Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Front Oncol ; 14: 1396285, 2024.
Article in English | MEDLINE | ID: mdl-38884086

ABSTRACT

Background: KRAS mutation status is a well-established independent prognostic factor in advanced non-small cell lung cancer (NSCLC), yet its role in early-stage disease is unclear. Here, we investigate the prognostic value of combining survival data on KRAS mutation status and tumor size in stage I-II NSCLC. Methods: We studied the combined impact of KRAS mutational status and tumor size on overall survival (OS) in patients with stage I-II NSCLC. We performed a retrospective study including 310 diagnosed patients with early (stage I-II) NSCLCs. All molecularly assessed patients diagnosed with stage I-II NSCLC between 2016-2018 in the Västra Götaland Region of western Sweden were screened in this multi-center retrospective study. The primary study outcome was overall survival. Results: Out of 310 patients with stage I-II NSCLC, 37% harbored an activating mutation in the KRAS gene. Our study confirmed staging and tumor size as prognostic factors. However, KRAS mutational status was not found to impact OS and there was no difference in the risk of death when combining KRAS mutational status and primary tumor size. Conclusions: In our patient cohort, KRAS mutations in combination with primary tumor size did not impact prognosis in stage I-II NSCLC.

2.
Dig Dis Sci ; 63(8): 2013-2021, 2018 08.
Article in English | MEDLINE | ID: mdl-29948557

ABSTRACT

The incidence of esophageal adenocarcinoma (EAC) and its precursor lesion Barrett's esophagus (BE) has been increasing steadily in the western world in recent decades. Understanding the cellular origins of BE and the conditions responsible for their malignant transformation would greatly facilitate risk assessment and identification of patients at risk of progression, but this topic remains a source of debate. Here, we review recent findings that have provided support for the gastroesophageal junction (GEJ) as the main source of stem cells that give rise to BE and EAC. These include both gastric cardia cells and transitional basal cells. Furthermore, we discuss the role of chronic injury and inflammation in a tumor microenvironment as a major factor in promoting stem cell expansion and proliferation as well as transformation of the GEJ-derived stem cells and progression to EAC. We conclude that there exists a large amount of empirical support for the GEJ as the likely source of BE stem cells. While BE seems to resemble a successful adaptation to esophageal damage, carcinogenesis appears as a consequence of natural selection at the level of GEJ stem cells, and later glands, that expand into the esophagus wherein the local ecology creates the selective landscape for cancer progression.


Subject(s)
Adenocarcinoma , Barrett Esophagus , Esophageal Neoplasms , Esophagogastric Junction/pathology , Stem Cells , Adenocarcinoma/etiology , Adenocarcinoma/pathology , Barrett Esophagus/complications , Barrett Esophagus/pathology , Disease Progression , Esophageal Neoplasms/etiology , Esophageal Neoplasms/pathology , Humans , Metaplasia , Stem Cells/pathology , Stem Cells/physiology
3.
Oncotarget ; 7(35): 56183-56192, 2016 Aug 30.
Article in English | MEDLINE | ID: mdl-27487143

ABSTRACT

The transcription factor Zinc finger protein 148 (Zfp148, ZBP-89, BFCOL, BERF1, htß) interacts physically with the tumor suppressor p53, but the significance of this interaction is not known. We recently showed that knockout of Zfp148 in mice leads to ectopic activation of p53 in some tissues and cultured fibroblasts, suggesting that Zfp148 represses p53 activity. Here we hypothesize that targeting Zfp148 would unleash p53 activity and protect against cancer development, and test this idea in the APCMin/+ mouse model of intestinal adenomas. Loss of one copy of Zfp148 markedly reduced tumor numbers and tumor-associated intestinal bleedings, and improved survival. Furthermore, after activation of ß-catenin-the initiating event in colorectal cancer-Zfp148 deficiency activated p53 and induced apoptosis in intestinal explants of APCMin/+ mice. The anti-tumor effect of targeting Zfp148 depended on p53, as Zfp148 deficiency did not affect tumor numbers in APCMin/+ mice lacking one or both copies of Trp53. The results suggest that Zfp148 controls the fate of newly transformed intestinal tumor cells by repressing p53 and that targeting Zfp148 might be useful in the treatment of colorectal cancer.


Subject(s)
Adenoma/pathology , Colorectal Neoplasms/pathology , DNA-Binding Proteins/metabolism , Gastrointestinal Hemorrhage/pathology , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Adenoma/mortality , Animals , Apoptosis , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Colorectal Neoplasms/mortality , DNA-Binding Proteins/genetics , Fibroblasts , Gastrointestinal Hemorrhage/mortality , Humans , Mice , Mice, Knockout , Neoplasms, Experimental/mortality , Neoplasms, Experimental/pathology , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , beta Catenin/metabolism
4.
Cell Metab ; 24(1): 41-50, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27320064

ABSTRACT

The gut microbiota is considered a metabolic "organ" that not only facilitates harvesting of nutrients and energy from the ingested food but also produces numerous metabolites that signal through their cognate receptors to regulate host metabolism. One such class of metabolites, bile acids, is produced in the liver from cholesterol and metabolized in the intestine by the gut microbiota. These bioconversions modulate the signaling properties of bile acids via the nuclear farnesoid X receptor and the G protein-coupled membrane receptor 5, which regulate numerous metabolic pathways in the host. Conversely, bile acids can modulate gut microbial composition both directly and indirectly through activation of innate immune genes in the small intestine. Thus, host metabolism can be affected through microbial modifications of bile acids, which lead to altered signaling via bile acid receptors, but also by altered microbiota composition.


Subject(s)
Bile Acids and Salts/metabolism , Intestinal Mucosa/metabolism , Intestines/microbiology , Microbiota , Animals , Bariatric Surgery , Diet , Humans , Receptors, Cytoplasmic and Nuclear/metabolism
5.
Nat Commun ; 6: 7629, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26134028

ABSTRACT

Bile acids are signalling molecules, which activate the transmembrane receptor TGR5 and the nuclear receptor FXR. BA sequestrants (BAS) complex bile acids in the intestinal lumen and decrease intestinal FXR activity. The BAS-BA complex also induces glucagon-like peptide-1 (GLP-1) production by L cells which potentiates ß-cell glucose-induced insulin secretion. Whether FXR is expressed in L cells and controls GLP-1 production is unknown. Here, we show that FXR activation in L cells decreases proglucagon expression by interfering with the glucose-responsive factor Carbohydrate-Responsive Element Binding Protein (ChREBP) and GLP-1 secretion by inhibiting glycolysis. In vivo, FXR deficiency increases GLP-1 gene expression and secretion in response to glucose hence improving glucose metabolism. Moreover, treatment of ob/ob mice with the BAS colesevelam increases intestinal proglucagon gene expression and improves glycaemia in a FXR-dependent manner. These findings identify the FXR/GLP-1 pathway as a new mechanism of BA control of glucose metabolism and a pharmacological target for type 2 diabetes.


Subject(s)
Enteroendocrine Cells/metabolism , Glucagon-Like Peptide 1/genetics , Intestinal Mucosa/metabolism , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Animals , Anticholesteremic Agents/pharmacology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Bile Acids and Salts/metabolism , Blood Glucose/metabolism , Colesevelam Hydrochloride/pharmacology , Colon/cytology , Colon/metabolism , Diet, High-Fat , Glucagon-Like Peptide 1/metabolism , Glycolysis , Humans , Ileum/cytology , Ileum/metabolism , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Intestines/cytology , Jejunum/cytology , Jejunum/metabolism , Mice , Mice, Knockout , Mice, Obese , Nuclear Proteins/metabolism , Obesity/genetics , Obesity/metabolism , Proglucagon/drug effects , Proglucagon/genetics , Proglucagon/metabolism , Receptors, G-Protein-Coupled/genetics , Sequestering Agents/pharmacology , Signal Transduction , Transcription Factors/metabolism
7.
Cell Metab ; 17(2): 225-35, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23395169

ABSTRACT

Bile acids are synthesized from cholesterol in the liver and further metabolized by the gut microbiota into secondary bile acids. Bile acid synthesis is under negative feedback control through activation of the nuclear receptor farnesoid X receptor (FXR) in the ileum and liver. Here we profiled the bile acid composition throughout the enterohepatic system in germ-free (GF) and conventionally raised (CONV-R) mice. We confirmed a dramatic reduction in muricholic acid, but not cholic acid, levels in CONV-R mice. Rederivation of Fxr-deficient mice as GF demonstrated that the gut microbiota regulated expression of fibroblast growth factor 15 in the ileum and cholesterol 7α-hydroxylase (CYP7A1) in the liver by FXR-dependent mechanisms. Importantly, we identified tauro-conjugated beta- and alpha-muricholic acids as FXR antagonists. These studies suggest that the gut microbiota not only regulates secondary bile acid metabolism but also inhibits bile acid synthesis in the liver by alleviating FXR inhibition in the ileum.


Subject(s)
Bile Acids and Salts/metabolism , Gastrointestinal Tract/microbiology , Metagenome , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Taurocholic Acid/analogs & derivatives , Absorption , Animals , Anti-Bacterial Agents/pharmacology , Cholesterol 7-alpha-Hydroxylase/genetics , Cholesterol 7-alpha-Hydroxylase/metabolism , Feedback, Physiological/drug effects , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Gastrointestinal Tract/drug effects , Gene Expression Profiling , Gene Expression Regulation/drug effects , Ileum/drug effects , Ileum/metabolism , Liver/drug effects , Liver/metabolism , Metagenome/drug effects , Metagenome/genetics , Mice , Models, Biological , Organ Specificity/drug effects , Organ Specificity/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Taurocholic Acid/metabolism , Taurocholic Acid/pharmacology
8.
Rhinology ; 44(3): 211-5, 2006 Sep.
Article in English | MEDLINE | ID: mdl-17020070

ABSTRACT

STATEMENT OF PROBLEM: Inverted papilloma (IP) is a proliferative lesion of the epithelium lining the sinonasal tract, characterized by marked propensity for recurrence and association with carcinoma. To determine a putative role of matrix metalloproteinase-2 (MMP-2) and MMP-9 in the establishment of IP, their expression was studied in IP. METHODS: Archived surgical specimens from 15 IPs were studied using immunohistochemistry and compared to 12 nasal polyps (NP), a model of chronic respiratory mucosal inflammation, and to 6 control nasal mucosa (CM) samples obtained from snorers during turbinectomy. Within IP, MMP-2 and -9 expression was compared between tumoral areas with hyperplastic epithelium and non tumoral areas with nonhyperplastic epithelium. RESULTS: In IP, MMP-2 and MMP-9 epithelial expression was not different compared to CM and NP. MMP-9 expression in submucosal inflammatory cells was not different between IP and CM or NP. However, within IP, a significantly increased number of MMP-9 positive inflammatory cells in the lamina propria adjacent to the hyperplastic epithelium was observed compared to the lamina propria adjacent to nonhyperplastic epithelium. CONCLUSION: Our findings suggest that MMP 9 expressing inflammatory cells may be involved in the pathophysiology of IP.


Subject(s)
Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Nose Neoplasms/metabolism , Papilloma, Inverted/metabolism , Case-Control Studies , Female , Humans , Male , Nasal Mucosa/metabolism , Nasal Mucosa/pathology , Nasal Polyps/metabolism , Nasal Polyps/pathology , Nose Neoplasms/pathology , Papilloma, Inverted/pathology
9.
FEBS Lett ; 500(1-2): 41-4, 2001 Jun 29.
Article in English | MEDLINE | ID: mdl-11434923

ABSTRACT

The tyrosine phosphatase r-PTPeta is able to suppress the malignant phenotype of rat thyroid tumorigenic cell lines. To identify r-PTPeta interacting proteins, a yeast two-hybrid screening was performed and an insert corresponding to the full-length syntenin cDNA was isolated. It encodes a protein containing two PDZ domains that mediates the binding of syntenin to proteins such as syndecan, proTGF-alpha, beta-ephrins and neurofascin. We show that r-PTPeta is able to interact with syntenin also in mammalian cells, and although syntenin is a tyrosine-phosphorylated protein it is not a substrate of r-PTPeta. The integrity of both PDZ domains of syntenin and the carboxy-terminal region of r-PTPeta are required for the interaction between syntenin and r-PTPeta.


Subject(s)
Carrier Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Protein Tyrosine Phosphatases/metabolism , Carrier Proteins/chemistry , Cells, Cultured , Humans , Precipitin Tests , Protein Structure, Tertiary , Receptor-Like Protein Tyrosine Phosphatases, Class 3 , Syntenins , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...