Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
Hepatology ; 71(3): 929-942, 2020 03.
Article in English | MEDLINE | ID: mdl-31692012

ABSTRACT

BACKGROUND AND AIMS: Aristolochic acid (AA) exposure has been statistically associated with human liver cancers. However, direct evidence of AA exposure-induced liver cancer is absent. This study aims to establish a direct causal relationship between AA exposure and liver cancers based on a mouse model and then explores the AA-mediated genomic alterations that could be implicated in human cancers with AA-associated mutational signature. APPROACH AND RESULTS: We subjected mice, including phosphatase and tensin homolog (Pten)-deficient ones, to aristolochic acid I (AAI) alone or a combination of AAI and CCl4 . Significantly, AAI exposure induced mouse liver cancers, including hepatocellular carcinoma (HCC) and combined HCC and intrahepatic cholangiocarcinoma, in a dose-dependent manner. Moreover, AAI exposure also enhanced tumorigenesis in these CCl4 -treated or Pten-deficient mice. AAI led to DNA damage and AAI-DNA adduct that could initiate liver cancers through characteristic adenine-to-thymine transversions, as indicated by comprehensive genomic analysis, which revealed recurrent mutations in Harvey rat sarcoma virus oncogene. Interestingly, an AA-associated mutational signature was mainly implicated in human liver cancers, especially from China. Moreover, we detected the AAI-DNA adduct in 25.8% (16/62) of paratumor liver tissues from randomly selected Chinese patients with HCC. Furthermore, based on phylogenetic analysis, the characteristic mutations were found in the initiating malignant clones in the AA-implicated mouse and human liver cancers where the mutations of tumor protein p53 and Janus kinase 1 were prone to be significantly enriched in the AA-affected human tumors. CONCLUSIONS: This study provides evidence for AA-induced liver cancer with the featured mutational processes during malignant clonal evolution, laying a solid foundation for the prevention and diagnosis of AA-associated human cancers, especially liver cancers.


Subject(s)
Aristolochic Acids/toxicity , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Mutation , Animals , Bile Duct Neoplasms/chemically induced , Bile Duct Neoplasms/genetics , Carbon Tetrachloride/toxicity , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Cholangiocarcinoma/chemically induced , Cholangiocarcinoma/genetics , DNA Damage , Dose-Response Relationship, Drug , Humans , Janus Kinase 1/genetics , Male , Mice , Mice, Inbred C57BL , Tumor Suppressor Protein p53/genetics , raf Kinases/physiology
2.
Br J Cancer ; 121(4): 332-339, 2019 08.
Article in English | MEDLINE | ID: mdl-31312030

ABSTRACT

BACKGROUND: A phase Ib study of binimetinib and capecitabine for gemcitabine-pretreated biliary tract cancer (BTC) patients was conducted. METHODS: Binimetinib and capecitabine were dosed twice daily on days 1-14, in 3-week cycles. In the dose-escalation (DE) part, three dose levels (DL) were tested (DL1: binimetinib/capecitabine, 15 mg/1000 mg/m2; DL2: 30 mg/1000 mg/m2; DL3: 30 mg/1250 mg/m2). RESULTS: In the DE part, nine patients were recruited and no dose-limiting toxicity was noted. Therefore, the recommended phase 2 dose was determined as DL3. In the expansion part, 25 patients were enrolled. In total, 34 patients, 25 (73.5%) and 9 patients (26.5%) were second-line and third-line settings, respectively. The 3-month progression-free survival (PFS) rate was 64.0%, and the median PFS and overall survival (OS) were 4.1 and 7.8 months. The objective response rate and disease control rate were 20.6% and 76.5%. In total, 68.4% of stable diseases were durable (> 12 weeks). Furthermore, patients with RAS/RAF/MEK/ERK pathway mutations (38.5%) showed significantly better tumour response (p = 0.028), PFS (5.4 vs. 3.5 months, p = 0.010) and OS (10.8 vs. 5.9 months, p = 0.160) than wild type. Most of the adverse events were grade 1/2 and manageable. CONCLUSIONS: A combination of binimetinib and capecitabine shows acceptable tolerability and promising antitumor efficacy for gemcitabine-pretreated BTC, especially in patients with RAS/RAF/MEK/ERK pathway mutations. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov (Identifier: NCT02773459).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biliary Tract Neoplasms/drug therapy , Extracellular Signal-Regulated MAP Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/genetics , Mutation , raf Kinases/genetics , ras Proteins/genetics , Aged , Benzimidazoles/administration & dosage , Benzimidazoles/adverse effects , Biliary Tract Neoplasms/genetics , Biliary Tract Neoplasms/mortality , Biliary Tract Neoplasms/psychology , Capecitabine/administration & dosage , Capecitabine/adverse effects , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/physiology , Female , Humans , Male , Middle Aged , Mitogen-Activated Protein Kinase Kinases/physiology , Quality of Life , Signal Transduction , raf Kinases/physiology , ras Proteins/physiology
3.
Nucleic Acids Res ; 45(18): 10518-10533, 2017 Oct 13.
Article in English | MEDLINE | ID: mdl-29048592

