Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cell Mol Life Sci ; 81(1): 262, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38878186

ABSTRACT

Through Smad3-dependent signalings, transforming growth factor-ß (TGF-ß) suppresses the development, maturation, cytokine productions and cytolytic functions of NK cells in cancer. Silencing Smad3 remarkably restores the cytotoxicity of NK-92 against cancer in TGF-ß-rich microenvironment, but its effects on the immunoregulatory functions of NK cells remain obscure. In this study, we identified Smad3 functioned as a transcriptional repressor for CSF2 (GM-CSF) in NK cells. Therefore, disrupting Smad3 largely mitigated TGF-ß-mediated suppression on GM-CSF production by NK cells. Furthermore, silencing GM-CSF in Smad3 knockout NK cells substantially impaired their anti-lung carcinoma effects. In-depth study demonstrated that NK-derived GM-CSF strengthened T cell immune responses by stimulating dendritic cell differentiation and M1 macrophage polarization. Meanwhile, NK-derived GM-CSF promoted the survival of neutrophils, which in turn facilitated the terminal maturation of NK cells, and subsequently boosted NK-cell mediated cytotoxicity against lung carcinoma. Thus, Smad3-silenced NK-92 (NK-92-S3KD) may serve as a promising immunoadjuvant therapy with clinical translational value given its robust cytotoxicity against malignant cells and immunostimulatory functions to reinforce the therapeutic effects of other immunotherapies.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor , Killer Cells, Natural , Lung Neoplasms , Smad3 Protein , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Smad3 Protein/metabolism , Smad3 Protein/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/genetics , Animals , Humans , Mice , Mice, Inbred C57BL , Cell Line, Tumor , Dendritic Cells/immunology , Dendritic Cells/metabolism , Transforming Growth Factor beta/metabolism , Cell Differentiation , Macrophages/metabolism , Macrophages/immunology , Signal Transduction
2.
J Exp Clin Cancer Res ; 43(1): 64, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38424636

ABSTRACT

Colorectal cancer (CRC) is a heterogenous malignancy underpinned by dysregulation of cellular signaling pathways. Previous literature has implicated aberrant JAK/STAT3 signal transduction in the development and progression of solid tumors. In this study we investigate the effectiveness of inhibiting JAK/STAT3 in diverse CRC models, establish in which contexts high pathway expression is prognostic and perform in depth analysis underlying phenotypes. In this study we investigated the use of JAK inhibitors for anti-cancer activity in CRC cell lines, mouse model organoids and patient-derived organoids. Immunohistochemical staining of the TransSCOT clinical trial cohort, and 2 independent large retrospective CRC patient cohorts was performed to assess the prognostic value of JAK/STAT3 expression. We performed mutational profiling, bulk RNASeq and NanoString GeoMx® spatial transcriptomics to unravel the underlying biology of aberrant signaling. Inhibition of signal transduction with JAK1/2 but not JAK2/3 inhibitors reduced cell viability in CRC cell lines, mouse, and patient derived organoids (PDOs). In PDOs, reduced Ki67 expression was observed post-treatment. A highly significant association between high JAK/STAT3 expression within tumor cells and reduced cancer-specific survival in patients with high stromal invasion (TSPhigh) was identified across 3 independent CRC patient cohorts, including the TrasnSCOT clinical trial cohort. Patients with high phosphorylated STAT3 (pSTAT3) within the TSPhigh group had higher influx of CD66b + cells and higher tumoral expression of PDL1. Bulk RNAseq of full section tumors showed enrichment of NFκB signaling and hypoxia in these cases. Spatial deconvolution through GeoMx® demonstrated higher expression of checkpoint and hypoxia-associated genes in the tumor (pan-cytokeratin positive) regions, and reduced lymphocyte receptor signaling in the TME (pan-cytokeratin- and αSMA-) and αSMA (pan-cytokeratin- and αSMA +) areas. Non-classical fibroblast signatures were detected across αSMA + regions in cases with high pSTAT3. Therefore, in this study we have shown that inhibition of JAK/STAT3 represents a promising therapeutic strategy for patients with stromal-rich CRC tumors. High expression of JAK/STAT3 proteins within both tumor and stromal cells predicts poor outcomes in CRC, and aberrant signaling is associated with distinct spatially-dependant differential gene expression.


Subject(s)
Colorectal Neoplasms , Humans , Animals , Mice , Retrospective Studies , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Signal Transduction , Hypoxia , Keratins/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Cell Line, Tumor
3.
Mol Cancer Res ; 22(2): 125-136, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-37889101

ABSTRACT

Exosomal long noncoding RNAs (lncRNA) derived from cancer cells are implicated in various processes, including cancer cell proliferation, metastasis, and immunomodulation. We investigated the role and underlying mechanism of exosome-transmitted lncRNA NEAT1 in the immune escape of multiple myeloma cells from natural killer (NK) cells. Multiple myeloma cells and samples from patients with multiple myeloma were obtained. The effects of multiple myeloma cell-derived exosomes (multiple myeloma exosomes) and exosomal NEAT1 on the functions of NK cells were evaluated using EdU staining, CCK-8, flow cytometry, and ELISA. Chromatin and RNA immunoprecipitation were performed to identify interactions between NEAT1, enhancer of Zeste Homolog 2 (EZH2), and pre-B-cell leukemia transcription factor 1 (PBX1). A xenograft tumor model was constructed to verify the effects of exosomal NEAT1 on tumor growth. qRT-PCR, Western blot analysis, and IHC were conducted to detect related genes. NEAT1 levels were upregulated in multiple myeloma tumor tissues, multiple myeloma cells, and multiple myeloma exosomes. Multiple myeloma exosomes suppressed cell proliferation, promoted apoptosis, reduced natural killer group 2, member D (NKG2D)-positive cells, and the production of TNFα) and interferon-gamma (IFN-γ) in NK cells, whereas NEAT1-silenced exosomes had little effect. NEAT1 silenced PBX1 by recruiting EZH2. PBX1 knockdown abrogated the effects of NEAT1-silenced exosomes on NK and multiple myeloma cells. NEAT1-silenced exosomes inhibited tumor growth in mice, decreased Ki67 and PD-L1, and increased NKG2D, TNFα, and IFNγ in tumor tissues. In summary, multiple myeloma cell-derived exosomal NEAT1 suppressed NK-cell activity by downregulating PBX1, promoting multiple myeloma cell immune escape. This study suggests a potential strategy for treating multiple myeloma. IMPLICATIONS: This study reveals that exosomal NEAT1 regulates EZH2/PBX1 axis to inhibit NK-cell activity, thereby promoting multiple myeloma cell immune escape, which offers a novel therapeutic potential for multiple myeloma.


Subject(s)
Exosomes , MicroRNAs , Multiple Myeloma , RNA, Long Noncoding , Animals , Humans , Mice , Cell Line, Tumor , Cell Proliferation , Enhancer of Zeste Homolog 2 Protein/genetics , Exosomes/genetics , Killer Cells, Natural , MicroRNAs/genetics , Multiple Myeloma/genetics , NK Cell Lectin-Like Receptor Subfamily K , Pre-B-Cell Leukemia Transcription Factor 1 , RNA, Long Noncoding/genetics , Tumor Necrosis Factor-alpha
4.
Biomaterials ; 288: 121730, 2022 09.
Article in English | MEDLINE | ID: mdl-35995622

ABSTRACT

Transforming growth factor ß (TGF-ß) is a well-known key mediator for the progression and metastasis of lung carcinoma. However, cost-effective anti-TGF-ß therapeutics for lung cancer remain to be explored. Specifically, the low efficacy in drug delivery greatly limits the clinical application of small molecular inhibitors of TGF-ß. In the present study, specific inhibitor of Smad3 (SIS3) is developed into a self-carried nanodrug (SCND-SIS3) using the reprecipitation method, which largely improves its solubility and bioavailability while reduces its nephrotoxicity. Compared to unmodified-SIS3, SCND-SIS3 demonstrates better anti-cancer effects through inducing tumor cell apoptosis, inhibiting angiogenesis, and boosting NK cell-mediated immune responses in syngeneic Lewis Lung Cancer (LLC) mouse model. Better still, it could achieve comparable anti-cancer effect with just one-fifth the dose of unmodified-SIS3. Mechanistically, RNA-sequencing analysis and cytokine array results unveil a TGF-ß/Smad3-dependent immunoregulatory landscape in NK cells. In particular, SCND-SIS3 promotes NK cell cytotoxicity by ameliorating Smad3-mediated transcriptional inhibition of Ndrg1. Furthermore, improved NK cell cytotoxicity by SCND-SIS3 is associated with higher expression of activation receptor Nkp46, and suppressed levels of Trib3 and TSP1 as compared with unmodified-SIS3. Taken together, SCND-SIS3 possesses superior anti-cancer effects with enhanced bioavailability and biocompatibility, therefore representing as a novel therapeutic strategy for lung carcinoma with promising clinical potential.


Subject(s)
Carcinoma , Lung Neoplasms , Nanoparticles , Animals , Carcinoma/drug therapy , Cell Line, Tumor , Isoquinolines/pharmacology , Isoquinolines/therapeutic use , Lung Neoplasms/drug therapy , Mice , Nanoparticles/therapeutic use , Pyridines/pharmacology , Pyrroles/therapeutic use , Signal Transduction , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism
5.
Mol Ther Oncolytics ; 20: 277-289, 2021 Mar 26.
Article in English | MEDLINE | ID: mdl-33614911

ABSTRACT

Transforming growth factor ß (TGF-ß) has been shown to promote tumor invasion and metastasis by activating the matrix metalloproteinases (MMPs); however, signaling mechanisms remain controversial and therapies targeting MMPs are still suboptimal. In the present study, we found that combined therapy with Asiatic acid (AA), a Smad7 agonist, and Naringenin (NG), a Smad3 inhibitor, effectively retrieved the balance between Smad3 and Smad7 signaling in the TGF-ß-rich tumor microenvironment and thus significantly suppressed tumor invasion and metastasis in mouse models of melanoma and lung carcinoma. Mechanistically, we unraveled that Smad3 acted as a transcriptional activator of MMP2 and as a transcriptional suppressor of tissue inhibitors of metalloproteinase-2 (TIMP2) via binding to 5' UTR of MMP2 and 3' UTR of TIMP2, respectively. Treatment with NG inhibited Smad3-mediated MMP2 transcription while increasing TIMP, whereas treatment with AA enhanced Smad7 to suppress TGF-ß/Smad3 signaling, as well as the activation of MMP2 by targeting the nuclear factor-κB (NF-κB)-membrane-type-1 MMP (MT1-MMP) axis. Therefore, the combination of AA and NG additively suppressed invasion and metastasis of melanoma and lung carcinoma by targeting TGF-ß/Smad-dependent MMP2 transcription, post-translational activation, and function.

6.
Cancer Immunol Res ; 8(8): 1004-1017, 2020 08.
Article in English | MEDLINE | ID: mdl-32532809

ABSTRACT

Tumor-associated macrophages (TAM) have important roles in cancer progression, but the signaling behind the formation of protumoral TAM remains understudied. Here, by single-cell RNA sequencing, we revealed that the pattern recognition receptor Mincle was highly expressed in TAM and significantly associated with mortality in patients with non-small cell lung cancer. Cancer cells markedly induced Mincle expression in bone marrow-derived macrophages (BMDM), thus promoting cancer progression in invasive lung carcinoma LLC and melanoma B16F10 in vivo and in vitro Mincle was predominately expressed in the M2-like TAM in non-small cell lung carcinoma and LLC tumors, and silencing of Mincle unexpectedly promoted M1-like phenotypes in vitro Mechanistically, we discovered a novel Mincle/Syk/NF-κB signaling pathway in TAM needed for executing their TLR4-independent protumoral activities. Adoptive transfer of Mincle-silenced BMDM significantly suppressed TAM-driven cancer progression in the LLC-bearing NOD/SCID mice. By modifying our well-established ultrasound microbubble-mediated gene transfer protocol, we demonstrated that tumor-specific silencing of Mincle effectively blocked Mincle/Syk/NF-κB signaling, therefore inhibiting the TAM-driven cancer progression in the syngeneic mouse cancer models. Thus, our findings highlight the function of Mincle as a novel immunotherapeutic target for cancer via blocking the Mincle/Syk/NF-κB circuit in TAM.


Subject(s)
Lectins, C-Type/metabolism , Lung Neoplasms/immunology , Melanoma/immunology , Membrane Proteins/metabolism , NF-kappa B/metabolism , Receptors, Immunologic/metabolism , Syk Kinase/metabolism , Tumor-Associated Macrophages/immunology , Aged , Animals , Cell Line, Tumor , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Lectins, C-Type/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Macrophages/immunology , Macrophages/pathology , Male , Melanoma/metabolism , Melanoma/pathology , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Middle Aged , NF-kappa B/immunology , Receptors, Immunologic/immunology , Signal Transduction , Syk Kinase/immunology , Tumor-Associated Macrophages/pathology
7.
Future Oncol ; 15(15): 1729-1744, 2019 May.
Article in English | MEDLINE | ID: mdl-31038361

ABSTRACT

Aims: To investigate roles of miR-29a-DNMT1-SOCS1 axis in cervical cancer invasion and migration. Materials & methods: The methylation level of SOCS1 was determined by methylation specific PCR. The cell apoptosis, proliferation, migration and invasion were examined by Annexin-V/PI staining, MTT and colony formation assays, plus scratch and transwell assays respectively. The expressions of epithelial-mesenchymal transition and NF-κB related proteins were determined by western blotting. Results: MiR-29a was downregulated, accompanied with DNMT1 upregulation and SOCS1 downregulation in cervical cancer tissues. MiR-29a suppressed DNMT1, inhibited SOCS1 promoter methylation and upregulated its expression. Moreover, miR-29a promoted cell apoptosis, suppressed proliferation, inhibited migration and invasion via inactivation of NF-κB signaling pathway in cervical cancer cells. Conclusion: MiR-29a-DNMT1-SOCS1 axis plays an important role on invasion and metastasis in cervical cancer via NF-κB signaling pathway.


Subject(s)
DNA Methylation , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , RNA Interference , Suppressor of Cytokine Signaling 1 Protein/genetics , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Adult , Aged , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Decitabine/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter , Humans , Middle Aged , NF-kappa B/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Staging
8.
Life Sci ; 231: 116335, 2019 Aug 15.
Article in English | MEDLINE | ID: mdl-30898647

ABSTRACT

BACKGROUND: PD-L1 enhanced the tumorigenesis and immune escape abilities of cancers. The upstream mechanisms of PD-L1 in regulating tumorigenesis and immune escape of diffuse large B cell lymphoma (DLBCL) remained unclear. METHODS: Human DLBCL cell line OCI-Ly10 and DLBCL patient samples were used in this study. MALAT1 was knocked down by shRNA. MiR-195 was inhibited by miR-195 inhibitor. Levels of MALAT1, PD-L1, miR-195 and CD8 were detected by RT-qPCR. Protein levels of PD-L1, Ras, p-ERK1/2, ERK1/2, Slug, E-cadherin, N-cadherin, Vimentin were detected by western blotting. The interaction between MALAT1 and miR-195, miR-195 and PD-L1 were detected by luciferase assay. OCI-Ly10 cell proliferation and apoptosis were detected by MTT and Annexin V/PI assays, respectively. Migration was detected by transwell assay. Cytotoxicity of CD8+ T cells was detected by LDH cytotoxicity kit. Proliferation and apoptosis of CD8+ T cell co-cultured with OCI-Ly10 cells were analyzed by CFSE and Annexin V/PI staining. RESULTS: MALAT1, PD-L1 and CD8 were up-regulated in DLBCL tissues while miR-195 was down-regulated. MiR-195 was negatively correlated with MALAT1 and PD-L1. MALAT1 could sponge miR-195 to regulate the expression of PD-L1. shMALAT1 treatment increased miR-195 level and decreased PD-L1 level. It also inhibited cell proliferation, migration and immune escape ability while increased apoptosis ratio of OCI-Ly10 cells. shMALAT1 treatment in OCI-Ly10 cells also promoted proliferation and inhibited apoptosis of CD8+ T cells. Knocking down of MALAT1 also suppressed EMT-like process via Ras/ERK signaling pathway. These effects were all rescued by miR-195 inhibitor. CONCLUSION: Long non-coding RNA MALAT1 sponged miR-195 to regulate proliferation, apoptosis and migration and immune escape abilities of DLBCL by regulation of PD-L1.


Subject(s)
Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/immunology , MicroRNAs/immunology , RNA, Long Noncoding/immunology , Tumor Escape/genetics , Apoptosis/physiology , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Carcinogenesis , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Cell Transformation, Neoplastic , Humans , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , MicroRNAs/genetics , RNA, Long Noncoding/genetics , RNA, Small Interfering/genetics , Signal Transduction , Tumor Escape/immunology
9.
Cancer Gene Ther ; 26(7-8): 195-207, 2019 07.
Article in English | MEDLINE | ID: mdl-30470842

ABSTRACT

Understanding the molecular mechanisms for the development of non-Hodgkin lymphoma (NHL) will improve our ability to cure the patients. qRT-PCR was applied for the examination of the efficiency of shRNA for DNMT1, the expression of suppressor genes, miRNA-152. The MTT analysis, cell cycle analysis, clonal formation, and apoptotic analysis were used to examine the functions of DNMT1 and miR-152 in lymphoma cells. Methylation-specific polymerase chain reaction (MSP) was used to examine the methylation of tumor suppressor genes. The dual luciferase assay and western blot were used to validate if DNMT1 is the target of miR-152. For the in vivo experiments, the lymphoma cells were injected into the nude mice for quantification of the tumor growth after transfection of miR-152 mimics. Knockdown of DNMT1 by shRNA (sh-DNMT1) in OCI-Ly10 and Granta-159 cells significantly upregulated the expression of tumor suppressor genes (SOCS3, BCL2L10, p16, p14, and SHP-1) via decreasing their methylation level. At the cellular level, we found sh-DNMT1 inhibited the proliferation, clonal formation and cell cycle progression and induced the cell apoptosis of lymphoma cells. Furthermore, we found miR-152 can downregulates the expression of DNMT1 via directly targeting the gene. Overexpression of miR-152 also increased the expression of tumor suppressor genes SOCS3 and SHP-1. And miR-152 also can inhibit the cell proliferation and induce the cell apoptosis. Moreover, we found overexpression of miR-152 significantly repressed the tumor growth with decreased DNMT1 expression and increased expression of tumor suppressor genes in vivo. Our study demonstrates that miR-152 can inhibit lymphoma growth via suppressing DNMT1-mediated silencing of SOCS3 and SHP-1. These data demonstrate a new mechanism for the development of NHL and this may provide a new therapeutic target for NHL.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/genetics , Lymphoma, Non-Hodgkin/genetics , MicroRNAs/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Suppressor of Cytokine Signaling 3 Protein/genetics , Animals , Apoptosis/physiology , Cell Line, Tumor , Cell Proliferation/physiology , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Down-Regulation , Gene Knockdown Techniques , Genes, Tumor Suppressor , HEK293 Cells , Heterografts , Humans , Lymphoma, Non-Hodgkin/metabolism , Lymphoma, Non-Hodgkin/pathology , Mice , MicroRNAs/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein/metabolism , Transfection
10.
Mol Ther ; 26(9): 2255-2266, 2018 09 05.
Article in English | MEDLINE | ID: mdl-30017880

ABSTRACT

Transforming growth factor ß1 (TGF-ß1) plays a promoting role in tumor growth via a mechanism associated with hyperactive Smad3 and suppressed Smad7 signaling in the tumor microenvironment. We report that retrieving the balance between Smad3 and Smad7 signaling with asiatic acid (AA, a Smad7 inducer) and naringenin (NG, a Smad3 inhibitor) effectively inhibited tumor progression in mouse models of invasive melanoma (B16F10) and lung carcinoma (LLC) by promoting natural killer (NK) cell development and cytotoxicity against cancer. Mechanistically, we found that Smad3 physically bound Id2 and IRF2 to suppress NK cell production and NK cell-mediated cytotoxicity against cancer. Treatment with AA and NG greatly inhibited Smad3 translation and phosphorylation while it restored Smad7 expression, and, therefore, it largely promoted NK cell differentiation, maturation, and cytotoxicity against cancer via Id2/IRF2-associated mechanisms. In contrast, silencing Id2 or IRF2 blunted the protective effects of AA and NG on NK cell-dependent anti-cancer activities. Thus, treatment with AA and NG produced an additive effect on inactivating TGF-ß1/Smad3 signaling, and, therefore, it suppressed melanoma and lung carcinoma growth by promoting NK cell immunity against cancer via a mechanism associated with Id2 and IRF2.


Subject(s)
Flavanones/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Pentacyclic Triterpenes/pharmacology , Smad3 Protein/metabolism , Smad7 Protein/metabolism , Animals , Blotting, Western , Cell Differentiation/drug effects , Cells, Cultured , Inhibitor of Differentiation Protein 2/metabolism , Interferon Regulatory Factor-2/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects
11.
Cancer Immunol Res ; 6(8): 965-977, 2018 08.
Article in English | MEDLINE | ID: mdl-29915022

ABSTRACT

Natural killer (NK) cells, early effectors in anticancer immunity, are paralyzed by TGFß1, an immunosuppressive cytokine produced by cancer cells. Development and activity of NK cells are largely inhibited in the Smad3-dependent tumor microenvironment. Here, we used genetic engineering to generate a stable SMAD3-silencing human NK cell line, NK-92-S3KD, whose cancer-killing activity and cytokine production were significantly enhanced under TGFß1-rich condition compared with the parental cell line. Interestingly, we identified that the IFNG gene is a direct E4BP4 target gene. Thus, silencing of SMAD3 allows upregulation of E4BP4 that subsequently promoting interferon-γ (IFNγ) production in the NK-92-S3KD cells. More importantly, NK-92-S3KD immunotherapy increases the production of not only IFNγ, but also granzyme B and perforin in tumors; therefore, inhibiting cancer progression in two xenograft mouse models with human hepatoma (HepG2) and melanoma (A375). Thus, the NK-92-S3KD cell line may be useful for the clinical immunotherapy of cancer. Cancer Immunol Res; 6(8); 965-77. ©2018 AACR.


Subject(s)
Immunotherapy/methods , Killer Cells, Natural/immunology , Liver Neoplasms, Experimental/therapy , Melanoma, Experimental/therapy , Smad3 Protein/genetics , Animals , Cell Line , Extracellular Matrix Proteins/immunology , Gene Silencing , Genetic Engineering/methods , Humans , Interferon-gamma/biosynthesis , Liver Neoplasms, Experimental/immunology , Melanoma, Experimental/immunology , Mice, Inbred NOD , Mice, Knockout , Smad3 Protein/immunology , Transforming Growth Factor beta/immunology , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
12.
J Vis Exp ; (131)2018 01 09.
Article in English | MEDLINE | ID: mdl-29364233

ABSTRACT

Natural killer (NK) cells belong to the innate immune system and are a first-line anti-cancer immune defense; however, they are suppressed in the tumor microenvironment and the underlying mechanism is still largely unknown. The lack of a consistent and reliable source of NK cells limits the research progress of NK cell immunity. Here, we report an in vitro system that can provide high quality and quantity of bone marrow-derived murine NK cells under a feeder-free condition. More importantly, we also demonstrate that siRNA-mediated gene silencing successfully inhibits the E4bp4-dependent NK cell maturation by using this system. Thus, this novel in vitro NK cell differentiating system is a biomaterial solution for immunity research.


Subject(s)
Cytological Techniques/methods , Killer Cells, Natural/cytology , Animals , Basic-Leucine Zipper Transcription Factors/antagonists & inhibitors , Bone Marrow Cells/cytology , Cell Differentiation/physiology , Cell Line , Gene Silencing , Killer Cells, Natural/physiology , Mice , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics
13.
Kidney Int ; 93(1): 173-187, 2018 01.
Article in English | MEDLINE | ID: mdl-29042082

ABSTRACT

Src activation has been associated with fibrogenesis after kidney injury. Macrophage-myofibroblast transition is a newly identified process to generate collagen-producing myofibroblasts locally in the kidney undergoing fibrosis in a TGF-ß/Smad3-dependent manner. The potential role of the macrophage-myofibroblast transition in Src-mediated renal fibrosis is unknown. In studying this by RNA sequencing at single-cell resolution, we uncovered a unique Src-centric regulatory gene network as a key underlying mechanism of macrophage-myofibroblast transition. A total of 501 differentially expressed genes associated with macrophage-myofibroblast transition were identified. However, Smad3-knockout largely reduced the transcriptome diversity. More importantly, inhibition of Src largely suppresses ureteral obstruction-induced macrophage-myofibroblast transition in the injured kidney in vivo along with transforming growth factor-ß1-induced elongated fibroblast-like morphology, α-smooth muscle actin expression and collagen production in bone marrow derived macrophages in vitro. Unexpectedly, we further uncovered that Src serves as a direct Smad3 target gene and also specifically up-regulated in macrophages during macrophage-myofibroblast transition. Thus, macrophage-myofibroblast transition contributes to Src-mediated tissue fibrosis. Hence, targeting Src may represent as a precision therapeutic strategy for macrophage-myofibroblast transition-driven fibrotic diseases.


Subject(s)
Cell Transdifferentiation , Cicatrix/enzymology , Kidney Diseases/enzymology , Kidney/enzymology , Macrophages/enzymology , Myofibroblasts/enzymology , src-Family Kinases/metabolism , Animals , Cell Transdifferentiation/drug effects , Cell Transdifferentiation/genetics , Cells, Cultured , Cicatrix/genetics , Cicatrix/pathology , Cicatrix/prevention & control , Disease Models, Animal , Fibrosis , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Kidney/drug effects , Kidney/pathology , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney Diseases/prevention & control , Macrophages/drug effects , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Myofibroblasts/drug effects , Myofibroblasts/pathology , Protein Kinase Inhibitors/pharmacology , Sequence Analysis, RNA , Signal Transduction , Single-Cell Analysis , Smad3 Protein/genetics , Smad3 Protein/metabolism , Ureteral Obstruction/drug therapy , Ureteral Obstruction/enzymology , Ureteral Obstruction/genetics , src-Family Kinases/genetics
14.
Nat Commun ; 8: 14677, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28262747

ABSTRACT

TGF-ß is known to influence tumour progression. Here we report an additional role of Smad3 in the tumour microenvironment regulating cancer progression. Deletion or inhibition of Smad3 in the tumour microenvironment suppresses tumour growth, invasion and metastasis in two syngeneic mouse tumour models. Smad3-/- bone marrow gives rise to an expanded NK cell population with enhanced tumour-suppressive activities in vivo, and promotes differentiation of NK cells ex vivo. We identify E4BP4/NFIL3 as a direct Smad3 target gene critical for NK cell differentiation. Smad3 suppresses transcription of IFN-γ via E4BP4 in a T-bet independent manner. Therefore disruption of Smad3 enhances both the E4BP4-mediated NK cell differentiation and anti-cancer effector functions in vivo and in vitro. Furthermore, systemic treatment with a Smad3 inhibitor SIS3 effectively suppresses cancer progression. In summary, suppression of NK cell-mediated immunosurveillance via the Smad3-E4BP4 axis contributes to cancer progression. We propose targeting Smad3-dependent tumour microenvironment may represent an effective anti-cancer strategy.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Killer Cells, Natural/metabolism , Neoplasms, Experimental/metabolism , Smad3 Protein/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Cell Differentiation/immunology , Cell Line, Tumor , Disease Progression , Female , Isoquinolines/pharmacology , Kaplan-Meier Estimate , Killer Cells, Natural/immunology , Male , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Pyridines/pharmacology , Pyrroles/pharmacology , RNA Interference , Smad3 Protein/antagonists & inhibitors , Smad3 Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...