Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Acta Pharmacol Sin ; 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38914677

ABSTRACT

Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have obvious advantages over MSC therapy. But the strong procoagulant properties of MSC-EVs pose a potential risk of thromboembolism, an issue that remains insufficiently explored. In this study, we systematically investigated the procoagulant activity of large EVs derived from human umbilical cord MSCs (UC-EVs) both in vitro and in vivo. UC-EVs were isolated from cell culture supernatants. Mice were injected with UC-EVs (0.125, 0.25, 0.5, 1, 2, 4 µg/g body weight) in 100 µL PBS via the tail vein. Behavior and mortality were monitored for 30 min after injection. We showed that these UC-EVs activated coagulation in a dose- and tissue factor-dependent manner. UC-EVs-induced coagulation in vitro could be inhibited by addition of tissue factor pathway inhibitor. Notably, intravenous administration of high doses of the UC-EVs (1 µg/g body weight or higher) led to rapid mortality due to multiple thrombus formations in lung tissue, platelets, and fibrinogen depletion, and prolonged prothrombin and activated partial thromboplastin times. Importantly, we demonstrated that pulmonary thromboembolism induced by the UC-EVs could be prevented by either reducing the infusion rate or by pre-injection of heparin, a known anticoagulant. In conclusion, this study elucidates the procoagulant characteristics and mechanisms of large UC-EVs, details the associated coagulation risk during intravenous delivery, sets a safe upper limit for intravenous dose, and offers effective strategies to prevent such mortal risks when high doses of large UC-EVs are needed for optimal therapeutic effects, with implications for the development and application of large UC-EV-based as well as other MSC-EV-based therapies.

2.
Acta Pharmacol Sin ; 45(1): 180-192, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37644132

ABSTRACT

Adhesion molecules play essential roles in the homeostatic regulation and malignant transformation of hematopoietic cells. The dysregulated expression of adhesion molecules in leukemic cells accelerates disease progression and the development of drug resistance. Thus, targeting adhesion molecules represents an attractive anti-leukemic therapeutic strategy. In this study, we investigated the prognostic role and functional significance of cytohesin-1 (CYTH1) in acute myeloid leukemia (AML). Analysis of AML patient data from the GEPIA and BloodSpot databases revealed that CYTH1 was significantly overexpressed in AML and independently correlated with prognosis. Functional assays using AML cell lines and an AML xenograft mouse model confirmed that CYTH1 depletion significantly inhibited the adhesion, migration, homing, and engraftment of leukemic cells, delaying disease progression and prolonging animal survival. The CYTH1 inhibitor SecinH3 exerted in vitro and in vivo anti-leukemic effects by disrupting leukemic adhesion and survival programs. In line with the CYTH1 knockdown results, targeting CYTH1 by SecinH3 suppressed integrin-associated adhesion signaling by reducing ITGB2 expression. SecinH3 treatment efficiently induced the apoptosis and inhibited the growth of a panel of AML cell lines (MOLM-13, MV4-11 and THP-1) with mixed-lineage leukemia gene rearrangement, partly by reducing the expression of the anti-apoptotic protein MCL1. Moreover, we showed that SecinH3 synergized with the BCL2-selective inhibitor ABT-199 (venetoclax) to inhibit the proliferation and promote the apoptosis of ABT-199-resistant leukemic cells. Taken together, our results not only shed light on the role of CYTH1 in cell-adhesion-mediated leukemogenesis but also propose a novel combination treatment strategy for AML.


Subject(s)
Antineoplastic Agents , Leukemia, Myeloid, Acute , Humans , Mice , Animals , Leukemia, Myeloid, Acute/drug therapy , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Adhesion Molecules , Disease Progression , Cell Line, Tumor
4.
Acta Pharmacol Sin ; 43(11): 2885-2894, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35365781

ABSTRACT

Few therapies can reverse the proangiogenic activity of senescent mesenchymal stromal/stem cells (MSCs). In this study, we investigated the effects of rapamycin on the proangiogenic ability of senescent human umbilical cord MSCs (UCMSCs). An in vitro replicative senescent cell model was established in cultured UCMSCs. We found that late passage (P25 or later) UCMSCs (LP-UCMSCs) exhibited impaired proangiogenic abilities. Treatment of P25 UCMSCs with rapamycin (900 nM) reversed the senescent phenotype and notably enhanced the proangiogenic activity of senescent UCMSCs. In a nude mouse model of hindlimb ischemia, intramuscular injection of rapamycin-treated P25 UCMSCs into the ischemic limb significantly promoted neovascularization and ischemic limb salvage. We further analyzed the changes in the expression of angiogenesis-associated genes in rapamycin-primed MSCs and found higher expression of several genes related to angiogenesis, such as VEGFR2 and CTGF/CCN2, in primed cells than in unprimed MSCs. Taken together, our data demonstrate that rapamycin is a potential drug to restore the proangiogenic activity of senescent MSCs, which is of importance in treating ischemic diseases and tissue engineering.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Humans , Mice , Animals , Limb Salvage , Hindlimb , Neovascularization, Physiologic , Sirolimus/pharmacology , Sirolimus/therapeutic use , Ischemia/therapy , Ischemia/metabolism , Mesenchymal Stem Cells/metabolism , Neovascularization, Pathologic/metabolism , Mice, Nude , Cells, Cultured
5.
Curr Med Sci ; 38(1): 35-42, 2018 Feb.
Article in English | MEDLINE | ID: mdl-30074149

ABSTRACT

Low-dose cytarabine combined with differentiating or DNA hypomethylating agents, such as vitamin D compounds, is a potential regimen to treat acute myeloid leukemia (AML) patients who are unfit for high-intensity chemotherapy. The present study aimed to determine which subset of AML would be most responsive to low-dose cytarabine with the differentiating agent 1,25-dihydroxyvitamin D3 (1,25-D3). Here, firstly, cBioPortal database was used and we found out that vitamin D receptor (VDR) was highly expressed in acute monocytic leukemia (M5) and high VDR expression was associated with a poor survival of AML patients. Then, we confirmed that 1,25-D3 at clinical available concentration could induce more significant differentiation in acute monocytic leukemia cell lines (U937, MOLM-13, THP-1) and blasts from M5 patients than in non-monocytic cell lines (KGla and K562) and blasts from M2 patient. Finally, it was shown that the combination of 1,25-D3 and low-dose cytarabine further increased the differentiating rate, growth inhibition and G0/G1 arrest, while mild changes were found in the apoptosis in acute monocytic leukemia cell lines. Our study demonstrates that the enhanced response of acute monocytic leukemia cells to low-dose cytarabine by 1,25-D3 might indicate a novel therapeutic direction for patients with acute monocytic leukemia, especially for elderly and frail ones.


Subject(s)
24,25-Dihydroxyvitamin D 3/pharmacology , Antineoplastic Agents/pharmacology , Cytarabine/pharmacology , Leukemia, Monocytic, Acute/drug therapy , Vitamins/pharmacology , 24,25-Dihydroxyvitamin D 3/administration & dosage , 24,25-Dihydroxyvitamin D 3/therapeutic use , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cytarabine/administration & dosage , Cytarabine/therapeutic use , Drug Synergism , Humans , Vitamins/administration & dosage , Vitamins/therapeutic use
6.
Acta Pharmacol Sin ; 38(11): 1475-1485, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28836580

ABSTRACT

The intercellular communication between leukemia cells and bone marrow mesenchymal stem cells (BM-MSCs) plays more important role in chronic myeloid leukemia (CML) than we previously understood. Recently, we found that microvesicles released from human leukemia cell line K562 (K562-MVs) containing BCR-ABL1 mRNA malignantly transformed normal hematopoietic transplants. Here, we investigated whether K562-MVs contribute to the transformation of human bone marrow mesenchymal stem cells (BM-MSCs). We showed that K562-MVs could be integrated into co-cultured normal BM-MSCs and dose-dependently enhanced the proliferation of BM-MSCs. Meanwhile, K562-MVs (400 ng/mL) significantly increased the expression of BCR-ABL1 in these BM-MSCs, accompanied by the enhanced secretion of TGF-ß1. These BM-MSCs in turn could trigger the TGF-ß1-dependent proliferation of K562 cells. Moreover, we confirmed the presence of BCR-ABL1 in circulating MVs from 11 CML patients. Compared to the normal BM-MSCs, the BM-MSCs from CML patients more effectively increased the BCR-ABL1 expression and TGF-ß1 secretion in K562 cells as well as the proliferation of K562 cells. Our findings enrich the mechanisms involved in the interaction between leukemia cells and BM-MSCs and provide novel ways to monitor minimal residual disease and worthwhile approaches to treat CML.


Subject(s)
Bone Marrow Cells/metabolism , Cell Communication , Cell Transformation, Neoplastic/genetics , Cell-Derived Microparticles/genetics , Fusion Proteins, bcr-abl/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mesenchymal Stem Cells/metabolism , Adolescent , Adult , Bone Marrow Cells/pathology , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cell-Derived Microparticles/pathology , Cell-Free Nucleic Acids/blood , Cell-Free Nucleic Acids/genetics , Coculture Techniques , Female , Fusion Proteins, bcr-abl/blood , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Mesenchymal Stem Cells/pathology , Middle Aged , RNA, Messenger/blood , RNA, Messenger/genetics , Time Factors , Transforming Growth Factor beta1/metabolism , Tumor Microenvironment , Young Adult
7.
Oncotarget ; 7(52): 86075-86086, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27863383

ABSTRACT

Accumulating evidences have shown that adipokines secreted from adipocytes contributes to tumor development, especially leptin. However, underlying mechanisms remain unclear. This study aims to explore the effect of leptin on development and chemoresistance in multiple myeloma cells and the potential mechanism. Analysis of levels of adipokines including leptin and adiponectin in 28 multiple myeloma patients identified significantly higher leptin compared with 28 normal controls(P < 0.05), and leptin level was positively correlated with clinical stage, IgG, ER, and ß2MG. Next, by using co-culture system of myeloma and adipocytes, and pharmacologic enhancement of leptin, we found that increased growth of myeloma cells and reduced toxicity of bortezomib were best observed at 50 ng/ml of leptin, along with increased expression of cyclinD1, Bcl-2 and decreased caspase-3 expression. We also found that phosphorylated AKT and STAT3 but not the proteins expression reached peak after 1h and 6h treatment of leptin, respectively. By using AG490, an agent blocking the phosphorylation of AKT and ERK, the proliferation of myeloma cells was inhibited, as well as the phosphorylation of AKT and STAT3, even adding leptin. Taken together, our study demonstrated that up-regulated leptin could stimulate proliferation of myeloma and reduce the anti-tumor effect of chemotherapy possibly via activating AKT and STAT3 pathways, and leptin might be one of the potential therapeutic targets for treating myeloma.


Subject(s)
Adipocytes/metabolism , Leptin/physiology , Multiple Myeloma/drug therapy , Adult , Aged , Cell Proliferation , Drug Resistance, Neoplasm , Female , Humans , Leptin/blood , Male , Middle Aged , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Proto-Oncogene Proteins c-akt/physiology , STAT3 Transcription Factor/physiology , Signal Transduction , Tyrphostins/pharmacology
8.
J Huazhong Univ Sci Technolog Med Sci ; 35(3): 343-349, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26072071

ABSTRACT

This study examined the mechanism of the inhibitory effect of parthenolide (PTL) on the activity of NF-κB in multiple myeloma (MM). Human multiple myeloma cell line RPMI 8226 cells were treated with or without different concentrations of PTL for various time periods, and then MTT assay was used to detect cell proliferation. Cell cycle and apoptosis were flow cytometrically detected. The level of protein ubiquitination was determined by using immunoprecipitation. Western blotting was employed to measure the level of total protein ubiquitination, the expression of IκB-α in cell plasma and the content of p65 in nucleus. The content of p65 in nucleus before and after PTL treatment was also examined with immunofluorescence. Exposure of RPMI 8226 cells to PTL attenuated the level of ubiquitinated Nemo, increased the expression of IκB-α and reduced the level of p65 in nucleus, finally leading to the decrease of the activity of NF-κB. PTL inhibited cell proliferation, induced apoptosis and blocked cell cycle. Furthermore, the levels of ubiquitinated tumor necrosis factor receptor-associated factor 6 (TRAF6) and total proteins were decreased after PTL treatment. By using Autodock software package, we predicted that PTL could bind to TRAF6 directly and tightly. Taken together, our findings suggest that PTL inhibits the activation of NF-κB signaling pathway via directly binding with TRAF6, thereby suppressing MM cell proliferation and inducing apoptosis.


Subject(s)
Multiple Myeloma/metabolism , NF-kappa B/antagonists & inhibitors , Sesquiterpenes/pharmacology , TNF Receptor-Associated Factor 6/metabolism , Apoptosis , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Multiple Myeloma/drug therapy , NF-kappa B/blood , Transcription Factor RelA/metabolism , Ubiquitination/drug effects
9.
Acta Pharmacol Sin ; 35(3): 381-93, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24374813

ABSTRACT

AIM: To investigate the effects of serum deprivation (SD) on microvesicles (MVs) secreted from human myeloma cells and the implications for disease progression. METHODS: RPMI 8226, U266, and KM3 human myeloma cells were incubated in medium containing 10% (non-SD) or 1% fetal bovine serum (SD) and MVs were isolated. The levels and size distribution of MVs were analyzed with flow cytometry. The protein profiles of MVs were studied using 2D SDS-PAGE, MALDI-TOF-MS, and Western blotting. NF-κB activation was analyzed using EMSA. Angiogenesis was examined in Eahy926 endothelial cells. RESULTS: Exposure of RPMI 8226 cells to SD for 24 h did not alter the number of apoptotic cells. However, SD increased the number of MVs from RPMI 8226, U266, and KM3 cells to 2.5-, 4.3-, and 3.8-fold, respectively. The size distribution of SD MVs was also significantly different from that of non-SD MVs. Three proteins ZNF224, SARM, and COBL in SD MVs were found to be up-regulated, which were involved in cell cycle regulation, signal transduction and metabolism, respectively. Co-culture of SD MVs and RPMI 8226 cells increased NF-κB activation in the target RPMI 8226 cells. Furthermore, SD MVs from RPMI 8226 cells significantly increased the microtubule formation capacity of Eahy926 endothelial cells compared with non-SD MVs. CONCLUSION: SD elevates the levels of microvesicles with different size distribution and selectively enriched proteins in human myeloma cells in vitro. The selectively enriched proteins, especially ZNF224, may play key roles in regulation of myeloma cells, allowing better adaptation to SD.


Subject(s)
Autocrine Communication , Cell-Derived Microparticles/metabolism , Culture Media, Serum-Free/metabolism , Multiple Myeloma/metabolism , Myeloma Proteins/metabolism , Paracrine Communication , Apoptosis , Blotting, Western , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Proliferation , Cell-Derived Microparticles/pathology , Coculture Techniques , Electrophoresis, Gel, Two-Dimensional , Endothelial Cells/metabolism , Flow Cytometry , Humans , Multiple Myeloma/blood , Multiple Myeloma/pathology , NF-kappa B/metabolism , Neovascularization, Physiologic , Proteomics/methods , Repressor Proteins/metabolism , Signal Transduction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Time Factors
10.
Acta Pharmacol Sin ; 35(2): 230-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24374814

ABSTRACT

AIM: To investigate whether human multiple myeloma (MM) cells secrete microvesicles (MVs) and whether the MVs secreted from MM cells (MM-MVs) promote angiogenesis. METHODS: RPMI8226 human MM cells and EA.hy926 human umbilical vein cells were used. MVs isolated from RPMI8226 cells were characterized under laser confocal microscopy, electron microscopy and with flow cytometry. The fusion of MM-MVs and EA.hy926 cells was studied under confocal microscopy, and the transfer of CD138 to EA.hy926 cells was demonstrated with flow cytometry. The proliferation, invasion and tube formation of EA.hy926 cells in vitro were evaluated using MTT, transwell migration and tube formation assays, respectively. The vasculization of EA.hy926 cells in vivo was studied using Matrigel plug assay. The expression of IL-6 and VEGF was analyzed with PCR and ELISA. RESULTS: MM-MVs from the RPMI 8226 cells had the characteristic cup-shape with diameter of 100-1000 nm. Most of the MM-MVs expressed phosphatidylserine and the myeloma cell marker CD138, confirming that they were derived from myeloma cells. After added to EA.hy926 cells, the MM-MVs transferred CD138 to the endothelial cells and significantly stimulated the endothelial cells to proliferate, invade, secrete IL-6 and VEGF, two key angiogenic factors of myeloma, and form tubes in vitro and in vivo. CONCLUSION: Our results confirm the presence of MVs in MM cells and support the idea that MM-MVs are newfound mediators for myeloma angiogenesis and may serve as a therapeutic target to treat MM.


Subject(s)
Multiple Myeloma/pathology , Neovascularization, Pathologic/pathology , Secretory Vesicles/pathology , Cell Line, Tumor , Humans , Umbilical Veins/pathology
11.
Cancer Invest ; 30(9): 646-56, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23020627

ABSTRACT

The immunosuppressive functions of multipotent mesenchymal stromal cells (MSCs) may give cancer cells a survival advantage. This study tests the hypothesis that MSCs protect leukemia cells from immune clearance. Our results demonstrate that MSCs are capable of inhibiting peripheral blood mononuclear cells (PBMNCs) proliferation and their migration toward leukemic cells by the reduction of CCL5 and CXCL12. In addition, we find that MSCs can inhibit the cytolytic functions of NK-cells and CTLs. TGF-ß1 secreted by MSCs is responsible for impaired CTLs and NK function by down-modulating surface NKG2D expression. These inhibitory functions of MSCs have negative effects on the CTLs or NK-mediated graft-versus-leukemia (GVL), particularly in the allogeneic hematopoietic stem cells transplantation setting.


Subject(s)
Killer Cells, Natural/immunology , Mesenchymal Stem Cells/immunology , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , T-Lymphocytes, Cytotoxic/immunology , Transforming Growth Factor beta1/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Chemokine CCL5/biosynthesis , Chemokine CXCL12/biosynthesis , Hematopoietic Stem Cell Transplantation , Humans , Immune Evasion , Killer Cells, Natural/metabolism , Leukemia/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , RNA Interference , RNA, Small Interfering , T-Lymphocytes, Cytotoxic/metabolism , Transforming Growth Factor beta1/genetics
12.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 20(2): 258-61, 2012 Apr.
Article in Chinese | MEDLINE | ID: mdl-22541077

ABSTRACT

This study was aimed to observe the effects of siRNA on Livin expression and function in K562 cells. Livin siRNA were designed and synthesized, then were transfected into K562 cells by using AMAXA nucle transfactor. Expressions of Livin mRNA and protein in transfected K562 cells was detected by RT-PCR and Western blot respectively. Non-transfected cells were used as control. The enhanced green fluorescent protein plasmid was used as positive control and the transfection efficiency was detected by flow cytometry. Cell apoptosis was measured by flow cytometry with Annexin V-FITC/PI double staining. The results showed that the transfection efficiency of electroporation method was about 50. The synthesized siRNA inhibited livin expression at both mRNA and protein levels. The rate of K562 cell apoptosis increased from (9.63 ± 0.89) in control group to (12.07 ± 1.39) and (27.41 ± 2.30) at 24 h and 48 h after transfection, respectively (P < 0.05). It is concluded that the siRNA can inhibit anti-apoptosis of livin gene via down-regulating livin gene expression, which may provide the new method for anti-leukemia study.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Apoptosis/genetics , Inhibitor of Apoptosis Proteins/genetics , Neoplasm Proteins/genetics , RNA, Small Interfering , Gene Expression Regulation, Leukemic , Humans , K562 Cells , RNA, Small Interfering/genetics
13.
Cancer Lett ; 317(2): 136-43, 2012 Apr 28.
Article in English | MEDLINE | ID: mdl-22240014

ABSTRACT

Mesenchymal stem cells (MSCs) have emerged as excellent candidates for clinical application because of their capabilities of immunomodulatory and supporting hematopoiesis. Nevertheless, it is unclear whether the characteristics of MSCs are altered in diseased states. In this study, we obtained and expanded MSCs from bone marrow of patients with myelodysplastic syndromes (MDS). Our results showed that MSCs derived from MDS (MDS-MSCs) were similar to normal adult bone marrow derived MSCs in morphology, growth property, surface epitopes, and differentiation ability in vitro. In addition, MDS-MSCs had normal karyotype and ultrastructure. However, MDS-MSCs appeared to be impaired in immunomodulatory and supporting hematopoiesis function. Although, MDS-MSCs could express hematopoietic cytokines and support hematopoiesis in long term culture, Real time quantitative polymerase chain reaction showed that the expression of hematopoietic cytokines in MDS-MSCs was much lower than that of normal adult derived MSCs. Moreover, MDS-MSCs showed reduced hematopoiesis support function, as compared to their normal counterparts. Lastly, the capacity of MDS-MSCs to inhibit T lymphocyte activation and proliferation was impaired in vitro. These results indicate that MDS-MSCs have impaired immunomodulatory and hematopoiesis support functions, suggesting their critical role in the pathogenesis of MDS.


Subject(s)
Bone Marrow Cells/immunology , Hematopoiesis/immunology , Mesenchymal Stem Cells/immunology , Myelodysplastic Syndromes/immunology , Adult , Aged , Bone Marrow Cells/metabolism , Bone Marrow Cells/ultrastructure , Cell Proliferation , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Female , Flow Cytometry , Gene Expression , Hematopoiesis/genetics , Humans , Immunophenotyping , Karyotype , Lymphocyte Activation/immunology , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/ultrastructure , Microscopy, Electron, Transmission , Middle Aged , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Young Adult
14.
Eur J Cancer ; 48(12): 1884-95, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22129888

ABSTRACT

Dendritic cells (DCs) are specialised antigen-presenting cells that play crucial roles in the initiation and regulation of immune responses. Recently, mesenchymal stem cells (MSCs) have gained further interest after demonstration of immunomodulatory effects on complicated interactions between T cells and even DCs. However, the mechanisms underlying these immunoregulatory effects of MSCs induced DCs are poorly understood. In addition, it is unclear whether the immunoregulatory functions of MSCs are altered in disease states. In this study, we showed that chronic myeloid leukaemia (CML) patients bone marrow derived MSCs (CML-MSC) could differentiate mature DCs (mDCs) into a distinct regulatory DC population, they had lower expression of CD40, CD80, CD83 and CD86. Similar to immature DCs (imDCs), CML-MSC induced DCs (CML-MSC-DCs) displayed powerful phagocytic capacity. Moreover, CML-MSC-DCs had the capacity to induce T cell anergy, another capacity of regulatory DCs. CML-MSC-DCs could inhibit the proliferation of T cells not only through TGF-ß1, but also by inducing the production of Treg cells or T-cell anergy. At last, CML-MSC-DCs could efficiently induce more CD4+CD25+Foxp3+Tregs from naive CD4+CD25-Foxp3-T cells than that of normal-MSC-DCs in vitro. CML-MSC-DCs derived TGF-ß1 was largely responsible for the increase in CD4+CD25+Foxp3+Tregs based on knockdown studies. The immunoregulatory effects of DCs induced by CML-MSCs enhance the potential use of autologous MSCs in cell therapy.


Subject(s)
Bone Marrow/immunology , Dendritic Cells/immunology , Immunomodulation , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Mesenchymal Stem Cells/immunology , Adult , Cells, Cultured , Coculture Techniques , Female , Humans , Male , Middle Aged , T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/immunology , Young Adult
15.
Immunol Invest ; 41(2): 183-98, 2012.
Article in English | MEDLINE | ID: mdl-21936678

ABSTRACT

Mesenchymal stem cells (MSCs) provide an excellent model for development of stem cell therapeutics, and their potential treatment in the immunopathogenic diseases have gained further interest after demonstration of immunomodulatory effects on complicated interactions between T cells and even dendritic cells (DCs). However, the mechanisms underlying these immunoregulatory effects of MSCs are poorly understood. In this study, we show that bone marrow derived MSCs can differentiate mature DCs (mDCs) into a distinct regulatory DC population. Compared with mDCs, they have lower expression of CD1a, CD80, CD86 and CD40, but higher expression of CD11b. MSCs induced DCs (MSC-DCs) can hardly stimulate T-cell proliferation even when MSC-DCs are stimulated by LPS. In addition, high endocytosic capacity, low immunogenicity, and strong immunoregulatory function of MSC-DCs are also observed. Moreover, MSC-DCs can efficiently generate CD4+CD25+Foxp3+ Treg cells from CD4+CD25-Foxp3-T cells. The inhibitory function of MSC-DCs is mediated not only through TGF-ß1, but also by inducing the production of Treg cells or T-cell anergy. These results demonstrate that the immunomodulatory effects of regulatory DCs induced by MSCs provide efficacious treatment for immunopathogenic diseases.


Subject(s)
Bone Marrow/immunology , Dendritic Cells/immunology , Immunomodulation , Mesenchymal Stem Cells/immunology , Adult , Antigens, CD/metabolism , Cell Communication , Cell Differentiation , Cells, Cultured , Endocytosis , Forkhead Transcription Factors/metabolism , Humans , Middle Aged , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta1/immunology , Young Adult
17.
Acta Pharmacol Sin ; 31(8): 999-1004, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20644548

ABSTRACT

AIM: To investigate the pharmacokinetics of imatinib in Chinese chronic myelogenous leukemia (CML) patients. METHODS: Fourty-six naïve Chinese CML patients treated with imatinib (400 and 600 mg daily, n=36 and 10, respectively) were recruited. The correlations of imatinib (400 mg) trough plasma concentrations (C(mins)) with the patients' characteristics and responses were analyzed. RESULTS: The overall mean (+/-SD, CV%) steady-state C(mins) for imatinib at 400 mg (n=36) and 600 mg (n=10) daily was 1325.61 ng/mL (+/-583.53 ng/mL; 44%) and 1550.90 ng/mL (+/-462.63 ng/mL; 30%), respectively, and no statistically significant differences were found between them (P=0.267). At 400 mg daily, female patients had significantly higher C(mins) than the male patients (P=0.048), and molecular responses were not correlated with imatinib C(mins), but they were correlated with time elapsed before imatinib therapy. CONCLUSION: The results suggest that Chinese CML patients have higher imatinib C(mins) than their Caucasian counterparts and that the optimal initial imatinib dose for them requires further investigation.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Piperazines/pharmacokinetics , Pyrimidines/pharmacokinetics , Adolescent , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Asian People , Benzamides , China , Dose-Response Relationship, Drug , Female , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/physiopathology , Male , Middle Aged , Piperazines/administration & dosage , Piperazines/therapeutic use , Pyrimidines/administration & dosage , Pyrimidines/therapeutic use , Treatment Outcome , Young Adult
18.
Acta Pharmacol Sin ; 30(10): 1471-8, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19767767

ABSTRACT

AIM: To analyze the results of idarubicin (IDA)- versus etoposide (VP16)-intensified myeloablative conditioning regimen in allogeneic hematopoietic stem cell transplantation (allo-SCT) for high-risk acute leukemia. METHODS: From January 2005 to June 2008, 48 consecutive patients (male: n=29; median age: 30 years, range 14-51 years) with high-risk acute leukemia underwent allo-SCT following an IDA- or VP16-intensified conditioning regimen. The conditioning regimens were modified BUCY2 (busulfan+cyclophosphamide) consisting of IDA (15 mg/m2 per day, days -12 to -10) or VP16 (25 mg/kg per day, days -3 to -2) and CY/TBI (cyclophosphamide/total body irradiation) intensified with IDA (15 mg/m2 per day, days -6 to -5) or VP16 (25 mg/kg per day, days -3 to -2) for acute myeloid leukemia and acute lymphoblastic leukemia, respectively. RESULTS: Between the two groups, no significant differences in terms of baseline characteristics, incidence of acute or chronic graft-versus-host disease (GVHD) or transplant-related mortality (TRM) (P=0.50) were observed. However, the IDA group demonstrated higher incidences of mucositis and Aspergillus pneumonia (P<0.01 and P=0.03, respectively). For the IDA and VP16 groups, relapse rates were 28% and 50%, respectively (P=0.13). For the same groups, the 2-year probabilities of leukemia-free survival (LFS) and overall survival (OS) were 72% versus 51% (P=0.04) and 74% versus 53% (P=0.04), respectively. CONCLUSION: This retrospective analysis suggests that conditioning regimens intensified with IDA can achieve better outcomes than conditioning regimens with VP16 in patients preparing to undergo allo-SCT for high-risk acute leukemia.


Subject(s)
Etoposide/administration & dosage , Hematopoietic Stem Cell Transplantation/methods , Idarubicin/administration & dosage , Leukemia/therapy , Transplantation Conditioning/methods , Acute Disease , Antineoplastic Combined Chemotherapy Protocols , Aspergillus/isolation & purification , Aspergillus/pathogenicity , Busulfan/administration & dosage , China , Combined Modality Therapy , Cyclophosphamide/administration & dosage , Cytarabine/administration & dosage , Disease-Free Survival , Female , Graft Survival , Graft vs Host Disease/prevention & control , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Hydroxyurea/administration & dosage , Immunosuppressive Agents/therapeutic use , Leukemia/mortality , Leukemia, Myeloid, Acute/therapy , Male , Methylprednisolone/administration & dosage , Mucositis/chemically induced , Pneumonia/chemically induced , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Premedication , Retrospective Studies , Risk Factors , Semustine/administration & dosage , Transplantation Conditioning/adverse effects , Transplantation, Homologous/adverse effects , Treatment Outcome , Whole-Body Irradiation
19.
Chin Med J (Engl) ; 121(18): 1770-4, 2008 Sep 20.
Article in English | MEDLINE | ID: mdl-19080355

ABSTRACT

BACKGROUND: Relapse remains an obstacle to successful allogeneic haematopoietic stem cell transplantation (allo-HSCT) for patients with acute leukaemia and no standard treatment is available. We assessed fludarabine and cytarabine with transfusion of donor haematopoietic stem cell in treating the relapse of acute leukaemia after allo-HSCT. METHODS: Seven patients, median age 34 years, with relapse of acute leukaemia after allo-HSCT received combination chemotherapy of fludarabine with cytarabine for 5 days. Five patients suffered from acute myeloid leukaemia (2 refractory) and 2 refractory acute lymphoblastic leukaemia. After the transplantation, the median relapse time was 110 days (range, 38 - 185 days). Two days after chemotherapy, 5 patients received infusion of donor's peripheral blood stem cells, mobilized by granulocyte colony stimulating factor. No prophylactic agents of graft versus host diseases were administered. RESULTS: Six patients achieved haematopoietic reconstitution. DNA sequence analysis at day 30 after treatment identified all as full donor chimera type. The median observation time was 189 days. After the treatment, the median time for neutrophilic granulocyte value = 0.5 x 10(9)/L and for platelet value = 20 x 10(9)/L were 13 days (range, 10 - 18 days) and 15 days (range, 11 - 24 days), respectively. Graft versus host disease occurred in 2 patients (acute) and 3 (chronic). Five patients suffered from pulmonary fungal infection (2 died), 3 haemorrhagic cystitis and 2 cytomegalovirus viraemia. The other patients died of leukaemia related deaths. Three patients with chronic graft versus host disease who had received donor peripheral blood stem cells reinfusion have survived for 375 days, 232 days and 195 days, respectively. CONCLUSIONS: Fludarabine with cytarabine plus the donor haematopoietic stem cell should be considered as an effective therapeutic regimen for relapse of acute leukaemia after allo-HSCT. The disease free state of patients may increase, though with high risk of secondary fungal infection.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute/therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Adolescent , Adult , Antimetabolites, Antineoplastic/administration & dosage , Antineoplastic Agents/administration & dosage , Cytarabine/administration & dosage , Female , Humans , Male , Middle Aged , Transplantation, Homologous , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives
20.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 15(5): 973-7, 2007 Oct.
Article in Chinese | MEDLINE | ID: mdl-17956672

ABSTRACT

The aim of this study was to investigate the effect of [(-)-epigallocatechin-3-gallate (EGCG)] on angiogenesis induced by multiple myeloma cell line KM3 and its mechanism. The effects of KM3 cell supernatant after being treated with EGCG in different concentrations on migration and vascular formation ability of endothelial cell line HUVEC were investigated through culture of MM cell line KM3 in vitro. The secretion level of vascular endothelial growth factor (VEGF) in KM3 cell supernatant and the expression level of VEGF mRNA in KM3 were detected by ELISA and RT-PCR respectively. The results indicated that the KM3 cell supernatant significantly induced endothelial cell migration and vascular formation in vitro. EGCG inhibited the effect of endothelial cell migration induced by KM3 cell supernatant, and the numbers of migrated cells were 414 +/- 27, 299 +/- 70, 202 +/- 42 and 116 +/- 13 at 5, 25, 50, 100 micromol/L respectively. The numbers of migrated cells showed negative correlation with the dose of EGCG (r = -0.952, p < 0.05). The areas of the capillary-like structures decreased while the concentrations of EGCG increased, 88343.9 +/- 3231.1 microm(2) at 25 micromol/L, 60897.5 +/- 914.1 microm2 at 50 micromol/L, which were significantly less than that in the control (p < 0.01) and showed negative correlation with the dose of EGCG (r = -0.888, p < 0.05). 48 hours after treatment with EGCG at concentrations of 5, 25, 50 and 100 micromol/L, the levels of VEGF in the culture supernatant were 1399.0 +/- 47.4, 660.1 +/- 5.7, 108.5 +/- 5.8 and 26.2 +/- 18.6 pg/ml respectively. Except 5 micromol/L, all the other groups showed significant changes while compared with the controls (p < 0.01). Furthermore, EGCG depressed the mRNA expression of VEGF in KM3 cells in a dose-dependent manner. It is concluded that the EGCG can significantly inhibit angiogenic ability of multiple myeloma KM3 cells, its pharmacological mechanism may be downregulation of VEGF mRNA expression and reduction of VEGF secretion.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Catechin/analogs & derivatives , Multiple Myeloma/blood supply , Vascular Endothelial Growth Factor A/metabolism , Catechin/pharmacology , Cell Line, Tumor , Down-Regulation , Humans , Multiple Myeloma/metabolism , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...