Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
1.
Oncogene ; 36(34): 4843-4858, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28414310

ABSTRACT

High CD44 expression is associated with enhanced malignant potential in esophageal squamous cell carcinoma (ESCC), among the deadliest of all human carcinomas. Although alterations in autophagy and CD44 expression are associated with poor patient outcomes in various cancer types, the relationship between autophagy and cells with high CD44 expression remains incompletely understood. In transformed oesophageal keratinocytes, CD44Low-CD24High (CD44L) cells give rise to CD44High-CD24-/Low (CD44H) cells via epithelial-mesenchymal transition (EMT) in response to transforming growth factor (TGF)-ß. We couple patient samples and xenotransplantation studies with this tractable in vitro system of CD44L to CD44H cell conversion to investigate the functional role of autophagy in generation of cells with high CD44 expression. We report that high expression of the autophagy marker cleaved LC3 expression correlates with poor clinical outcome in ESCC. In ESCC xenograft tumours, pharmacological autophagy inhibition with chloroquine derivatives depletes cells with high CD44 expression while promoting oxidative stress. Autophagic flux impairment during EMT-mediated CD44L to CD44H cell conversion in vitro induces mitochondrial dysfunction, oxidative stress and cell death. During CD44H cell generation, transformed keratinocytes display evidence of mitophagy, including mitochondrial fragmentation, decreased mitochondrial content and mitochondrial translocation of Parkin, essential in mitophagy. RNA interference-mediated Parkin depletion attenuates CD44H cell generation. These data suggest that autophagy facilitates EMT-mediated CD44H generation via modulation of redox homeostasis and Parkin-dependent mitochondrial clearance. This is the first report to implicate mitophagy in regulation of tumour cells with high CD44 expression, representing a potential novel therapeutic avenue in cancers where EMT and CD44H cells have been implicated, including ESCC.


Subject(s)
Autophagy/physiology , Hyaluronan Receptors/metabolism , Mitochondria/physiology , Oxidative Stress/physiology , Ubiquitin-Protein Ligases/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition/physiology , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Humans , Keratinocytes/metabolism , Keratinocytes/physiology , Mitochondria/metabolism , Oxidation-Reduction , RNA Interference/physiology , Transforming Growth Factor beta/metabolism
2.
Oncogene ; 35(41): 5337-5349, 2016 10 13.
Article in English | MEDLINE | ID: mdl-26923327

ABSTRACT

Esophageal cancer is a deadly disease, ranking sixth among all cancers in mortality. Despite incremental advances in diagnostics and therapeutics, esophageal cancer still carries a poor prognosis, and thus, there remains a need to elucidate the molecular mechanisms underlying this disease. There is accumulating evidence that a comprehensive understanding of the molecular composition of esophageal cancer requires attention to not only tumor cells but also the tumor microenvironment (TME), which contains diverse cell populations, signaling factors and structural molecules that interact with tumor cells and support all stages of tumorigenesis. In esophageal cancer, environmental exposures can trigger chronic inflammation, which leads to constitutive activation of pro-inflammatory signaling pathways that promote survival and proliferation. Antitumor immunity is attenuated by cell populations such as myeloid-derived suppressor cells and regulatory T cells, as well as immune checkpoints like programmed death-1. Other immune cells such as tumor-associated macrophages can have other pro-tumorigenic functions, including the induction of angiogenesis and tumor cell invasion. Cancer-associated fibroblasts secrete growth factors and alter the extracellular matrix to create a tumor niche and enhance tumor cell migration and metastasis. Further study of how these TME components relate to the different stages of tumor progression in each esophageal cancer subtype will lead to development of novel and specific TME-targeting therapeutic strategies, which offer considerable potential especially in the setting of combination therapy.


