Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 152
Filter
Add more filters

Publication year range
1.
Mol Carcinog ; 62(2): 145-159, 2023 02.
Article in English | MEDLINE | ID: mdl-36218231

ABSTRACT

Doublecortin like kinase 1 (DCLK1) plays a crucial role in several cancers including colon and pancreatic adenocarcinomas. However, its role in squamous cell carcinoma (SCC) remains unknown. To this end, we examined DCLK1 expression in head and neck SCC (HNSCC) and anal SCC (ASCC). We found that DCLK1 is elevated in patient SCC tissue, which correlated with cancer progression and poorer overall survival. Furthermore, DCLK1 expression is significantly elevated in human papilloma virus negative HNSCC, which are typically aggressive with poor responses to therapy. To understand the role of DCLK1 in tumorigenesis, we used specific shRNA to suppress DCLK1 expression. This significantly reduced tumor growth, spheroid formation, and migration of HNSCC cancer cells. To further the translational relevance of our studies, we sought to identify a selective DCLK1 inhibitor. Current attempts to target DCLK1 using pharmacologic approaches have relied on nonspecific suppression of DCLK1 kinase activity. Here, we demonstrate that DiFiD (3,5-bis [2,4-difluorobenzylidene]-4-piperidone) binds to DCLK1 with high selectivity. Moreover, DiFiD mediated suppression of DCLK1 led to G2/M arrest and apoptosis and significantly suppressed tumor growth of HNSCC xenografts and ASCC patient derived xenografts, supporting that DCLK1 is critical for SCC growth.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Humans , Apoptosis , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Doublecortin-Like Kinases , G2 Phase Cell Cycle Checkpoints , Intracellular Signaling Peptides and Proteins/genetics , Protein Serine-Threonine Kinases/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics , Animals
2.
J Pathol ; 256(2): 186-201, 2022 02.
Article in English | MEDLINE | ID: mdl-34714554

ABSTRACT

Due to widespread adoption of screening mammography, there has been a significant increase in new diagnoses of ductal carcinoma in situ (DCIS). However, DCIS prognosis remains unclear. To address this gap, we developed an in vivo model, Mouse-INtraDuctal (MIND), in which patient-derived DCIS epithelial cells are injected intraductally and allowed to progress naturally in mice. Similar to human DCIS, the cancer cells formed in situ lesions inside the mouse mammary ducts and mimicked all histologic subtypes including micropapillary, papillary, cribriform, solid, and comedo. Among 37 patient samples injected into 202 xenografts, at median duration of 9 months, 20 samples (54%) injected into 95 xenografts showed in vivo invasive progression, while 17 (46%) samples injected into 107 xenografts remained non-invasive. Among the 20 samples that showed invasive progression, nine samples injected into 54 xenografts exhibited a mixed pattern in which some xenografts showed invasive progression while others remained non-invasive. Among the clinically relevant biomarkers, only elevated progesterone receptor expression in patient DCIS and the extent of in vivo growth in xenografts predicted an invasive outcome. The Tempus XT assay was used on 16 patient DCIS formalin-fixed, paraffin-embedded sections including eight DCISs that showed invasive progression, five DCISs that remained non-invasive, and three DCISs that showed a mixed pattern in the xenografts. Analysis of the frequency of cancer-related pathogenic mutations among the groups showed no significant differences (KW: p > 0.05). There were also no differences in the frequency of high, moderate, or low severity mutations (KW; p > 0.05). These results suggest that genetic changes in the DCIS are not the primary driver for the development of invasive disease. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Epithelial Cells/pathology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/metabolism , Cell Movement , Cell Proliferation , Disease Progression , Epithelial Cells/metabolism , Epithelial Cells/transplantation , Female , Heterografts , Humans , Mice, Inbred NOD , Mice, SCID , Mutation , Neoplasm Invasiveness , Neoplasm Transplantation , Receptors, Progesterone/metabolism , Time Factors
3.
Breast Cancer Res ; 24(1): 68, 2022 10 18.
Article in English | MEDLINE | ID: mdl-36258226

ABSTRACT

BACKGROUND: Ductal carcinoma in situ (DCIS) is the most common type of in situ premalignant breast cancers. What drives DCIS to invasive breast cancer is unclear. Basal-like invasive breast cancers are aggressive. We have previously shown that NRAS is highly expressed selectively in basal-like subtypes of invasive breast cancers and can promote their growth and progression. In this study, we investigated whether NRAS expression at the DCIS stage can control transition from luminal DCIS to basal-like invasive breast cancers. METHODS: Wilcoxon rank-sum test was performed to assess expression of NRAS in DCIS compared to invasive breast tumors in patients. NRAS mRNA levels were also determined by fluorescence in situ hybridization in patient tumor microarrays (TMAs) with concurrent normal, DCIS, and invasive breast cancer, and association of NRAS mRNA levels with DCIS and invasive breast cancer was assessed by paired Wilcoxon signed-rank test. Pearson's correlation was calculated between NRAS mRNA levels and basal biomarkers in the TMAs, as well as in patient datasets. RNA-seq data were generated in cell lines, and unsupervised hierarchical clustering was performed after combining with RNA-seq data from a previously published patient cohort. RESULTS: Invasive breast cancers showed higher NRAS mRNA levels compared to DCIS samples. These NRAShigh lesions were also enriched with basal-like features, such as basal gene expression signatures, lower ER, and higher p53 protein and Ki67 levels. We have shown previously that NRAS drives aggressive features in DCIS-like and basal-like SUM102PT cells. Here, we found that NRAS-silencing induced a shift to a luminal gene expression pattern. Conversely, NRAS overexpression in the luminal DCIS SUM225 cells induced a basal-like gene expression pattern, as well as an epithelial-to-mesenchymal transition signature. Furthermore, these cells formed disorganized mammospheres containing cell masses with an apparent reduction in adhesion. CONCLUSIONS: These data suggest that elevated NRAS levels in DCIS are not only a marker but can also control the emergence of basal-like features leading to more aggressive tumor activity, thus supporting the therapeutic hypothesis that targeting NRAS and/or downstream pathways may block disease progression for a subset of DCIS patients with high NRAS.


Subject(s)
Breast Neoplasms , Carcinoma, Ductal, Breast , Carcinoma, Intraductal, Noninfiltrating , Humans , Female , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/pathology , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Carcinoma, Ductal, Breast/pathology , Tumor Suppressor Protein p53/genetics , Breast Neoplasms/pathology , In Situ Hybridization, Fluorescence , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , RNA, Messenger , Disease Progression , Membrane Proteins/genetics , Membrane Proteins/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism
4.
Gastroenterology ; 158(5): 1433-1449.e27, 2020 04.
Article in English | MEDLINE | ID: mdl-31786131

ABSTRACT

BACKGROUND & AIMS: Prolactin (PRL) signaling is up-regulated in hormone-responsive cancers. The PRL receptor (PRLR) is a class I cytokine receptor that signals via the Janus kinase (JAK)-signal transducer and activator of transcription and mitogen-activated protein kinase pathways to regulate cell proliferation, migration, stem cell features, and apoptosis. Patients with pancreatic ductal adenocarcinoma (PDAC) have high plasma levels of PRL. We investigated whether PRLR signaling contributes to the growth of pancreatic tumors in mice. METHODS: We used immunohistochemical analyses to compare levels of PRL and PRLR in multitumor tissue microarrays. We used structure-based virtual screening and fragment-based drug discovery to identify compounds likely to bind PRLR and interfere with its signaling. Human pancreatic cell lines (AsPC-1, BxPC-3, Panc-1, and MiaPaCa-2), with or without knockdown of PRLR (clustered regularly interspaced short palindromic repeats or small hairpin RNA), were incubated with PRL or penfluridol and analyzed in proliferation and spheroid formation. C57BL/6 mice were given injections of UNKC-6141 cells, with or without knockdown of PRLR, into pancreas, and tumor development was monitored for 4 weeks, with some mice receiving penfluridol treatment for 21 days. Human pancreatic tumor tissues were implanted into interscapular fat pads of NSG mice, and mice were given injections of penfluridol daily for 28 days. Nude mice were given injections of Panc-1 cells, xenograft tumors were grown for 2 weeks, and mice were then given intraperitoneal penfluridol for 35 days. Tumors were collected from mice and analyzed by histology, immunohistochemistry, and immunoblots. RESULTS: Levels of PRLR were increased in PDAC compared with nontumor pancreatic tissues. Incubation of pancreatic cell lines with PRL activated signaling via JAK2-signal transducer and activator of transcription 3 and extracellular signal-regulated kinase, as well as formation of pancospheres and cell migration; these activities were not observed in cells with PRLR knockdown. Pancreatic cancer cells with PRLR knockdown formed significantly smaller tumors in mice. We identified several diphenylbutylpiperidine-class antipsychotic drugs as agents that decreased PRL-induced JAK2 signaling; incubation of pancreatic cancer cells with these compounds reduced their proliferation and formation of panco spheres. Injections of 1 of these compounds, penfluridol, slowed the growth of xenograft tumors in the different mouse models, reducing proliferation and inducing autophagy of the tumor cells. CONCLUSIONS: Levels of PRLR are increased in PDAC, and exposure to PRL increases proliferation and migration of pancreatic cancer cells. Antipsychotic drugs, such as penfluridol, block PRL signaling in pancreatic cancer cells to reduce their proliferation, induce autophagy, and slow the growth of xenograft tumors in mice. These drugs might be tested in patients with PDAC.


Subject(s)
Antipsychotic Agents/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Pancreatic Neoplasms/drug therapy , Penfluridol/pharmacology , Prolactin/metabolism , Receptors, Prolactin/antagonists & inhibitors , Animals , Antipsychotic Agents/therapeutic use , Autophagy/drug effects , Carcinoma, Pancreatic Ductal/blood , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Discovery , Gene Knockdown Techniques , Humans , Injections, Intraperitoneal , Janus Kinase 2/metabolism , Male , Mice , Pancreas/pathology , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/pathology , Penfluridol/therapeutic use , Prolactin/blood , Receptors, Prolactin/genetics , Receptors, Prolactin/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Spheroids, Cellular , Tissue Array Analysis , Xenograft Model Antitumor Assays
5.
Gastroenterology ; 157(6): 1646-1659.e11, 2019 12.
Article in English | MEDLINE | ID: mdl-31442435

ABSTRACT

BACKGROUND & AIMS: The histone lysine demethylase 3A (KDM3A) demethylates H3K9me1 and H3K9Me2 to increase gene transcription and is upregulated in tumors, including pancreatic tumors. We investigated its activities in pancreatic cancer cell lines and its regulation of the gene encoding doublecortin calmodulin-like kinase 1 (DCLK1), a marker of cancer stem cells. METHODS: We knocked down KDM3A in MiaPaCa-2 and S2-007 pancreatic cancer cell lines and overexpressed KDM3A in HPNE cells (human noncancerous pancreatic ductal cell line); we evaluated cell migration, invasion, and spheroid formation under hypoxic and normoxic conditions. Nude mice were given orthotopic injections of S2-007 cells, with or without (control) knockdown of KDM3A, and HPNE cells, with or without (control) overexpression of KDM3A; tumor growth was assessed. We analyzed pancreatic tumor tissues from mice and pancreatic cancer cell lines by immunohistochemistry and immunoblotting. We performed RNA-sequencing analysis of MiaPaCa-2 and S2-007 cells with knockdown of KDM3A and evaluated localization of DCLK1 and KDM3A by immunofluorescence. We analyzed the cancer genome atlas for levels of KDM3A and DCLK1 messenger RNA in human pancreatic ductal adenocarcinoma (PDAC) tissues and association with patient survival time. RESULTS: Levels of KDM3A were increased in human pancreatic tumor tissues and cell lines, compared with adjacent nontumor pancreatic tissues, such as islet and acinar cells. Knockdown of KDM3A in S2-007 cells significantly reduced colony formation, invasion, migration, and spheroid formation, compared with control cells, and slowed growth of orthotopic tumors in mice. We identified KDM3A-binding sites in the DCLK1 promoter; S2-007 cells with knockdown of KDM3A had reduced levels of DCLK1. HPNE cells that overexpressed KDM3A formed foci and spheres in culture and formed tumors and metastases in mice, whereas control HPNE cells did not. Hypoxia induced sphere formation and increased levels of KDM3A in S2-007 cells and in HPNE cells that overexpressed DCLK1, but not control HPNE cells. Levels of KDM3A and DCLK1 messenger RNA were higher in human PDAC than nontumor pancreatic tissues and correlated with shorter survival times of patients. CONCLUSIONS: We found human PDAC samples and pancreatic cancer cell lines to overexpress KDM3A. KDM3A increases expression of DCLK1, and levels of both proteins are increased in human PDAC samples. Knockdown of KDM3A in pancreatic cancer cell lines reduced their invasive and sphere-forming activities in culture and formation of orthotopic tumors in mice. Hypoxia increased expression of KDM3A in pancreatic cancer cells. Strategies to disrupt this pathway might be developed for treatment of pancreatic cancer.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/genetics , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Pancreatic Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , Animals , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , DNA Methylation , Datasets as Topic , Doublecortin-Like Kinases , Female , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Male , Mice , Middle Aged , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Promoter Regions, Genetic/genetics , Protein Serine-Threonine Kinases/metabolism , Survival Analysis , Up-Regulation , Xenograft Model Antitumor Assays
6.
Int J Gynecol Cancer ; 28(7): 1271-1277, 2018 09.
Article in English | MEDLINE | ID: mdl-30036218

ABSTRACT

OBJECTIVE: Undifferentiated endometrial carcinoma (UEC) represents a recently recognized and rare diagnosis that is commonly misclassified on histopathologic evaluation. These cancers account for less than 10% of carefully reviewed series of endometrial cancers from academic medical centers. We reviewed a single-institutional experience with the management of UEC focusing on clinicopathologic characteristics and treatment outcomes. METHODS: The medical records of all patients treated for histologically proven endometrial carcinoma between 2007 through 2016 were reviewed. Analysis was limited to 24 consecutive patients with histologically proven endometrial carcinomas that had at least a component of undifferentiated carcinoma on central pathology review. All patients were initially treated by definitive surgical resection. Grade 3 endometrioid carcinomas treated over the same period were used as a control group. The Kaplan-Meier method was used to estimate survival outcomes. RESULTS: The median age at diagnosis was 66 years (range, 37-74 years). Ten patients presented with locally advanced or metastatic disease (42%). Fifteen patients (63%) received adjuvant chemotherapy with carboplatin and paclitaxel, 12 patients (50%) received adjuvant pelvic external beam radiation, and 10 patients (42%) received adjuvant vaginal cuff brachytherapy. With a median follow-up of 14 months (range, 0.5-115 months), 4 patients (21%) had developed disease recurrence and/or progression, 2 patients (11%) had died of disease, and 1 patient died of treatment complications. Twelve patients (63%) were alive with no evidence of disease at last contact. Outcomes were comparable to those with grade 3 endometrioid carcinoma. CONCLUSIONS: Our data are consistent with prior studies demonstrating that UEC represents a rare clinical entity characterized by high rates of locally advanced disease at presentation. However, survival outcomes appear to be comparable to other high-grade endometrial cancers. Further studies investigating optimal adjuvant therapy in these patients are warranted.


Subject(s)
Carcinoma, Endometrioid/pathology , Carcinoma, Endometrioid/therapy , Endometrial Neoplasms/pathology , Endometrial Neoplasms/therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biopsy , Brachytherapy , Carboplatin/administration & dosage , Carcinoma, Endometrioid/surgery , Chemotherapy, Adjuvant , Endometrial Neoplasms/surgery , Female , Humans , Lymph Nodes/pathology , Middle Aged , Neoplasm Grading , Neoplasm Recurrence, Local/pathology , Paclitaxel/administration & dosage , Radiotherapy, Adjuvant , Retrospective Studies , Survival Rate
7.
Int J Mol Sci ; 18(3)2017 Mar 16.
Article in English | MEDLINE | ID: mdl-28300755

ABSTRACT

Osteosarcoma (OS) is an aggressive malignancy of bone affecting children, adolescents and young adults. Understanding vitamin D metabolism and vitamin D regulated genes in OS is an important aspect of vitamin D/cancer paradigm, and in evaluating vitamin D as adjuvant therapy for human OS. Vitamin D treatment of 143B OS cells induced significant and novel changes in the expression of genes that regulate: (a) inflammation and immunity; (b) formation of reactive oxygen species, metabolism of cyclic nucleotides, sterols, vitamins and mineral (calcium), quantity of gap junctions and skeletogenesis; (c) bone mineral density; and (d) cell viability of skeletal cells, aggregation of bone cancer cells and exocytosis of secretory vesicles. Ingenuity pathway analysis revealed significant reduction in Runx2 target genes such as fibroblast growth factor -1, -12 (FGF1 and FGF12), bone morphogenetic factor-1 (BMP1), SWI/SNF related, matrix associated actin dependent regulator of chromatin subfamily a, member 4 (SMARCA4), Matrix extracellular phosphoglycoprotein (MEPE), Integrin, ß4 (ITGBP4), Matrix Metalloproteinase -1, -28 (MMP1 and MMP28), and signal transducer and activator of transcription-4 (STAT4) in vitamin D treated 143B OS cells. These genes interact with the inflammation, oxidative stress and membrane vesicle biogenesis gene networks. Vitamin D not only inhibited the expression of Runx2 target genes MMP1, MMP28 and kallikrein related peptidase-7 (KLK7), but also migration and invasion of 143B OS cells. Vitamin D regulated Runx2 target genes or their products represent potential therapeutic targets and laboratory biomarkers for applications in translational oncology.


Subject(s)
Core Binding Factor Alpha 1 Subunit/genetics , Gene Regulatory Networks , Osteosarcoma/metabolism , Oxidative Stress , Transport Vesicles/genetics , Vitamin D/pharmacology , Vitamins/pharmacology , Bone Morphogenetic Protein 1/genetics , Bone Morphogenetic Protein 1/metabolism , Cell Line, Tumor , Core Binding Factor Alpha 1 Subunit/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Inflammation/genetics , Integrin beta4/genetics , Integrin beta4/metabolism , Kallikreins/genetics , Kallikreins/metabolism , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transport Vesicles/metabolism
8.
Am J Physiol Gastrointest Liver Physiol ; 310(5): G295-302, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26744468

ABSTRACT

Farnesoid X receptor (FXR) belongs to the nuclear receptor superfamily with its endogenous ligands bile acids. Mice with whole body FXR deficiency develop liver tumors spontaneously, but the underlying mechanism is unclear. Moreover, it is unknown whether FXR deficiency in liver alone serves as a tumor initiator or promoter during liver carcinogenesis. This study aims to evaluate the effects of hepatocyte-specific FXR deficiency (FXR(hep-/-)) in liver tumor formation. The results showed that FXR(hep-/-) mice did not show spontaneous liver tumorigenesis with aging (up to 24 mo of age). Therefore FXR(hep-/-) mice were fed a bile acid (cholic acid)-containing diet alone or along with a liver tumor initiator, diethylnitrosamine (DEN). Thirty weeks later, no tumors were found in wild-type or FXR(hep-/-) mice without any treatment or with DEN only. However, with cholic acid, while only some wild-type mice developed tumors, all FXR(hep-/-) mice presented with severe liver injury and tumors. Interestingly, FXR(hep-/-) mouse livers increased basal expression of tumor suppressor p53 protein, apoptosis, and decreased basal cyclin D1 expression, which may prevent tumor development in FXR(hep-/-) mice. However, cholic acid feeding reversed these effects in FXR(hep-/-) mice, which is associated with an increased cyclin D1 and decreased cell cycle inhibitors. More in-depth analysis indicates that the increased in cell growth might result from disturbance of the MAPK and JAK/Stat3 signaling pathways. In conclusion, this study shows that hepatic FXR deficiency may only serve as a tumor initiator, and increased bile acids is required for tumor formation likely by promoting cell proliferation.


Subject(s)
Cholic Acid/metabolism , Cyclin D1/metabolism , Hepatocytes , Liver Neoplasms , Receptors, Cytoplasmic and Nuclear , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Carcinogenesis/metabolism , Cell Proliferation/drug effects , Diethylnitrosamine/pharmacology , Hepatocytes/metabolism , Hepatocytes/pathology , Janus Kinases/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase Kinases/metabolism , Models, Animal , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects
9.
Ann Diagn Pathol ; 21: 47-52, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27040931

ABSTRACT

Despite the known benefits of the use of radioactive seed localization (RSL), few studies have looked at the resultant pathologic marginal status of these lumpectomy specimens, especially in regard to different definitions of close/positive margins. We compared the marginal status of lumpectomy specimens removed by either RSL or conventional wire localization (CWL) techniques. A total of 106 lumpectomy specimens including 62 by CWL and 44 by RSL for invasive ductal and lobular carcinomas were compared. Data on gross and microscopic surgical margin status, tumor type and grade, and demographic information were retrospectively collected. There was no difference between the techniques in terms of tumor characteristics including size, histologic grade, lymph node positivity, or age. Although the distributions are very similar between CWL and RSL specimens for final marginal assessments (P=.69), there is a (modest) statistically significant difference in the distribution for margin classifications based on gross assessments (P=.040), specifically more RSL specimens exhibiting tumor within 1mm of the closest margin. Concordance between gross and microscopic lesion measurements is highest for invasive ductal carcinoma grade 3 for both CWL and RSL lumpectomies (78.6% and 80.0%). This study shows that there were no significant marginal status differences between RSL and CWL lumpectomy specimens with invasive carcinoma. Rather, what was relevant is whether the entire specimen could be classified as having negative/close margins. Significant workflow challenges in surgical pathology laboratories are expected with the adoption of the RSL process.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/pathology , Carcinoma, Lobular/pathology , Mastectomy, Segmental/methods , Adult , Aged , Aged, 80 and over , Breast/pathology , Breast Neoplasms/surgery , Carcinoma, Ductal, Breast/surgery , Carcinoma, Intraductal, Noninfiltrating/surgery , Carcinoma, Lobular/surgery , Female , Humans , Iodine Radioisotopes , Margins of Excision , Middle Aged , Neoplasm Grading , Retrospective Studies
10.
Ann Diagn Pathol ; 22: 54-7, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27180061

