Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Nat Commun ; 12(1): 3044, 2021 05 24.
Article in English | MEDLINE | ID: mdl-34031415

ABSTRACT

Unlike other malignancies, therapeutic options in pancreatic ductal adenocarcinoma (PDAC) are largely limited to cytotoxic chemotherapy without the benefit of molecular markers predicting response. Here we report tumor-cell-intrinsic chromatin accessibility patterns of treatment-naïve surgically resected PDAC tumors that were subsequently treated with (Gem)/Abraxane adjuvant chemotherapy. By ATAC-seq analyses of EpCAM+ PDAC malignant epithelial cells sorted from 54 freshly resected human tumors, we show here the discovery of a signature of 1092 chromatin loci displaying differential accessibility between patients with disease free survival (DFS) < 1 year and patients with DFS > 1 year. Analyzing transcription factor (TF) binding motifs within these loci, we identify two TFs (ZKSCAN1 and HNF1b) displaying differential nuclear localization between patients with short vs. long DFS. We further develop a chromatin accessibility microarray methodology termed "ATAC-array", an easy-to-use platform obviating the time and cost of next generation sequencing. Applying this methodology to the original ATAC-seq libraries as well as independent libraries generated from patient-derived organoids, we validate ATAC-array technology in both the original ATAC-seq cohort as well as in an independent validation cohort. We conclude that PDAC prognosis can be predicted by ATAC-array, which represents a low-cost, clinically feasible technology for assessing chromatin accessibility profiles.


Subject(s)
Chromatin Immunoprecipitation Sequencing/methods , Chromatin , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/genetics , Biomarkers, Tumor , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Nucleus , Hepatocyte Nuclear Factor 1-beta/genetics , High-Throughput Nucleotide Sequencing/methods , Humans , Kruppel-Like Transcription Factors/genetics , Pancreatic Neoplasms/metabolism , Prognosis , Transcription Factors , Transcriptome , Pancreatic Neoplasms
2.
Oncogene ; 35(32): 4282-8, 2016 08 11.
Article in English | MEDLINE | ID: mdl-26592447

ABSTRACT

Pancreatic cancer is one of the most lethal malignancies, with virtually all patients eventually succumbing to their disease. Mutations in p53 have been documented in >50% of pancreatic cancers. Owing to the high incidence of p53 mutations in PanIN 3 lesions and pancreatic tumors, we interrogated the comparative ability of adult pancreatic acinar and ductal cells to respond to oncogenic Kras and mutant Tp53(R172H) using Hnf1b:CreER(T2) and Mist1:CreER(T2) mice. These studies involved co-activation of a membrane-tethered GFP lineage label, allowing for direct visualization and isolation of cells undergoing Kras and mutant p53 activation. Kras activation in Mist1(+) adult acinar cells resulted in brisk PanIN formation, whereas no evidence of pancreatic neoplasia was observed for up to 6 months following Kras activation in Hnf1beta(+) adult ductal cells. In contrast to the lack of response to oncogenic Kras alone, simultaneous activation of Kras and mutant p53 in adult ductal epithelium generated invasive PDAC in 75% of mice as early as 2.5 months after tamoxifen administration. These data demonstrate that pancreatic ductal cells, whereas exhibiting relative resistance to oncogenic Kras alone, can serve as an effective cell of origin for pancreatic ductal adenocarcinoma in the setting of gain-of-function mutations in p53.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Mutation , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Tumor Suppressor Protein p53/genetics , Animals , Carcinogenesis , Cell Line, Tumor , Cellular Senescence , Humans , Mice , Phosphoproteins/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction
3.
Oncogene ; 32(45): 5253-60, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-23416985

ABSTRACT

Efficient metastasis is believed as the result of multiple genetic, epigenetic and/or post-translational events in the lifetime of a carcinoma. At the genetic level, these events may be categorized into those that occur during carcinogenesis, and those that occur during subclonal evolution. This review summarizes current knowledge of the genetics of pancreatic cancer from its initiation within a normal cell until the time that is has disseminated to distant organs, many features of which can be extrapolated to other solid tumor types. The implications of these findings to personalize genome analyses of an individual patient's tumor are also discussed.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Pancreatic Ductal/genetics , Neoplasm Metastasis/genetics , Pancreatic Neoplasms/genetics , Clonal Evolution , Disease Progression , Genes, p16 , Humans , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Smad4 Protein/genetics , Tumor Suppressor Protein p53/genetics , ras Proteins/genetics
4.
Br J Cancer ; 103(5): 649-55, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20664591

ABSTRACT

BACKGROUND: The purpose of this work was to determine the efficacy of inhibiting mammalian target of rapamycin (mTOR) in pancreatic cancer preclinical models and translate preclinical observations to the clinic. METHODS: Temsirolimus (20 mg Kg(-1) daily) was administered to freshly generated pancreatic cancer xenografts. Tumour growth inhibition was determined after 28 days. Xenografts were characterised at baseline by gene expression and comparative genomic hybridisation. Patients with advanced, gemcitabine-resistant pancreatic cancer were treated with sirolimus (5 mg daily). The primary end point was 6-month survival rate (6mSR). Correlative studies included immunohistochemistry assessment of pathway expression in baseline tumours, drug pharmacokinetics (PKs), response assessment by FDG-PET and pharmacodynamic effects in peripheral-blood mononuclear cells (PBMCs). RESULTS: In all, 4 of 17 xenografts (23%) responded to treatment. Sensitive tumours were characterised by gene copy number variations and overexpression of genes leading to activation of the PI3K/Akt/mTOR pathway. Activation of p70S6K correlated with drug activity in the preclinical studies. Sirolimus was well tolerated in the clinic, showed predictable PKs, exerted pathway inhibition in post-treatment PBMCs and resulted in a 6mSR of 26%. No correlation, however, was found between activated p70S6K in tumour tissues and anti-tumour effects. CONCLUSION: Sirolimus activity in pancreatic cancer was marginal and not predicted by the selected biomarker.


Subject(s)
Adenocarcinoma/drug therapy , Antibiotics, Antineoplastic/therapeutic use , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Sirolimus/therapeutic use , Adult , Aged , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Signal Transduction , TOR Serine-Threonine Kinases , Xenograft Model Antitumor Assays
5.
Clin Genet ; 72(6): 568-73, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17924967

ABSTRACT

LKB1/STK11 germline inactivations are identified in the majority (66-94%) of Peutz-Jeghers syndrome (PJS) patients. Therefore, defects in other genes or so far unidentified ways of LKB1 inactivation may cause PJS. The genes encoding the MARK proteins, homologues of the Par1 polarity protein that associates with Par4/Lkb1, were analyzed in this study because of their link to LKB1 and cell polarity. The genetic defect underlying PJS was determined through analysis of both LKB1 and all four MARK genes. LKB1 point mutations and small deletions were identified in 18 of 23 PJS families using direct sequencing and multiplex ligation-dependent probe amplification analysis identified exon deletions in 3 of 23 families. In total, 91% of the studied families showed LKB1 inactivation. Furthermore, a MARK1, MARK2, MARK3 and MARK4 mutation analysis and an MARK4 quantitative multiplex polymerase chain reaction analysis to identify exon deletions on another eight PJS families without identified LKB1 germline mutation did not identify mutations in the MARK genes. LKB1 defects are the major cause of PJS and genes of the MARK family do not represent alternative PJS genes. Other mechanisms of inactivation of LKB1 may cause PJS in the remaining families.


Subject(s)
Multigene Family , Peutz-Jeghers Syndrome/genetics , Protein Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinase Kinases , Exons , Female , Germ-Line Mutation , Humans , Introns , Male , Peutz-Jeghers Syndrome/enzymology , Sequence Deletion
7.
Am J Pathol ; 159(6): 2017-22, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11733352

ABSTRACT

Despite the growing awareness of intraductal papillary-mucinous neoplasms (IPMNs) of the pancreas among clinicians, the molecular features of IPMNs have not been well characterized. Previous reports suggest that inactivation of the STK11/LKB1, a tumor-suppressor gene responsible for Peutz-Jeghers syndrome (PJS), plays a role in the pathogenesis of gastrointestinal hamartomas as well as several cancers, including pancreatic adenocarcinoma. Using polymerase chain reaction amplification of five microsatellite markers from the 19p13.3 region harboring the STK11/LKB1 gene, we analyzed DNA from 22 IPMNs for loss of heterozygosity (LOH). LOH at 19p13.3 was identified in 2 of 2 (100%) IPMNs from patients with PJS and 5 of 20 (25%) from patients lacking features of PJS (7 of 22, 32% overall). Sequencing analysis of the STK11/LKB1 gene in these IPMNs with LOH revealed a germline mutation in one IPMN that arose in a patient with PJS and a somatic mutation in 1 of the 20 sporadic IPMNs. None of the 22 IPMNs showed hypermethylation of the STK11/LKB1 gene. These results suggest that the STK11/LKB1 gene is involved in the pathogenesis of some IPMNs.


Subject(s)
Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Papillary/genetics , Pancreatic Neoplasms/genetics , Peutz-Jeghers Syndrome/genetics , Protein Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinase Kinases , Adenocarcinoma, Mucinous/pathology , Adenocarcinoma, Papillary/pathology , Chromosomes, Human, Pair 19/genetics , DNA Methylation , DNA Mutational Analysis , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , Gene Silencing , Germ-Line Mutation , Humans , Loss of Heterozygosity , Mutation , Pancreatic Ducts/pathology , Pancreatic Neoplasms/pathology , Peutz-Jeghers Syndrome/pathology
8.
Clin Cancer Res ; 7(12): 3862-8, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11751476

ABSTRACT

PURPOSE: Effective new markers of pancreatic carcinoma are urgently needed. In a previous analysis of gene expression in pancreatic adenocarcinoma using serial analysis of gene expression (SAGE), we found that the tag for the mesothelin mRNA transcript was present in seven of eight SAGE libraries derived from pancreatic carcinomas but not in the two SAGE libraries derived from normal pancreatic duct epithelial cells. In this study, we evaluate the potential utility of mesothelin as a tumor marker for pancreatic adenocarcinoma. EXPERIMENTAL DESIGN: Mesothelin mRNA expression was evaluated in pancreatic adenocarcinomas using reverse-transcription PCR (RT-PCR) and in situ hybridization, whereas mesothelin protein expression was evaluated by immunohistochemistry. RESULTS: Using an online SAGE database (http://www.ncbi.nlm.gov/SAGE), we found the tag for mesothelin to be consistently present in the mesothelioma, ovarian cancer, and pancreatic cancer libraries but not in normal pancreas libraries. Mesothelin mRNA expression was confirmed by in situ hybridization in 4 of 4 resected primary pancreatic adenocarcinomas and by RT-PCR in 18 of 20 pancreatic cancer cell lines, whereas mesothelin protein expression was confirmed by immunohistochemistry in all 60 resected primary pancreatic adenocarcinomas studied. The adjacent normal pancreas in these 60 cases did not label, or at most only rare benign pancreatic ducts showed weak labeling for mesothelin. CONCLUSIONS: Mesothelin is a new marker for pancreatic adenocarcinoma identified by gene expression analysis. Mesothelin overexpression in pancreatic adenocarcinoma has potential diagnostic, imaging, and therapeutic implications.


Subject(s)
Adenocarcinoma/genetics , Antigens, Neoplasm/genetics , Biomarkers, Tumor/analysis , Gene Expression Regulation, Neoplastic , Membrane Glycoproteins/genetics , Pancreatic Neoplasms/genetics , Adenocarcinoma/chemistry , Adenocarcinoma/pathology , Antigens, Neoplasm/analysis , GPI-Linked Proteins , Humans , Immunohistochemistry , In Situ Hybridization , Membrane Glycoproteins/analysis , Mesothelin , Online Systems , Pancreatic Neoplasms/chemistry , Pancreatic Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Tumor Cells, Cultured
9.
Cancer J ; 7(4): 251-8, 2001.
Article in English | MEDLINE | ID: mdl-11561601

ABSTRACT

Until recently, pancreatic cancer was a poorly understood disease. Research in the past decade has shown conclusively, however, that pancreatic cancer is primarily genetic in nature. Inactivation with a variety of tumor-suppressor genes such as p16, DPC4, and p53, coupled with activation of oncogenes such as K-ras, are a few of the mutations that trigger the growth of cancerous cells. Understanding these mutations is critical to a better understanding of familial pancreatic cancer and to the development of gene-based screening tests and therapies. In this article, we review the genetic alterations identified in pancreatic cancer and provide examples of how this information can be applied to patient care.


Subject(s)
DNA, Mitochondrial/genetics , Genes, Tumor Suppressor/physiology , Oncogenes/physiology , Pancreatic Neoplasms/genetics , Humans , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology
10.
Ann Surg ; 234(3): 313-21; discussion 321-2, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11524584

ABSTRACT

OBJECTIVE: To assess the authors' experience with intraductal papillary mucinous neoplasms of the pancreas (IPMNs). SUMMARY BACKGROUND DATA: Intraductal papillary mucinous neoplasms of the pancreas are being recognized with increasing frequency. METHODS: All patients who underwent pancreatic resection for an IPMN at the Johns Hopkins Hospital between January 1987 and December 2000 were studied. The data were compared with those of 702 concurrent patients with infiltrating ductal adenocarcinoma of the pancreas not associated with an IPMN resected by pancreaticoduodenectomy. RESULTS: In the 13-year time period, 60 patients underwent pancreatic resection for IPMNs, with 40 patients undergoing resection in the past 3 years. Mean age at presentation was 67.4 +/- 1.4 years. The most common presenting symptom in patients with IPMNs was abdominal pain (59%). Most IPMNs were in the head of the pancreas or diffusely involved the gland, with 70% being resected via pancreaticoduodenectomy, 22% via total pancreatectomy, and 8% via distal pancreatectomy. Twenty-two patients (37%) had IPMNs with an associated infiltrating adenocarcinoma. In a subset of IPMNs immunohistochemically stained for the Dpc4 protein (n = 50), all of the intraductal or noninvasive components strongly expressed Dpc4, whereas 84% of associated infiltrating cancers expressed Dpc4. The 5-year survival rate for all patients with IPMNs (n = 60) was 57%. CONCLUSION: Intraductal papillary mucinous neoplasms represent a distinct clinicopathologic entity being recognized with increasing frequency. IPMNs are clinically, histologically, and genetically disparate from pancreatic ductal adenocarcinomas. The distinct clinical features, the presumably long in situ or noninvasive phase, and the good long-term survival of patients with IPMNs offer a unique opportunity for early diagnosis, curative resection, and further studies of the molecular genetics and natural history of these unusual neoplasms.


Subject(s)
Adenocarcinoma, Mucinous/pathology , Carcinoma, Papillary/pathology , Pancreatic Neoplasms/pathology , Adenocarcinoma, Mucinous/mortality , Adenocarcinoma, Mucinous/surgery , Adult , Aged , Aged, 80 and over , Carcinoma, Papillary/mortality , Carcinoma, Papillary/surgery , Female , Humans , Immunohistochemistry , Male , Middle Aged , Pancreatectomy , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/surgery , Pancreaticoduodenectomy , Retrospective Studies , Survival Rate
11.
Am J Surg Pathol ; 25(8): 1067-73, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11474292

ABSTRACT

Colchicine is an alkaloid with antimitotic ability used to treat a variety of medical conditions. Colchicine toxicity can result in multiorgan failure and death. The histopathologic features of colchicine toxicity in gastrointestinal biopsies have not been reported. Twenty-one gastrointestinal mucosal biopsies obtained from nine patients receiving oral colchicine therapy were studied. Immunohistochemical staining for Ki67 proliferation antigen was performed, and medical records of each patient were reviewed. All patients had a history of gout. Four patients with chronic renal failure also had clinical evidence of colchicine toxicity, and the other five patients did not. Distinct morphologic changes, seen as metaphase mitoses, epithelial pseudostratification, and loss of polarity, were seen in biopsy material from 4 of 4 (100%) patients with clinical colchicine toxicity. Three of these four cases (75%) also contained abundant crypt apoptotic bodies. These morphologic features were best seen in the biopsies from duodenum and gastric antrum, with relative sparing of the gastric body in the upper gastrointestinal tract. Ki67 staining demonstrated an expansion of the proliferating region in three available cases with clinical colchicine toxicity. These distinctive morphologic features were not seen in the five patients without clinical colchicine toxicity. These results indicate that colchicine toxicity can produce diagnostic morphologic features in gastrointestinal mucosal biopsies. Recognition of these features is important because colchicine toxicity can be fatal if undiagnosed clinically.


Subject(s)
Colchicine/adverse effects , Intestinal Diseases/chemically induced , Intestinal Mucosa/drug effects , Adult , Aged , Aged, 80 and over , Apoptosis , Biopsy , Colon/drug effects , Colon/pathology , Duodenum/drug effects , Duodenum/pathology , Female , Humans , Immunohistochemistry , Intestinal Diseases/metabolism , Intestinal Diseases/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Ki-67 Antigen/metabolism , Male , Middle Aged , Mitosis , Pyloric Antrum/drug effects , Pyloric Antrum/pathology , Rectum/drug effects , Rectum/pathology , Retrospective Studies
12.
Am J Surg Pathol ; 24(11): 1544-8, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11075857

ABSTRACT

DPC4 (MADH4, SMAD4) is a nuclear transcription factor shown to be genetically inactivated in over half of infiltrating ductal adenocarcinomas of the pancreas. Immunohistochemical labeling for the DPC4 gene product using a monoclonal antibody has recently been shown to be an extremely sensitive and specific marker for DPC4 gene alterations in pancreatic adenocarcinomas. Mucinous cystic neoplasms (MCNs) are a biologically less aggressive subtype of pancreatic neoplasm that may show benign, borderline, or overtly malignant features. However, the role of DPC4 inactivation in the development of MCNs has not been examined. The immunohistochemical expression of Dpc4 protein was therefore examined in 36 mucinous cystic neoplasms using this previously characterized monoclonal antibody. The 36 mucinous cystic neoplasms studied included 23 adenomas, 1 tumor with borderline potential, 5 tumors with carcinoma in situ, and 7 invasive carcinomas. Twenty-nine (100%) of the 29 noninvasive mucinous cystic neoplasms strongly expressed Dpc4 in the neoplastic epithelium. In striking contrast, only one (14%) of seven infiltrating carcinomas expressed Dpc4 in the neoplastic epithelium (p = 0.0001). The adjacent stroma retained expression of this protein in all 36 cases. In invasive MCNs with loss of Dpc4 expression, areas of carcinoma in situ were identified in the same paraffin sections, and these areas of carcinoma in situ retained expression of Dpc4. The frequent loss of Dpc4 expression in invasive MCNs indicates that genetic inactivation of Dpc4 occurs late in the neoplastic progression of these tumors and suggests a relationship to the development of invasion.


Subject(s)
Adenocarcinoma, Mucinous/metabolism , Adenoma/metabolism , Carcinoma in Situ/metabolism , DNA-Binding Proteins/metabolism , Genes, Tumor Suppressor , Pancreatic Neoplasms/metabolism , Trans-Activators/metabolism , Adenocarcinoma, Mucinous/secondary , Adenocarcinoma, Mucinous/surgery , Adenoma/pathology , Adenoma/surgery , Carcinoma in Situ/pathology , Carcinoma in Situ/surgery , Disease Progression , Female , Fluorescent Antibody Technique, Indirect , Follow-Up Studies , Humans , Lymph Nodes/pathology , Lymphatic Metastasis , Male , Middle Aged , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , Smad4 Protein
13.
Am J Pathol ; 157(3): 755-61, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10980115

ABSTRACT

DPC4 (MADH4, SMAD4) encodes a nuclear transcription factor shown to be genetically inactivated in over one-half of conventional infiltrating ductal adenocarcinomas of the pancreas. Intraductal papillary mucinous neoplasms (IPMNs) of the pancreas have been suggested to be distinct neoplasms with a significantly less aggressive course than conventional ductal adenocarcinomas of the pancreas, but molecular comparisons of these tumor types have previously been impaired by technical difficulties. Recently, immunohistochemical labeling for the DPC4 gene product has been shown to be an extremely sensitive and specific marker for DPC4 gene alterations in pancreatic adenocarcinomas. Therefore, we analyzed the immunohistochemical expression of Dpc4 protein in 79 IPMNs using a previously characterized monoclonal antibody. Twenty-nine of the IPMNs also had an associated infiltrating adenocarcinoma available for analysis. The labeling patterns observed were compared to those we have previously reported for conventional ductal carcinomas. All 79 of the intraductal components of the IPMNs strongly expressed Dpc4 protein. In 77 of the 79 cases (97%), the labeling was diffusely positive, and in 2 of the 79 (3%) the labeling was focally positive. Dpc4 expression was seen in 28 (97%) of the associated 29 invasive cancers. The one infiltrating carcinoma that showed loss of Dpc4 expression was associated with an intraductal component which showed focal loss of Dpc4 expression. The strong and almost universal expression of Dpc4 in IPMNs contrasts sharply with the loss of Dpc4 expression seen in approximately 30% of in situ adenocarcinomas of the pancreas (so-called pancreatic intraepithelial neoplasms, grade 3; P: < 0.001) and in 55% of pancreatic duct carcinomas (P: < 0.0001). Differences in Dpc4 expression between IPMNs and ductal carcinomas suggest a fundamental genetic difference in tumorigenesis, which may relate to the significantly better clinical outcomes observed for IPMNs.


Subject(s)
Adenocarcinoma, Mucinous/metabolism , Carcinoma, Pancreatic Ductal/metabolism , DNA-Binding Proteins/metabolism , Pancreatic Neoplasms/metabolism , Trans-Activators/metabolism , Adenocarcinoma, Mucinous/secondary , Adenoma/metabolism , Adenoma/pathology , Aged , Carcinoma in Situ/metabolism , Carcinoma in Situ/pathology , Carcinoma, Pancreatic Ductal/secondary , DNA-Binding Proteins/genetics , Female , Fluorescent Antibody Technique, Indirect , Genes, Tumor Suppressor , Humans , Lymph Nodes/pathology , Lymphatic Metastasis , Male , Pancreas/metabolism , Pancreas/pathology , Pancreas/surgery , Pancreatic Neoplasms/pathology , Smad4 Protein , Trans-Activators/genetics
14.
Cancer Res ; 60(7): 2002-6, 2000 Apr 01.
Article in English | MEDLINE | ID: mdl-10766191

ABSTRACT

Infiltrating adenocarcinomas of the pancreas are believed to arise from histologically identifiable intraductal precursors [pancreatic intraepithelial neoplasias (PanINs)] that undergo a series of architectural, cytological, and genetic changes. The role of DPC4 tumor suppressor gene inactivation in this progression has not been defined. Immunohistochemistry for the Dpc4 protein in formalin-fixed, paraffin-embedded tissue is a sensitive and specific marker for DPC4 gene status, providing a tool to examine DPC4 status in these putative precursor lesions. A total of 188 PanINs were identified in 40 pancreata, 38 (95%) of which also contained an infiltrating adenocarcinoma. Sections containing these 188 duct lesions were labeled with a monoclonal antibody to Dpc4. All 82 flat (PanIN-1A), all 54 papillary (PanIN-1B), and all 23 atypical papillary (PanIN-2) intraductal lesions expressed Dpc4. In contrast, 9 of 29 (31%) severely atypical lesions (PanIN-3 lesions, carcinomas in situ) did not. The difference in Dpc4 expression between histologically low-grade (PanIN-1 and -2) and histologically high-grade (PanIN-3) duct lesions was statistically significant (P < 0.0001). In three cases, the pattern of Dpc4 expression in the PanIN-3 lesions did not match the pattern of expression in the associated infiltrating carcinomas, indicating that these high-grade lesions did not simply represent infiltrating carcinoma growing along benign ducts. Loss of Dpc4 expression occurs biologically late in the neoplastic progression that leads to the development of infiltrating pancreatic cancer, at the stage of histologically recognizable carcinoma.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , DNA-Binding Proteins/genetics , Gene Silencing , Genes, Tumor Suppressor , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Trans-Activators/genetics , Adenocarcinoma/surgery , DNA-Binding Proteins/analysis , Disease Progression , Humans , Pancreatic Ducts/pathology , Pancreatic Neoplasms/surgery , Smad4 Protein , Trans-Activators/analysis
15.
Thyroid ; 9(6): 569-77, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10411119

ABSTRACT

Cathepsin B (CB) is involved in the hydrolysis of thyroglobulin (Tg) and thought to be regulated by thyroid stimulating hormone (TSH) in the normal thyroid. Our analyses of 91 thyroid tissues from 71 patients with Graves' disease (GD), multinodular goiter (MNG), papillary carcinoma (PC), or follicular carcinoma (FC), demonstrated a 2-fold increase in expression of CB in GD and an average increase of 1.5-fold in MNG (varying from 10-fold below normal to 6-fold above normal in MNG nodules), as might be predicted by the altered functional status of thyroid follicular cells in those diseases. However, CB activity was not downregulated in conjunction with the known "blocking effect" of malignancy on many thyroid functions, but rather increased on average 9-fold in papillary carcinomas (n = 33), and also showed a marked increase in 2 follicular carcinomas. Activity measurements were confirmed by CB protein detection on Western blot with moderately increased CB protein levels demonstrated in GD, variable expression in nodules of MNG, and markedly increased protein expression in carcinomas. In all diseased states, increased protein was detected primarily as overexpression of the 27 kd heavy chain of 2-chain mature CB and less frequently as overexpression of 31 kd single-chain mature CB. However, an additional 35 kd protein form was noted in 3 of 9 PCs, 1 of 2 FCs, and 1 of 4 GD cases but in none of 10 MNG cases. In conjunction with elevated CB activity plus additional protein bands on Western blots, altered patterns of CB immunohistochemical staining were observed, irrespective of the type of thyroid disease, suggesting certain common changes in CB expression, posttranslational processing, and vesicular trafficking. In summary, GD and MNG thyroid tissues demonstrated altered CB expression in keeping with predicted functional changes in thyroid follicular cells, while increased CB expression in carcinomas indicated a more pathological role for CB in thyroid cancers, possibly related to the processes of invasion or metastasis.


Subject(s)
Cathepsin B/metabolism , Goiter, Nodular/enzymology , Graves Disease/enzymology , Proteins/metabolism , Thyroid Neoplasms/enzymology , Blotting, Western , Goiter, Nodular/metabolism , Goiter, Nodular/pathology , Graves Disease/metabolism , Graves Disease/pathology , Humans , Immunohistochemistry , Subcellular Fractions/enzymology , Subcellular Fractions/metabolism , Thyroid Hormones/metabolism , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology
16.
Dis Colon Rectum ; 42(1): 24-30, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10211516

ABSTRACT

PURPOSE: Cytomegaloviral enterocolitis is an uncommon disorder that can complicate inflammatory bowel disease. It is more common in patients with acquired immunodeficiency syndrome and can occur in patients on immunosuppressive therapy for autoimmune or inflammatory diseases and in allograft recipients. Mortality rates of up to 71 percent have been reported for cytomegaloviral enterocolitis. The aims of this study were 1) to identify the presentation, underlying medical conditions, treatment, and outcome of patients with cytomegaloviral enterocolitis and 2) to determine the prevalence of this infection in patients undergoing intestinal resection for inflammatory bowel disease. METHODS: A retrospective chart review of patients with pathologic evidence of cytomegaloviral enterocolitis from 1985 through 1996 was performed. To determine the prevalence of this condition, the hospital discharge database was searched for the diagnoses of ulcerative colitis and Crohn's disease in patients who underwent bowel resection. RESULTS: 93 patients (mean age, 44 years; 66 percent males) had cytomegaloviral infection in the small intestine (n = 6), large intestine (n = 86), or appendix (n = 1). Patients with acquired immunodeficiency syndrome (n = 42), with ulcerative colitis (n = 11), with Crohn's disease (n = 11), receiving organ transplant (n = 12), receiving bone marrow transplant (n = 8), and in other immunosuppressed states (n = 11) comprised this study. Seventeen patients (18 percent) underwent intestinal resection, and the remaining 76 patients were treated medically. Abdominal pain (77 vs. 37 percent; P < 0.01) and gastrointestinal bleeding (65 vs. 34 percent; P < 0.05) were more common presenting symptoms in patients who required resection than patients in the medically managed group. Mortality was 17.6 percent in the surgically managed group and 14.5 percent in the patients who were managed medically. The median duration of ulcerative colitis in patients with coexisting cytomegaloviral infection was 12 months. The prevalence of cytomegaloviral enterocolitis was 4.6 percent in patients with ulcerative colitis and 0.8 percent in patients with Crohn's disease. CONCLUSIONS: These data suggest that cytomegaloviral infection more frequently complicates ulcerative colitis than Crohn's disease. Furthermore, a short and fulminant course of ulcerative colitis may indicate coexisting cytomegaloviral infection. The overall low mortality in this retrospective study suggests that aggressive medical and surgical treatment improves survival in patients with cytomegaloviral enterocolitis.


Subject(s)
Cytomegalovirus Infections/complications , Enterocolitis/complications , Inflammatory Bowel Diseases/complications , Acquired Immunodeficiency Syndrome/complications , Adult , Aged , Aged, 80 and over , Colitis, Ulcerative/complications , Crohn Disease/complications , Cytomegalovirus Infections/mortality , Cytomegalovirus Infections/pathology , Enterocolitis/mortality , Enterocolitis/pathology , Female , Humans , Immunocompromised Host , Male , Middle Aged , Retrospective Studies
17.
J Biol Chem ; 272(46): 29190-9, 1997 Nov 14.
Article in English | MEDLINE | ID: mdl-9360997

ABSTRACT

Western blots, enzyme assays, protein glycosylation studies, and immunohistochemical staining were used to characterize cathepsin B expression at successive stages of colorectal tumor progression. In normal colon mucosa and premalignant adenomas, cathepsin B expression was predominantly due to mature two-chain protein detected on Western blots as the nonglycosylated 27-kDa form, with overexpression of this protein occurring in only 4 of 18 adenomas. Overexpression increased significantly in Dukes A and B carcinomas (26 of 37 cases), with cathepsin B protein generally detectable in carcinomas as a combination of both 27-kDa nonglycosylated and 28-kDa glycosylated mature two-chain forms. Glycosylated cathepsin B protein in carcinoma extracts was sensitive to PNGase F but resistant to Endo H, indicating a pattern consistent with complex rather than high mannose type glycosylation. When sorted by advancing tumor stage, peak expression of cathepsin B protein occurred in carcinomas involved in local invasion compared with adenomas or metastatic cancers. At all stages, cathepsin B activity correlated significantly with the levels of heavy chain mature cathepsin B protein (r = 0.6682, p < 0.0001) irrespective of glycosylation. Immunohistochemical staining of cathepsin B protein revealed fine diffuse cytoplasmic staining in both adenomas and carcinomas compared with coarse granular cytoplasmic staining (typical of lysosomes) seen in matched normal mucosa. Our results demonstrate several sequential, apparently independent changes in cathepsin B expression during colorectal tumor progression including early changes in subcellular localization, up-regulation of cathepsin B protein and activity in invasive cancers, and altered protein glycosylation detected in malignant tumors at all stages.


Subject(s)
Cathepsin B/metabolism , Colorectal Neoplasms/enzymology , Adenoma/enzymology , Adenoma/pathology , Blotting, Western , Carcinoma/enzymology , Carcinoma/pathology , Colorectal Neoplasms/pathology , Disease Progression , Enzyme Activation , Glycosylation , Humans , Intestinal Mucosa/enzymology , Subcellular Fractions/enzymology
18.
In Vivo ; 11(3): 209-16, 1997.
Article in English | MEDLINE | ID: mdl-9239513

ABSTRACT

Clinicopathologic staging of colorectal cancers cannot always predict aggressiveness of prognosis for a particular patient. We have used activity assays for cysteine proteinases, cathepsins B, L and H (CB, CL and CH) and matrix metalloproteinases, MMP-2 and MMP-9, to identify several distinctive, reproducible proteolytic profiles in a large set of colorectal carcinomas. We observed that individual proteinases demonstrated specific and distinct levels of activity at different cancer stages, possible reflecting non-random steps in a proteolytic cascade related to tumor development. We also observed that individual colon cancers fell into relatively few categories when characterized for the combined expression of three proteinases: CB, CL and MMP-9. Four proteolytic profiles, designated "Early", "Middle", "Late", and "High", could be used to define almost 80% of the colorectal carcinomas analyzed. Such profiles, based on the expression of several proteinases in a given tumor, provided information independent of clinical stage and may identify crucial variations in tumor behavior.


Subject(s)
Biomarkers, Tumor , Colorectal Neoplasms/enzymology , Endopeptidases , Cathepsin B/metabolism , Cathepsin H , Cathepsin L , Cathepsins/metabolism , Collagenases/metabolism , Colorectal Neoplasms/chemistry , Colorectal Neoplasms/diagnosis , Cysteine Endopeptidases/metabolism , Gelatinases/metabolism , Humans , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 9 , Metalloendopeptidases/metabolism , Neoplasm Staging
SELECTION OF CITATIONS
SEARCH DETAIL
...