Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Proc Natl Acad Sci U S A ; 112(44): E5916-25, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26489647

ABSTRACT

Fibrolamellar hepatocellular carcinoma (FLHCC) tumors all carry a deletion of ∼ 400 kb in chromosome 19, resulting in a fusion of the genes for the heat shock protein, DNAJ (Hsp40) homolog, subfamily B, member 1, DNAJB1, and the catalytic subunit of protein kinase A, PRKACA. The resulting chimeric transcript produces a fusion protein that retains kinase activity. No other recurrent genomic alterations have been identified. Here we characterize the molecular pathogenesis of FLHCC with transcriptome sequencing (RNA sequencing). Differential expression (tumor vs. adjacent normal tissue) was detected for more than 3,500 genes (log2 fold change ≥ 1, false discovery rate ≤ 0.01), many of which were distinct from those found in hepatocellular carcinoma. Expression of several known oncogenes, such as ErbB2 and Aurora Kinase A, was increased in tumor samples. These and other dysregulated genes may serve as potential targets for therapeutic intervention.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Transcriptome , Gene Expression Regulation, Neoplastic , Humans , Polymerase Chain Reaction
2.
Mod Pathol ; 30(12): 1760-1772, 2017 12.
Article in English | MEDLINE | ID: mdl-28776573

ABSTRACT

Intraductal tubulopapillary neoplasm is a relatively recently described member of the pancreatic intraductal neoplasm family. The more common member of this family, intraductal papillary mucinous neoplasm, often carries genetic alterations typical of pancreatic infiltrating ductal adenocarcinoma (KRAS, TP53, and CDKN2A) but additionally has mutations in GNAS and RNF43 genes. However, the genetic characteristics of intraductal tubulopapillary neoplasm have not been well characterized. Twenty-two intraductal tubulopapillary neoplasms were analyzed by either targeted next-generation sequencing, which enabled the identification of sequence mutations, copy number alterations, and selected structural rearrangements involving all targeted (≥300) genes, or whole-exome sequencing. Three of these intraductal tubulopapillary neoplasms were also subjected to whole-genome sequencing. All intraductal tubulopapillary neoplasms revealed the characteristic histologic (cellular intraductal nodules of back-to-back tubular glands lined by predominantly cuboidal cells with atypical nuclei and no obvious intracellular mucin) and immunohistochemical (immunolabeled with MUC1 and MUC6 but were negative for MUC2 and MUC5AC) features. By genomic analyses, there was loss of CDKN2A in 5/20 (25%) of these cases. However, the majority of the previously reported intraductal papillary mucinous neoplasm-related alterations were absent. Moreover, in contrast to most ductal neoplasms of the pancreas, MAP-kinase pathway was not involved. In fact, 2/22 (9%) of intraductal tubulopapillary neoplasms did not reveal any mutations in the tested genes. However, certain chromatin remodeling genes (MLL1, MLL2, MLL3, BAP1, PBRM1, EED, and ATRX) were found to be mutated in 7/22 (32%) of intraductal tubulopapillary neoplasms and 27% harbored phosphatidylinositol 3-kinase (PI3K) pathway (PIK3CA, PIK3CB, INPP4A, and PTEN) mutations. In addition, 4/18 (18%) of intraductal tubulopapillary neoplasms had FGFR2 fusions (FGFR2-CEP55, FGFR2-SASS6, DISP1-FGFR2, FGFR2-TXLNA, and FGFR2-VCL) and 1/18 (5.5%) had STRN-ALK fusion. Intraductal tubulopapillary neoplasm is a distinct clinicopathologic entity in the pancreas. Although its intraductal nature and some clinicopathologic features resemble those of intraductal papillary mucinous neoplasm, our results suggest that intraductal tubulopapillary neoplasm has distinguishing genetic characteristics. Some of these mutated genes are potentially targetable. Future functional studies will be needed to determine the consequences of these gene alterations.


