Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Cell Journal [Yakhteh]. 2017; 19 (1): 137-145
in English | IMEMR | ID: emr-185800

ABSTRACT

Objective: Adipose derived stem cells [ASCs], as one of the important stromal cells in the tumor microenvironment, are determined with immunomodulatory effects. The principle aim of this study was to evaluate the immunosuppressive effects of ASCs on natural killer [NK] cells


Materials and Methods: In this experimental study, we assessed the expressions of indolamine 2, 3-dioxygenase [IDO1], IDO2 and human leukocyte antigen-G5 [HLA-G5] in ASCs isolated from breast cancer patients with different stages as well as normal individuals, using quantitative reverse transcriptase-polymerase chain reaction [qRT-PCR]. Immunomodulatory effects of ASCs on the expression of CD16, CD56, CD69, NKG2D, NKp30, NKG2A and NKp44 was also assessed in peripheral blood lymphocytes [PBLs] by flow-cytometry


Results: Our result showed that IDO1, IDO2 and HLA-G5 had higher mRNA expressions in ASCs isolated from breast cancer patients than those from normal individuals [P>0.05]. mRNA expression of these molecules were higher in ASCs isolated from breast cancer patients with stage III tumors than those with stage II. The indirect culture of ASCs isolated from breast cancer patients and normal individuals with activated PBLs significantly reduced NKG2D+ and CD69+ NK cells [P<0.05]


Conclusion: Results of the present study suggest more evidences for the immunosuppression of ASCs on NK cells, providing conditions in favor of tumor immune evasion


Subject(s)
Adult , Female , Humans , Middle Aged , Adipose Tissue/cytology , Mesenchymal Stem Cells/immunology , Killer Cells, Natural/immunology , Immunomodulation , Immunosuppression Therapy , Iran
3.
Biol. Res ; 48: 1-8, 2015. ilus, graf, tab
Article in English | LILACS | ID: lil-734619

ABSTRACT

BACKGROUND: Mesenchymal stem cells (MSCs) are considered the best candidate in stem cells therapy due to their multipotent differentiation ability, low expression of co-stimulatory molecules (CD80, CD86, CD34 and HLA-II) and immunosuppression effects on in vivo immune responses. MSCs were now widely used in clinical trials but received no encourage results. The major problem was the fate of engrafted MSCs in vivo could not be defined. Some studies indicated that MSCs could induce immune response and result in the damage and rejection of MSCs. As toll like receptors (TLRs) are important in inducing of immune responses, in this study we study the role of TLR7 in mediating the immune status of MSCs isolated from umbilical cord. RESULTS: Our results indicated that TLR7 agonist Imiquimod could increase the proliferation of PBMC isolated from healthy human volunteers and release of lactate dehydrogenase (LDH) in supernatant from PBMC-UCMSCs co-culture system. Flow cytometry and quantitative PCR also confirmed the regulated expression of surface co-stimulatory molecules and pro-inflammatory genes (IL-6, IL-8, IL-12, TGF-β and TNF-α). And the down-regulation expression of stem cell markers also confirmed the loss of stemness of UCMSCs. We also found that the osteo-differentiation ability of UCMSCs was enhanced in the presence of Imiquimod. CONCLUSION: To our knowledge, this is the first report that activation of TLR7 pathway increases the immunogenicity of UCMSCs. Extensive researches have now been conducted to study whether the change of immune status will be help in tumor rejection based on the tumor-tropism of MSCs.


Subject(s)
Humans , Adjuvants, Immunologic/pharmacology , Aminoquinolines/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/immunology , /agonists , Antigens, CD/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Flow Cytometry , Gene Expression Regulation/drug effects , /analysis , /analysis , /analysis , L-Lactate Dehydrogenase/drug effects , L-Lactate Dehydrogenase , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Membrane Proteins/drug effects , Osteogenesis/drug effects , Real-Time Polymerase Chain Reaction , Transforming Growth Factor beta/analysis , Tumor Necrosis Factor-alpha/analysis
4.
The Korean Journal of Internal Medicine ; : 580-589, 2015.
Article in English | WPRIM | ID: wpr-216634

ABSTRACT

Currently, the most effective treatment for end-stage liver fibrosis is liver transplantation; however, transplantation is limited by a shortage of donor organs, surgical complications, immunological rejection, and high medical costs. Recently, mesenchymal stem cell (MSC) therapy has been suggested as an effective alternate approach for the treatment of hepatic diseases. MSCs have the potential to differentiate into hepatocytes, and therapeutic value exists in their immune-modulatory properties and secretion of trophic factors, such as growth factors and cytokines. In addition, MSCs can suppress inflammatory responses, reduce hepatocyte apoptosis, increase hepatocyte regeneration, regress liver fibrosis and enhance liver functionality. Despite these advantages, issues remain; MSCs also have fibrogenic potential and the capacity to promote tumor cell growth and oncogenicity. This paper summarizes the properties of MSCs for regenerative medicine and their therapeutic mechanisms and clinical application in the treatment of liver fibrosis. We also present several outstanding risks, including their fibrogenic potential and their capacity to promote pre-existing tumor cell growth and oncogenicity.


Subject(s)
Animals , Humans , Cell Differentiation , Cell Proliferation , Hepatocytes/immunology , Liver/immunology , Liver Cirrhosis/diagnosis , Liver Regeneration , Mesenchymal Stem Cell Transplantation/adverse effects , Mesenchymal Stem Cells/immunology , Phenotype , Regenerative Medicine/methods , Risk Factors , Signal Transduction , Treatment Outcome
5.
Egyptian Journal of Hospital Medicine [The]. 2014; 57 (October): 403-417
in English | IMEMR | ID: emr-160239

ABSTRACT

The ideal stem cell for use in functional tissue engineering needs to be abundantly available, harvested with minimal morbidity, differentiated reliably down various pathways and able to be transplanted safely and efficaciously. Adult human adipose tissue contains a population of mesenchymal stem cells; adipose-derived stem cells, which seem to fulfil most, if not all, of these criteria. In this work, we investigated the immunogenicity properties of human adipose-derived mesenchymal stem cells [HAMSCs] and their effect on monocytes differentiation. The HAMSCs have been isolated and specified. Human peripheral blood mononuclear cells [PBMCs] were isolated and passed through a column with magnetic beads coated with anti-CD14 antibody. CD14+ ve cells were isolated and cultured independently or co-cultured with HAMSCs in the presence of cytokines [IL-4, Granulocyte-macrophage colony-stimulating factor [GM-CSF]] to induce their differentiation into dendritic cells [DCs]. Their further maturation was induced by LPS added on the 6[th] day of culture. The major part of the independently cultured cells [CD14+ ve] was found to express the markers which are considered to be specific for the mature dendritic cells such as Human leukocyte antigen-DR [HLA-DR] [40.44%] and low percentage of cells [6.9%]. Nevertheless dendritic cells of monocyte origin [mDCs] co-cultured with HAMSCs showed significant shifts in the pattern of surface markers. The percentage of HLA-DR cells was much lower [6.44%] compared to control cultures [p < 0.001]. Similarly, the secretion of IL-10 by DCs was up-regulated in co-cultures of HAMSCs and DCs. The results show that human adipose-tissue mesenchymal stem cells [HAMSCs] could inhibit the differentiation of the blood monocytes into dendritic cells


Subject(s)
Humans , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/physiology , Mesenchymal Stem Cells/ultrastructure , Monocytes/ultrastructure , Lipopolysaccharide Receptors
6.
Korean Journal of Ophthalmology ; : 246-256, 2014.
Article in English | WPRIM | ID: wpr-51380

ABSTRACT

PURPOSE: This study investigated interleukin (IL)-17-secreting cell involvement in sterile inflammation, and evaluated the effect of mesenchymal stem cells (MSCs) on IL-17-secreting cell immunologic profiling. METHODS: Twenty mice were sacrificed at time points of 6 hours, 1 day, 1 week, and 3 weeks (each group, n = 5) after the cornea was chemically injured with 0.5N NaOH; IL-17 changes in the cornea were evaluated using enzyme-linked immunosorbent assay. Further, IL-17 secreting cells were assessed in the cervical lymph nodes by a flow cytometer. Rat MSCs were applied intraperitoneally in a burn model (n = 10), IL-17-secreting T helper 17 (Th17) cell and non-Th17 cell changes were checked using a flow cytometer in both cornea and cervical lymph nodes at 1week, and compared with those in the positive control (n = 10). RESULTS: IL-17 was highest in the cornea at 1 week, while, in the cervical lymph nodes, IL-17-secreting cells showed early increase at 6 hours, and maintained the increase through 1 day to 1 week, and levels returned to the basal level at 3 weeks. Specifically, the non-Th17 cells secreted IL-17 earlier than the Th17 cells. When the MSCs were applied, IL-17 secretion was reduced in CD3(+)CD4(-)CD8(-), CD3(+)CD4(+)CD8(-), and CD3(+) CD4(-)CD8(+) cells of the cervical lymph nodes by 53.7%, 43.8%, and 50.8%, respectively. However, in the cornea, IL-17 secretion of CD3(+)CD4(-)CD8(-) cells was completely blocked. CONCLUSIONS: The results indicated that both IL-17-secreting non-Th17 and Th17 cells were involved in the chemical burn model, and MSCs appeared to mainly modulate non-Th17 cells and also partially suppress the Th17 cells.


Subject(s)
Animals , Male , Mice , Burns, Chemical/immunology , Cells, Cultured , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Eye Burns/immunology , Flow Cytometry , Immunity, Cellular , Interleukin-17/metabolism , Mesenchymal Stem Cells/immunology , Mice, Inbred C57BL
7.
Rev. bras. hematol. hemoter ; 35(1): 62-67, 2013.
Article in English | LILACS | ID: lil-670462

ABSTRACT

Although bone marrow is the main source, mesenchymal stem cells have already been isolated from various other tissues, such as the liver, pancreas, adipose tissue, peripheral blood and dental pulp. These plastic adherent cells are morphologically similar to fibroblasts and have a high proliferative potential. This special group of cells possesses two essential characteristics: self-renewal and differentiation, with appropriate stimuli, into various cell types. Mesenchymal stem cells are considered immunologically privileged, since they do not express costimulatory molecules, required for complete T cell activation, on their surface. Several studies have shown that these cells exert an immunosuppressive effect on cells from both innate and acquired immunity systems. Mesenchymal stem cells can regulate the immune response in vitro by inhibiting the maturation of dendritic cells, as well as by suppressing the proliferation and function of T and B lymphocytes and natural killer cells. These special properties of mesenchymal stem cells make them a promising strategy in the treatment of immune mediated disorders, such as graft-versus-host disease and autoimmune diseases, as well as in regenerative medicine. The understanding of immune regulation mechanisms of mesenchymal stem cells, and also those involved in the differentiation of these cells in various lineages is primordial for their successful and safe application in different areas of medicine.


Subject(s)
Humans , Autoimmune Diseases/therapy , Stem Cells , Cell Differentiation , Immunomodulation , Mesenchymal Stem Cells/immunology , Immune System
8.
The Korean Journal of Internal Medicine ; : 387-402, 2013.
Article in English | WPRIM | ID: wpr-53537

ABSTRACT

Mesenchymal stem cells (MSCs) are self-renewing, multipotent progenitor cells with multilineage potential to differentiate into cell types of mesodermal origin, such as adipocytes, osteocytes, and chondrocytes. In addition, MSCs can migrate to sites of inflammation and exert potent immunosuppressive and anti-inflammatory effects through interactions between lymphocytes associated with both the innate and adaptive immune system. Along with these unique therapeutic properties, their ease of accessibility and expansion suggest that use of MSCs may be a useful therapeutic approach for various disorders. In the clinical setting, MSCs are being explored in trials of various conditions, including orthopedic injuries, graft versus host disease following bone marrow transplantation, cardiovascular diseases, autoimmune diseases, and liver diseases. Furthermore, genetic modification of MSCs to overexpress antitumor genes has provided prospects for clinical use as anticancer therapy. Here, we highlight the currently reported uses of MSCs in clinical trials and discuss their efficacy as well as their limitations.


Subject(s)
Animals , Humans , Cell Differentiation , Cell Lineage , Cell Movement , Cell Proliferation , Gene Expression Regulation , Mesenchymal Stem Cell Transplantation/adverse effects , Mesenchymal Stem Cells/immunology , Regeneration , Regenerative Medicine/methods , Treatment Outcome
9.
Biol. Res ; 45(3): 269-277, 2012. ilus, graf
Article in English | LILACS | ID: lil-659284

ABSTRACT

Mesenchymal stem cells (MSCs) are now known to display not only stem cell multipotency, but also robust antiinflammatory and regenerative properties. After widespread in-vitro and in-vivo preclinical testing, autologous and allogeneic MSCs have been applied in a range of immune mediated conditions, including graft versus host disease, Crohn's disease, multiple sclerosis, refractory systemic lupus erythematosus and systemic sclerosis. Current data suggests that MSCs may not only replace diseased tissues, but also exert several trophic, regenerative and antiinflammatory effects. While the clinical outcome in case reports and phase I-II trials seems occasionally striking, these limited results point to the need to perform controlled multicenter trials. Future advances from stem cell science can be expected to pinpoint significant MSC subpopulations and/or stem cell markers for improved regenerative or immunoregulatory properties.


Subject(s)
Humans , Autoimmune Diseases/therapy , Mesenchymal Stem Cell Transplantation/methods , Clinical Trials as Topic , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL