Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cell Stem Cell ; 22(6): 879-892.e6, 2018 Jun 01.
Article in English | MEDLINE | ID: mdl-29804890

ABSTRACT

Hematopoietic stem cells (HSCs) sustain hematopoiesis throughout life. HSCs exit dormancy to restore hemostasis in response to stressful events, such as acute blood loss, and must return to a quiescent state to prevent their exhaustion and resulting bone marrow failure. HSC activation is driven in part through the phosphatidylinositol 3-kinase (PI3K)/AKT/mTORC1 signaling pathway, but less is known about the cell-intrinsic pathways that control HSC dormancy. Here, we delineate an ERK-dependent, rate-limiting feedback mechanism that controls HSC fitness and their re-entry into quiescence. We show that the MEK/ERK and PI3K pathways are synchronously activated in HSCs during emergency hematopoiesis and that feedback phosphorylation of MEK1 by activated ERK counterbalances AKT/mTORC1 activation. Genetic or chemical ablation of this feedback loop tilts the balance between HSC dormancy and activation, increasing differentiated cell output and accelerating HSC exhaustion. These results suggest that MEK inhibitors developed for cancer therapy may find additional utility in controlling HSC activation.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/enzymology , Animals , Cells, Cultured , Coculture Techniques , Female , Humans , MAP Kinase Kinase 1/deficiency , MAP Kinase Kinase 1/metabolism , Male , Membrane Potential, Mitochondrial , Mice , Reactive Oxygen Species/metabolism
3.
Mol Cell ; 50(1): 43-55, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23453810

ABSTRACT

The Raf/MEK/ERK and PI3K/Akt pathways are prominent effectors of oncogenic Ras. These pathways negatively regulate each other, but the mechanism involved is incompletely understood. We now identify MEK1 as an essential regulator of lipid/protein phosphatase PTEN, through which it controls phosphatidylinositol-3-phosphate accumulation and AKT signaling. MEK1 ablation stabilizes AKT activation and, in vivo, causes a lupus-like autoimmune disease and myeloproliferation. Mechanistically, MEK1 is necessary for PTEN membrane recruitment as part of a ternary complex containing the multidomain adaptor MAGI1. Complex formation is independent of MEK1 kinase activity but requires phosphorylation of T292 on MEK1 by activated ERK. Thus, inhibiting the ERK pathway reduces PTEN membrane recruitment, increasing phosphatidylinositol-3-phosphate accumulation and AKT activation. Our data offer a conceptual framework for the observation that activation of the PI3K pathway frequently mediate resistance to MEK inhibitors and for the promising results obtained by combined MEK/PI3K inhibition in preclinical cancer models.


Subject(s)
Cell Membrane/enzymology , Fibroblasts/enzymology , Lymphocytes/enzymology , MAP Kinase Kinase 1/metabolism , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Self Tolerance , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autoimmune Diseases/enzymology , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , COS Cells , Cell Adhesion Molecules , Cell Death , Cell Membrane/drug effects , Cell Membrane/immunology , Cell Membrane/pathology , Chlorocebus aethiops , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/immunology , Fibroblasts/pathology , Genotype , Guanylate Kinases , Lymphocyte Activation , Lymphocytes/immunology , Lymphocytes/pathology , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/deficiency , MAP Kinase Kinase 1/genetics , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Multienzyme Complexes , Myeloproliferative Disorders/enzymology , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/immunology , Phenotype , Phosphatidylinositol Phosphates/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Transport , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , RNA Interference , Signal Transduction , Time Factors , Transfection , Tyrosine
4.
Neuron ; 75(6): 940-2, 2012 Sep 20.
Article in English | MEDLINE | ID: mdl-22998861

ABSTRACT

In this issue of Neuron, Li et al. (2012) show that the neuron/glia cell fate switch of cortical progenitors is regulated by MEK1 and MEK2. The observations resonate with recent studies on the genesis of low-grade astrocytomas and highlight neuronal support functions of astrocytes in the postnatal brain.


Subject(s)
Brain , Gene Expression Regulation, Developmental/genetics , MAP Kinase Kinase 1/deficiency , MAP Kinase Kinase 2/deficiency , Neuroglia/physiology , Animals
5.
Neuron ; 75(6): 1035-50, 2012 Sep 20.
Article in English | MEDLINE | ID: mdl-22998872

ABSTRACT

We have defined functions of MEK in regulating gliogenesis in developing cerebral cortex using loss- and gain-of-function mouse genetics. Radial progenitors deficient in both Mek1 and Mek2 fail to transition to the gliogenic mode in late embryogenesis, and astrocyte and oligodendroglial precursors fail to appear. In exploring mechanisms, we found that the key cytokine-regulated gliogenic pathway is attenuated. Further, the Ets transcription family member Etv5/Erm is strongly regulated by MEK and Erm overexpression can rescue the gliogenic potential of Mek-deleted progenitors. Remarkably, Mek1/2-deleted mice surviving postnatally exhibit cortices almost devoid of astrocytes and oligodendroglia and exhibit neurodegeneration. Conversely, expression of constitutively active MEK1 leads to a major increase in numbers of astrocytes in the adult brain. We conclude that MEK is essential for acquisition of gliogenic competence by radial progenitors and that levels of MEK activity regulate gliogenesis in the developing cortex.


Subject(s)
Brain , Gene Expression Regulation, Developmental/genetics , MAP Kinase Kinase 1/deficiency , MAP Kinase Kinase 2/deficiency , Neuroglia/physiology , Age Factors , Animals , Animals, Newborn , Brain/cytology , Brain/embryology , Brain/growth & development , Cell Count , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Ciliary Neurotrophic Factor/pharmacology , DNA-Binding Proteins/metabolism , Electroporation , Embryo, Mammalian , Excitatory Amino Acid Transporter 1/metabolism , Eye Proteins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Ki-67 Antigen/metabolism , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 2/genetics , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , PAX6 Transcription Factor , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism , Signal Transduction/genetics , Stem Cells/drug effects , Stem Cells/physiology , Transcription Factors/metabolism
6.
Cancer Cell ; 19(5): 652-63, 2011 May 17.
Article in English | MEDLINE | ID: mdl-21514245

ABSTRACT

We have investigated the role of individual members of the Raf/Mek/Erk cascade in the onset of K-Ras oncogene-driven non-small cell lung carcinoma (NSCLC). Ablation of Erk1 or Erk2 in K-Ras oncogene-expressing lung cells had no significant effect due to compensatory activities. Yet, elimination of both Erk kinases completely blocked tumor development. Similar results were obtained with Mek kinases. Ablation of B-Raf had no significant effect on tumor development. However, c-Raf expression was absolutely essential for the onset of NSCLC. Interestingly, concomitant elimination of c-Raf and B-Raf in adult mice had no deleterious consequences for normal homeostasis. These results indicate that c-Raf plays a unique role in mediating K-Ras signaling and makes it a suitable target for therapeutic intervention.


Subject(s)
Carcinoma, Non-Small-Cell Lung/enzymology , Genes, ras , Lung Neoplasms/enzymology , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Animals , Apoptosis , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Cellular Senescence , Fibroblasts/enzymology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , MAP Kinase Kinase 1/deficiency , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 2/deficiency , MAP Kinase Kinase 2/genetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/deficiency , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/deficiency , Mitogen-Activated Protein Kinase 3/genetics , Proto-Oncogene Proteins B-raf/deficiency , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-raf/deficiency , Proto-Oncogene Proteins c-raf/genetics , RNA Interference , Signal Transduction , Time Factors , Transfection , Tumor Burden
7.
Cancer Res ; 69(9): 3772-8, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19383924

ABSTRACT

The Ras/Raf/Mek/Erk mitogen-activated protein kinase pathway regulates fundamental processes in normal and malignant cells, including proliferation, differentiation, and cell survival. Mutations in this pathway have been associated with carcinogenesis and developmental disorders, making Mek1 and Mek2 prime therapeutic targets. In this study, we examined the requirement for Mek1 and Mek2 in skin neoplasia using the two-step 7,12-dimethylbenz(a)anthracene/12-O-tetradecanoylphorbol-13-acetate (DMBA/TPA) skin carcinogenesis model. Mice lacking epidermal Mek1 protein develop fewer papillomas than both wild-type and Mek2-null mice following DMBA/TPA treatment. Mek1 knockout mice had smaller papillomas, delayed tumor onset, and half the tumor burden of wild-type mice. Loss of one Mek1 allele, however, did not affect tumor development, indicating that one Mek1 allele is sufficient for normal papilloma formation. No difference in TPA-induced hyperproliferation, inflammation, or Erk activation was observed between wild-type, conditional Mek1 knockout, and Mek2-null mice, indicating that Mek1 findings were not due to a general failure of these processes. These data show that Mek1 is important for skin tumor development and that Mek2 cannot compensate for the loss of Mek1 function in this setting.


Subject(s)
Cell Transformation, Neoplastic/metabolism , MAP Kinase Kinase 1/deficiency , MAP Kinase Kinase 2/deficiency , Skin Neoplasms/enzymology , 9,10-Dimethyl-1,2-benzanthracene , Animals , Cell Differentiation/physiology , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/genetics , Genotype , Hyperplasia , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 2/genetics , Mice , Mice, Knockout , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oncogene Protein v-akt/metabolism , Papilloma/chemically induced , Papilloma/enzymology , Papilloma/genetics , Phosphorylation , Skin/drug effects , Skin/enzymology , Skin/pathology , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Tetradecanoylphorbol Acetate
8.
Development ; 136(8): 1363-74, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19304888

ABSTRACT

The mammalian genome contains two ERK/MAP kinase kinase genes, Map2k1 and Map2k2, which encode dual-specificity kinases responsible for ERK/MAP kinase activation. In the mouse, loss of Map2k1 function causes embryonic lethality, whereas Map2k2 mutants survive with a normal lifespan, suggesting that Map2k1 masks the phenotype due to the Map2k2 mutation. To uncover the specific function of MAP2K2 and the threshold requirement of MAP2K proteins during embryo formation, we have successively ablated the Map2k gene functions. We report here that Map2k2 haploinsufficiency affects the normal development of placenta in the absence of one Map2k1 allele. Most Map2k1(+/-)Map2k2(+/-) embryos die during gestation because of placenta defects restricted to extra-embryonic tissues. The impaired viability of Map2k1(+/-)Map2k2(+/-) embryos can be rescued when the Map2k1 deletion is restricted to the embryonic tissues. The severity of the placenta phenotype is dependent on the number of Map2k mutant alleles, the deletion of the Map2k1 allele being more deleterious. Moreover, the deletion of one or both Map2k2 alleles in the context of one null Map2k1 allele leads to the formation of multinucleated trophoblast giant (MTG) cells. Genetic experiments indicate that these structures are derived from Gcm1-expressing syncytiotrophoblasts (SynT), which are affected in their ability to form the uniform SynT layer II lining the maternal sinuses. Thus, even though Map2k1 plays a predominant role, these results enlighten the function of Map2k2 in placenta development.


Subject(s)
MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Placentation , Trophoblasts/metabolism , Alleles , Animals , Enzyme Activation , Female , Gene Expression Regulation, Developmental , MAP Kinase Kinase 1/deficiency , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 2/deficiency , MAP Kinase Kinase 2/genetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Mutation/genetics , Phenotype , Placenta/embryology , Placenta/metabolism , Placentation/genetics , Pregnancy
9.
Am J Respir Cell Mol Biol ; 38(5): 618-26, 2008 May.
Article in English | MEDLINE | ID: mdl-18192500

ABSTRACT

The extracellular signal-regulated kinases 1 and 2 (ERK1/2) are phosphorylated after inhalation of asbestos. The effect of blocking this signaling pathway in lung epithelium is unclear. Asbestos-exposed transgenic mice expressing a dominant-negative mitogen-activated protein kinase kinase-1 (dnMEK1) (i.e., the upstream kinase necessary for phosphorylation of ERK1/2) targeted to lung epithelium exhibited morphologic and molecular changes in lung. Transgene-positive (Tg+) (i.e., dnMEK1) and transgene-negative (Tg-) littermates were exposed to crocidolite asbestos for 2, 4, 9, and 32 days or maintained in clean air (sham controls). Distal bronchiolar epithelium was isolated using laser capture microdissection and mRNA analyzed for molecular markers of proliferation and Clara cell secretory protein (CCSP). Lungs and bronchoalveolar lavage fluids were analyzed for inflammatory and proliferative changes and molecular markers of fibrogenesis. Distal bronchiolar epithelium of asbestos-exposed wild-type mice showed increased expression of c-fos at 2 days. Elevated mRNA levels of histone H3 and numbers of Ki-67-labeled proliferating bronchiolar epithelial cells were decreased at 4 days in asbestos-exposed Tg+ mice. At 32 days, distal bronchioles normally composed of Clara cells in asbestos-exposed Tg+ mouse lungs exhibited nonreplicating ciliated and mucin-secreting cells as well as decreased mRNA levels of CCSP. Gene expression (procollagen 3-a-1, procollagen 1-a-1, and IL-6) linked to fibrogenesis was also increased in lung homogenates of asbestos-exposed Tg- mice, but reduced in asbestos-exposed Tg+ mice. These results suggest a critical role of MEK1 signaling in epithelial cell proliferation and lung remodeling after toxic injury.


Subject(s)
Asbestos, Crocidolite/toxicity , Asbestosis/enzymology , Asbestosis/pathology , Cell Proliferation/drug effects , Lung/enzymology , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Signaling System/physiology , Respiratory Mucosa/enzymology , Animals , Asbestosis/prevention & control , Extracellular Signal-Regulated MAP Kinases/metabolism , Lung/pathology , MAP Kinase Kinase 1/deficiency , MAP Kinase Kinase 1/genetics , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Respiratory Mucosa/drug effects , Respiratory Mucosa/pathology
10.
Int J Oncol ; 29(6): 1573-80, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17088999

ABSTRACT

The role of mitogen-activated protein/extracellular signal-regulated kinase kinase (MEK) signaling in estrogen receptor positive (ER(+)) MCF-7 breast carcinoma cells is not well understood. We depleted MEK by cotransfection of MEK1 and MEK2 siRNA duplexes in a MCF-7 derived line (MCF-7/ lacZ, ML-20) and determined its effect on serum, 17beta-estradiol (E(2)), and growth factor induced DNA synthesis. MEK knockdown did not decrease fetal bovine serum-induced DNA synthesis in ML-20 cells although it did inhibit DNA synthesis induced by estrogen-stripped calf serum (CCS) suggesting that MEK activation plays an important role in growth signaling induced by serum components other than estrogen. Consistent with this notion, MEK knockdown only modestly decreased DNA synthesis induced by E(2)-supplemented CCS medium in ML-20 cells. Similarly, MEK knockdown only caused moderate decreases in DNA synthesis induced by fibroblast growth factor-1 (FGF-1) or heregulin-beta1 (HRGbeta1) in this media. Also, there were only minimal effects of MEK knockdown in cells treated with growth factor-supplemented serum-free medium. Although MEK depletion inhibited ERK1/2 phosphorylation induced by CCS in these cells, that induced by growth factor supplemented CCS media was relatively unaffected. Similarly, ERK1/2 phosphorylation induced by growth factor-supplemented serum-free media was also relatively unaffected by MEK depletion. These results suggest that pathways regulating DNA synthesis induced by serum in MCF-7 cells are significantly more dependent on constitutive MEK levels than that induced by E(2) or growth factors.


Subject(s)
Breast Neoplasms/metabolism , DNA, Neoplasm/biosynthesis , Estradiol/pharmacology , Intercellular Signaling Peptides and Proteins/pharmacology , MAP Kinase Kinase 1/deficiency , MAP Kinase Kinase 2/deficiency , MAP Kinase Kinase Kinases/antagonists & inhibitors , Breast Neoplasms/genetics , Bromodeoxyuridine/metabolism , Culture Media, Serum-Free , DNA, Neoplasm/antagonists & inhibitors , Fibroblast Growth Factor 1/antagonists & inhibitors , Fibroblast Growth Factor 1/pharmacology , Humans , Insulin-Like Growth Factor I/antagonists & inhibitors , Insulin-Like Growth Factor I/pharmacology , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/genetics , MAP Kinase Kinase 2/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neuregulin-1/antagonists & inhibitors , Neuregulin-1/pharmacology , Nucleic Acid Synthesis Inhibitors/pharmacology , Phosphorylation , RNA, Small Interfering/genetics , Serum , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...