Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Genome Res ; 29(1): 1-17, 2019 01.
Article in English | MEDLINE | ID: mdl-30563911

ABSTRACT

Human papillomavirus (HPV) is a necessary but insufficient cause of a subset of oral squamous cell carcinomas (OSCCs) that is increasing markedly in frequency. To identify contributory, secondary genetic alterations in these cancers, we used comprehensive genomics methods to compare 149 HPV-positive and 335 HPV-negative OSCC tumor/normal pairs. Different behavioral risk factors underlying the two OSCC types were reflected in distinctive genomic mutational signatures. In HPV-positive OSCCs, the signatures of APOBEC cytosine deaminase editing, associated with anti-viral immunity, were strongly linked to overall mutational burden. In contrast, in HPV-negative OSCCs, T>C substitutions in the sequence context 5'-ATN-3' correlated with tobacco exposure. Universal expression of HPV E6*1 and E7 oncogenes was a sine qua non of HPV-positive OSCCs. Significant enrichment of somatic mutations was confirmed or newly identified in PIK3CA, KMT2D, FGFR3, FBXW7, DDX3X, PTEN, TRAF3, RB1, CYLD, RIPK4, ZNF750, EP300, CASZ1, TAF5, RBL1, IFNGR1, and NFKBIA Of these, many affect host pathways already targeted by HPV oncoproteins, including the p53 and pRB pathways, or disrupt host defenses against viral infections, including interferon (IFN) and nuclear factor kappa B signaling. Frequent copy number changes were associated with concordant changes in gene expression. Chr 11q (including CCND1) and 14q (including DICER1 and AKT1) were recurrently lost in HPV-positive OSCCs, in contrast to their gains in HPV-negative OSCCs. High-ranking variant allele fractions implicated ZNF750, PIK3CA, and EP300 mutations as candidate driver events in HPV-positive cancers. We conclude that virus-host interactions cooperatively shape the unique genetic features of these cancers, distinguishing them from their HPV-negative counterparts.


Subject(s)
Carcinoma, Squamous Cell , Mouth Neoplasms , Neoplasm Proteins , Oncogene Proteins, Viral , Papillomavirus Infections , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/virology , Female , Humans , Male , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Mouth Neoplasms/virology , Mutation , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Oncogene Proteins, Viral/biosynthesis , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , Papillomaviridae/metabolism
2.
J Natl Compr Canc Netw ; 19(10): 1122-1132, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34666312

ABSTRACT

Identifying individuals with hereditary syndromes allows for timely cancer surveillance, opportunities for risk reduction, and syndrome-specific management. Establishing criteria for hereditary cancer risk assessment allows for the identification of individuals who are carriers of pathogenic genetic variants. The NCCN Guidelines for Genetic/Familial High-Risk Assessment: Colorectal provides recommendations for the assessment and management of patients at risk for or diagnosed with high-risk colorectal cancer syndromes. The NCCN Genetic/Familial High-Risk Assessment: Colorectal panel meets annually to evaluate and update their recommendations based on their clinical expertise and new scientific data. These NCCN Guidelines Insights focus on familial adenomatous polyposis (FAP)/attenuated familial adenomatous polyposis (AFAP) syndrome and considerations for management of duodenal neoplasia.


Subject(s)
Adenomatous Polyposis Coli , Colorectal Neoplasms , Adenomatous Polyposis Coli/diagnosis , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/therapy , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , Heterozygote , Humans , Risk Factors
3.
Transpl Int ; 33(2): 142-148, 2020 02.
Article in English | MEDLINE | ID: mdl-31523865

ABSTRACT

Intestinal transplant recipients experience a high rate of renal complications secondary to dehydration due to increased ostomy output. It is hypothesized that inclusion of donor colon in the intestinal allograft may improve renal function in patients without functional native colon by improving fluid absorption. A single-center retrospective study of intestinal transplant recipients compared outcomes of patients receiving en bloc colon as part an intestinal allograft (ICTx), and those not receiving colon (CCNTx), as well as a control group of intestinal transplant recipients with functional native colon (ITx). Forty-seven patients (ICTx n = 17, CCNTx n = 15, ITx n = 15) were studied. One-year post-transplant renal function, as measured by change in glomerular filtration rate (GFR) and blood urea nitrogen (BUN) from baseline, was superior in ICTx (mean delta-GFR of -1.31 and delta-BUN of -1.46) compared to CCNTx (-6.54 and 17.54, P = 0.05 and P = 0.17, respectively) and similar to the ITx controls (0.55 and 2.09). Recipients of donor colon experienced a higher rate of ileostomy reversal when compared to CCNTx (62.5% vs. 20%, P = 0.0008), which was similar to the ITx controls (60%). These findings support the inclusion of en bloc donor colon in the intestinal allograft for recipients without functional native colon.


Subject(s)
Colon/transplantation , Intestines/transplantation , Kidney/physiology , Allografts , Glomerular Filtration Rate , Humans , Ileostomy , Kidney/physiopathology , Retrospective Studies
4.
Int J Surg Pathol ; 30(4): 443-447, 2022 Jun.
Article in English | MEDLINE | ID: mdl-34894817

ABSTRACT

COVID-19, the syndrome caused by the novel coronavirus SARS-CoV-2, has spread throughout the world, causing the death of at least three million people. For the over 81 million who have recovered, however, the long-term effects are only beginning to manifest. We performed a bilateral lung transplant on a 31-year-old male patient for chronic hypoxic respiratory failure, severe pulmonary hypertension and radiographically identified pulmonary fibrosis five months after an acute COVID-19 infection. The explant demonstrated moderate pulmonary vascular remodeling with intimal thickening and medial hypertrophy throughout, consistent with pulmonary hypertension. The parenchyma demonstrated an organizing lung injury in the proliferative phase, with severe fibrosis, histiocytic proliferation, type II pneumocyte hyperplasia, and alveolar loss consistent with known COVID-19 pneumonia complications.This report highlights a novel histologic finding in severe, chronic COVID-19. Although the findings in acute COVID-19 pneumonia have been well-examined at autopsy, the chronic course of this complex disease is not yet understood. The case presented herein suggests that COVID-induced pulmonary hypertension may become more common as more patients survive severe SARS-CoV-2-related pneumonia. Pulmonologists and pulmonary pathologists should be aware of this possible association and look for the clinical, radiographic, and histologic criteria in the appropriate clinical setting.


Subject(s)
COVID-19 , Hypertension, Pulmonary , Hypertension , Adult , Autopsy , COVID-19/complications , Humans , Hypertension, Pulmonary/etiology , Lung/diagnostic imaging , Lung/pathology , Male , SARS-CoV-2
5.
Cancer Biomark ; 33(2): 219-235, 2022.
Article in English | MEDLINE | ID: mdl-35213363

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is a formidable challenge for patients and clinicians. OBJECTIVE: To analyze the distribution of 31 different markers in tumor and stromal portions of the tumor microenvironment (TME) and identify immune cell populations to better understand how neoplastic, non-malignant structural, and immune cells, diversify the TME and influence PDAC progression. METHODS: Whole slide imaging (WSI) and cyclic multiplexed-immunofluorescence (MxIF) was used to collect 31 different markers over the course of nine distinctive imaging series of human PDAC samples. Image registration and machine learning algorithms were developed to largely automate an imaging analysis pipeline identifying distinct cell types in the TME. RESULTS: A random forest algorithm accurately predicted tumor and stromal-rich areas with 87% accuracy using 31 markers and 77% accuracy using only five markers. Top tumor-predictive markers guided downstream analyses to identify immune populations effectively invading into the tumor, including dendritic cells, CD4+ T cells, and multiple immunoregulatory subtypes. CONCLUSIONS: Immunoprofiling of PDAC to identify differential distribution of immune cells in the TME is critical for understanding disease progression, response and/or resistance to treatment, and the development of new treatment strategies.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Machine Learning , Pancreatic Neoplasms/metabolism , Stromal Cells/metabolism , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Female , Fluorescent Antibody Technique , Humans , Image Interpretation, Computer-Assisted , Male , Middle Aged , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Tumor Microenvironment/immunology
6.
Cancer Lett ; 503: 91-102, 2021 04 10.
Article in English | MEDLINE | ID: mdl-33485947

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is highly lethal. MUC4 (mucin4) is a heavily glycosylated protein aberrantly expressed in PDAC and promotes tumorigenesis via an unknown mechanism. To assess this, we genetically knocked out (KO) MUC4 in PDAC cells that did not express and did express truncated O-glycans (Tn/STn) using CRISPR/Cas9 technology. We found that MUC4 knockout cells possess less tumorigenicity in vitro and in vivo, which was further reduced in PDAC cells that express aberrant overexpression of truncated O-glycans. Also, MUC4KO cells showed a further reduction of epidermal growth factor receptors (ErbB) and their downstream signaling pathways in truncated O-glycan expressing PDAC cells. Tn-MUC4 specific 3B11 antibody inhibited MUC4-induced ErbB receptor and its downstream signaling cascades. MUC4 knockout differentially regulates apoptosis and cell cycle arrest in branched and truncated O-glycan expressing PDAC cells. Additionally, MUC4KO cells were found to be more sensitive to gemcitabine treatment. They possessed the upregulated expression of hENT1 and hCNT3 compared to parental cells, which were further affected in cells with aberrant O-glycosylation. Taken together, our results indicate that MUC4 enhances the malignant properties and gemcitabine resistance in PDAC tumors that aberrantly overexpress truncated O-glycans via altering ErbB/AKT signaling cascades and expression of nucleoside transporters, respectively.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Drug Resistance, Neoplasm , Mucin-4/genetics , Pancreatic Neoplasms/pathology , Polysaccharides/metabolism , Animals , Apoptosis , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Cycle , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Equilibrative Nucleoside Transporter 1/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Humans , Membrane Transport Proteins/metabolism , Mice , Mucin-4/metabolism , Neoplasm Transplantation , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Gemcitabine
7.
Article in English | MEDLINE | ID: mdl-34250417

ABSTRACT

Hereditary cancer syndromes infer high cancer risks and require intensive surveillance. Identification of high-risk individuals among patients with colorectal cancer (CRC) needs improvement. METHODS: Three thousand three hundred ten unselected adults who underwent surgical resection for primary invasive CRC were prospectively accrued from 51 hospitals across Ohio between January 1, 2013, and December 31, 2016. Universal Tumor screening (UTS) for mismatch repair (MMR) deficiency was performed for all, and pathogenic germline variants (PGVs) were identified using multigene panel testing (MGPT) in those who met at least one inclusion criterion: MMR deficiency, diagnosed < 50 years, multiple primary tumors (CRC or endometrial cancer), or with a first-degree relative with CRC or endometrial cancer. RESULTS: Five hundred twenty-five patients (15.9%) had MMR deficiency. Two hundred thirty-four of 3,310 (7.1%; 16% of the 1,462 who received MGPT) had 248 PGVs in cancer susceptibility genes. One hundred forty-two (4.3%) had a PGV in an MMR gene, and 101 (3.1%) had a PGV in a non-MMR gene. Ten with Lynch syndrome (LS) also had a non-MMR PGV and were included in both groups. Two (0.06%) had constitutional MLH1 hypermethylation. Of unexplained MMR-deficient patients, 88.4% (76 of 86) had double somatic MMR mutations. Testing for only MMR genes in MMR-deficient patients would have missed 18 non-MMR gene PGVs (7.3% of total PGVs identified). Had UTS been the only method used to screen for hereditary cancer syndromes, 38.6% (91 of 236) would have been missed, including 6.3% (9 of 144) of those with LS. These results have treatment implications as 5.3% (175 of 3,310) had PGVs in genes with therapeutic targets. CONCLUSION: UTS alone is insufficient for identifying a large proportion of CRC patients with hereditary syndromes, including some with LS. At a minimum, 7.1% of individuals with CRC have a PGV and pan-cancer MGPT should be considered for all patients with CRC.


Subject(s)
Colorectal Neoplasms/diagnosis , Early Detection of Cancer , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/genetics , Colorectal Neoplasms/prevention & control , Female , Humans , Male , Middle Aged , Neoplastic Syndromes, Hereditary/diagnosis , Ohio , Prospective Studies
8.
J Contam Hydrol ; 235: 103728, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33069942

ABSTRACT

A complex mixture of dissolved organic contaminants, emanating from a many decades-old, residual, dense non-aqueous phase liquid (DNAPL) source, migrates through unconfined, moderately heterogeneous, glacial-derived sediments and sedimentary rock in a residential area of Dane County, Wisconsin, USA. A portion of this contaminant plume intersects a large man-made pond, roughly 400 m downgradient of the source zone. Depth-discrete, multilevel groundwater sampling, detailed sedimentological logs, and hydraulic head profiles were used to delineate the spatial distribution of hydraulic, geologic, organic contaminant, and redox hydrochemical conditions within the established plume along two transects immediately upgradient of the pond. Twenty-one contaminants were detected and classified into four major contaminant groups: chlorinated ethenes, chlorinated ethanes, aromatics (BTEX: benzene, toluene, ethylbenzene, xylene), and aliphatic ketones. Within the glacial sediments and shallow bedrock, zones of reductive dechlorination of chlorinated ethenes and ethanes were juxtaposed with zones of BTEX and ketone degradation. Spatial heterogeneity in the concentration and distribution of contaminant groups and redox conditions was observed over lateral distances of tens of meters and vertical distances of tens of centimeters along the two transects. Although the site was situated in a complex glacial depositional environment, lithologic and hydraulic heterogeneity surprisingly only had a modest influence on the spatial distribution of plume contaminants. Depth-discrete sampling along paired, closely spaced transects (~20 m apart) was essential to assess internal plume composition/concentration evolution along flow paths with strong attenuation over short migration distances. This study shows how paired, highly resolved transects can enhance understanding of transverse and longitudinal variability in areas where contaminant-induced redox conditions control reaction zones and strong plume attenuation.


Subject(s)
Groundwater , Water Pollutants, Chemical , Aged , Ethane , Geology , Humans , Water Pollutants, Chemical/analysis
9.
Biochim Biophys Acta Rev Cancer ; 1873(1): 188318, 2020 01.
Article in English | MEDLINE | ID: mdl-31676330

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is an incredibly deadly disease with a 5-year survival rate of 9%. The presence of pancreatic cystic lesions (PCLs) confers an increased likelihood of future pancreatic cancer in patients placing them in a high-risk category. Discerning concurrent malignancy and risk of future PCL progression to cancer must be carefully and accurately determined to improve survival outcomes and avoid unnecessary morbidity of pancreatic resection. Unfortunately, current image-based guidelines are inadequate to distinguish benign from malignant lesions. There continues to be a need for accurate molecular and imaging biomarker(s) capable of identifying malignant PCLs and predicting the malignant potential of PCLs to enable risk stratification and effective intervention management. This review provides an update on the current status of biomarkers from pancreatic cystic fluid, pancreatic juice, and seromic molecular analyses and discusses the potential of radiomics for differentiating PCLs harboring cancer from those that do not.


Subject(s)
Carcinoma, Pancreatic Ductal/diagnosis , Early Detection of Cancer/methods , Pancreatic Cyst/diagnosis , Pancreatic Neoplasms/diagnosis , Precancerous Conditions/diagnosis , Biomarkers, Tumor/analysis , Disease Progression , Pancreas/pathology , Risk Factors , Survival Analysis
10.
Clin Cancer Res ; 14(19): 5995-6004, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18829478

ABSTRACT

PURPOSE: We investigated the contribution of Sonic hedgehog (SHH) to pancreatic cancer progression. EXPERIMENTAL DESIGN: We expressed SHH in a transformed primary ductal-derived epithelial cell line from the human pancreas, transformed hTert-HPNE (T-HPNE), and evaluated the effects on tumor growth. We also directly inhibited the activity of SHH in vivo by administering a blocking antibody to mice challenged orthotopically with the Capan-2 pancreatic cancer cell line, which is known to express SHH and form moderately differentiated tumors in nude mice. RESULTS: Our data provide evidence that expression of SHH influences tumor growth by contributing to the formation of desmoplasia in pancreatic cancer. We further show that SHH affects the differentiation and motility of human pancreatic stellate cells and fibroblasts. CONCLUSIONS: These data suggest that SHH contributes to the formation of desmoplasia in pancreatic cancer, an important component of the tumor microenvironment.


Subject(s)
Fibrosis/pathology , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/physiology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/therapy , Animals , Cell Line, Transformed , Cell Line, Tumor , Cell Transformation, Neoplastic , Fibroblasts/metabolism , Fibrosis/metabolism , Hedgehog Proteins/metabolism , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Pancreas/metabolism
11.
Cancer Res ; 67(11): 5201-10, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17545600

ABSTRACT

MUC1 is a heterodimeric transmembrane glycoprotein that is overexpressed and aberrantly glycosylated in ductal adenocarcinomas. Differential phosphorylation of the MUC1 cytoplasmic tail (MUC1CT) has been associated with signaling events that influence the proliferation and metastasis of cancer cells. We identified a novel tyrosine phosphorylation site (HGRYVPP) in the MUC1CT by mass spectrometric analysis of MUC1 from human pancreatic adenocarcinoma cell lines. Analyses in vitro and in vivo showed that platelet-derived growth factor receptor beta (PDGFRbeta) catalyzed phosphorylation of this site and of tyrosine in the RDTYHPM site. Stimulation of S2-013.MUC1F cells with PDGF-BB increased nuclear colocalization of MUC1CT and beta-catenin. PDGF-BB stimulation had no significant effect on cell proliferation rate; however, it enhanced invasion in vitro through Matrigel and in vivo tumor growth and metastases. Invasive properties of the cells were significantly altered on expression of phosphorylation-abrogating or phosphorylation-mimicking mutations at these sites. We propose that interactions of MUC1 and PDGFRbeta induce signal transduction events that influence the metastatic properties of pancreatic adenocarcinoma.


Subject(s)
Adenocarcinoma/metabolism , Antigens, Neoplasm/metabolism , Mucins/metabolism , Pancreatic Neoplasms/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites , Cell Line, Tumor , Cell Nucleus/metabolism , Female , Humans , Mass Spectrometry , Mice , Mice, Nude , Molecular Sequence Data , Mucin-1 , Neoplasm Invasiveness , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Phosphorylation , Tyrosine/metabolism , beta Catenin/metabolism
12.
J Nutr Biochem ; 64: 110-120, 2019 02.
Article in English | MEDLINE | ID: mdl-30476878

ABSTRACT

Occupational agricultural dust exposure can cause severe lung injury, including COPD and asthma exacerbations. Cell-derived extracellular vesicles can mediate inflammatory responses and immune activation, but the contribution of diet-derived extracellular vesicles to these processes is poorly understood. We investigated whether bovine milk-derived extracellular vesicles modulate inflammatory responses to agricultural dust exposures in a murine model. C57BL/6 mice were fed either an extracellular vesicle-enriched modification of the AIN-93G diet with lyophilized bovine milk (EV) or a control diet wherein the milk was presonicated, disrupting the milk extracellular vesicles and thereby leading to RNA degradation (DEV). Mice were maintained on the diets for 5-7 weeks and challenged with a single (acute) intranasal instillation of a 12.5% organic dust extract (DE) or with 15 instillations over 3 weeks (repetitive exposure model). Through these investigations, we identified significant interactions between diet and DE when considering numerous inflammatory outcomes, including lavage inflammatory cytokine levels and cellular infiltration into the lung airways. DE-treated peritoneal macrophages also demonstrated altered polarization, with EV-fed mouse macrophages exhibiting an M1 shift compared to an M2 phenotype in DEV-fed mice (IL-6, TNF, IL-12/23 all significantly elevated, and IL-10 and arginase decreased in EV macrophages, ex vivo). In complementary in vitro studies, mouse macrophages treated with purified milk-derived EV were found to express similar polarization phenotypes upon DE stimulation. These results suggest a role for dietary extracellular vesicles in the modulation of lung inflammation in response to organic dust which may involve macrophage phenotype polarization.


Subject(s)
Dust , Environmental Exposure/adverse effects , Extracellular Vesicles , Macrophages, Alveolar/immunology , Macrophages, Peritoneal/immunology , Milk , Animals , Cattle , Cell Polarity , Cells, Cultured , Housing, Animal , Immunity, Innate , Inflammation/diet therapy , Inflammation/etiology , Inflammation/pathology , Macrophages, Alveolar/pathology , Macrophages, Peritoneal/pathology , Male , Mice, Inbred C57BL , Milk/chemistry , Milk/cytology
13.
Cancers (Basel) ; 12(1)2019 Dec 18.
Article in English | MEDLINE | ID: mdl-31861288

ABSTRACT

Pancreatic cancer is the third leading cause of cancer-related deaths in the USA. Pancreatic tumors are characterized by enhanced glycolytic metabolism promoted by a hypoxic tumor microenvironment and a resultant acidic milieu. The metabolic reprogramming allows cancer cells to survive hostile microenvironments. Through the analysis of the principal metabolic pathways, we identified the specific metabolites that are altered during pancreatic cancer progression in the spontaneous progression (KPC) mouse model. Genetically engineered mice exhibited metabolic alterations during PanINs formation, even before the tumor development. To account for other cells in the tumor microenvironment and to focus on metabolic adaptations concerning tumorigenic cells only, we compared the metabolic profile of KPC and orthotopic tumors with those obtained from KPC-tumor derived cell lines. We observed significant upregulation of glycolysis and the pentose phosphate pathway metabolites even at the early stages of pathogenesis. Other biosynthetic pathways also demonstrated a few common perturbations. While some of the metabolic changes in tumor cells are not detectable in orthotopic and spontaneous tumors, a significant number of tumor cell-intrinsic metabolic alterations are readily detectable in the animal models. Overall, we identified that metabolic alterations in precancerous lesions are maintained during cancer development and are largely mirrored by cancer cells in culture conditions.

14.
J Bone Miner Res ; 34(9): 1733-1743, 2019 09.
Article in English | MEDLINE | ID: mdl-30995344

ABSTRACT

Rheumatoid arthritis (RA) is characterized by extra-articular involvement including lung disease, yet the mechanisms linking the two conditions are poorly understood. The collagen-induced arthritis (CIA) model was combined with the organic dust extract (ODE) airway inflammatory model to assess bone/joint-lung inflammatory outcomes. DBA/1J mice were intranasally treated with saline or ODE daily for 5 weeks. CIA was induced on days 1 and 21. Treatment groups included sham (saline injection/saline inhalation), CIA (CIA/saline), ODE (saline/ODE), and CIA + ODE (CIA/ODE). Arthritis inflammatory scores, bones, bronchoalveolar lavage fluid, lung tissues, and serum were assessed. In DBA/1J male mice, arthritis was increased in CIA + ODE > CIA > ODE versus sham. Micro-computed tomography (µCT) demonstrated that loss of BMD and volume and deterioration of bone microarchitecture was greatest in CIA + ODE. However, ODE-induced airway neutrophil influx and inflammatory cytokine/chemokine levels in lavage fluids were increased in ODE > CIA + ODE versus sham. Activated lung CD11c+ CD11b+ macrophages were increased in ODE > CIA + ODE > CIA pattern, whereas lung hyaluronan, fibronectin, and amphiregulin levels were greatest in CIA + ODE. Serum autoantibody and inflammatory marker concentrations varied among experimental groups. Compared with male mice, female mice showed less articular and pulmonary disease. The interaction of inhalation-induced airway inflammation and arthritis induction resulted in compartmentalized responses with the greatest degree of arthritis and bone loss in male mice with combined exposures. Data also support suppression of the lung inflammatory response, but increases in extracellular matrix protein deposition/interstitial disease in the setting of arthritis. This coexposure model could be exploited to better understand and treat RA-lung disease. © 2019 American Society for Bone and Mineral Research.


Subject(s)
Arthritis, Experimental/complications , Arthritis, Rheumatoid/complications , Dust , Inflammation/complications , Lung Diseases/etiology , Lung/pathology , Animals , Arthritis, Experimental/blood , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/pathology , Autoantibodies/blood , Biomarkers/blood , Cancellous Bone/pathology , Collagen , Extracellular Matrix Proteins/metabolism , Female , Inflammation/blood , Inflammation/pathology , Joints/pathology , Lung Diseases/blood , Lung Diseases/pathology , Male , Mice , Staining and Labeling
16.
Pancreas ; 47(5): 556-560, 2018.
Article in English | MEDLINE | ID: mdl-29683969

ABSTRACT

OBJECTIVE: The aim of this study is to determine the diagnostic accuracy of preoperative evaluation to detect main pancreatic duct involvement in pancreatic cystic lesions thus differentiating mixed intraductal papillary mucinous neoplasm (IPMN) from branch duct (BD)-IPMN. METHODS: The pathology database of pancreatic resections from 2000 to 2014 was reviewed. Main pancreatic duct-IPMNs and IPMNs with intracystic mass/nodules were excluded. The preoperative test characteristics were analyzed using surgical histopathology as the "gold standard." RESULTS: Sixty BD-IPMNs and 23 mixed-IPMNs were identified. Mixed-IPMNs were larger (mean [standard deviation], 4.14 [2.9] vs 2.74 [1.9] mm; P = 0.03) and demonstrated frequent high-grade dysplasia/adenocarcinoma (43% vs 12%, P = 0.004) than BD-IPMNs. Endoscopic ultrasound (EUS) (sensitivity, 80%; specificity, 78%; accuracy, 79%) had the best diagnostic accuracy, whereas magnetic resonance imaging (MRI) (sensitivity, 83%; specificity, 63%; accuracy, 68%) had the highest sensitivity for the diagnosis of mixed-IPMN. A combination of EUS and MRI reached maximum sensitivity but with decreased accuracy (sensitivity, 100%; specificity, 64%; accuracy, 67%). The area under the curve for receiver operation curve was 0.71 whereas the optimal cyst size to detect main duct involvement was 3 cm. CONCLUSIONS: For preoperative evaluation of pancreatic cystic lesions without evidence of intracystic nodules, a combination of MRI and EUS should be considered for improved detection of main duct involvement.


Subject(s)
Adenocarcinoma, Mucinous/diagnostic imaging , Carcinoma, Pancreatic Ductal/diagnostic imaging , Carcinoma, Papillary/diagnostic imaging , Diagnostic Imaging/methods , Pancreatic Ducts/diagnostic imaging , Pancreatic Neoplasms/diagnostic imaging , Aged , Diagnosis, Differential , Endosonography , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Preoperative Period , Sensitivity and Specificity
17.
Clin Cancer Res ; 12(10): 2976-87, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16707592

ABSTRACT

MUC1 is a highly glycosylated, type I transmembrane protein expressed by normal ductal epithelial cells of the pancreas, breast, lung, and gastrointestinal tract, and overexpressed in many cases of adenocarcinoma. We down-regulated MUC1 expression by RNA interference and investigated the effects on malignant and metastatic potential of a human pancreatic cancer cell line, S2-013. MUC1-suppressed clones, S2-013.MTII.C1 and S2-013.MTII.C2, were established by targeting a sequence 3,151 bp from the initiation codon and characterized in vitro for proliferation, invasion, and adhesion. We evaluated the effects of MUC1 suppression in vivo on tumor growth and metastatic properties following implantation into the cecum or pancreas of athymic mice. MUC1-suppressed clones showed significantly decreased proliferation in vitro and in vivo. Global gene expression was evaluated by oligonucleotide microarray analysis. Surprisingly, genes predicted to increase doubling times (cyclin B1 and cyclin D3) were overexpressed in MUC1-suppressed clones. There were alterations in expression of several genes that may affect the malignant properties of pancreatic cancer. Adhesion of MUC1-suppressed cells in vitro to type IV collagen and fibronectin was slightly increased, and adhesion was slightly decreased to type I collagen and laminin. Results of implantation to cecum and pancreas showed significant reduction of metastasis to lymph nodes, lung, or peritoneal sites compared with S2-013.gfp-neo control cells. These results support the hypothesis that MUC1 contributes significantly to growth and metastasis, and that down-regulation of MUC1 protein expression decreases the metastatic potential of pancreatic adenocarcinoma.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/physiology , Mucins/biosynthesis , Mucins/physiology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , RNA Interference , Animals , Cell Adhesion , Cell Proliferation , Down-Regulation , Female , Gene Expression Profiling , Humans , Mice , Mice, Nude , Mucin-1 , Neoplasm Metastasis , Neoplasms, Experimental , Phenotype , RNA, Small Interfering , Tumor Cells, Cultured
18.
Diagn Pathol ; 12(1): 24, 2017 Mar 04.
Article in English | MEDLINE | ID: mdl-28259170

ABSTRACT

BACKGROUND: Microsatellite-unstable colorectal cancers (CRC) that are due to deficient DNA mismatch repair (dMMR) represent approximately 15% of all CRCs in the United States. These microsatellite-unstable CRCs represent a heterogenous group of diseases with distinct oncogenesis pathways. There are overlapping clinicopathologic features between some of these groups, but many important differences are present. Therefore, determination of the etiology for the dMMR is vital for proper patient management and follow-up. MAIN BODY: Epigenetic inactivation of MLH1 MMR gene (sporadic microsatellite-unstable CRC) and germline mutation in an MMR gene (Lynch syndrome, LS) are the two most common mechanisms in the pathogenesis of microsatellite instability in CRC. However, in a subset of dMMR CRC cases that are identified by screening tests, no known LS-associated genetic alterations are appreciated by current genetic analysis. When the etiology for dMMR is unclear, it leads to patient anxiety and creates challenges for clinical management. CONCLUSION: It is critical to distinguish LS patients from other patients with tumors due to dMMR, so that the proper screening protocol can be employed for the patients and their families, with the goal to save lives while avoiding unnecessary anxiety and costs. This review summarizes the major pathogenesis pathways of dMMR CRCs, their clinicopathologic features, and practical screening suggestions. In addition, we include frequently asked questions for MMR immunohistochemistry interpretation.


Subject(s)
Colorectal Neoplasms/genetics , DNA Mismatch Repair/genetics , Microsatellite Instability , Colon/pathology , Colorectal Neoplasms/diagnosis , Humans , Immunohistochemistry , Pathologists , Rectum/pathology
20.
World J Gastroenterol ; 23(18): 3338-3348, 2017 May 14.
Article in English | MEDLINE | ID: mdl-28566895

ABSTRACT

AIM: To investigate the reproducibility of the in vivo endoscopic ultrasound (EUS) - guided needle based confocal endomicroscopy (nCLE) image patterns in an ex vivo setting and compare these to surgical histopathology for characterizing pancreatic cystic lesions (PCLs). METHODS: In a prospective study evaluating EUS-nCLE for evaluation of PCLs, 10 subjects underwent an in vivo nCLE (AQ-Flex nCLE miniprobe; Cellvizio, MaunaKea, Paris, France) during EUS and ex vivo probe based CLE (pCLE) of the PCL (Gastroflex ultrahigh definition probe, Cellvizio) after surgical resection. Biopsies were obtained from ex vivo CLE-imaged areas for comparative histopathology. All subjects received intravenous fluorescein prior to EUS and pancreatic surgery for in vivo and ex vivo CLE imaging respectively. RESULTS: A total of 10 subjects (mean age 53 ± 12 years; 5 female) with a mean PCL size of 34.8 ± 14.3 mm were enrolled. Surgical histopathology confirmed 2 intraductal papillary mucinous neoplasms (IPMNs), 3 mucinous cystic neoplasms (MCNs), 2 cystic neuroendocrine tumors (cystic-NETs), 1 serous cystadenoma (SCA), and 2 squamous lined PCLs. Characteristic in vivo nCLE image patterns included papillary projections for IPMNs, horizon-type epithelial bands for MCNs, nests and trabeculae of cells for cystic-NETs, and a "fern pattern" of vascularity for SCA. Identical image patterns were observed during ex vivo pCLE imaging of the surgically resected PCLs. Both in vivo and ex vivo CLE imaging findings correlated with surgical histopathology. CONCLUSION: In vivo nCLE patterns are reproducible in ex vivo pCLE for all major neoplastic PCLs. These findings add further support the application of EUS-nCLE as an imaging biomarker in the diagnosis of PCLs.


Subject(s)
Microscopy, Confocal/methods , Pancreatic Cyst/diagnostic imaging , Adult , Aged , Biomarkers, Tumor/metabolism , Biopsy , Cross-Sectional Studies , Cystadenoma, Serous/diagnostic imaging , Cystadenoma, Serous/pathology , Endoscopic Ultrasound-Guided Fine Needle Aspiration , Female , Humans , Image Processing, Computer-Assisted , Male , Middle Aged , Pancreatic Cyst/pathology , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/pathology , Prospective Studies , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL