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










Publication year range
1.
Neuro Oncol ; 20(5): 621-631, 2018 04 09.
Article in English | MEDLINE | ID: mdl-29401256

ABSTRACT

Background: Glioblastoma (GBM) is one of the most aggressive human brain tumors, with a median survival of 15-18 months. There is a desperate need to find novel therapeutic targets. Various receptor protein kinases have been identified as potential targets; however, response rates in clinical studies have been somewhat disappointing. Targeting the spleen tyrosine kinase (SYK), which acts downstream of a range of oncogenic receptors, may therefore show more promising results. Methods: Kinase expression of brain tumor samples including GBM and low-grade tumors were compared with normal brain and normal human astrocytes by microarray analysis. Furthermore, SYK, LYN, SLP76, and PLCG2 protein expressions were analyzed by immunohistochemistry, western blot, and immunofluorescence of additional GBM patient samples, murine glioma samples, and cell lines. SYK was then blocked chemically and genetically in vitro and in vivo in 2 different mouse models. Multiphoton intravital imaging and multicolor flow cytometry were performed in a syngeneic immunocompetent C57BL/6J mouse GL261 glioma model to study the effect of these inhibitors on the tumor microenvironment. Results: SYK, LYN, SLP76, and PLCG2 were found expressed in human and murine glioma samples and cell lines. SYK inhibition blocked proliferation, migration, and colony formation. Flow cytometric and multiphoton imaging imply that targeting SYK in vivo attenuated GBM tumor growth and invasiveness and reduced B and CD11b+ cell mobility and infiltration. Conclusions: Our data suggest that gliomas express a SYK signaling network important in glioma progression, inhibition of which results in reduced invasion with slower tumor progression.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Movement , Cell Proliferation , Disease Models, Animal , Glioblastoma/pathology , Syk Kinase/metabolism , Tumor Microenvironment , Animals , Apoptosis , Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Female , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Prognosis , Syk Kinase/genetics , Tumor Cells, Cultured
2.
Oncotarget ; 8(41): 69204-69218, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-29050198

ABSTRACT

BRAF inhibitors (BRAFi) and the combination therapy of BRAF and MEK inhibitors (MEKi) were recently approved for therapy of metastatic melanomas harbouring the oncogenic BRAFV600 mutation. Although these therapies have shown pronounced therapeutic efficacy, the limited durability of the response indicates an acquired drug resistance that still remains mechanistically poorly understood at the molecular level. We conducted transcriptome gene profiling in BRAFi-treated melanoma cells and identified that Mer tyrosine kinase (MerTK) is specifically upregulated. MerTK overexpression was demonstrated not only in melanomas resistant to BRAFi monotherapy (5 out of 10 samples from melanoma patients) but also in melanoma resistant to BRAFi+MEKi (1 out of 3), although MEKi alone does not affect MerTK. Mechanistically, BRAFi-induced activation of Zeb2 stimulates MerTK in BRAFV600 melanoma through mTORC1-triggered activation of autophagy. Co-targeting MerTK and BRAFV600 significantly reduced tumour burden in xenografted mice, which was pheno-copied by co-inhibition of autophagy and mutant BRAFV600.

3.
Sci Signal ; 8(397): ra100, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26443704

ABSTRACT

The serine and threonine kinase MST1 is the mammalian homolog of Hippo. MST1 is a critical mediator of the migration, adhesion, and survival of T cells; however, these functions of MST1 are independent of signaling by its typical effectors, the kinase LATS and the transcriptional coactivator YAP. The kinase NDR1, a member of the same family of kinases as LATS, functions as a tumor suppressor by preventing T cell lymphomagenesis, which suggests that it may play a role in T cell homeostasis. We generated and characterized mice with a T cell-specific double knockout of Ndr1 and Ndr2 (Ndr DKO). Compared with control mice, Ndr DKO mice exhibited a substantial reduction in the number of naïve T cells in their secondary lymphoid organs. Mature single-positive thymocytes accumulated in the thymus in Ndr DKO mice. We also found that NDRs acted downstream of MST1 to mediate the egress of mature thymocytes from the thymus, as well as the interstitial migration of naïve T cells within popliteal lymph nodes. Together, our findings indicate that the kinases NDR1 and NDR2 function as downstream effectors of MST1 to mediate thymocyte egress and T cell migration.


Subject(s)
Lymphocytes/cytology , Lymphopenia/enzymology , Protein Serine-Threonine Kinases/physiology , Thymocytes/cytology , Thymus Gland/pathology , Transendothelial and Transepithelial Migration/physiology , Actins/physiology , Animals , Apoptosis , Cell Movement , Chemotaxis , Cytoskeleton/ultrastructure , Lymphocyte Count , Lymphoid Tissue/pathology , Lymphopenia/pathology , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , T-Lymphocyte Subsets/cytology , rho GTP-Binding Proteins/physiology
4.
PLoS One ; 10(8): e0136566, 2015.
Article in English | MEDLINE | ID: mdl-26305214

ABSTRACT

Studies of mammalian tissue culture cells indicate that the conserved and distinct NDR isoforms, NDR1 and NDR2, play essential cell biological roles. However, mice lacking either Ndr1 or Ndr2 alone develop normally. Here, we studied the physiological consequences of inactivating both NDR1 and NDR2 in mice, showing that the lack of both Ndr1/Ndr2 (called Ndr1/2-double null mutants) causes embryonic lethality. In support of compensatory roles for NDR1 and NDR2, total protein and activating phosphorylation levels of the remaining NDR isoform were elevated in mice lacking either Ndr1 or Ndr2. Mice retaining one single wild-type Ndr allele were viable and fertile. Ndr1/2-double null embryos displayed multiple phenotypes causing a developmental delay from embryonic day E8.5 onwards. While NDR kinases are not required for notochord formation, the somites of Ndr1/2-double null embryos were smaller, irregularly shaped and unevenly spaced along the anterior-posterior axis. Genes implicated in somitogenesis were down-regulated and the normally symmetric expression of Lunatic fringe, a component of the Notch pathway, showed a left-right bias in the last forming somite in 50% of all Ndr1/2-double null embryos. In addition, Ndr1/2-double null embryos developed a heart defect that manifests itself as pericardial edemas, obstructed heart tubes and arrest of cardiac looping. The resulting cardiac insufficiency is the likely cause of the lethality of Ndr1/2-double null embryos around E10. Taken together, we show that NDR kinases compensate for each other in vivo in mouse embryos, explaining why mice deficient for either Ndr1 or Ndr2 are viable. Ndr1/2-double null embryos show defects in somitogenesis and cardiac looping, which reveals their essential functions and shows that the NDR kinases are critically required during the early phase of organogenesis.


Subject(s)
Organogenesis/genetics , Protein Serine-Threonine Kinases/genetics , Proteins/genetics , Adaptor Proteins, Signal Transducing , Animals , Embryonic Development/genetics , Humans , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/biosynthesis , Signal Transduction
5.
Curr Biol ; 25(3): 296-305, 2015 Feb 02.
Article in English | MEDLINE | ID: mdl-25601544

ABSTRACT

BACKGROUND: Phosphorylation of the transcriptional coactivator YAP1 is a key event in defining Hippo signaling outputs. Previous studies demonstrated that phosphorylation of YAP1 at serine 127 (S127) sequesters YAP1 in the cytoplasm and consequently inhibits YAP1 transcriptional activity. Mammalian tissue-culture experiments suggest that downstream of MST1/2 signaling, LATS1/2 function as YAP1-S127 kinases. However, studies of Mst1/2 knockout mouse models revealed that the identity of the physiological YAP1-S127 kinase(s) in certain tissues, such as the intestine, remains unknown. RESULTS: We show that mammalian NDR1/2 kinases phosphorylate YAP1 on S127 and thereby negatively regulate YAP1 activity in tissue-cultured cells. By studying NDR1/2-deficient mice, we demonstrate the in vivo relevance of NDR1/2-mediated regulation of YAP1. Specifically, upon loss of NDR1/2 in the intestinal epithelium, endogenous S127 phosphorylation is decreased whereas total YAP1 levels are increased. Significantly, ablation of NDR1/2 from the intestinal epithelium renders mice exquisitely sensitive to chemically induced colon carcinogenesis. Analysis of human colon cancer samples further revealed that NDR2 and YAP1 protein expression are inversely correlated in the majority of samples with high YAP1 expression. Collectively, we report NDR1/2 as physiological YAP1-S127 kinases that might function as tumor suppressors upstream of YAP1 in human colorectal cancer. CONCLUSIONS: We establish mammalian NDR1/2 as bona fide kinases that target YAP1 on S127 in vitro and in vivo. Our findings therefore have important implications for a broad range of research efforts aimed at decoding and eventually manipulating YAP1 biology in cancer settings, regenerative medicine, and possibly also noncancer human diseases.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Gene Expression Regulation, Neoplastic/physiology , Intestinal Mucosa/enzymology , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Blotting, Western , Cell Cycle Proteins , Cell Proliferation , Gene Expression Regulation, Neoplastic/genetics , Histological Techniques , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Serine/metabolism , YAP-Signaling Proteins
6.
Liver Int ; 35(4): 1354-66, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24845341

ABSTRACT

BACKGROUND & AIMS: Non-alcoholic fatty liver disease (NAFLD) is a major health problem and occurs frequently in the context of metabolic syndrome and type 2 diabetes mellitus. Hepatocyte-specific Pten-deficiency in mice was shown previously to result in hepatic steatosis due to hyperactivated AKT2. However, the role of peripheral insulin-sensitive tissues on PTEN- and AKT2-dependent accumulation of hepatic lipids has not been addressed. METHODS: Effects of systemically perturbed PTEN/AKT2 signalling on hepatic lipid content were studied in Pten-haplodeficient (Pten(+/-) /Akt2(+/+) ) mice and Pten-haplodeficient mice lacking Akt2 (Pten(+/-) /Akt2(-/-) ). The liver and skeletal muscle were characterized by histology and/or analysis of insulin signalling. To assess the effects of AKT2 activity in skeletal muscle on hepatic lipid content, AKT2 mutants were expressed in skeletal muscle of Pten(+/+) /Akt2(+/+) and Pten(+/-) /Akt2(+/+) mice using adeno-associated virus 8. RESULTS: Pten(+/-) /Akt2(+/+) mice were found to have a more than 2-fold reduction in hepatic lipid content, at a level similar to that observed in Pten(+/-) /Akt2(-/-) mice. Insulin signalling in the livers of Pten(+/-) /Akt2(+/+) mice was enhanced, indicating that extrahepatic factors prevent lipid accumulation. The skeletal muscle of Pten(+/-) /Akt2(+/+) mice also showed enhanced insulin signalling. Skeletal muscle-specific expression of constitutively active AKT2 reduced hepatic lipid content in Pten(+/+) /Akt2(+/+) mice, and dominant negative AKT2 led to an increase in accumulation of hepatic lipids in both Pten(+/+) /Akt2(+/+) and Pten(+/-) /Akt2(+/+) mice. CONCLUSION: Our results demonstrate that AKT2 activity in skeletal muscle critically affects lipid accumulation in the livers of Pten(+/+) /Akt2(+/+) and Pten(+/-) /Akt2(+/+) mice, and emphasize the role of skeletal muscle in the pathology of NAFLD.


Subject(s)
Haploinsufficiency , Lipid Metabolism , Liver/metabolism , Muscle, Skeletal/enzymology , Non-alcoholic Fatty Liver Disease/prevention & control , PTEN Phosphohydrolase/deficiency , Proto-Oncogene Proteins c-akt/metabolism , Animals , Blood Glucose/metabolism , Disease Models, Animal , Enzyme Activation , Genotype , Glycogen/metabolism , Insulin/blood , Liver/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/pathology , Mutation , Non-alcoholic Fatty Liver Disease/enzymology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , PTEN Phosphohydrolase/genetics , Phenotype , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction , Time Factors
7.
Diabetologia ; 57(12): 2576-85, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25205222

ABSTRACT

AIMS/HYPOTHESIS: Insulin resistance, a major contributor to the pathogenesis of type 2 diabetes, leads to increased hepatic glucose production (HGP) owing to an impaired ability of insulin to suppress hepatic gluconeogenesis. Nuclear receptor oestrogen-related receptor γ (ERRγ) is a major transcriptional regulator of hepatic gluconeogenesis. In this study, we investigated insulin-dependent post-translational modifications (PTMs) altering the transcriptional activity of ERRγ for the regulation of hepatic gluconeogenesis. METHODS: We examined insulin-dependent phosphorylation and subcellular localisation of ERRγ in cultured cells and in the liver of C57/BL6, leptin receptor-deficient (db/db), liver-specific insulin receptor knockout (LIRKO) and protein kinase B (PKB) ß-deficient (Pkbß (-/-)) mice. To demonstrate the role of ERRγ in the inhibitory action of insulin on hepatic gluconeogenesis, we carried out an insulin tolerance test in C57/BL6 mice expressing wild-type or phosphorylation-deficient mutant ERRγ. RESULTS: We demonstrated that insulin suppressed the transcriptional activity of ERRγ by promoting PKB/Akt-mediated phosphorylation of ERRγ at S179 and by eliciting translocation of ERRγ from the nucleus to the cytoplasm through interaction with 14-3-3, impairing its ability to promote hepatic gluconeogenesis. In addition, db/db, LIRKO and Pkbß (-/-) mice displayed enhanced ERRγ transcriptional activity due to a block in PKBß-mediated ERRγ phosphorylation during refeeding. Finally, the phosphorylation-deficient mutant ERRγ S179A was resistant to the inhibitory action of insulin on HGP. CONCLUSIONS/INTERPRETATION: These results suggest that ERRγ is a major contributor to insulin action in maintaining hepatic glucose homeostasis.


Subject(s)
Gluconeogenesis/drug effects , Insulin/pharmacology , Liver/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Estrogen/metabolism , Signal Transduction/drug effects , Animals , Diabetes Mellitus, Type 2/metabolism , Gene Expression Regulation/drug effects , Gluconeogenesis/physiology , Liver/metabolism , Mice , Mice, Knockout , Phosphorylation/drug effects , Receptor, Insulin/genetics , Receptor, Insulin/metabolism
8.
Cancer Res ; 74(14): 3779-89, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24872389

ABSTRACT

New therapeutic targets are needed that circumvent inherent therapeutic resistance of glioblastoma multiforme (GBM). Here, we report such a candidate target in the uncharacterized adaptor protein hMOB3, which we show is upregulated in GBM. In a search for its biochemical function, we found that hMOB3 specifically interacts with MST1 kinase in response to apoptotic stimuli and cell-cell contact. Moreover, hMOB3 negatively regulated apoptotic signaling by MST1 in GBM cells by inhibiting the MST1 cleavage-based activation process. Physical interaction between hMOB3 and MST1 was essential for this process. In vivo investigations established that hMOB3 sustains GBM cell growth at high cell density and promotes tumorigenesis. Our results suggest hMOB3 as a candidate therapeutic target for the treatment of malignant gliomas.


Subject(s)
Apoptosis , Glioblastoma/metabolism , Glioblastoma/pathology , Hepatocyte Growth Factor/metabolism , Microtubule-Associated Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Gene Expression , Glioblastoma/genetics , Heterografts , Humans , Immunohistochemistry , Microtubule-Associated Proteins/genetics , Protein Binding , Proteolysis , Tumor Burden
9.
J Clin Invest ; 124(2): 742-54, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24401275

ABSTRACT

High levels of mammalian target of rapamycin complex 1 (mTORC1) activity in malignant gliomas promote tumor progression, suggesting that targeting mTORC1 has potential as a therapeutic strategy. Remarkably, clinical trials in patients with glioma revealed that rapamycin analogs (rapalogs) have limited efficacy, indicating activation of resistance mechanisms. Targeted depletion of MAPK-interacting Ser/Thr kinase 1 (MNK1) sensitizes glioma cells to the mTORC1 inhibitor rapamycin through an indistinct mechanism. Here, we analyzed how MNK1 and mTORC1 signaling pathways regulate the assembly of translation initiation complexes, using the cap analog m7GTP to enrich for initiation complexes in glioma cells followed by mass spectrometry-based quantitative proteomics. Association of eukaryotic translation initiation factor 4E (eIF4E) with eIF4E-binding protein 1 (4EBP1) was regulated by the mTORC1 pathway, whereas pharmacological blocking of MNK activity by CGP57380 or MNK1 knockdown, along with mTORC1 inhibition by RAD001, increased 4EBP1 binding to eIF4E. Furthermore, combined MNK1 and mTORC1 inhibition profoundly inhibited 4EBP1 phosphorylation at Ser65, protein synthesis and proliferation in glioma cells, and reduced tumor growth in an orthotopic glioblastoma (GBM) mouse model. Immunohistochemical analysis of GBM samples revealed increased 4EBP1 phosphorylation. Taken together, our data indicate that rapalog-activated MNK1 signaling promotes glioma growth through regulation of 4EBP1 and indicate a molecular cross-talk between the mTORC1 and MNK1 pathways that has potential to be exploited therapeutically.


Subject(s)
Brain Neoplasms/drug therapy , Eukaryotic Initiation Factor-4E/metabolism , Glioma/drug therapy , Multiprotein Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/metabolism , Aniline Compounds/chemistry , Animals , Brain Neoplasms/metabolism , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Glioma/metabolism , Humans , Immunohistochemistry , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Mice, Nude , Neoplasm Transplantation , Phosphorylation , Protein Binding , Protein Biosynthesis , Purines/chemistry , Signal Transduction , Sirolimus/analogs & derivatives
10.
Mol Cell Biol ; 32(14): 2685-97, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22586271

ABSTRACT

Acyl coenzyme A (acyl-CoA) thioesterases hydrolyze thioester bonds in acyl-CoA metabolites. The majority of mammalian thioesterases are α/ß-hydrolases and have been studied extensively. A second class of Hotdog-fold enzymes has been less well described. Here, we present a structural and functional analysis of a new mammalian mitochondrial thioesterase, Them5. Them5 and its paralog, Them4, adopt the classical Hotdog-fold structure and form homodimers in crystals. In vitro, Them5 shows strong thioesterase activity with long-chain acyl-CoAs. Loss of Them5 specifically alters the remodeling process of the mitochondrial phospholipid cardiolipin. Them5(-/-) mice show deregulation of lipid metabolism and the development of fatty liver, exacerbated by a high-fat diet. Consequently, mitochondrial morphology is affected, and functions such as respiration and ß-oxidation are impaired. The novel mitochondrial acyl-CoA thioesterase Them5 has a critical and specific role in the cardiolipin remodeling process, connecting it to the development of fatty liver and related conditions.


Subject(s)
Cardiolipins/metabolism , Fatty Liver/etiology , Fatty Liver/metabolism , Palmitoyl-CoA Hydrolase/metabolism , Thiolester Hydrolases/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , Crystallography, X-Ray , Dimerization , Fatty Liver/enzymology , HEK293 Cells , Humans , In Vitro Techniques , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mitochondria, Liver/metabolism , Models, Biological , Models, Molecular , Molecular Sequence Data , Palmitoyl-CoA Hydrolase/chemistry , Palmitoyl-CoA Hydrolase/genetics , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thiolester Hydrolases/chemistry , Thiolester Hydrolases/deficiency , Thiolester Hydrolases/genetics
11.
Cancer Discov ; 2(3): 248-59, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22585995

