Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
J Cell Physiol ; 234(5): 6731-6745, 2019 05.
Article in English | MEDLINE | ID: mdl-30273442

ABSTRACT

The Ras/mitogen-activated protein kinase (MAPK) pathway controls fundamental cellular processes such as proliferation, differentiation, and apoptosis. The dual-specificity phosphatase 6 (DUSP6) regulates cytoplasmic MAPK signaling by dephosphorylating and inactivating extracellular signal-regulated kinase (ERK1/2) MAPK. To determine the role of DUSP6 in the maintenance of intestinal homeostasis, we characterized the intestinal epithelial phenotype of Dusp6 knockout (KO) mice under normal, oncogenic, and proinflammatory conditions. Our results show that loss of Dusp6 increased crypt depth and epithelial cell proliferation without altering colonic architecture. Crypt regeneration capacity was also enhanced, as revealed by ex vivo Dusp6 KO organoid cultures. Additionally, loss of Dusp6 induced goblet cell expansion without affecting enteroendocrine and absorptive cell differentiation. Our data also demonstrate that Dusp6 KO mice were protected from acute dextran sulfate sodium-induced colitis, as opposed to wild-type mice. In addition, Dusp6 gene deletion markedly enhanced tumor load in Apc Min/+ mice. Decreased DUSP6 expression by RNA interference in HT29 colorectal cancer cells enhanced ERK1/2 activation levels and promoted both anchorage-independent growth in soft agar as well as invasion through Matrigel. Finally, DUSP6 mRNA expression in human colorectal tumors was decreased in advanced stage tumors compared with paired normal tissues. These results demonstrate that DUSP6 phosphatase, by controlling ERK1/2 activation, regulates colonic inflammatory responses, and protects the intestinal epithelium against oncogenic stress.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/physiology , Cell Transformation, Neoplastic/metabolism , Colon/pathology , Colorectal Neoplasms/metabolism , Dual Specificity Phosphatase 6/metabolism , Animals , Apoptosis/physiology , Colitis/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Dextran Sulfate , Dual Specificity Phosphatase 6/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Intestinal Mucosa/metabolism , Mice, Transgenic
2.
Bioconjug Chem ; 30(6): 1665-1676, 2019 06 19.
Article in English | MEDLINE | ID: mdl-31045351

ABSTRACT

To reply to as yet unmet medical needs to treat osteosarcoma, a form of primary bone cancer, we conceived the 12b80 compound by covalently conjugating antineoplastic compound doxorubicin to a bone targeting hydroxybisphosphonate vector and turned it into a prodrug through a custom linker designed to specifically trigger doxorubicin release in acidic bone tumor microenvironment. Synthesis of 12b80 was thoroughly optimized to be produced at gram scale. 12b80 was evaluated in vitro for high bone support affinity, specific release of doxorubicin in acidic condition, lower cytotoxicity, and cellular uptake of the prodrug. In vivo in rodents, 12b80 displayed rapid and sustained targeting of bone tissue and tumor-associated heterotopic bone and permitted a higher doxorubicin payload in tumor bone environment compared to nonvectorized doxorubicin. Consequently, 12b80 showed much lower toxicity compared to doxorubicin, promoted strong antitumor effects on rodent orthotopic osteosarcoma, displayed a dose-response therapeutic effect, and was more potent than doxorubicin/zoledronate combination.


Subject(s)
Antibiotics, Antineoplastic/chemistry , Bone Neoplasms/drug therapy , Diphosphonates/chemistry , Doxorubicin/analogs & derivatives , Osteosarcoma/drug therapy , Animals , Antibiotics, Antineoplastic/chemical synthesis , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/therapeutic use , Bone Neoplasms/pathology , Cell Line, Tumor , Chemistry Techniques, Synthetic , Diphosphonates/chemical synthesis , Diphosphonates/pharmacokinetics , Diphosphonates/therapeutic use , Doxorubicin/chemical synthesis , Doxorubicin/pharmacokinetics , Doxorubicin/therapeutic use , Female , Mice, Nude , Osteosarcoma/pathology , Rats
3.
Mol Carcinog ; 55(5): 671-87, 2016 May.
Article in English | MEDLINE | ID: mdl-25808857

ABSTRACT

Cathepsin B is a cysteine proteinase that primarily functions as an endopeptidase within endolysosomal compartments in normal cells. However, during tumoral expansion, the regulation of cathepsin B can be altered at multiple levels, thereby resulting in its overexpression and export outside of the cell. This may suggest a possible role of cathepsin B in alterations leading to cancer progression. The aim of this study was to determine the contribution of intracellular and extracellular cathepsin B in growth, tumorigenesis, and invasion of colorectal cancer (CRC) cells. Results show that mRNA and activated levels of cathepsin B were both increased in human adenomas and in CRCs of all stages. Treatment of CRC cells with the highly selective and non-permeant cathepsin B inhibitor Ca074 revealed that extracellular cathepsin B actively contributed to the invasiveness of human CRC cells while not essential for their growth in soft agar. Cathepsin B silencing by RNAi in human CRC cells inhibited their growth in soft agar, as well as their invasion capacity, tumoral expansion, and metastatic spread in immunodeficient mice. Higher levels of the cell cycle inhibitor p27(Kip1) were observed in cathepsin B-deficient tumors as well as an increase in cyclin B1. Finally, cathepsin B colocalized with p27(Kip1) within the lysosomes and efficiently degraded the inhibitor. In conclusion, the present data demonstrate that cathepsin B is a significant factor in colorectal tumor development, invasion, and metastatic spreading and may, therefore, represent a potential pharmacological target for colorectal tumor therapy.


Subject(s)
Carcinogenesis/genetics , Cathepsin B/genetics , Cathepsin B/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Animals , Caco-2 Cells , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Dipeptides/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , HEK293 Cells , HT29 Cells , Humans , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation
4.
BMC Cell Biol ; 14: 33, 2013 Aug 06.
Article in English | MEDLINE | ID: mdl-23919615

ABSTRACT

BACKGROUND: The transcription factor E2F4 controls proliferation of normal and cancerous intestinal epithelial cells. E2F4 localization in normal human intestinal epithelial cells (HIEC) is cell cycle-dependent, being cytoplasmic in quiescent differentiated cells but nuclear in proliferative cells. However, the intracellular signaling mechanisms regulating such E2F4 localization remain unknown. RESULTS: Treatment of quiescent HIEC with serum induced ERK1/2 activation, E2F4 phosphorylation, E2F4 nuclear translocation and G1/S phase transition while inhibition of MEK/ERK signaling by U0126 prevented these events. Stimulation of HIEC with epidermal growth factor (EGF) also led to the activation of ERK1/2 but, in contrast to serum or lysophosphatidic acid (LPA), EGF failed to induce E2F4 phosphorylation, E2F4 nuclear translocation and G1/S phase transition. Furthermore, Akt and GSK3ß phosphorylation levels were markedly enhanced in serum- or LPA-stimulated HIEC but not by EGF. Importantly, E2F4 phosphorylation, E2F4 nuclear translocation and G1/S phase transition were all observed in response to EGF when GSK3 activity was concomitantly inhibited by SB216763. Finally, E2F4 was found to be overexpressed, phosphorylated and nuclear localized in epithelial cells from human colorectal adenomas exhibiting mutations in APC and KRAS or BRAF genes, known to deregulate GSK3/ß-catenin and MEK/ERK signaling, respectively. CONCLUSIONS: The present results indicate that MEK/ERK activation and GSK3 inhibition are both required for E2F4 phosphorylation as well as its nuclear translocation and S phase entry in HIEC. This finding suggests that dysregulated E2F4 nuclear localization may be an instigating event leading to hyperproliferation and hence, of tumor initiation and promotion in the colon and rectum.


Subject(s)
Cell Proliferation/drug effects , E2F4 Transcription Factor/drug effects , E2F4 Transcription Factor/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , MAP Kinase Signaling System/physiology , Mitogens/pharmacology , Transcription, Genetic/drug effects , Adenoma/metabolism , Adenoma/pathology , Butadienes/pharmacology , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Line , Cells, Cultured , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , E2F4 Transcription Factor/genetics , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/drug effects , Humans , Intestinal Mucosa/drug effects , Lysophospholipids/pharmacology , MAP Kinase Signaling System/drug effects , Nitriles/pharmacology , Phosphorylation/drug effects , Phosphorylation/physiology , Transcription, Genetic/physiology
5.
Carcinogenesis ; 33(3): 529-37, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22201186

ABSTRACT

Recent evidences suggest that the activity of glycogen synthase kinase-3 (GSK3) contributes to the tumorigenic potential of pancreatic cancer cells through modulation of cell proliferation and survival. However, further investigations are needed to identify GSK3-dependent mechanisms involved in the control of pancreatic cancer cell proliferation and survival. This study was undertaken to provide further support for a role of GSK3 in pancreatic cancer cell growth as well as to identify new cellular and molecular mechanisms involved. Herein, we demonstrate that prolonged inhibition of GSK3 triggers an apoptotic response only in human pancreatic cancer cells but not in human non-transformed pancreatic epithelial cells. We show that prolonged inhibition of GSK3 activity increases Bim messenger RNA and protein expressions. Moreover, we provide evidence that activation of the c-jun N-terminal kinase (JNK) pathway is necessary for the GSK3 inhibition-mediated increase in Bim expression and apoptotic response. Finally, we demonstrate that concomitant inhibition of GSK3 potentiates the death ligand-induced apoptotic response in pancreatic cancer cells but not in non-transformed pancreatic epithelial cells and that this effect also requires JNK activity. Considering that different approaches leading to stimulation of death receptor signaling are under clinical trials for treatment of unresectable or metastatic pancreatic cancer, inhibition of GSK3 could represent an attractive new avenue to improve their effectiveness.


Subject(s)
Apoptosis , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Pancreatic Neoplasms/metabolism , Anthracenes/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Indoles/pharmacology , MAP Kinase Signaling System , Maleimides/pharmacology , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Pancreas/metabolism , Pancreatic Neoplasms/pathology , Phosphorylation , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pyridines/pharmacology , Pyrimidines/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Death Domain/metabolism
6.
Mol Cancer ; 9: 271, 2010 Oct 13.
Article in English | MEDLINE | ID: mdl-20942929

ABSTRACT

BACKGROUND: Among the most harmful of all genetic abnormalities that appear in colorectal cancer (CRC) development are mutations of KRAS and its downstream effector BRAF as they result in abnormal extracellular signal-related kinase (ERK) signaling. In a previous report, we had shown that expression of a constitutive active mutant of MEK1 (caMEK) in normal rat intestinal epithelial cells (IECs) induced morphological transformation associated with epithelial to mesenchymal transition, growth in soft agar, invasion and metastases in nude mice. Results from microarrays comparing control to caMEK-expressing IECs identified the gene encoding for serpinE2, a serine protease inhibitor, as a potential target of activated MEK1. RESULTS: 1- RT-PCR and western blot analyses confirmed the strong up-regulation of serpinE2 expression and secretion by IECs expressing oncogenic MEK, Ras or BRAF. 2- Interestingly, serpinE2 mRNA and protein were also markedly enhanced in human CRC cells exhibiting mutation in KRAS and BRAF. 3- RNAi directed against serpinE2 in caMEK-transformed rat IECs or in human CRC cell lines HCT116 and LoVo markedly decreased foci formation, anchorage-independent growth in soft agarose, cell migration and tumor formation in nude mice. 4- Treatment of CRC cell lines with U0126 markedly reduced serpinE2 mRNA levels, indicating that expression of serpinE2 is likely dependent of ERK activity. 5- Finally, Q-PCR analyses demonstrated that mRNA levels of serpinE2 were markedly increased in human adenomas in comparison to healthy adjacent tissues and in colorectal tumors, regardless of tumor stage and grade. CONCLUSIONS: Our data indicate that serpinE2 is up-regulated by oncogenic activation of Ras, BRAF and MEK1 and contributes to pro-neoplastic actions of ERK signaling in intestinal epithelial cells. Hence, serpinE2 may be a potential therapeutic target for colorectal cancer treatment.


Subject(s)
Colorectal Neoplasms/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Serpin E2/metabolism , Animals , Blotting, Western , Butadienes/pharmacology , Cell Line, Tumor , Colorectal Neoplasms/genetics , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/genetics , HCT116 Cells , Humans , In Vitro Techniques , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , Mice , Mice, Nude , Nitriles/pharmacology , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , RNA Interference , Rats , Reverse Transcriptase Polymerase Chain Reaction , Serpin E2/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , ras Proteins/genetics , ras Proteins/metabolism
7.
Oncotarget ; 11(46): 4281-4292, 2020 Nov 17.
Article in English | MEDLINE | ID: mdl-33245733

ABSTRACT

PURPOSE: 12b80 combines doxorubicin bound to a bone targeting hydroxybisphosphonate vector using a pH-sensitive linker, designed to specifically trigger doxorubicin release in an acidic bone tumor microenvironment. This phase I study aimed to determine the safety and toxicity profiles of 12b80 in dogs with naturally occurring osteosarcoma, with the objective to translate findings from dogs to humans. EXPERIMENTAL DESIGN: Ten client-owned dogs with osteosarcoma were enrolled in an accelerated dose-titration design followed by 3 + 3 design. Dogs received three cycles of 12b80 intravenous injection at 4 mg/kg (n = 1), 6 mg/kg (n = 2), 8 mg/kg (n = 3), and 10 mg/kg (n = 4). Endpoints included safety, tolerability, maximum tolerated dose (MTD), and dose-limiting toxicity (DLT). RESULTS: The MTD of 12b80 was 8 mg/kg (i.e., equivalent dose of doxorubicin of 110 mg/m2, range: 93-126). Most adverse events included grade ≤ 2 gastrointestinal disorders and hypersensitivity reactions. No hematological or cardiac DLT were observed at any dose tested. CONCLUSIONS: In dogs, 12b80 is overall well tolerated and expends the MTD of doxorubicin up to four times the standard dose of 30 mg/m2. These results demonstrate the potential therapeutic benefit of 12b80 in canine and human osteosarcoma.

8.
Acta Oncol ; 47(8): 1502-12, 2008.
Article in English | MEDLINE | ID: mdl-18607838

ABSTRACT

BACKGROUND: The gene encoding cortactin, CTTN (locus 11q13), an actin-binding substrate of Src kinases, is frequently amplified in breast and head and neck squamous cell carcinomas (HNSCC) and cortactin overexpression is thought to contribute in a significant way to the invasive phenotype of these tumors. Elevated Epidermal Growth Factor receptor (EGFR) expression is also commonly observed in HNSCC and has been associated with poor prognosis and resistance to cytotoxic agents, including ionizing radiation. It has been suggested that cortactin overexpression may increase EGFR levels in these tumors by affecting receptor downregulation, however we recently found by multivariate analysis, that cortactin expression status remained an independent prognostic factor for local recurrence, disease-free survival, and overall survival. MATERIAL AND METHODS: To examine the potential link between cortactin overexpression and EGFR status, we compared cortactin and EGFR levels in a series of tumor lines derived from HNSCC. RNAi-mediated silencing was performed in cortactin overexpressing cells and in vivo tumoral potential with respect to cortactin and EGFR status was analyzed. RESULTS AND DISCUSSION: Cortactin and EGFR levels were not strictly coupled in these lines and cortactin depletion did not decrease steady state receptor levels, although it did affect the epithelial to mesenchymal phenotypic conversion of cells. These results, together with clinical findings point to the existence of an EGFR-independent role of cortactin in HNSCC that may have important implications regarding the design of targeted therapies to combat tumor spread.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cortactin/metabolism , ErbB Receptors/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Animals , Blotting, Southern , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cells, Cultured , Cortactin/genetics , ErbB Receptors/genetics , Female , Fluorescent Antibody Technique , Gene Amplification , Head and Neck Neoplasms/genetics , Humans , Immunoenzyme Techniques , Kidney/metabolism , Kidney/pathology , Mice , Mice, Nude , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology
9.
FASEB J ; 20(9): 1489-91, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16723384

ABSTRACT

Extracellular matrix (ECM) receptors of the integrin family initiate changes in cell shape and motility by recruiting signaling components that coordinate these events. Integrin-linked kinase (ILK) is one such partner of beta1 integrins that participates in dynamic rearrangement of cell-matrix adhesions and cell spreading by mechanisms that are not well understood. To further elucidate the role of ILK in these events, we engineered a chimeric molecule comprising ILK fused to a membrane-targeted green fluorescent protein (ILK-GFP-F). ILK-GFP-F is highly enriched in cell-matrix adhesions, and its expression in fibroblasts leads to an accumulation of focal adhesions (2-5 microm) and elongated adhesions (>5 microm). ILK-GFP-F enhances cell spreading on fibronectin and induces a constitutive increase in the levels of GTP-bound Rac-1. Conversely, ILK knock-down by siRNA transfection decreases active Rac-1. Endogenous ILK was found to associate with PKL (paxillin kinase linker) and the Rac/Cdc42 guanine nucleotide exchange factor betaPIX. Further, expression of a dominant negative betaPIX mutant reversed the increase in active Rac-1 levels of ILK-GFP-F-expressing cells, thus placing betaPIX in the pathway leading from ILK to Rac-1 activation. However, expression of constitutively active Rac only partially restores the spreading defects of ILK-depleted cells, suggesting that an additional ILK-dependent signal is required for cell spreading.


Subject(s)
Endothelium, Vascular/physiology , Extracellular Matrix/physiology , Protein Serine-Threonine Kinases/physiology , rac1 GTP-Binding Protein/metabolism , Animals , Aorta , Cattle , Cells, Cultured , Endothelium, Vascular/cytology , Genetic Vectors , Kinetics , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology , Transfection , rac1 GTP-Binding Protein/genetics
10.
Oncogene ; 22(43): 6785-93, 2003 Oct 02.
Article in English | MEDLINE | ID: mdl-14555991

ABSTRACT

Bim is a proapoptotic member of the Bcl-2 family that shares only the BH3 domain with this family. Three Bim proteins Bim-EL, Bim-L and Bim-S are synthesized from the same transcript. We report here that Bim-EL when phosphorylated by Erk1/2 is rapidly degraded via the proteasome pathway. Using different cellular models we evidence that serine 69 is both necessary and sufficient for Erk1/2-mediated phosphorylation and degradation of Bim-EL. In K562 cells, Phorbol 12-myristate 13-acetate activates Erk1/2 and consequently increases Bim-EL phosphorylation and degradation by the proteasome, resulting in cell survival, while the Bcr-Abl inhibitor imatinib abrogates Bim-EL phosphorylation and degradation and induces caspase activation and apoptosis. We also show that Bim-EL(S69G) promotes apoptosis more efficiently than Bim-EL-WT in K562 cells. Altogether, our findings demonstrate that phosphorylation of Bim-EL by Erk1/2 on serine 69 selectively leads to its proteasomal degradation and therefore represents a new and important mechanism of Bim regulation.


Subject(s)
Apoptosis , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cysteine Endopeptidases/metabolism , Membrane Proteins , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Multienzyme Complexes/metabolism , Proto-Oncogene Proteins , Serine/chemistry , Amino Acid Sequence , Antineoplastic Agents, Hormonal/pharmacology , Apoptosis Regulatory Proteins , Bcl-2-Like Protein 11 , Binding Sites , Caspases/metabolism , Cell Line , Cell Survival , Gene Expression Regulation , Glutathione Transferase/metabolism , Humans , K562 Cells , Lymphoma/metabolism , Mitogen-Activated Protein Kinase 3 , Models, Biological , Molecular Sequence Data , Phosphorylation , Proteasome Endopeptidase Complex , Protein Isoforms , Proto-Oncogene Proteins c-raf/metabolism , Tamoxifen/pharmacology , Tetradecanoylphorbol Acetate , Time Factors , Transfection , Tumor Cells, Cultured
11.
Mol Cell Biol ; 33(11): 2275-84, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23530062

ABSTRACT

Polymorphisms of PTPN11 encoding SHP-2 are biomarkers for ulcerative colitis (UC) susceptibility. However, their functional relevance is unknown. We thus investigated the role of epithelial SHP-2 in the control of intestinal homeostasis. Mice with an intestinal epithelial cell-specific SHP-2 deletion (SHP-2(IEC-KO) mice) were generated. Control and SHP-2(IEC-KO) mice were monitored for clinical symptoms and sacrificed for histological staining and Western blot analyses. Cytokines and chemokines, as well as intestinal permeability, were quantified. SHP-2 mRNA expression was evaluated in control and UC patients. SHP-2(IEC-KO) mice showed growth retardation compared to control littermates and rapidly developed severe colitis. Colon architecture was markedly altered with infiltration of immune cells, crypt abscesses, neutrophil accumulation, and reduced goblet cell numbers. Decreased expression of claudins was associated with enhanced intestinal permeability in mutant SHP-2(IEC-KO) mice. Inflammatory transcription factors Stat3 and NF-κB were hyperactivated early in the mutant colonic epithelium. Levels of several epithelial chemokines and cytokines were markedly enhanced in SHP-2(IEC-KO) mice. Of note, antibiotic treatment remarkably impaired the development of colitis in SHP-2(IEC-KO) mice. Finally, SHP-2 mRNA levels were significantly reduced in intestinal biopsy specimens from UC patients. Our results establish intestinal epithelial SHP-2 as a critical determinant for prevention of gut inflammation.


Subject(s)
Colitis, Ulcerative/enzymology , Colitis/genetics , Intestines/physiopathology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Colitis/drug therapy , Colitis/physiopathology , Colitis, Ulcerative/genetics , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Female , Humans , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/metabolism , Permeability , Pregnancy , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
12.
J Signal Transduct ; 2011: 834948, 2011.
Article in English | MEDLINE | ID: mdl-21637382

ABSTRACT

In many cell types, growth factor removal induces the release of cytochrome-c from mitochondria that leads to activation of caspase-9 in the apoptosome complex. Here, we show that sustained stimulation of the Raf-1/MAPK1,3 pathway prevents caspase-9 activation induced by serum depletion in CCL39/ΔRaf-1:ER fibroblasts. The protective effect mediated by Raf-1 is sensitive to MEK inhibition that is sufficient to induce caspase-9 cleavage in exponentially growing cells. Raf-1 activation does not inhibit the release of cytochrome-c from mitochondria while preventing caspase-9 activation. Gel filtration chromatography analysis of apoptosome formation in cells shows that Raf-1/MAPK1,3 activation does not interfere with APAF-1 oligomerization and recruitment of caspase 9. Raf-1-mediated caspase-9 inhibition is sensitive to emetine, indicating that the protective mechanism requires protein synthesis. However, the Raf/MAPK1,3 pathway does not regulate XIAP. Taken together, these results indicate that the Raf-1/MAPK1,3 pathway controls an apoptosis regulator that prevents caspase-9 activation in the apoptosome complex.

13.
FEBS J ; 277(1): 2-21, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19843174

ABSTRACT

The Ras/Raf/extracellular signal-regulated kinase (ERK) signaling pathway plays a crucial role in almost all cell functions and therefore requires exquisite control of its spatiotemporal activity. Depending on the cell type and stimulus, ERK activity will mediate different antiproliferative events, such as apoptosis, autophagy and senescence in vitro and in vivo. ERK activity can promote either intrinsic or extrinsic apoptotic pathways by induction of mitochondrial cytochrome c release or caspase-8 activation, permanent cell cycle arrest or autophagic vacuolization. These unusual effects require sustained ERK activity in specific subcellular compartments and could depend on the presence of reactive oxygen species. We will summarize the mechanisms involved in Ras/Raf/ERK antiproliferative functions.


Subject(s)
Cell Death/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , Animals , Apoptosis/physiology , Autophagy/physiology , Caspase 8/physiology , Cellular Senescence/physiology , Cytochromes c/physiology , Humans , Lysosomes/physiology , MAP Kinase Signaling System , Models, Biological , Proto-Oncogene Proteins c-bcl-2/physiology , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/physiology , raf Kinases/physiology , ras Proteins/physiology
14.
Pigment Cell Melanoma Res ; 21(5): 534-44, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18715233

ABSTRACT

Somatic activating mutations of BRAF are the earliest and most common genetic abnormality detected in the genesis of human melanoma. However, the mechanism(s) by which activated BRAF promotes melanoma cell cycle progression and/or survival remain unclear. Here we demonstrate that expression of BIM, a pro-apoptotic member of the BCL-2 family, is inhibited by BRAF-->MEK-->ERK signaling in mouse and human melanocytes and in human melanoma cells. Trophic factor deprivation of melanocytes leads to elevated BIM expression. However, re-addition of trophic factors or activation of a conditional form of BRAF(V600E) leads to rapid inhibition of BIM expression. In both cases, inhibition of BIM expression was dependent on the activity of MEK1/2 and the proteasome. Consistent with these observations, pharmacological inhibition of BRAF(V600E) or MEK1/2 in human melanoma cells (using PLX4720 and CI-1040 respectively) led to a striking elevation of BIM expression. Re-activation of BRAF-->MEK-->ERK signaling led to phosphorylation of BIM-EL on serine 69 and its subsequent degradation. Interestingly, endogenous expression of BIM in melanoma cells was insufficient to induce apoptosis unless combined with serum deprivation. Under these circumstances, inhibition of BIM expression by RNA interference provided partial protection from apoptosis. These data suggest that regulation of BIM expression by BRAF-->MEK-->ERK signaling is one mechanism by which oncogenic BRAF(V600E) can influence the aberrant physiology of melanoma cells.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Melanoma/metabolism , Melanoma/pathology , Membrane Proteins/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Animals , Apoptosis/physiology , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Cell Line, Tumor , Cell Survival , Culture Media, Serum-Free , Humans , Intercellular Signaling Peptides and Proteins/metabolism , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase 2/metabolism , Melanocytes/cytology , Melanocytes/metabolism , Membrane Proteins/genetics , Mice , Mitochondria/metabolism , Point Mutation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL