Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Front Cell Dev Biol ; 10: 821875, 2022.
Article in English | MEDLINE | ID: mdl-35237602

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is one of the most lethal human cancers. Transforming Growth Factor Beta (TGF-ß) is a cytokine that switches from a tumor-suppressor at early stages to a tumor promoter in the late stages of tumor development, by yet unknown mechanisms. Tumor associated MUC1 is aberrantly glycosylated and overexpressed in >80% of PDAs and is associated with poor prognosis. MUC1 expression is found in the early stages of PDA development with subsequent increase in later stages. Analysis of human PDA samples from TCGA database showed significant differences in gene expression and survival profiles between low and high MUC1 samples. Further, high MUC1 expression was found to positively correlate to TGF-ßRII expression and negatively correlate to TGF-ßRI expression in PDA cell lines. We hypothesized that MUC1 overexpression induces TGF-ß mediated non-canonical signaling pathways which is known to be associated with poor prognosis. In this study, we report that MUC1 overexpression in PDA cells directly activates the JNK pathway in response to TGF-ß, and leads to increased cell viability via up-regulation and stabilization of c-Myc. Conversely, in low MUC1 expressing PDA cells, TGF-ß preserves its tumor-suppressive function and inhibits phosphorylation of JNK and stabilization of c-Myc. Knockdown of MUC1 in PDA cells also results in decreased phosphorylation of JNK and c-Myc in response to TGF-ß treatment. Taken together, the results indicate that overexpression of MUC1 plays a significant role in switching the TGF-ß function from a tumor-suppressor to a tumor promoter by directly activating JNK. Lastly, we report that high-MUC1 PDA tumors respond to TGF-ß neutralizing antibody in vivo showing significantly reduced tumor growth while low-MUC1 tumors do not respond to TGF-ß neutralizing antibody further confirming our hypothesis.

2.
Int J Mol Sci ; 22(11)2021 May 25.
Article in English | MEDLINE | ID: mdl-34070449

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are immature myeloid cells that are responsible for immunosuppression in tumor microenvironment. Here we report the impact of mucin 1 (MUC1), a transmembrane glycoprotein, on proliferation and functional activity of MDSCs. To determine the role of MUC1 in MDSC phenotype, we analyzed MDSCs derived from wild type (WT) and MUC1-knockout (MUC1KO) mice bearing syngeneic pancreatic (KCKO) or breast (C57MG) tumors. We observed enhanced tumor growth of pancreatic and breast tumors in the MUC1KO mice compared to the WT mice. Enhanced tumor growth in the MUC1KO mice was associated with increased numbers of suppressive MDSCs and T regulatory (Tregs) cells in the tumor microenvironment. Compared to the WT host, MUC1KO host showed higher levels of iNOS, ARG1, and TGF-ß, thus promoting proliferation of MDSCs with an immature and immune suppressive phenotype. When co-cultured with effector T cells, MDSCs from MUC1KO mice led to higher repression of IL-2 and IFN-γ production by T cells as compared to MDSCs from WT mice. Lastly, MDSCs from MUC1KO mice showed higher levels of c-Myc and activated pSTAT3 as compared to MDSCs from WT mice, suggesting increased survival, proliferation, and prevention of maturation of MDSCs in the MUC1KO host. We report diminished T cell function in the KO versus WT mice. In summary, the data suggest that MUC1 may regulate signaling pathways that are critical to maintain the immunosuppressive properties of MDSCs.


Subject(s)
Breast Neoplasms/metabolism , Mucin-1/metabolism , Myeloid-Derived Suppressor Cells/immunology , Pancreatic Neoplasms/metabolism , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/immunology , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Coculture Techniques , Disease Models, Animal , Female , Interferon-gamma/metabolism , Interleukin-2/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucin-1/genetics , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins c-myc/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Spleen/cytology , Spleen/metabolism , Transforming Growth Factor beta/blood , Tumor Microenvironment/genetics
3.
PLoS One ; 14(11): e0224309, 2019.
Article in English | MEDLINE | ID: mdl-31693710

ABSTRACT

In recent years, vaccines against tumor antigens have shown potential for combating invasive cancers, including primary tumors and metastatic lesions. This is particularly pertinent for breast cancer, which is the second-leading cause of cancer-related death in women. MUC1 is a glycoprotein that is normally expressed on glandular epithelium, but is overexpressed and under-glycosylated in most human cancers, including the majority of breast cancers. This under-glycosylation exposes the MUC1 protein core on the tumor-associated form of the protein. We have previously shown that a vaccine consisting of MUC1 core peptides stimulates a tumor-specific immune response. However, this immune response is dampened by the immunosuppressive microenvironment within breast tumors. Thus, in the present study, we investigated the effectiveness of MUC1 vaccination in combination with four different drugs that inhibit different components of the COX pathway: indomethacin (COX-1 and COX-2 inhibitor), celecoxib (COX-2 inhibitor), 1-methyl tryptophan (indoleamine 2,3 dioxygenase inhibitor), and AH6809 (prostaglandin E2 receptor antagonist). These treatment regimens were explored for the treatment of orthotopic MUC1-expressing breast tumors in mice transgenic for human MUC1. We found that the combination of vaccine and indomethacin resulted in a significant reduction in tumor burden. Indomethacin did not increase tumor-specific immune responses over vaccine alone, but rather appeared to reduce the proliferation and increase apoptosis of tumor cells, thus rendering them susceptible to immune cell killing.


Subject(s)
Breast Neoplasms/therapy , Cancer Vaccines/administration & dosage , Cyclooxygenase Inhibitors/administration & dosage , Indomethacin/administration & dosage , Mammary Neoplasms, Experimental/therapy , Mucin-1/immunology , Animals , Breast Neoplasms/immunology , Cancer Vaccines/immunology , Combined Modality Therapy/methods , Female , Humans , Immunogenicity, Vaccine/drug effects , Immunogenicity, Vaccine/immunology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/immunology , Mice , Mice, Transgenic , Mucin-1/genetics , Tumor Burden/drug effects , Tumor Burden/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
4.
Oncotarget ; 9(6): 6897-6910, 2018 Jan 23.
Article in English | MEDLINE | ID: mdl-29467938

ABSTRACT

Pancreatic Ductal Adenocarcinoma (PDA) has a mortality rate that nearly matches its incidence rate. Transforming Growth Factor Beta (TGF-ß) is a cytokine with a dual role in tumor development switching from a tumor suppressor to a tumor promoter. There is limited knowledge of how TGF-ß function switches during tumorigenesis. Mucin 1 (MUC1) is an aberrantly glycosylated, membrane-bound, glycoprotein that is overexpressed in >80% of PDA cases and is associated with poor prognosis. In PDA, MUC1 promotes tumor progression and metastasis via signaling through its cytoplasmic tail (MUC1-CT) and interacting with other oncogenic signaling molecules. We hypothesize that high levels of MUC1 in PDA may be partly responsible for the TGF-ß functional switch during oncogenesis. We report that overexpression of MUC1 in BxPC3 human PDA cells (BxPC3.MUC1) enhances the induction of epithelial to mesenchymal transition leading to increased invasiveness in response to exogenous TGF-ß1. Simultaneously, these cells resist TGF-ß induced apoptosis by downregulating levels of cleaved caspases. We show that mutating the tyrosines in MUC1-CT to phenylalanine reverses the TGF-ß induced invasiveness. This suggests that the tyrosine residues in MUC1-CT are required for TGF-ß induced invasion. Some of these tyrosines are phosphorylated by the tyrosine kinase c-Src. Thus, treatment of BxPC3.MUC1 cells with a c-Src inhibitor (PP2) significantly reduces TGF-ß induced invasiveness. Similar observations were confirmed in the Chinese hamster ovarian (CHO) cell line. Data strongly suggests that MUC1 may regulate TGF-ß function in PDA cells and thus have potential clinical relevance in the use of TGF-ß inhibitors in clinical trials.

5.
Methods Mol Biol ; 1428: 231-43, 2016.
Article in English | MEDLINE | ID: mdl-27236803

ABSTRACT

Dendritic cell (DC)-based vaccines are commonly used for cancer immunotherapy. To prepare vaccines, DCs are pulsed or transfected with either: (a) defined peptides of tumor-associated antigens, (b) total protein isolated from the tumor cell, (c) autologous total RNA isolated from the tumor cell, (d) synthetic tumor-antigen-encoding mRNA, or (e) genes that encode for specific tumor-associated antigens. Introduction of tumor-associated antigen(s) and subsequent generation of mature DCs that can stimulate tumor-antigen-specific cytotoxic T lymphocytes comprise the critical steps of cancer vaccine preparation. Here, we described a method of: (a) preparing and delivering synthetic FOXP3 mRNA into human DCs, (b) generating mature DCs,


Subject(s)
Dendritic Cells/cytology , Forkhead Transcription Factors/genetics , Inflammatory Breast Neoplasms/immunology , RNA, Messenger/immunology , T-Lymphocytes, Cytotoxic/cytology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Cell Line, Tumor , Cell- and Tissue-Based Therapy , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/metabolism , Electroporation , Female , Forkhead Transcription Factors/immunology , Humans , Immunotherapy , Inflammatory Breast Neoplasms/genetics , Inflammatory Breast Neoplasms/therapy , T-Lymphocytes, Cytotoxic/metabolism , Transfection
6.
Pharmacol Ther ; 163: 94-108, 2016 07.
Article in English | MEDLINE | ID: mdl-27063403

ABSTRACT

Cancer cells propagated in three-dimensional (3D) culture systems exhibit physiologically relevant cell-cell and cell-matrix interactions, gene expression and signaling pathway profiles, heterogeneity and structural complexity that reflect in vivo tumors. In recent years, development of various 3D models has improved the study of host-tumor interaction and use of high-throughput screening platforms for anti-cancer drug discovery and development. This review attempts to summarize the various 3D culture systems, with an emphasis on the most well characterized and widely applied model - multicellular tumor spheroids. This review also highlights the various techniques to generate tumor spheroids, methods to characterize them, and its applicability in cancer research.


Subject(s)
Cell Communication/physiology , Cell Culture Techniques/methods , Neoplasms/physiopathology , Spheroids, Cellular/physiology , Antineoplastic Agents/pharmacology , Cell Movement/physiology , Flow Cytometry , Humans , Hydrogels/chemistry , Microscopy, Confocal , Microscopy, Electron, Scanning , Neoplasm Invasiveness/physiopathology , Tumor Cells, Cultured , Tumor Microenvironment/physiology
7.
Pancreas ; 44(6): 909-17, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26035123

ABSTRACT

OBJECTIVE: Eighty percent of pancreatic ductal adenocarcinomas (PDAs) overexpress mucin 1 (MUC1), a transmembrane mucin glycoprotein. MUC1(high) PDA patients also express high levels of cyclooxygenase 2 (COX-2) and show poor prognosis. The cytoplasmic tail of MUC1 (MUC1-CT) partakes in oncogenic signaling, resulting in accelerated cancer progression. Our aim was to understand the regulation of Cox-2 expression by MUC1. METHODS: Levels of COX-2 and MUC1 were determined in MUC1(-/-), MUC1(low), and MUC1(high) PDA cells and tumors using reverse transcriptase-polymerase chain reaction, Western blot, and immunohistochemistry. Proliferative and invasive potential was assessed using MTT and Boyden chamber assays. Chromatin immunoprecipitation was performed to evaluate binding of MUC1-CT to the promoter of COX-2 gene. RESULTS: Significantly higher levels of COX-2 mRNA and protein were detected in MUC1(high) versus MUC1(low/null) cells, which were recapitulated in vivo. In addition, deletion of MUC1 gene and transient knockdown of MUC1 led to decreased COX-2 level. Also, MUC1-CT associated with the COX-2 promoter at ∼1000 base pairs upstream of the transcription start site, the same gene locus where nuclear factor κB p65 associates with the COX-2 promoter. CONCLUSIONS: Data supports a novel regulation of COX-2 gene by MUC1 in PDA, the intervention of which may lead to a better therapeutic targeting in PDA patients.


Subject(s)
Carcinoma, Pancreatic Ductal/enzymology , Cyclooxygenase 2/metabolism , Mucin-1/metabolism , Pancreatic Neoplasms/enzymology , Animals , Antineoplastic Agents/pharmacology , Binding Sites , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation/drug effects , Cyclooxygenase 2/genetics , Cyclooxygenase 2 Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Mice, Nude , Mucin-1/genetics , Neoplasm Invasiveness , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Promoter Regions, Genetic , RNA Interference , RNA, Messenger/metabolism , Transcription Factor RelA/metabolism , Transfection , Xenograft Model Antitumor Assays
8.
Front Immunol ; 5: 67, 2014.
Article in English | MEDLINE | ID: mdl-24605110

ABSTRACT

Mucin 1 (MUC1) is a transmembrane mucin glycoprotein that is over-expressed and aberrantly glycosylated in >80% of human pancreatic ductal adenocarcinoma (PDA) and is associated with poor prognosis. To understand the role of MUC1 in PDA, we have recently developed two mouse models of spontaneous PDA, one that expresses full-length human MUC1 transgene (KCM mice) and one that is null for MUC1 (KCKO mice). We have previously reported that KCM mice express high levels of myeloid derived suppressor cells (MDSCs) in their tumors and develop highly aggressive PDA. To further understand the underlying mechanism for high MDSC levels in KCM-tumors, we generated primary cell lines from KCM and KCKO-tumors. In this study, we report that MDSCs derived using KCM cells express significantly higher levels of arginase 1 and inducible nitric oxide synthase (markers associated with immune suppression) and lower levels of CD115 (a marker associated with maturation of myeloid cells) as compared to KCKO-derived MDSCs. Functionally, KCM-derived MDSCs secrete significantly higher levels of urea and nitric oxide (NO) when co-cultured with normal splenic cells as compared to KCKO-derived MDSCs. Data indicates that KCM-derived MDSCs remain immature and are more suppressive as compared to KCKO-derived MDSCs. This was further corroborated in vivo where MDSCs isolated from KCM-tumor-bearing mice retained their immature state and were highly suppressive as compared to MDSCs derived from KCKO-tumor-bearing mice. Finally, we show that KCM cells secrete significantly higher levels of prostaglandin E2 (PGE2), a COX-2 metabolite and a known driver of suppressive MDSCs as compared to KCKO cells. Thus, inhibiting PGE2 with a specific COX-2 inhibitor reverses the immunosuppressive and immature phenotype of KCM-derived MDSCs. This is the first report that clearly suggests a functional role of pancreatic tumor-associated MUC1 in the development of functional MDSCs.

9.
Trends Mol Med ; 20(6): 332-42, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24667139

ABSTRACT

The transmembrane glycoprotein Mucin 1 (MUC1) is aberrantly glycosylated and overexpressed in a variety of epithelial cancers, and plays a crucial role in progression of the disease. Tumor-associated MUC1 differs from the MUC1 expressed in normal cells with regard to its biochemical features, cellular distribution, and function. In cancer cells, MUC1 participates in intracellular signal transduction pathways and regulates the expression of its target genes at both the transcriptional and post-transcriptional levels. This review highlights the structural and functional differences that exist between normal and tumor-associated MUC1. We also discuss the recent advances made in the use of MUC1 as a biomarker and therapeutic target for cancer.


Subject(s)
Antigens, Neoplasm/metabolism , Mucin-1/metabolism , Cell Proliferation , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Humans , Mucin-1/chemistry , Mucin-1/genetics , Neoplastic Stem Cells/metabolism , Signal Transduction
10.
Biol Open ; 2(1): 1-9, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23336071

ABSTRACT

The abundance of Myc protein must be exquisitely controlled to avoid growth abnormalities caused by too much or too little Myc. An intriguing mode of regulation exists in which Myc protein itself leads to reduction in its abundance. We show here that dMyc binds to the miR-308 locus and increases its expression. Using our gain-of-function approach, we show that an increase in miR-308 causes a destabilization of dMyc mRNA and reduced dMyc protein levels. In vivo knockdown of miR-308 confirmed the regulation of dMyc levels in embryos. This regulatory loop is crucial for maintaining appropriate dMyc levels and normal development. Perturbation of the loop, either by elevated miR-308 or elevated dMyc, caused lethality. Combining elevated levels of both, therefore restoring balance between miR-308 and dMyc levels, resulted in lower apoptotic activity and suppression of lethality. These results reveal a sensitive feedback mechanism that is crucial to prevent the pathologies caused by abnormal levels of dMyc.

SELECTION OF CITATIONS
SEARCH DETAIL
...