Subject(s)
Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Papillary/genetics , Biomarkers, Tumor/genetics , Carcinoma, Pancreatic Ductal/genetics , Pancreatic Neoplasms/genetics , Adenocarcinoma, Mucinous/pathology , Adenocarcinoma, Papillary/pathology , Adult , Aged , Carcinoma, Pancreatic Ductal/pathology , DNA Mutational Analysis , Female , Humans , Male , Middle Aged , Pancreatic Neoplasms/pathology , Young Adult
3.
Proc Natl Acad Sci U S A ; 110(33): 13481-6, 2013 Aug 13.
Article in English | MEDLINE | ID: mdl-23901115

ABSTRACT

Synonymous mutations, which do not alter the protein sequence, have been shown to affect protein function [Sauna ZE, Kimchi-Sarfaty C (2011) Nat Rev Genet 12(10):683-691]. However, synonymous mutations are rarely investigated in the cancer genomics field. We used whole-genome and -exome sequencing to identify somatic mutations in 29 melanoma samples. Validation of one synonymous somatic mutation in BCL2L12 in 285 samples identified 12 cases that harbored the recurrent F17F mutation. This mutation led to increased BCL2L12 mRNA and protein levels because of differential targeting of WT and mutant BCL2L12 by hsa-miR-671-5p. Protein made from mutant BCL2L12 transcript bound p53, inhibited UV-induced apoptosis more efficiently than WT BCL2L12, and reduced endogenous p53 target gene transcription. This report shows selection of a recurrent somatic synonymous mutation in cancer. Our data indicate that silent alterations have a role to play in human cancer, emphasizing the importance of their investigation in future cancer genome studies.


Subject(s)
Apoptosis/genetics , Gene Expression Regulation/genetics , Genome, Human/genetics , Melanoma/genetics , Muscle Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Base Sequence , Blotting, Western , DNA Primers/genetics , Exome/genetics , Genetic Vectors/genetics , HEK293 Cells , Humans , Immunoprecipitation , Lentivirus , MicroRNAs/genetics , Molecular Sequence Data , Muscle Proteins/metabolism , Mutation/genetics , Polymorphism, Single Nucleotide/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA , Tumor Suppressor Protein p53/metabolism
4.
PLoS Genet ; 8(8): e1002871, 2012.
Article in English | MEDLINE | ID: mdl-22912592

ABSTRACT

Much emphasis has been placed on the identification, functional characterization, and therapeutic potential of somatic variants in tumor genomes. However, the majority of somatic variants lie outside coding regions and their role in cancer progression remains to be determined. In order to establish a system to test the functional importance of non-coding somatic variants in cancer, we created a low-passage cell culture of a metastatic melanoma tumor sample. As a foundation for interpreting functional assays, we performed whole-genome sequencing and analysis of this cell culture, the metastatic tumor from which it was derived, and the patient-matched normal genomes. When comparing somatic mutations identified in the cell culture and tissue genomes, we observe concordance at the majority of single nucleotide variants, whereas copy number changes are more variable. To understand the functional impact of non-coding somatic variation, we leveraged functional data generated by the ENCODE Project Consortium. We analyzed regulatory regions derived from multiple different cell types and found that melanocyte-specific regions are among the most depleted for somatic mutation accumulation. Significant depletion in other cell types suggests the metastatic melanoma cells de-differentiated to a more basal regulatory state. Experimental identification of genome-wide regulatory sites in two different melanoma samples supports this observation. Together, these results show that mutation accumulation in metastatic melanoma is nonrandom across the genome and that a de-differentiated regulatory architecture is common among different samples. Our findings enable identification of the underlying genetic components of melanoma and define the differences between a tissue-derived tumor sample and the cell culture created from it. Such information helps establish a broader mechanistic understanding of the linkage between non-coding genomic variations and the cellular evolution of cancer.


Subject(s)
Cell Dedifferentiation/genetics , DNA, Intergenic , Melanoma/genetics , Neoplasm Metastasis , Polymorphism, Single Nucleotide , Adult , DNA Copy Number Variations , Genome, Human , Genome-Wide Association Study , Humans , Male , Melanocytes/metabolism , Melanocytes/pathology , Primary Cell Culture , Regulatory Sequences, Nucleic Acid , Tumor Cells, Cultured
5.
J Cutan Pathol ; 41(8): 663-71, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24628623

ABSTRACT

Phacomatosis pigmentokeratotica (PPK) is characterized by the co-existence of epidermal nevi and large segmental speckled lentiginous nevi of the papulosa type. PPK, previously explained as 'twin spot' mosaicism due to the postzygotic crossing-over of two homozygous recessive mutations, has recently been shown to derive from one postzygotic activating RAS mutation. Epidermal nevi, including those in PPK, are known to give rise to neoplasms such as trichoblastoma and basal cell carcinoma. Within speckled lentiginous nevi, Spitz nevi and melanoma have been well documented. We report a case of PPK with a combined melanocytic and adnexal neoplasm presenting where the nevi conjoined. Using next-generation sequencing techniques, we were able to identify the same HRAS G13R mutation within both components of the tumor, and to show the absence of additional mutated modifier genes in a panel of 300 cancer-related genes. Given the genetic findings in this rare tumor-type, we suggest that this case may be used as a model for understanding the development of biphenotypic neoplasia or intratumoral heterogeneity in some cases.


Subject(s)
Mutation , Neoplasms, Complex and Mixed/genetics , Neoplasms, Complex and Mixed/pathology , Nevus, Pigmented/genetics , Nevus, Pigmented/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Adolescent , DNA Mutational Analysis , Female , High-Throughput Nucleotide Sequencing , Humans , Laser Capture Microdissection , Melanoma/genetics , Melanoma/pathology , Sweat Gland Neoplasms/genetics , Sweat Gland Neoplasms/pathology
6.
Cancer Cell ; 42(2): 209-224.e9, 2024 02 12.
Article in English | MEDLINE | ID: mdl-38215748

ABSTRACT

Although immunotherapy with PD-(L)1 blockade is routine for lung cancer, little is known about acquired resistance. Among 1,201 patients with non-small cell lung cancer (NSCLC) treated with PD-(L)1 blockade, acquired resistance is common, occurring in >60% of initial responders. Acquired resistance shows differential expression of inflammation and interferon (IFN) signaling. Relapsed tumors can be separated by upregulated or stable expression of IFNγ response genes. Upregulation of IFNγ response genes is associated with putative routes of resistance characterized by signatures of persistent IFN signaling, immune dysfunction, and mutations in antigen presentation genes which can be recapitulated in multiple murine models of acquired resistance to PD-(L)1 blockade after in vitro IFNγ treatment. Acquired resistance to PD-(L)1 blockade in NSCLC is associated with an ongoing, but altered IFN response. The persistently inflamed, rather than excluded or deserted, tumor microenvironment of acquired resistance may inform therapeutic strategies to effectively reprogram and reverse acquired resistance.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Animals , Mice , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Signal Transduction , Immunotherapy , Antigen Presentation , B7-H1 Antigen/metabolism , Tumor Microenvironment
7.
Sci Transl Med ; 15(707): eadf7006, 2023 08 02.
Article in English | MEDLINE | ID: mdl-37531417

ABSTRACT

In lung and prostate adenocarcinomas, neuroendocrine (NE) transformation to an aggressive derivative resembling small cell lung cancer (SCLC) is associated with poor prognosis. We previously described dependency of SCLC on the nuclear transporter exportin 1. Here, we explored the role of exportin 1 in NE transformation. We observed up-regulated exportin 1 in lung and prostate pretransformation adenocarcinomas. Exportin 1 was up-regulated after genetic inactivation of TP53 and RB1 in lung and prostate adenocarcinoma cell lines, accompanied by increased sensitivity to the exportin 1 inhibitor selinexor in vitro. Exportin 1 inhibition prevented NE transformation in different TP53/RB1-inactivated prostate adenocarcinoma xenograft models that acquire NE features upon treatment with the aromatase inhibitor enzalutamide and extended response to the EGFR inhibitor osimertinib in a lung cancer transformation patient-derived xenograft (PDX) model exhibiting combined adenocarcinoma/SCLC histology. Ectopic SOX2 expression restored the enzalutamide-promoted NE phenotype on adenocarcinoma-to-NE transformation xenograft models despite selinexor treatment. Selinexor sensitized NE-transformed lung and prostate small cell carcinoma PDXs to standard cytotoxics. Together, these data nominate exportin 1 inhibition as a potential therapeutic target to constrain lineage plasticity and prevent or treat NE transformation in lung and prostate adenocarcinoma.


Subject(s)
Adenocarcinoma , Lung Neoplasms , Prostatic Neoplasms , SOXB1 Transcription Factors , Small Cell Lung Carcinoma , Humans , Male , Adenocarcinoma/pathology , Down-Regulation , Lung Neoplasms/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Small Cell Lung Carcinoma/genetics , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Animals , Exportin 1 Protein
8.
Cancer Cell ; 41(4): 776-790.e7, 2023 04 10.
Article in English | MEDLINE | ID: mdl-37001526

ABSTRACT

Paired single-cell RNA and T cell receptor sequencing (scRNA/TCR-seq) has allowed for enhanced resolution of clonal T cell dynamics in cancer. Here, we report a scRNA/TCR-seq analysis of 187,650 T cells from 31 tissue regions, including tumor, adjacent normal tissues, and lymph nodes (LN), from three patients with non-small cell lung cancer after immune checkpoint blockade (ICB). Regions with viable cancer cells are enriched for exhausted CD8+ T cells, regulatory CD4+ T cells (Treg), and follicular helper CD4+ T cells (TFH). Tracking T cell clonotypes across tissues, combined with neoantigen specificity assays, reveals that TFH and tumor-specific exhausted CD8+ T cells are clonally linked to TCF7+SELL+ progenitors in tumor draining LNs, and progressive exhaustion trajectories of CD8+ T, Treg, and TFH cells with proximity to the tumor microenvironment. Finally, longitudinal tracking of tumor-specific CD8+ and CD4+ T cell clones reveals persistence in the peripheral blood for years after ICB therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , CD8-Positive T-Lymphocytes , Carcinoma, Non-Small-Cell Lung/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Receptors, Antigen, T-Cell , Clone Cells , Tumor Microenvironment
9.
Cancer Res ; 83(22): 3796-3812, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37812025

ABSTRACT

Multiple large-scale genomic profiling efforts have been undertaken in osteosarcoma to define the genomic drivers of tumorigenesis, therapeutic response, and disease recurrence. The spatial and temporal intratumor heterogeneity could also play a role in promoting tumor growth and treatment resistance. We conducted longitudinal whole-genome sequencing of 37 tumor samples from 8 patients with relapsed or refractory osteosarcoma. Each patient had at least one sample from a primary site and a metastatic or relapse site. Subclonal copy-number alterations were identified in all patients except one. In 5 patients, subclones from the primary tumor emerged and dominated at subsequent relapses. MYC gain/amplification was enriched in the treatment-resistant clones in 6 of 7 patients with multiple clones. Amplifications in other potential driver genes, such as CCNE1, RAD21, VEGFA, and IGF1R, were also observed in the resistant copy-number clones. A chromosomal duplication timing analysis revealed that complex genomic rearrangements typically occurred prior to diagnosis, supporting a macroevolutionary model of evolution, where a large number of genomic aberrations are acquired over a short period of time followed by clonal selection, as opposed to ongoing evolution. A mutational signature analysis of recurrent tumors revealed that homologous repair deficiency (HRD)-related SBS3 increases at each time point in patients with recurrent disease, suggesting that HRD continues to be an active mutagenic process after diagnosis. Overall, by examining the clonal relationships between temporally and spatially separated samples from patients with relapsed/refractory osteosarcoma, this study sheds light on the intratumor heterogeneity and potential drivers of treatment resistance in this disease. SIGNIFICANCE: The chemoresistant population in recurrent osteosarcoma is subclonal at diagnosis, emerges at the time of primary resection due to selective pressure from neoadjuvant chemotherapy, and is characterized by unique oncogenic amplifications.


Subject(s)
Bone Neoplasms , Osteosarcoma , Humans , Osteosarcoma/genetics , Whole Genome Sequencing , Genomics , Bone Neoplasms/genetics , Recurrence , DNA Copy Number Variations , Mutation
10.
J Hematol Oncol ; 14(1): 170, 2021 10 16.
Article in English | MEDLINE | ID: mdl-34656143

ABSTRACT

BACKGROUND: Lineage plasticity, the ability to transdifferentiate among distinct phenotypic identities, facilitates therapeutic resistance in cancer. In lung adenocarcinomas (LUADs), this phenomenon includes small cell and squamous cell (LUSC) histologic transformation in the context of acquired resistance to targeted inhibition of driver mutations. LUAD-to-LUSC transdifferentiation, occurring in up to 9% of EGFR-mutant patients relapsed on osimertinib, is associated with notably poor prognosis. We hypothesized that multi-parameter profiling of the components of mixed histology (LUAD/LUSC) tumors could provide insight into factors licensing lineage plasticity between these histologies. METHODS: We performed genomic, epigenomics, transcriptomics and protein analyses of microdissected LUAD and LUSC components from mixed histology tumors, pre-/post-transformation tumors and reference non-transformed LUAD and LUSC samples. We validated our findings through genetic manipulation of preclinical models in vitro and in vivo and performed patient-derived xenograft (PDX) treatments to validate potential therapeutic targets in a LUAD PDX model acquiring LUSC features after osimertinib treatment. RESULTS: Our data suggest that LUSC transdifferentiation is primarily driven by transcriptional reprogramming rather than mutational events. We observed consistent relative upregulation of PI3K/AKT, MYC and PRC2 pathway genes. Concurrent activation of PI3K/AKT and MYC induced squamous features in EGFR-mutant LUAD preclinical models. Pharmacologic inhibition of EZH1/2 in combination with osimertinib prevented relapse with squamous-features in an EGFR-mutant patient-derived xenograft model, and inhibition of EZH1/2 or PI3K/AKT signaling re-sensitized resistant squamous-like tumors to osimertinib. CONCLUSIONS: Our findings provide the first comprehensive molecular characterization of LUSC transdifferentiation, suggesting putative drivers and potential therapeutic targets to constrain or prevent lineage plasticity.


Subject(s)
Adenocarcinoma of Lung/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/pathology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Transdifferentiation , Humans , Mice, Inbred NOD , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-myc/genetics , Signal Transduction , Transcriptome
11.
Cancer Cell ; 39(11): 1479-1496.e18, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34653364

ABSTRACT

Small cell lung cancer (SCLC) is an aggressive malignancy that includes subtypes defined by differential expression of ASCL1, NEUROD1, and POU2F3 (SCLC-A, -N, and -P, respectively). To define the heterogeneity of tumors and their associated microenvironments across subtypes, we sequenced 155,098 transcriptomes from 21 human biospecimens, including 54,523 SCLC transcriptomes. We observe greater tumor diversity in SCLC than lung adenocarcinoma, driven by canonical, intermediate, and admixed subtypes. We discover a PLCG2-high SCLC phenotype with stem-like, pro-metastatic features that recurs across subtypes and predicts worse overall survival. SCLC exhibits greater immune sequestration and less immune infiltration than lung adenocarcinoma, and SCLC-N shows less immune infiltrate and greater T cell dysfunction than SCLC-A. We identify a profibrotic, immunosuppressive monocyte/macrophage population in SCLC tumors that is particularly associated with the recurrent, PLCG2-high subpopulation.


Subject(s)
Gene Expression Profiling/methods , Lung Neoplasms/genetics , Phospholipase C gamma/genetics , Small Cell Lung Carcinoma/genetics , Cell Plasticity , Humans , Neoplasm Metastasis , Prognosis , Sequence Analysis, RNA , Single-Cell Analysis , Survival Analysis
12.
Cancer Discov ; 11(12): 3028-3047, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34155000

ABSTRACT

Lineage plasticity is implicated in treatment resistance in multiple cancers. In lung adenocarcinomas (LUAD) amenable to targeted therapy, transformation to small cell lung cancer (SCLC) is a recognized resistance mechanism. Defining molecular mechanisms of neuroendocrine (NE) transformation in lung cancer has been limited by a paucity of pre/posttransformation clinical samples. Detailed genomic, epigenomic, transcriptomic, and protein characterization of combined LUAD/SCLC tumors, as well as pre/posttransformation samples, supports that NE transformation is primarily driven by transcriptional reprogramming rather than mutational events. We identify genomic contexts in which NE transformation is favored, including frequent loss of the 3p chromosome arm. We observed enhanced expression of genes involved in the PRC2 complex and PI3K/AKT and NOTCH pathways. Pharmacologic inhibition of the PI3K/AKT pathway delayed tumor growth and NE transformation in an EGFR-mutant patient-derived xenograft model. Our findings define a novel landscape of potential drivers and therapeutic vulnerabilities of NE transformation in lung cancer. SIGNIFICANCE: The difficulty in collection of transformation samples has precluded the performance of molecular analyses, and thus little is known about the lineage plasticity mechanisms leading to LUAD-to-SCLC transformation. Here, we describe biological pathways dysregulated upon transformation and identify potential predictors and potential therapeutic vulnerabilities of NE transformation in the lung. See related commentary by Meador and Lovly, p. 2962. This article is highlighted in the In This Issue feature, p. 2945.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Neuroendocrine Tumors , Small Cell Lung Carcinoma , Adenocarcinoma of Lung/drug therapy , Humans , Lung Neoplasms/drug therapy , Mutation , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/pathology , Phosphatidylinositol 3-Kinases/genetics , Small Cell Lung Carcinoma/pathology
13.
Lab Invest ; 89(5): 489-97, 2009 May.
Article in English | MEDLINE | ID: mdl-19308045

ABSTRACT

The pathobiology of chronic pancreatitis (CP) remains enigmatic despite remarkable progress made recently in uncovering key mechanisms involved in the initiation and progression of the disease. CP is increasingly thought of as a multifactorial disorder. Apoptosis plays a role in parenchymal destruction, the pathological hallmark of CP. The apoptotic mechanisms preferentially target the exocrine compartment, leaving endocrine islets relatively intact for a prolonged period. Exocrine cells shed their 'immunoprivileged' status, express death receptors, and are rendered susceptible to apoptosis induced by death ligands on infiltrating lymphocytes, and released locally by activated pancreatic stellate cells. Islet cells retain their 'immunoprivileged' status and activate anti-apoptotic programs through NF-kappaB. Ductal changes, including distortion, dilatation, and pancreatic ductal hypertension in the setting of CP, induce genomic damage and increased cell turnover. In addition, signaling mechanisms that play a role in the development of embryonic pancreas are reinstated, thus, playing a role in repair, regeneration, and transformation. This, in turn, leads to acino-ductal metaplasia (ADM) and pancreatic intraepithelial neoplasia (PanIN). Some of these pathways are activated in pancreatic cancer. We attempt to integrate the current knowledge and major concepts in the pathogenesis of CP and to explain the mechanism of differential cell loss. We also discuss the possible implications of signaling pathway activation in pancreatic inflammation, relevant to the cellular transformation that leads to pancreatic neoplasia.


Subject(s)
Pancreatic Neoplasms/etiology , Pancreatitis, Chronic/physiopathology , Cyclooxygenase 2/metabolism , Hedgehog Proteins/metabolism , Humans , Inhibitor of Apoptosis Proteins/physiology , NF-kappa B/physiology , Pancreatic Ducts/metabolism , Pancreatitis, Chronic/etiology , Pancreatitis, Chronic/immunology , Receptors, Death Domain/physiology , Receptors, Notch/metabolism , Signal Transduction
14.
Oncotarget ; 9(12): 10211-10227, 2018 Feb 13.
Article in English | MEDLINE | ID: mdl-29535801

ABSTRACT

Fibrolamellar hepatocellular carcinoma (FLC) is a rare primary liver cancer found in adolescents and young adults without underlying liver disease. A deletion of ~400 kD has been found in one copy of chromosome 19 in the tumor tissue of all patients tested. This produces a fusion of the genes DNAJB1 and PRKACA which, in turn, produces a chimeric transcript and protein. Transcriptomic analysis of the tumor has shown upregulation of various oncologically relevant pathways, including EGF/ErbB, Aurora Kinase A, pak21 and wnt. To explore other factors that may contribute to oncogenesis, we examined the microRNA (miRNA) and long non-coding RNA (lncRNA) expression in FLC. The non-coding RNA expression profile in tumor tissue samples is distinctly different from the adjacent normal liver and from other liver tumors. Furthermore, miRZip knock down or over expression of certain miRNAs led to changes in the levels of coding genes that recapitulated changes observed in FLC, suggesting mechanistically that the changes in the cellular levels of miRNA are not merely correlative. Thus, in addition to serving as diagnostic tools for FLC, non-coding RNAs may serve as therapeutic targets.

16.
Verh Dtsch Ges Pathol ; 90: 159-67, 2006.
Article in German | MEDLINE | ID: mdl-17867593

ABSTRACT

In chronic pancreatitis (CP), fibrous replacement of exocrine tissue spares islets. There is local production of IFNgamma and death ligands by inflammatory cells as well as TGFbeta and TRAIL by pancreatic stellate cells (PSCs), along with functional death receptor neo-expression and apoptosis in exocrine but not in endocrine cells. Moreover, islets are strongly induced for TRAIL-receptor(R)-4 lacking a functional death domain. TRAIL-R4 signalling in T-cells induces NFkappaB transcription factors which activate anti-apoptotic programs. Whether TRAIL elicits this response in endocrine cells, we tested human insulinoma cell line CM and determined NFkappaB subunits transcripts and NFkappaB dependent inhibitor of apoptosis proteins (IAPs) in normal pancreas (NP) and CP. We treated CM with cytokines, determined TRAIL-R expression by flow cytometry, graded degree of fibrosis in CP specimens, microdissected epithelial compartments, performed real time PCRs for NFkappaB subunits transcripts, and immunohistochemistry for IKK-gamma, IkappaB-alpha, RelA, survivin, and cIAP1. In CM, TGFbeta/IFNgamma/TRAIL induced TRAIL-R4 surface expression. TRAIL/ IFNgamma, upregulated NFkappaB subunits and survivin while down-modulating 1kappaBalpha. NP epithelia had low RNA levels of NFkappaB subunits. These were increased in parenchymal areas of CP with severe fibrosis and most intensely in islets. The NFkappaB regulated proteins IkappaBalpha, survivin, and cIAP1 were found in corresponding sites, again, at highest levels in islets surrounded by fibrosis. In CP, islets not only evade immune attack by non-exposure of functional death receptors in presence of TRAIL-R4. They also neo-express NFkappaB subunits, survivin, and cIAP1. This apoptosis-inhibitory security program might be enforced by PSC-derived TRAIL.


Subject(s)
Genes, Intracisternal A-Particle/genetics , Islets of Langerhans/pathology , NF-kappa B/genetics , Pancreatitis/pathology , Chronic Disease , Epithelial Cells/cytology , Epithelial Cells/pathology , Gene Expression Regulation , Humans , Inhibitor of Apoptosis Proteins/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics
17.
Lab Invest ; 85(10): 1263-75, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16127431

ABSTRACT

In advanced chronic pancreatitis (CP), islets are preserved even in the midst of scarring. We recently showed in CP local production of interferon (IFN)gamma, transforming growth factor (TGF)beta and death receptor ligand TRAIL (tumor necrosis factor-related apoptosis-inducing ligand), along with functional death receptor neoexpression and apoptosis in exocrine but not in endocrine cells. However, islets are strongly induced for TRAIL-receptor (R)-4 lacking the functional death domain. TRAIL-R4 signaling in T cells induces NFkappaB, which activates antiapoptotic programs. Here, we demonstrate that in insulinoma cells CM, TGFbeta/IFNgamma/TRAIL in combination induced TRAIL-R4 surface expression. TRAIL/IFNgamma upregulated NFkappaB subunits and its target gene survivin while downmodulating IkappaB alpha mRNA. RelA transcriptional activity increased upon stimulation with IFNgamma and IFNgamma/TRAIL. In situ, normal pancreatic epithelia had low mRNA levels of NFkappaB subunits. These were higher in parenchymal areas of CP with severe fibrosis and highest in islets. NFkappaB-regulated proteins IkappaB alpha, survivin and another apoptosis inhibitor, cIAP1, were found in corresponding sites, again at highest levels in islets surrounded by fibrosis. In conclusion, islets in CP not only evade immune attack by nonexposure of functional death receptors in the presence of TRAIL-R4 but also additionally neoexpress NFkappaB and its target genes, survivin and cIAP1, to protect themselves from apoptosis.


Subject(s)
Inhibitor of Apoptosis Proteins/biosynthesis , Islets of Langerhans/pathology , Microtubule-Associated Proteins/biosynthesis , NF-kappa B/biosynthesis , Neoplasm Proteins/biosynthesis , Pancreatitis, Chronic/pathology , Apoptosis , Apoptosis Regulatory Proteins/biosynthesis , Cell Line, Tumor , Epithelium/metabolism , Fibrosis , Gene Expression Regulation , Humans , I-kappa B Kinase/metabolism , Insulinoma , Interferon-gamma/physiology , Islets of Langerhans/metabolism , Membrane Glycoproteins/biosynthesis , Pancreatitis, Chronic/metabolism , Protein Subunits/biosynthesis , Receptors, Tumor Necrosis Factor/biosynthesis , Survivin , TNF-Related Apoptosis-Inducing Ligand , Transcription Factor RelA/biosynthesis , Transforming Growth Factor beta/physiology , Tumor Necrosis Factor Decoy Receptors , Tumor Necrosis Factor-alpha/biosynthesis , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL