Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cancer Res Commun ; 2(3): 131-145, 2022 03 10.
Article in English | MEDLINE | ID: mdl-36466034

ABSTRACT

Targeting the human epidermal growth factor receptor 2 (HER2) became a landmark in the treatment of HER2-driven breast cancer. Nonetheless, the clinical efficacy of anti-HER2 therapies can be short-lived and a significant proportion of patients ultimately develop metastatic disease and die. One striking consequence of oncogenic activation of HER2 in breast cancer cells is the constitutive activation of the extracellular-regulated protein kinase 5 (ERK5) through its hyperphosphorylation. In this study, we sought to decipher the significance of this unique molecular signature in promoting therapeutic resistance to anti-HER2 agents. We found that a small-molecule inhibitor of ERK5 suppressed the phosphorylation of the retinoblastoma protein (RB) in HER2 positive breast cancer cells. As a result, ERK5 inhibition enhanced the anti-proliferative activity of single-agent anti-HER2 therapy in resistant breast cancer cell lines by causing a G1 cell cycle arrest. Moreover, ERK5 knockdown restored the anti-tumor activity of the anti-HER2 agent lapatinib in human breast cancer xenografts. Taken together, these findings support the therapeutic potential of ERK5 inhibitors to improve the clinical benefit that patients receive from targeted HER2 therapies.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Humans , Female , Breast Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Protein Kinases/therapeutic use , Quinazolines/pharmacology , Cell Cycle
2.
J Med Chem ; 65(6): 4783-4797, 2022 03 24.
Article in English | MEDLINE | ID: mdl-35290041

ABSTRACT

A set of meta-substituted 3-arylisoquinolinones have been identified that show substantial cytotoxicity in breast, liver, lung and colon cancer cell lines; these are up to 700-fold more active than the corresponding para analogues. These compounds were initially proposed as inhibitors of N-ribosyl dihydronicotinamide (NRH): quinone oxidoreductase 2 (NQO2) but were found to be inactive against the enzyme. Instead, COMPARE analysis suggested that 6-fluoro-3-(meta-fluorophenyl)isoquinolin-1(2H)-one (4) could mimic colchicine and interact with microtubules, a recognized target for cancer therapy. Subsequent docking, molecular dynamics simulations, and free energy analysis further suggested that compound 4 bound well into the colchicine-binding pocket of tubulin. Indeed, 4 suppressed tubulin polymerization, caused G2/M cell cycle arrest, and induced apoptosis. Also, 4 inhibited the formation of endothelial cell capillary-like tubes and further disrupted the structure of preestablished tubes; the effects were not observed with para analogue 5. In accordance with this, the computed free energy of binding of 5 to tubulin was lower in magnitude than that for 4 and appeared to arise in part from the inability of the para substituent to occupy a tubulin subpocket, which is possible in the meta orientation. In conclusion, the antiproliferative potential of the novel 3-arylisoquinolinones is markedly influenced by a subtle change in the structure (meta versus para). The meta-substituted isoquinolinone 4 is a microtubule-destabilizing agent with potential tumor-selectivity and antiangiogenic and vascular disrupting features.


Subject(s)
Antineoplastic Agents , Tubulin , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation , Colchicine/metabolism , Drug Screening Assays, Antitumor , Microtubules , Molecular Structure , Structure-Activity Relationship , Tubulin/metabolism , Tubulin Modulators/chemistry
3.
Oncogene ; 40(23): 3929-3941, 2021 06.
Article in English | MEDLINE | ID: mdl-33981002

ABSTRACT

There is overwhelming clinical evidence that the extracellular-regulated protein kinase 5 (ERK5) is significantly dysregulated in human breast cancer. However, there is no definite understanding of the requirement of ERK5 in tumor growth and metastasis due to very limited characterization of the pathway in disease models. In this study, we report that a high level of ERK5 is a predictive marker of metastatic breast cancer. Mechanistically, our in vitro data revealed that ERK5 was critical for maintaining the invasive capability of triple-negative breast cancer (TNBC) cells through focal adhesion protein kinase (FAK) activation. Specifically, we found that phosphorylation of FAK at Tyr397 was controlled by a kinase-independent function of ERK5. Accordingly, silencing ERK5 in mammary tumor grafts impaired FAK phosphorylation at Tyr397 and suppressed TNBC cell metastasis to the lung without preventing tumor growth. Collectively, these results establish a functional relationship between ERK5 and FAK signaling in promoting malignancy. Thus, targeting the oncogenic ERK5-FAK axis represents a promising therapeutic strategy for breast cancer exhibiting aggressive clinical behavior.


Subject(s)
Focal Adhesion Kinase 1/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Triple Negative Breast Neoplasms/enzymology , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/biosynthesis , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion/physiology , Cell Line, Tumor , Disease Progression , Female , Heterografts , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 7/biosynthesis , Mitogen-Activated Protein Kinase 7/genetics , Neoplasm Invasiveness , Phosphorylation , Prognosis , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
4.
Cancers (Basel) ; 13(5)2021 Feb 24.
Article in English | MEDLINE | ID: mdl-33668183

ABSTRACT

Brain tumours kill more children and adults under 40 than any other cancer, with approximately half of primary brain tumours being diagnosed as high-grade malignancies known as glioblastomas. Despite de-bulking surgery combined with chemo-/radiotherapy regimens, the mean survival for these patients is only around 15 months, with less than 10% surviving over 5 years. This dismal prognosis highlights the urgent need to develop novel agents to improve the treatment of these tumours. To address this need, we carried out a human kinome siRNA screen to identify potential drug targets that augment the effectiveness of temozolomide (TMZ)-the standard-of-care chemotherapeutic agent used to treat glioblastoma. From this we identified ERK5/MAPK7, which we subsequently validated using a range of siRNA and small molecule inhibitors within a panel of glioma cells. Mechanistically, we find that ERK5 promotes efficient repair of TMZ-induced DNA lesions to confer cell survival and clonogenic capacity. Finally, using several glioblastoma patient cohorts we provide target validation data for ERK5 as a novel drug target, revealing that heightened ERK5 expression at both the mRNA and protein level is associated with increased tumour grade and poorer patient survival. Collectively, these findings provide a foundation to develop clinically effective ERK5 targeting strategies in glioblastomas and establish much-needed enhancement of the therapeutic repertoire used to treat this currently incurable disease.

5.
Oncogene ; 39(33): 5553-5569, 2020 08.
Article in English | MEDLINE | ID: mdl-32655131

ABSTRACT

Metastasis is the leading cause of cancer-related death. This multistage process involves contribution from both tumour cells and the tumour stroma to release metastatic cells into the circulation. Circulating tumour cells (CTCs) survive circulatory cytotoxicity, extravasate and colonise secondary sites effecting metastatic outcome. Reprogramming the transcriptomic landscape is a metastatic hallmark, but detecting underlying master regulators that drive pathological gene expression is a key challenge, especially in childhood cancer. Here we used whole tumour plus single-cell RNA-sequencing in primary bone cancer and CTCs to perform weighted gene co-expression network analysis to systematically detect coordinated changes in metastatic transcript expression. This approach with comparisons applied to data collected from cell line models, clinical samples and xenograft mouse models revealed mitogen-activated protein kinase 7/matrix metallopeptidase 9 (MAPK7/MMP9) signalling as a driver for primary bone cancer metastasis. RNA interference knockdown of MAPK7 reduces proliferation, colony formation, migration, tumour growth, macrophage residency/polarisation and lung metastasis. Parallel to these observations were reduction of activated interleukins IL1B, IL6, IL8 plus mesenchymal markers VIM and VEGF in response to MAPK7 loss. Our results implicate a newly discovered, multidimensional MAPK7/MMP9 signalling hub in primary bone cancer metastasis that is clinically actionable.


Subject(s)
Bone Neoplasms/complications , Mitogen-Activated Protein Kinase 7/metabolism , Animals , Bone Neoplasms/genetics , Humans , Matrix Metalloproteinase 9/metabolism , Mice , Neoplasm Metastasis
6.
Cancer Res ; 80(16): 3319-3330, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32561530

ABSTRACT

The presence of immunosuppressive macrophages that become activated in the tumor microenvironment constitutes a major factor responsible for tumor growth and malignancy. In line with this knowledge, we report here that macrophage proliferation is a significant feature of advanced stages of cancer. Moreover, we have found that a high proportion of proliferating macrophages in human tumors express ERK5. ERK5 was required for supporting the proliferation of macrophages in tumor grafts in mice. Furthermore, myeloid ERK5 deficiency negatively impacted the proliferation of both resident and infiltrated macrophages in metastatic lung nodules. ERK5 maintained the capacity of macrophages to proliferate by suppressing p21 expression to halt their differentiation program. Collectively, these data provide insight into the mechanism underpinning macrophage proliferation to support malignant tumor development, thereby strengthening the value of ERK5-targeted therapies to restore antitumor immunity through the blockade of protumorigenic macrophage activation. SIGNIFICANCE: These findings offer a new rationale for anti-ERK5 therapy to improve cancer patient outcomes by blocking the proliferative activity of tumor macrophages.


Subject(s)
Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Tumor-Associated Macrophages/metabolism , Animals , Cell Differentiation , Humans , Ki-67 Antigen/analysis , Melanoma/secondary , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 7/antagonists & inhibitors , Mitogen-Activated Protein Kinase 7/deficiency , Tumor-Associated Macrophages/cytology
7.
Br J Cancer ; 122(1): 62-71, 2020 01.
Article in English | MEDLINE | ID: mdl-31819173

ABSTRACT

BACKGROUND: High expression of Ki67, a proliferation marker, is associated with reduced endometrial cancer-specific survival. Pre-surgical metformin reduces tumour Ki-67 expression in some women with endometrial cancer. Metformin's anti-cancer activity may relate to effects on cellular energy metabolism. Since tumour hypoxia and glucose availability are major cellular redox determinants, we evaluated their role in endometrial cancer response to metformin. METHODS: Endometrial cancer biopsies from women treated with pre-surgical metformin were tested for the hypoxia markers, HIF-1α and CA-9. Endometrial cancer cell lines were treated with metformin in variable glucose concentrations in normoxia or hypoxia and cell viability, mitochondrial biogenesis, function and energy metabolism were assessed. RESULTS: In women treated with metformin (n = 28), Ki-67 response was lower in hypoxic tumours. Metformin showed minimal cytostatic effects towards Ishikawa and HEC1A cells in conventional medium (25 mM glucose). In low glucose (5.5 mM), a dose-dependent cytostatic effect was observed in normoxia but attenuated in hypoxia. Tumours treated with metformin showed increased mitochondrial mass (n = 25), while in cultured cells metformin decreased mitochondrial function. Metformin targets mitochondrial respiration, however, in hypoxic, high glucose conditions, there was a switch to glycolytic metabolism and decreased metformin response. CONCLUSIONS: Understanding the metabolic adaptations of endometrial tumours may identify patients likely to derive clinical benefit from metformin.


Subject(s)
Cell Hypoxia/drug effects , Cytostatic Agents/therapeutic use , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/metabolism , Hyperglycemia/metabolism , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Carbonic Anhydrase IX/genetics , Carbonic Anhydrase IX/metabolism , Cell Hypoxia/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytostatic Agents/administration & dosage , Endometrial Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Glucose/metabolism , Humans , Hypoglycemic Agents/administration & dosage , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ki-67 Antigen/metabolism , Metformin/administration & dosage , Metformin/adverse effects , Mitochondria/drug effects , Mitochondria/metabolism , Preoperative Care/methods , Signal Transduction/drug effects , Treatment Outcome
8.
Proc Natl Acad Sci U S A ; 115(12): E2801-E2810, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29507229

ABSTRACT

Owing to the prevalence of tumor-associated macrophages (TAMs) in cancer and their unique influence upon disease progression and malignancy, macrophage-targeted interventions have attracted notable attention in cancer immunotherapy. However, tractable targets to reduce TAM activities remain very few and far between because the signaling mechanisms underpinning protumor macrophage phenotypes are largely unknown. Here, we have investigated the role of the extracellular-regulated protein kinase 5 (ERK5) as a determinant of macrophage polarity. We report that the growth of carcinoma grafts was halted in myeloid ERK5-deficient mice. Coincidentally, targeting ERK5 in macrophages induced a transcriptional switch in favor of proinflammatory mediators. Further molecular analyses demonstrated that activation of the signal transducer and activator of transcription 3 (STAT3) via Tyr705 phosphorylation was impaired in erk5-deleted TAMs. Our study thus suggests that blocking ERK5 constitutes a treatment strategy to reprogram macrophages toward an antitumor state by inhibiting STAT3-induced gene expression.


Subject(s)
Macrophages/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Neoplasms/metabolism , Neoplasms/pathology , STAT3 Transcription Factor/metabolism , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Cell Polarity , Humans , Macrophages/pathology , Mice, Inbred C57BL , Mice, Transgenic , Mitogen-Activated Protein Kinase 7/genetics , Phosphorylation , Receptors, Cell Surface/metabolism , STAT3 Transcription Factor/genetics , Tyrosine/metabolism , Xenograft Model Antitumor Assays
9.
Cancer Res ; 76(4): 787-95, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26659574

ABSTRACT

There is a clinical need for noninvasive biomarkers of tumor hypoxia for prognostic and predictive studies, radiotherapy planning, and therapy monitoring. Oxygen-enhanced MRI (OE-MRI) is an emerging imaging technique for quantifying the spatial distribution and extent of tumor oxygen delivery in vivo. In OE-MRI, the longitudinal relaxation rate of protons (ΔR1) changes in proportion to the concentration of molecular oxygen dissolved in plasma or interstitial tissue fluid. Therefore, well-oxygenated tissues show positive ΔR1. We hypothesized that the fraction of tumor tissue refractory to oxygen challenge (lack of positive ΔR1, termed "Oxy-R fraction") would be a robust biomarker of hypoxia in models with varying vascular and hypoxic features. Here, we demonstrate that OE-MRI signals are accurate, precise, and sensitive to changes in tumor pO2 in highly vascular 786-0 renal cancer xenografts. Furthermore, we show that Oxy-R fraction can quantify the hypoxic fraction in multiple models with differing hypoxic and vascular phenotypes, when used in combination with measurements of tumor perfusion. Finally, Oxy-R fraction can detect dynamic changes in hypoxia induced by the vasomodulator agent hydralazine. In contrast, more conventional biomarkers of hypoxia (derived from blood oxygenation-level dependent MRI and dynamic contrast-enhanced MRI) did not relate to tumor hypoxia consistently. Our results show that the Oxy-R fraction accurately quantifies tumor hypoxia noninvasively and is immediately translatable to the clinic.


Subject(s)
Magnetic Resonance Imaging/methods , Neoplasms/diagnostic imaging , Oxygen/chemistry , Cell Hypoxia , Humans , Prognosis , Radiography
10.
Cancer Res ; 75(4): 742-53, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25649771

ABSTRACT

Chronic inflammation is a hallmark of many cancers, yet the pathogenic mechanisms that distinguish cancer-associated inflammation from benign persistent inflammation are still mainly unclear. Here, we report that the protein kinase ERK5 controls the expression of a specific subset of inflammatory mediators in the mouse epidermis, which triggers the recruitment of inflammatory cells needed to support skin carcinogenesis. Accordingly, inactivation of ERK5 in keratinocytes prevents inflammation-driven tumorigenesis in this model. In addition, we found that anti-ERK5 therapy cooperates synergistically with existing antimitotic regimens, enabling efficacy of subtherapeutic doses. Collectively, our findings identified ERK5 as a mediator of cancer-associated inflammation in the setting of epidermal carcinogenesis. Considering that ERK5 is expressed in almost all tumor types, our findings suggest that targeting tumor-associated inflammation via anti-ERK5 therapy may have broad implications for the treatment of human tumors.


Subject(s)
Carcinogenesis/genetics , Inflammation/genetics , Mitogen-Activated Protein Kinase 7/biosynthesis , Skin Neoplasms/genetics , Animals , Carcinogens/toxicity , Epidermis/metabolism , Epidermis/pathology , Gene Expression Regulation, Neoplastic , Humans , Inflammation/chemically induced , Inflammation/complications , Inflammation/pathology , Keratinocytes/metabolism , Keratinocytes/pathology , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 7/genetics , Skin Neoplasms/chemically induced , Skin Neoplasms/etiology , Skin Neoplasms/pathology
11.
Cancer Res ; 74(12): 3344-56, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24713432

ABSTRACT

The c-Jun N-terminal protein kinase (JNK) and its two direct activators, namely the mitogen-activated protein kinase (MAPK) kinase 4 (MKK4) and MKK7, constitute a signaling node frequently mutated in human pancreatic ductal adenocarcinoma (PDAC). Here we demonstrate the cooperative interaction of endogenous expression of Kras(G12D) with loss-of-function mutations in mkk4 or both, mkk4 and mkk7 genes in the pancreas. More specifically, impaired JNK signaling in a subpopulation of Pdx1-expressing cells dramatically accelerated the appearance of Kras(G12D)-induced acinar-to-ductal metaplasia and pancreatic intraepithelial neoplasias, which rapidly progressed to invasive PDAC within 10 weeks of age. Furthermore, inactivation of mkk4/mkk7 compromised acinar regeneration following acute inflammatory stress by locking damaged exocrine cells in a permanently de-differentiated state. Therefore, we propose that JNK signaling exerts its tumor suppressive function in the pancreas by antagonizing the metaplastic conversion of acinar cells toward a ductal fate capable of responding to oncogenic stimulation.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 7/genetics , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Acinar Cells/enzymology , Animals , Carcinogenesis/metabolism , Carcinoma, Pancreatic Ductal/genetics , Cell Dedifferentiation , MAP Kinase Kinase 4/metabolism , MAP Kinase Kinase 7/metabolism , MAP Kinase Signaling System , Mice , Mice, Transgenic , Mutation, Missense , Pancreas/enzymology , Pancreas/pathology , Pancreas/physiopathology , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Regeneration
12.
Cell Signal ; 24(12): 2360-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22917534

ABSTRACT

Activation of the extracellular-regulated protein kinase 5 (ERK5) has been associated with mitogenic signal transduction. However, conflicting findings have challenged the idea that ERK5 is a critical regulator of cell proliferation. We have addressed this issue by testing the effect of the conditional loss of ERK5 in primary fibroblasts. We have discovered that ERK5 suppressed the expression of the cyclin dependent protein kinase (CDKs) inhibitors, p21 and p27, by decreasing mRNA and protein stability, respectively. As a result, low level CDK2 activity detected in ERK5-deficient cells correlated with a defect in G1 to S phase transition of the cell cycle. Similarly, we found that the malignant MDA-MB-231 human breast cancer cell line was dependent on ERK5 to proliferate. We propose that ERK5 blocks p21 expression in MDA-MB-231 cells via a mechanism that implicates c-Myc-dependent transcriptional regulation of the miR-17-92 cluster. Together with evidence that cancer patients with poor prognosis display a high level of expression of components of the ERK5 signaling pathway, these findings support the hypothesis that ERK5 can be a potential target for cancer therapy.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Animals , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase 2/metabolism , Down-Regulation , Fibroblasts/cytology , Fibroblasts/metabolism , G1 Phase , Mice , MicroRNAs/metabolism , Proto-Oncogene Proteins c-myc/metabolism , S Phase , Transcription, Genetic
13.
Cancer Res ; 70(14): 5797-806, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20610622

ABSTRACT

The mitogen-activated protein kinase (MAPK) kinase 4 (MKK4) is a nonredundant component of stress-activated MAPK signaling modules. Its function in tumorigenesis remains highly controversial with some studies indicating that MKK4 is a tumor suppressor, whereas others have reported a pro-oncogenic role. To clarify the role of MKK4 in cancer, we have created a novel mouse model to test the effect of the specific loss of MKK4 in the epidermis on the formation of papillomas caused by activated ras mutation. We have discovered that skin-specific MKK4-deficient mice are resistant to carcinogen-induced tumorigenesis. One mechanism by which MKK4 promotes cell proliferation and the formation of tumors is by increasing epidermal growth factor receptor expression through the c-Jun NH(2)-terminal protein kinase/c-Jun signaling pathway. Together, our results provide the first genetic demonstration that MKK4 is essential to mediate the oncogenic effect of Ras in vivo, thereby validating MKK4 as a potential drug target for cancer therapy.


Subject(s)
Cell Transformation, Neoplastic/metabolism , MAP Kinase Kinase 4/metabolism , Skin Neoplasms/enzymology , 9,10-Dimethyl-1,2-benzanthracene , Animals , Carcinogens , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/genetics , Enzyme Activation , Female , Genes, ras , Inbreeding , MAP Kinase Kinase 4/deficiency , MAP Kinase Kinase 4/genetics , Male , Mice , Skin/enzymology , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics
14.
FEBS Lett ; 580(5): 1320-6, 2006 Feb 20.
Article in English | MEDLINE | ID: mdl-16458303

ABSTRACT

The signaling mechanism by which JNK affects mitochondria is critical to initiate apoptosis. Here we show that the absence of JNK provides a partial resistance to the toxic effect of the heavy metal cadmium. Both wild type and jnk-/- fibroblasts undergoing death exhibit cytosolic cytochrome c but, unlike wild type cells, the JNK-deficient fibroblasts do not display increased caspase activity and DNA fragmentation. The absence of apoptotic death correlates with a specific defect in activation of Bax. We conclude that JNK-dependent regulation of Bax is essential to mediate the apoptotic release of cytochrome c regardless of Bid and Bim activation.


Subject(s)
Apoptosis , JNK Mitogen-Activated Protein Kinases/metabolism , Mitochondria/physiology , Signal Transduction , bcl-2-Associated X Protein/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , BH3 Interacting Domain Death Agonist Protein/metabolism , Bcl-2-Like Protein 11 , Cadmium/toxicity , Cells, Cultured , Cytochromes c/metabolism , Enzyme Activation , Fibroblasts/cytology , JNK Mitogen-Activated Protein Kinases/deficiency , Membrane Proteins/metabolism , Mice , Mice, Knockout , Proto-Oncogene Proteins/metabolism , Signal Transduction/physiology
15.
Mol Cell Biol ; 25(1): 336-45, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15601854

ABSTRACT

To elucidate the physiological significance of MEK5 in vivo, we have examined the effect of mek5 gene elimination in mice. Heterozygous mice appear to be healthy and were fertile. However, mek5(-/-) embryos die at approximately embryonic day 10.5 (E10.5). The phenotype of the mek5(-/-) embryos includes abnormal cardiac development as well as a marked decrease in proliferation and an increase in apoptosis in the heart, head, and dorsal regions of the mutant embryos. The absence of MEK5 does not affect cell cycle progression but sensitizes mouse embryonic fibroblasts (MEFs) to the ability of sorbitol to enhance caspase 3 activity. Further studies with mek5(-/-) MEFs indicate that MEK5 is required for mediating extracellular signal-regulated kinase 5 (ERK5) activation and for the regulation of the transcriptional activity of myocyte enhancer factor 2. Overall, this is the first study to rigorously establish the role of MEK5 in vivo as an activator of ERK5 and as an essential regulator of cell survival that is required for normal embryonic development.


Subject(s)
DNA-Binding Proteins/genetics , MAP Kinase Kinase 5/genetics , MAP Kinase Kinase 5/physiology , Mitogen-Activated Protein Kinase 7/genetics , Transcription Factors/genetics , Animals , Apoptosis , Blotting, Southern , Caspase 3 , Caspases/metabolism , Cell Death , Cell Proliferation , Cell Survival , Cells, Cultured , Enzyme Activation , Fibroblasts/metabolism , Flow Cytometry , Gene Deletion , Genes, Reporter , Genetic Vectors , Genotype , Heterozygote , Immunoblotting , Immunohistochemistry , In Situ Nick-End Labeling , Luciferases/metabolism , MEF2 Transcription Factors , Mice , Mice, Knockout , Models, Genetic , Mutation , Myocardium/metabolism , Myogenic Regulatory Factors , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , Tissue Distribution , Transcription, Genetic , Transcriptional Activation , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL
...