Subject(s)
Esophageal Neoplasms/genetics , Inflammation/genetics , Neovascularization, Pathologic/genetics , Tumor Microenvironment/genetics , Apoptosis/genetics , Apoptosis/immunology , Cell Cycle Checkpoints/genetics , Cell Cycle Checkpoints/immunology , Esophageal Neoplasms/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Neoplasm Invasiveness/immunology , Neovascularization, Pathologic/pathology , T-Lymphocytes, Regulatory/immunology
3.
Oncogene ; 35(29): 3880-6, 2016 07 21.
Article in English | MEDLINE | ID: mdl-26592448

ABSTRACT

Epidermal growth factor receptor (EGFR) signaling has a critical role in oncogenic Kras-driven pancreatic carcinogenesis. However, the downstream targets of this signaling network are largely unknown. We developed a novel model system utilizing murine primary pancreatic ductal epithelial cells (PDECs), genetically engineered to allow time-specific expression of oncogenic Kras(G12D) from the endogenous promoter. We show that primary PDECs are susceptible to Kras(G12D)-driven transformation and form pancreatic ductal adenocarcinomas in vivo after Cdkn2a inactivation. In addition, we demonstrate that activation of Kras(G12D) induces an EGFR signaling loop to drive proliferation. Interestingly, pharmacological inhibition of EGFR fails to decrease Kras(G12D)-activated ERK or PI3K signaling. Instead our data provide novel evidence that EGFR signaling is needed to activate the oncogenic and pro-proliferative transcription factor c-MYC. EGFR and c-MYC have been shown to be essential for pancreatic carcinogenesis. Importantly, our data link both pathways and thereby explain the crucial role of EGFR for Kras(G12D)-driven carcinogenesis in the pancreas.


Subject(s)
Epithelial Cells/metabolism , ErbB Receptors/metabolism , Pancreatic Ducts/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Epithelial Cells/transplantation , ErbB Receptors/genetics , Gene Expression Profiling/methods , Immunoblotting , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , Mutation , Pancreatic Ducts/cytology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics
4.
Oncogene ; 34(18): 2347-59, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-24931169

ABSTRACT

Notch activity regulates tumor biology in a context-dependent and complex manner. Notch may act as an oncogene or a tumor-suppressor gene even within the same tumor type. Recently, Notch signaling has been implicated in cellular senescence. Yet, it remains unclear as to how cellular senescence checkpoint functions may interact with Notch-mediated oncogenic and tumor-suppressor activities. Herein, we used genetically engineered human esophageal keratinocytes and esophageal squamous cell carcinoma cells to delineate the functional consequences of Notch activation and inhibition along with pharmacological intervention and RNA interference experiments. When expressed in a tetracycline-inducible manner, the ectopically expressed activated form of Notch1 (ICN1) displayed oncogene-like characteristics inducing cellular senescence corroborated by the induction of G0/G1 cell-cycle arrest, Rb dephosphorylation, flat and enlarged cell morphology and senescence-associated ß-galactosidase activity. Notch-induced senescence involves canonical CSL/RBPJ-dependent transcriptional activity and the p16(INK4A)-Rb pathway. Loss of p16(INK4A) or the presence of human papilloma virus (HPV) E6/E7 oncogene products not only prevented ICN1 from inducing senescence but permitted ICN1 to facilitate anchorage-independent colony formation and xenograft tumor growth with increased cell proliferation and reduced squamous-cell differentiation. Moreover, Notch1 appears to mediate replicative senescence as well as transforming growth factor-ß-induced cellular senescence in non-transformed cells and that HPV E6/E7 targets Notch1 for inactivation to prevent senescence, revealing a tumor-suppressor attribute of endogenous Notch1. In aggregate, cellular senescence checkpoint functions may influence dichotomous Notch activities in the neoplastic context.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Receptor, Notch1/metabolism , Retinoblastoma Protein/metabolism , Signal Transduction , Cell Cycle Checkpoints , Cell Transformation, Viral , Cells, Cultured , Cellular Senescence , Esophageal Squamous Cell Carcinoma , Esophagus/cytology , Esophagus/metabolism , Humans , Keratinocytes/metabolism , Phosphorylation , Transforming Growth Factor beta/metabolism , Viral Proteins/metabolism
5.
Oncogenesis ; 2: e59, 2013 Aug 05.
Article in English | MEDLINE | ID: mdl-23917221

ABSTRACT

Periostin (POSTN), a matricellular protein, has been reported to be important in supporting tumor cell dissemination. However, the molecular mechanisms underlying POSTN function within the tumor microenvironment are poorly understood. In this study, we observe that the inducible knockdown of POSTN decreases esophageal squamous cell carcinoma (ESCC) tumor growth in vivo and demonstrate that POSTN cooperates with a conformational missense p53 mutation to enhance invasion. Pathway analyses reveal that invasive esophageal cells expressing POSTN and p53(R175H) mutation display activation of signal transducer and activator of transcription 1 (STAT1) target genes, suggesting that the induction of STAT1 and STAT1-related genes could foster a permissive microenvironment that facilitates invasion of esophageal epithelial cells into the extracellular matrix. Genetic knockdown of STAT1 in transformed esophageal epithelial cells underscores the importance of STAT1 in promoting invasion. Furthermore, we find that STAT1 is activated in ESCC xenograft tumors, but this activation is attenuated with inducible knockdown of POSTN in ESCC tumors. Overall, these results highlight the novel molecular mechanisms supporting the capacity of POSTN in mediating tumor invasion during ESCC development and have implications of therapeutic strategies targeting the tumor microenvironment.

6.
Br J Cancer ; 108(4): 755-61, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23322204

ABSTRACT

Matricellular proteins have been classified as a family of non-structural matrix proteins capable of modulating a variety of biological processes within the extracellular matrix (ECM). These proteins are expressed dynamically and their cellular functions are highly dependent upon cues from the local environment. Recent studies have shown an increasing appreciation of the key roles these ECM proteins play within the tumour microenvironment. Induced by either tumour cells or tumour stromal components, matricellular proteins initiate downstream signalling events that lead to proliferation, invasion, matrix remodelling and dissemination to pre-metastatic niches in other organs. In this review, we summarise and discuss the current knowledge of the diverse roles these proteins play within the microenvironment that influences tumour progression and potential for future therapies targeting the tumour microenvironment.


Subject(s)
Extracellular Matrix Proteins/physiology , Neoplasms/pathology , Tumor Microenvironment , Extracellular Matrix/metabolism , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/metabolism , Signal Transduction
7.
Cancer Gene Ther ; 18(12): 871-83, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21869822

ABSTRACT

Esophageal carcinoma is the most rapidly increasing tumor in the United States and has a dismal 15% 5-year survival. Immunotherapy has been proposed to improve patient outcomes; however, no immunocompetent esophageal carcinoma model exists to date to test this approach. We developed two mouse models of esophageal cancer by inoculating immunocompetent mice with syngeneic esophageal cell lines transformed by cyclin-D1 or mutant HRAS(G12V) and loss of p53. Similar to humans, surgery and adjuvant chemotherapy (cisplatin and 5-fluorouracil) demonstrated limited efficacy. Gene-mediated cyototoxic immunotherapy (adenoviral vector carrying the herpes simplex virus thymidine kinase gene in combination with the prodrug ganciclovir; AdV-tk/GCV) demonstrated high levels of in vitro transduction and efficacy. Using in vivo syngeneic esophageal carcinoma models, combining surgery, chemotherapy and AdV-tk/GCV improved survival (P=0.007) and decreased disease recurrence (P<0.001). Mechanistic studies suggested that AdV-tk/GCV mediated a direct cytotoxic effect and an increased intra-tumoral trafficking of CD8 T cells (8.15% vs 14.89%, P=0.02). These data provide the first preclinical evidence that augmenting standard of care with immunotherapy may improve outcomes in the management of esophageal carcinoma.


Subject(s)
Carcinoma/therapy , Esophageal Neoplasms/therapy , Genetic Therapy/methods , Immunotherapy/methods , Neoadjuvant Therapy/methods , Neoplasms, Experimental/therapy , Animals , Cell Line, Tumor , Cell Survival , Cyclin D1/genetics , Cytotoxicity, Immunologic , Female , Genes, ras/genetics , Genetic Vectors , Humans , Mice , Mice, Inbred C57BL , Neoplasm Recurrence, Local/prevention & control , Simplexvirus/genetics , Thymidine Kinase/genetics , Treatment Outcome , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
8.
Oncogene ; 30(40): 4185-93, 2011 Oct 06.
Article in English | MEDLINE | ID: mdl-21625210

ABSTRACT

Lin28b is an RNA-binding protein that inhibits biogenesis of let-7 microRNAs. LIN28B is overexpressed in diverse cancers, yet a specific role in the molecular pathogenesis of colon cancer has to be elucidated. We have determined that human colon tumors exhibit decreased levels of mature let-7 isoforms and increased expression of LIN28B. To determine LIN28B's mechanistic role in colon cancer, we expressed LIN28B in immortalized colonic epithelial cells and human colon cancer cell lines. We found that LIN28B promotes cell migration, invasion and transforms immortalized colonic epithelial cells. In addition, constitutive LIN28B expression increases expression of intestinal stem cell markers LGR5 and PROM1 in the presence of let-7 restoration. This may occur as a result of Lin28b protein binding LGR5 and PROM1 mRNA, suggesting that a subset of LIN28B functions is independent of its ability to repress let-7. Our findings establish a new role for LIN28B in human colon cancer pathogenesis, and suggest LIN28B post-transcriptionally regulates LGR5 and PROM1 through a let-7-independent mechanism.


Subject(s)
Cell Movement/physiology , Colonic Neoplasms/pathology , DNA-Binding Proteins/physiology , MicroRNAs/physiology , Neoplasm Invasiveness , Cell Line, Tumor , Humans , RNA-Binding Proteins
9.
Oncogene ; 29(29): 4170-82, 2010 Jul 22.
Article in English | MEDLINE | ID: mdl-20498638

ABSTRACT

Variable drug responses among malignant cells within individual tumors may represent a barrier to their eradication using chemotherapy. Carcinoma cells expressing mesenchymal markers resist conventional and epidermal growth factor receptor (EGFR)-targeted chemotherapy. In this study, we evaluated whether mesenchymal-like sub-populations within human squamous cell carcinomas (SCCs) with predominantly epithelial features contribute to overall therapy resistance. We identified a mesenchymal-like subset expressing low E-cadherin (Ecad-lo) and high vimentin within the upper aerodigestive tract SCCs. This subset was both isolated from the cell lines and was identified in xenografts and primary clinical specimens. The Ecad-lo subset contained more low-turnover cells, correlating with resistance to the conventional chemotherapeutic paclitaxel in vitro. Epidermal growth factor induced less stimulation of the mitogen-activated protein kinase and phosphatidylinositol-3-kinase pathways in Ecad-lo cells, which was likely due to lower EGFR expression in this subset and correlated with in vivo resistance to the EGFR-targeted antibody, cetuximab. The Ecad-lo and high E-cadherin subsets were dynamic in phenotype, showing the capacity to repopulate each other from single-cell clones. Taken together, these results provide evidence for a low-turnover, mesenchymal-like sub-population in SCCs with diminished EGFR pathway function and intrinsic resistance to conventional and EGFR-targeted chemotherapies.


Subject(s)
Carcinoma, Squamous Cell/pathology , Mesoderm/pathology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , Cetuximab , Drug Resistance, Neoplasm , Humans , MAP Kinase Signaling System , Paclitaxel/pharmacology , Phenotype , Phosphatidylinositol 3-Kinases/physiology
10.
Oncogene ; 26(16): 2353-64, 2007 Apr 05.
Article in English | MEDLINE | ID: mdl-17043653

ABSTRACT

Epidermal growth factor receptor (EGFR) overexpression and activation is critical in the initiation and progression of cancers, especially those of epithelial origin. EGFR activation is associated with the induction of divergent signal transduction pathways and a gamut of cellular processes; however, the cell-type and tissue-type specificity conferred by certain pathways remains to be elucidated. In the context of the esophageal epithelium, a prototype stratified squamous epithelium, EGFR overexpression is relevant in the earliest events of carcinogenesis as modeled in a three-dimensional organotypic culture system. We demonstrate that the phosphatidylinositol 3-kinase (PI3K)/AKT signaling pathway, and not the MEK/MAPK (mitogen-activated protein kinase) pathway, is preferentially activated in EGFR-mediated esophageal epithelial hyperplasia, a premalignant lesion. The hyperplasia was abolished with direct inhibition of PI3K and of AKT but not with inhibition of the MAPK pathway. With the introduction of an inducible AKT vector in both primary and immortalized esophageal epithelial cells, we find that AKT overexpression and activation is permissive for complete epithelial formation in organotypic culture, but imposes a growth constraint in cells grown in monolayer. In organotypic culture, AKT mediates changes related to cell shape and size with an expansion of the differentiated compartment.


Subject(s)
Esophagus/cytology , Oncogene Protein v-akt/physiology , Cell Differentiation , Cell Division , Cells, Cultured , Cellular Senescence , Epithelial Cells/cytology , ErbB Receptors/physiology , Genetic Vectors , Humans , Immunohistochemistry , Organ Culture Techniques , Retroviridae
11.
J Clin Oncol ; 22(23): 4730-6, 2004 Dec 01.
Article in English | MEDLINE | ID: mdl-15570073

ABSTRACT

PURPOSE: In March 2001, the National Colorectal Cancer Research Alliance (NCCRA) and OncoLink (http://www.oncolink.org) established a database to facilitate patient enrollment onto clinical trials. This study describes the population registering with the database and identifies discrepancies between individuals registering through the Internet and those registering through a telephone call center. METHODS: Participants registered with the NCCRA/OncoLink database through the Internet or a telephone call center. All participants entering the database completed a questionnaire regarding basic demographics, colon cancer risk factors, and indicated how they became aware of the database. Comparisons were made between individuals registering through the Internet and those registering through the telephone call center. RESULTS: A total of 2,162 participants registered during the first 16 months of the database. Most patients registered through the Internet rather than the telephone call center (88% v 12%; P < .001). More females than males registered (73% v 27%; P < .001). The majority (89%) were white. Participants registering through the Internet were younger than those registering through the call center (mean, 48.8 v 55.0 years; P < .001). There was no difference between the two groups with regard to sex or ethnicity. CONCLUSION: The Internet has the potential to increase the likelihood that interested individuals find appropriate clinical trials. Some of the discrepancies that are known to exist for access to the Internet were also seen for those registering with the database through the Internet. Despite these differences, the potential to increase clinical trial enrollment with this type of Internet-based database is high.


Subject(s)
Clinical Trials as Topic , Colorectal Neoplasms/therapy , Internet , Registries , Telephone , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Colorectal Neoplasms/diagnosis , Databases, Factual , Female , Humans , Male , Middle Aged , Patient Selection , Probability , Research , Sensitivity and Specificity , Sex Factors , Societies, Medical , United States
13.
Crit Rev Oral Biol Med ; 13(1): 51-61, 2002.
Article in English | MEDLINE | ID: mdl-12097237

ABSTRACT

The dysregulation of the molecular events governing cell cycle control is emerging as a central theme of oral carcinogenesis. Regulatory pathways responding to extracellular signaling or intracellular stress and DNA damage converge on the cell cycle apparatus. Abrogation of mitogenic and anti-mitogenic response regulatory proteins, such as the retinoblastoma tumor suppressor protein (pRB), cyclin D1, cyclin-dependent kinase (CDK) 6, and CDK inhibitors (p21(WAF1/CIP1), p27(KIP1), and p16(INK4a)), occur frequently in human oral cancers. Cellular responses to metabolic stress or genomic damage through p53 and related pathways that block cell cycle progression are also altered during oral carcinogenesis. In addition, new pathways and cell cycle regulatory proteins, such as p12(DOC-1), are being discovered. The multistep process of oral carcinogenesis likely involves functional alteration of cell cycle regulatory members combined with escape from cellular senescence and apoptotic signaling pathways. Detailing the molecular alterations and understanding the functional consequences of the dysregulation of the cell cycle apparatus in the malignant oral keratinocyte will uncover novel diagnostic and therapeutic approaches.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Cycle Proteins/metabolism , Cell Cycle/physiology , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Alphapapillomavirus/physiology , Apoptosis , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/virology , Cell Proliferation , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Humans , Mouth Neoplasms/genetics , Mouth Neoplasms/virology , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism
14.
Gastroenterology ; 121(5): 1035, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11706826

Subject(s)
Research/economics , Humans
15.
J Clin Invest ; 108(5): 725-32, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11544278

ABSTRACT

The immortalization of human cells is a critical step in multistep carcinogenesis. Oral-esophageal carcinomas, a model system to investigate molecular mechanisms underlying squamous carcinogenesis, frequently involve cyclin D1 overexpression and inactivation of the p53 tumor suppressor. Therefore, our goal was to establish the functional role of cyclin D1 overexpression and p53 inactivation in the immortalization of primary human oral squamous epithelial cells (keratinocytes) as an important step toward malignant transformation. Cyclin D1 overexpression alone was found to induce extension of the replicative life span of normal oral keratinocytes, whereas the combination of cyclin D1 overexpression and p53 inactivation led to their immortalization. This study also demonstrates that immortalization of oral keratinocytes can be independent of telomerase activation, involving an alternative pathway of telomere maintenance (ALT).


Subject(s)
Carcinoma, Squamous Cell/etiology , Cell Transformation, Neoplastic , Cyclin D1/genetics , Genes, p53 , Keratinocytes/physiology , Mouth Neoplasms/etiology , Carcinoma, Squamous Cell/enzymology , Cell Cycle , Cell Division , Cell Line , Cyclin D1/metabolism , Humans , Keratinocytes/cytology , Keratinocytes/enzymology , Mouth/cytology , Mouth Neoplasms/enzymology , Mutation , Telomerase/physiology , Transduction, Genetic
16.
Cancer Res ; 61(17): 6467-73, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11522642

ABSTRACT

The p53 gene family, comprising p53, p63, and p73, has overlapping and distinctive functional roles. These members share structural similarities allowing for dynamic interplay in the activation of genes that are important in development and key cellular functions, such as the induction of apoptosis. Whereas p53 is a classical tumor suppressor gene, p63 and p73 do not share this feature in cancer formation and progression. The compensation in the expression level of these members in a background that is deficient for one of them has not been examined previously. Given the importance of p63 in the development and differentiation of oral-esophageal stratified squamous epithelia and the absence of oral-esophageal tumors in p53-null mice, we postulated and describe herein that p63 expression is associated with the loss of p53 in a p53-deficient background. Both full-length and amino-truncated forms of p63 are expressed and increased in oral-esophageal epithelia of p53-null mice when compared with wild-type mice, and the induction of p21 may potentially be preserved through the increase of p63.


Subject(s)
Esophagus/metabolism , Membrane Proteins , Mouth Mucosa/metabolism , Phosphoproteins/biosynthesis , Trans-Activators/biosynthesis , Tumor Suppressor Protein p53/deficiency , Animals , Cell Differentiation/physiology , Cells, Cultured , DNA-Binding Proteins , Epithelium/metabolism , Epithelium/physiology , Esophagus/cytology , Esophagus/physiology , Genes, Tumor Suppressor , Genes, p53/physiology , Humans , Immunohistochemistry , Mice , Mouth Mucosa/cytology , Mouth Mucosa/physiology , Phosphoproteins/genetics , Trans-Activators/genetics , Transcription Factors , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins
17.
Gastroenterology ; 120(6): 1319, 2001 May.
Article in English | MEDLINE | ID: mdl-11339232
18.
Gastroenterology ; 120(7): 1720-8, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11375953

ABSTRACT

BACKGROUND & AIMS: Keratins are intermediate filaments that are critical in cytoskeletal organization. Their roles in cellular processes are underscored by inherited human diseases in which germline mutations of keratins are found, as well as by transgenic and knockout mouse models that recapitulate those diseases. Keratin 19 (K19) has unique structural properties and developmental and spatial expression patterns. This suggests that K19 expression may correlate with important cell fate decisions in gastrointestinal tract epithelia. METHODS: We used mouse K19 5' untranslated region and promoter sequences and fused it to the lacZ reporter gene in a transgene construct. Characterization was by beta-galactosidase expression and X-gal histochemistry in gastrointestinal epithelia. Because endogenous K19 protein is transcriptionally regulated by the Kruppel-like transcription factor 4 (KLF4), we determined the spatial expression patterns of KLF4 and K19 in relationship to the lacZ reporter gene product. RESULTS: K19-lacZ transgenic mice were found to have reporter gene expression in an epithelial-specific pattern. Expression was restricted to ductal epithelial cells in the pancreas, surface colonocytes, small intestinal villi, and gastric isthmus cells. Transgene expression correlated with K19 and KLF4 protein expression in the pancreas and stomach and was overlapping in the small and large intestine. CONCLUSIONS: The K19 promoter may be a useful tool to study epithelial cell biology and subsequent transdifferentiation programs, particularly the pancreas and stomach.


Subject(s)
Gene Targeting , Keratins/genetics , Promoter Regions, Genetic , Animals , Colon/chemistry , Gastric Mucosa/chemistry , Immunohistochemistry , Keratins/analysis , Kruppel-Like Factor 4 , Mice , Mice, Transgenic , Pancreatic Ducts/chemistry
19.
Gastroenterology ; 120(6): 1321, 2001 May.
Article in English | MEDLINE | ID: mdl-11313300
SELECTION OF CITATIONS
SEARCH DETAIL
...