Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Nature ; 623(7985): 157-166, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37853118

ABSTRACT

Immunotherapy failures can result from the highly suppressive tumour microenvironment that characterizes aggressive forms of cancer such as recurrent glioblastoma (rGBM)1,2. Here we report the results of a first-in-human phase I trial in 41 patients with rGBM who were injected with CAN-3110-an oncolytic herpes virus (oHSV)3. In contrast to other clinical oHSVs, CAN-3110 retains the viral neurovirulence ICP34.5 gene transcribed by a nestin promoter; nestin is overexpressed in GBM and other invasive tumours, but not in the adult brain or healthy differentiated tissue4. These modifications confer CAN-3110 with preferential tumour replication. No dose-limiting toxicities were encountered. Positive HSV1 serology was significantly associated with both improved survival and clearance of CAN-3110 from injected tumours. Survival after treatment, particularly in individuals seropositive for HSV1, was significantly associated with (1) changes in tumour/PBMC T cell counts and clonal diversity, (2) peripheral expansion/contraction of specific T cell clonotypes; and (3) tumour transcriptomic signatures of immune activation. These results provide human validation that intralesional oHSV treatment enhances anticancer immune responses even in immunosuppressive tumour microenvironments, particularly in individuals with cognate serology to the injected virus. This provides a biological rationale for use of this oncolytic modality in cancers that are otherwise unresponsive to immunotherapy (ClinicalTrials.gov: NCT03152318 ).


Subject(s)
Brain Neoplasms , Glioblastoma , Herpesvirus 1, Human , Oncolytic Virotherapy , Oncolytic Viruses , Humans , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Glioblastoma/immunology , Glioblastoma/pathology , Nestin/genetics , Oncolytic Virotherapy/adverse effects , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology , Oncolytic Viruses/physiology , Reproducibility of Results , Survival Analysis , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Treatment Outcome , Tumor Microenvironment/immunology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/physiology
2.
Sci Rep ; 11(1): 14120, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34239022

ABSTRACT

Transforming growth factor beta (TGF-ß) is the main cytokine responsible for the induction of the epithelial-mesenchymal transition of breast cancer cells, which is a hallmark of tumor transformation to the metastatic phenotype. Recently, research demonstrated that the chemokine CCL2 gene expression level directly correlates with the TGF-ß activity in breast cancer patients. CCL2 attracts tumor-associated macrophages and is, therefore, considered as an important inductor of breast cancer progression; however, the precise mechanisms underlying its regulation by TGF-ß are unknown. Here, we studied the behavior of the CCL2 gene in MDA-MB-231 and HCC1937 breast cancer cells representing mesenchymal-like phenotype activated by TGF-ß. Using bioinformatics, deletion screening and point mutagenesis, we identified binding sites in the CCL2 promoter and candidate transcription factors responsible for its regulation by TGF-ß. Among these factors, only the knock-down of EGR1 and RXRA made CCL2 promoter activity independent of TGF-ß. These factors also demonstrated binding to the CCL2 promoter in a TGF-ß-dependent manner in a chromatin immunoprecipitation assay, and point mutations in the EGR1 and RXRA binding sites totally abolished the effect of TGF-ß. Our results highlight the key role of EGR1 and RXRA transcription factors in the regulation of CCL2 gene in response to TGF-ß pathway.


Subject(s)
Chemokine CCL2/metabolism , Early Growth Response Protein 1/metabolism , Retinoid X Receptor alpha/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Triple Negative Breast Neoplasms/metabolism , Base Sequence , Binding Sites , Cell Line, Tumor , Chemokine CCL2/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Models, Biological , Point Mutation/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
3.
Biochim Biophys Acta Mol Basis Dis ; 1866(3): 165626, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31785408

ABSTRACT

Toll-like receptor 4 (TLR4) is an innate immunity receptor predominantly expressed on myeloid cells and involved in the development of various diseases, many of them with complex genetics. Here we present data on functionality of single nucleotide polymorphism rs7873784 located in the 3'-untranslated region (3'-UTR) of TLR4 gene and associated with various pathologies involving chronic inflammation. We demonstrate that TLR4 3'-UTR strongly enhanced the activity of TLR4 promoter in U937 human monocytic cell line while minor rs7873784(C) allele created a binding site for transcription factor PU.1 (encoded by SPI1 gene), a known regulator of TLR4 expression. Increased binding of PU.1 further augmented the TLR4 transcription while PU.1 knockdown or complete disruption of the PU.1 binding site abrogated the effect. We hypothesize that additional functional PU.1 site may increase TLR4 expression in individuals carrying minor C variant of rs7873784 and modulate the development of certain pathologies, such as rheumatoid arthritis and type-2 diabetes mellitus.


Subject(s)
Arthritis, Rheumatoid/genetics , Diabetes Mellitus, Type 2/genetics , Genetic Predisposition to Disease/genetics , Polymorphism, Single Nucleotide/genetics , Proto-Oncogene Proteins/genetics , Toll-Like Receptor 4/genetics , Trans-Activators/genetics , 3' Untranslated Regions/genetics , Alleles , Cell Line , Cell Line, Tumor , HEK293 Cells , Humans , Promoter Regions, Genetic/genetics , U937 Cells
4.
Int J Mol Sci ; 19(12)2018 Dec 09.
Article in English | MEDLINE | ID: mdl-30544846

ABSTRACT

Interleukin 33 (IL-33) is a cytokine constitutively expressed by various cells of barrier tissues that contribute to the development of inflammatory immune responses. According to its function as an alarmin secreted by lung and airway epithelium, IL-33 plays a significant role in pathogenesis of allergic disorders. IL-33 is strongly involved in the pathogenesis of asthma, anaphylaxis, allergy and dermatitis, and genetic variations in IL33 locus are associated with increased susceptibility to asthma. Genome-wide association studies have identified risk "T" allele of the single-nucleotide polymorphism rs4742170 located in putative IL33 enhancer area as susceptible variant for development of specific wheezing phenotype in early childhood. Here, we demonstrate that risk "T" rs4742170 allele disrupts binding of glucocorticoid receptor (GR) transcription factor to IL33 putative enhancer. The IL33 promoter/enhancer constructs containing either 4742170 (T) allele or point mutations in the GR-binding site, were significantly more active and did not respond to cortisol in a pulmonary epithelial cell line. At the same time, the constructs containing rs4742170 (C) allele with a functional GR-binding site were less active and further inhibitable by cortisol. The latter effect was GR-dependent as it was completely abolished by GR-specific siRNA. This mechanism may explain the negative effect of the rs4742170 (T) risk allele on the development of wheezing phenotype that strongly correlates with allergic sensitization in childhood.


Subject(s)
Alleles , Enhancer Elements, Genetic/genetics , Interleukin-33/genetics , Introns/genetics , Polymorphism, Single Nucleotide/genetics , Receptors, Glucocorticoid/metabolism , Respiratory Sounds/genetics , Base Sequence , Binding Sites , Cell Line, Tumor , Child, Preschool , Humans , Hydrocortisone/pharmacology , Phenotype , Phosphorylation/drug effects , Promoter Regions, Genetic
5.
Int J Mol Sci ; 19(10)2018 Sep 25.
Article in English | MEDLINE | ID: mdl-30257479

ABSTRACT

Cytokine interleukin 33 (IL-33) is constitutively expressed by epithelial barrier cells, and promotes the development of humoral immune responses. Along with other proinflammatory mediators released by the epithelium of airways and lungs, it plays an important role in a number of respiratory pathologies. In particular, IL-33 significantly contributes to pathogenesis of allergy and asthma; genetic variations in the IL33 locus are associated with increased susceptibility to asthma. Large-scale genome-wide association studies have identified minor "G" allele of the single-nucleotide polymorphism rs928413, located in the IL33 promoter area, as a susceptible variant for early childhood and atopic asthma development. Here, we demonstrate that the rs928413(G) allele creates a binding site for the cAMP response element-binding protein 1 (CREB1) transcription factor. In a pulmonary epithelial cell line, activation of CREB1, presumably via the p38 mitogen-activated protein kinases (MAPK) cascade, activates the IL33 promoter containing the rs928413(G) allele specifically and in a CREB1-dependent manner. This mechanism may explain the negative effect of the rs928413 minor "G" allele on asthma development.


Subject(s)
Asthma/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Interleukin-33/genetics , Polymorphism, Single Nucleotide , Alleles , Asthma/metabolism , Cell Line, Tumor , Child , Epithelial Cells/metabolism , Genetic Predisposition to Disease , Humans , Lung/cytology , Lung/metabolism , MAP Kinase Signaling System , Promoter Regions, Genetic , Protein Binding , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...