Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 22(3): 580-7, 2014 Jun.
Article in Chinese | MEDLINE | ID: mdl-24989258

ABSTRACT

The genome instability and tumorigenicity of induced pluripotent stem cells (iPSC) hinder their great potentials for clinical application. Using episomal vectors to generate iPSC is the best way to solve safety issues at present. This method is simple and the exogenous gene was not integrated into the host genome. However, the reprogramming efficiency for this method is very low and thus limits its usage. This study was purposed to improve episomal method for generating induced pluripotent stem cells from cord blood mononuclear cells (CB MNC), to establish integration-free iPSC technology system, and to lay the foundation for individualized iPSC for future clinical uses. To improve the reprogramming efficiency for iPSC, episomal method was used at various combinations of episomal vectors, pre-stimulating culture mediums and oxygen condition were tested to optimize the method. The results showed that using erythroid culture medium for culturing 8 days, transfecting with episomal vectors with SFFV (spleen focus forming virus) promoter under the hypoxic condition (3%), CB MNC could be mostly efficiently reprogrammed with the efficiency 0.12%. Furthermore, the results showed that erythroblasts (CD36(+)CD71(+)CD235a(low)) were the cells that are reprogrammed with high efficiency after culture for 8 days. It is concluded that a highly efficient and safe method for generation of integration-free iPSC is successfully established, which is useable in clinical study.


Subject(s)
Cell Culture Techniques/methods , Induced Pluripotent Stem Cells/cytology , Plasmids , Cellular Reprogramming , Genetic Vectors , Humans , Transfection
2.
Acta Pharmacol Sin ; 34(8): 1025-35, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23852085

ABSTRACT

AIM: Toll-like receptor 2 (TLR2) signaling plays a critical role in the initiation of atherosclerosis. The aim of this study was to investigate whether blocking TLR2 activity could produce therapeutic effects on advanced atherosclerosis. METHODS: Forty-week old apolipoprotein E-deficient (ApoE(-/-)) mice fed on a normal diet were intravenously injected with a TLR2-neutralizing antibody or with an isotype-matched IgG for 18 weeks. Double-knockout ApoE(-/-)Tlr2(-/-) mice were taken as a positive control. At the end of the treatments, the plasma lipid levels were measured, and the plaque morphology, pro-inflammatory cytokines expression and apoptosis in arteries were analyzed. In the second part of this study, 6-week old ApoE(-/-) and ApoE(-/-)Tlr2(-/-) mice fed on a high-cholesterol diet for 12 to 24 weeks, the expression levels of TLR2 and apoptotic markers in arteries were examined. RESULTS: Blockade of TLR2 activity with TLR2-neutralizing antibody or knockout of Tlr2 gene did not alter the plasma lipid levels in ApoE(-/-) mice. However, the pharmacologic and genetic manipulations significantly reduced the plaque size and vessel stenosis, and increased plaque stability in the brachiocephalic arteries. The protective effects of TLR2 antagonism were associated with the suppressed expression of pro-inflammatory cytokines IL-6 and TNF-α and the inactivation of transcription factors NF-κB and Stat3. In addition, blocking TLR2 activity attenuated ER stress-induced macrophage apoptosis in the brachiocephalic arteries, which could promote the resolution of necrotic cores in advanced atherosclerosis. Moreover, high-cholesterol diet more prominently accelerated atherosclerotic formation and increased the expression of pro-apoptotic protein CHOP and apoptosis in ApoE(-/-) mice than in ApoE(-/-)Tlr2(-/-) mice. CONCLUSION: The pharmacologic or genetic blockade of TLR2 activity diminishes and stabilizes advanced atherosclerotic lesions in ApoE(-/-) mice. Thus, targeting TLR2 signaling may be a promising therapeutic strategy against advanced atherosclerosis.


Subject(s)
Apolipoproteins E/deficiency , Plaque, Atherosclerotic/drug therapy , Plaque, Atherosclerotic/metabolism , Toll-Like Receptor 2/antagonists & inhibitors , Toll-Like Receptor 2/metabolism , Animals , Antibodies, Neutralizing/pharmacology , Antibodies, Neutralizing/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic/pathology , Random Allocation
3.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 21(2): 268-72, 2013 Apr.
Article in Chinese | MEDLINE | ID: mdl-23628014

ABSTRACT

mTOR (mammalian target of rapamycin) is the center for cellular activities. It controls many cell activities via inhibiting apoptosis and promoting cell growth. Rheb can activate mTOR signaling pathway and participate in genesis and development of multiple cancers. This study was purposed to explore the underlying role of Rheb in human myeloid leukemia by using the myeloid leukemia cell lines. Two myeloid leukemia cell lines HL-60 and K562 overexpressing Rheb were established with retrovirus containing Rheb. The mRNA and protein expressions of Rheb were determined by Real-Time PCR and Western blot respectively. Cell proliferation rate was examined by CCK-8 assay and apoptosis rate was analyzed using Annexin V and 7-AAD double-staining. The results showed that Rheb was overexpressed in both HL-60 and K562 cell lines. The Rheb overexpression cell lines were successfully established. It is found that overexpression of Rheb could promote cell growth. Furthermore, the overexpression of Rheb could accelerate cells entering into G2/M phase (P < 0.01), while did not affect the apoptosis. It is concluded that Rheb overexpression promotes myeloid leukemia cell proliferation through accelerating cell cycle progression.


Subject(s)
Cell Cycle , Cell Proliferation , Monomeric GTP-Binding Proteins/metabolism , Neuropeptides/metabolism , HL-60 Cells , Humans , K562 Cells , Ras Homolog Enriched in Brain Protein , Signal Transduction
4.
Acta Pharmacol Sin ; 33(7): 873-8, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22659623

ABSTRACT

AIM: To explore the signalling pathways involved in aldosterone-induced inflammation and fibrosis in rat vascular smooth muscle cells (VSMCs). METHODS: Using Western blotting and real-time RT-PCR, we investigated the effects of aldosterone on the expression of cyclooxygenase-2 (Cox-2) and IL-6, two important proinflammatory factors, and TGFß1, a critical profibrotic factor, in VSMCs. RESULTS: Aldosterone treatment significantly increased the expression of Cox-2 and IL-6 and activation of p38MAPK and NF-κB. The expression of both Cox-2 and IL-6 could be blocked by the mineralocorticoid receptor (MR) antagonist spironolactone and the p38MAPK inhibitor SB203580. Also, the rapid phosphorylation of p38MAPK could be suppressed by SB203580 but not by spironolactone, implicating in nongenomic effects of aldosterone. Similar to SB203580 and spironolactone, the NF-κB inhibitor α-p-tosyl-L-lysine chloromethyl ketone (TLCK) markedly attenuated expression of Cox-2, indicating that MR, p38MAPK and NF-κB are associated with aldosterone-induced inflammatory responses. Furthermore, aldosterone enhanced expression of TGFß1 in rat VSMCs. This result may be related to activation of the MR/ERK-Sp1 signalling pathway because PD98059, an ERK1/2 inhibitor, significantly blocked the rapid phosphorylation of ERK1/2 and function of Sp1 and led to reduced expression of TGFß1. Spironolactone was also shown to significantly inhibit TGFß1 and Sp1 expression but not ERK1/2 phosphorylation. CONCLUSION: These results suggest that aldosterone-induced inflammatory responses and fibrotic responses may be mediated by the MR/p38MAPK-NF-κB pathways and the MR/ERK-Sp1 pathways in VSMCs, respectively.


Subject(s)
Aldosterone/immunology , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocytes, Smooth Muscle/immunology , NF-kappa B/immunology , Protein Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/immunology , Animals , Cell Line , Cell Nucleolus/immunology , Cell Nucleolus/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/immunology , Interleukin-6/genetics , Interleukin-6/immunology , MAP Kinase Signaling System/drug effects , Muscle, Smooth, Vascular/cytology , Phosphorylation , Protein Kinases/genetics , RNA, Messenger/genetics , Rats , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/immunology , Up-Regulation
5.
Acta Pharmacol Sin ; 33(4): 503-12, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22426694

ABSTRACT

AIM: To develop a rational immunotherapy against tumor metastasis by combining a Toll-like-receptor 2 (TLR2)-neutralizing antibody with a TLR9 agonist CpG ODN, and then investigate the mechanism of action for this combinational regimen. METHODS: After mouse melanoma B16-F10 cell inoculation, female C57BL/6 mice were treated with either CpG ODN (0.5 mg/kg) or the anti-TLR2 antibody (200 µg/kg), or with a combination of the two agents. Pulmonary metastases were evaluated by counting metastatic nodes on the lung surface using anatomical microscopy. Flow cytometry was used to evaluate the cytotoxicity of the immune cells in tumor-draining lymph nodes, the cell population in the spleen, and the infiltration of immune cells within the lungs. Cytokine and enzyme expression in the lung tissue was evaluated using ELISA or immunostaining. RESULTS: Anti-metastatic effects were detected in mice treated with either CpG ODN or the anti-TLR2 antibody alone. However, treatment with CpG ODN plus the anti-TLR2 antibody synergistically suppressed the metastasis as compared with treatment with either single agent. The combinational treatment resulted in enhanced infiltration of natural killer cells and cytotoxic T cells, reduced recruitment of type 2 macrophages and Tregs, and decreased expression of immunosuppressive factors including TGF-ß1, cyclooxygenase-2 and indoleamine 2,3-dioxygenase, thus stimulated tumor cytotoxicity and suppressed metastasis. The anti-metastatic effect of the combinational regimen was further confirmed in spontaneous metastatic mouse model of Lewis lung carcinoma. CONCLUSION: Our studies suggest that combining a TLR9 agonist with an anti-TLR2 antibody, which eliminates immunosuppressive factors from the tumor environment, is critical for an effective anti-metastatic immunotherapy.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Antibodies, Neutralizing/therapeutic use , Lung Neoplasms/secondary , Melanoma, Experimental/therapy , Neoplasm Metastasis/prevention & control , Oligodeoxyribonucleotides/therapeutic use , Toll-Like Receptor 2/immunology , Toll-Like Receptor 9/agonists , Animals , Antibodies, Neutralizing/immunology , Female , Immunotherapy/methods , Lung/drug effects , Lung/pathology , Lung Neoplasms/pathology , Lung Neoplasms/prevention & control , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL
6.
Am J Pathol ; 180(1): 275-92, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22062220

ABSTRACT

Pulmonary fibrosis is an inflammation-driven lung disease with a poor prognosis and no cure. Here we report that basal toll-like receptor 4 (TLR4) activity is critical for the resolution of acute and chronic inflammation and pulmonary fibrosis in mouse models of lung injury. We found that genetic or pharmacologic inhibition of TLR4 exacerbates bleomycin-induced pulmonary inflammation, fibrosis, dysfunction, and animal death through promoting formation of an immunosuppressive tissue microenvironment and attenuating autophagy-associated degradation of collagen and cell death in the fibrotic lung tissues. In contrast, pharmacologic activation of TLR4 resulted in a quick resolution of acute inflammation, reversed the established pulmonary fibrosis, improved lung function, and rescued mice from death. Similarly, blocking TLR4 impaired the resolution of silica-induced chronic inflammation and fibrosis. Importantly, altering autophagic activity could reverse the TLR4-regulated lung inflammation, fibrosis, dysfunction, and animal death. Rapamycin, an autophagy activator, reversed the effects of TLR4 antagonism. In contrast, inhibition of autophagy by 3-methyladenine reversed the proresolving and antifibrotic roles of TLR4 agonists and increased animal death. These results not only highlight a pivotal role for TLR4-mediated basal immunity, particularly autophagic activity, in the proresolution of inflammation and fibrosis after chemical-induced lung injury but also provide proof for the concept for activating TLR4 signaling, particularly TLR4-mediated autophagy, as a novel therapeutic strategy against chronic fibroproliferative diseases that are unresponsive to current therapy.


Subject(s)
Acute Lung Injury/physiopathology , Idiopathic Pulmonary Fibrosis/physiopathology , Lung Injury/physiopathology , Pneumonia/physiopathology , Toll-Like Receptor 4/physiology , Acute Lung Injury/pathology , Animals , Apoptosis/physiology , Autophagy/physiology , Idiopathic Pulmonary Fibrosis/pathology , Lung Injury/pathology , MAP Kinase Signaling System/physiology , Mice , Pneumonia/pathology , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/deficiency
7.
PLoS One ; 6(9): e24705, 2011.
Article in English | MEDLINE | ID: mdl-21931823

ABSTRACT

BACKGROUND: Immunotherapy is often recommended as an adjuvant treatment to reduce the chance of cancer recurrence or metastasis. Interestingly, timing is very important for a successful immunotherapy against metastasis, although the precise mechanism is still unknown. METHODS AND FINDINGS: Using a mouse model of melanoma metastasis induced by intravenous injection of B16-F10 cells, we investigated the mechanism responsible for the diverse efficacy of the prophylactic or therapeutic TLR4 and TLR9 agonist complex against metastasis. We found that the activation of TLR4 and TLR9 prevented, but did not reverse, metastasis because the potency of this combination was neither sufficient to overcome the tumor cell-educated immune tolerance nor to induce efficacious autophagy in tumor cells. The prophylactic application of the complex promoted antimetastatic immunity, leading to the autophagy-associated death of melanoma cells via IFNγ/STAT1 activation and attenuated tumor metastasis. IFNγ neutralization reversed the prophylactic benefit induced by the complex by suppressing STAT1 activation and attenuating autophagy in mice. However, the therapeutic application of the complex did not suppress metastasis because the complex could not reverse tumor cell-induced STAT3 activation and neither activate IFNγ/STAT1 signaling and autophagy. Suppressing STAT3 activation with the JAK/STAT antagonist AG490 restored the antimetastatic effect of the TLR4/9 agonist complex. Activation of autophagy after tumor inoculation by using rapamycin, with or without the TLR4/9 agonist complex, could suppress metastasis. CONCLUSION AND SIGNIFICANCE: Our studies suggest that activation of IFNγ/STAT1 signaling and induction of autophagy are critical for an efficacious anti-metastatic immunotherapy and that autophagy activators may overcome the timing barrier for immunotherapy against metastasis.


Subject(s)
Immunotherapy/methods , Interferon-gamma/metabolism , STAT1 Transcription Factor/metabolism , Animals , Apoptosis/physiology , Autophagy/physiology , Blotting, Western , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Interferon-gamma/genetics , Lung/metabolism , Lung/pathology , Lung/ultrastructure , Melanoma/complications , Melanoma/therapy , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Neoplasm Metastasis/prevention & control , Neoplasm Metastasis/therapy , STAT1 Transcription Factor/genetics , Time Factors
8.
J Immunol ; 187(6): 3003-14, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21841134

ABSTRACT

Pulmonary fibrosis is the pathologic basis for a variety of incurable human chronic lung diseases. IL-17A, a glycoprotein secreted from IL-17-producing cells, has recently been shown to be a proinflammatory cytokine involved in chronic inflammation and autoimmune disease. In this study, we report that IL-17A increased the synthesis and secretion of collagen and promoted the epithelial-mesenchymal transition in alveolar epithelial cells in a TGF-ß1-dependent manner. Using in vivo fibrotic models, we found IL-17A expression to be elevated and IL-17A-associated signaling pathways to be activated in fibrotic lung tissues. Neutralization of IL-17A in vivo promoted the resolution of bleomycin-induced acute inflammation, attenuated pulmonary fibrosis, and increased survival. Additionally, IL-17A antagonism inhibited silica-induced chronic inflammation and pulmonary fibrosis. Targeting IL-17A resulted in a shift of the suppressive immune response in fibrotic lung tissue toward a Th1-type immune response, and it effectively induced autophagy, which promoted the autophagic degradation of collagen and autophagy-associated cell death. Moreover, IL-17A was found to attenuate the starvation-induced autophagy, and autophagy modulators regulated collagen degradation in the alveolar epithelial cells in a TGF-ß1-independent manner. Administration of 3-methylamphetamine, an autophagy inhibitor, reversed the therapeutic efficacy of IL-17A antagonism in pulmonary fibrosis. Our studies indicate that IL-17A participates in the development and progression of pulmonary fibrosis in both TGF-ß1-dependent and -independent manners and that the components of the IL-17A signaling pathway are potential therapeutic targets for the treatment of fibroproliferative lung diseases.


Subject(s)
Interleukin-17/metabolism , Pneumonia/metabolism , Pulmonary Fibrosis/metabolism , Signal Transduction/immunology , Transforming Growth Factor beta1/metabolism , Animals , Autophagy , Blotting, Western , Bronchoalveolar Lavage Fluid/chemistry , Cell Separation , Collagen/biosynthesis , Epithelial-Mesenchymal Transition/immunology , Flow Cytometry , Interleukin-17/antagonists & inhibitors , Interleukin-17/immunology , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Pneumonia/immunology , Pneumonia/pathology , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/pathology , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta1/immunology
9.
Acta Pharmacol Sin ; 32(8): 1045-54, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21706041

ABSTRACT

AIM: To explore the pathogenic role of Th17 cells and interleukin-17A (IL-17A)-associated signaling pathways in spontaneous pulmonary emphysema induced by a Toll-like receptor 4 mutant (TLR4(mut)). METHODS: Lungs were obtained from wild-type (WT) or TLR4mut mice that were treated with or without recombinant mouse IL-17A (1 µg·kg(-1)·d(-1), ip) from the age of 3 weeks to 3 months. Pulmonary emphysema was determined using histology, immunochemistry, and biochemical analysis. T cell polarization was determined with flow cytometry, the levels of cytokines were measured using ELISA, and the levels of IL-17A-associated signaling molecules were detected using Western blot. RESULTS: Compared to WT mice, 3 month-old TLR4(mut) mice were characterized by significantly reduced infiltration of Th17 cells into lungs (2.49%±1.13 % νs 5.26%±1.39%), and significantly reduced expression levels of IL-17A (3.66±0.99 pg/µg νs 10.67±1.65 pg/µg), IL-23 (12.43±1.28 pg/µg νs 28.71±2.57 pg/µg) and IL-6 (51.82±5.45 pg/µg νs 92.73±10.91 pg/µg) in bronchoalveolar lavage fluid. In addition, p38 MAPK phosphorylation and AP-1 expression were decreased to 27%±9% and 51%±8%, respectively, of that in WT mice. Treatment of TLR4(mut) mice with IL-17A increased the infiltration of Th17 cells into lungs and expression levels of IL-17A, IL-6, and IL-23 in bronchoalveolar lavage fluid, attenuated MDA and apoptosis, and improved emphysema accompanied with increased phosphorylation of p38 MAPK and expression of AP-1. CONCLUSION: Th17 cells, in particular the cytokine IL-17A, play a crucial role in the pathogenesis of TLR4(mut)-induced spontaneous pulmonary emphysema. Both of them are potential targets for therapeutic strategies for pulmonary emphysema.


Subject(s)
Interleukin-17/metabolism , Pulmonary Emphysema/genetics , Pulmonary Emphysema/metabolism , Th17 Cells/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Animals , Mice , Mice, Inbred C3H , Mutation/genetics
10.
Yao Xue Xue Bao ; 45(8): 976-86, 2010 Aug.
Article in English | MEDLINE | ID: mdl-21348427

ABSTRACT

Anti-cancer drug bleomycin (BLM) can cause acute lung injury (ALI) which often results in pulmonary fibrosis due to a failure of resolving acute inflammatory response. The aim of this study is to investigate whether toll-like receptor (TLR) 2 mediates BLM-induced ALI, inflammation and fibrosis. BLM-induced dendritic cells (DCs) maturation was analyzed by flow cytometry and cytokine secretion was detected by the ELISA method. The expression and activity of p38 and ERK MAPK were determined with Western blotting. The roles of TLR2 in ALI, inflammation and fibrosis were investigated in C57BL/6 mice administered intratracheally with BLM. The results demonstrated that BLM-administered mice had higher expression of TLR2 (P<0.001) and its signaling molecules. Blocking TLR2 significantly inhibited the maturation of DCs and reversed BLM-stimulated secretion of cytokines in DCs, such as IL-6 (P<0.001), IL-17 (P<0.05) and IL-23 (P<0.05). TLR2 inhibition attenuated BLM-induced increase of inflammatory cells in bronchoalveolar lavage fluid (BALF), and reversed the immunosuppressive microenvironment by enhancing TH1 response (P<0.05) and inhibiting TH2 (P<0.001), Treg (P<0.01) and TH17 (P<0.01) responses. Importantly, blocking TLR2 in vivo significantly protected BLM-administered mice from pulmonary injury, inflammation and fibrosis and subsequently increased BLM-induced animal survival (from 50% to 92%). Therefore, TLR2 is a novel potential target for ALI and pulmonary fibrosis.


Subject(s)
Acute Lung Injury/metabolism , Cytokines/metabolism , Inflammation/metabolism , Pulmonary Fibrosis/metabolism , Toll-Like Receptor 2/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Animals , Bleomycin/toxicity , Bronchoalveolar Lavage Fluid , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/metabolism , Inflammation/chemically induced , Inflammation/pathology , Interleukin-17/metabolism , Interleukin-23/metabolism , Interleukin-6/metabolism , Lung/metabolism , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , T-Lymphocytes, Regulatory/drug effects , Th1 Cells/drug effects , Th2 Cells/drug effects , Toll-Like Receptor 2/physiology
11.
PLoS One ; 4(8): e6520, 2009 Aug 05.
Article in English | MEDLINE | ID: mdl-19654875

ABSTRACT

BACKGROUND: Metastasis is the most pivotal cause of mortality in cancer patients. Immune tolerance plays a crucial role in tumor progression and metastasis. METHODS AND FINDINGS: In this study, we investigated the potential roles and mechanisms of TLR2 signaling on tumor metastasis in a mouse model of intravenously injected B16 melanoma cells. Multiple subtypes of TLRs were expressed on B16 cells and several human cancer cell lines; TLR2 mediated the invasive activity of these cells. High metastatic B16 cells released more heat shock protein 60 than poor metastatic B16-F1 cells. Importantly, heat shock protein 60 released by tumor cells caused a persistent activation of TLR2 and was critical in the constitutive activation of transcription factor Stat3, leading to the release of immunosuppressive cytokines and chemokines. Moreover, targeting TLR2 markedly reduced pulmonary metastases and increased the survival of B16-bearing mice by reversing B16 cells induced immunosuppressive microenvironment and restoring tumor-killing cells such as CD8(+) T cells and M1 macrophages. Combining an anti-TLR2 antibody and a cytotoxic agent, gemcitabine, provided a further improvement in the survival of tumor-bearing mice. CONCLUSIONS AND SIGNIFICANCE: Our results demonstrate that TLR2 is an attractive target against metastasis and that targeting immunosuppressive microenvironment using anti-TLR2 antibody is a novel therapeutic strategy for combating a life-threatening metastasis.


Subject(s)
Lung Neoplasms/pathology , Melanoma, Experimental/pathology , Neoplasm Metastasis/prevention & control , Toll-Like Receptor 2/antagonists & inhibitors , Animals , Chaperonin 60/metabolism , Chemokines/metabolism , Cytokines/metabolism , Lung Neoplasms/metabolism , Melanoma, Experimental/metabolism , Mice , Toll-Like Receptor 2/immunology , Toll-Like Receptor 2/physiology
12.
J Immunol ; 182(1): 692-702, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19109203

ABSTRACT

Pulmonary fibrosis is a consequence of chronic lung injury and is associated with a high mortality. Despite the pathogenesis of pulmonary fibrosis remaining as an enigma, immune responses play a critical role in the deregulation of wound healing process after lung injury, which leads to fibrosis. Accumulating evidence argues the rationales for current treatments of pulmonary fibrosis using immunosuppressive agents such as corticosteroids. In this study, we report that bleomycin (BLM), a well-known fibrogenic agent functioning as a TLR2 agonist, induced the maturation of dendritic cells and release of cytokines. The BLM activation of TLR2 mediated a time-dependent alteration of immune responses in the lung. These responses resulted in an increase in the tissue-infiltrating proinflammatory cells and cytokines in the early period initially following BLM exposure and an increase in the tissue-infiltrating suppressive immune cells and factors during the later period following BLM exposure. TLR2 deficiency, however, reduced pulmonary inflammation, injury, and subsequently attenuated pulmonary fibrosis. Targeting TLR2 by a TLR2-neutralizing Ab not only markedly decreased animal death but also protected animals from the development of pulmonary fibrosis and reversed the established pulmonary fibrosis through regulating BLM-induced immunosuppressive microenvironments. Our studies suggest that TLR2 is a promising target for the development of therapeutic agents against pulmonary fibrosis and that eliminating immunosuppressive cells and factors via immunostimulants is a novel strategy for fibro-proliferative diseases. Moreover, combining BLM with an anti-TLR2 Ab or TLR2 antagonist for cancer therapy will improve the BLM therapeutic profile by enhancing anti-cancer efficacy and reducing systemic inflammation and pulmonary fibrosis.


Subject(s)
Bleomycin/toxicity , Immunosuppressive Agents , Inflammation Mediators/antagonists & inhibitors , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/prevention & control , Toll-Like Receptor 2/antagonists & inhibitors , Toll-Like Receptor 2/deficiency , Animals , Bleomycin/adverse effects , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Movement/drug effects , Cell Movement/immunology , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/pathology , Disease Models, Animal , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/toxicity , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Intubation, Intratracheal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/pathology , Toll-Like Receptor 2/agonists , Toll-Like Receptor 2/physiology
13.
Sheng Li Ke Xue Jin Zhan ; 40(4): 297-302, 2009 Oct.
Article in Chinese | MEDLINE | ID: mdl-21417026

ABSTRACT

B cells are typically characterized by their ability to regulate the immune responses through presenting antigens and producing antibodies. However, a novel B cell subset, named regulatory B cells (Bregs), has been identified. As Tregs, the Bregs are capable of performing both pathogenic and regulatory functions by production of suppressive cytokines, such as IL-10 or TGF-beta1, or by interaction with pathogen T cells or other immune cells. Recent studies indicate that the Bregs play a critical role in the development and resolution of multiple chronic diseases, including inflammatory bowel disease, rheumatoid arthritis, and experimental autoimmune encephalomyelitis. The identification and the clarification of action mechanisms of the Bregs will greatly contribute to understanding the mechanisms of immune tolerance comprehensively and deeply, and to develop the rational therapeutic strategies for arthritis, diabetes, multiple sclerosis, infectious diseases and cancer, etc. In this review, we summarized the recent insights of identification, characterizations, development, and regulation mechanisms of Bregs and these cells' contribution to the pathogenesis of inflammatory diseases.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Immune Tolerance , Animals , Humans , Inflammation
14.
Sheng Li Ke Xue Jin Zhan ; 39(2): 114-8, 2008 Apr.
Article in Chinese | MEDLINE | ID: mdl-18664173

ABSTRACT

Toll-like receptors (TLRs) are widely expressed in the innate and adaptive immune system. They initiate host defense against endogenous and exogenous pathogens containing conserved pathogen associated molecular patterns. TLRs are critical bridges between the innate and adaptive immunity, especially the cellular immunity mediated by T cells. Emerging evidence indicated that B cells express almost all subtypes of TLRs. TLRs play critical role not only in regulation of proliferation, maturation and function of B cells, but also in pathogenesis of diseases, such as systemic lupus erythematosus and chronic lymphocytic leukemia. Targeting TLRs of B cells is a promising therapeutic strategy for these disorders.


Subject(s)
B-Lymphocytes/metabolism , Toll-Like Receptors/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/physiology , Humans , Signal Transduction , Toll-Like Receptors/immunology , Toll-Like Receptors/physiology
15.
J Immunol ; 180(11): 7349-57, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18490734

ABSTRACT

Hypertension-induced cardiovascular hypertrophy and fibrosis are critical in the development of heart failure. The activity of TLRs has been found to be involved in the development of pressure overload-induced myocardial hypertrophy and cardiac fibrosis. We wondered whether vaccine bacillus Calmette-Guérin (BCG), which activated TLR4 to elicit immune responses, modulated the pressure overload-stimulated cardiovascular hypertrophy and cardiac fibrosis in the murine models of abdominal aortic constriction (AAC)-induced hypertension. Before or after AAC, animals received BCG, TLR4 agonist, IFN-gamma, or TLR4 antagonist i.p. BCG and TLR4 agonist significantly prevented AAC-induced cardiovascular hypertrophy and reactive cardiac fibrosis with no changes in hemodynamics. Moreover, TLR4 antagonist reversed the BCG- and TLR4 agonist-induced actions of anti-cardiovascular hypertrophy and cardiac fibrosis. BCG decreased the expression of TLR2 or TLR4 on the heart tissue but TLR4 agonist increased the expression of TLR2 or TLR4 on the immune cells that infiltrate into the heart tissue. This led to an increased expression ratio of IFN-gamma/TGF-beta in the heart. The cardiac protective effects of BCG and TLR4 agonist are related to their regulation of ERK-Akt and p38-NF-kappaB signal pathways in the heart. In conclusion, the activity of TLR4 plays a critical role in the mediation of pressure overload-induced myocardial hypertrophy and fibrosis. The regulation of immune responses by BCG and TLR4 agonist has a great potential for the prevention and treatment of hypertension-induced myocardial hypertrophy and cardiac fibrosis.


Subject(s)
BCG Vaccine/immunology , Cardiomegaly/immunology , Interferon-gamma/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Acyltransferases/pharmacology , Animals , BCG Vaccine/administration & dosage , Cardiomegaly/prevention & control , Cardiovascular System/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Escherichia coli Proteins/pharmacology , Fibrosis/immunology , Fibrosis/prevention & control , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Interferon-gamma/immunology , Lipopolysaccharides/immunology , Male , Mice , Mice, Inbred Strains , Mitogen-Activated Protein Kinase Kinases/metabolism , Myocardium/pathology , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/immunology , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism
16.
Acta Pharmacol Sin ; 25(10): 1292-8, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15456530

ABSTRACT

AIM: To prepare neutralizing monoclonal antibody (mAb) against extracellular immunoglobulin (Ig)-like domain III of vascular endothelial growth factor receptor KDR and study its biological activity. METHODS: Soluble KDR Ig domain III (KDR-III) fusion protein was expressed in E Coli and purified from the bacterial periplasmic extracts via an affinity chromatography. Monoclonal antibodies against KDR-III were prepared by hybridoma technique. ELISA and FACS analysis were used to identify its specificity. Immunoprecipitation and [3H]-thymidine incorporation assay were also used to detect the activity of anti-KDR mAb blocking the phosphorylation of KDR tyrosine kinase receptor and the influence on vascular endothelial growth factor-induced mitogenesis of human endothelial cells. RESULTS: A monoclonal antibody, Ycom1D3 (IgG1), was generated from a mouse immunized with the recombinant KDR-III protein. Ycom1D3 bound specifically to both the soluble KDR-III and the cell-surface expressed KDR. Ycom1D3 effectively blocked VEGF/KDR interaction and inhibited VEGF-stimulated KDR activation in human endothelial cells. Furthermore, the antibody efficiently neutralized VEGF-induced mitogenesis of human endothelial cells. CONCLUSION: Our results suggest that the anti-KDR mAb, Ycom1D3, has potential applications in the treatment of cancer and other diseases where pathological angiogenesis is involved.


Subject(s)
Antibodies, Monoclonal/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Antibodies, Monoclonal/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Endothelial Cells/metabolism , Escherichia coli/metabolism , Humans , Immunoglobulin Fragments/immunology , Immunoglobulin Fragments/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mitosis/drug effects , Phosphorylation , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Umbilical Veins/cytology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/immunology
17.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 9(4): 343-346, 2001 Dec.
Article in Chinese | MEDLINE | ID: mdl-12578576

ABSTRACT

To explore the antitumor activities of KRN7000 in a NS-1 myeloma-bearing mice and the underlying mechanisms, cytotoxic activities of the spleen cells treated with KRN7000 in vivo and in vitro were examined with Yac-1 (a NK-sensitive cell line) and NS-1 (a NK-insensitive cell line) as the targeted cells. The life span of NS-1 myeloma-bearing mice was observed after treating with KRN7000 and combined with cyclophosphamide (CTX). Furthermore, toxicities of KRN7000 administration on liver and kidney were evaluated. The results showed that KRN7000 enhanced NK cytotoxic activities of spleen cells. Around 20% of NS-1-bearing mice survived after KRN7000 administration and the survival rate reached up to 80% of NS-1-bearing mice when KRN7000 was used in combination with CTX at a dose of 100 mg/kg (P < 0.005). KRN7000, at a dose of 100 micro g/kg, had no toxic effects on liver and kidney. These findings suggest that KRN7000 might be a promosing agent in tumor immunotherapy.

SELECTION OF CITATIONS
SEARCH DETAIL
...