1
|
Pagotto VPF, Cacere PV, Altran SC, Dantas VANC, Furuya TK, Santos DLS, Alves MJF, Uno M, Camargo CP, Gemperli R. Sequential Applications of Adipose Tissue-derived Stem Cells Enhance Healing in Complex Wounds: Experimental Model. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2025; 13:e6718. [PMID: 40291637 PMCID: PMC12026421 DOI: 10.1097/gox.0000000000006718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 03/05/2025] [Indexed: 04/30/2025]
Abstract
Background Complex wounds represent a challenge to global health, incurring significant healthcare costs and compromising patients' quality of life. Recently, research has investigated the potential of adipose tissue-derived stem cells (ASC) for therapeutic stem cells' immunomodulatory properties. This study aimed to evaluate the effect of sequential applications of ASC in an experimental model of a complex wound. Methods Thirty male Wistar rats were subjected to a complex wound model of enterocutaneous fistula. After 4 weeks, the rats were divided into 3 groups: control, 2 applications of culture medium, and 2 applications of 1 × 106 allogeneic ASC. The animals were euthanized and analyzed 4 weeks after the first application regarding the macroscopic reduction of the wound, histopathologic changes, and gene expression related to wound healing (Cd68, Il1rap, Il10, Mmp3, Mmp9, and Tnf). Results Animals treated with ASC showed a reduction in wound diameter of 68% compared with the control group (P = 0.002) and a reduction of 65% compared with the culture medium group (P = 0.011). The ASC group also showed a more than 100% increase in blood vessel count compared with the control group (P = 0.02). Gene expression analysis showed a decrease in the Mmp9 levels in the ASC group compared with the control group. Conclusions This study demonstrated that the treatment with ASC improves the healing of enterocutaneous fistula wounds.
Collapse
Affiliation(s)
- Vitor P F Pagotto
- From the Microsurgery and Plastic Surgery Laboratory (LIM04), Faculdade de Medicina, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Paula V Cacere
- From the Microsurgery and Plastic Surgery Laboratory (LIM04), Faculdade de Medicina, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Silvana C Altran
- From the Microsurgery and Plastic Surgery Laboratory (LIM04), Faculdade de Medicina, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Viviane A N C Dantas
- Multidisciplinary Research Center, School of Arts, Sciences and Humanities, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Tatiane K Furuya
- Center for Translational Research in Oncology (LIM24), Instituto do Câncer do Estado de São Paulo (ICESP), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, São Paulo, Brazil
- Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Deborah L S Santos
- Comissão de Residência Multiprofissional (COREMU), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, São Paulo, Brazil
| | - Maria José F Alves
- Center for Translational Research in Oncology (LIM24), Instituto do Câncer do Estado de São Paulo (ICESP), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, São Paulo, Brazil
- Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Miyuki Uno
- Center for Translational Research in Oncology (LIM24), Instituto do Câncer do Estado de São Paulo (ICESP), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, São Paulo, Brazil
- Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Cristina P Camargo
- From the Microsurgery and Plastic Surgery Laboratory (LIM04), Faculdade de Medicina, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Rolf Gemperli
- From the Microsurgery and Plastic Surgery Laboratory (LIM04), Faculdade de Medicina, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| |
Collapse
|
2
|
Wang X, Shang D, Chen J, Cheng S, Chen D, Zhang Z, Liu C, Yu J, Cao H, Li L, Li L. Serum metabolomics reveals the effectiveness of human placental mesenchymal stem cell therapy for Crohn's disease. Talanta 2024; 277:126442. [PMID: 38897006 DOI: 10.1016/j.talanta.2024.126442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 06/10/2024] [Accepted: 06/15/2024] [Indexed: 06/21/2024]
Abstract
Mesenchymal stem cell (MSC) therapy offers a promising cure for Crohn's disease (CD), however, its therapeutic effects vary significantly due to individual differences. Therefore, identifying easily detectable biomarkers is essential to assess the efficacy of MSC therapy. In this study, SAMP1/Yit mice were used as a model of CD, which develop spontaneous chronic ileitis, closely resembling the characteristics present in CD patients. Serum metabolic alterations during treatment were analyzed, through the application of differential 12C-/13C-dansylation labeling liquid chromatography-mass spectrometry. Based on the significant differences and time-varying trends of serum amine/phenol-containing metabolites abundance between the control group, the model group, and the treatment group, four serum biomarkers were ultimately screened for evaluating the efficacy of MSC treatment for CD, namely 4-hydroxyphenylpyruvate, 4-hydroxyphenylacetaldehyde, caffeate, and N-acetyltryptamine, whose abundances both increased in the serum of CD model mice and decreased after MSC treatment. These metabolic alterations were associated with tyrosine metabolism, which was validated by the dysregulation of related enzymes. The discovery of biomarkers may help to improve the targeting and effectiveness of treatment and provide innovative prospects for the clinical application of MSC for CD.
Collapse
Affiliation(s)
- Xiao Wang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan City 250117, China
| | - Dandan Shang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan City 250117, China
| | - Junyao Chen
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Sheng Cheng
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Deying Chen
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Zhehua Zhang
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Chaoxu Liu
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
| | - Jiong Yu
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan City 250117, China; State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China; Zhejiang Key Laboratory for Diagnosis and Treatment of Physic-chemical and Aging-related Injuries, 79 Qingchun Rd, Hangzhou City 310003, China.
| | - Hongcui Cao
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan City 250117, China; State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China; Zhejiang Key Laboratory for Diagnosis and Treatment of Physic-chemical and Aging-related Injuries, 79 Qingchun Rd, Hangzhou City 310003, China.
| | - Liang Li
- Department of Chemistry, University of Alberta, Edmonton, Alberta, T6G 2G2, Canada
| | - Lanjuan Li
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan City 250117, China; State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China
| |
Collapse
|
3
|
Francesca T, Elena LP, Barbara DSA, Marta DS, Marco T, Maria CA, Carmela P, Francesco M, Francesco D, Adriana C, Serena M. An analysis of the immunomodulatory properties of human spheroids from adipose-derived stem cells. Life Sci 2023; 321:121610. [PMID: 36948391 DOI: 10.1016/j.lfs.2023.121610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 03/09/2023] [Accepted: 03/17/2023] [Indexed: 03/24/2023]
Abstract
AIMS Current methods to induce tolerance following allotransplantation or in autoimmunity carry significant morbidity, and research is very active in investigating alternative methods which could minimize toxicity. Spheroids from adipose stem cells (SASCs) are increasingly gaining interest, they hold a great proliferative and differentiating potential. An immunomodulatory effect has not been investigated on SASCs yet. In this study, we analysed the immunomodulatory properties of SASCs and compared them to ADSCs. MAIN METHODS Adipose stem cells (SASCs and ADSCs) and peripheral blood mononuclear cells (PBMCs) were collected from healthy individuals. We analysed the cytokine production and proliferation of T cells co-cultured with adipose samples or conditioned medium. KEY FINDINGS SASCs modulated cytokines production and proliferation of heterologous and autologous T cells. In the heterologous assays, we observed a reduction of IFNγ and IL-17 production and an increase of IL-9 in γδ T cells. The soluble factors present in SASCs sovranatants were also able to induce a slight reduction of IFNγ and an increase of IL-9, IL-10 and IL-17 while they could not modulate the proliferative ability of γδ T cells. In the autologous assays, we observed a reduction of the proliferative ability of T cells in co-culture at different ratios with SASCs. Analysis of the SASCs secretome showed an increased IL-5, IL-10, IL-4 and IL-13 production compared to the ADSCs one, demonstrating greater anti-inflammatory properties. SIGNIFICANCE Our preliminary results support the idea that SASCs exert more pronounced biological immune modulation compared to the classical adherent ADSCs, especially in heterologous experimental settings.
Collapse
Affiliation(s)
- Toia Francesca
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Plastic and Reconstructive Surgery Section, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy; Section of Plastic and Reconstructive Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy; Plastic and Reconstructive Unit, Department of Oncology, Azienda Ospedaliera Universitaria Policlinico "Paolo Giaccone", 90127 Palermo, Italy
| | - Lo Presti Elena
- National Research Council (CNR)-Institute for Biomedical Research and Innovation (IRIB), 90146 Palermo, Italy
| | - Di Stefano Anna Barbara
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Plastic and Reconstructive Surgery Section, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy.
| | - Di Simone Marta
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy; Department of Biomedicine, Neurosciences and Advanced Diagnosis, University of Palermo, Palermo, Italy
| | - Trapani Marco
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Plastic and Reconstructive Surgery Section, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Corsale Anna Maria
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy; Department of Biomedicine, Neurosciences and Advanced Diagnosis, University of Palermo, Palermo, Italy
| | - Picone Carmela
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy; Department of Biomedicine, Neurosciences and Advanced Diagnosis, University of Palermo, Palermo, Italy
| | - Moschella Francesco
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Plastic and Reconstructive Surgery Section, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Dieli Francesco
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy; Department of Biomedicine, Neurosciences and Advanced Diagnosis, University of Palermo, Palermo, Italy
| | - Cordova Adriana
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Plastic and Reconstructive Surgery Section, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy; Section of Plastic and Reconstructive Surgery, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy; Plastic and Reconstructive Unit, Department of Oncology, Azienda Ospedaliera Universitaria Policlinico "Paolo Giaccone", 90127 Palermo, Italy
| | - Meraviglia Serena
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR), University of Palermo, Palermo, Italy; Department of Biomedicine, Neurosciences and Advanced Diagnosis, University of Palermo, Palermo, Italy
| |
Collapse
|
4
|
Isolation and characterization of novel peptides from fermented products of Lactobacillus for ulcerative colitis prevention and treatment. FOOD SCIENCE AND HUMAN WELLNESS 2022. [DOI: 10.1016/j.fshw.2022.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
5
|
Ruiz-Magaña MJ, Llorca T, Martinez-Aguilar R, Abadia-Molina AC, Ruiz-Ruiz C, Olivares EG. Stromal cells of the endometrium and decidua: in search of a name and an identity. Biol Reprod 2022; 107:1166-1176. [PMID: 35947987 DOI: 10.1093/biolre/ioac158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 11/14/2022] Open
Abstract
Human endometrial and decidual stromal cells are the same cells in different environments (non-pregnancy and pregnancy, respectively). Although some authors consider decidual stromal cells to arise solely from the differentiation of endometrial stromal cells, this is a debatable issue given that decidualization processes do not end with the formation of the decidua, as shown by the presence of stromal cells from both the endometrium and decidua in both undifferentiated (non-decidualized) and decidualized states. Furthermore, recent functional and transcriptomic results have shown that there are differences in the decidualization process of endometrial and decidual stromal cells, with the latter having a greater decidualization capacity than the former. These differences suggest that in the terminology and study of their characteristics, endometrial and decidual stromal cells should be clearly distinguished, as should their undifferentiated or decidualized status. There is, however, considerable confusion in the designation and identification of uterine stromal cells. This confusion may impede a judicious understanding of the functional processes in normal and pathological situations. In the present article we analyse the different terms used in the literature for different types of uterine stromal cells, and propose that a combination of differentiation status (undifferentiated, decidualized) and localization (endometrium, decidua) criteria should be used to arrive at a set of accurate, unambiguous terms. The cell identity of uterine stromal cells is also a debatable issue: phenotypic, functional and transcriptomic studies in recent decades have related these cells to different established cells. We discuss the relevance of these associations in normal and pathological situations.
Collapse
Affiliation(s)
- Maria Jose Ruiz-Magaña
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Armilla, Granada, Spain
| | - Tatiana Llorca
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Armilla, Granada, Spain.,Departamento de Bioquímica y Biología Molecular III e Inmunología, Universidad de Granada, Granada, Spain
| | - Rocio Martinez-Aguilar
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Armilla, Granada, Spain.,Departamento de Bioquímica y Biología Molecular III e Inmunología, Universidad de Granada, Granada, Spain
| | - Ana Clara Abadia-Molina
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Armilla, Granada, Spain.,Departamento de Bioquímica y Biología Molecular III e Inmunología, Universidad de Granada, Granada, Spain
| | - Carmen Ruiz-Ruiz
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Armilla, Granada, Spain.,Departamento de Bioquímica y Biología Molecular III e Inmunología, Universidad de Granada, Granada, Spain
| | - Enrique G Olivares
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Armilla, Granada, Spain.,Departamento de Bioquímica y Biología Molecular III e Inmunología, Universidad de Granada, Granada, Spain.,Unidad de Gestión Clínica Laboratorios, Complejo Hospitalario Universitario de Granada, Granada, Spain
| |
Collapse
|
6
|
Elhag DA, Kumar M, Saadaoui M, Akobeng AK, Al-Mudahka F, Elawad M, Al Khodor S. Inflammatory Bowel Disease Treatments and Predictive Biomarkers of Therapeutic Response. Int J Mol Sci 2022; 23:ijms23136966. [PMID: 35805965 PMCID: PMC9266456 DOI: 10.3390/ijms23136966] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/05/2022] [Accepted: 06/06/2022] [Indexed: 02/08/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic immune-mediated inflammation of the gastrointestinal tract with a highly heterogeneous presentation. It has a relapsing and remitting clinical course that necessitates lifelong monitoring and treatment. Although the availability of a variety of effective therapeutic options including immunomodulators and biologics (such as TNF, CAM inhibitors) has led to a paradigm shift in the treatment outcomes and clinical management of IBD patients, some patients still either fail to respond or lose their responsiveness to therapy over time. Therefore, according to the recent Selecting Therapeutic Targets in Inflammatory Bowel Disease (STRIDE-II) recommendations, continuous disease monitoring from symptomatic relief to endoscopic healing along with short- and long-term therapeutic responses are critical for providing IBD patients with a tailored therapy algorithm. Moreover, considering the high unmet need for novel therapeutic approaches for IBD patients, various new modulators of cytokine signaling events (for example, JAK/TYK inhibitors), inhibitors of cytokines (for example IL-12/IL-23, IL-22, IL-36, and IL-6 inhibitors), anti-adhesion and migration strategies (for example, β7 integrin, sphingosine 1-phosphate receptors, and stem cells), as well as microbial-based therapeutics to decolonize the bed buds (for example, fecal microbiota transplantation and bacterial inhibitors) are currently being evaluated in different phases of controlled clinical trials. This review aims to offer a comprehensive overview of available treatment options and emerging therapeutic approaches for IBD patients. Furthermore, predictive biomarkers for monitoring the therapeutic response to different IBD therapies are also discussed.