ABSTRACT

The yeast 2-µm plasmid is a remarkable genetic parasite, managing efficient maintenance at high-copy number with minimal impact on the host. Equal partitioning of the plasmid upon host cell division requires plasmid proteins Rep1 and Rep2 and the plasmid STB locus. The Rep proteins and the plasmid-encoded Raf protein also regulate plasmid gene transcription. In this study, protein interaction assays, sequence analyses and mutational approaches were used to identify domains and residues in Rep2 and Raf required for association with Rep1 and Rep2 and to delineate the Rep2 DNA-binding domain. Rep2 and Raf displayed similarities in interactions with Rep1 and Rep2, in having Rep1 promote their STB association in vivo, and in stabilizing Rep protein levels. Rep2 mutants impaired for self-association were competent for transcriptional repression while those deficient for Rep1 association were not. Surprisingly, Rep2 mutants impaired for either Rep1 interaction or self-association were able to maintain efficient plasmid inheritance provided Raf was present and competent for Rep protein interaction. Our findings provide insight into the Rep protein complexes required for partitioning and transcriptional repression, and suggest that in addition to its transcriptional function, Raf stabilization of Rep partitioning proteins contributes to the remarkable persistence of the 2-µm plasmid.


Subject(s)
Plasmids/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Trans-Activators/metabolism , raf Kinases/metabolism , raf Kinases/physiology , Cell Division , Inheritance Patterns , Organisms, Genetically Modified , Protein Binding , Protein Stability
4.
Genes Dev ; 24(24): 2800-11, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21159820

ABSTRACT

Organization into polarized three-dimensional structures defines whether epithelial cells are normal or malignant. In a model of morphogenesis, we show that inhibiting key signaling pathways in human breast cancer cells leads to "phenotypic reversion" of the malignant cells. Using architecture as an endpoint, we report that, in all cases, signaling through Raf/MEK/ERK disrupted tissue polarity via matrix metalloproteinase9 (MMP9) activity. Induction of Raf or activation of an engineered, functionally inducible MMP9 in nonmalignant cells led to loss of tissue polarity, and reinitiated proliferation. Conversely, inhibition of Raf or MMP9 with small molecule inhibitors or shRNAs restored the ability of cancer cells to form polarized quiescent structures. Silencing MMP9 expression also reduced tumor growth dramatically in a murine xenograft model. LC-MS/MS analysis comparing conditioned medium from nonmalignant cells with or without active MMP9 revealed laminin 111 (LM1) as an important target of MMP9. LM1 has been implicated in acinar morphogenesis; thus, its degradation by MMP9 provides a mechanism for loss of tissue polarity and reinitiation of growth associated with MMP9 activity. These findings underscore the importance of the dynamic reciprocity between the extracellular matrix integrity, tissue polarity, and Raf/MEK/ERK and MMP9 activities, providing an axis for either tissue homeostasis or malignant progression.


Subject(s)
Breast Neoplasms/pathology , Breast/cytology , Matrix Metalloproteinase 9/metabolism , Morphogenesis , raf Kinases/physiology , Animals , Cell Culture Techniques , Cell Polarity , Cell Proliferation , Humans , Laminin/metabolism , Mice , Neoplasms, Experimental , Signal Transduction , Transplantation, Heterologous
5.
Biochim Biophys Acta ; 1863(4): 770-84, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26844774

ABSTRACT

Melanoma has markedly increased worldwide during the past several decades in the Caucasian population and is responsible for 80% of skin cancer deaths. Considering that metastatic melanoma is almost completely resistant to most current therapies and is linked with a poor patient prognosis, it is crucial to further investigate potential molecular targets. Major cell-autonomous drivers in the pathogenesis of this disease include the classical MAPK (i.e., RAS-RAF-MEK-ERK), WNT, and PI3K signaling pathways. These pathways play a major role in defining the progression of melanoma, and some have been the subject of recent pharmacological strategies to treat this belligerent disease. This review describes the latest advances in the understanding of melanoma progression and the major molecular pathways involved. In addition, we discuss the roles of emerging molecular players that are involved in melanoma pathogenesis, including the functional role of the melanoma tumor antigen, p97/MFI2 (melanotransferrin).


Subject(s)
Melanoma/genetics , Melanoma/pathology , Oncogenes/physiology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Animals , Disease Progression , Genes, ras/physiology , Humans , MAP Kinase Signaling System/physiology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/genetics , Wnt Signaling Pathway/physiology , raf Kinases/physiology
6.
J Hepatol ; 63(3): 651-60, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25865556

ABSTRACT

BACKGROUND & AIMS: We have previously reported that Shp2, a tyrosine phosphatase previously known as a pro-leukemogenic molecule, suppresses the initiation of hepatocellular carcinoma (HCC). However, the role of Shp2 in HCC progression remains obscure. METHODS: Shp2 expression was determined in human HCC using real-time PCR, immunoblotting and immunohistochemistry. Clinical significance of Shp2 expression was analyzed in 301 HCC tissues with clinico-pathological characteristics and follow-up information. Short hairpin RNA was utilized to investigate the function of Shp2 in hepatoma cell behavior. Role of Shp2 in HCC progression was monitored through nude mice xenograft assay. Kinase activity assay and co-immunoprecipitation were used for mechanism analysis. RESULTS: Elevated expression of Shp2 was detected in 65.9% (394/598) of human HCCs, and its levels were even higher in metastasized foci. Overexpression of Shp2 correlated well with the malignant clinico-pathological characteristics of HCC and predicted the poor prognosis of patients. Interference of Shp2 expression suppressed the proliferation of hepatoma cells in vitro and inhibited the growth of HCC xenografts in vivo. Down-regulation of Shp2 attenuated the adhesion and migration of hepatoma cells and diminished metastasized HCC formation in mice. Our data demonstrated that Shp2 promotes HCC growth and metastasis by coordinately activating Ras/Raf/Erk pathway and PI3-K/Akt/mTOR cascade. Moreover, down-regulation of Shp2 enhanced the sensitivity of hepatoma cells upon sorafenib treatment, and patients with low Shp2 expression exhibited superior prognosis to sorafenib. CONCLUSIONS: Shp2 promotes the progression of HCC and may serve as a prognostic biomarker for patients.


Subject(s)
Carcinoma, Hepatocellular/etiology , Liver Neoplasms/etiology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , Animals , Carcinoma, Hepatocellular/mortality , Cell Line, Tumor , Cell Proliferation , Disease Progression , Humans , Liver Neoplasms/mortality , MAP Kinase Signaling System , Mice , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Phenylurea Compounds/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Prognosis , Sorafenib , raf Kinases/physiology
7.
Br J Cancer ; 110(10): 2479-88, 2014 May 13.
Article in English | MEDLINE | ID: mdl-24762959

ABSTRACT

BACKGROUND: Mesothelioma is a notoriously chemotherapy-resistant neoplasm, as is evident in the dismal overall survival for patients with those of asbestos-associated disease. We previously demonstrated co-activation of multiple receptor tyrosine kinases (RTKs), including epidermal growth factor receptor (EGFR), MET, and AXL in mesothelioma cell lines, suggesting that these kinases could serve as novel therapeutic targets. Although clinical trials have not shown activity for EGFR inhibitors in mesothelioma, concurrent inhibition of various activated RTKs has pro-apoptotic and anti-proliferative effects in mesothelioma cell lines. Thus, we hypothesised that a coordinated network of multi-RTK activation contributes to mesothelioma tumorigenesis. METHODS: Activation of PI3K/AKT/mTOR, Raf/MAPK, and co-activation of RTKs were evaluated in mesotheliomas. Effects of RTK and downstream inhibitors/shRNAs were assessed by measuring mesothelioma cell viability/growth, apoptosis, activation of signalling intermediates, expression of cell-cycle checkpoints, and cell-cycle alterations. RESULTS: We demonstrate activation of the PI3K/AKT/p70S6K and RAF/MEK/MAPK pathways in mesothelioma, but not in non-neoplastic mesothelial cells. The AKT activation, but not MAPK activation, was dependent on coordinated activation of RTKs EGFR, MET, and AXL. In addition, PI3K/AKT/mTOR pathway inhibition recapitulated the anti-proliferative effects of concurrent inhibition of EGFR, MET, and AXL. Dual targeting of PI3K/mTOR by BEZ235 or a combination of RAD001 and AKT knockdown had a greater effect on mesothelioma proliferation and viability than inhibition of individual activated RTKs or downstream signalling intermediates. Inhibition of PI3K/AKT was also associated with MDM2-p53 cell-cycle regulation. CONCLUSIONS: These findings show that PI3K/AKT/mTOR is a crucial survival pathway downstream of multiple activated RTKs in mesothelioma, underscoring that PI3K/mTOR is a compelling target for therapeutic intervention.


Subject(s)
Antineoplastic Agents/pharmacology , Mesothelioma/enzymology , Neoplasm Proteins/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Butadienes/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Chromones/pharmacology , Drug Screening Assays, Antitumor , Enzyme Activation/drug effects , Everolimus , Humans , Imidazoles/pharmacology , Indazoles/pharmacology , MAP Kinase Signaling System , Mesothelioma/pathology , Molecular Targeted Therapy , Morpholines/pharmacology , Neoplasm Proteins/physiology , Nitriles/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Quinolines/pharmacology , RNA Interference , RNA, Small Interfering/pharmacology , Receptor Protein-Tyrosine Kinases/physiology , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Sulfonamides/pharmacology , TOR Serine-Threonine Kinases/physiology , raf Kinases/physiology
8.
Cancer Cell ; 10(6): 459-72, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17157787

ABSTRACT

Oncogene-induced senescence functions to limit tumor development. However, a complete understanding of the signals that trigger this type of senescence is currently lacking. We found that mutations affecting NF1, Raf, and Ras induce a global negative feedback response that potently suppresses Ras and/or its effectors. Moreover, these signals promote senescence by inhibiting the Ras/PI3K pathway, which can impact the senescence machinery through HDM2 and FOXO. This negative feedback program is regulated in part by RasGEFs, Sprouty proteins, RasGAPs, and MKPs. Moreover, these signals function in vivo in benign human tumors. Thus, the ultimate response to the aberrant activation of the Ras pathway is a multifaceted negative feedback signaling network that terminates the oncogenic signal and participates in the senescence response.


Subject(s)
Cellular Senescence , Genes, ras/physiology , Signal Transduction/physiology , Animals , Cells, Cultured , Feedback , Genes, Neurofibromatosis 1/physiology , Genes, Retinoblastoma/physiology , Genes, p53/physiology , Humans , Mice , Neoplasms/genetics , Neoplasms/pathology , Phosphatidylinositol 3-Kinases/physiology , Stem Cells/pathology , raf Kinases/physiology
9.
Cancer Cell ; 9(1): 9-12, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16413467

ABSTRACT

RAF research is booming since the discovery of mutant B-RAF in approximately 8% of human cancer. One reason for the excitement is the availability of RAF-targeted therapies. RAF inhibitors have been developed because RAF functions at a convergence point of signal transduction. Two recent papers by the groups of Rosen and Marais dramatically advance our understanding of RAF oncogenes in human tumors. The results confirm that the mitogenic cascade (RAF-MEK-ERK) is essential for RAF transformation, that RAF kinases work in concert, and that RAF-transformed cells are hooked on MEK, making them sensitive to growth inhibition by kinase inhibitors.


Subject(s)
Mitogen-Activated Protein Kinases/physiology , Signal Transduction/physiology , raf Kinases/physiology , Animals , Cell Proliferation/drug effects , Enzyme Activation , Genes, ras , Humans , Mutation , raf Kinases/antagonists & inhibitors , raf Kinases/genetics
10.
Am J Pathol ; 180(6): 2462-78, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22546478

ABSTRACT

Lesions displaying a variety of dysplastic changes precede invasive oral and epidermal squamous cell carcinoma (SCC); however, there are no histopathological criteria for either confirming or staging premalignancy. SCCs and dysplasias frequently contain cells that abnormally express the γ2 subunit of laminin-332. We developed cell culture models to investigate γ2 dysregulation. Normal human keratinocytes displayed density-dependent repression of γ2, whereas premalignant keratinocytes and SCC cells overexpressed γ2 and secreted laminin assembly intermediates. Neoplastic cells had hyperactive EGFR/MAPK(ERK) signaling coordinate with overexpressed γ2, and EGFR and MEK inhibitors normalized γ2 expression. Keratinocytes engineered to express HPV16 E6 or activated mutant HRAS, cRAF1, or MEK1 lost density repression of γ2 and shared with neoplastic cells signaling abnormalities downstream of ERK, including increased phosphorylation of S6 and eIF4 translation factors. Notably, qPCR results revealed that γ2 overexpression was not accompanied by increased γ2 mRNA levels, consistent with ERK-dependent, eIF4B-mediated translation initiation of the stem-looped, 5'-untranslated region of γ2 mRNA in neoplastic cells. Inhibitors of MEK, but not of TORC1/2, blocked S6 and eIF4B phosphorylation and γ2 overexpression. Immunostaining of oral dysplasias identified γ2 overexpression occurring within fields of basal cells that had elevated p-S6 levels. These results reveal a causal relationship between ERK-dependent translation factor activation and laminin γ2 dysregulation and identify new markers of preinvasive neoplastic change during progression to SCC.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Laminin/biosynthesis , Mitogen-Activated Protein Kinase Kinases/physiology , Mouth Neoplasms/metabolism , Precancerous Conditions/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Disease Progression , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic/physiology , Humans , Keratinocytes/metabolism , Laminin/genetics , MAP Kinase Signaling System/physiology , Mouth Neoplasms/enzymology , Mouth Neoplasms/pathology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Precancerous Conditions/enzymology , Precancerous Conditions/pathology , Protein Modification, Translational/physiology , Signal Transduction/physiology , Tumor Cells, Cultured , raf Kinases/physiology , ras Proteins/physiology
11.
Carcinogenesis ; 33(12): 2467-76, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23027619

ABSTRACT

Interleukin-11 (IL-11), which belongs to a class of IL6-type cytokines, plays an important role in inflammation, motility and invasion in cancer. The ras mutation is frequently found in human cancer, but little is known regarding the transcriptional activation of the IL-11 gene by the Ras signal pathway in tumour cells. In this study, we investigated the role of Ras in the regulation of IL-11 using two different cell model systems: mouse NIH3T3 cells over-expressing oncogenic Ras with a tet-on system and Capan-1 human pancreatic carcinoma cells harbouring a K-ras mutation. We found that IL-11 expression was up-regulated at the transcriptional level by oncogenic Ras. Activation of the AP-1 response element, located between -153 and -30 in the 5'-regulatory region of the IL-11 gene, was necessary for oncogenic Ras-induced IL-11 promoter activation. AP-1 proteins, including Fra-1 and Fra-2, were up-regulated through the Raf/MEK and phosphatidylinositol 3-kinase (PI3K)/Akt pathways by oncogenic Ras. Knockdown of Fra-1 by siRNA in NIH3T3 or Capan-1 cells strongly attenuated oncogenic Ras-induced IL-11 expression. Additionally, inhibition of JNK, p38 and Stat3 abrogated oncogenic Ras-induced IL-11 expression. These results suggest that both the PI3K and Raf pathways are necessary for the expression of IL-11 in oncogenic Ras-mutated cells, and that JNK, p38 and Stat3 also contribute to oncogenic Ras-induced IL-11 expression.


Subject(s)
Gene Expression Regulation , Genes, ras/physiology , Interleukin-11/genetics , Animals , HEK293 Cells , Humans , Mice , Mutation , NIH 3T3 Cells , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/physiology , Promoter Regions, Genetic , Proto-Oncogene Proteins c-fos/physiology , Transcription Factor AP-1/physiology , Transcription, Genetic , raf Kinases/physiology
12.
Br J Cancer ; 104(2): 229-34, 2011 Jan 18.
Article in English | MEDLINE | ID: mdl-21081934

ABSTRACT

The epidermis is the outermost layer of the body and protects it from environmental insults. This crucial function is sustained by a continuous process of self-renewal involving the carefully balanced proliferation and differentiation of progenitor cells constantly replacing the mature cells at the surface of the epidermis. Genetic changes in the signalling pathways controlling keratinocyte proliferation and differentiation disrupt this balance and lead to pathological changes including carcinogenesis. This review discusses the role of Ras, an oncogene critically involved in the development of skin neoplasia, and its downstream effector Raf in epidermal homeostasis and tumourigenesis. In particular, we will focus on the recently established role of Raf-1 as the decisive element that, by restraining keratinocyte differentiation, allows the development and maintenance of Ras-driven tumours.


Subject(s)
Cell Transformation, Neoplastic , Epidermis/growth & development , Proto-Oncogene Proteins p21(ras)/physiology , raf Kinases/physiology , Animals , Humans
13.
BMC Neurosci ; 12: 109, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22044770

ABSTRACT

BACKGROUND: Cigarette smoking enhances the risk of stroke. However, the underlying molecular mechanisms are largely unknown. The present study established an in vivo rat secondhand cigarette smoking (SHS) model and examined the hypothesis that SHS upregulates endothelin receptors with increased cerebrovascular contraction via the Raf/extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinases (MAPK) pathway. RESULTS: Rats were exposed to SHS for up to 8 weeks. The cerebral artery vasoconstriction was recorded by a sensitive myograph. The mRNA and protein expressions for endothelin receptors in cerebral arteries were studied by real-time PCR and Western blot. Compared to fresh air exposed rats, cerebral arteries from SHS rats exhibited stronger contractile responses (P < 0.05) mediated by endothelin type A (ETA) receptors. The expressions of mRNA and protein for ETA receptors in the cerebral arteries from SHS rats were higher (P < 0.05) than that in control. SHS did not affect endothelin type B (ETB) receptor-mediated contractions, mRNA or protein levels. The results suggest that SHS upregulates ETA, but not ETB receptors in vivo. After SHS exposure, the mRNA levels of Raf-1 and ERK1/2, the protein expression of phosphorylated (p)-Raf-1 and p-ERK1/2 were increased (P < 0.05). Raf-1 inhibitor, GW5074 suppressed the enhanced ETA receptor-mediated contraction, mRNA and protein levels induced by SHS. In addition, GW5074 inhibited the SHS-caused increased mRNA and phosphorylated protein levels of Raf-1 and ERK1/2, suggesting that SHS induces activation of the Raf/ERK/MAPK pathway. CONCLUSIONS: SHS upregulates cerebrovascular ETA receptors via the Raf/ERK/MAPK pathway, which provides novel understanding of mechanisms involved in SHS-associated stroke.


Subject(s)
Cerebrovascular Circulation/physiology , MAP Kinase Signaling System/physiology , Receptor, Endothelin A/biosynthesis , Stroke/enzymology , Stroke/etiology , Tobacco Smoke Pollution/adverse effects , Up-Regulation/physiology , raf Kinases/physiology , Animals , Male , Rats , Rats, Sprague-Dawley , Receptor, Endothelin A/genetics , Stroke/metabolism , raf Kinases/antagonists & inhibitors , raf Kinases/genetics
14.
BMC Neurosci ; 12: 107, 2011 Oct 27.
Article in English | MEDLINE | ID: mdl-22032648

ABSTRACT

BACKGROUND: Late cerebral ischemia carries high morbidity and mortality after subarachnoid hemorrhage (SAH) due to reduced cerebral blood flow (CBF) and the subsequent cerebral ischemia which is associated with upregulation of contractile receptors in the vascular smooth muscle cells (SMC) via activation of mitogen-activated protein kinase (MAPK) of the extracellular signal-regulated kinase (ERK)1/2 signal pathway. We hypothesize that SAH initiates cerebrovascular ERK1/2 activation, resulting in receptor upregulation. The raf inhibitor will inhibit the molecular events upstream ERK1/2 and may provide a therapeutic window for treatment of cerebral ischemia after SAH. RESULTS: Here we demonstrate that SAH increases the phosphorylation level of ERK1/2 in cerebral vessels and reduces the neurology score in rats in additional with the CBF measured by an autoradiographic method. The intracisternal administration of SB-386023-b, a specific inhibitor of raf, given 6 h after SAH, aborts the receptor changes and protects the brain from the development of late cerebral ischemia at 48 h. This is accompanied by reduced phosphorylation of ERK1/2 in cerebrovascular SMC. SAH per se enhances contractile responses to endothelin-1 (ET-1), 5-carboxamidotryptamine (5-CT) and angiotensin II (Ang II), upregulates ETB, 5-HT1B and AT1 receptor mRNA and protein levels. Treatment with SB-386023-b given as late as at 6 h but not at 12 h after the SAH significantly decreased the receptor upregulation, the reduction in CBF and the neurology score. CONCLUSION: These results provide evidence for a role of the ERK1/2 pathway in regulation of expression of cerebrovascular SMC receptors. It is suggested that raf inhibition may reduce late cerebral ischemia after SAH and provides a realistic time window for therapy.


Subject(s)
Cerebrovascular Circulation/drug effects , Protein Kinase Inhibitors/pharmacology , Subarachnoid Hemorrhage/drug therapy , Subarachnoid Hemorrhage/enzymology , Vasospasm, Intracranial/drug therapy , Vasospasm, Intracranial/enzymology , raf Kinases/antagonists & inhibitors , raf Kinases/metabolism , Animals , Cerebrovascular Circulation/physiology , Disease Models, Animal , Enzyme Activation/drug effects , Enzyme Activation/physiology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Protein Kinase Inhibitors/therapeutic use , Rats , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/physiopathology , Vasospasm, Intracranial/physiopathology , raf Kinases/physiology
15.
Curr Top Microbiol Immunol ; 346: 143-69, 2010.
Article in English | MEDLINE | ID: mdl-20563706

ABSTRACT

Ras proteins are key regulators of signalling cascades, controlling many processes such as proliferation, differentiation and apoptosis. Mutations in these proteins or in their effectors, activators and regulators are associated with pathological conditions, particularly the development of various forms of human cancer. RAS proteins signal through direct interaction with a number of effector enzymes, one of the best characterized being type I phosphatidylinositol (PI) 3-kinases. Although the ability of RAS to control PI 3-kinase has long been well established in cultured cells, evidence for a role of the interaction of endogenous RAS with PI 3-kinase in normal and malignant cell growth in vivo has only been obtained recently. Mice with mutations in the PI 3-kinase catalytic p110a isoform that block its ability to interact with RAS are highly resistant to endogenous KRAS oncogene induced lung tumourigenesis and HRAS oncogene induced skin carcinogenesis. Cells from these mice show proliferative defects and selective disruption of signalling from certain growth factors to PI 3-kinase, while the mice also display delayed development of the lymphatic vasculature. The interaction of RAS with p110a is thus required in vivo for some normal growth factor signalling and also for RAS-driven tumour formation. RAS family members were among the first oncogenes identified over 40 years ago. In the late 1960s, the rat-derived Harvey and Kirsten murine sarcoma retroviruses were discovered and subsequently shown to promote cancer formation through related oncogenes, termed RAS (from rat sarcoma virus). The central role of RAS proteins in human cancer is highlighted by the large number of tumours in which they are activated by mutation: approximately 20% of human cancers carry a mutation in RAS proteins. Because of the complex signalling network in which RAS operates, with multiple activators and effectors, each with a different pattern of tissue-specific expression and a distinct set of intracellular functions, one of the critical issues concerns the specific role of each effector in RAS-driven oncogenesis. In this chapter, we summarize current knowledge about how RAS regulates one of its best-known effectors, phosphoinositide 3-kinase (PI3K).


Subject(s)
Phosphatidylinositol 3-Kinases/physiology , Renin-Angiotensin System/physiology , Animals , Humans , Neoplasms/etiology , Signal Transduction , raf Kinases/physiology
16.
Nat Neurosci ; 10(5): 598-607, 2007 May.
Article in English | MEDLINE | ID: mdl-17396120

ABSTRACT

To define the role of the Raf serine/threonine kinases in nervous system development, we conditionally targeted B-Raf and C-Raf, two of the three known mammalian Raf homologs, using a mouse line expressing Cre recombinase driven by a nestin promoter. Targeting of B-Raf, but not C-Raf, markedly attenuated baseline phosphorylation of Erk in neural tissues and led to growth retardation. Conditional elimination of B-Raf in dorsal root ganglion (DRG) neurons did not interfere with survival, but instead caused marked reduction in expression of the glial cell line-derived neurotrophic factor receptor Ret at postnatal stages, associated with a profound reduction in levels of transcription factor CBF-beta. Elimination of both alleles of Braf, which encodes B-Raf, and one allele of Raf1, which encodes C-Raf, affected DRG neuron maturation as well as proprioceptive axon projection toward the ventral horn in the spinal cord. Finally, conditional elimination of all Braf and Raf1 alleles strongly reduced neurotrophin-dependent axon growth in vitro as well as cutaneous axon terminal arborization in vivo. We conclude that Raf function is crucial for several aspects of DRG neuron development, including differentiation and axon growth.


Subject(s)
Axons/physiology , Cell Differentiation/physiology , Neurons, Afferent/cytology , Neurons, Afferent/physiology , Signal Transduction/physiology , raf Kinases/physiology , Animals , Cell Differentiation/drug effects , Cell Survival , Cells, Cultured , Embryo, Mammalian , Exons , Ganglia, Spinal/cytology , Gene Expression/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/physiology , Nerve Growth Factor/pharmacology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons, Afferent/drug effects , Rats , Signal Transduction/genetics , Stem Cells/drug effects , Transfection , raf Kinases/genetics
17.
Carcinogenesis ; 31(7): 1165-74, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20047953

ABSTRACT

Some 25 years ago, Raf was discovered as the transforming principle shared by a murine sarcoma and an avian carcinoma virus. Thus, Raf and tumorigenesis have been connected from the very beginning. Ten years later, the work of many groups instated Raf as the link between Ras, the oncogene most frequently mutated in human cancers, and the mitogen-activated protein kinase/extracellular signal-regulated kinase kinase (MEK/ERK) module, which with its manifold substrates can contribute to different aspects of carcinogenesis. Finally, the discovery of activating B-Raf mutations in a subset of human cancers, notably melanomas, conclusively established Raf as a major player in tumor development. Recent studies in animal models now show that endogenous C-Raf is essential for the development and maintenance of Ras-induced epidermal tumors. Surprisingly, the role of C-Raf in this case is not that of an mitogen-activated protein kinase activator, but rather that of an endogenous inhibitor of Rho signaling, expanding the range of tumor-related Raf targets. This review focuses on old and new targets of Raf in tumorigenesis.


Subject(s)
Neoplasms/etiology , raf Kinases/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , Genes, ras , Humans , MAP Kinase Kinase Kinase 5/physiology , Mitogen-Activated Protein Kinase Kinases/physiology , Proto-Oncogene Proteins B-raf/physiology , Proto-Oncogene Proteins c-raf/physiology , Signal Transduction
18.
Immunopharmacol Immunotoxicol ; 32(1): 82-91, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19839729

ABSTRACT

BACKGROUND AND AIM: Pentoxifylline (PTX) has been proven to be an inhibitor of fMLP-induced neutrophil (PMN) oxidative burst and is thought to function by increasing cAMP and Protein kinase A (PKA). We hypothesized that PTX diminishes production of the neutrophil respiratory burst by both PKA-dependent and independent mechanisms. MATERIAL AND METHODS: Human neutrophils were isolated and stimulated with fMLP (1microM) alone or in combination with PTX (2mM). PMN activation was determined by the cytochrome C reduction method in the presence and absence of p38 MAPK (SB203580), ERK (PD98059), and PKA inhibitors (H89). Western blot analysis of Ras, Raf, p38 MAPK, ERK, and Akt was performed in PMNs exposed to fMLP and PTX. Cell membranes were fractionated to measure membrane-associated p47 phox. Treated cells were imaged using confocal microscopy to examine changes in localization of Akt and p47phox. RESULTS: PTX produced a decrease in oxidative burst that was diminished but not abrogated by H89 exposure. The reduction in Ras, Raf, and Akt activation seen with PTX was not effected by the presence of H89. The ability of PTX to attenuate phosphorylation of p38 MAPK and ERK was significantly decreased in the presence of H89, suggesting a PKA-dependent mechanisms. Membrane fractions of neutrophils demonstrate that PTX decreased membrane-associated p47phox, thus diminishing the ability to generate oxidative burst. PTX also decreased membrane localization of Akt and p47phox by confocal microscopy. CONCLUSIONS: PTX attenuates activation of signaling molecules involved in activation of p47phox and suppress the subsequent assembly of the NADPH machinery through both PKA-dependent and PKA-independent mechanisms.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , NADPH Oxidases/metabolism , Neutrophils/drug effects , Pentoxifylline/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Respiratory Burst/drug effects , Down-Regulation , Humans , MAP Kinase Signaling System/drug effects , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/metabolism , Proto-Oncogene Proteins c-akt/metabolism , raf Kinases/physiology , ras Proteins/physiology
19.
J Hematol Oncol ; 13(1): 113, 2020 08 17.
Article in English | MEDLINE | ID: mdl-32807225

ABSTRACT

Cancer is characterized as a complex disease caused by coordinated alterations of multiple signaling pathways. The Ras/RAF/MEK/ERK (MAPK) signaling is one of the best-defined pathways in cancer biology, and its hyperactivation is responsible for over 40% human cancer cases. To drive carcinogenesis, this signaling promotes cellular overgrowth by turning on proliferative genes, and simultaneously enables cells to overcome metabolic stress by inhibiting AMPK signaling, a key singular node of cellular metabolism. Recent studies have shown that AMPK signaling can also reversibly regulate hyperactive MAPK signaling in cancer cells by phosphorylating its key components, RAF/KSR family kinases, which affects not only carcinogenesis but also the outcomes of targeted cancer therapies against the MAPK signaling. In this review, we will summarize the current proceedings of how MAPK-AMPK signalings interplay with each other in cancer biology, as well as its implications in clinic cancer treatment with MAPK inhibition and AMPK modulators, and discuss the exploitation of combinatory therapies targeting both MAPK and AMPK as a novel therapeutic intervention.


Subject(s)
Adenylate Kinase/physiology , MAP Kinase Signaling System/physiology , Molecular Targeted Therapy , Neoplasm Proteins/physiology , Neoplasms/enzymology , Amino Acids/metabolism , Antineoplastic Agents/therapeutic use , Autophagy , Cell Differentiation/physiology , Cell Division/physiology , Clinical Trials as Topic , Drug Synergism , Energy Metabolism , Enzyme Activation , Homeostasis , Humans , MAP Kinase Signaling System/drug effects , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Phosphorylation , Protein Kinase Inhibitors/therapeutic use , Protein Processing, Post-Translational , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/physiology , Tumor Suppressor Proteins/physiology , raf Kinases/antagonists & inhibitors , raf Kinases/genetics , raf Kinases/physiology
20.
Cancer Sci ; 100(1): 117-25, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19037990

ABSTRACT

The extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK-MAPK) is critical in human malignancies. It remained to be established whether DNA methyltransferases (Dnmt) and proliferating cell nuclear antigen (PCNA) involved in DNA methylation during RAF-transformed cell proliferation. The plasmid of constitutively active RAF was used to transfect gastric cell GES-1 and cancer cell AGS. RAF promoted cell proliferation, growth in soft agar and induced cell cycle progress faster than empty plasmid by accelerating G1/S transition in both cell lines, a massive induction of cyclin D1 and PCNA expression was observed, along with reduced expression of p16INK4A, p21WAF1 and p27KIP1. Methylation-specific polymerase chain reaction and bisulfite sequencing showed that the promoter of p16INK4A was methylated in RAF-transformed cells, treatment with 5-aza-dC or PD98059 restored the expression of p16INK4A, increased p21WAF1 and p27KIP1 partially, associated with upregulation of the activity of Dnmt in RAF-transformed cell GES-1, and also decreased the hypermethylation status of p16INK4A, but not all CpG islands of p21WAF1 and p27KIP1. These data suggest that RAF may induce cell proliferation through hypermethylation of tumor suppressor gene p16INK4A, while the epigenetic inactivation of p21WAF1 and p27KIP1 may be not a key factor in RAF-transformed cells.


Subject(s)
DNA Methylation , Gastric Mucosa/pathology , Genes, Tumor Suppressor , Promoter Regions, Genetic , Stomach Neoplasms/etiology , raf Kinases/physiology , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Extracellular Signal-Regulated MAP Kinases/physiology , Flavonoids/pharmacology , Gastric Mucosa/metabolism , Genes, p16 , Humans , Phosphorylation , Stomach Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL