [Indexed: 12/13/2022] Open
Abstract
Human amniotic fluid stem cells (hAFS) have shown a distinct secretory profile and significant regenerative potential in several preclinical models of disease. Nevertheless, little is known about the detailed characterization of their secretome. Herein we show for the first time that hAFS actively release extracellular vesicles (EV) endowed with significant paracrine potential and regenerative effect. c‐KIT+ hAFS were isolated from leftover samples of amniotic fluid from prenatal screening and stimulated to enhance EV release (24 hours 20% O2 versus 1% O2 preconditioning). The capacity of the c‐KIT+ hAFS‐derived EV (hAFS‐EV) to induce proliferation, survival, immunomodulation, and angiogenesis were investigated in vitro and in vivo. The hAFS‐EV regenerative potential was also assessed in a model of skeletal muscle atrophy (HSA‐Cre, SmnF7/F7 mice), in which mouse AFS transplantation was previously shown to enhance muscle strength and survival. hAFS secreted EV ranged from 50 up to 1,000 nm in size. In vitro analysis defined their role as biological mediators of regenerative, paracrine effects while their modulatory role in decreasing skeletal muscle inflammation in vivo was shown for the first time. Hypoxic preconditioning significantly induced the enrichment of exosomes endowed with regenerative microRNAs within the hAFS‐EV. In conclusion, this is the first study showing that c‐KIT+ hAFS dynamically release EV endowed with remarkable paracrine potential, thus representing an appealing tool for future regenerative therapy. Stem Cells Translational Medicine2017;6:1340–1355
Collapse
Affiliation(s)
- Carolina Balbi
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Martina Piccoli
- Stem Cells and Regenerative Medicine Laboratory, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Lucio Barile
- Laboratory of Molecular and Cellular Cardiology, CardioCentro Ticino Foundation_CCT, Lugano Switzerland
| | - Andrea Papait
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Andrea Armirotti
- Drug Discovery and Development Department, IIT-Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| | - Elisa Principi
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Daniele Reverberi
- Molecular Pathology Unit, IRCCS AOU San Martino - IST National Institute for Cancer Research, Genova, Italy
| | - Luisa Pascucci
- Veterinary Medicine Department, University of Perugia, Perugia, Italy
| | - Pamela Becherini
- Molecular Biology Laboratory, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Luigi Varesio
- Molecular Biology Laboratory, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Massimo Mogni
- Human Genetics Laboratory, E.O. Ospedali Galliera, Genova, Italy
| | | | - Tiziano Bandiera
- Drug Discovery and Development Department, IIT-Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| | - Michela Pozzobon
- Stem Cells and Regenerative Medicine Laboratory, Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy.,Department of Woman and Child Health, University of Padova, Padova, Italy
| | - Ranieri Cancedda
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy
| | - Sveva Bollini
- Regenerative Medicine Laboratory, Department of Experimental Medicine, University of Genova, Genova, Italy
| |
Collapse
|
799
|
Somal A, Bhat IA, B I, Singh AP, Panda BSK, Desingu PA, Pandey S, Bharti MK, Pal A, Saikumar G, Chandra V, Sharma GT. Impact of Cryopreservation on Caprine Fetal Adnexa Derived Stem Cells and Its Evaluation for Growth Kinetics, Phenotypic Characterization, and Wound Healing Potential in Xenogenic Rat Model. J Cell Physiol 2017; 232:2186-2200. [PMID: 27966782 DOI: 10.1002/jcp.25731] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 12/13/2016] [Indexed: 12/12/2022]
Abstract
This study was conducted to know the impact of cryopreservation on caprine fetal adnexa derived mesenchymal stem cells (MSCs) on the basic stem cell characteristics. Gravid caprine uteri (2-3 months) were collected from local abattoir to derive (amniotic fluid [cAF], amniotic sac [cAS], Wharton's jelly [cWJ], and cord blood [cCB]) MSCs and expanded in vitro. Cells were cryopreserved at 3rd passage (P3) using 10% DMSO. Post-thaw viability and cellular properties were assessed. Cells were expanded to determine growth kinetics, tri-lineage differentiation, localization, and molecular expression of MSCs and pluripotency markers; thereafter, these cells were transplanted in the full-thickness (2 × 2cm2 ) rat skin wound to determine their wound healing potential. The post-thaw (pt) growth kinetics study suggested that cWJ MSCs expanded more rapidly with faster population doubling time (PDT) than that of other fetal adnexa MSCs. The relative mRNA expression of surface antigens (CD73, CD90, and CD 105) and pluripotency markers (Oct4, KLF, and cMyc) was higher in cWJ MSCs in comparison to cAS, cAF, and cCB MSCs post-thaw. The percent wound contraction on 7th day was more than 50% for all the MSC-treated groups (pre and post-thaw), against 39.55% in the control group. On day 28th, 99% and more wound contraction was observed in cAF, cAF-pt, cAS-pt, cWJ, cWJ-pt, and cCB, MSCs with better scores for epithelization, neovascularization, and collagen characteristics at a non-significant level. It is concluded that these MSCs could be successfully cryopreserved without altering their stemness and wound healing properties. J. Cell. Physiol. 232: 2186-2200, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Anjali Somal
- Division of Physiology and Climatology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Irfan A Bhat
- Division of Physiology and Climatology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Indu B
- Division of Physiology and Climatology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Anuj P Singh
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Bibhudatta S K Panda
- Division of Physiology and Climatology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Perumal A Desingu
- Division of Veterinary Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Sriti Pandey
- Division of Physiology and Climatology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Mukesh K Bharti
- Division of Physiology and Climatology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Amar Pal
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Guttula Saikumar
- Division of Veterinary Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Vikash Chandra
- Division of Physiology and Climatology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Guttula Taru Sharma
- Division of Physiology and Climatology, ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| |
Collapse
|
800
|
Ragni E, Banfi F, Barilani M, Cherubini A, Parazzi V, Larghi P, Dolo V, Bollati V, Lazzari L. Extracellular Vesicle-Shuttled mRNA in Mesenchymal Stem Cell Communication. Stem Cells 2017; 35:1093-1105. [PMID: 28164431 DOI: 10.1002/stem.2557] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 11/23/2016] [Accepted: 11/27/2016] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cells (MSC) are multipotent cells able to differentiate into several cell types, hence providing cell reservoirs for therapeutic applications. The absence of detectable MSC homing at injury sites suggests that paracrine functions could, at least in part, be mediated by extracellular vesicles (EVs); EVs are newly identified players that are studied mainly as predictive or diagnostic biomarkers. Together with their clinical interests, EVs have recently come to the fore for their role in cell-to-cell communication. In this context, we investigated gene-based communication mechanisms in EVs generated by bone marrow and umbilical cord blood MSC (BMMSC and CBMSC, respectively). Both MSC types released vesicles with similar physical properties, although CBMSC were able to secrete EVs with faster kinetics. A pattern of preferentially incorporated EV transcripts was detected with respect to random internalization from the cytosol, after a validated normalization procedure was established. In the paradigm where EVs act as bioeffectors educating target cells, we demonstrated that kidney tubular cells lacking IL-10 expression and exposed to BMMSC-EVs and CBMSC-EVs acquired the IL-10 mRNA, which was efficiently translated into the corresponding protein. These findings suggest that horizontal mRNA transfer through EVs is a new mechanism in the MSC restoring ability observed in vivo that is here further demonstrated in an in vitro rescue model after acute cisplatin injury of tubular cells. Stem Cells 2017;35:1093-1105.
Collapse
Affiliation(s)
- Enrico Ragni
- Cell Factory, Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Federica Banfi
- Cell Factory, Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Mario Barilani
- Cell Factory, Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Industrial Engineering, University of Padova, Padova, Italy
| | - Alessandro Cherubini
- Cell Factory, Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valentina Parazzi
- Cell Factory, Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Paola Larghi
- Autoimmunity Program, Istituto Nazionale di Genetica Molecolare "Romeo Ed Enrica Invernizzi", Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Vincenza Dolo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Valentina Bollati
- EPIGET - Epidemiology, Epigenetics and Toxicology Lab, Department of Clinical Sciences and Community Health, University of Milano, Milan, Italy.,Epidemiology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lorenza Lazzari
- Cell Factory, Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|