Collapse
Affiliation(s)
- Duaa Ahmed Elhag
- Research Department, Sidra Medicine, Doha 26999, Qatar; (D.A.E.); (M.K.); (M.S.)
| | - Manoj Kumar
- Research Department, Sidra Medicine, Doha 26999, Qatar; (D.A.E.); (M.K.); (M.S.)
| | - Marwa Saadaoui
- Research Department, Sidra Medicine, Doha 26999, Qatar; (D.A.E.); (M.K.); (M.S.)
| | - Anthony K. Akobeng
- Division of Gastroenterology, Hepatology and Nutrition, Sidra Medicine, Doha 26999, Qatar; (A.K.A.); (F.A.-M.); (M.E.)
| | - Fatma Al-Mudahka
- Division of Gastroenterology, Hepatology and Nutrition, Sidra Medicine, Doha 26999, Qatar; (A.K.A.); (F.A.-M.); (M.E.)
| | - Mamoun Elawad
- Division of Gastroenterology, Hepatology and Nutrition, Sidra Medicine, Doha 26999, Qatar; (A.K.A.); (F.A.-M.); (M.E.)
| | - Souhaila Al Khodor
- Research Department, Sidra Medicine, Doha 26999, Qatar; (D.A.E.); (M.K.); (M.S.)
- Correspondence:
| |
Collapse
|
7
|
El-Nakeep S, Shawky A, Abbas SF, Abdel Latif O. Stem cell transplantation for induction of remission in medically refractory Crohn's disease. Cochrane Database Syst Rev 2022; 5:CD013070. [PMID: 35556242 PMCID: PMC9099217 DOI: 10.1002/14651858.cd013070.pub2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Crohn's disease (CD) is an inflammatory bowel disease that causes inflammation and stricture, of any part of the mucosa and the gut wall. It forms skip lesions, sparing the areas in between the affected parts of the gastrointestinal tract. Crohn's disease could have one of three complications; fistula, intestinal obstruction due to stricture, or gastrointestinal inflammation presenting as severe diarrhoea. Stem cell therapy (SCT) is an innovative treatment that has been recently used in CD. The exact role of SCT in CD is still unclear. Stem cells modify the immunity of the patients or act as a "reset tool" for the immune system as in the case of systemically-injected stem cells, or regenerate the affected area of necrotic and inflammatory tissue as in the case of local injection into the lesion. Stem cells are a wide variety of cells including pluripotent stem cells or differentiated stem cells. The hazards range from rejection to symptomatic manifestations as fever or increase infection. OBJECTIVES: The objective of this Cochrane systematic review is to assess the effects of stem cell transplantation compared to standard of care alone or with placebo on efficacy and safety outcomes in patients with refractory CD. SEARCH METHODS We searched MEDLINE, Embase, Cochrane Central Register of Controlled Trials (CENTRAL), and clinical trial registries (Clinicaltrials.gov, World Health Organization-International Clinical Trials Registry Platform WHO ICTRP) from inception to 19 March 2021, without any language, publication year, or publication status restrictions. In addition, we searched references of included studies and review articles for further references. An update of the published studies was done during the writing of the review. SELECTION CRITERIA We included only randomised controlled trials (RCTs) that assessed the effectiveness and safety of SCT in refractory CD versus standard care alone (control) or with placebo. DATA COLLECTION AND ANALYSIS Two review authors (SEN and SFA) independently screened the studies retrieved from the search results for inclusion, extracted data and assessed the risk of bias. Any disagreement was resolved through a consensus between the authors. We used standard methodological procedures expected by Cochrane. MAIN RESULTS We conducted our search on 19 March 2021 and identified 639 records. We added two records by a manual search of the published reviews on the topic to a total of 641 records. The Covidence program removed 125 duplicates making a total of 516 reports. Two review authors (SEN and SFA) screened titles and abstracts and excluded 451 records with the remaining 65 for full-text records screened independently by the two authors; only 18 studies were considered for inclusion. We included seven RCTs with a total of 442 participants for the meta-analysis. The intervention group included 234 patients, and the control group included 208 patients. Nine trials are ongoing and, two abstracts are awaiting classification. All patients in the control and intervention groups received the standard therapy for CD. Only three studies used blinding methods for the control group in the form of a placebo, with one study of the three stated that the blinding method was inefficient. The patients and personnel were aware of the intervention in the rest of the four studies as they were open-label trials. However, the effect of unblinding was balanced by the low risk of detection bias in five of the included studies. The evidence is uncertain about the effect of SCT on achieving clinical remission as compared to control/placebo (risk ratio (RR) 1.88, 95% Confidence Interval (CI) 0.80 to 4.41; 3 studies; low-certainty evidence). The evidence is very uncertain about the effect of SCT on achieving Crohn's Disease Activity Index (CDAI) <150 at 24 weeks compared to control (RR1.02 95% CI 0.67 to 1.56; 4 studies; very-low certainty evidence). SCT is likely to achieve fistula closure as compared to the control/placebo both in the short term (RR 1.48, 95% CI 1.12 to 1.96); low-certainty evidence) and in the long term (RR 1.42, 95% CI 1.09 to 1.87; 4 studies; low-certainty evidence) follow-up. The evidence is very uncertain about the effect of SCT to cause no difference in the number of total adverse events as compared to the control/placebo (RR 0.99, 95% CI [0.88 to 1.13); 4 studies; very-low-certainty evidence). However, SCT is likely to increase the number of serious adverse events as compared to the control/placebo (RR 1.22, 95% CI 0.88 to 1.67; 7 studies; low-certainty evidence). The evidence is very uncertain about the effect of SCT to decrease the withdrawal due to adverse events as compared to the control/placebo (RR 0.78, 95% CI 0.32 to 1.89; 3 studies; very-low certainty evidence). Funding by pharmaceutical companies was found in three studies, with one including more than 50% of our studied population. AUTHORS' CONCLUSIONS SCT shows an uncertain effect on clinical remission with low certainty of evidence. SCT shows an uncertain effect on CDAI score to reach <150 after 24 weeks of treatment, with very low certainty evidence. SCT shows beneficial effects on fistula-closure during short and long-term follow-up with low-certainty evidence in both outcomes. There was no change in the total number of adverse events with SCT as compared to control, with very low certainty evidence. While there was a moderate effect on increasing the number of serious adverse events in the SCT group, as compared to the control with low-certainty evidence. Withdrawal due to adverse events was slightly higher in the control group with very low certainty evidence. All the participants were refractory to standard medical treatment, but the number of participants was small, this may limit the generalizability of the results. Further research is needed for validation. More objective outcomes are needed in the assessment of stem cell effectiveness in the treatment of Crohn's disease, especially the intestinal CD subtype; with standardization of the dose, methods of stem cell preparation, route of administration, and inclusion criteria to the studies to achieve clear results.
Collapse
Affiliation(s)
- Sarah El-Nakeep
- Gastroenterology and Hepatology Unit, Internal Medicine Department , Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ahmed Shawky
- Gastroenterology and Hepatology Unit, Internal Medicine Department , Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sara F Abbas
- Extended Medical Program, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Osama Abdel Latif
- Allergy and Clinical Immunology Unit, Internal Medicine Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
8
|
Kouchakian MR, Baghban N, Moniri SF, Baghban M, Bakhshalizadeh S, Najafzadeh V, Safaei Z, Izanlou S, Khoradmehr A, Nabipour I, Shirazi R, Tamadon A. The Clinical Trials of Mesenchymal Stromal Cells Therapy. Stem Cells Int 2021; 2021:1634782. [PMID: 34745268 PMCID: PMC8566082 DOI: 10.1155/2021/1634782] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 08/22/2021] [Accepted: 10/05/2021] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are a heterogeneous population of adult stem cells, which are multipotent and possess the ability to differentiate/transdifferentiate into mesodermal and nonmesodermal cell lineages. MSCs display broad immunomodulatory properties since they are capable of secreting growth factors and chemotactic cytokines. Safety, accessibility, and isolation from patients without ethical concern make MSCs valuable sources for cell therapy approaches in autoimmune, inflammatory, and degenerative diseases. Many studies have been conducted on the application of MSCs as a new therapy, but it seems that a low percentage of them is related to clinical trials, especially completed clinical trials. Considering the importance of clinical trials to develop this type of therapy as a new treatment, the current paper is aimed at describing characteristics of MSCs and reviewing relevant clinical studies registered on the NIH database during 2016-2020 to discuss recent advances on MSC-based therapeutic approaches being used in different diseases.
Collapse
Affiliation(s)
- Mohammad Reza Kouchakian
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Neda Baghban
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Seyedeh Farzaneh Moniri
- Department of Anatomical Sciences, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mandana Baghban
- Department of Obstetrics and Gynecology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shabnam Bakhshalizadeh
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Vahid Najafzadeh
- Department of Veterinary and Animal Sciences, Anatomy & Biochemistry Section, University of Copenhagen, Copenhagen, Denmark
| | - Zahra Safaei
- Department of Obstetrics and Gynecology, School of Medicine, Amir Al Mo'menin Hospital, Amir Al Mo'menin IVF Center, Arak University of Medical Sciences, Arak, Iran
| | - Safoura Izanlou
- Department of Nursing, School of Nursing, Larestan University of Medical Sciences, Larestan, Iran
| | - Arezoo Khoradmehr
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Iraj Nabipour
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Reza Shirazi
- Department of Anatomy, School of Medical Sciences, Medicine & Health, UNSW Sydney, Sydney, Australia
| | - Amin Tamadon
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| |
Collapse
|
9
|
Mallis P, Michalopoulos E, Chatzistamatiou T, Giokas CS. Interplay between mesenchymal stromal cells and immune system: clinical applications in immune-related diseases. EXPLORATION OF IMMUNOLOGY 2021. [DOI: 10.37349/ei.2021.00010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/07/2021] [Indexed: 11/08/2024]
Abstract
Mesenchymal stromal cells (MSCs) are a mesodermal stem cell population, with known self-renewal and multilineage differentiation properties. In the last century, MSCs have been widely used in regenerative medicine and tissue engineering approaches. MSCs initially were isolated from bone marrow aspirates, but currently have been identified in a great number of tissues of the human body. Besides their utilization in regenerative medicine, MSCs possess significant immunoregulatory/immunosuppressive properties, through interaction with the cells of innate and adaptive immunity. MSCs can exert their immunomodulatory properties with either cell-cell contact or via paracrine secretion of molecules, such as cytokines, growth factors and chemokines. Of particular importance, the MSCs’ immunomodulatory properties are explored as promising therapeutic strategies in immune-related disorders, such as autoimmune diseases, graft versus host disease, cancer. MSCs may also have an additional impact on coronavirus disease-19 (COVID-19), by attenuating the severe symptoms of this disorder. Nowadays, a great number of clinical trials, of MSC-mediated therapies are evaluated for their therapeutic potential. In this review, the current knowledge on cellular and molecular mechanisms involved in MSC-mediated immunomodulation were highlighted. Also, the most important aspects, regarding their potential application in immune-related diseases, will be highlighted. The broad application of MSCs has emerged their role as key immunomodulatory players, therefore their utilization in many disease situations is full of possibilities for future clinical treatment.
Collapse
Affiliation(s)
- Panagiotis Mallis
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, 11527 Athens, Greece
| | - Efstathios Michalopoulos
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, 11527 Athens, Greece
| | - Theofanis Chatzistamatiou
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, 11527 Athens, Greece 2Histocompatibility & Immunogenetics Lab, Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, 11527 Athens, Greece
| | | |
Collapse
|
10
|
Crippa S, Santi L, Berti M, De Ponti G, Bernardo ME. Role of ex vivo Expanded Mesenchymal Stromal Cells in Determining Hematopoietic Stem Cell Transplantation Outcome. Front Cell Dev Biol 2021; 9:663316. [PMID: 34017834 PMCID: PMC8129582 DOI: 10.3389/fcell.2021.663316] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 03/17/2021] [Indexed: 02/06/2023] Open
Abstract
Overall, the human organism requires the production of ∼1 trillion new blood cells per day. Such goal is achieved via hematopoiesis occurring within the bone marrow (BM) under the tight regulation of hematopoietic stem and progenitor cell (HSPC) homeostasis made by the BM microenvironment. The BM niche is defined by the close interactions of HSPCs and non-hematopoietic cells of different origin, which control the maintenance of HSPCs and orchestrate hematopoiesis in response to the body’s requirements. The activity of the BM niche is regulated by specific signaling pathways in physiological conditions and in case of stress, including the one induced by the HSPC transplantation (HSCT) procedures. HSCT is the curative option for several hematological and non-hematological diseases, despite being associated with early and late complications, mainly due to a low level of HSPC engraftment, impaired hematopoietic recovery, immune-mediated graft rejection, and graft-versus-host disease (GvHD) in case of allogenic transplant. Mesenchymal stromal cells (MSCs) are key elements of the BM niche, regulating HSPC homeostasis by direct contact and secreting several paracrine factors. In this review, we will explore the several mechanisms through which MSCs impact on the supportive activity of the BM niche and regulate HSPC homeostasis. We will further discuss how the growing understanding of such mechanisms have impacted, under a clinical point of view, on the transplantation field. In more recent years, these results have instructed the design of clinical trials to ameliorate the outcome of HSCT, especially in the allogenic setting, and when low doses of HSPCs were available for transplantation.
