Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Head Neck ; 37(4): 530-6, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24532155

ABSTRACT

BACKGROUND: Gastroesophageal reflux (GER) contributes to periprosthetic leakage after prosthetic voice rehabilitation. However, underlying mechanisms are unclear, and markers predicting anti-reflux therapy response are missing. METHODS: We assessed epithelial-mesenchymal transition in 148 consecutive biopsies from 44 patients with/without fistula enlargement under dual-probe pH monitoring before and after proton-pump inhibitor (PPI) therapy applying immunohistochemistry. Results were correlated with reflux intensity and clinical and histologic findings. RESULTS: Epithelial-mesenchymal transition correlated with GER in all samples, and patients with fistula enlargement showed higher epithelial-mesenchymal transition scores. Contrary to patients without enlargement, epithelial-mesenchymal transition scores did not regress during therapy in this group. Furthermore, pretherapeutic epithelial-mesenchymal transition scores were lower in therapy responders than in nonresponders without reaching significance (p = .07). CONCLUSION: We demonstrate that epithelial-mesenchymal transition correlates with severity of GER and presence of periprosthetic fistula enlargement in patients who underwent prosthetic voice rehabilitation, but epithelial-mesenchymal transition seems to be reversible upon PPI treatment in early stages only.


Subject(s)
Epithelial-Mesenchymal Transition , Gastroesophageal Reflux/complications , Adult , Aged , Aged, 80 and over , Esophageal pH Monitoring , Female , Gastroesophageal Reflux/drug therapy , Humans , Immunohistochemistry , Larynx, Artificial , Male , Middle Aged , Proton Pump Inhibitors/therapeutic use , Tracheoesophageal Fistula/complications
2.
Mol Cancer ; 13: 145, 2014 Jun 09.
Article in English | MEDLINE | ID: mdl-24913355

ABSTRACT

BACKGROUND: The Abelson tyrosine kinase (c-Abl) inhibitor STI571 (Glivec®) has been shown to effectively inhibit colorectal cancer cell migration and invasion. The c-Abl substrate abelson interactor 1 (Abi1) is a key regulator of actin reorganization and upregulated in colorectal carcinoma. The specific role of Abi1 in relation to extracellular matrix degradation and effects of targeting Abi1 phosphorylation have not yet been examined. Here, we investigated the role of Abi1 in relation to invasive properties in colorectal cancer. METHODS AND RESULTS: In 56 primary human colorectal carcinoma samples, we found overexpression of Abi1 in 39% at the invasive edge of the tumour, associated with an infiltrative phenotype and high-grade tumour cell budding (p = 0.001). To explore the role of Abi1 in vitro, we employed the Abi1 expressing and KRAS-mutated CHD1 model and performed matrix degradation assays that showed Abi1 localization at specific sites of matrix degradation. Moreover, quantification of matrix dissolution demonstrated suppression after RNAi knockdown of Abi1 by 95% (p = 0.001). Importantly, treatment with STI571 did abolish Abi1 Y435-phosphorylation, suppressed the matrix dissolution, decreased fibronectin attachment, and suppressed cell invasion through reconstituted extracellular matrix. CONCLUSION: Our data indicate that phosphorylated Abi1 contributes to the invasive properties of colorectal cancer.


Subject(s)
Actins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Adhesion/genetics , Colorectal Neoplasms/genetics , Cytoskeletal Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Aged , Aged, 80 and over , Benzamides/administration & dosage , Cell Movement/genetics , Colorectal Neoplasms/pathology , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Extracellular Matrix/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Imatinib Mesylate , Male , Middle Aged , Phosphorylation/genetics , Piperazines/administration & dosage , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Pyrimidines/administration & dosage , ras Proteins/genetics
3.
Biochem J ; 450(2): 333-43, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23167255

ABSTRACT

APP (amyloid precursor protein) and LRP1 (low-density lipoprotein receptor-related protein 1) have been implicated in the pathogenesis of AD (Alzheimer's disease). They are functionally linked by Fe65, a PTB (phosphotyrosine-binding)-domain-containing adaptor protein that binds to intracellular NPxY-motifs of APP and LRP1, thereby influencing expression levels, cellular trafficking and processing. Additionally, Fe65 has been reported to mediate nuclear signalling in combination with intracellular domains of APP and LRP1. We have previously identified another adaptor protein, GULP1 (engulfment adaptor PTB-domain-containing 1). In the present study we characterize and compare nuclear trafficking and transactivation of GULP1 and Fe65 together with APP and LRP1 and report differential nuclear trafficking of adaptors when APP or LRP1 are co-expressed. The observed effects were additionally supported by a reporter-plasmid-based transactivation assay. The results from the present study indicate that Fe65 might have signalling properties together with APP and LRP1, whereas GULP1 only mediates LRP1 transactivation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Adaptor Proteins, Signal Transducing/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Binding Sites , Humans , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Signal Transduction , Transcriptional Activation , Transfection
4.
PLoS One ; 7(7): e40671, 2012.
Article in English | MEDLINE | ID: mdl-22808230

ABSTRACT

BACKGROUND: Abelson interactor 1 (Abi1) is an important regulator of actin dynamics during cytoskeletal reorganization. In this study, our aim was to investigate the expression of Abi1 in colonic mucosa with and without inflammation, colonic polyps, colorectal carcinomas (CRC) and metastases as well as in CRC cell lines with respect to BRAF/KRAS mutation status and to find out whether introduction of KRAS mutation or stimulation with TNFalpha enhances Abi1 protein expression in CRC cells. METHODOLOGY/PRINCIPAL FINDINGS: We immunohistochemically analyzed Abi1 protein expression in 126 tissue specimens from 95 patients and in 5 colorectal carcinoma cell lines with different mutation status by western immunoblotting. We found that Abi1 expression correlated positively with KRAS, but not BRAF mutation status in the examined tissue samples. Furthermore, Abi1 is overexpressed in inflammatory mucosa, sessile serrated polyps and adenomas, tubular adenomas, invasive CRC and CRC metastasis when compared to healthy mucosa and BRAF-mutated as well as KRAS wild-type hyperplastic polyps. Abi1 expression in carcinoma was independent of microsatellite stability of the tumor. Abi1 protein expression correlated with KRAS mutation in the analyzed CRC cell lines, and upregulation of Abi1 could be induced by TNFalpha treatment as well as transfection of wild-type CRC cells with mutant KRAS. The overexpression of Abi1 could be abolished by treatment with the PI3K-inhibitor Wortmannin after KRAS transfection. CONCLUSIONS/SIGNIFICANCE: Our results support a role for Abi1 as a downstream target of inflammatory response and adenomatous change as well as oncogenic KRAS mutation via PI3K, but not BRAF activation. Furthermore, they highlight a possible role for Abi1 as a marker for early KRAS mutation in hyperplastic polyps. Since the protein is a key player in actin dynamics, our data encourages further studies concerning the exact role of Abi1 in actin reorganization upon enhanced KRAS/PI3K signalling during colonic tumorigenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenoma/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Cytoskeletal Proteins/metabolism , Inflammation/pathology , Mutation/genetics , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , Adenoma/genetics , Aged , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Female , Humans , Immunohistochemistry , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Male , Middle Aged , Models, Biological , Neoplasm Metastasis , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras) , Transfection , Tumor Necrosis Factor-alpha/pharmacology
5.
Neurobiol Aging ; 33(4): 732-43, 2012 Apr.
Article in English | MEDLINE | ID: mdl-20674096

ABSTRACT

Previous studies identified engulfment adapter phosphotyrosine binding (PTB) domain containing 1 (GULP1) as an NPXY-motif interactor of low-density lipoprotein receptor-related protein 1 (LRP1) and suggested a potential relevance in Alzheimer's disease (AD). Since AD associated proteins amyloid-ß A4 precursor protein (APP) and LRP1 were shown to interact with the PTB domain of Fe65 and several other adapters via their intracellular NPXY-motifs, we examined a possible interaction of GULP1 PTB domain with the YENPTY-motif of APP. Here we demonstrate that GULP1 is present in human hippocampal and neocortical neurons. Confocal live cell imaging revealed that coexpressed and endogenous GULP1 colocalizes with APP in the Golgi and endoplasmic reticulum. Analysis of the interacting domains by co-immunoprecipitation of point and deletion mutants revealed that the interaction depends on the PTB domain of GULP1 and the YENPTY-motif of APP. Coexpression of GULP1 affected APP cell surface localization and suppressed generation of Aß40/42 and sAPPα. Taken together, these data identify GULP1 as a novel neuronal APP interacting protein that alters trafficking and processing of APP.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Amyloid beta-Protein Precursor/metabolism , Gene Expression Regulation/physiology , Neurons/metabolism , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Motifs/genetics , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Biotinylation , Cells, Cultured , Embryo, Mammalian , Endoplasmic Reticulum/metabolism , Gene Expression Regulation/genetics , Golgi Apparatus/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hippocampus/cytology , Humans , Immunoprecipitation , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Mice , Microscopy, Confocal , Neocortex/cytology , Neurons/ultrastructure , Peptide Fragments/metabolism , Protein Transport/genetics , Protein Transport/physiology , Transfection
6.
Am J Pathol ; 176(3): 1409-20, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20075202

ABSTRACT

Transgenic mice expressing human mutated superoxide dismutase 1 (SOD1) linked to familial forms of amyotrophic lateral sclerosis are frequently used as a disease model. We used the SOD1G93A mouse in a cross-breeding strategy to study the function of physiological prion protein (Prp). SOD1G93APrp-/- mice exhibited a significantly reduced life span, and an earlier onset and accelerated progression of disease, as compared with SOD1G93APrp+/+ mice. Additionally, during disease progression, SOD1G93APrp-/- mice showed impaired rotarod performance, lower body weight, and reduced muscle strength. Histologically, SOD1G93APrp-/- mice showed reduced numbers of spinal cord motor neurons and extended areas occupied by large vacuoles early in the course of the disease. Analysis of spinal cord homogenates revealed no differences in SOD1 activity. Using an unbiased proteomic approach, a marked reduction of glial fibrillary acidic protein and enhanced levels of collapsing response mediator protein 2 and creatine kinase were detected in SOD1G93APrp-/- versus SOD1G93A mice. In the course of disease, Bcl-2 decreases, nuclear factor-kappaB increases, and Akt is activated, but these changes were largely unaffected by Prp expression. Exclusively in double-transgenic mice, we detected a significant increase in extracellular signal-regulated kinase 2 activation at clinical onset. We propose that Prp has a beneficial role in the SOD1G93A amyotrophic lateral sclerosis mouse model by influencing neuronal and/or glial factors involved in antioxidative defense, rather than anti-apoptotic signaling.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Neuroprotective Agents/metabolism , Prions/metabolism , Amyotrophic Lateral Sclerosis/enzymology , Amyotrophic Lateral Sclerosis/pathology , Animals , Brain/enzymology , Brain/pathology , Breeding , Cell Count , DNA/metabolism , Disease Models, Animal , Disease Progression , Enzyme Activation , Female , Glial Fibrillary Acidic Protein/metabolism , Humans , Male , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/metabolism , Motor Neurons/pathology , Prion Proteins , Spinal Cord/enzymology , Spinal Cord/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Survival Analysis , Transgenes/genetics , Vacuoles/metabolism
7.
J Neurochem ; 99(2): 426-37, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17029597

ABSTRACT

The modulation of cell proliferation in neural progenitor cells (NPCs) is believed to play a role in neuronal regeneration. Recent studies showed that K(+) channel activity influenced cell proliferation. Therefore, we examined NPCs for K(+) channels and tested whether NPC self renewing can be modulated by synthetic K(+) channel modulators. The whole-cell K(+) current was partly K(+) dependent and showed a cumulative inactivating component. Two tetra-ethyl-ammonium ion (TEA)-sensitive K(+) currents with different voltage dependencies ( = 65 microm, E(50) = -0.3 +/- 1.3 mV and = 8 mm, E(50) = -15.2 +/- 2.8 mV) and an almost TEA-insensitive current were identified. Kaliotoxin blocked approximately 50% of the entire K(+) currents (IC(50) = 0.25 nm). These properties resembled functional characteristics of K(v)1.4, K(v)1.3 and K(v)3.1 channels. Transcripts for these channels, as well as proteins for K(v)1.3 and K(v)3.1, were identified. Immunocytochemical staining revealed K(v)1.3 and K(v)3.1 K(+) channel expression in almost all NPCs. The blockage of K(v)3.1 by low concentrations of TEA, as well as the blockage of K(v)1.3 by Psora-4, increased NPC proliferation. These findings underline the regulatory role of K(+) channels on the cell cycle and imply that K(+) channel modulators might be used therapeutically to activate endogenous NPCs.


Subject(s)
Cell Proliferation/drug effects , Kv1.3 Potassium Channel/antagonists & inhibitors , Neurons/metabolism , Shaw Potassium Channels/antagonists & inhibitors , Stem Cells/metabolism , Animals , Cell Culture Techniques/methods , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Immunohistochemistry , Kv1.3 Potassium Channel/genetics , Kv1.3 Potassium Channel/metabolism , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mesencephalon/cytology , Mesencephalon/growth & development , Mesencephalon/metabolism , Neurons/cytology , Neurons/drug effects , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , RNA, Messenger/analysis , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Scorpion Venoms/pharmacology , Shaw Potassium Channels/genetics , Shaw Potassium Channels/metabolism , Stem Cells/cytology , Stem Cells/drug effects , Tetraethylammonium/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL