Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Publication year range
1.
Int J Stem Cells ; 15(4): 405-414, 2022 Nov 30.
Article in English | MEDLINE | ID: mdl-35220282

ABSTRACT

Background and Objectives: Chronic inflammation of bone tissue often results in bone defects and hazards to tissue repair and regeneration. Cannabidiol (CBD) is a natural cannabinoid with multiple biological activities, including anti-inflammatory and osteogenic potential. This study aimed to investigate the efficacy and mechanisms of CBD in the promotion of bone marrow mesenchymal stem cells (BMSCs) osteogenic differentiation in the inflammatory microenvironment. Methods and Results: BMSCs isolated from C57BL/6 mice, expressed stem cell characteristic surface markers and presented multidirectional differentiation potential. The CCK-8 assay was applied to evaluate the effects of CBD on BMSCs' vitality, and demonstrating the safety of CBD on BMSCs. Then, BMSCs were stimulated with lipopolysaccharide (LPS) to induce inflammatory microenvironment. We found that CBD intervention down-regulated mRNA expression levels of inflammatory cytokines and promoted cells proliferation in LPS-treated BMSCs, also reversed the protein and mRNA levels downregulation of osteogenic markers caused by LPS treatment. Moreover, CBD intervention activated the cannabinoid receptor 2 (CB2) and the p38 mitogen-activated protein kinase (MAPK) signaling pathway. While AM630, a selective CB2 inhibitor, reduced phosphorylated (p)-p38 levels. In addition, AM630 and SB530689, a selective p38 MAPK inhibitor, attenuated the enhancement of osteogenic markers expression levels by CBD in inflammatory microenvironment, respectively. Conclusions: CBD promoted osteogenic differentiation of BMSCs via the CB2/p38 MAPK signaling pathway in the inflammatory microenvironment.

2.
J Cell Mol Med ; 20(8): 1550-60, 2016 08.
Article in English | MEDLINE | ID: mdl-27028905

ABSTRACT

Mesenchymal stem cells (MSCs) possess an immunoregulatory capacity and are a therapeutic target for many inflammation-related diseases. However, the detailed mechanisms of MSC-mediated immunosuppression remain unclear. In this study, we provide new information to partly explain the molecular mechanisms of immunoregulation by MSCs. Specifically, we found that A20 expression was induced in MSCs by inflammatory cytokines. Knockdown of A20 in MSCs resulted in increased proliferation and reduced adipogenesis, and partly reversed the suppressive effect of MSCs on T cell proliferation in vitro and inhibited tumour growth in vivo. Mechanistic studies indicated that knockdown of A20 in MSCs inhibited activation of the p38 mitogen-activated protein kinase (MAPK) pathway, which potently promoted the production of tumour necrosis factor (TNF)-α and inhibited the production of interleukin (IL)-10. Collectively, these data reveal a crucial role of A20 in regulating the immunomodulatory activities of MSCs by controlling the expression of TNF-α and IL-10 in an inflammatory environment. These findings provide novel insights into the pathogenesis of various inflammatory-associated diseases, and are a new reference for the future development of treatments for such afflictions.


Subject(s)
Mesenchymal Stem Cells/immunology , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Adipogenesis , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Differentiation , Cell Proliferation , Cell Shape , Cytokines/metabolism , Dinoprostone/metabolism , Gene Knockdown Techniques , Immunosuppression Therapy , Inflammation Mediators/metabolism , Intercellular Adhesion Molecule-1/metabolism , MAP Kinase Signaling System , Male , Mesenchymal Stem Cells/cytology , Mice, Inbred C57BL , Nitric Oxide/metabolism , Phenotype , Programmed Cell Death 1 Receptor/metabolism , RNA, Small Interfering/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
3.
Sci Rep ; 6: 22211, 2016 Feb 26.
Article in English | MEDLINE | ID: mdl-26915790

ABSTRACT

Deubiquitinase MYSM1 has been shown to play a critical role in hematopoietic cell differentiation and hematopoietic stem cell (HSC) maintenance. Mesenchymal stem cells (MSCs) are multipotent stromal cells within the bone marrow. MSCs are progenitors to osteoblasts, chondrocytes, adipocytes, and myocytes. Although, MSCs have been extensively studied, the roles of MYSM1 in these cells remain unclear. Here we describe the function of MYSM1 on MSC maintenance and differentiation. In this report, we found that Mysm1-/- mice had a lower bone mass both in long bone and calvaria compared with their control counterpart. Preosteoblasts from Mysm1-/- mice did not show changes in proliferation or osteogenesis when compared to WT mice. Conversely, Mysm1-/- MSCs showed enhanced autonomous differentiation and accelerated adipogenesis. Our results demonstrate that MYSM1 plays a critical role in MSC maintenance and differentiation. This study also underscores the biological significance of deubiquitinase activity in MSC function. Mysm1 may represent a potential therapeutic target for controlling MSC lineage differentiation, and possibly for the treatment of metabolic bone diseases such as osteoporosis.


Subject(s)
Adipocytes/cytology , Adipogenesis/genetics , Endopeptidases/genetics , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Osteogenesis/genetics , 3T3 Cells , Animals , Bone Density/genetics , Bone and Bones/abnormalities , Cell Lineage/genetics , Endopeptidases/deficiency , Mice , Mice, Knockout , Osteoporosis/genetics , Trans-Activators , Ubiquitin-Specific Proteases
4.
Stem Cells Int ; 2016: 1628352, 2016.
Article in English | MEDLINE | ID: mdl-26649045

ABSTRACT

As stromal cells and recently confirmed mesenchymal stem cells, OP9 cells support hematopoiesis stem cell (HSC) differentiation into the B lymphocyte lineage, yet Delta-like-1 (DL1) overexpressing OP9 (OP9DL1) cells promote the development of early T lymphocytes from HSC. However, the immunomodulatory capacity of OP9 or OP9DL1 on mature B and T cell proliferation has not been elucidated. Here, we show that OP9 and OP9DL1 have similar proliferation capacities and immunophenotypes except DL1 expression. Compared with OP9, OP9DL1 displayed more osteogenesis and less adipogenesis when cultured in the respective induction media. Both OP9 and OP9DL1 inhibited mature B and T cell proliferation. Furthermore, OP9 showed stronger inhibition on B cell proliferation and OP9DL1 exhibited stronger inhibition on T cell proliferation. With stimulation, both OP9 and OP9DL1 showed increased nitrate oxide (NO) production. The NO levels of OP9 were higher than that of OP9DL1 when stimulated with TNFα/IFNγ or LPS/IL4. Taken together, our study reveals a previously unrecognized role of OP9 and OP9DL1 in mature B and T cell proliferation. DL1 overexpression alone changed the properties of OP9 cells in addition to their role in early B cell development.

5.
Age (Dordr) ; 37(2): 16, 2015.
Article in English | MEDLINE | ID: mdl-25693923

ABSTRACT

Mesenchymal stem cells (MSCs) are known to have many notable features, especially their multiple differentiation ability and immunoregulatory capacity. MSCs are important stem cells in the bone marrow (BM), and their characteristics are affected by the BM microenvironment. However, effects of the BM microenvironment on the properties of MSCs are not well understood. In this study, we found that BM from aged mice decreased MSC colony formation. Flow cytometry data showed that the proportion of B220(+) cells in BM from aged mice was significantly lower than that in BM from young mice, while the proportion of CD11b(+), CD3(+), Gr-1(+), or F4/80(+) cells are on the contrary. CD11b(+), B220(+), and Ter119(+) cells from aged mice were not the subsets that decreased MSC colony formation. We further demonstrated that both BM from aged mice and young mice exhibited similar effects on the proliferation of murine MSC cell line C3H10T1/2. However, when cocultured with BM from aged mice, C3H10T1/2 showed slower migration ability. In addition, we found that phosphorylation of JNK (c-Jun N-terminal kinases) in C3H10T1/2 cocultured with BM from aged mice was lower than that in C3H10T1/2 cocultured with BM from young mice. Collectively, our data revealed that BM from aged mice could decrease the migration of MSCs from their niche through regulating the JNK pathway.


Subject(s)
Aging/physiology , Bone Marrow Cells/cytology , Cell Movement/physiology , Mesenchymal Stem Cells/cytology , Stem Cell Niche/physiology , Animals , Bone Marrow Cells/physiology , Cell Differentiation , Cell Migration Assays , Coculture Techniques , Female , Flow Cytometry , JNK Mitogen-Activated Protein Kinases/analysis , Male , Mice, Inbred C57BL , Phosphorylation
6.
PLoS One ; 9(12): e115720, 2014.
Article in English | MEDLINE | ID: mdl-25549354

ABSTRACT

The clinical acute graft-versus-host disease (GvHD)-therapy of mesenchymal stem cells (MSCs) is not as satisfactory as expected. Secondary lymphoid organs (SLOs) are the major niches serve to initiate immune responses or induce tolerance. Our previous study showed that CCR7 guide murine MSC line C3H10T1/2 migrating to SLOs. In this study, CCR7 gene was engineered into murine MSCs by lentivirus transfection system (MSCs/CCR7). The immunomodulatory mechanism of MSCs/CCR7 was further investigated. Provoked by inflammatory cytokines, MSCs/CCR7 increased the secretion of nitric oxide and calmed down the T cell immune response in vitro. Immunofluorescent staining results showed that transfused MSCs/CCR7 can migrate to and relocate at the appropriate T cell-rich zones within SLOs in vivo. MSCs/CCR7 displayed enhanced effect in prolonging the survival and alleviating the clinical scores of the GvHD mice than normal MSCs. Owing to the critical relocation sites, MSCs/CCR7 co-infusion potently made the T cells in SLOs more naïve like, thus control T cells trafficking from SLOs to the target organs. Through spoiling the fourth supplemental Billingham's tenet, MSCs/CCR7 potently inhibited the development of GvHD. The study here provides a novel therapeutic strategy of MSCs/CCR7 infusion at a low dosage to give potent immunomodulatory effect for clinical immune disease therapy.


Subject(s)
Graft vs Host Disease/prevention & control , Mesenchymal Stem Cells/physiology , Receptors, CCR7/physiology , Animals , Cell Movement , Immunomodulation , Male , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/metabolism , Receptors, CCR7/genetics , Receptors, CCR7/metabolism
7.
PLoS One ; 9(5): e97256, 2014.
Article in English | MEDLINE | ID: mdl-24826993

ABSTRACT

Mesenchymal stem cells (MSCs) have been shown to be highly immunosuppressive and have been employed to treat various immune disorders. However, the mechanisms underlying the immunosuppressive capacity of MSCs are not fully understood. We found the suppressor of cytokine signaling 1 (SOCS1) was induced in MSCs treated with inflammatory cytokines. Knockdown of SOCS1 did not bring much difference on the proliferation and differentiation properties of MSCs. However, MSCs with SOCS1 knockdown exhibited enhanced immunosuppressive capacity, showing as inhibiting T cell proliferation at extremely low ratio (MSC to T) in vitro, significantly promoting tumor growth and inhibiting delayed-type hypersensitivity response in vivo. We further demonstrated that SOCS1 inhibited the immunosuppressive capacity of MSCs by reducing inducible nitric oxide synthase (iNOS) expression. Additionally, we found the significantly lower SOCS1 expression and higher nitric oxide (NO) production in MSCs isolated from synovial fluid of rheumatoid arthritis patients. Collectively, our data revealed a novel role of SOCS1 in regulating the immune modulatory activities of MSCs.


Subject(s)
Immunologic Factors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Nitric Oxide/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Cell Differentiation/genetics , Cell Line , Cell Proliferation/genetics , Cytokines/genetics , Cytokines/metabolism , Humans , Immunologic Factors/genetics , Immunosuppression Therapy/methods , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Suppressor of Cytokine Signaling Proteins/genetics , T-Lymphocytes/metabolism
8.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 21(6): 1563-7, 2013 Dec.
Article in Chinese | MEDLINE | ID: mdl-24370049

ABSTRACT

This study was purposed to establish a convenient and efficient method for isolating and culturing mouse bone marrow mesenchymal stem cells (MSC). The femurs and tibias of mouse were taken under sterile condition. MSC were isolated and cultured with flushing- out bone marrow or collagenase-digested bone fragment or bone marrow plus bone fragment. MSC colony number and size were compared. Immunophenotype and differentiation ability were tested to identify MSC. The results showed that colonies from bone marrow plus bone fragment group came out earliest and the colony number was 20 ± 4 at day 4; there were 11.5 ± 2.5 colonies in collagenase-digested bone fragment group and 9.5 ± 1.5 in flushing- out bone marrow group. The total cell yields of MSC after passaging showed best in bone marrow plus bone fragment group. Flow cytometry data showed the cultured cells expressed Sca-1, CD44 and CD29, not expressed pan-leukocyte surface marker CD45 and endothelial cell marker CD31. The isolated and cultured MSC could differentiate into osteoblast at the osteogenic differentiation condition, or adipocyte at adipogenic differentiation condition. It is concluded that the method of bone marrow plus bone fragment is convenient and efficient for isolating and culturing MSC.


Subject(s)
Bone Marrow Cells/cytology , Cell Culture Techniques/methods , Cell Separation/methods , Mesenchymal Stem Cells/cytology , Animals , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...