Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Oncogene ; 41(48): 5147-5159, 2022 11.
Article in English | MEDLINE | ID: mdl-36271032

ABSTRACT

MUC16, membrane-bound mucin, plays an oncogenic role in pancreatic ductal adenocarcinoma (PDAC). However, the pathological role of MUC16 in the PDAC progression, tumor microenvironment, and metastasis in cooperation with KrasG12D and Trp53R172H mutations remains unknown. Deletion of Muc16 with activating mutations KrasG12D/+ and Trp53R172H/+ in mice significantly decreased progression and prolonged overall survival in KrasG12D/+; Trp53R172H/+; Pdx-1-Cre; Muc16-/- (KPCM) and KrasG12D/+; Pdx-1-Cre; Muc16-/- (KCM), as compared to KrasG12D/+; Trp53R172H/+; Pdx-1-Cre (KPC) and KrasG12D/+; Pdx-1-Cre (KC) mice, respectively. Muc16 knockout pancreatic tumor (KPCM) displays decreased tumor microenvironment factors and significantly reduced incidence of liver and lung metastasis compared to KPC. Furthermore, in silico data analysis showed a positive correlation of MUC16 with activated stroma and metastasis-associated genes. KPCM mouse syngeneic cells had significantly lower metastatic and endothelial cell binding abilities than KPC cells. Similarly, KPCM organoids significantly decreased the growth rate compared to KPC organoids. Interestingly, RNA-seq data revealed that the cytoskeletal proteins Actg2, Myh11, and Pdlim3 were downregulated in KPCM tumors. Further knockdown of these genes showed reduced metastatic potential. Overall, our results demonstrate that Muc16 alters the tumor microenvironment factors during pancreatic cancer progression and metastasis by changing the expression of Actg2, Myh11, and Pdlim3 genes.


Subject(s)
Carcinoma, Pancreatic Ductal , Mucins , Pancreatic Neoplasms , Animals , Mice , Carcinogenesis , Carcinoma, Pancreatic Ductal/pathology , Mucins/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Tumor Microenvironment/genetics , Pancreatic Neoplasms
2.
Analyst ; 146(16): 5150-5159, 2021 Aug 09.
Article in English | MEDLINE | ID: mdl-34286712

ABSTRACT

Label-free radiation pressure force analysis using a microfluidic platform is applied to the differential detection of innate immune cell activation. Murine-derived peritoneal macrophages (IC-21) are used as a model system and the activation of IC-21 cells by lipopolysaccharide (LPS) and interferon gamma (IFN-γ) to M1 pro-inflammatory phenotype is confirmed by RNA gene sequencing and nitric oxide production. The mean cell size determined by radiation pressure force analysis increases slightly after the activation (4 to 6%) and the calculated percentage of population overlaps between the control and the activated group after 14 and 24 h stimulations are at 79% and 77%. Meanwhile the mean cell velocity decreases more significantly after the activation (14% to 15%) and the calculated percentage of population overlaps between the control and the activated group after 14 and 24 h stimulations are only at 14% and 13%. The results demonstrate that the majority of the activated cells acquire a lower velocity than the cells from the control group without changes in cell size. For comparison label-free flow cytometry analysis of living IC-21 cells under the same stimulation conditions are performed and the results show population shifts towards larger values in both forward scatter and side scatter, but the calculated percentage of population overlaps in all case are significant (70% to 83%). Cell images obtained during radiation pressure force analysis by a CCD camera, and by optical microscopy and atomic force microscopy (AFM) reveal correlations between the cell activation by LPS/IFN-γ, the increase in cell complexity and surface roughness, and enhanced back scattered light by the activated cells. The unique relationship predicted by Mie's theory between the radiation pressure force exerted on the cell and the angular distribution of the scattered light by the cell which is influenced by its size, complexity, and surface conditions, endows the cell velocity based measurement by radiation pressure force analysis with high sensitivity in differentiating immune cell activation.


Subject(s)
Lipopolysaccharides , Macrophages, Peritoneal , Animals , Interferon-gamma , Lipopolysaccharides/toxicity , Mice , Microscopy, Atomic Force , Nitric Oxide
3.
Mater Sci Eng C Mater Biol Appl ; 120: 111685, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33545847

ABSTRACT

We present a low-cost, easy-to-implement platform for printing materials and interfacing them with eukaryotic cells. We show that thermal or chemical reduction of a graphene oxide thin film allows water-assisted delamination of the film from glass or plastic. The chemical and physical properties and permeability of the resulting film are dependent on the method of reduction and deposition of the graphene oxide, with thermal reduction removing more oxidized carbon functionality than chemical reduction. We also developed a method to attach the films onto cell surfaces using a thin layer of gelatin as an adhesive. In general, the films are highly impermeable to nutrients and we observed a significant amount of cell death when gelatin was not used; gelatin enables diffusion of nutrients for sustained cell viability. The combination of nanoscale membranes with a low melting point biopolymer allows us to reversibly interface cells with cargo transferred by graphene oxide while maintaining cell viability. To demonstrate delivery of electronic structures, we modified a commercial off-the-shelf printer to print a silver-based ink directly onto the reduced graphene oxide films which we then transferred to the surface of the cells.


Subject(s)
Gelatin , Graphite , Electronics
4.
Biol Open ; 9(9)2020 09 07.
Article in English | MEDLINE | ID: mdl-32709695

ABSTRACT

Pancreatic cancer (PC) is acquired postnatally; to mimic this scenario, we developed an inducible KrasG12D; Ptf1a-CreER™ (iKC) mouse model, in which Kras is activated postnatally at week 16 upon tamoxifen (TAM) administration. Upon TAM treatment, iKC mice develop pancreatic intraepithelial neoplasia (PanIN) lesions and PC with metastasis at the fourth and fortieth weeks, respectively, and exhibited acinar-to-ductal metaplasia (ADM) and transdifferentiation. Kras activation upregulated the transcription factors Ncoa3, p-cJun and FoxM1, which in turn upregulated expression of transmembrane mucins (Muc1, Muc4 and Muc16) and secretory mucin (Muc5Ac). Interestingly, knockdown of KrasG12D in multiple PC cell lines resulted in downregulation of MUC1, MUC4, MUC5AC and MUC16. In addition, iKC mice exhibited ADM and transdifferentiation. Our results show that the iKC mouse more closely mimics human PC development and can be used to investigate pancreatic ductal adenocarcinoma (PDAC) biomarkers, early onset of PDAC, and ADM. The iKC model can also be used for preclinical strategies such as targeting mucin axis alone or in combination with neo-adjuvant, immunotherapeutic approaches and to monitor chemotherapy response.


Subject(s)
Carcinoma, Pancreatic Ductal/etiology , Carcinoma, Pancreatic Ductal/pathology , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/genetics , Mucins/genetics , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/pathology , Tamoxifen/adverse effects , Animals , Biomarkers , Carcinoma, Pancreatic Ductal/metabolism , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Disease Progression , Disease Susceptibility , Gene Expression , Immunohistochemistry , Mice , Mucins/metabolism , Multigene Family , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Transcription Factors
5.
ACS Omega ; 4(7): 12938-12947, 2019 Jul 31.
Article in English | MEDLINE | ID: mdl-31460420

ABSTRACT

Flow-through optical chromatography (FT-OC), an advanced mode of optical chromatography, achieved baseline separation of a mixture of silica microparticles (SiO2, 1.00 and 2.50 µm) and a mixture of polystyrene microparticles (PS, 1.00, 2.00, and 3.00 µm) based on particle size. Comparisons made between experimentally determined velocities for the microparticles and theoretically derived velocities from Mie theory and Stokes' law validated the data collection setup and the data analysis for FT-OC. A population shift in live macrophages (cell line IC-21, ATCC TIB-186) responding to environmental stimuli was sensitively detected by FT-OC. The average velocity of macrophages stressed by nutritional deprivation was decreased considerably together with a small but statistically significant increase in cell size. Mie scattering calculations demonstrated that the small increase in cell size of macrophages stressed by nutritional deprivation was not entirely responsible for this decrease. Confocal fluorescence microscopy and atomic force microscopy (AFM) studies revealed morphological changes of macrophages induced by nutritional deprivation, and these changes were more likely responsible for the decrease in average velocity detected by FT-OC. Confocal Raman microspectroscopy was used to shed light upon biochemical transformations of macrophages suffering from nutritional deprivation.

6.
F1000Res ; 6: 1834, 2017.
Article in English | MEDLINE | ID: mdl-30109018

ABSTRACT

Background: Shipwrecks serve as a rich source for novel microbial populations that have largely remained undiscovered. Low temperatures, lack of sunlight, and the availability of substrates derived from the shipwreck's hull and cargo may provide an environment in which microbes can develop unique metabolic adaptations.   Methods: To test our hypothesis that shipwrecks could influence the microbial population involved in denitrification when a consortium is grown in the laboratory, we collected samples proximate to two steel shipwrecks in the northern Gulf of Mexico. Then under laboratory conditions, we grew two independent denitrifying microbial consortia. Each consortium was grown by using the BART assay system and analyzed based on growth kinetics, ion chromatography and 16S amplicon sequencing. Results: Both denitrifying consortia were different from each other based on varied growth profiles, rates of nitrate utilization and 16S amplicon sequencing. Conclusions: Our observations conclude that the laboratory grown water column microbial consortia from deep-sea shipwrecks in the Gulf of Mexico are able to undergo aggressive denitrification.

7.
Genes Cancer ; 7(3-4): 110-124, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27382435

ABSTRACT

MUC16, a heavily glycosylated type-I transmembrane mucin is overexpressed in several cancers including pancreatic ductal adenocarcinoma (PDAC). Previously, we have shown that MUC16 is significantly overexpressed in human PDAC tissues. However, the functional consequences and its role in PDAC is poorly understood. Here, we show that MUC16 knockdown decreases PDAC cell proliferation, colony formation and migration in vitro. Also, MUC16 knockdown decreases the tumor formation and metastasis in orthotopic xenograft mouse model. Mechanistically, immunoprecipitation and immunofluorescence analyses confirms MUC16 interaction with galectin-3 and mesothelin in PDAC cells. Adhesion assay displayed decreased cell attachment of MUC16 knockdown cells with recombinant galectin-1 and galectin-3 protein. Further, CRISPR/Cas9-mediated MUC16 knockout cells show decreased tumor-associated carbohydrate antigens (T and Tn) in PDAC cells. Importantly, carbohydrate antigens were decreased in the region that corresponds to MUC16 and suggests for the decreased MUC16-galectin interactions. Co-immunoprecipitation also revealed a novel interaction between MUC16 and FAK in PDAC cells. Interestingly, we observed decreased expression of mesenchymal and increased expression of epithelial markers in MUC16-silenced cells. Additionally, MUC16 loss showed a decreased FAK-mediated Akt and ERK/MAPK activation. Altogether, these findings suggest that MUC16-focal adhesion signaling may play a critical role in facilitating PDAC growth and metastasis.

8.
Oncotarget ; 6(25): 21085-99, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26035354

ABSTRACT

Several studies have demonstrated that MUC4 is involved in progression and metastasis of pancreatic cancer (PC). Here, we report that HER3/MUC4 interaction in HER2 low cells is critical in driving pancreatic tumorigenesis. Upon HER2 knockdown, we observed elevated expression of HER3 and MUC4 and their interactions, which was confirmed by immunoprecipitation and bioinformatics analyses. In paired human PC tissues, higher percentage of HER3 positivity (10/33, 30.3%; p = 0.001) was observed than HER2 (5/33, 15.1%; p = 0.031), which was further confirmed in spontaneous mice (KPC; KrasG12D; Trp53R172H/+; Pdx-Cre) tumors of different weeks. Mechanistically, increased phosphorylation of ERK and expression of PI3K and c-Myc were observed in HER2 knockdown cells, suggesting a positive role for HER3/MUC4 in HER2 low cells. Further, HER2 knockdown resulted in increased proliferation, motility and tumorigenicity of PC cells. Consistently, transient knockdown of HER3 by siRNA in HER2 knockdown cells led to decreased proliferation. These observations led us to conclude that HER3 interacts with MUC4 to promote proliferation in HER2 low PC cells. Further, deficiency of both HER2 and HER3 leads to decreased proliferation of PC cells. Hence targeting these newly identified HER3/MUC4 signals would improve the PC patients survival by intercepting MUC4 mediated oncogenic signaling.


Subject(s)
Gene Expression Regulation, Neoplastic , Mucin-4/metabolism , Pancreatic Neoplasms/pathology , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Amino Acid Sequence , Animals , Carcinogenesis , Cell Cycle , Cell Proliferation , Disease Progression , Female , Humans , Mice , Mice, Nude , Molecular Sequence Data , Neoplasm Metastasis , Neoplasm Transplantation , Pancreatic Neoplasms/metabolism , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-myc/metabolism , Sequence Homology, Amino Acid , Signal Transduction
9.
Oncotarget ; 6(22): 19118-31, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26046375

ABSTRACT

MUC16, a transmembrane mucin, facilitates pancreatic adenocarcinoma progression and metastasis. In the current studies, we observed that MUC16 knockdown pancreatic cancer cells exhibit reduced glucose uptake and lactate secretion along with reduced migration and invasion potential, which can be restored by supplementing the culture media with lactate, an end product of aerobic glycolysis. MUC16 knockdown leads to inhibition of mTOR activity and reduced expression of its downstream target c-MYC, a key player in cellular growth, proliferation and metabolism. Ectopic expression of c-MYC in MUC16 knockdown pancreatic cancer cells restores the altered cellular physiology. Our LC-MS/MS based metabolomics studies indicate global metabolic alterations in MUC16 knockdown pancreatic cancer cells, as compared to the controls. Specifically, glycolytic and nucleotide metabolite pools were significantly decreased. We observed similar metabolic alterations that correlated with MUC16 expression in primary tumor tissue specimens from human pancreatic adenocarcinoma cancer patients. Overall, our results demonstrate that MUC16 plays an important role in metabolic reprogramming of pancreatic cancer cells by increasing glycolysis and enhancing motility and invasiveness.


Subject(s)
CA-125 Antigen/metabolism , Membrane Proteins/metabolism , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/metabolism , TOR Serine-Threonine Kinases/metabolism , CA-125 Antigen/genetics , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Gene Knockdown Techniques , Glucose/metabolism , Humans , Lactic Acid/metabolism , Membrane Proteins/genetics , Neoplasm Invasiveness , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , TOR Serine-Threonine Kinases/genetics , Transfection
10.
Oncotarget ; 6(7): 5164-81, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25686822

ABSTRACT

Transmembrane proteins MUC4, EGFR and HER2 are shown to be critical in invasion and metastasis of pancreatic cancer. Besides, we and others have demonstrated de novo expression of MUC4 in ~70-90% of pancreatic cancer patients and its stabilizing effects on HER2 downstream signaling in pancreatic cancer. Here, we found that use of canertinib or afatinib resulted in reduction of MUC4 and abrogation of in vitro and in vivo oncogenic functions of MUC4 in pancreatic cancer cells. Notably, silencing of EGFR family member in pancreatic cancer cells decreased MUC4 expression through reduced phospho-STAT1. Furthermore, canertinib and afatinib treatment also inhibited proliferation, migration and survival of pancreatic cancer cells by attenuation of signaling events including pERK1/2 (T202/Y204), cyclin D1, cyclin A, pFAK (Y925) and pAKT (Ser473). Using in vivo bioluminescent imaging, we demonstrated that canertinib treatment significantly reduced tumor burden (P=0.0164) and metastasis to various organs. Further, reduced expression of MUC4 and EGFR family members were confirmed in xenografts. Our results for the first time demonstrated the targeting of EGFR family members along with MUC4 by using pan-EGFR inhibitors. In conclusion, our studies will enhance the translational acquaintance of pan-EGFR inhibitors for combinational therapies to combat against lethal pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/prevention & control , Cell Movement/drug effects , ErbB Receptors/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Mucin-4/antagonists & inhibitors , Pancreatic Neoplasms/prevention & control , STAT1 Transcription Factor/antagonists & inhibitors , Afatinib , Animals , Apoptosis/drug effects , Blotting, Western , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/secondary , Cell Proliferation/drug effects , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Mice , Mice, Nude , Morpholines/pharmacology , Mucin-4/genetics , Mucin-4/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Quinazolines/pharmacology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Oncotarget ; 6(4): 2064-75, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25576918

ABSTRACT

Amyloid precursor-like protein 2 (APLP2) is aberrantly expressed in pancreatic cancer. Here we showed that APLP2 is increased in pancreatic cancer metastases, particularly in metastatic lesions found in the diaphragm and intestine. Examination of matched human primary tumor-liver metastasis pairs showed that 38.1% of the patients had positive APLP2 expression in both the primary tumor and the corresponding liver metastasis. Stable knock-down of APLP2 expression (with inducible shRNA) in pancreatic cancer cells reduced the ability of these cells to migrate and invade. Loss of APLP2 decreased cortical actin and increased intracellular actin filaments in pancreatic cancer cells. Down-regulation of APLP2 decreased the weight and metastasis of orthotopically transplanted pancreatic tumors in nude mice.


Subject(s)
Actin Cytoskeleton/metabolism , Amyloid beta-Protein Precursor/metabolism , Cell Proliferation , Nerve Tissue Proteins/metabolism , Pancreatic Neoplasms/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Anti-Bacterial Agents/pharmacology , Cell Line, Tumor , Cell Movement , Down-Regulation/drug effects , Doxycycline/pharmacology , Female , Flow Cytometry , Humans , Immunoblotting , Immunohistochemistry , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice, Nude , Nerve Tissue Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , RNA Interference , Tumor Burden/drug effects , Tumor Burden/genetics , Xenograft Model Antitumor Assays
12.
Adv Drug Deliv Rev ; 81: 16-33, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25453266

ABSTRACT

Despite considerable progress being made in understanding pancreatic cancer (PC) pathogenesis, it still remains the 10th most often diagnosed malignancy in the world and 4th leading cause of cancer related deaths in the United States with a five year survival rate of only 6%. The aggressive nature, lack of early diagnostic and prognostic markers, late clinical presentation, and limited efficacy of existing treatment regimens make PC a lethal cancer with high mortality and poor prognosis. Therefore, novel reliable biomarkers and molecular targets are urgently needed to combat this deadly disease. MicroRNAs (miRNAs) are short (19-24 nucleotides) non-coding RNA molecules implicated in the regulation of gene expression at post-transcriptional level and play significant roles in various physiological and pathological conditions. Aberrant expression of miRNAs has been reported in several cancers including PC and is implicated in PC pathogenesis and progression, suggesting their utility in diagnosis, prognosis and therapy. In this review, we summarize the role of several miRNAs that regulate various oncogenes (KRAS) and tumor suppressor genes (p53, p16, SMAD4, etc.) involved in PC development, their prospective roles as diagnostic and prognostic markers and as a therapeutic targets.


Subject(s)
Biomarkers, Tumor/genetics , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Animals , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Molecular Targeted Therapy , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/therapy , Prognosis , Survival Rate
13.
J Thorac Oncol ; 10(1): 19-27, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25319180

ABSTRACT

Aberrant expression of mucins is associated with cancer development and metastasis. An overexpression of few mucins contributes to oncogenesis by enhancing cancer cell growth and providing constitutive survival signals. This review focuses on the importance of mucins both in the normal bronchial epithelial cells and the malignant tumors of the lung and their contribution in the diagnosis and prognosis of lung cancer patients. During lung cancer progression, mucins either alone or through their interaction with many receptor tyrosine kinases mediate cell signals for growth and survival of cancer cells. Also, stage-specific expression of certain mucins, like MUC1, is associated with poor prognosis from lung cancer. Thus, mucins are emerging as attractive targets for developing novel therapeutic approaches for lung cancer. Several strategies targeting mucin expression and function are currently being investigated to control lung cancer progression.


Subject(s)
Lung Neoplasms/diagnosis , Lung Neoplasms/therapy , Mucins/metabolism , Disease Progression , Humans , Immunohistochemistry , Lung Neoplasms/metabolism , Prognosis
14.
FASEB J ; 28(10): 4183-99, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25002120

ABSTRACT

MUC16 is a high-molecular-weight glycoprotein that is expressed by the various epithelial cell surfaces of the human body to protect the cell layer from a myriad of insults. It is the largest mucin known to date, with an ∼22,152 aa sequence. Structurally, MUC16 is characterized into 3 distinct domains: the amino terminal, the tandem repeat, and the carboxyl terminal domain, with each domain having unique attributes. The extracellular portion of MUC16 is shed into the bloodstream and serves as a biomarker for diagnosing and monitoring patients with cancer; however, its functional role in cancer is yet to be elucidated. Several factors contribute to this challenge, which include the large protein size; the extensive glycosylation that the protein undergoes, which confers functional heterogeneity; lack of specific antibodies that detect the unique domains of MUC16; and the existence of splicing variants. Despite these limitations, MUC16 has been established as a molecule of significant application in cancer. Hence, in this review, we discuss the various aspects of MUC16, which include its discovery, structure, and biological significance both in benign and malignant conditions with an attempt to dissect its functional relevance


Subject(s)
CA-125 Antigen/metabolism , Membrane Proteins/metabolism , Neoplasms/metabolism , Animals , CA-125 Antigen/genetics , Epithelial Cells/metabolism , Humans , Membrane Proteins/genetics , Neoplasms/diagnosis
15.
Arch Pathol Lab Med ; 137(4): 546-51, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23544943

ABSTRACT

CONTEXT: Diagnoses rendered as atypical/suspicious for malignancy on fine-needle aspiration (FNA) of pancreatic mass lesions range from 2% to 29% in various studies. We have identified the expression of 3 genes, MUC4, MUC16, and NGAL that are highly upregulated in pancreatic adenocarcinoma. In this study, we analyzed the expression of these markers in FNA samples to determine whether they could improve sensitivity and specificity. OBJECTIVE: To evaluate the utility of MUC4, MUC16, and NGAL in the evaluation of pancreatic FNA specimens. DESIGN: Records of pancreatic FNAs performed during 10 consecutive years were reviewed. Unstained sections from corresponding cell blocks were immunostained for MUC4, MUC16, and NGAL (polyclonal). Immunostaining was assessed using the H-score (range, 0-3). Any case with an H-score of >0.5 was considered positive. RESULTS: Cases were classified using cytomorphologic criteria as adenocarcinoma (31 of 64; 48.4%), benign (17 of 64; 26.6%), and atypical/suspicious (16 of 64; 25%). On follow-up, all cases (100%; 31 of 31) diagnosed as carcinoma on cytology were confirmed on biopsy/resection samples or by clinical follow-up (such as unresectable disease). Of the cases diagnosed as atypical/suspicious, 69% (11 of 16) were found to be positive for adenocarcinoma and 31% (5 of 16) were benign on subsequent follow-up. Overall sensitivity and specificity, respectively, for the various markers for the detection of pancreatic adenocarcinoma were as follows: MUC4 (74% and 100%), MUC16 (62.9% and 100%), and NGAL (61.3% and 58.8%). In cases that were atypical/suspicious on cytology, expression of MUC4 and MUC16 was 100% specific for carcinoma with sensitivities of 63.6% and 66.7%, respectively. CONCLUSION: Immunocytochemistry for MUC4 and MUC16 appears to be a useful adjunct in the classification of pancreatic FNA samples, especially in cases that are equivocal (atypical/suspicious) for adenocarcinoma on cytomorphologic assessment.


Subject(s)
Adenocarcinoma/pathology , CA-125 Antigen/metabolism , Cytodiagnosis/methods , Immunohistochemistry/methods , Membrane Proteins/metabolism , Mucin-4/metabolism , Pancreatic Neoplasms/pathology , Adenocarcinoma/metabolism , Biomarkers, Tumor/metabolism , Biopsy, Fine-Needle , Humans , Pancreatic Neoplasms/metabolism , Predictive Value of Tests , Reproducibility of Results , Up-Regulation
16.
J Hematol Oncol ; 5: 68, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-23102107

ABSTRACT

BACKGROUND: Pancreatic cancer (PC) is a lethal malignancy primarily driven by activated Kras mutations and characterized by the deregulation of several genes including mucins. Previous studies on mucins have identified their significant role in both benign and malignant human diseases including PC progression and metastasis. However, the initiation of MUC expression during PC remains unknown because of lack of early stage tumor tissues from PC patients. METHODS: In the present study, we have evaluated stage specific expression patterns of mucins during mouse PC progression in (Kras(G12D);Pdx1-Cre (KC)) murine PC model from pancreatic intraepithelial neoplasia (PanIN) to pancreatic ductal adenocarcinoma (PDAC) by immunohistochemistry and quantitative real-time PCR. RESULTS: In agreement with previous studies on human PC, we observed a progressive increase in the expression of mucins particularly Muc1, Muc4 and Muc5AC in the pancreas of KC (as early as PanIN I) mice with advancement of PanIN lesions and PDAC both at mRNA and protein levels. Additionally, mucin expression correlated with the increased expression of inflammatory cytokines IFN-γ (p < 0.0062), CXCL1 (p < 0.00014) and CXCL2 (p < 0.08) in the pancreas of KC mice, which are known to induce mucin expression. Further, we also observed progressive increase in inflammation in pancreas of KC mice from 10 to 50 weeks of age as indicated by the increase in the macrophage infiltration. Overall, this study corroborates with previous human studies that indicated the aberrant overexpression of MUC1, MUC4 and MUC5AC mucins during the progression of PC. CONCLUSIONS: Our study reinforces the potential utility of the KC murine model for determining the functional role of mucins in PC pathogenesis by crossing KC mice with corresponding mucin knockout mice and evaluating mucin based diagnostic and therapeutic approaches for lethal PC.


Subject(s)
Mucins/biosynthesis , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Animals , Disease Models, Animal , Disease Progression , Genotype , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Mucins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/therapy , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
17.
Carcinogenesis ; 33(10): 1953-64, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22791819

ABSTRACT

MUC4 is a type-1 transmembrane mucin differentially expressed in multiple cancers and has previously been shown to potentiate progression and metastasis of pancreatic cancer. In this study, we investigated the molecular mechanisms associated with the MUC4-induced invasion and metastasis in pancreatic cancer. Stable silencing of MUC4 in multiple pancreatic cancer cells resulted in the downregulation of N-cadherin and its interacting partner fibroblast growth factor receptor 1 (FGFR1) through downregulation of partly by pFAK, pMKK7, pJNK and pc-Jun pathway and partly through PI-3K/Akt pathway. The downregulation of FGFR1 in turn led to downregulation of pAkt, pERK1/2, pNF-κB, pIkBα, uPA, MMP-9, vimentin, N-cadherin, Twist, Slug and Zeb1 and upregulation of E-cadherin, Occludin, Cytokeratin-18 and Caspase-9 in MUC4 knockdown BXPC3 and Capan1 cells compared with scramble vector transfected cells. Further, downregulation of FGFR1 was associated with a significant change in morphology and reorganization of the actin-cytoskeleton, leading to a significant decrease in motility (P < 0.00001) and invasion (P < 0.0001) in vitro and decreased tumorigenicity and incidence of metastasis in vivo upon orthotopic implantation in the athymic mice. Taken together, the results of the present study suggest that MUC4 promotes invasion and metastasis by FGFR1 stabilization through the N-cadherin upregulation.


Subject(s)
Epithelial-Mesenchymal Transition , Mucin-4/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Animals , Cadherins/metabolism , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Gene Knockdown Techniques , Humans , Mice , Mice, Nude , Mucin-4/genetics , RNA, Small Interfering/pharmacology , Transplantation, Heterologous , Up-Regulation
18.
Expert Opin Ther Targets ; 16(1): 15-31, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22239438

ABSTRACT

INTRODUCTION: Cancer is a devastating disease; however, several therapeutic advances have recently been made, wherein EGFR and its family members have emerged as useful biomarkers and therapeutic targets. EGFR, a transmembrane glycoprotein is a member of the ERBB receptor tyrosine kinase superfamily. EGFR binds to its cognate ligand EGF, which further induces tyrosine phosphorylation and receptor dimerization with other family members leading to enhanced uncontrolled proliferation. Several anti-EGFR therapies such as monoclonal antibodies and tyrosine kinase inhibitors have been developed, which has enabled clinicians to identify and treat specific patient cohorts. AREAS COVERED: This review covers the basic mechanism of EGFR activation and the role of EGFR signaling in cancer progression. Furthermore, current developments made toward targeting the EGFR signaling pathway for the treatment of epithelial cancers and a summary of the various anti-EGFR therapeutic agents that are currently in use are also presented in this review. EXPERT OPINION: EGFR signaling is a part of a complex network that has been the target of effective cancer therapies. However, a further understanding of the system is required to develop an effective anticancer regimen. A combination therapy that comprises an anti-EGFR and a chemotherapeutic/chemopreventive agent will exhibit a multi-pronged approach that can be developed into a highly attractive and specific molecular oriented remedy.


Subject(s)
Antineoplastic Agents/therapeutic use , ErbB Receptors/metabolism , Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/therapeutic use , Humans , Neoplasms/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction/drug effects
19.
PLoS One ; 6(10): e26839, 2011.
Article in English | MEDLINE | ID: mdl-22066010

ABSTRACT

MUC16 (CA125) belongs to a family of high-molecular weight O-glycosylated proteins known as mucins. While MUC16 is well known as a biomarker in ovarian cancer, its expression pattern in pancreatic cancer (PC), the fourth leading cause of cancer related deaths in the United States, remains unknown. The aim of our study was to analyze the expression of MUC16 during the initiation, progression and metastasis of PC for possible implication in PC diagnosis, prognosis and therapy. In this study, a microarray containing tissues from healthy and PC patients was used to investigate the differential protein expression of MUC16 in PC. MUC16 mRNA levels were also measured by RT-PCR in the normal human pancreatic, pancreatitis, and PC tissues. To investigate its expression pattern during PC metastasis, tissue samples from the primary pancreatic tumor and metastases (from the same patient) in the lymph nodes, liver, lung and omentum from Stage IV PC patients were analyzed. To determine its association in the initiation of PC, tissues from PC patients containing pre-neoplastic lesions of varying grades were stained for MUC16. Finally, MUC16 expression was analyzed in 18 human PC cell lines. MUC16 is not expressed in the normal pancreatic ducts and is strongly upregulated in PC and detected in pancreatitis tissue. It is first detected in the high-grade pre-neoplastic lesions preceding invasive adenocarcinoma, suggesting that its upregulation is a late event during the initiation of this disease. MUC16 expression appears to be stronger in metastatic lesions when compared to the primary tumor, suggesting a role in PC metastasis. We have also identified PC cell lines that express MUC16, which can be used in future studies to elucidate its functional role in PC. Altogether, our results reveal that MUC16 expression is significantly increased in PC and could play a potential role in the progression of this disease.


Subject(s)
CA-125 Antigen/metabolism , Membrane Proteins/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Blotting, Western , CA-125 Antigen/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Membrane Proteins/genetics , Microscopy, Confocal , Neoplasm Metastasis , Pancreatic Ducts/metabolism , Pancreatic Ducts/pathology , Pancreatic Neoplasms/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...