Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Microorganisms ; 12(4)2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38674581

ABSTRACT

Global rewiring of bacterial gene expressions in response to environmental cues is mediated by regulatory proteins such as the CsrA global regulator from E. coli. Several direct mRNA and sRNA targets of this protein have been identified; however, high-throughput studies suggest an expanded RNA targetome for this protein. In this work, we demonstrate that CsrA can extend its network by directly binding and regulating the evgA and acnA transcripts, encoding for regulatory proteins. CsrA represses EvgA and AcnA expression and disrupting the CsrA binding sites of evgA and acnA, results in broader gene expression changes to stress response networks. Specifically, altering CsrA-evgA binding impacts the genes related to acidic stress adaptation, and disrupting the CsrA-acnA interaction affects the genes involved in metal-induced oxidative stress responses. We show that these interactions are biologically relevant, as evidenced by the improved tolerance of evgA and acnA genomic mutants depleted of CsrA binding sites when challenged with acid and metal ions, respectively. We conclude that EvgA and AcnA are intermediate regulatory hubs through which CsrA can expand its regulatory role. The indirect CsrA regulation of gene networks coordinated by EvgA and AcnA likely contributes to optimizing cellular resources to promote exponential growth in the absence of stress.

2.
Cancer Res ; 73(19): 5974-84, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23943799

ABSTRACT

The Notch pathway has been implicated in a number of malignancies with different roles that are cell- and tissue-type dependent. Notch1 is a putative oncogene in non-small cell lung cancer (NSCLC) and activation of the pathway represents a negative prognostic factor. To establish the role of Notch1 in lung adenocarcinoma, we directly assessed its requirement in Kras-induced tumorigenesis in vivo using an autochthonous model of lung adenocarcinoma with concomitant expression of oncogenic Kras and deletion of Notch1. We found that Notch1 function is required for tumor initiation via suppression of p53-mediated apoptosis through the regulation of p53 stability. These findings implicate Notch1 as a critical effector in Kras-driven lung adenocarcinoma and as a regulator of p53 at a posttranslational level. Moreover, our study provides new insights to explain, at a molecular level, the correlation between Notch1 activity and poor prognosis in patients with NSCLC carrying wild-type p53. This information is critical for design and implementation of new therapeutic strategies in this cohort of patients representing 50% of NSCLC cases.


Subject(s)
Adenocarcinoma/pathology , Apoptosis , Cell Transformation, Neoplastic , Lung Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/physiology , Receptor, Notch1/physiology , Tumor Suppressor Protein p53/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Blotting, Western , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle , Cell Proliferation , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Knockout , Mutation/genetics , Signal Transduction , Tumor Cells, Cultured , Tumor Suppressor Protein p53/chemistry
3.
Trends Mol Med ; 19(5): 320-7, 2013 May.
Article in English | MEDLINE | ID: mdl-23545339

ABSTRACT

The Notch signaling pathways are known to play critical roles during pancreatic development, but it remains unclear what functions are important in the adult organ. One area of debate is the role of Notch signaling in the development of pancreatic ductal adenocarcinoma (PDAC) and proposed precursor lesions, pancreatic intraepithelial neoplasia (PanIN). Initial studies revealed that Notch signaling is reactivated during PDAC initiation and development, suggesting that Notch promotes PDAC and may therefore represent a target for drug development. However, more recent work reveals a tumor suppressive role for Notch receptors in the context of PanIN development. Here, we summarize the current literature describing Notch signaling in the development of PDAC, and discuss the potential of the Notch pathway as a therapeutic target.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Pancreatic Neoplasms/metabolism , Receptors, Notch/metabolism , Signal Transduction , Animals , Carcinoma, Pancreatic Ductal/genetics , Disease Models, Animal , Humans , Mice , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Pancreatic Neoplasms/genetics , Receptors, Notch/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
4.
PLoS One ; 7(12): e52133, 2012.
Article in English | MEDLINE | ID: mdl-23284900

ABSTRACT

Pancreatic ductal adenocarcinoma is believed to arise from precursor lesions termed pancreatic intraepithelial neoplasia (PanIN). Mouse models have demonstrated that targeted expression of activated K-ras to mature acinar cells in the pancreas induces the spontaneous development of PanIN lesions; implying acinar-to-ductal metaplasia (ADM) is a key event in this process. Recent studies suggest Notch signaling is a key regulator of ADM. To assess if Notch1 is required for K-ras driven ADM we employed both an in vivo mouse model and in vitro explant culture system, in which an oncogenic allele of K-ras is activated and Notch1 is deleted simultaneously in acinar cells. Our results demonstrate that oncogenic K-ras is sufficient to drive ADM both in vitro and in vivo but that loss of Notch1 has a minimal effect on this process. Interestingly, while loss of Notch1 in vivo does not affect the severity of PanIN lesions observed, the overall numbers of lesions were greater in mice with deleted Notch1. This suggests Notch1 deletion renders acinar cells more susceptible to formation of K-ras-induced PanINs.


Subject(s)
Acinar Cells/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Genes, ras , Receptor, Notch1/genetics , Animals , Carcinoma in Situ/genetics , Carcinoma in Situ/metabolism , Carcinoma in Situ/pathology , Carcinoma, Pancreatic Ductal/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Gene Deletion , Metaplasia , Mice , Pancreatitis/genetics , Pancreatitis/metabolism , Pancreatitis/pathology , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Receptor, Notch1/metabolism , Signal Transduction
5.
Cancer Cell ; 19(4): 527-40, 2011 Apr 12.
Article in English | MEDLINE | ID: mdl-21481793

ABSTRACT

The Merlin/NF2 tumor suppressor restrains cell growth and tumorigenesis by controlling contact-dependent inhibition of proliferation. We have identified a tight-junction-associated protein complex comprising Merlin, Angiomotin, Patj, and Pals1. We demonstrate that Angiomotin functions downstream of Merlin and upstream of Rich1, a small GTPase Activating Protein, as a positive regulator of Rac1. Merlin, through competitive binding to Angiomotin, releases Rich1 from the Angiomotin-inhibitory complex, allowing Rich1 to inactivate Rac1, ultimately leading to attenuation of Rac1 and Ras-MAPK pathways. Patient-derived Merlin mutants show diminished binding capacities to Angiomotin and are unable to dissociate Rich1 from Angiomotin or inhibit MAPK signaling. Depletion of Angiomotin in Nf2(-/-) Schwann cells attenuates the Ras-MAPK signaling pathway, impedes cellular proliferation in vitro and tumorigenesis in vivo.


Subject(s)
Intercellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Microfilament Proteins/physiology , Neurofibromin 2/physiology , Signal Transduction/physiology , Tight Junctions/physiology , Tumor Suppressor Proteins/physiology , Amino Acid Sequence , Angiomotins , Animals , Cell Proliferation , GTPase-Activating Proteins/physiology , Humans , MAP Kinase Signaling System , Mice , Mice, SCID , Molecular Sequence Data , Nucleoside-Phosphate Kinase/physiology , Peripheral Nerves/chemistry , Schwann Cells/chemistry , Tight Junction Proteins , rac1 GTP-Binding Protein/physiology
6.
Cancer Res ; 70(11): 4280-6, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20484026

ABSTRACT

K-ras is the most commonly mutated oncogene in pancreatic cancer and its activation in murine models is sufficient to recapitulate the spectrum of lesions seen in human pancreatic ductal adenocarcinoma (PDAC). Recent studies suggest that Notch receptor signaling becomes reactivated in a subset of PDACs, leading to the hypothesis that Notch1 functions as an oncogene in this setting. To determine whether Notch1 is required for K-ras-induced tumorigenesis, we used a mouse model in which an oncogenic allele of K-ras is activated and Notch1 is deleted simultaneously in the pancreas. Unexpectedly, the loss of Notch1 in this model resulted in increased tumor incidence and progression, implying that Notch1 can function as a tumor suppressor gene in PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Genes, Tumor Suppressor , Genes, ras , Pancreatic Neoplasms/genetics , Receptor, Notch1/genetics , Animals , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Disease Progression , Gene Deletion , Gene Expression Regulation, Neoplastic , Mice , Mice, Transgenic , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Receptor, Notch1/deficiency , Receptor, Notch1/metabolism , Signal Transduction , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...