Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 3178, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38609378

ABSTRACT

Chemo-immunotherapy combinations have been regarded as one of the most practical ways to improve immunotherapy response in cancer patients. In this study, we integrate the transcriptomics data from anti-PD-1-treated tumors and compound-treated cancer cell lines to systematically screen for chemo-immunotherapy synergisms in silico. Through analyzing anti-PD-1 induced expression changes in patient tumors, we develop a shift ability score to measure if a chemotherapy or a small molecule inhibitor treatment can shift anti-PD-1 resistance in tumor cells. By applying shift ability analysis to 41,321 compounds and 16,853 shRNA treated cancer cell lines transcriptomic data, we characterize the landscape of chemo-immunotherapy synergism and experimentally validated a mitochondrial RNA-dependent mechanism for drug-induced immune activation in tumor. Our study represents an effort to mechanistically characterize chemo-immunotherapy synergism and will facilitate future pre-clinical and clinical studies.


Subject(s)
Immunotherapy , Neoplasms , Humans , Drug Therapy, Combination , Cell Line , Gene Expression Profiling , Neoplasms/drug therapy , Neoplasms/genetics
2.
Res Sq ; 2023 Sep 14.
Article in English | MEDLINE | ID: mdl-37790509

ABSTRACT

Chemo-immunotherapy combinations have been regarded as one of the most practical ways to improve immunotherapy response in cancer patients. In this study, we integrated the transcriptomics data from immunotherapy-treated tumors and compound-treated cell lines to systematically identify chemo-immunotherapy synergisms and their underlying mechanisms. Through analyzing anti-PD-1 treatment induced expression changes in patient tumors, we developed a shift ability score that can measure whether a chemotherapy treatment shifts anti-PD-1 response. By applying the shift ability analysis on 41,321 compounds and 16,853 shRNA treated cancer cell line expression profiles, we characterized a systematic landscape of chemo-immunotherapy synergism and prioritized 17 potential synergy targets. Further investigation of the treatment induced transcriptomic data revealed that a mitophagy-dsRNA-MAVS-dependent activation of type I IFN signaling may be a novel mechanism for chemo-immunotherapy synergism. Our study represents the first comprehensive effort to mechanistically characterize chemo-immunotherapy synergism and will facilitate future pre-clinical and clinical studies.

3.
Sci Adv ; 8(49): eadd0005, 2022 12 09.
Article in English | MEDLINE | ID: mdl-36475797

ABSTRACT

The majority of lncRNAs' roles in tumor immunology remain elusive. This project performed a CRISPR activation screening of 9744 lncRNAs in melanoma cells cocultured with human CD8+ T cells. We identified 16 lncRNAs potentially regulating tumor immune response. Further integrative analysis using tumor immunogenomics data revealed that IL10RB-DT and LINC01198 are significantly correlated with tumor immune response and survival in melanoma and breast cancer. Specifically, IL10RB-DT suppresses CD8+ T cells activation via inhibiting IFN-γ-JAK-STAT1 signaling and antigen presentation in melanoma and breast cancer cells. On the other hand, LINC01198's up-regulation sensitizes the killing of tumor cells by CD8+ T cells. Mechanistically, LINC01198 interacts and activates NF-κB component p65 to trigger the type I and type II interferon responses in melanoma and breast cancer cells. Our study systematically characterized novel lncRNAs involved in tumor immune response.


Subject(s)
Breast Neoplasms , Melanoma , RNA, Long Noncoding , Humans , Female , RNA, Long Noncoding/genetics , CD8-Positive T-Lymphocytes , Gain of Function Mutation , Immunity , Melanoma/genetics , Breast Neoplasms/genetics
4.
Mol Carcinog ; 61(12): 1073-1081, 2022 12.
Article in English | MEDLINE | ID: mdl-36161729

ABSTRACT

Lentivirus-based transduction systems are widely used in biological science and cancer biology, including cancer immunotherapy. However, in in vivo transplanted tumor model, the immunogenicity of these transduced cells was not appropriately addressed. Here, we used empty vector-transduced mouse melanoma (B16) and carcinoma (lewis lung carcinoma) cells transplanted tumor model to study the immune response due to the transduction processes. We showed that the overall in vivo tumor growth rate gets reduced in transduced cells only in immune-competent mice but not in nude mice. This data indicate the involvement of the immune system in the in vivo tumor growth restriction in the transduced group. Further studies showed that specific activation of CD8+ T cells might be responsible for restricted tumor growth. Mechanistically, transduced tumor cells show the higher activity of type I interferon, which might play an essential role in this activation. Overall, our data indicate the modulation of the immune system by lentiviral vector transduced tumor cells, which required further studies to explore the mechanisms and better understand the biological significance. Our data also indicate the importance of considering the immunogenicity of transduced cells when analyzing in vivo results, especially in studies related to immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Mice , Animals , Transduction, Genetic , Genetic Vectors/genetics , Mice, Nude , Dendritic Cells , Lentivirus/genetics
5.
Inflamm Res ; 68(12): 1011-1024, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31489459

ABSTRACT

BACKGROUND: Polarized macrophages induce fibrosis through multiple mechanisms, including a process termed epithelial-to-mesenchymal transition (EMT). Mesenchymal cells contribute to the excessive accumulation of fibrous connective tissues, leading to organ failure. This study was aimed to investigate the effect of tannic acid (TA), a natural dietary polyphenol on M1 macrophage-induced EMT and its underlying mechanisms. MATERIALS: First, we induced M1 polarization in macrophage cell lines (RAW 264.7 and THP-1). Then, the conditioned-medium (CM) from these polarized macrophages was used to induce EMT in the human adenocarcinomic alveolar epithelial (A549) cells. We also analysed the role of TA on macrophage polarization. RESULTS: We found that TA pre-treated CM did not induce EMT in epithelial cells. Further, TA pre-treated CM showed diminished activation of MAPK in epithelial cells. Subsequently, TA was shown to inhibit LPS-induced M1 polarization in macrophages by directly targeting toll-like receptor 4 (TLR4), thereby repressing LPS binding to TLR4/MD2 complex and subsequent signal transduction. CONCLUSION: It was concluded that TA prevented M1 macrophage-induced EMT by suppressing the macrophage polarization possibly through inhibiting the formation of LPS-TLR4/MD2 complex and blockage of subsequent downstream signal activation. Further, our findings may provide beneficial information to develop new therapeutic strategies against chronic inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Lung/cytology , Macrophage Activation/drug effects , Tannins/pharmacology , Toll-Like Receptor 4/metabolism , A549 Cells , Animals , Fibrosis , Humans , Lipopolysaccharides/pharmacology , Mice , RAW 264.7 Cells , THP-1 Cells
6.
J Cell Physiol ; 234(5): 6463-6476, 2019 05.
Article in English | MEDLINE | ID: mdl-30246289

ABSTRACT

Acute lung injury (ALI) and its severe form acute respiratory distress syndrome (ARDS) remain a major cause of morbidity and mortality in critically ill patients, and no specific therapies are still available to control the mortality rate. Thus, we explored the preventive and therapeutic effects of tannic acid (TA), a natural polyphenol in the context of ALI. We used in vivo and in vitro models, respectively, using lipopolysaccharide (LPS) to induce ALI in mice and exposing J774 and BEAS-2B cells to LPS. In both preventive and therapeutic approaches, TA attenuated LPS-induced histopathological alterations, lipid peroxidation, lung permeability, infiltration of inflammatory cells, and the expression of proinflammatory mediators. In addition, in-vitro study showed that TA treatment could reduce the expression of proinflammatory mediators. Further studies revealed that TA-dampened inflammatory responses by downregulating the LPS-induced toll-like receptor 4 (TLR4) expression and inhibiting extracellular-signal-regulated kinase (ERK)1/2 and p38 mitogen-activated protein kinase (MAPK) activation. Furthermore, cells treated with the inhibitors of ERK1/2 (PD98059) and p38 (SB203580) mitigated the expression of cytokines induced by LPS, thus suggesting that ERK1/2 and p38 activity are required for the inflammatory response. In conclusion, TA could attenuate LPS-induced inflammation and may be a potential therapeutic agent for ALI-associated inflammation in clinical settings.


Subject(s)
Acute Lung Injury/pathology , Mitogen-Activated Protein Kinases/biosynthesis , Tannins/pharmacology , Toll-Like Receptor 4/biosynthesis , Acute Lung Injury/metabolism , Animals , Disease Models, Animal , Down-Regulation , Male , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinases/drug effects , Toll-Like Receptor 4/drug effects
7.
J Cell Biochem ; 119(8): 6732-6742, 2018 08.
Article in English | MEDLINE | ID: mdl-29665059

ABSTRACT

In response to tissue injury, fibroblasts migrate into the wound, where they undergo proliferation and differentiation. The persistence of these differentiated fibroblasts (myofibroblasts) is associated with excessive scarring in various organs. We aimed to investigate the effects of Tannic acid (TA) on fibroblast proliferation and differentiation, and found that TA inhibited fibroblast differentiation as assessed by reduced expression of α-smooth muscle actin, N-cadherin, and type-1-collagen. TA also prevented the TGF-ß1-induced alteration in the expression of two classes of genes involved in the remodeling of extracellular matrix (ECM) proteins, namely matrix metalloproteinases (Mmp-2 and -9) and tissue inhibitors of metalloproteinases (Timp-1 and -3). Further, TA suppressed TGF-ß1-induced cell proliferation and induced cell cycle arrest at G0/G1 phase via targeting Cyclins expression. Finally, TA exerted its inhibitory effects by decreasing the phosphorylation of Smad and ERK signaling. In sum, our results suggesting that TA may be a potential therapeutic agent for pathological fibrosis.


Subject(s)
Cell Differentiation/drug effects , G1 Phase/drug effects , Myofibroblasts/metabolism , Resting Phase, Cell Cycle/drug effects , Actins/biosynthesis , Animals , Cadherins/biosynthesis , Collagen Type I/biosynthesis , Extracellular Matrix/metabolism , Fibrosis , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Myofibroblasts/pathology , NIH 3T3 Cells , Tannins , Transforming Growth Factor beta1/pharmacology
8.
Clin Rev Allergy Immunol ; 54(3): 480-492, 2018 Jun.
Article in English | MEDLINE | ID: mdl-27677501

ABSTRACT

Lack of markers of subclinical disease state and clinical phenotype other than pulmonary function test has made the diagnosis and interventions of environmental respiratory diseases a major challenge. MicroRNAs (miRNAs), small non-coding single stranded RNAs, have emerged as potential disease-modifier in various environmental respiratory diseases. They can also be found in various body fluids and are remarkably stable. Because of their high stability, disease-specific expression, and the ease to detect and quantify them have raised the potential of miRNAs in body fluids to be useful clinical diagnostic biomarkers for lung disease phenotyping. In the present review, we provide a comprehensive overview of progress made in identifying miRNAs in various body fluids including blood, serum, plasma, bronchoalveolar lavage (BAL) fluid, and sputum as biomarkers for a wide range of human respiratory diseases such as acute lung injury/acute respiratory distress syndrome (ALI/ARDS), idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD), and asthma. Finally, we discuss several challenges remain to be concerned and suggest few disease-specific and non-specific miRNAs to become part of future clinical practice.


Subject(s)
Asthma/genetics , Circulating MicroRNA , Idiopathic Pulmonary Fibrosis/genetics , Lung/pathology , MicroRNAs/genetics , Pulmonary Disease, Chronic Obstructive/genetics , Respiratory Distress Syndrome/genetics , Asthma/diagnosis , Biomarkers/metabolism , Bronchoalveolar Lavage Fluid/immunology , Extracellular Space , Humans , Idiopathic Pulmonary Fibrosis/diagnosis , Pulmonary Disease, Chronic Obstructive/diagnosis , Respiratory Distress Syndrome/diagnosis
9.
J Cell Physiol ; 233(3): 2513-2525, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28771711

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and an irreversible lung disorder characterized by the accumulation of fibroblasts and myofibroblasts in the extracellular matrix. The transforming growth factor-ß1 (TGF-ß1)-induced epithelial-to-mesenchymal transition (EMT) is thought to be one of the possible sources for a substantial increase in the number of fibroblasts/myofibroblasts in IPF lungs. Tannic acid (TA), a natural dietary polyphenolic compound has been shown to possess diverse pharmacological effects. However, whether TA can inhibit TGF-ß1-mediated EMT in lung epithelial cells remains enigmatic. Both the human adenocarcinomic alveolar epithelial (A549) and normal bronchial epithelial (BEAS-2B) cells were treated with TGF-ß1 with or without TA. Results showed that TA addition, markedly inhibited TGF-ß1-induced EMT as assessed by reduced expression of N-cadherin, type-1-collagen, fibronectin, and vimentin. Furthermore, TA inhibited TGF-ß1-induced cell proliferation through inducing cell cycle arrest at G0/G1 phase. TGF-ß1-induced increase in the phosphorylation of Smad (Smad2 and 3), Akt as well as that of mitogen activated protein kinase (ERK1/2, JNK1/2, and p38) mediators was effectively inhibited by TA. On the other hand, TA reduced the TGF-ß1-induced increase in TGF-ß receptors expression. Using molecular docking approach, FTIR, HPLC and Western blot analyses, we further identified the direct binding of TA to TGF-ß1. Finally, we conclude that TA might directly interact with TGF-ß1, thereby repressing TGF-ß signaling and subsequent EMT process in lung epithelial cells. Further animal studies are needed to clarify its potential therapeutic benefit in pulmonary fibrosis.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Idiopathic Pulmonary Fibrosis/drug therapy , Lung Neoplasms/drug therapy , Lung/drug effects , Signal Transduction/drug effects , Tannins/pharmacology , Transforming Growth Factor beta1/metabolism , A549 Cells , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Antineoplastic Agents/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lung/metabolism , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Molecular Docking Simulation , Protein Binding , Tannins/metabolism , Time Factors , Transforming Growth Factor beta1/pharmacology
10.
Pharmacol Rep ; 69(3): 426-431, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28288400

ABSTRACT

BACKGROUND: Epithelial mesenchymal transition (EMT) is a process through which epithelial cells undergo multiple biochemical changes, causing them to differentiate into a mesenchymal-cell phenotype. This process has been shown to contribute to the development of fibrotic diseases. C-phycocyanin (C-PC) is a phycobiliprotein extracted from Spirulina platensis. This study was done to investigate the effect of C-PC on transforming growth factor-ß1 (TGF-ß1)-induced EMT and an EMT associated proliferation in human epithelial cell lines. METHODS: Human adenocarcinoma cell line, A549 and breast cancer cell line, MCF-7 were treated with TGF-ß1, and EMT-related genes expression, cell proliferation and cell cycle arrest were examined. RESULTS: C-PC suppressed the EMT as assessed by reduced expression of vimentin, type-1-collagen and fibronectin, and increased E-cadherin expression in TGF-ß1 treated cells. Further, TGF-ß1 treatment induced cell cycle arrest in S and G2/M phase in A549 cells. However, TGF-ß1-mediated cell cycle arrest was significantly reversed by combined treatment with C-PC. CONCLUSIONS: The overall data suggested that C-PC suppresses TGF- ß1-induced EMT and warrants further in vivo studies for future evaluation of C-PC as a potential antifibrotic agent.


Subject(s)
Cell Proliferation/drug effects , Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Phycocyanin/pharmacology , A549 Cells , Adenocarcinoma/metabolism , Breast Neoplasms/metabolism , Cell Cycle Checkpoints/drug effects , Epithelial Cells/metabolism , Female , Gene Expression Regulation/drug effects , Humans , MCF-7 Cells , Phycocyanin/isolation & purification , Transforming Growth Factor beta1/administration & dosage
11.
J Cell Physiol ; 231(10): 2097-106, 2016 10.
Article in English | MEDLINE | ID: mdl-26790856

ABSTRACT

The acute respiratory distress syndrome (ARDS), a severe form of acute lung injury (ALI), is a very common condition associated with critically ill patients, which causes substantial morbidity and mortality worldwide. Despite decades of research, effective therapeutic strategies for clinical ALI/ARDS are not available. In recent years, microRNAs (miRNAs), small non-coding molecules have emerged as a major area of biomedical research as they post-transcriptionally regulate gene expression in diverse biological and pathological processes, including ALI/ARDS. In this context, this present review summarizes a large body of evidence implicating miRNAs and their target molecules in ALI/ARDS originating largely from studies using animal and cell culture model systems of ALI/ARDS. We have also focused on the involvement of miRNAs in macrophage polarization, which play a critical role in regulating the pathogenesis of ALI/ARDS. Finally, the possible future directions that might lead to novel therapeutic strategies for the treatment of ALI/ARDS are also reviewed. J. Cell. Physiol. 231: 2097-2106, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Acute Lung Injury/genetics , Gene Expression/genetics , MicroRNAs/genetics , Protein Processing, Post-Translational/genetics , Respiratory Distress Syndrome/genetics , Acute Lung Injury/metabolism , Acute Lung Injury/therapy , Animals , Disease Models, Animal , Humans , Respiratory Distress Syndrome/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...