Collapse
Affiliation(s)
- Stefania Crippa
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ludovica Santi
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Margherita Berti
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giada De Ponti
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| | - Maria Ester Bernardo
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Pediatric Immunohematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, Milan, Italy.,University Vita-Salute San Raffaele, Faculty of Medicine, Milan, Italy
| |
Collapse
|
11
|
Stromal-vascular fraction of adipose tissue as an alternative source of cellular material for regenerative medicine. UKRAINIAN BIOCHEMICAL JOURNAL 2021. [DOI: 10.15407/ubj93.01.040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Adipose tissue is the most convenient source of cellular material for regenerative medicine as it can be obtained in significant quantities via cosmetic liposuction, lipoaspiration of subcutaneous fat or by excision of fat deposits. Adipose tissue consists of adipocytes and cells, which are the part of the stromal-vascular fraction (SVF). Different cell populations can be isolated from SVF, among which the population of adipose tissue stem cells (adipose-derived stem cells, ADSC) is especially important for regenerative medicine. SVF can be obtained relatively easily from adipose tissue (adipose tissue is an alternative to bone marrow in terms of being a source of stem cells) and used to treat various pathologies. Recent studies show that SVF not only has a therapeutic effect similar to that of ADSC, but in some cases is even more effective. The article provides the analysis of the main methods of SVF obtainment, characteristics of SVF cellular composition, its potential for use in clinical medicine and its main advantages over other sources of cellular material, including ADSC cultured in vitro, for regenerative medicine. Keywords: adipocytes, adipose-derived stem cells, regenerative medicine, stromal-vascular fraction
Collapse
|
12
|
Abstract
AbstractInflammatory bowel disease, which mainly involves Crohn's disease and ulcerative rectocolitis, is an inflammatory condition of the mucosa that can afflict any segment of the gastrointestinal tract. Despite the fact that the existing therapies result in improvement in patient's symptomatology and quality of life, there is no curative treatment. Surgical treatment involves complex procedures associated with high morbidity and mortality rates. In this context, cell therapy with stem cells has emerged as a treatment with broad potential applicability. In this study, we intended to verify the efficacy of transplantation of adipose tissue-derived stem cells in rats with intestinal inflammation induced by trinitrobenzenesulfonic acid. The cell population was isolated from the adipose tissue of inguinal region of rats and processed for culture by mechanical dissociation. The animals were evaluated with respect to clinical and biochemical aspects, as well as by macroscopic, microscopic and histological analyses. In the experimental model of bowel inflammation by 2,4,6-trinitrobenzenesulfonic acid, the infusion of adipose tissue significantly reduced the presence of adhesions in the colon and adjacent organs and decreased the activity of myeloperoxidase, a marker of neutrophil infiltration in the injured mucosa. The results suggest that cell therapy with adipose tissue can promote and/or accelerate the regeneration of damaged intestinal mucosa. It is concluded that the presence of adhesions and the determination of myeloperoxidase activity provide indications that adipose tissue can promote and/or accelerate the regeneration of inflammatory bowel mucosa.
Collapse
|
13
|
Sharma P, Kumar A, Dey AD, Behl T, Chadha S. Stem cells and growth factors-based delivery approaches for chronic wound repair and regeneration: A promise to heal from within. Life Sci 2021; 268:118932. [PMID: 33400933 DOI: 10.1016/j.lfs.2020.118932] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/03/2020] [Accepted: 12/12/2020] [Indexed: 02/06/2023]
Abstract
The sophisticated chain of cellular and molecular episodes during wound healing includes cell migration, cell proliferation, deposition of extracellular matrix, and remodelling and are onerous to replicate. Encapsulation of growth factors (GFs) and Stem cell-based (SCs) has been proclaimed to accelerate healing by transforming every phase associated with wound healing to enhance skin regeneration. Therapeutic application of mesenchymal stem cells (MSCs), embryonic stem cells (ESCs) and induced pluripotent stem cells (PSCs) provides aid in wound fixing, tissue integrity restoration and function of impaired tissue. Several scientific studies have established the essential role GFs in wound healing and their reduced degree in the chronic wound. The overall limitation includes half-life, unfriendly microhabitat abundant with protease, and inadequate delivery approaches results in decreased delivery of effective amounts in a suitable time-based fashion. Advancements in the area of reformative medicine as well as tissue engineering have offered techniques competent of dispensing SCs and GFs in site-oriented manner. The progress in nanotechnology-based approaches attracts researcher to study and evaluate the potential of this SCs and GFs based therapy in chronic wounds. These techniques embrace the polymeric regime viz., nano-formulations, hydrogels, liposomes, scaffolds, nanofibers, metallic nanoparticles, lipid-based nanoparticles and dendrimers that have established better retort through targeting tissues when GFs and SCs are transported via these humans made devices. Assumed the current problems, improvements in delivery approaches and difficulties offered by chronic wounds, we hope to show that encapsulation of SCs and GFs loaded nanoformulations therapies is the rational next step in improving wound care.
Collapse
Affiliation(s)
- Preety Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Asmita Deka Dey
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Swati Chadha
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
14
|
Abdullah M, Kegel S, Gunasekaran M, Saha P, Fu X, Mishra R, Sharma S, Sunjay Kaushal. Stem Cell Therapy in Single-Ventricle Physiology: Recent Progress and Future Directions. Semin Thorac Cardiovasc Surg Pediatr Card Surg Annu 2021; 24:67-76. [PMID: 34116785 DOI: 10.1053/j.pcsu.2021.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 02/08/2021] [Accepted: 03/09/2021] [Indexed: 12/29/2022]
Abstract
Current surgical and medical treatment options for single ventricle physiology conditions remain palliative. On the long term, despite treatment, the systemic ventricle has a significant risk of developing failure. There are unmet needs to develop novel treatment modalities to help ameliorate the ventricular dysfunction. Advances in the field of stem cell therapy have been promising for the treatment of heart failure. Numerous stem cell populations have been identified. Preclinical studies in small and large animal models provide evidence for effectiveness of this treatment modality and reveal several mechanisms of action by which stem cells exert their effect. Many clinical trials have been designed to further investigate the therapeutic potential that stem cell therapy may hold for pediatric populations with single ventricle physiology. In this review, we discuss the stem cell types used in these populations, some preclinical studies, and the clinical trials of stem cell therapy in single ventricle patients.
Collapse
Affiliation(s)
| | - Samantha Kegel
- University of Maryland School of Medicine, Baltimore, Maryland
| | - Muthukumar Gunasekaran
- Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Feinburg School of Medicine, Chicago, Illinois
| | - Progyaparamita Saha
- Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Feinburg School of Medicine, Chicago, Illinois
| | - Xuebin Fu
- Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Feinburg School of Medicine, Chicago, Illinois
| | - Rachana Mishra
- Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Feinburg School of Medicine, Chicago, Illinois
| | - Sudhish Sharma
- Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Feinburg School of Medicine, Chicago, Illinois
| | - Sunjay Kaushal
- Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Feinburg School of Medicine, Chicago, Illinois.
| |
Collapse
|
15
|
Singh S, Varshney A, Borkar N, Jindal A, Padhi P, Ahmed I, Srivastava N. Clinical Utility of Stem Cells in Congenital Anomalies: New Horizons in Pediatric Surgery. Indian J Surg 2020. [DOI: 10.1007/s12262-020-02264-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
16
|
Hou H, Zhang L, Duan L, Liu Y, Han Z, Li Z, Cao X. Spatio-Temporal Metabolokinetics and Efficacy of Human Placenta-Derived Mesenchymal Stem/Stromal Cells on Mice with Refractory Crohn's-like Enterocutaneous Fistula. Stem Cell Rev Rep 2020; 16:1292-1304. [PMID: 33011925 DOI: 10.1007/s12015-020-10053-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2020] [Indexed: 12/14/2022]
Abstract
Crohn's disease (CD) with externally fistulizing openings indicates the aggressive and relapsing manifestation and results in undesirable long-term outcomes of patients. MSC-based approach combined with multidisciplinary strategy has mandated a redefinition of the administration and management of numerous recurrent and refractory diseases whereas the spatio-temporal evaluation of the metabolokinetics and efficacy of MSCs on intractable CD with enterocutaneous fistula (EF) are largely inaccessible and dauntingly complex. Herein, we primitively established dual-fluorescence expressing placenta-derived MSCs (DF-MSCs) and explored their multidimensional attributes, including cytomorphology, immunophenotying, multilineage differentiation and long-term proliferation, together with the recognition of bifluorescence intensity (BLI). Then, with the aid of in vivo living imaging, clinicopathological or inflammatory cytokine examinations and in vitro analyses, we systematically and meticulously dissected the metabolokinetics and curative effect of MSCs on mice with refractory Crohn's-like EF (EF mice), together with revealing the underlying mechanism including reactive oxygen species (ROS) and neovascularization. Strikingly, the DF-MSCs exhibited stabilized BLI and biological properties. The spatio-temporal distribution and therapeutic process of MSCs in EF mice were intuitively delineated. Meanwhile, our data indicated the curative mechanisms of DF-MSCs by simultaneously downregulating ROS and accelerating neovascularization. Collectively, we systematically illuminated the spatio-temporal biofunction and mechanism of DF-MSCs on EF mice. Our findings have supplied new references for safety and effectiveness assessments as well as the establishment of guidelines for optimal administrations of MSC-based cytotherapy in preclinical studies, which collectively indicates the prospect of P-MSC administration in clinical trials during a wide spectrum of disease remodeling including the fistulizing CD. Graphical abstract.
Collapse
Affiliation(s)
- Huixing Hou
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Leisheng Zhang
- The Postdoctoral Research Station, School of Medicine, Nankai University, Tianjin, 300071, China. .,State Key Laboratory of Experimental Hematology & National Clinical Research Center for Blood Disease, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, People's Republic of China. .,Precision Medicine Division, Health-Biotech (Tianjin) Stem Cell Research Institute Co., Ltd, Tianjin, 301700, China.
| | - Liyun Duan
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Yuanyuan Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, 300052, China
| | - Zhongchao Han
- State Key Laboratory of Experimental Hematology & National Clinical Research Center for Blood Disease, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, People's Republic of China.,Precision Medicine Division, Health-Biotech (Tianjin) Stem Cell Research Institute Co., Ltd, Tianjin, 301700, China
| | - Zongjin Li
- The Postdoctoral Research Station, School of Medicine, Nankai University, Tianjin, 300071, China.
| | - Xiaocang Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, 300052, China.
| |
Collapse
|
17
|
Bozorgmehr M, Gurung S, Darzi S, Nikoo S, Kazemnejad S, Zarnani AH, Gargett CE. Endometrial and Menstrual Blood Mesenchymal Stem/Stromal Cells: Biological Properties and Clinical Application. Front Cell Dev Biol 2020; 8:497. [PMID: 32742977 PMCID: PMC7364758 DOI: 10.3389/fcell.2020.00497] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/25/2020] [Indexed: 12/11/2022] Open
Abstract
A highly proliferative mesenchymal stem/stromal cell (MSC) population was recently discovered in the dynamic, cyclically regenerating human endometrium as clonogenic stromal cells that fulfilled the International Society for Cellular Therapy (ISCT) criteria. Specific surface markers enriching for clonogenic endometrial MSC (eMSC), CD140b and CD146 co-expression, and the single marker SUSD2, showed their perivascular identity in the endometrium, including the layer which sheds during menstruation. Indeed, cells with MSC properties have been identified in menstrual fluid and commonly termed menstrual blood stem/stromal cells (MenSC). MenSC are generally retrieved from menstrual fluid as plastic adherent cells, similar to bone marrow MSC (bmMSC). While eMSC and MenSC share several biological features with bmMSC, they also show some differences in immunophenotype, proliferation and differentiation capacities. Here we review the phenotype and functions of eMSC and MenSC, with a focus on recent studies. Similar to other MSC, eMSC and MenSC exert immunomodulatory and anti-inflammatory impacts on key cells of the innate and adaptive immune system. These include macrophages, T cells and NK cells, both in vitro and in small and large animal models. These properties suggest eMSC and MenSC as additional sources of MSC for cell therapies in regenerative medicine as well as immune-mediated disorders and inflammatory diseases. Their easy acquisition via an office-based biopsy or collected from menstrual effluent makes eMSC and MenSC attractive sources of MSC for clinical applications. In preparation for clinical translation, a serum-free culture protocol was established for eMSC which includes a small molecule TGFβ receptor inhibitor that prevents spontaneous differentiation, apoptosis, senescence, maintains the clonogenic SUSD2+ population and enhances their potency, suggesting potential for cell-therapies and regenerative medicine. However, standardization of MenSC isolation protocols and culture conditions are major issues requiring further research to maximize their potential for clinical application. Future research will also address crucial safety aspects of eMSC and MenSC to ensure these protocols produce cell products free from tumorigenicity and toxicity. Although a wealth of data on the biological properties of eMSC and MenSC has recently been published, it will be important to address their mechanism of action in preclinical models of human disease.
Collapse
Affiliation(s)
- Mahmood Bozorgmehr
- Reproductive Immunology Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Shanti Gurung
- Centre for Reproductive Health, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Saeedeh Darzi
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Shohreh Nikoo
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Somaieh Kazemnejad
- Nanobitechnology Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Amir-Hassan Zarnani
- Reproductive Immunology Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Caroline E. Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
18
|
Cimino M, Parreira P, Bidarra SJ, Gonçalves RM, Barrias CC, Martins MCL. Effect of surface chemistry on hMSC growth under xeno-free conditions. Colloids Surf B Biointerfaces 2020; 189:110836. [DOI: 10.1016/j.colsurfb.2020.110836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 01/17/2020] [Accepted: 01/29/2020] [Indexed: 01/05/2023]
|
19
|
Eljarrah A, Gergues M, Pobiarzyn PW, Sandiford OA, Rameshwar P. Therapeutic Potential of Mesenchymal Stem Cells in Immune-Mediated Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1201:93-108. [PMID: 31898783 DOI: 10.1007/978-3-030-31206-0_5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that can self-renew and differentiate into cells of all germ layers. MSCs can be easily attracted to the site of tissue insult with high levels of inflammatory mediators. The general ability of MSCs to migrate at the sites of tissue injury suggested an innate ability for these cells to be involved in baseline tissue repair. The bone marrow is one of the primary sources of MSCs, though they can be ubiquitous. An attractive property of MSCs for clinical application is their ability to cross allogeneic barrier. However, alone, MSCs are not immune suppressive cells. Rather, they can be licensed by the tissue microenvironment to become immune suppressor cells. Immune suppressor functions of MSCs include those that blunt cytotoxicity of natural killer cells, suppression of T-cell proliferation, and "veto" function. MSCs, as third-party cells, suppress the immune response that generally recapitulates graft-versus-host disease (GvHD) responses. Based on the plastic functions of MSCs, these cells have dominated the field of cell-based therapies, such as anti-inflammatory and drug delivery. Here, we focus on the potential use of MSC for immunological disorders such as Crohn's disease and GvHD.
Collapse
Affiliation(s)
- Adam Eljarrah
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
- Rutgers School of Graduate School at New Jersey Medical School, Newark, NJ, USA
| | - Marina Gergues
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
- Rutgers School of Graduate School at New Jersey Medical School, Newark, NJ, USA
| | - Piotr W Pobiarzyn
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
- Rutgers School of Graduate School at New Jersey Medical School, Newark, NJ, USA
| | - Oleta A Sandiford
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
- Rutgers School of Graduate School at New Jersey Medical School, Newark, NJ, USA
| | - Pranela Rameshwar
- Department of Medicine - Division of Hematology/Oncology, New Jersey Medical School, Rutgers School of Biomedical Health Science, Newark, NJ, USA.
| |
Collapse
|
20
|
Ahn SY, Maeng YS, Kim YR, Choe YH, Hwang HS, Hyun YM. In vivo monitoring of dynamic interaction between neutrophil and human umbilical cord blood-derived mesenchymal stem cell in mouse liver during sepsis. Stem Cell Res Ther 2020; 11:44. [PMID: 32014040 PMCID: PMC6998265 DOI: 10.1186/s13287-020-1559-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/30/2019] [Accepted: 01/09/2020] [Indexed: 12/24/2022] Open
Abstract
Background Sepsis is a global inflammatory disease that causes death. It has been reported that mesenchymal stem cell (MSC) treatment can attenuate inflammatory and septic symptoms. In this study, we investigated how interactions between neutrophils and human umbilical cord blood (hUCB)-MSCs in the liver of septic mice are involved in mitigating sepsis that is mediated by MSCs. Accordingly, we aimed to determine whether hUCB-MSC application could be an appropriate treatment for sepsis. Methods To induce septic condition, lipopolysaccharide (LPS) was intraperitoneally (i.p.) injected into mice 24 h after the intravenous (i.v.) injection of saline or hUCB-MSCs. To determine the effect of hUCB-MSCs on the immune response during sepsis, histologic analysis, immunoassays, and two-photon intravital imaging were performed 6 h post-LPS injection. For the survival study, mice were monitored for 6 days after LPS injection. Results The injection (i.v.) of hUCB-MSCs alleviated the severity of LPS-induced sepsis by increasing IL-10 levels (p < 0.001) and decreasing mortality (p < 0.05) in septic mice. In addition, this significantly reduced the recruitment of neutrophils (p < 0.001) to the liver. In hUCB-MSC-treated condition, we also observed several distinct patterns of dynamic interactions between neutrophils and hUCB-MSCs in the inflamed mouse liver, as well as vigorous interactions between hepatic stellate cells (HSCs or ito cells) and hUCB-MSCs. Interestingly, hUCB-MSCs that originated from humans were not recognized as foreign in the mouse body and consequently did not cause graft rejection. Conclusions These distinct interaction patterns between innate immune cells and hUCB-MSCs demonstrated that hUCB-MSCs have beneficial effects against LPS-induced sepsis through associations with neutrophils. In addition, the immunomodulatory properties of hUCB-MSCs might enable immune evasion in the host. Taken together, our results suggest the prospects of hUCB-MSCs as a therapeutic tool to inhibit inflammation and alleviate pathological immune responses such as sepsis.
Collapse
Affiliation(s)
- Sung Yong Ahn
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong-Sun Maeng
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul, Republic of Korea.,Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yu Rim Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea.,BK21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Ho Choe
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea.,BK21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Han Sung Hwang
- Department of Obstetrics and Gynecology, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Republic of Korea.
| | - Young-Min Hyun
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea. .,BK21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
21
|
Dragoo JL, Guzman RA. Evaluation of the Consistency and Composition of Commercially Available Bone Marrow Aspirate Concentrate Systems. Orthop J Sports Med 2020; 8:2325967119893634. [PMID: 32010732 PMCID: PMC6970477 DOI: 10.1177/2325967119893634] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 09/10/2019] [Indexed: 12/13/2022] Open
Abstract
Background: Bone marrow aspirate (BMA) concentrate (BMAC) has gained popularity as a treatment modality for various orthopaedic conditions; however, there are still inconsistencies in its reported therapeutic efficacy. This may be because of the many different commercial BMAC preparation systems used clinically, which generate dissimilar concentrate products. Purpose: To compare 3 commercially available BMAC preparation systems: Harvest SmartPrep 3, Biomet BioCUE, and Arthrex Angel. We evaluated the consistency of each of these systems and compared the composition of their concentrate products. Study Design: Controlled laboratory study. Methods: A total of 10 patients donated whole blood and BMA, which were combined and processed with the 3 different BMAC preparation systems. Samples were taken before and after processing for the measurement of white blood cells (WBC), platelets (PLT), CD34+ cells, and colony-forming unit–fibroblast (CFU-F). To evaluate consistency, the variances of cell yield and concentration increase from baseline for each cell type were compared between systems. To compare concentrate product composition, differences between the systems’ mean cell yield and concentration increase from baseline for each cell type were evaluated. Results: The Harvest system (variance, 0.25) concentrated WBC more consistently than the Arthrex system (variance, 3.25) (P = .024), but no other differences in consistency were noted between the 3 systems. The Harvest system recovered the greatest percentage of CFU-F (82.4% ± 18.2%), CD34+ cells (81.1% ± 28.5%), and WBC (77.3% ± 8.6%), whereas the Biomet system recovered the greatest percentage of PLTs (92.9% ± 27.3%). The Arthrex system concentrated PLT to the greatest degree (11.10 ± 2.05 times baseline), while the Biomet system concentrated WBC to the greatest degree (5.99 ± 1.04 times baseline). Conclusion: The consistency of the 3 systems was similar for all but 1 of the evaluated cell types. However, the composition of the concentrate products differed across systems. This may grant each system unique advantages without having to sacrifice reproducibility. Clinical Relevance: Understanding the consistency of different BMAC preparation systems and their product makeup may aid in determining optimal therapeutic doses of different cell types.
Collapse
Affiliation(s)
- Jason L Dragoo
- Department of Orthopaedic Surgery, Stanford University Medical Center, Stanford, California, USA
| | - Roberto A Guzman
- Department of Orthopaedic Surgery, Stanford University Medical Center, Stanford, California, USA
| |
Collapse
|
22
|
Kalinec GM, Cohn W, Whitelegge JP, Faull KF, Kalinec F. Preliminary Characterization of Extracellular Vesicles From Auditory HEI-OC1 Cells. Ann Otol Rhinol Laryngol 2019; 128:52S-60S. [DOI: 10.1177/0003489419836226] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Objectives: Isolate, purify, and characterize extracellular vesicles (EVs) obtained from auditory HEI-OC1 cells, and evaluate their suitability for intracochlear transport and delivery of pharmacological drugs and/or pro-resolution mediators of acute inflammatory processes. Methods: HEI-OC1 EVs were isolated and purified using the exoEasy Maxi Kit, and their size was evaluated by nanoparticle tracking techniques. Bottom-up proteomics of the EVs, either freshly obtained or stored for up to 4 months at −20°C, was performed by LC-ESI-MS/MS. LC-ESI-MS/MS-MRM was used to measure the loading of dexamethasone inside EVs following co-incubation at room temperature for 1 hour with and without 5 minutes sonication. Results: Routinely, we were able to obtain purified fractions of >2 × 109 EVs/mL, with diameters varying between 50 and 800 nm. Bottom-up proteomics showed that among the most abundant EVs proteins, 19.2% were cytoplasmic, 17.2% were membrane localized, 12.3% were cytosolic, and 14.6% were nucleolar. No significant differences between fresh and stored EVs were detected. Importantly, co-incubation of HEI-OC1 EVs (1 × 108 EVs/mL) with dexamethasone (10 mM) resulted in the incorporation of 10.1 ± 1.9 nM dexamethasone per milliliter of EVs suspension. Conclusions: Altogether, the results suggest that EVs from HEI-OC1 cells could be advantageously used as biological nanocarriers for the delivery of specific molecules and pharmacological drugs into the inner ear.
Collapse
Affiliation(s)
- Gilda M. Kalinec
- Department of Head and Neck Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Whitaker Cohn
- Pasarow Mass Spectrometry Laboratory, Jane and Terry Semel Institute for Neuroscience and Human Behavior and Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Julian P. Whitelegge
- Pasarow Mass Spectrometry Laboratory, Jane and Terry Semel Institute for Neuroscience and Human Behavior and Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Kym F. Faull
- Pasarow Mass Spectrometry Laboratory, Jane and Terry Semel Institute for Neuroscience and Human Behavior and Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Federico Kalinec
- Department of Head and Neck Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
23
|
Nourian Dehkordi A, Mirahmadi Babaheydari F, Chehelgerdi M, Raeisi Dehkordi S. Skin tissue engineering: wound healing based on stem-cell-based therapeutic strategies. Stem Cell Res Ther 2019; 10:111. [PMID: 30922387 PMCID: PMC6440165 DOI: 10.1186/s13287-019-1212-2] [Citation(s) in RCA: 310] [Impact Index Per Article: 51.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Normal wound healing is a dynamic and complex multiple phase process involving coordinated interactions between growth factors, cytokines, chemokines, and various cells. Any failure in these phases may lead wounds to become chronic and have abnormal scar formation. Chronic wounds affect patients' quality of life, since they require repetitive treatments and incur considerable medical costs. Thus, much effort has been focused on developing novel therapeutic approaches for wound treatment. Stem-cell-based therapeutic strategies have been proposed to treat these wounds. They have shown considerable potential for improving the rate and quality of wound healing and regenerating the skin. However, there are many challenges for using stem cells in skin regeneration. In this review, we present some sets of the data published on using embryonic stem cells, induced pluripotent stem cells, and adult stem cells in healing wounds. Additionally, we will discuss the different angles whereby these cells can contribute to their unique features and show the current drawbacks.
Collapse
Affiliation(s)
- Azar Nourian Dehkordi
- Department of Stem Cell and Regenerative Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Fatemeh Mirahmadi Babaheydari
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad Chehelgerdi
- Biotechnology Research Center, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | | |
Collapse
|
24
|
Gunawardena TNA, Rahman MT, Abdullah BJJ, Abu Kasim NH. Conditioned media derived from mesenchymal stem cell cultures: The next generation for regenerative medicine. J Tissue Eng Regen Med 2019; 13:569-586. [PMID: 30644175 DOI: 10.1002/term.2806] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 10/26/2018] [Accepted: 12/17/2018] [Indexed: 12/13/2022]
Abstract
Recent studies suggest that the main driving force behind the therapeutic activity observed in mesenchymal stem cells (MSCs) are the paracrine factors secreted by these cells. These biomolecules also trigger antiapoptotic events to prevent further degeneration of the diseased organ through paracrine signalling mechanisms. In comparison with the normal physiological conditions, an increased paracrine gradient is observed within the peripheral system of diseased organs that enhances the migration of tissue-specific MSCs towards the site of infection or injury to promote healing. Thus, upon administration of conditioned media derived from mesenchymal stem cell cultures (MSC-CM) could contribute in maintaining the increased paracrine factor gradient between the diseased organ and the stem cell niche in order to speed up the process of recovery. Based on the principle of the paracrine signalling mechanism, MSC-CM, also referred as the secretome of the MSCs, is a rich source of the paracrine factors and are being studied extensively for a wide range of regenerative therapies such as myocardial infarction, stroke, bone regeneration, hair growth, and wound healing. This article highlights the current technological applications and advances of MSC-CM with the aim to appraise its future potential as a regenerative therapeutic agent.
Collapse
Affiliation(s)
| | - Mohammad Tariqur Rahman
- Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | | | - Noor Hayaty Abu Kasim
- Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia.,Regenerative Dentistry Research Group, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
25
|
Jo H, Eom YW, Kim HS, Park HJ, Kim HM, Cho MY. Regulatory Dendritic Cells Induced by Mesenchymal Stem Cells Ameliorate Dextran Sodium Sulfate-Induced Chronic Colitis in Mice. Gut Liver 2019; 12:664-673. [PMID: 29938461 PMCID: PMC6254613 DOI: 10.5009/gnl18072] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/11/2018] [Accepted: 04/23/2018] [Indexed: 12/11/2022] Open
Abstract
Background/Aims Regulatory dendritic cells (rDCs), which can be induced by mesenchymal stem cells (MSCs), play an important role in inducing and maintaining homeostasis of regulatory T cells and exhibit anti-inflammatory functions. In this study, we investigated whether MSCs could differentiate DCs into rDCs and compared the therapeutic effects of rDCs and MSCs on dextran sodium sulfate (DSS)-induced chronic colitis mice. Methods Immature DCs (imDCs) and lipopolysaccharide (LPS)-treated mature DCs (mDCs) were co-cultured with MSCs for 48 hours, and then the profiles of surface markers and cytokines and regulatory roles of these DCs for primary splenocytes were analyzed. In addition, the therapeutic effects of MSCs and DCs co-cultured with MSCs were compared in chronic colitis mice. Results After co-culture of imDCs (MSC-DCs) or LPS-treated mDCs (LPS+MSC-DCs) with MSCs, the expression of CD11c, CD80, CD86, interleukin 6 (IL-6), tumor necrosis factor-α (TNF-α), and interferon-γ (IFN-γ), was decreased, but that of CD11b, IL-10, and transforming growth factor-β (TGF-β) was increased. Furthermore, MSC-DCs and LPS+MSC-DCs induced the expression of CD4, CD25, and Foxp3 in primary splenocytes isolated from mice. In DSS-induced colitis mice, MSCs and MSC-DCs increased colon length, body weight, and survival rate and induced histological improvement. Moreover, in the colon tissues, the expression of IL-6, TNF-α, and IFN-γ decreased, but that of IL-10, TGF-β, and Foxp3 increased in the MSC- and MSC-DC-injected groups. Conclusions Our data suggest that MSCs differentiate DCs into rDCs, which ameliorate chronic colitis. Thus, rDCs stimulated by MSCs may be therapeutically useful for the treatment of chronic inflammatory diseases.
Collapse
Affiliation(s)
- Hannah Jo
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Young Woo Eom
- Cell Therapy and Tissue Engineering Center, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Hyun-Soo Kim
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Hong Jun Park
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Hee Man Kim
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Mee-Yon Cho
- Department of Pathology, Yonsei University Wonju College of Medicine, Wonju, Korea
| |
Collapse
|
26
|
Salmenkari H, Laitinen A, Forsgård RA, Holappa M, Lindén J, Pasanen L, Korhonen M, Korpela R, Nystedt J. The use of unlicensed bone marrow-derived platelet lysate-expanded mesenchymal stromal cells in colitis: a pre-clinical study. Cytotherapy 2019; 21:175-188. [PMID: 30611671 DOI: 10.1016/j.jcyt.2018.11.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/04/2018] [Accepted: 11/13/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) are a promising candidate for treatment of inflammatory disorders, but their efficacy in human inflammatory bowel diseases (IBDs) has been inconsistent. Comparing the results from various pre-clinical and clinical IBD studies is also challenging due to a large variation in study designs. METHODS In this comparative pre-clinical study, we compared two administration routes and investigated the safety and feasibility of both fresh and cryopreserved platelet-lysate-expanded human bone marrow-derived MSCs without additional licensing in a dextran sodium sulfate (DSS) colitis mouse model both in the acute and regenerative phases of colitis. Body weight, macroscopic score for inflammation and colonic interleukin (IL)-1β and tumor necrosis factor (TNF)α concentrations were determined in both phases of colitis. Additionally, histopathology was assessed and Il-1β and Agtr1a messenger RNA (mRNA) levels and angiotensin-converting enzyme (ACE) protein levels were measured in the colon in the regenerative phase of colitis. RESULTS Intravenously administered MSCs exhibited modest anti-inflammatory capacity in the acute phase of colitis by reducing IL-1β protein levels in the inflamed colon. There were no clear improvements in mice treated with fresh or cryopreserved unlicensed MSCs according to weight monitoring results, histopathology and macroscopic score results. Pro-inflammatory ACE protein expression and shedding were reduced by cryopreserved MSCs in the colon. CONCLUSIONS In conclusion, we observed a good safety profile for bone marrow-derived platelet lysate-expanded MSCs in a mouse pre-clinical colitis model, but the therapeutic effect of MSCs prepared without additional licensing (i.e. such as MSCs are administered in graft-versus-host disease) was modest in the chosen in vivo model system and limited to biochemical improvements in cytokines without a clear benefit in histopathology or body weight development.
Collapse
Affiliation(s)
- Hanne Salmenkari
- Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Anita Laitinen
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Richard A Forsgård
- Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mervi Holappa
- Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jere Lindén
- Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Lauri Pasanen
- Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Matti Korhonen
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Riitta Korpela
- Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Johanna Nystedt
- Advanced Cell Therapy Centre, Finnish Red Cross Blood Service, Helsinki, Finland.
| |
Collapse
|
27
|
Fitzsimmons REB, Mazurek MS, Soos A, Simmons CA. Mesenchymal Stromal/Stem Cells in Regenerative Medicine and Tissue Engineering. Stem Cells Int 2018; 2018:8031718. [PMID: 30210552 PMCID: PMC6120267 DOI: 10.1155/2018/8031718] [Citation(s) in RCA: 231] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 05/31/2018] [Accepted: 07/17/2018] [Indexed: 02/08/2023] Open
Abstract
As a result of over five decades of investigation, mesenchymal stromal/stem cells (MSCs) have emerged as a versatile and frequently utilized cell source in the fields of regenerative medicine and tissue engineering. In this review, we summarize the history of MSC research from the initial discovery of their multipotency to the more recent recognition of their perivascular identity in vivo and their extraordinary capacity for immunomodulation and angiogenic signaling. As well, we discuss long-standing questions regarding their developmental origins and their capacity for differentiation toward a range of cell lineages. We also highlight important considerations and potential risks involved with their isolation, ex vivo expansion, and clinical use. Overall, this review aims to serve as an overview of the breadth of research that has demonstrated the utility of MSCs in a wide range of clinical contexts and continues to unravel the mechanisms by which these cells exert their therapeutic effects.
Collapse
Affiliation(s)
- Ross E. B. Fitzsimmons
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, Canada M5S 3G9
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Ave, Toronto, ON, Canada M5G 1M1
| | - Matthew S. Mazurek
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, Calgary, AB, Canada T2N 4Z6
| | - Agnes Soos
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, Canada M5S 3G9
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Ave, Toronto, ON, Canada M5G 1M1
| | - Craig A. Simmons
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON, Canada M5S 3G9
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Ave, Toronto, ON, Canada M5G 1M1
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, ON, Canada M5S 3G8
| |
Collapse
|
28
|
Pelagalli A, Nardelli A, Lucarelli E, Zannetti A, Brunetti A. Autocrine signals increase ovine mesenchymal stem cells migration through Aquaporin-1 and CXCR4 overexpression. J Cell Physiol 2018; 233:6241-6249. [PMID: 29345324 DOI: 10.1002/jcp.26493] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 01/17/2018] [Indexed: 12/13/2022]
Abstract
Sheep is a relevant large animal model that is frequently used to test innovative tissue engineering (TE) approaches especially for bone reconstruction. Mesenchymal stem cells (MSCs) are used in TE applications because they represent key component of adult tissue repair. Importantly, MSCs from different species show similar characteristics, which facilitated their application in translational studies using animal models. Nowadays, many researches are focusing on the use of ovine mesenchymal stem cells (oMSCs) in orthopedic preclinical settings for regenerative medicine purposes. Therefore, there is a need to amplify our knowledge on the mechanisms underlying the behaviour of these cells. Recently, several studies have shown that MSC function is largely dependent on factors that MSCs release in the environment, as well as, in conditioned medium (CM). It has been demonstrated that MSCs through autocrine and paracrine signals are able to stimulate proliferation, migration, and differentiation of different type of cells including themselves. In this study, we investigated the effects of the CM produced by oMSCs on oMSCs themselves and we explored the signal pathways involved. We observed that CM caused an enhancement of oMSC migration. Furthermore, we found that CM increased levels of two membrane proteins involved in cell migration, Aquaporin 1 (AQP1), and C-X-C chemokine receptor type 4 (CXCR4), and activated Akt and Erk intracellular signal pathways. In conclusion, taken together our results suggest the high potential of autologous CM as a promising tool to modulate behaviour of MSCs thus improving their use in therapeutically approaches.
Collapse
Affiliation(s)
- Alessandra Pelagalli
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
- Institute of Biostructure and Bioimaging, National Research Council, Naples, Italy
| | - Anna Nardelli
- Institute of Biostructure and Bioimaging, National Research Council, Naples, Italy
| | - Enrico Lucarelli
- Osteoarticolar Regeneration Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Antonella Zannetti
- Institute of Biostructure and Bioimaging, National Research Council, Naples, Italy
| | - Arturo Brunetti
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
29
|
El‐Nakeep S, Abdel Latif O, Shawky A, Nabhan AF. Stem cell transplantation for induction of remission in medically refractory Crohn’s disease. Cochrane Database Syst Rev 2018; 2018:CD013070. [PMCID: PMC6513462 DOI: 10.1002/14651858.cd013070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
This is a protocol for a Cochrane Review (Intervention). The objectives are as follows: The objective of this review is to assess the efficacy and safety of stem cell transplantation for induction of remission in active Crohn's disease.
Collapse
Affiliation(s)
- Sarah El‐Nakeep
- Faculty of Medicine, Ain Shams UniversityGastroenterology and Hepatology Unit, Department of Internal MedicineCairoEgypt
| | - Osama Abdel Latif
- Faculty of Medicine, Ain Shams UniversityAllergy and Clinical Immunology Unit, Department of Internal MedicineCairoEgypt
| | - Ahmed Shawky
- Faculty of Medicine, Ain Shams UniversityGastroenterology and Hepatology Unit, Department of Internal MedicineCairoEgypt
| | - Ashraf F Nabhan
- Ain Shams UniversityDepartment of Obstetrics and Gynaecology, Faculty of Medicine16 Ali Fahmi Kamel StreetHeliopolisCairoEgypt11351
| |
Collapse
|
30
|
Mesenchymal Stem Cell-Based Immunomodulation: Properties and Clinical Application. Stem Cells Int 2018; 2018:3057624. [PMID: 30013600 PMCID: PMC6022321 DOI: 10.1155/2018/3057624] [Citation(s) in RCA: 338] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 05/29/2018] [Indexed: 02/05/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent stem cells characterized by self-renewal, production of clonal cell populations, and multilineage differentiation. They exist in nearly all tissues and play a significant role in tissue repair and regeneration. Additionally, MSCs possess wide immunoregulatory properties via interaction with immune cells in both innate and adaptive immune systems, leading to immunosuppression of various effector functions. Numerous bioactive molecules secreted by MSCs, particularly cytokines, growth factors, and chemokines, exert autocrine/paracrine effects that modulate the physiological processes of MSCs. These invaluable virtues of MSCs provide new insight into potential treatments for tissue damage and inflammation. In particular, their extensive immunosuppressive properties are being explored for promising therapeutic application in immune disorders. Recently, clinical trials for MSC-mediated therapies have rapidly developed for immune-related diseases following reports from preclinical studies declaring their therapeutic safety and efficacy. Though immunotherapy of MSCs remains controversial, these clinical trials pave the way for their widespread therapeutic application in immune-based diseases. In this review, we will summarize and update the latest research findings and clinical trials on MSC-based immunomodulation.
Collapse
|
31
|
Nguyen DC, Lewis HC, Joyner C, Warren V, Xiao H, Kissick HT, Wu R, Galipeau J, Lee FEH. Extracellular vesicles from bone marrow-derived mesenchymal stromal cells support ex vivo survival of human antibody secreting cells. J Extracell Vesicles 2018; 7:1463778. [PMID: 29713426 PMCID: PMC5917896 DOI: 10.1080/20013078.2018.1463778] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 04/04/2018] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) from bone marrow (BM)-derived mesenchymal stromal cells (BM-MSC) are novel mechanisms of cell-cell communication over short and long distances. BM-MSC have been shown to support human antibody secreting cells (ASC) survival ex vivo, but whether the crosstalk between the MSC-ASC interaction can occur via EVs is not known. Thus, we evaluated the role of EVs in ASC survival and IgG secretion. EVs were isolated from irradiated and non-irradiated primary BM-MSC and were quantified. They were further characterized by electron microscopy (EM) and CD63 and CD81 immuno-gold EM staining. Human ASC were isolated via fluorescence-activated cell sorting (FACS) and cultured ex vivo with the EV fractions, the EV-reduced fractions, or conventional media. IgG Elispots were used to measure the survival and functionality of the ASC. Contents of the EV fractions were evaluated by proteomics. We saw that both irradiated and non-irradiated MSC secretome preparations afforded vesicles of a size consistent with EVs. Both preparations appeared comparable in EM morphology and CD63 and CD81 immuno-gold EM. Both irradiated and non-irradiated EV fractions supported ASC function, at 88% and 90%, respectively, by day 3. In contrast, conventional media maintained only 4% ASC survival by day 3. To identify the specific factors that provided in vitro ASC support, we compared proteomes of the irradiated and non-irradiated EV fractions with conventional media. Pathway analysis of these proteins identified factors involved in the vesicle-mediated delivery of integrin signalling proteins. These findings indicate that BM-MSC EVs provide an effective support system for ASC survival and IgG secretion.
Collapse
Affiliation(s)
- Doan C. Nguyen
- Division of Pulmonary Allergy, Critical Care, & Sleep Medicine, Emory University, Atlanta, GA, USA
| | - Holly C. Lewis
- Departments of Pediatrics and Hematology & Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Chester Joyner
- International Center for Malaria Research, Education and Development, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Vivien Warren
- Division of Pulmonary Allergy, Critical Care, & Sleep Medicine, Emory University, Atlanta, GA, USA
| | - Haopeng Xiao
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
| | - Haydn T. Kissick
- Emory Vaccine Center and Department of Urology, Emory University, Atlanta, GA, USA
| | - Ronghu Wu
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA
| | - Jacques Galipeau
- Department of Medicine and University of Wisconsin Carbone Cancer Center, University of Wisconsin in Madison, Madison, WI, USA
| | - F. Eun-Hyung Lee
- Division of Pulmonary Allergy, Critical Care, & Sleep Medicine, Emory University, Atlanta, GA, USA
| |
Collapse
|
32
|
Mesenchymal stromal cells (MSCs) and colorectal cancer: a troublesome twosome for the anti-tumour immune response? Oncotarget 2018; 7:60752-60774. [PMID: 27542276 PMCID: PMC5312417 DOI: 10.18632/oncotarget.11354] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 07/09/2016] [Indexed: 12/18/2022] Open
Abstract
The tumour microenvironment (TME) is an important factor in determining the growth and metastasis of colorectal cancer, and can aid tumours by both establishing an immunosuppressive milieu, allowing the tumour avoid immune clearance, and by hampering the efficacy of various therapeutic regimens. The tumour microenvironment is composed of many cell types including tumour, stromal, endothelial and immune cell populations. It is widely accepted that cells present in the TME acquire distinct functional phenotypes that promote tumorigenesis. One such cell type is the mesenchymal stromal cell (MSC). Evidence suggests that MSCs exert effects in the colorectal tumour microenvironment including the promotion of angiogenesis, invasion and metastasis. MSCs immunomodulatory capacity may represent another largely unexplored central feature of MSCs tumour promoting capacity. There is considerable evidence to suggest that MSCs and their secreted factors can influence the innate and adaptive immune responses. MSC-immune cell interactions can skew the proliferation and functional activity of T-cells, dendritic cells, natural killer cells and macrophages, which could favour tumour growth and enable tumours to evade immune cell clearance. A better understanding of the interactions between the malignant cancer cell and stromal components of the TME is key to the development of more specific and efficacious therapies for colorectal cancer. Here, we review and explore MSC- mediated mechanisms of suppressing anti-tumour immune responses in the colon tumour microenvironment. Elucidation of the precise mechanism of immunomodulation exerted by tumour-educated MSCs is critical to inhibiting immunosuppression and immune evasion established by the TME, thus providing an opportunity for targeted and efficacious immunotherapy for colorectal cancer growth and metastasis.
Collapse
|
33
|
García-Manrique P, Matos M, Gutiérrez G, Pazos C, Blanco-López MC. Therapeutic biomaterials based on extracellular vesicles: classification of bio-engineering and mimetic preparation routes. J Extracell Vesicles 2018; 7:1422676. [PMID: 29372017 PMCID: PMC5774402 DOI: 10.1080/20013078.2017.1422676] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 12/22/2017] [Indexed: 12/19/2022] Open
Abstract
Extracellular vesicles (EVs) are emerging as novel theranostic tools. Limitations related to clinical uses are leading to a new research area on design and manufacture of artificial EVs. Several strategies have been reported in order to produce artificial EVs, but there has not yet been a clear criterion by which to differentiate these novel biomaterials. In this paper, we suggest for the first time a systematic classification of the terms used to build up the artificial EV landscape, based on the preparation method. This could be useful to guide the derivation to clinical trial routes and to clarify the literature. According to our classification, we have reviewed the main strategies reported to date for their preparation, including key points such as: cargo loading, surface targeting strategies, purification steps, generation of membrane fragments for the construction of biomimetic materials, preparation of synthetic membranes inspired in EV composition and subsequent surface decoration.
Collapse
Affiliation(s)
- Pablo García-Manrique
- Department of Physical and Analytical Chemistry, University of Oviedo, Oviedo, Spain
- Department of Chemical and Environmental Engineering, University of Oviedo, Oviedo, Spain
| | - María Matos
- Department of Chemical and Environmental Engineering, University of Oviedo, Oviedo, Spain
| | - Gemma Gutiérrez
- Department of Chemical and Environmental Engineering, University of Oviedo, Oviedo, Spain
| | - Carmen Pazos
- Department of Chemical and Environmental Engineering, University of Oviedo, Oviedo, Spain
| | | |
Collapse
|
34
|
Cimino M, Gonçalves RM, Bauman E, Barroso-Vilares M, Logarinho E, Barrias CC, Martins MCL. Optimization of the use of a pharmaceutical grade xeno-free medium for in vitro expansion of human mesenchymal stem/stromal cells. J Tissue Eng Regen Med 2017; 12:e1785-e1795. [PMID: 29024519 DOI: 10.1002/term.2588] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 09/23/2017] [Accepted: 10/03/2017] [Indexed: 01/04/2023]
Abstract
Human bone marrow-derived mesenchymal stem/stromal cells (hMSCs) are considered promising therapeutic agents in the field of cell therapy and regenerative medicine, mainly due to their relative facility to be isolated, multi-differentiation potential, and immunomodulatory role. However, their application in clinics requires a crucial step of in vitro expansion. Most of the protocols for hMSCs in vitro culture use foetal bovine serum as medium supplement that, being from animal origin, presents several safety concerns and may initiate xenogeneic immune responses after cells transplantation. This work reports the optimization of a pharmaceutical-grade xeno-free strategy for hMSCs in vitro expansion based on the supplementation of basal medium with a pharmaceutical-grade human plasma-derived supplement for cell culture (SCC) and 2 human growth factors (bFGF and TGFβ1), plus a coating of human plasma fibronectin (Fn). After 4 weeks in culture, this strategy improves hMSCs expansion yield about 4.3-fold in comparison with foetal bovine serum supplementation and 4.5-fold compared with a commercially available xeno-free medium. hMSCs expanded in SCC-based formulation maintained their phenotype and differentiation capacity into osteogenic, adipogenic, and chondrogenic lineages, without alterations in cell karyotype. Overall, the SCC-based medium appears to be an excellent alternative for the xeno-free expansion of hMSCs as therapeutic agents for clinical applications.
Collapse
Affiliation(s)
- M Cimino
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto (UP), Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, University of Porto (UP), Porto, Portugal
| | - R M Gonçalves
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto (UP), Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, University of Porto (UP), Porto, Portugal.,ICBAS-Instituto de Ciências Biomédicas Abel Salazar, University of Porto (UP), Porto, Portugal
| | - E Bauman
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto (UP), Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, University of Porto (UP), Porto, Portugal
| | - M Barroso-Vilares
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto (UP), Porto, Portugal.,IBMC-Instituto de Biologia Molecular e Celular, University of Porto (UP), Porto, Portugal
| | - E Logarinho
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto (UP), Porto, Portugal.,IBMC-Instituto de Biologia Molecular e Celular, University of Porto (UP), Porto, Portugal
| | - C C Barrias
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto (UP), Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, University of Porto (UP), Porto, Portugal.,ICBAS-Instituto de Ciências Biomédicas Abel Salazar, University of Porto (UP), Porto, Portugal
| | - M C L Martins
- i3S, Instituto de Investigação e Inovação em Saúde, University of Porto (UP), Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, University of Porto (UP), Porto, Portugal.,ICBAS-Instituto de Ciências Biomédicas Abel Salazar, University of Porto (UP), Porto, Portugal
| |
Collapse
|
35
|
Wawryk-Gawda E, Wojcik B. Application of mesenchymal stem cells in paediatrics. CURRENT ISSUES IN PHARMACY AND MEDICAL SCIENCES 2017. [DOI: 10.1515/cipms-2017-0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
Mesenchymal stem cells (MSC) were described by Friedenstein in the 1970s as being a group of bone marrow non-hematopoietic cells that are the source of fibroblasts. Since then, knowledge about the therapeutic potential of MSCs has significantly increased. MSCs are currently used for the treatment of many diseases, both in adults and children. MSCs are used successfully in the case of autoimmune diseases, including rheumatic diseases, diabetes mellitus type 1, gastroenterological and neurological diseases. Moreover, treatment of such organ disorders as damage or hypoxia through application of MSC therapy has shown to be satisfactory. In addition, there are some types of congenital disorders, including osteogenesis imperfecta and Spinal Muscular Atrophy, that may be treated with cellular therapy. Most studies showed no other adverse effects than fever. Our study is an analysis that particularly focuses on the registered trials and results of MSCs application to under 18 patients with acute, chronic, recurrent, resistance and corticosteroids types of Graft-versus-Host Disease (GvHD). Stem cells currently play an important role in the treatment of many diseases. Long-term studies conducted on animals have shown that cell therapy is both effective and safe. The number of indications for use of these cells in the course of treatment of people is constantly increasing. The results of subsequent studies provide important data justifying the application of MSCs in the course of treatment of many diseases whose treatment is ineffective when utilizing other approaches.
Collapse
Affiliation(s)
- Ewelina Wawryk-Gawda
- Chair and Department Histology and Embryology with Experimental Cytology Unit, Medical University of Lublin, Radziwillowska 11, 20-080 Lublin , Poland
| | - Beata Wojcik
- Clinic and Department of Pediatric Hematology and Oncology and Transplantology University Children’s Hospital in Lublin, Prof. Antoniego Gebali 6, Lublin , Poland
| |
Collapse
|
36
|
Nikiforou M, Willburger C, de Jong AE, Kloosterboer N, Jellema RK, Ophelders DRMG, Steinbusch HWM, Kramer BW, Wolfs TGAM. Global hypoxia-ischemia induced inflammation and structural changes in the preterm ovine gut which were not ameliorated by mesenchymal stem cell treatment. Mol Med 2016; 22:244-257. [PMID: 27257938 PMCID: PMC5023518 DOI: 10.2119/molmed.2015.00252] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 04/11/2016] [Indexed: 12/13/2022] Open
Abstract
Perinatal asphyxia, a condition of impaired gas exchange during birth, leads to fetal hypoxia-ischemia (HI) and is associated with postnatal adverse outcomes including intestinal dysmotility and necrotizing enterocolitis (NEC). Evidence from adult animal models of transient, locally-induced intestinal HI has shown that inflammation is essential in HI-induced injury of the gut. Importantly, mesenchymal stem cell (MSC) treatment prevented this HI-induced intestinal damage. We therefore assessed whether fetal global HI induced inflammation, injury and developmental changes in the gut and whether intravenous MSC administration ameliorated these HI-induced adverse intestinal effects. In a preclinical ovine model, fetuses were subjected to umbilical cord occlusion (UCO), with or without MSC treatment, and sacrificed 7 days after UCO. Global HI increased the number of myeloperoxidase positive cells in the mucosa, upregulated mRNA levels of interleukin (IL)-1β and IL-17 in gut tissue and caused T-cell invasion in the intestinal muscle layer. Intestinal inflammation following global HI was associated with increased Ki67+ cells in the muscularis and subsequent muscle hyperplasia. Global HI caused distortion of glial fibrillary acidic protein immunoreactivity in the enteric glial cells and increased synaptophysin and serotonin expression in the myenteric ganglia. Intravenous MSC treatment did not ameliorate these HI-induced adverse intestinal events. Global HI resulted in intestinal inflammation and enteric nervous system abnormalities which are clinically associated with postnatal complications including feeding intolerance, altered gastrointestinal transit and NEC. The intestinal histopathological changes were not prevented by intravenous MSC treatment directly after HI, indicating that alternative treatment regimens for cell-based therapies should be explored.
Collapse
Affiliation(s)
- Maria Nikiforou
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Carolin Willburger
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Anja E de Jong
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Nico Kloosterboer
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Reint K Jellema
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Daan RMG Ophelders
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Harry WM Steinbusch
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Boris W Kramer
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- School of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Tim GAM Wolfs
- Department of Pediatrics, Maastricht University Medical Center, Maastricht, The Netherlands
- School of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
37
|
Burrello J, Monticone S, Gai C, Gomez Y, Kholia S, Camussi G. Stem Cell-Derived Extracellular Vesicles and Immune-Modulation. Front Cell Dev Biol 2016; 4:83. [PMID: 27597941 PMCID: PMC4992732 DOI: 10.3389/fcell.2016.00083] [Citation(s) in RCA: 191] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 08/02/2016] [Indexed: 12/13/2022] Open
Abstract
Extra-cellular vesicles (EVs) are bilayer membrane structures enriched with proteins, nucleic acids, and other active molecules and have been implicated in many physiological and pathological processes over the past decade. Recently, evidence suggests EVs to play a more dichotomic role in the regulation of the immune system, whereby an immune response may be enhanced or supressed by EVs depending on their cell of origin and its functional state. EVs derived from antigen (Ag)-presenting cells for instance, have been involved in both innate and acquired (or adaptive) immune responses, as Ag carriers or presenters, or as vehicles for delivering active signaling molecules. On the other hand, tumor and stem cell derived EVs have been identified to exert an inhibitory effect on immune responses by carrying immuno-modulatory effectors, such as transcriptional factors, non-coding RNA (Species), and cytokines. In addition, stem cell-derived EVs have also been reported to impair dendritic cell maturation and to regulate the activation, differentiation, and proliferation of B cells. They have been shown to control natural killer cell activity and to suppress the innate immune response (IIR). Studies reporting the role of EVs on T lymphocyte modulation are controversial. Discrepancy in literature may be due to stem cell culture conditions, methods of EV purification, EV molecular content, and functional state of both parental and target cells. However, mesenchymal stem cell-derived EVs were shown to play a more suppressive role by shifting T cells from an activated to a T regulatory phenotype. In this review, we will discuss how stem cell-derived EVs may contribute toward the modulation of the immune response. Collectively, stem cell-derived EVs mainly exhibit an inhibitory effect on the immune system.
Collapse
Affiliation(s)
- Jacopo Burrello
- Stem Cell Laboratory, Department of Medical Sciences, University of Torino Torino, Italy
| | - Silvia Monticone
- Stem Cell Laboratory, Department of Medical Sciences, University of Torino Torino, Italy
| | - Chiara Gai
- Stem Cell Laboratory, Department of Medical Sciences, University of Torino Torino, Italy
| | - Yonathan Gomez
- Stem Cell Laboratory, Department of Medical Sciences, University of Torino Torino, Italy
| | - Sharad Kholia
- Stem Cell Laboratory, Department of Medical Sciences, University of Torino Torino, Italy
| | - Giovanni Camussi
- Stem Cell Laboratory, Department of Medical Sciences, University of Torino Torino, Italy
| |
Collapse
|
38
|
Zhou A, Li M, He B, Feng W, Huang F, Xu B, Dunker AK, Balch C, Li B, Liu Y, Wang Y. Lipopolysaccharide treatment induces genome-wide pre-mRNA splicing pattern changes in mouse bone marrow stromal stem cells. BMC Genomics 2016; 17 Suppl 7:509. [PMID: 27557078 PMCID: PMC5001229 DOI: 10.1186/s12864-016-2898-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Lipopolysaccharide (LPS) is a gram-negative bacterial antigen that triggers a series of cellular responses. LPS pre-conditioning was previously shown to improve the therapeutic efficacy of bone marrow stromal cells/bone-marrow derived mesenchymal stem cells (BMSCs) for repairing ischemic, injured tissue. RESULTS In this study, we systematically evaluated the effects of LPS treatment on genome-wide splicing pattern changes in mouse BMSCs by comparing transcriptome sequencing data from control vs. LPS-treated samples, revealing 197 exons whose BMSC splicing patterns were altered by LPS. Functional analysis of these alternatively spliced genes demonstrated significant enrichment of phosphoproteins, zinc finger proteins, and proteins undergoing acetylation. Additional bioinformatics analysis strongly suggest that LPS-induced alternatively spliced exons could have major effects on protein functions by disrupting key protein functional domains, protein-protein interactions, and post-translational modifications. CONCLUSION Although it is still to be determined whether such proteome modifications improve BMSC therapeutic efficacy, our comprehensive splicing characterizations provide greater understanding of the intracellular mechanisms that underlie the therapeutic potential of BMSCs.
Collapse
Affiliation(s)
- Ao Zhou
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Bioinformatics Program, Indiana University School of Informatics, Indianapolis, IN, 46202, USA
| | - Meng Li
- College of Automation, Harbin Engineering University, Harbin, Heilongjiang, China
| | - Bo He
- College of Automation, Harbin Engineering University, Harbin, Heilongjiang, China
| | - Weixing Feng
- College of Automation, Harbin Engineering University, Harbin, Heilongjiang, China
| | - Fei Huang
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Bing Xu
- Department of Medical and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, China
| | - A Keith Dunker
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Curt Balch
- Bioscience Advising, Indianapolis, IN, 46227, USA
| | - Baiyan Li
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yunlong Liu
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Department of Medical and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yue Wang
- Department of Medical and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
39
|
Gimeno ML, Fuertes F, Barcala Tabarrozzi AE, Attorressi AI, Cucchiani R, Corrales L, Oliveira TC, Sogayar MC, Labriola L, Dewey RA, Perone MJ. Pluripotent Nontumorigenic Adipose Tissue-Derived Muse Cells have Immunomodulatory Capacity Mediated by Transforming Growth Factor-β1. Stem Cells Transl Med 2016; 6:161-173. [PMID: 28170177 PMCID: PMC5442729 DOI: 10.5966/sctm.2016-0014] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 06/07/2016] [Indexed: 02/06/2023] Open
Abstract
Adult mesenchymal stromal cell‐based interventions have shown promising results in a broad range of diseases. However, their use has faced limited effectiveness owing to the low survival rates and susceptibility to environmental stress on transplantation. We describe the cellular and molecular characteristics of multilineage‐differentiating stress‐enduring (Muse) cells derived from adipose tissue (AT), a subpopulation of pluripotent stem cells isolated from human lipoaspirates. Muse‐AT cells were efficiently obtained using a simple, fast, and affordable procedure, avoiding cell sorting and genetic manipulation methods. Muse‐AT cells isolated under severe cellular stress, expressed pluripotency stem cell markers and spontaneously differentiated into the three germ lineages. Muse‐AT cells grown as spheroids have a limited proliferation rate, a diameter of ∼15 µm, and ultrastructural organization similar to that of embryonic stem cells. Muse‐AT cells evidenced high stage‐specific embryonic antigen‐3 (SSEA‐3) expression (∼60% of cells) after 7–10 days growing in suspension and did not form teratomas when injected into immunodeficient mice. SSEA‐3+‐Muse‐AT cells expressed CD105, CD29, CD73, human leukocyte antigen (HLA) class I, CD44, and CD90 and low levels of HLA class II, CD45, and CD34. Using lipopolysaccharide‐stimulated macrophages and antigen‐challenged T‐cell assays, we have shown that Muse‐AT cells have anti‐inflammatory activities downregulating the secretion of proinflammatory cytokines, such as interferon‐γ and tumor necrosis factor‐α. Muse‐AT cells spontaneously gained transforming growth factor‐β1 expression that, in a phosphorylated SMAD2‐dependent manner, might prove pivotal in their observed immunoregulatory activity through decreased expression of T‐box transcription factor in T cells. Collectively, the present study has demonstrated the feasibility and efficiency of obtaining Muse‐AT cells that can potentially be harnessed as immunoregulators to treat immune‐related disorders. Stem Cells Translational Medicine2017;6:161–173
Collapse
Affiliation(s)
- María L. Gimeno
- Instituto de Investigación en Biomedicina de Buenos Aires, National Scientific and Technical Research Council (CONICET), Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Florencia Fuertes
- Instituto de Investigación en Biomedicina de Buenos Aires, National Scientific and Technical Research Council (CONICET), Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Andres E. Barcala Tabarrozzi
- Instituto de Investigación en Biomedicina de Buenos Aires, National Scientific and Technical Research Council (CONICET), Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Alejandra I. Attorressi
- Instituto de Investigación en Biomedicina de Buenos Aires, National Scientific and Technical Research Council (CONICET), Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | | | - Luis Corrales
- Servicio de Cirugía Plástica, Hospital Austral, Derqui, Argentina
| | - Talita C. Oliveira
- Biochemistry Department, Chemistry Institute, University of São Paulo, São Paulo, Brasil
| | - Mari C. Sogayar
- Cell and Molecular Therapy Center (Núcleo de Terapia Celular e Molecular/NETCEM), School of Medicine, University of São Paulo, São Paulo, Brasil
| | - Leticia Labriola
- Biochemistry Department, Chemistry Institute, University of São Paulo, São Paulo, Brasil
| | - Ricardo A. Dewey
- Laboratorio de Terapia Génica y Células Madre, Instituto de Investigaciones Biotecnológicas–Instituto Tecnológico de Chascomús (IIB‐INTECH), National Scientific and Technical Research Council, National University of General San Martin, Chascomús, Argentina
| | - Marcelo J. Perone
- Instituto de Investigación en Biomedicina de Buenos Aires, National Scientific and Technical Research Council (CONICET), Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| |
Collapse
|
40
|
Abstract
Despite recent therapeutic advances, patients with Crohn's disease (CD) continue to experience high recurrence with cumulative structural damage and ultimate loss of nutritional autonomy. With short bowel syndrome, strictures, and enteric fistulae being the underlying pathology, CD is the second common indication for home parenteral nutrition (HPN). With development of intestinal failure, nutritional management including HPN is required as a rescue therapy. Unfortunately, some patients do not escape the HPN-associated complications. Therefore, the concept of gut rehabilitation has evolved as part of the algorithmic management of these patients, with transplantation being the ultimate life-saving therapy. With type 2 intestinal failure, comprehensive rehabilitative measures including nutritional care, pharmacologic manipulation, autologous reconstruction, and bowel lengthening is often successful, particularly in patients with quiescent disease. With type 3 intestinal failure, transplantation is the only life-saving treatment for patients with HPN failure and intractable disease. With CD being the second common indication for transplantation in adults, survival outcome continues to improve because of surgical innovation, novel immunosuppression, and better postoperative care. Despite being a rescue therapy, the procedure has achieved survival rates similar to other solid organs, and comparable to those who continue to receive HPN therapy. With similar technical, immunologic, and infectious complications, survival is similar in the CD and non-CD recipients. Full nutritional autonomy is achievable in most survivors with better quality of life and long-term cost-effectiveness. CD recurrence is rare with no impact on graft function. Further progress is anticipated with new insights into the pathogenesis of CD and mechanisms of transplant tolerance.
Collapse
|
41
|
Shroff G. Human Embryonic Stem Cell Therapy in Crohn's Disease: A Case Report. AMERICAN JOURNAL OF CASE REPORTS 2016; 17:124-8. [PMID: 26923312 PMCID: PMC4774574 DOI: 10.12659/ajcr.896512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Patient: Male, 21 Final Diagnosis: Crohn’s disease Symptoms: Intolerance to specific foods • abdominal pain and diarrhea Medication: Human embryonic stem cell therapy Clinical Procedure: Human embryonic stem cell transplantation Specialty: Gastroenterology
Collapse
Affiliation(s)
- Geeta Shroff
- Stem Cell Therapy, Nutech Mediworld, New Delhi, India
| |
Collapse
|
42
|
Abstract
Kidney transplantation is the best treatment for end-stage renal disease, but its implementation is limited by organ shortage and immune rejection. Side effects of current immunosuppressive drugs, such as nephrotoxicity, opportunistic infection, and tumorigenic potential, influence long-term graft outcomes. In recent years, continued research and subsequent discoveries concerning the properties and potential utilization of mesenchymal stem cells (MSCs) have aroused considerable interest and expectations. Biological characteristics of MSCs, including multi-lineage differentiation, homing potential, paracrine effect and immunomodulation, have opened new horizons for applications in kidney transplantation. However, many studies have shown that the biological activity of MSCs depends on internal inflammatory conditions, and the safety and efficacy of the clinical application of MSCs remain controversial. This review summarizes the findings of a large number of studies and aims to provide an objective viewpoint based on a comprehensive analysis of the presently established benefits and obstacles of implementing MSC-based therapy in kidney transplantation, and to promote its clinical translation.
Collapse
Affiliation(s)
- Cheng Chen
- Department of Urology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, PR China
| | - Jianquan Hou
- Department of Urology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, PR China.
| |
Collapse
|
43
|
Carvedilol protects bone marrow stem cells against hydrogen peroxide-induced cell death via PI3K-AKT pathway. Biomed Pharmacother 2016; 78:257-263. [PMID: 26898450 DOI: 10.1016/j.biopha.2016.01.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 01/03/2016] [Accepted: 01/13/2016] [Indexed: 02/06/2023] Open
Abstract
Carvedilol, a nonselective β-adrenergic receptor blocker, has been reported to exert potent anti-oxidative activities. In the present study, we aimed to investigate the effects of carvedilol against hydrogen peroxide (H2O2)-induced bone marrow-derived mesenchymal stem cells (BMSCs) death, which imitate the microenvironment surrounding transplanted cells in the injured spinal cord in vitro. Carvedilol significantly reduced H2O2-induced reactive oxygen species production, apoptosis and subsequent cell death. LY294002, the PI3K inhibitor, blocked the protective effects and up-regulation of Akt phosphorylation of carvedilol. Together, our results showed that carvedilol protects H2O2-induced BMSCs cell death partly through PI3K-Akt pathway, suggesting carvedilol could be used in combination with BMSCs for the treatment of spinal cord injury by improving the cell survival and oxidative stress microenvironments.
Collapse
|
44
|
Lener T, Gimona M, Aigner L, Börger V, Buzas E, Camussi G, Chaput N, Chatterjee D, Court FA, Del Portillo HA, O'Driscoll L, Fais S, Falcon-Perez JM, Felderhoff-Mueser U, Fraile L, Gho YS, Görgens A, Gupta RC, Hendrix A, Hermann DM, Hill AF, Hochberg F, Horn PA, de Kleijn D, Kordelas L, Kramer BW, Krämer-Albers EM, Laner-Plamberger S, Laitinen S, Leonardi T, Lorenowicz MJ, Lim SK, Lötvall J, Maguire CA, Marcilla A, Nazarenko I, Ochiya T, Patel T, Pedersen S, Pocsfalvi G, Pluchino S, Quesenberry P, Reischl IG, Rivera FJ, Sanzenbacher R, Schallmoser K, Slaper-Cortenbach I, Strunk D, Tonn T, Vader P, van Balkom BWM, Wauben M, Andaloussi SE, Théry C, Rohde E, Giebel B. Applying extracellular vesicles based therapeutics in clinical trials - an ISEV position paper. J Extracell Vesicles 2015; 4:30087. [PMID: 26725829 PMCID: PMC4698466 DOI: 10.3402/jev.v4.30087] [Citation(s) in RCA: 1051] [Impact Index Per Article: 105.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 12/11/2015] [Accepted: 12/13/2015] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs), such as exosomes and microvesicles, are released by different cell types and participate in physiological and pathophysiological processes. EVs mediate intercellular communication as cell-derived extracellular signalling organelles that transmit specific information from their cell of origin to their target cells. As a result of these properties, EVs of defined cell types may serve as novel tools for various therapeutic approaches, including (a) anti-tumour therapy, (b) pathogen vaccination, (c) immune-modulatory and regenerative therapies and (d) drug delivery. The translation of EVs into clinical therapies requires the categorization of EV-based therapeutics in compliance with existing regulatory frameworks. As the classification defines subsequent requirements for manufacturing, quality control and clinical investigation, it is of major importance to define whether EVs are considered the active drug components or primarily serve as drug delivery vehicles. For an effective and particularly safe translation of EV-based therapies into clinical practice, a high level of cooperation between researchers, clinicians and competent authorities is essential. In this position statement, basic and clinical scientists, as members of the International Society for Extracellular Vesicles (ISEV) and of the European Cooperation in Science and Technology (COST) program of the European Union, namely European Network on Microvesicles and Exosomes in Health and Disease (ME-HaD), summarize recent developments and the current knowledge of EV-based therapies. Aspects of safety and regulatory requirements that must be considered for pharmaceutical manufacturing and clinical application are highlighted. Production and quality control processes are discussed. Strategies to promote the therapeutic application of EVs in future clinical studies are addressed.
Collapse
Affiliation(s)
- Thomas Lener
- Spinal Cord Injury & Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), Salzburg, Austria
- Department of Blood Group Serology and Transfusion Medicine, University Hospital, Salzburger Landeskliniken GesmbH (SALK), Salzburg, Austria
| | - Mario Gimona
- Spinal Cord Injury & Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), Salzburg, Austria
- Department of Blood Group Serology and Transfusion Medicine, University Hospital, Salzburger Landeskliniken GesmbH (SALK), Salzburg, Austria
| | - Ludwig Aigner
- Spinal Cord Injury & Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), Salzburg, Austria
| | - Verena Börger
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Edit Buzas
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Giovanni Camussi
- Molecular Biotechnology Center, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Nathalie Chaput
- Laboratory of Immunomonitoring in Oncology, UMS 3655 CNRS/US23 Inserm, Villejuif, France
- Centre of Clinical Investigation in Biotherapy CICBT 1248, Institut Gustave Roussy, Villejuif, France
| | - Devasis Chatterjee
- Division of Hematology & Oncology, Rhode Island Hospital, Providence, RI, USA
- The Alpert Medical School of Brown University, Providence, RI, USA
| | - Felipe A Court
- Department of Physiology, Faculty of Biology, Pontificia-Universidad Católica de Chile, Santiago, Chile
| | - Hernando A Del Portillo
- ICREA at Barcelona Centre for International Health Research (CRESIB), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigació Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland
- Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Stefano Fais
- Anti-Tumor Drugs Section, Department of Therapeutic Research and Medicines Evaluation, National Institute of Health (ISS), Rome, Italy
| | - Juan M Falcon-Perez
- Metabolomics Unit, CIC bioGUNE, CIBERehd, Bizkaia Technology Park, Derio, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Ursula Felderhoff-Mueser
- Department of Paediatrics I, Neonatology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Lorenzo Fraile
- Departament de Producció Animal, ETSEA, Universitat de Lleida, Lleida, Spain
| | - Yong Song Gho
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - André Görgens
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ramesh C Gupta
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - An Hendrix
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University Hospital, Ghent, Belgium
| | - Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Andrew F Hill
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | | | - Peter A Horn
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | | | - Lambros Kordelas
- Department of Bone Marrow Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Boris W Kramer
- Experimental Perinatology/Neonatology, School of Mental Health and Neuroscience, School of Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Eva-Maria Krämer-Albers
- Molecular Cell Biology and Focus Program Translational Neurosciences, University of Mainz, Mainz, Germany
| | - Sandra Laner-Plamberger
- Spinal Cord Injury & Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), Salzburg, Austria
- Department of Blood Group Serology and Transfusion Medicine, University Hospital, Salzburger Landeskliniken GesmbH (SALK), Salzburg, Austria
| | - Saara Laitinen
- Research and Cell Services, Finnish Red Cross Blood Service, Helsinki, Finland
| | - Tommaso Leonardi
- Division of Stem Cell Neurobiology, Department of Clinical Neurosciences, Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
| | - Magdalena J Lorenowicz
- Department of Cell Biology, Center for Molecular Medicine, University Medical Center, Utrecht, The Netherlands
| | - Sai Kiang Lim
- Institute of Medical Biology, Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine, the Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Casey A Maguire
- Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Antonio Marcilla
- Dpto. Biología Celular y Parasitologia, Facultat de Farmacia, Universitat de Valencia, Valencia, Spain
- Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Universitat de València-Health Research Institute La Fe, Valencia, Spain
| | - Irina Nazarenko
- Institute for Environmental Health Sciences and Hospital Infection Control Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | - Takahiro Ochiya
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo, Japan
| | - Tushar Patel
- Departments of Transplantation and Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Shona Pedersen
- Centre for Cardiovascular Research, Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg University, Aalborg, Denmark
| | - Gabriella Pocsfalvi
- Mass Spectrometry and Proteomics, Institute of Biosciences and BioResources, National Research Council of Italy, Naples, Italy
| | - Stefano Pluchino
- Division of Stem Cell Neurobiology, Department of Clinical Neurosciences, Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Peter Quesenberry
- Division of Hematology & Oncology, Rhode Island Hospital, Providence, RI, USA
- The Alpert Medical School of Brown University, Providence, RI, USA
| | - Ilona G Reischl
- BASG - Bundesamt für Sicherheit im Gesundheitswesen - Federal Office for Safety in Health Care, AGES - Agentur für Gesundheit und Ernährungssicherheit - Austrian Agency for Health and Food Safety, Institut Überwachung - Institute Surveillance, Wien, Austria
| | - Francisco J Rivera
- Institute of Molecular Regenerative Medicine, Spinal Cord Injury & Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), Salzburg, Austria
| | - Ralf Sanzenbacher
- Ralf Sanzenbacher, Paul-Ehrlich-Institut, Bundesinstitut für Impfstoffe und biomedizinische Arzneimittel, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Katharina Schallmoser
- Spinal Cord Injury & Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), Salzburg, Austria
- Department of Blood Group Serology and Transfusion Medicine, University Hospital, Salzburger Landeskliniken GesmbH (SALK), Salzburg, Austria
| | - Ineke Slaper-Cortenbach
- Cell Therapy Facility, Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dirk Strunk
- Experimental & Clinical Cell Therapy Institute, Spinal Cord Injury & Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Torsten Tonn
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
| | - Pieter Vader
- Laboratory of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Bas W M van Balkom
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marca Wauben
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Samir El Andaloussi
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Clotilde Théry
- Centre of Clinical Investigation in Biotherapy CICBT 1248, Institut Gustave Roussy, Villejuif, France
- INSERM U932, Institut Curie, Paris, France
| | - Eva Rohde
- Spinal Cord Injury & Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), Salzburg, Austria
- Department of Blood Group Serology and Transfusion Medicine, University Hospital, Salzburger Landeskliniken GesmbH (SALK), Salzburg, Austria;
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany;
| |
Collapse
|
45
|
The Modulatory Effects of Mesenchymal Stem Cells on Osteoclastogenesis. Stem Cells Int 2015; 2016:1908365. [PMID: 26823668 PMCID: PMC4707367 DOI: 10.1155/2016/1908365] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 09/21/2015] [Indexed: 12/14/2022] Open
Abstract
The effect of mesenchymal stem cells (MSCs) on bone formation has been extensively demonstrated through several in vitro and in vivo studies. However, few studies addressed the effect of MSCs on osteoclastogenesis and bone resorption. Under physiological conditions, MSCs support osteoclastogenesis through producing the main osteoclastogenic cytokines, RANKL and M-CSF. However, during inflammation, MSCs suppress osteoclast formation and activity, partly via secretion of the key anti-osteoclastogenic factor, osteoprotegerin (OPG). In vitro, co-culture of MSCs with osteoclasts in the presence of high concentrations of osteoclast-inducing factors might reflect the in vivo inflammatory pathology and prompt MSCs to exert an osteoclastogenic suppressive effect. MSCs thus seem to have a dual effect, by stimulating or inhibiting osteoclastogenesis, depending on the inflammatory milieu. This effect of MSCs on osteoclast formation seems to mirror the effect of MSCs on other immune cells, and may be exploited for the therapeutic potential of MSCs in bone loss associated inflammatory diseases.
Collapse
|
46
|
Nguyen A, Guo J, Banyard DA, Fadavi D, Toranto JD, Wirth GA, Paydar KZ, Evans GRD, Widgerow AD. Stromal vascular fraction: A regenerative reality? Part 1: Current concepts and review of the literature. J Plast Reconstr Aesthet Surg 2015; 69:170-9. [PMID: 26565755 DOI: 10.1016/j.bjps.2015.10.015] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 08/25/2015] [Accepted: 10/13/2015] [Indexed: 12/21/2022]
Abstract
Stromal Vascular Fraction (SVF) is a heterogeneous collection of cells contained within adipose tissue that is traditionally isolated using enzymes such as collagenase. With the removal of adipose cells, connective tissue and blood from lipoaspirate, comes the SVF, a mix including mesenchymal stem cells, endothelial precursor cells, T regulatory cells, macrophages, smooth muscle cells, pericytes and preadipocytes. In part 1 of our 2-part series, we review the literature with regards to the intensifying interest that has shifted toward this mixture of cells, particularly due to its component synergy and translational potential. Trials assessing the regenerative potential of cultured Adipose Derived Stem Cells (ADSCs) and SVF demonstrate that SVF is comparably effective in treating conditions ranging from radiation injuries, burn wounds and diabetes, amongst others. Aside from their use in chronic conditions, SVF enrichment of fat grafts has proven a major advance in maintaining fat graft volume and viability. Many SVF studies are currently in preclinical phases or are moving to human trials. Overall, regenerative cell therapy based on SVF is at an early investigative stage but its potential for clinical application is enormous.
Collapse
Affiliation(s)
- Andrew Nguyen
- Center for Tissue Engineering, Department of Plastic Surgery, University of California, Irvine, USA
| | - James Guo
- Center for Tissue Engineering, Department of Plastic Surgery, University of California, Irvine, USA
| | - Derek A Banyard
- Center for Tissue Engineering, Department of Plastic Surgery, University of California, Irvine, USA
| | - Darya Fadavi
- Center for Tissue Engineering, Department of Plastic Surgery, University of California, Irvine, USA
| | - Jason D Toranto
- Center for Tissue Engineering, Department of Plastic Surgery, University of California, Irvine, USA
| | - Garrett A Wirth
- Center for Tissue Engineering, Department of Plastic Surgery, University of California, Irvine, USA
| | - Keyianoosh Z Paydar
- Center for Tissue Engineering, Department of Plastic Surgery, University of California, Irvine, USA
| | - Gregory R D Evans
- Department of Plastic Surgery, University of California, Irvine, USA
| | - Alan D Widgerow
- Center for Tissue Engineering, Department of Plastic Surgery, University of California, Irvine, USA.
| |
Collapse
|
47
|
Roubeix C, Godefroy D, Mias C, Sapienza A, Riancho L, Degardin J, Fradot V, Ivkovic I, Picaud S, Sennlaub F, Denoyer A, Rostene W, Sahel JA, Parsadaniantz SM, Brignole-Baudouin F, Baudouin C. Intraocular pressure reduction and neuroprotection conferred by bone marrow-derived mesenchymal stem cells in an animal model of glaucoma. Stem Cell Res Ther 2015; 6:177. [PMID: 26377305 PMCID: PMC4574127 DOI: 10.1186/s13287-015-0168-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 01/28/2015] [Accepted: 08/21/2015] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Glaucoma is a sight-threatening retinal neuropathy associated with elevated intraocular pressure (IOP) due to degeneration and fibrosis of the trabecular meshwork (TM). Glaucoma medications aim to reduce IOP without targeting the specific TM pathology, Bone-marrow mesenchymal stem cells (MSCs) are used today in various clinical studies. Here, we investigated the potential of MSCs therapy in an glaucoma-like ocular hypertension (OHT) model and decipher in vitro the effects of MSCs on primary human trabecular meshwork cells. METHODS Ocular hypertension model was performed by cauterization of 3 episcleral veins (EVC) of Long-Evans male rat eyes. MSCs were isolated from rat bone marrow, amplified in vitro and tagged with quantum dot nanocrystals. Animals were distributed as 1) MSCs group receiving 5.10(5)cells/6μl Minimum Essential Medium and 2) MEM group receiving 6μl MEM (n = 10 each). Injections were performed into the anterior chamber of 20 days-hypertensive eyes and IOP was monitored twice a week for 4 weeks. At the end of experiment, cell distribution in the anterior segment was examined in confocal microscopy on flat mounted corneas. Moreover, we tested in vitro effects of MSCs conditioned medium (MSC-CM) on primary human trabecular meshwork cells (hTM cells) using Akt activation, myosin phosphorylation and TGF-β2-dependent profibrotic phenotype in hTM cells. RESULTS We demonstrated a rapid and long-lasting in vivo effect of MSCs transplantation that significantly reduced IOP in hypertensive eyes induced by EVC. MSCs were located to the ciliary processes and the TM. Enumeration of RGCs on whole flat-mounted retina highlighted a protective effect of MSCs on RGCs death. In vitro, MSC-CM promotes: (i) hTM cells survival by activating the antiapoptotic pathway, Akt, (ii) hTM cells relaxation as analyzed by the decrease in myosin phosphorylation and (iii) inhibition of TGF-β2-dependent profibrotic phenotype acquisition in hTM cells. CONCLUSIONS MSCs injection in the ocular anterior chamber in a rat model of OHT provides neuroprotective effect in the glaucoma pathophysiology via TM protection. These results demonstrate that MSCs constitute promising tool for treating ocular hypertension and retinal cell degeneration.
Collapse
Affiliation(s)
- Christophe Roubeix
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
| | - David Godefroy
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
| | - Céline Mias
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR 1048, 31432 Toulouse cedex 4, France, Toulouse, France.
| | - Anaïs Sapienza
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
| | - Luisa Riancho
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
| | - Julie Degardin
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
| | - Valérie Fradot
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
| | - Ivana Ivkovic
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
| | - Serge Picaud
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
| | - Florian Sennlaub
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
| | - Alexandre Denoyer
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-DHOS CIC 503, Paris, F-75012, France.
| | - William Rostene
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
| | - José Alain Sahel
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-DHOS CIC 503, Paris, F-75012, France.
| | - Stéphane Melik Parsadaniantz
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
| | - Françoise Brignole-Baudouin
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-DHOS CIC 503, Paris, F-75012, France.
- University Paris Descartes, Sorbonne Paris Cité, Paris, F-75006, France.
- Faculté de Pharmacie de Paris, University Paris Descartes, Sorbonne Paris Cité, Paris, F-75006, France.
| | - Christophe Baudouin
- INSERM, U968, Paris, F-75012, France.
- UPMC Université Paris 06, UMR_S 968, Institut de la Vision, Paris, F-75012, France.
- CNRS, UMR_7210, Paris, F-75012, France.
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-DHOS CIC 503, Paris, F-75012, France.
- Department of Ophthalmology, Hôpital Ambroise Pare, AP HP, Boulogne, F-92100, France.
- University Versailles St Quentin en Yvelines, Montigny-Le-Bretonneux, F-78180, France.
| |
Collapse
|
48
|
Kavanagh DPJ, Suresh S, Newsome PN, Frampton J, Kalia N. Pretreatment of Mesenchymal Stem Cells Manipulates Their Vasculoprotective Potential While Not Altering Their Homing Within the Injured Gut. Stem Cells 2015; 33:2785-97. [PMID: 26124062 PMCID: PMC4737111 DOI: 10.1002/stem.2061] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 04/20/2015] [Indexed: 12/15/2022]
Abstract
Mesenchymal stem cells (MSCs) have shown therapeutic promise in many experimental and clinical models of inflammation. However, a commonly reported feature of MSC transplantation is poor homing to injured tissues. Previously, we have shown that pretreatment with cytokines/chemical factors enhances hematopoietic SC adhesion within intestinal microvasculature following ischemia-reperfusion (IR) injury. Using intravital microscopy, the ability of similar pretreatment strategies to enhance the recruitment of murine MSCs to murine intestinal microvasculature following IR injury was investigated. Primary MSCs were isolated from bone marrow and selected on the basis of platelet-derived growth factor receptor-α and SC antigen-1 positivity (PDGFRα(+) /Sca-1(+) ). MSC recruitment was similar in IR injured gut mucosa when compared with sham operated controls, with limited cell adhesion observed. MSCs appeared contorted in microvessels, suggesting physical entrapment. Although not recruited specifically by injury, MSC administration significantly reduced neutrophil recruitment and improved tissue perfusion in the severely injured jejunum. Vasculoprotective effects were not demonstrated in the lesser injured ileum. Pretreatment of MSCs with tumor necrosis factor (TNF)-α, CXCL12, interferon (IFN)-γ, or hydrogen peroxide did not enhance their intestinal recruitment. In fact, TNFα and IFNγ removed the previous therapeutic ability of transplanted MSCs to reduce neutrophil infiltration and improve perfusion in the jejunum. We provide direct evidence that MSCs can rapidly limit leukocyte recruitment and improve tissue perfusion following intestinal IR injury. However, this study also highlights complexities associated with strategies to improve MSC therapeutic efficacy. Future studies using cytokine/chemical pretreatments to enhance MSC recruitment/function require careful consideration and validation to ensure therapeutic function is not impeded.
Collapse
Affiliation(s)
- Dean P J Kavanagh
- Centre for Cardiovascular Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Shankar Suresh
- NIHR Centre for Liver Research and Biomedical Research Unit, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Philip N Newsome
- NIHR Centre for Liver Research and Biomedical Research Unit, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Jon Frampton
- School of Immunity and Infection, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Neena Kalia
- Centre for Cardiovascular Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| |
Collapse
|
49
|
Umbilical Cord-Derived Mesenchymal Stem Cells Inhibit Cadherin-11 Expression by Fibroblast-Like Synoviocytes in Rheumatoid Arthritis. J Immunol Res 2015; 2015:137695. [PMID: 26090476 PMCID: PMC4451296 DOI: 10.1155/2015/137695] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 03/18/2015] [Accepted: 03/26/2015] [Indexed: 02/06/2023] Open
Abstract
This study aimed to determine whether umbilical cord-derived mesenchymal stem cells (UCMSC) regulate Cadherin-11 (CDH11) expression by fibroblast-like synoviocytes (FLS) in rheumatoid arthritis (RA). FLS were isolated from the synovium of RA and osteoarthritis (OA) patients. FLS from RA patients were cocultured with UCMSC in a transwell system. CDH11 mRNA levels in FLS were tested, and levels of soluble factors expressed by UCMSC, such as indoleamine 2,3-dioxygenase (IDO), hepatocyte growth factor (HGF), and interleukin- (IL-) 10, were determined. IDO, HGF, and IL-10 were upregulated in cocultures, so that appropriate inhibitors were added before determination of CDH11 expression. The effects of UCMSC on arthritis were investigated in the collagen-induced arthritis (CIA) model in Wistar rats. FLS from RA patients expressed higher CDH11 levels than those from OA patients, and this effect was suppressed by UCMSC. The inhibitory effect of UCMSC on CDH11 expression by FLS was abolished by suppression of IL-10 activity. CDH11 expression in synovial tissues was higher in the context of CIA than under basal conditions, and this effect was prevented by UCMSC administration. IL-10 mediates the inhibitory effect of UCMSC on CDH11 expression by FLS, and this mechanism might be targeted to ameliorate arthritis.
Collapse
|
50
|
Arnberg F, Lundberg J, Olsson A, Samén E, Jaff N, Jussing E, Dahlén U, Nava S, Axelsson R, Ringdén O, Kaipe H, Holmin S. Intra-arterial Administration of Placenta-Derived Decidual Stromal Cells to the Superior Mesenteric Artery in the Rabbit: Distribution of Cells, Feasibility, and Safety. Cell Transplant 2015; 25:401-10. [PMID: 25976072 DOI: 10.3727/096368915x688191] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Selective administration of mesenchymal stromal cells to the mesenteric arteries is a potential technique to overcome pulmonary trapping and increase the density of transplanted cells in extensive mural inflammation of the intestine, such as in inflammatory bowel disease and graft-versus-host disease. We injected 5 × 10(6) (111)In-oxine-labeled human decidual stromal cells (DSCs) to the rabbit superior mesenteric artery (SMA) using clinical routine catheters guided by an angiographical system under sterile conditions. We used longitudinal single-photon emission tomography at 6 h and at 1, 2, and 5 days to assess trafficking and distribution of DSCs. We used digital subtraction angiography, computed tomography, and hematoxylin and eosin stainings to determine biodistribution of cells and to assess safety end points. We found that selective injection of human DSCs to the rabbit SMA does not result in acute embolic complications. Furthermore, we found that IV administration resulted in extensive retention of the radiolabeled DSCs in the lungs, corroborating previous studies on pulmonary trapping. In sharp contrast, selective injections to the SMA resulted in uptake distributed in the intestine supplied by the SMA and in the liver, indicating that this approach could significantly increase the fraction of injected DSCs reaching the target tissue.
Collapse
Affiliation(s)
- Fabian Arnberg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|