ABSTRACT

The prognostic significance of peritoneal washing cytology in gynecologic neoplasms is controversial. The presence of neoplastic cells in peritoneal washings is currently part of the Federation of Gynecology and Obstetrics and American Joint Committee on Cancer TNM staging systems in cases of ovarian and fallopian tube neoplasms without metastasis beyond the pelvis. In this study, we retrospectively reviewed all cases of ovarian and fallopian tube neoplasms in which cytologic studies were performed. The utility of cytology in tumor staging and the relationship between cytology results and patient outcome are studied. All cases of ovarian and fallopian tube neoplasms in our institution between July 2002 and July 2012 were reviewed. Primary tumor characteristics including type and pelvic extension were collected, categorized, and correlated with peritoneal washing cytology. Final tumor staging was reviewed and the impact of positive cytology was evaluated. A total of 120 cases of ovarian and fallopian tube neoplasms without extrapelvic metastasis were identified within the study period. Peritoneal washing cytology was positive in 24% (29/120) of neoplasms and upstaged the tumor 83% (24/29) of the time when positive. Overall, 20% (24/120) of reviewed cases were upstaged based on positive cytology results. Peritoneal washing cytology remains a useful staging tool for ovarian and fallopian tube neoplasms limited to the pelvic cavity. Positive cytology results in upstaging in a significant proportion of the cases regardless of the tumor type. A larger study is needed to analyze follow-up data to determine if upstaging based on positive cytology adversely affects outcome.


Subject(s)
Endometrial Neoplasms/pathology , Fallopian Tube Neoplasms/diagnosis , Fallopian Tube Neoplasms/pathology , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Cytodiagnosis/methods , Endometrial Neoplasms/diagnosis , Female , Humans , Middle Aged , Neoplasm Staging , Peritoneal Neoplasms/diagnosis , Peritoneal Neoplasms/secondary , Prognosis , Retrospective Studies
11.
Breast Cancer Res ; 17: 128, 2015 Sep 17.
Article in English | MEDLINE | ID: mdl-26384318

ABSTRACT

INTRODUCTION: There are an estimated 60,000 new cases of ductal carcinoma in situ (DCIS) each year. A lack of understanding in DCIS pathobiology has led to overtreatment of more than half of patients. We profiled the temporal molecular changes during DCIS transition to invasive ductal carcinoma (IDC) using in vivo DCIS progression models. These studies identified B cell lymphoma-9 (BCL9) as a potential molecular driver of early invasion. BCL9 is a newly found co-activator of Wnt-stimulated ß-catenin-mediated transcription. BCL9 has been shown to promote progression of multiple myeloma and colon carcinoma. However BCL9 role in breast cancer had not been previously recognized. METHODS: Microarray and RNA sequencing were utilized to characterize the sequential changes in mRNA expression during DCIS invasive transition. BCL9-shRNA knockdown was performed to assess the role of BCL9 in in vivo invasion, epithelial-mesenchymal transition (EMT) and canonical Wnt-signaling. Immunofluorescence of 28 patient samples was used to assess a correlation between the expression of BCL9 and biomarkers of high risk DCIS. The cancer genome atlas data were analyzed to assess the status of BCL9 gene alterations in breast cancers. RESULTS: Analysis of BCL9, by RNA and protein showed BCL9 up-regulation to be associated with DCIS transition to IDC. Analysis of patient DCIS revealed a significant correlation between high nuclear BCL9 and pathologic characteristics associated with DCIS recurrence: Estrogen receptor (ER) and progesterone receptor (PR) negative, high nuclear grade, and high human epidermal growth factor receptor2 (HER2). In vivo silencing of BCL9 resulted in the inhibition of DCIS invasion and reversal of EMT. Analysis of the TCGA data showed BCL9 to be altered in 26 % of breast cancers. This is a significant alteration when compared to HER2 (ERBB2) gene (19 %) and estrogen receptor (ESR1) gene (8 %). A significantly higher proportion of basal like invasive breast cancers compared to luminal breast cancers showed BCL9 amplification. CONCLUSION: BCL9 is a molecular driver of DCIS invasive progression and may predispose to the development of basal like invasive breast cancers. As such, BCL9 has the potential to serve as a biomarker of high risk DCIS and as a therapeutic target for prevention of IDC.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Intraductal, Noninfiltrating/genetics , Carcinoma, Intraductal, Noninfiltrating/pathology , Neoplasm Proteins/genetics , Transcriptome/genetics , Animals , Biomarkers, Tumor/genetics , Carcinoma, Ductal, Breast/pathology , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Receptor, ErbB-2/genetics , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics , Transcription Factors , Transcription, Genetic/genetics , Up-Regulation/genetics , Wnt Proteins/genetics , beta Catenin/genetics
12.
Mol Carcinog ; 54(12): 1772-85, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25418856

ABSTRACT

Molecular vulnerabilities represent promising candidates for the development of targeted therapies that hold the promise to overcome the challenges encountered with non-targeted chemotherapy for the treatment of ovarian cancer. Through a synthetic lethality screen, we previously identified pleiotrophin (PTN) as a molecular vulnerability in ovarian cancer and showed that siRNA-mediated PTN knockdown induced apoptotic cell death in epithelial ovarian cancer (EOC) cells. Although, it is well known that PTN elicits its pro-tumorigenic effects through its receptor, protein tyrosine phosphatase receptor Z1 (PTPRZ1), little is known about the potential importance of this pathway in the pathogenesis of ovarian cancer. In this study, we show that PTN is expressed, produced, and secreted in a panel of EOC cell lines. PTN levels in serous ovarian tumor tissues are on average 3.5-fold higher relative to normal tissue and PTN is detectable in serum samples of patients with EOC. PTPRZ1 is also expressed and produced by EOC cells and is found to be up-regulated in serous ovarian tumor tissue relative to normal ovarian surface epithelial tissue (P < 0.05). Gene silencing of PTPRZ1 in EOC cell lines using siRNA-mediated knockdown shows that PTPRZ1 is essential for viability and results in significant apoptosis with no effect on the cell cycle phase distribution. In order to determine how PTN mediates survival, we silenced the gene using siRNA mediated knockdown and performed expression profiling of 36 survival-related genes. Through computational mapping of the differentially expressed genes, members of the MAPK (mitogen-activated protein kinase) family were found to be likely effectors of PTN signaling in EOC cells. Our results provide the first experimental evidence that PTN and its signaling components may be of significance in the pathogenesis of epithelial ovarian cancer and provide a rationale for clinical evaluation of MAPK inhibitors in PTN and/or PTPRZ1 expressing ovarian tumors.


Subject(s)
Carrier Proteins/genetics , Cytokines/genetics , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 5/genetics , Signal Transduction/genetics , Adult , Aged , Aged, 80 and over , Apoptosis/genetics , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Silencing/physiology , Humans , Middle Aged , Mitogen-Activated Protein Kinases/genetics , RNA, Small Interfering/genetics
13.
Anal Biochem ; 486: 70-4, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26142221

ABSTRACT

Osteosarcoma, the most common type of primary bone cancer, is the second highest cause of cancer-related death in pediatric patients. To understand the mechanisms behind osteosarcoma progression and to discover novel therapeutic strategies for this disease, a reliable and appropriate mouse model is essential. For this purpose, osteosarcoma cells need to be injected into the bone marrow. Previously, the intratibial and intrafemoral injection methods were reported; however, the major drawback of these methods is the potential leakage of tumor cells from the injection site during or after these procedures. To overcome this, we have established an improved method to minimize leakage in an orthotopic mouse model of osteosarcoma. By taking advantage of the anatomical benefits of the femur with less bowing and larger medullary cavity than those of the tibia, osteosarcoma cells are injected directly into the femoral cavity following reaming of its intramedullary space. To prevent potential leakage of tumor cells during and after the surgery, the injection site is sealed with bone wax. This method requires a minor surgery of approximately 15min under anesthesia. Our established orthotopic osteosarcoma model could serve as a valuable and reliable tool for examining progression of various types of bone tumors.


Subject(s)
Bone Neoplasms/pathology , Cell Transformation, Neoplastic , Femur , Injections/methods , Osteosarcoma/pathology , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Mice
14.
Acta Cytol ; 59(2): 187-200, 2015.
Article in English | MEDLINE | ID: mdl-25967603

ABSTRACT

OBJECTIVE: To date, the impact of digital imaging on routine cytology remains far from perfect. Cellblock (CB) preparations from Pap samples have been shown to be diagnostically valuable. We evaluated the validity of utilizing whole-slide imaging (WSI) prepared from Pap CBs as a screening tool. STUDY DESIGN: A total of 1,110 CB slides prepared from residual Pap samples were analyzed - 563 normal, 282 atypical squamous cells of undetermined significance (ASCUS), 12 atypical squamous cells-cannot exclude high-grade squamous intraepithelial lesion, 188 low-grade squamous intraepithelial lesions (LSIL), 36 high-grade squamous intraepithelial lesions (HSIL), 25 atypical glandular cells of undetermined significance, 1 adenocarcinoma in situ, 2 invasive adenocarcinomas, and 1 squamous cell carcinoma. Virtual slides were obtained using the Aperio system. Test performance characteristics of liquid-based samples and WSI from CB samples were compared. RESULTS: Average sensitivity and specificity of the five WSI reviewers was 58.3 and 85.1% for ASCUS, respectively, 54.1 and 93.9% for LSIL, and 51.8 and 98.8% for HSIL. Overall WSI sensitivity and specificity for detecting lesions was 82.1 and 86.2%, respectively. Agreement (kappa values) between WSI reviewers was 0.56 for ASCUS, 0.69 for LSIL, 0.67 for HSIL, and 0.74 for negative samples. CONCLUSIONS: WSI of CB preparations is a feasible method to achieve high-quality specimen preparations. It is as sensitive as liquid-based methods and appears to be highly specific for the detection of LSIL and HSIL.


Subject(s)
Adenocarcinoma/pathology , Atypical Squamous Cells of the Cervix , Carcinoma, Squamous Cell/pathology , Cervix Uteri/pathology , Image Interpretation, Computer-Assisted , Papanicolaou Test , Squamous Intraepithelial Lesions of the Cervix/pathology , Uterine Cervical Dysplasia/pathology , Uterine Cervical Neoplasms/pathology , Vaginal Smears , Adolescent , Adult , Aged , Aged, 80 and over , Automation, Laboratory , Feasibility Studies , Female , Humans , Middle Aged , Neoplasm Grading , Observer Variation , Predictive Value of Tests , Reproducibility of Results , Retrospective Studies , Young Adult
15.
Carcinogenesis ; 35(4): 795-806, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24265293

ABSTRACT

Prolactin (PRL) is a secretory cytokine produced by various tissues. Binding to the cognate PRL receptor (PRLR), it activates intracellular signaling via janus kinase (JAK), extracellular signal-regulated kinase (ERK) and signal transducer and activator of transcription (STAT) proteins. PRL regulates diverse activities under normal and abnormal conditions, including malignancies. Previous clinical data suggest serum PRL levels are elevated in colorectal cancer (CRC) patients. In this study, we first determined the expression of PRL and PRLR in colon cancer tissue and cell lines. Higher levels of PRLR expression were observed in the cancer cells and cell lines compared with normal colonic epithelial cells. Incubation of colon cancer cells with PRL-induced JAK2, STAT3 and ERK1/2 phosphorylation and increased expression of Jagged 1, which is a Notch-1 receptor ligand. Notch signaling regulates CRC stem cell population. We observed increased accumulation of the cleaved/active form of Notch-1 receptor (Notch intracellular domain) and increased expression of Notch responsive genes HEY1, HES1 and stem cell marker genes DCLK1, LGR5, ALDH1 and CD44. Finally, inhibiting PRL induced JAK2-STAT3 and JAK2-ERK1/2 using AG490 and PD98059, respectively, leads to complete abrogation of Notch signaling, suggesting a role for this pathway in regulating CRC stem cells. Together, our results demonstrate that cytokine signaling induced by PRL is active in colorectal cancers and may provide a novel target for therapeutic intervention.


Subject(s)
Colonic Neoplasms/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Janus Kinase 2/metabolism , Neoplastic Stem Cells/metabolism , Prolactin/metabolism , Receptors, Notch/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Base Sequence , Colonic Neoplasms/metabolism , DNA Primers , Humans , Phosphorylation , Polymerase Chain Reaction
16.
Ann Diagn Pathol ; 18(6): 333-42, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25263387

ABSTRACT

The diagnosis of intraductal carcinoma (IDC) of the prostate remains subjective because 3 sets of diagnostic criteria are in use. An internet survey was compiled from 38 photomicrographs showing duct proliferations: 14 signed out as high-grade prostatic intraepithelial neoplasia (HGPIN), 17 IDC, and 7 invasive cribriform/ductal carcinoma. Each image was assessed for the presence of 9 histologic criteria ascribed to IDC. Thirty-nine respondents were asked to rate images as (1) benign/reactive, (2) HGPIN, (3) borderline between HGPIN and IDC, (4) IDC, or (5) invasive cribriform/ductal carcinoma. Intraclass correlation coefficient was 0.68. There was 70% overall agreement with HGPIN, 43% with IDC, and 73% with invasive carcinoma (P < .001, χ(2)). Respondents considered 19 (50%) of 38 cases as IDC candidates, of which 5 (26%) had a two-thirds consensus for IDC; two-thirds consensus for either borderline or IDC was reached in 9 (47%). Two-thirds consensus other than IDC was reached in the remaining 19 of 38 cases, with 15 supporting HGPIN and 4 supporting invasive carcinoma. Findings that differed across diagnostic categories were lumen-spanning neoplastic cells (P < .001), 2× benign duct diameters (P < .001), duct space contours (round, irregular, and branched) (P < .001), papillary growth (P = .048), dense cribriform or solid growth (both P = .023), and comedonecrosis (P = .015). When the 19 of 38 images that attained consensus for HGPIN or invasive carcinoma were removed from consideration, lack of IDC consensus was most often attributable to only loose cribriform growth (5/19), central nuclear maturation (5/19), or comedonecrosis (3/19). Of the 9 histologic criteria, only 1 retained significant correlation with a consensus diagnosis of IDC: the presence of solid areas (P = .038). One case that attained IDC consensus had less than 2× duct enlargement yet still had severe nuclear atypia and nucleomegaly. Six fold nuclear enlargement was not significant (P = .083), although no image had both 6× nuclei and papillary or loose cribriform growth: a combination postulated as sufficient criteria for IDC. Finally, 20.5% of respondents agreed that an isolated diagnosis of IDC on needle biopsy warrants definitive therapy, 20.5% disagreed, and 59.0% considered the decision to depend upon clinicopathologic variables. Although IDC diagnosis remains challenging, we propose these criteria: a lumen-spanning proliferation of neoplastic cells in preexisting ducts with a dense cribriform or partial solid growth pattern. Solid growth, in any part of the duct space, emerges as the most reproducible finding to rule in a diagnosis of IDC. Comedonecrosis is a rarer finding, but in most cases, it should rule in IDC. Duct space enlargement to greater than 2× the diameter of the largest, adjacent benign spaces is usually present in IDC, although there may be rare exceptions.


Subject(s)
Carcinoma, Intraductal, Noninfiltrating/pathology , Prostatic Neoplasms/pathology , Diagnosis, Differential , Health Surveys , Humans , Male , Neoplasm Grading , Observer Variation , Physicians , Prognosis , Prostatic Intraepithelial Neoplasia/pathology , Reproducibility of Results
17.
Autops Case Rep ; 14: e2024488, 2024.
Article in English | MEDLINE | ID: mdl-38803483

ABSTRACT

Primary cardiac tumors are rare. The cardiac sarcomas are the most common malignant cardiac tumors. These tumors have a dismal prognosis with an overall median survival of 25 months. Clinical features include dyspnea, arrhythmias, pericardial effusions, heart failure, and sudden cardiac death. The diagnosis is often challenging. Therefore, the cardiac imaging workup plays a central role in addition to a high clinical suspicion in the setting of atypical presentations that do not respond to standard therapies. The echocardiography, computed tomography, and cardiac MRI are crucial in clinching the diagnosis. Multimodal treatment with surgery, chemotherapy, and radiotherapy has been shown to improve outcomes, as opposed to using either of these modalities alone. We describe the case of a 30-year-old gentleman with COVID-19 infection who developed recurrent hemorrhagic pericardial effusions refractory to standard treatment and was eventually diagnosed as a case of pericardial angiosarcoma after his biopsy revealed the diagnosis and staging was performed using PET-CT-FDG scan. Our case re-emphasizes the importance of considering a malignant etiology early in the course of the disease presentation, especially in recurrent hemorrhagic effusions despite an inflammatory cytologic diagnosis of fluid. It also highlights the place for cardiac CT and MRI to ascertain the location and spread and to plan the further course of treatment. If diagnosed early, the estimated survival time can be prolonged by instituting a multimodal approach.

18.
Cancers (Basel) ; 16(4)2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38398152

ABSTRACT

BACKGROUND AND AIMS: There is limited literature on sample adequacy for molecular testing in pancreatic ductal adenocarcinoma obtained via endoscopic ultrasound (EUS) fine-needle aspiration (FNA) versus EUS fine-needle biopsy (FNB). We aimed to compare these two modalities regarding sample adequacy for molecular and genomic sequencing. METHODS: We reviewed all patients with pancreatic ductal adenocarcinoma who underwent EUS at Saint Luke's Hospital from 2018 to 2021. The patients were categorized based on the method of EUS tissue acquisition, specifically FNA or FNB. A comprehensive evaluation was conducted for all cases by cytotechnologists. RESULTS: Out of 132 patients who underwent EUS-guided biopsies, 76 opted for FNA, 48 opted for FNB, and 8 opted for a combination of both. The average number of passes required for FNB and FNA was 2.58 ± 1.06 and 2.49 ± 1.07, respectively (p = 0.704), indicating no significant difference. Interestingly, 71.4% (35) of FNB-obtained samples were deemed adequate for molecular testing, surpassing the 32.1% (26) adequacy observed with FNA (p < 0.001). Additionally, 46.4% (26) of FNB-obtained samples were considered adequate for genomic testing, a notable improvement over the 23.8% (20) adequacy observed with FNA (p = 0.005). CONCLUSION: Although the number of passes required for cytologic diagnosis did not differ significantly between EUS-FNB and EUS-FNA, the former demonstrated superiority in obtaining samples adequate for molecular testing. Tumor surface area and cellularity were crucial parameters in determining sample adequacy for molecular testing, irrespective of the chosen tissue acquisition modality.

19.
Cell Death Dis ; 15(5): 362, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38796478

ABSTRACT

Advanced epithelial ovarian cancer (EOC) survival rates are dishearteningly low, with ~25% surviving beyond 5 years. Evidence suggests that cancer stem cells contribute to acquired chemoresistance and tumor recurrence. Here, we show that IRAK1 is upregulated in EOC tissues, and enhanced expression correlates with poorer overall survival. Moreover, low molecular weight hyaluronic acid, which is abundant in malignant ascites from patients with advanced EOC, induced IRAK1 phosphorylation leading to STAT3 activation and enhanced spheroid formation. Knockdown of IRAK1 impaired tumor growth in peritoneal disease models, and impaired HA-induced spheroid growth and STAT3 phosphorylation. Finally, we determined that TCS2210, a known inducer of neuronal differentiation in mesenchymal stem cells, is a selective inhibitor of IRAK1. TCS2210 significantly inhibited EOC growth in vitro and in vivo both as monotherapy, and in combination with cisplatin. Collectively, these data demonstrate IRAK1 as a druggable target for EOC.


Subject(s)
Carcinoma, Ovarian Epithelial , Hyaluronic Acid , Interleukin-1 Receptor-Associated Kinases , Neoplastic Stem Cells , Ovarian Neoplasms , STAT3 Transcription Factor , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Humans , STAT3 Transcription Factor/metabolism , Female , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/genetics , Hyaluronic Acid/metabolism , Hyaluronic Acid/pharmacology , Animals , Ovarian Neoplasms/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Cell Line, Tumor , Mice , Cisplatin/pharmacology , Mice, Nude , Phosphorylation/drug effects , Cell Proliferation/drug effects , Molecular Weight , Xenograft Model Antitumor Assays
20.
Am J Physiol Gastrointest Liver Physiol ; 305(11): G829-37, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24091600

ABSTRACT

Farnesoid X receptor (FXR, Nr1h4) is a ligand-activated transcription factor belonging to the nuclear receptor superfamily. FXR is essential in maintaining bile acid (BA) homeostasis, and FXR(-/-) mice develop cholestasis, inflammation, and spontaneous liver tumors. The signal transducer and activator of transcription 3 (STAT3) is well known to regulate liver growth, and STAT3 is feedback inhibited by its target gene, the suppressor of cytokine signaling 3 (SOCS3). Strong activation of STAT3 was detected in FXR(-/-) mouse livers. However, the mechanism of STAT3 activation with FXR deficiency remains elusive. Wild-type (WT) and FXR(-/-) mice were used to detect STAT3 pathway activation in the liver. In vivo BA feeding or deprivation was used to determine the role of BAs in STAT3 activation, and in vitro molecular approaches were used to determine the direct transcriptional regulation of SOCS3 by FXR. STAT3 was activated in FXR(-/-) but not WT mice. BA feeding increased, but deprivation by cholestyramine reduced, serum inflammatory markers and STAT3 activation. Furthermore, the Socs3 gene was determined as a direct FXR target gene. The elevated BAs and inflammation, along with reduced SOCS3, collectively contribute to the activation of the STAT3 signaling pathway in the liver of FXR(-/-) mice. This study suggests that the constitutive activation of STAT3 may be a mechanism of liver carcinogenesis in FXR(-/-) mice.


Subject(s)
Liver/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , STAT3 Transcription Factor/metabolism , Animals , Bile Acids and Salts/deficiency , Bile Acids and Salts/metabolism , Bile Acids and Salts/pharmacology , Cholestyramine Resin/pharmacology , Cytokines/blood , Cytokines/metabolism , Liver/drug effects , Mice , Mice, Knockout , Receptors, Cytoplasmic and Nuclear/genetics , STAT3 Transcription Factor/genetics , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL