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










Publication year range
1.
J Immunol ; 194(9): 4298-308, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25825436

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) can influence the destiny of hematopoietic stem/progenitor cells (HSCs) and exert broadly immunomodulatory effects on immune cells. However, how MSCs regulate the differentiation of regulatory dendritic cells (regDCs) from HSCs remains incompletely understood. In this study, we show that mouse bone marrow-derived Sca-1(+)Lin(-)CD117(-) MSCs can drive HSCs to differentiate into a novel IFN regulatory factor (IRF)8-controlled regDC population (Sca(+) BM-MSC-driven DC [sBM-DCs]) when cocultured without exogenous cytokines. The Notch pathway plays a critical role in the generation of the sBM-DCs by controlling IRF8 expression in an RBP-J-dependent way. We observed a high level of H3K27me3 methylation and a low level of H3K4me3 methylation at the Irf8 promoter during sBM-DC induction. Importantly, infusion of sBM-DCs could alleviate colitis in mice with inflammatory bowel disease by inhibiting lymphocyte proliferation and increasing the numbers of CD4(+)CD25(+) regulatory T cells. Thus, these data infer a possible mechanism for the development of regDCs and further support the role of MSCs in treating immune disorders.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Interferon Regulatory Factors/metabolism , Mesenchymal Stem Cells/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals , Antigens, Ly/metabolism , Cell Differentiation , Cytokines/metabolism , Dendritic Cells/cytology , Disease Models, Animal , Gene Expression , Histones/metabolism , Immunomodulation , Inflammation Mediators/metabolism , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Interferon Regulatory Factors/genetics , Membrane Proteins/metabolism , Mice , Models, Biological , Phenotype , Proto-Oncogene Proteins c-kit/metabolism
2.
Stem Cell Rev Rep ; 10(2): 295-303, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24390934

ABSTRACT

BACKGROUND AND PURPOSE: Intravascular injection of mesenchymal stem cells (MSCs) has been found to cause considerable vascular obstructions which may lead to serious outcomes, particularly after intra-arterial injection. However, the underlying mechanisms have been poorly understood. METHODS: In this study, we fractionated MSCs that had been cultured in monolayer for six passages into small (average diameter = 17.9 µm) and large (average diameter 30.4 µm) populations according to their sizes, and examined their vascular obstructions after intra-internal carotid artery injection in rats and mice in comparison with MSCs derived from 3D spheroids which were uniformly smaller in size (average diameter 12.6 µm). RESULTS: We found that 3D MSCs did not cause detectable infarct in the brain as evidenced by MRI scan and TTC stain, 2D MSCs in small size caused a microinfarct in one of five animals, which was co-localized to the area of entrapped MSCs (labeled with DiI), while 2D MSCs in large size caused much larger infarcts in all five animals, and substantial amounts of DiI-positive MSCs were found in the infarct. Meanwhile, corresponding neurological defects were observed in the animals with stroke. In consistence, injection of 2D MSCs (average diameter 26.5) caused a marked loss of cortical neurons and their axons in Thy1-GFP transgenic mice and the activation of microglia in CX3CR1-GFP transgenic mice in the area with MSC entrapment. CONCLUSIONS: Our results suggest that the size of MSCs is a significant cause of MSC caused vascular obstructions and stroke.


Subject(s)
Mesenchymal Stem Cell Transplantation/adverse effects , Mesenchymal Stem Cells/physiology , Stroke/etiology , Animals , Cell Separation , Cell Size , Cerebral Cortex/pathology , Humans , Injections, Intra-Arterial , Iris/blood supply , Iris/pathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Rats , Rats, Sprague-Dawley , Retina/pathology , Retinal Vessels/pathology , Stroke/pathology
3.
Stem Cells Dev ; 23(9): 978-89, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24341685

ABSTRACT

The therapeutic effect of mesenchymal stem cells (MSCs) in tissue repair/regeneration is substantially dampened by the loss of primitive properties and poor engraftment to target organs. In this study, the multipotency and cell sizes of human MSCs, which had been expanded in monolayer culture for several passages, were dramatically restored after an episode of three-dimensional (3D) spheroid culture. Unlike MSCs derived from monolayer, which caused embolism and blindness, MSCs derived from 3D spheroids did not cause vascular obstructions, after intra-carotid artery infusion in rats. Importantly, intra-carotid infusion of 1 million 3D spheroid MSCs in rats 24 h after middle cerebral artery occlusion and reperfusion resulted in engraftment of the cells into the lesion and significant (over 70%) reduction of infarct size along with restoration of neurologic function. Moreover, the enhanced effect of spheroid MSCs was coincided with significantly increased differentiation of the MSCs into neurons and markedly increased number of endogenous glial fibrillary acidic protein-positive neural progenitors in the peri-infarct boundary zone. However, the similarly administered monolayer MSCs resulted in a modest functional improvement. Our results suggest that 3D MSCs, in combination with intra-carotid delivery, may represent a novel therapeutic approach of MSCs for stroke.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Spheroids, Cellular/metabolism , Stroke/therapy , Animals , Cells, Cultured , Disease Models, Animal , Female , Heterografts , Mesenchymal Stem Cells/cytology , Rats , Rats, Sprague-Dawley , Spheroids, Cellular/cytology , Spheroids, Cellular/transplantation , Stroke/metabolism
4.
J Cell Biochem ; 114(6): 1374-84, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23239100

ABSTRACT

The ERK-MAPK signaling pathway plays a pivotal role during mesenchymal stem cell (MSC) differentiation. Studies have demonstrated that ERK-MAPK promotes adipogenesis and osteogenesis through the phosphorylation of differentiation-associated transcription factors and that it is the only active signaling in all three lineages (adipogenic, chondrogenic, and osteogenic) during MSC differentiation. Recent studies pointed to the significant roles of microRNA-21 (miR-21) during several physiological and pathological processes, especially stem cell fate determination. The miR-21 expression pattern is also correlated with ERK-MAPK activity. Here, we found that miR-21 expression is elevated and associated with an increased differentiation potential in MSCs during adipogenesis and osteogenesis. The overexpression of miR-21 elevated the expression level of the differentiation-associated genes PPARγ and Cbfa-1 during MSC differentiation, whereas miR-21 knockdown reduced the expression level of both genes. The ERK-MAPK signaling pathway activity had an increasing tendency to respond to miR-21 upregulation and a decreasing tendency to respond to miR-21 down-regulation during the first 4 days of adipogenesis and osteogenesis. Our data indicate that miR-21 modulated ERK-MAPK signaling activity by repressing SPRY2 expression, a known regulator of the receptor tyrosine kinase (RTK) signaling pathway, to affect the duration and magnitude of ERK-MAPK activity. The ERK-MAPK signaling pathway was regulated by Sprouty2 (SPRY2) expression via a miR-21-mediated mechanism during MSC differentiation.


Subject(s)
Adipogenesis , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System , Membrane Proteins/metabolism , Mesenchymal Stem Cells/physiology , MicroRNAs/physiology , RNA Interference , 3' Untranslated Regions , Adipose Tissue/cytology , Adult , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Base Sequence , Binding Sites , Cell Separation , Cells, Cultured , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Membrane Proteins/genetics , Osteogenesis , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
5.
Exp Biol Med (Maywood) ; 237(9): 1110-6, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22956625

ABSTRACT

Enhancer of zeste homolog 2 (EZH2) is crucially involved in epigenetic silencing by acting as a histone methyltransferase. Although EZH2 is overexpressed in many solid cancers, the role of EZH2 in B-cell acute lymphoblastic leukemia (B-ALL) remains largely unexplored. In a microarray experiment, we found that EZH2 was significantly upregulated in Nalm-6 cells and this was associated with the silencing of tumor suppressor genes p21, p53 and phosphatase and tensin homolog (PTEN). The abnormal expression of these genes was further confirmed by quantitative realtime polymerase chain reaction and Western blot analysis on Nalm-6 cells. Chromatin immunoprecipitation assay showed that EZH2 and H3K27me3 were both enriched in the promoter region of PTEN and p21 in Nalm-6 cells but not in normal B cells. Functional analysis showed that siRNA-mediated EZH2 knockdown led to decreased proliferation and increased apoptosis of Nalm-6 cells, accompanied by the reactivation of PTEN and p21 expression. Furthermore, we found that EZH2 inhibitor deazaneplanocin A promoted vincristine sulfate-induced apoptosis of Nalm-6 cells. Taken together, our data suggest that EZH2 is overexpressed in B-ALL and promotes the progression of B-ALL by directly mediating the inactivation of tumor suppressor genes p21 and PTEN, and could serve as a potential epigenetic target for B-ALL therapy.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/genetics , PTEN Phosphohydrolase/genetics , Polycomb Repressive Complex 2/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Apoptosis , B-Lymphocytes/metabolism , Cell Line, Transformed , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Enhancer of Zeste Homolog 2 Protein , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Histones/metabolism , Humans , PTEN Phosphohydrolase/metabolism , Polycomb Repressive Complex 2/antagonists & inhibitors , Polycomb Repressive Complex 2/genetics , RNA Interference , RNA, Small Interfering , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation , Vincristine/pharmacology
6.
J Immunol ; 189(3): 1182-92, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22753940

ABSTRACT

Suppression of immune response by mesenchymal stem/stromal cells (MSCs) is well documented. However, their regulatory effects on immune cells, especially regulatory dendritic cells, are not fully understood. We have identified a novel Sca-1(+)Lin(-)CD117(-) MSC population isolated from mouse embryonic fibroblasts (MEF) that suppressed lymphocyte proliferation in vitro. Moreover, the Sca-1(+)Lin(-)CD117(-) MEF-MSCs induced hematopoietic stem/progenitor cells to differentiate into novel regulatory dendritic cells (DCs) (Sca-1(+)Lin(-)CD117(-) MEF-MSC-induced DCs) when cocultured in the absence of exogenous cytokines. Small interfering RNA silencing showed that Sca-1(+)Lin(-)CD117(-) MEF-MSCs induced the generation of Sca-1(+)Lin(-)CD117(-) MEF-MSC-induced DCs via IL-10-activated SOCS3, whose expression was regulated by the JAK-STAT pathway. We observed a high degree of H3K4me3 modification mediated by MLL1 and a relatively low degree of H3K27me3 modification regulated by SUZ12 on the promoter of SOCS3 during SOCS3 activation. Importantly, infusion of Sca-1(+)CD117(-)Lin(-) MEF-MSCs suppressed the inflammatory response by increasing DCs with a regulatory phenotype. Thus, our results shed new light on the role of MSCs in modulating regulatory DC production and support the clinical application of MSCs to reduce the inflammatory response in numerous disease states.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Interleukin-10/physiology , Mesenchymal Stem Cells/immunology , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Cell Line , Coculture Techniques , Embryonic Stem Cells/immunology , Embryonic Stem Cells/metabolism , Fibroblasts/immunology , Fibroblasts/metabolism , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , MAP Kinase Signaling System/immunology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred BALB C , Stromal Cells/immunology , Stromal Cells/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/physiology , Up-Regulation/immunology
7.
Exp Hematol ; 40(9): 761-70, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22634392

ABSTRACT

Recent findings indicate that mesenchymal stem cells (MSCs) may act as a regulator of Th17 cell differentiation, however, the underlying mechanism is still under debate. To investigate the underlying mechanisms of MSCs' regulatory effect, mouse bone marrow-derived MSCs were cocultured with mouse CD4(+)CD25(low)CD44(low)CD62L(high) T cells in vitro, and the proportion of induced Th17 cells, cytokines secretion, and transcription factors expression were examined by flow cytometry, enzyme-linked immunosorbent assay, quantitative reverse transcription polymerase chain reaction, and Western blotting. For the first time, our results showed that bone marrow-derived MSCs were able to inhibit Th17 cell differentiation via interleukin (IL)-10 secretion as the Th17 cell proportion was significantly regained when IL-10 was neutralized, or expression of IL-10 by bone marrow-derived MSCs was downregulated by RNA interference technique. Furthermore, IL-10 may suppress expression of Rorγt, the key transcription factor for Th17 cells, both by activating suppressor of cytokine signaling 3 through signal transducers and activators of transcription 5 phosphorylation, and decreasing signal transducers and activators of transcription 3 binding, which is at the promoter of Rorγt. Thus, our results demonstrate the inhibitory effect of MSCs on Th17 cells differentiation, and suggest increased IL-10 secretion might be the key factor.


Subject(s)
Cell Differentiation , Interleukin-10/metabolism , Mesenchymal Stem Cells/metabolism , Th17 Cells/metabolism , Animals , Blotting, Western , Cells, Cultured , Coculture Techniques , Flow Cytometry , Interleukin-10/genetics , Mice , Mice, Inbred BALB C , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism
8.
Stem Cell Rev Rep ; 8(1): 243-50, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21706142

ABSTRACT

A growing body of preclinical evidence suggests that mesenchymal stem cells (MSCs) are effective for the structural and functional recovery of the infracted heart. Accordingly, clinical trials are underway to determine the benefit of MSC-based therapies. While systemic administration of MSCs is an attractive strategy, and is the route currently used for the administration of MSCs in clinical studies for myocardial infarction, the majority of infused cells do not appear to localize to infracted myocardium in animal studies. Recently, important progress has been made in identifying chemokine receptors critical for the migration and homing of MSCs. Here, we review recent literature regarding mechanisms of MSC homing and recruitment to the ischemic myocardium, and discuss potential influences of low engraftment rates of systemically administered MSCs to the infracted heart tissue on the effects of MSC-based therapies on myocardial infarction.


Subject(s)
Chemokines/physiology , Chemotaxis , Mesenchymal Stem Cells/physiology , Myocardium/cytology , Animals , Bone Marrow Cells/cytology , Chemokine CXCL12/metabolism , Humans , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Myocardial Infarction/therapy , Myocardium/metabolism , Receptors, CXCR4/metabolism , Receptors, Chemokine/metabolism , Receptors, Chemokine/physiology
9.
PLoS One ; 6(6): e20526, 2011.
Article in English | MEDLINE | ID: mdl-21694780

ABSTRACT

BACKGROUND: Mesenchymal stem cells (MSCs) hold great promise for the treatment of difficult diseases. As MSCs represent a rare cell population, ex vivo expansion of MSCs is indispensable to obtain sufficient amounts of cells for therapies and tissue engineering. However, spontaneous differentiation and aging of MSCs occur during expansion and the molecular mechanisms involved have been poorly understood. METHODOLOGY/PRINCIPAL FINDINGS: Human MSCs in early and late passages were examined for their expression of genes involved in osteogenesis to determine their spontaneous differentiation towards osteoblasts in vitro, and of genes involved in self-renewal and proliferation for multipotent differentiation potential. In parallel, promoter DNA methylation and hostone H3 acetylation levels were determined. We found that MSCs underwent aging and spontaneous osteogenic differentiation upon regular culture expansion, with progressive downregulation of TERT and upregulation of osteogenic genes such as Runx2 and ALP. Meanwhile, the expression of genes associated with stem cell self-renewal such as Oct4 and Sox2 declined markedly. Notably, the altered expression of these genes were closely associated with epigenetic dysregulation of histone H3 acetylation in K9 and K14, but not with methylation of CpG islands in the promoter regions of most of these genes. bFGF promoted MSC proliferation and suppressed its spontaneous osteogenic differentiation, with corresponding changes in histone H3 acetylation in TERT, Oct4, Sox2, Runx2 and ALP genes. CONCLUSIONS/SIGNIFICANCE: Our results indicate that histone H3 acetylation, which can be modulated by extrinsic signals, plays a key role in regulating MSC aging and differentiation.


Subject(s)
Cell Differentiation/genetics , Cellular Senescence/genetics , Epigenesis, Genetic , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Acetylation/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Cells, Cultured , Cellular Senescence/drug effects , DNA Methylation/drug effects , Down-Regulation/drug effects , Down-Regulation/genetics , Estrogens/pharmacology , Fibroblast Growth Factor 2/pharmacology , Flow Cytometry , Histones/metabolism , Humans , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/metabolism , Osteogenesis/drug effects , Osteogenesis/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism
10.
Exp Hematol ; 39(6): 608-16, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21288479

ABSTRACT

MicroRNAs (miRNAs) are short non-coding RNAs involved in post-trascriptional regulation of gene expression and diverse biological activities. They are crucial for self-renewal and behavior of embryonic stem cells, but their role in mesenchymal stem cells has been poorly understood. Recently emerging evidence suggests that miRNAs are closely involved in controlling key steps of mesenchymal stem cell differentiation into certain cell lineages. This review focuses on miRNAs identified recently that regulate mesenchymal stem cell differentiation and other activities.


Subject(s)
Cell Differentiation/physiology , Mesenchymal Stem Cells/cytology , MicroRNAs/physiology , Animals , Cell Lineage , Cellular Senescence , Humans , Wound Healing
11.
Exp Hematol ; 39(2): 214-224.e1, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21078360

ABSTRACT

OBJECTIVE: Cyclosporine A (CsA), known as an effective immunosuppressive agent, is widely used in clinical fields. Mesenchymal stem cells may exert immunomodulatory effects on the immune system, but the exact mechanisms underlying them remain controversial. Here we investigated whether human adipose tissue-derived mesenchymal stem cells (AMSCs) facilitate in vitro the immunomodulatory effects of CsA and we explored the molecule mechanisms that may be involved. MATERIALS AND METHODS: Proliferation of T lymphocytes was measured by uptake of (3)H-thymidine. Transcription and production of interleukin-2 and interferon-γ were evaluated by real-time quantitative polymerase chain reaction, reverse transcription polymerase chain reaction, and enzyme-linked immunosorbent assay. Nuclear factor-κB (NF-κB) was assayed by Western blotting and electrophoretic mobility shift assay. Expression of Jagged-1, Jagged-2, and Delta-1 of AMSCs were surveyed by flow cytometric analysis and Western blotting. RESULTS: The combination of moderate-dose AMSCs and low-dose CsA was significantly more powerful than moderate-dose AMSCs or large-dose CsA alone in suppressing transcription and production of interleukin-2 and interferon-γ, activation of NF-κB, and proliferation of T lymphocytes. In addition, AMSCs expressed a high level of Jagged-1, which induced activation of Notch signaling in T lymphocytes, thus reducing NF-κB activity. Anti-Jagged-1 neutralizing antibody and N [N-(3, 5-difluorophenacetyl-L-alanyl)]-S-phenylglycine t-butyl ester could reverse this trend. CONCLUSIONS: Human AMSCs facilitate the immunosuppressive effect of CsA on T lymphocytes through Jagged-1/Notch-related inhibition of NF-κB signaling. The combination of AMSCs and CsA represents a rationale therapeutic approach aimed to prevent adverse effects of CsA while maintaining its adequate immunosuppressive effect. Expression of Jagged-1 on AMSCs may provide an effective mechanism for the immunomodulatory activity of AMSCs via direct cell-cell interaction.


Subject(s)
Adipose Tissue/cytology , Calcium-Binding Proteins/metabolism , Cyclosporine/pharmacology , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mesenchymal Stem Cells/metabolism , NF-kappa B/physiology , Signal Transduction/drug effects , T-Lymphocytes/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Gene Expression Regulation , Humans , Immunosuppressive Agents/pharmacology , Jagged-1 Protein , Protein Binding , Serrate-Jagged Proteins , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
12.
Stem Cells ; 28(5): 905-15, 2010 May.
Article in English | MEDLINE | ID: mdl-20474078

ABSTRACT

Our understanding of the role of bone marrow (BM)-derived cells in cutaneous homeostasis and wound healing had long been limited to the contribution of inflammatory cells. Recent studies, however, suggest that the BM contributes a significant proportion of noninflammatory cells to the skin, which are present primarily in the dermis in fibroblast-like morphology and in the epidermis in a keratinocyte phenotype; and the number of these BM-derived cells increases markedly after wounding. More recently, several studies indicate that mesenchymal stem cells derived from the BM could significantly impact wound healing in diabetic and nondiabetic animals, through cell differentiation and the release of paracrine factors, implying a profound therapeutic potential. This review discusses the most recent understanding of the contribution of BM-derived noninflammatory cells to cutaneous homeostasis and wound healing.


Subject(s)
Bone Marrow Cells/physiology , Bone Marrow Transplantation/methods , Dermatologic Surgical Procedures , Regeneration/physiology , Skin/cytology , Stem Cells/physiology , Wound Healing/physiology , Animals , Bone Marrow Cells/cytology , Bone Marrow Transplantation/trends , Humans , Skin/injuries , Stem Cells/cytology
13.
Blood ; 113(1): 46-57, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18832657

ABSTRACT

Mesenchymal stem cells (MSCs), in addition to their multilineage differentiation, exert immunomodulatory effects on immune cells, even dendritic cells (DCs). However, whether they influence the destiny of full mature DCs (maDCs) remains controversial. Here we report that MSCs vigorously promote proliferation of maDCs, significantly reduce their expression of Ia, CD11c, CD80, CD86, and CD40 while increasing CD11b expression. Interestingly, though these phenotypes clearly suggest their skew to immature status, bacterial lipopolysaccharide (LPS) stimulation could not reverse this trend. Moreover, high endocytosic capacity, low immunogenicity, and strong immunoregulatory function of MSC-treated maDCs (MSC-DCs) were also observed. Furthermore we found that MSCs, partly via cell-cell contact, drive maDCs to differentiate into a novel Jagged-2-dependent regulatory DC population and escape their apoptotic fate. These results further support the role of MSCs in preventing rejection in organ transplantation and treatment of autoimmune disease.


Subject(s)
Cell Communication/immunology , Dendritic Cells/cytology , Dendritic Cells/metabolism , Membrane Proteins/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Animals , Apoptosis/immunology , B7-2 Antigen/metabolism , CD11b Antigen/metabolism , CD11c Antigen/metabolism , CD40 Antigens/metabolism , Cell Differentiation/immunology , Cell Division/immunology , Cells, Cultured , Coculture Techniques , Green Fluorescent Proteins/genetics , Histocompatibility Antigens Class II/metabolism , Immunophenotyping , Inflammation/immunology , Jagged-2 Protein , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic
14.
DNA Cell Biol ; 24(7): 458-63, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16008514

ABSTRACT

Bone marrow-derived mesenchymal stem cells (bMSCs) can differentiate into a number of different cell/tissue types, and also possess immunoregulatory functions. The present study was undertaken to elucidate the exact immunoregulatory effects of allogeneic bMSCs on T- and B-lymphocyte proliferation, activation, and function maturation of BXSB mice, which has been considered as a experimental model for human systemic lupus erythematosus (SLE). We determined that bMSCs from BALB/c mice had inhibitory effects on BXSB mice T-lymphocyte proliferation, but no inhibitory effect on their activation. In addition, they had a significant inhibitory and stimulatory effect on IL-4- and IFN-gamma-producing T cells, respectively. Also, bMSCs had inhibitory effects on the proliferation, activation, and IgG secretion of B lymphocytes. In addition, BALB/c bMSCs had an enhancing effect on CD40 expression and inhibitory effects on CD40 ligand (CD40L) ectopic hyperexpression on B cells from BXSB mice.


Subject(s)
B-Lymphocytes/immunology , Bone Marrow Cells/cytology , Mesenchymal Stem Cells/immunology , T-Lymphocytes/immunology , Transplantation, Homologous/immunology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , CD40 Antigens/metabolism , CD40 Ligand/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Concanavalin A/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Flow Cytometry , Genetic Markers/drug effects , Immunoglobulin G/metabolism , Interferon-gamma/metabolism , Interleukin-4/metabolism , Lipopolysaccharides/pharmacology , Lupus Erythematosus, Systemic/immunology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Mitogens/pharmacology , Spleen/cytology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects
15.
Tissue Eng ; 11(1-2): 110-9, 2005.
Article in English | MEDLINE | ID: mdl-15738666

ABSTRACT

Stem cell plasticity has created great interest because of its potential therapeutic application in degenerative or inherited diseases. Transplantation of bone marrow-derived stem cells was shown to give rise to cells of muscle, liver, nerve, endothelium, epithelium, and so on. But there are still disputes about stem cell plasticity, especially concerning the contribution of bone marrow-derived cells to skin cells. In this study, CM-DiI fluorescence-labeled Flk-(1+) bone marrow mesenchymal stem cells (bMSCs) of BALB/c mice (H-2Kd, white) were transplanted into lethally irradiated C57BL/6 mice (H-2Kb, black). By fluorescence tracing, we found that donor cells could migrate and take residency at the skin, which was confirmed by Y chromosome-specific PCR and Southern blot. The recipient mice grew white hairs about 40 days later and white hairs could spread over the body. Immunochemistry staining and RT-PCR demonstrated that skin tissue within the white hair regions was largely composed of donor-derived H-2Kd cells, including stem cells and committed cells. Furthermore, most skin cells cultured from white hair skin originated from the donor. Thus, our findings provide direct evidence that bone marrow-derived cells can give rise to functional skin cells and regenerate skin tissue. These may have important scientific implications in stem cell biology and transplantation therapy for skin tissue injury.


Subject(s)
Bone Marrow Cells/cytology , Mesenchymal Stem Cells/cytology , Skin/cytology , Stem Cell Transplantation , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cells, Cultured , Female , Graft Survival , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Radiation Injuries, Experimental , Time Factors , Whole-Body Irradiation
16.
Blood ; 105(7): 2733-40, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15591120

ABSTRACT

Overwhelming evidence from leukemia research has shown that the clonal population of neoplastic cells exhibits marked heterogeneity with respect to proliferation and differentiation. There are rare stem cells within the leukemic population that possess extensive proliferation and self-renewal capacity not found in the majority of the leukemic cells. These leukemic stem cells are necessary and sufficient to maintain the leukemia. Interestingly, the BCR/ABL fusion gene, which is present in chronic myelogenous leukemia (CML), was also detected in the endothelial cells of patients with CML, suggesting that CML might originate from hemangioblastic progenitor cells that can give rise to both blood cells and endothelial cells. Here we isolated fetal liver kinase-1-positive (Flk1+) cells carrying the BCR/ABL fusion gene from the bone marrow of 17 Philadelphia chromosome-positive (Ph+) patients with CML and found that these cells could differentiate into malignant blood cells and phenotypically defined endothelial cells at the single-cell level. These findings provide direct evidence for the first time that rearrangement of the BCR/ABL gene might happen at or even before the level of hemangioblastic progenitor cells, thus resulting in detection of the BCR/ABL fusion gene in both blood and endothelial cells.


Subject(s)
Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Adolescent , Adult , Animals , Antigens, CD34/metabolism , Cell Division , Cell Transformation, Neoplastic/immunology , Cells, Cultured , Clone Cells , Endothelial Cells/cytology , Endothelial Cells/physiology , Female , Fusion Proteins, bcr-abl/genetics , Hematopoiesis , Humans , Liver/cytology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasm Transplantation , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
17.
Exp Hematol ; 32(9): 861-7, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15345288

ABSTRACT

OBJECTIVE: To investigate the possibility of flk-1+Sca-1- bone marrow-derived mesenchymal stem cells (bMSCs) to induce stable mixed chimerism and donor-specific graft tolerance. METHODS: Allogeneic flk-1+Sca-1- bMSCs and syngeneic bone marrow (BM) cells were cotransplanted into lethally irradiated (8.5 Gy) recipient mice. FACS was used to analyze the chimerism 150 days later. Donor-type skin transplantation was performed to observe donor-specific immunotolerance in recipient mice. Mixed lymphocyte reaction (MLR) and mitogen proliferative assays were performed to evaluate proliferative response of splenocytes from recipient mice. RESULTS: More than 5% donor-derived CD3+ cells were detected in splenocytes of recipient mice. Long-term survival of donor-type skin grafts was observed. MLR and mitogen proliferative assays showed that recipient mice had low immunoresponse to donor cells but retained normal ConA-induced proliferative response compared with normal mice. CONCLUSION: Our results show for the first time that induction of stable mixed hematopoietic chimerism can be achieved with allogeneic flk-1+Sca-1- bMSC transplantation, which leads to permanent donor-specific immunotolerance in allogeneic host and results in long-term allogeneic skin graft acceptance.


Subject(s)
Antigens, Ly/analysis , Bone Marrow Cells/cytology , Immune Tolerance , Membrane Proteins/analysis , Mesenchymal Stem Cell Transplantation , Skin Transplantation/immunology , Transplantation Chimera , Vascular Endothelial Growth Factor Receptor-2/analysis , Animals , Female , Lymphocyte Culture Test, Mixed , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Transplantation, Homologous
18.
Stem Cells Dev ; 13(3): 263-71, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15186722

ABSTRACT

Mesenchymal stem cells (MSCs) reportedly inhibit the mixed lymphocyte reaction. Whether this effect is mediated by dendritic cells (DCs) is still unknown. In this study, we used an in vitro model to observe the effects of MSCs and their supernatants on the development of monocyte-derived DCs. Phenotypes and the endocytosic ability of harvested DCs were determined by flow cytometry; interleukin 12 (IL-12) secreted by DCs was evaluated by enzyme-linked immunosorbent assay (ELISA); and the antigen-presenting function of DCs was evaluated by MLR. Our results show that MSCs inhibit the up-regulation of CD1a, CD40, CD80, CD86, and HLA-DR during DC differentiation and prevent an increase of CD40, CD86, and CD83 expression during DC maturation. MSCs supernatants had no effect on DCs differentiation, but they inhibited the up-regulation of CD83 during maturation. Both MSCs and their supernatants interfered with endocytosis of DCs, decreased their capacity to secret IL-12 and activate alloreactive T cells. Thus, effects of MSCs on DCs contribute to immunoregulation and development.


Subject(s)
Cell Differentiation/physiology , Dendritic Cells/immunology , Mesenchymal Stem Cells/physiology , Monocytes/immunology , Animals , Antigens, CD/immunology , Cell Proliferation , Cells, Cultured , Dendritic Cells/cytology , HLA-DR Antigens/immunology , Humans , Interleukin-12/immunology , Mesenchymal Stem Cells/cytology , Monocytes/cytology , T-Lymphocytes/immunology
19.
World J Gastroenterol ; 10(7): 950-3, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15052672

ABSTRACT

AIM: To study the synergistic effects of calmodulin (CaM) antagonist O-4-ethoxyl-butyl-berbamine (EBB) and pegylated liposomal doxorubicin (PLD) on hepatoma-22 (H(22)) in vivo. METHODS: Hepatoma model was established in 50 Balb/c mice by inoculating H(22) cells (2.5 x 10(6)) subcutaneously into the right backs of the mice. These mice were divided into 5 groups, and treated with saline only, PLD only, doxorubicin (Dox) only, PLD plus EBB and Dox plus EBB, respectively. In the treatment groups, mice were given 5 intravenous of PLD or Dox on days 0, 3, 6, 9 and 12. The first dosage of PLD or Dox was 4.5 mg/kg, the other 4 injections was 1 mg/kg. EBB (5 mg/kg) was coadministered with PLD or Dox in the corresponding groups. The effect of drugs on the life spans of hepatoma-bearing mice and tumor response to the drugs were recorded. Dox levels in the hepatoma cells were measured by a fluorescence assay. Light microscopy was performed to determine the histopathological changes in the major organs of these tumor-bearing mice. The MTT method was used to analyze the effect of Dox or PLD alone, Dox in combination with EBB, or PLD in combination with EBB on the growth of H(22) cells in an in vitro experiment. RESULTS: EBB (5 mg/kg) significantly augmented the antitumor activity of Dox or PLD, remarkably prolonged the median survival time. The median survival time was 18.2 d for control group, but 89.2 d for PLD+EBB group and 70.1 d for Dox+EBB group, respectively. However, Dox alone did not show any remarkable antitumor activity, and the median survival time was just 29.7 d. Addition of EBB to Dox or PLD significantly increased the level of Dox in H(22) cells in vivo. Moreover, EBB diminished liver toxicity of Dox and PLD. In vitro, EBB reduced the IC50 value of Dox or PLD on H(22) cells from 0.050+/-0.006 mg/L and 0.054+/-0.004 mg/L to 0.012+/-0.002 mg/L and 0.013+/-0.002 mg/L, respectively (P<0.01). CONCLUSION: EBB and liposomization could improve the therapeutic efficacy of Dox in liver cancer, while decreasing its liver toxicity.


Subject(s)
Antineoplastic Agents/pharmacology , Benzylisoquinolines/pharmacology , Calmodulin/antagonists & inhibitors , Carcinoma, Hepatocellular/pathology , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacology , Liver Neoplasms/pathology , Animals , Drug Synergism , Mice , Mice, Inbred BALB C
20.
Mol Cell Neurosci ; 22(1): 49-61, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12595238

ABSTRACT

In vitro, cyclic AMP (cAMP) elevation alters neuronal responsiveness to diffusible growth factors and myelin-associated inhibitory molecules. Here we used an established in vivo model of adult central nervous system injury to investigate the effects of elevated cAMP on neuronal survival and axonal regeneration. We studied the effects of intraocular injections of neurotrophic factors and/or a cAMP analogue (CPT-cAMP) on the regeneration of axotomized rat retinal ganglion cell (RGC) axons into peripheral nerve autografts. Elevation of cAMP alone did not significantly increase RGC survival or the number of regenerating RGCs. Ciliary neurotrophic factor increased RGC viability and axonal regrowth, the latter effect substantially enhanced by coapplication with CPT-cAMP. Under these conditions over 60% of surviving RGCs regenerated their axons. Neurotrophin-4/5 injections also increased RGC viability, but there was reduced long-distance axonal regrowth into grafts, an effect partially ameliorated by cAMP elevation. Thus, cAMP can act cooperatively with appropriate neurotrophic factors to promote axonal regeneration in the injured adult mammalian central nervous system.


Subject(s)
Ciliary Neurotrophic Factor/metabolism , Cyclic AMP/analogs & derivatives , Cyclic AMP/metabolism , Nerve Regeneration/drug effects , Retina/drug effects , Retina/growth & development , Retinal Ganglion Cells/drug effects , Stilbamidines , Up-Regulation/physiology , Animals , Animals, Newborn , Axotomy , Brain Tissue Transplantation , Carbocyanines , Cell Survival/drug effects , Cell Survival/physiology , Ciliary Neurotrophic Factor/pharmacology , Cyclic AMP/pharmacology , Disease Models, Animal , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Growth Cones/drug effects , Growth Cones/metabolism , Growth Cones/ultrastructure , Nerve Growth Factors/metabolism , Nerve Growth Factors/pharmacology , Nerve Regeneration/physiology , Peripheral Nerves/transplantation , Rats , Rats, Sprague-Dawley , Retina/injuries , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Thionucleotides/pharmacology , Tubulin/metabolism , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...