ABSTRACT

UNLABELLED: Metastatic breast tumor cells display an epithelial-mesenchymal transition (EMT) that increases cell motility, invasion, and dissemination. Although the transcription factor Twist1 has been shown to contribute to EMT and cancer metastasis, the signaling pathways regulating Twist1 activity are poorly understood. Here, we show that Twist1 is ubiquitously phosphorylated in 90% of 1,532 invasive human breast tumors. Akt/protein kinase B (PKB)-mediated Twist1 phosphorylation promotes EMT and breast cancer metastasis by modulating its transcriptional target TGF-ß2, leading to enhanced TGF-ß receptor signaling, which in turn maintains hyperactive phosphoinositide 3-kinase (PI3K)/Akt signaling. Preventing phosphorylation of Twist1, as well as depletion of TGF-ß2, significantly impaired the metastatic potential of cancer cells in vivo, indicating a key role of phosphorylated Twist1 (phospho-Twist1) in mediating cross-talk between the PI3K/Akt and TGF-ß/Smad signaling axes that supports metastatic tumor development. Our results describe a novel signaling event linking PI3K/Akt hyperactivation in tumor cells to direct regulation of Twist1 activation and tumor metastasis. SIGNIFICANCE: We identified the first phospho-Twist1 transcriptional target TGF-ß2, which mediates cross-talk between PI3K/Akt and TGF-ß signaling and promotes tumor metastasis. Our results thus illustrate a direct role of PI3K/Akt signaling in metastatic cancer development and suggest that Twist1 phosphorylation could be a potential therapeutic target in clinical cancer treatment.


Subject(s)
Breast Neoplasms/metabolism , Nuclear Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Transforming Growth Factor beta/metabolism , Twist-Related Protein 1/metabolism , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Phosphorylation , Signal Transduction
12.
Dis Model Mech ; 5(3): 403-11, 2012 May.
Article in English | MEDLINE | ID: mdl-22275470

ABSTRACT

Ovarian cysts affect women of all ages and decrease fertility. In particular, polycystic ovarian syndrome (PCOS), in which multiple follicular cysts develop, affects 5-10% of women of reproductive age and can result in infertility. Current non-invasive treatments for PCOS can resolve cysts and restore fertility, but unresponsive patients must undergo severe ovarian wedge resection and resort to in vitro fertilization. PCOS is related to the deregulation of leutinizing hormone (LH) signaling at various levels of the hypothalamic-pituitary-ovarian axis and resultant hyperproduction of androgens. Because insulin resistance and compensatory hyperinsulinemia are observed in 50-70% of individuals with PCOS, deregulated insulin signaling in the ovary is considered an important factor in the disease. Here we report that aged mice specifically lacking the PKBß (also known as Akt2) isoform that is crucial for insulin signaling develop increased testosterone levels and ovarian cysts, both of which are also observed in insulin-resistant PCOS patients. Young PKBß knockout mice were used to model PCOS by treatment with LH and exhibited a cyst area that was threefold greater than in controls, but without hyperinsulinemia. Thus, loss of PKBß might predispose mice to ovarian cysts independently of hyperactive insulin signaling. Targeted therapeutic augmentation of specific PKBß signaling could therefore provide a new avenue for the treatment and management of ovarian cysts.


Subject(s)
Genetic Predisposition to Disease , Polycystic Ovary Syndrome/enzymology , Polycystic Ovary Syndrome/pathology , Proto-Oncogene Proteins c-akt/deficiency , Aging/drug effects , Aging/pathology , Animals , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Deletion , Humans , Hyperplasia , Isoenzymes/deficiency , Isoenzymes/metabolism , Luteinizing Hormone/administration & dosage , Luteinizing Hormone/pharmacology , Mice , Mice, Knockout , Models, Biological , Polycystic Ovary Syndrome/genetics , Proto-Oncogene Proteins c-akt/metabolism , Reproduction/drug effects , Signal Transduction/drug effects , Steroids/metabolism , Theca Cells/drug effects , Theca Cells/pathology
13.
Sci Signal ; 3(126): ra47, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20551432

ABSTRACT

Defective apoptosis contributes to the development of various human malignancies. The kinases nuclear Dbf2-related 1 (NDR1) and NDR2 mediate apoptosis downstream of the tumor suppressor proteins RASSF1A (Ras association domain family member 1A) and MST1 (mammalian Ste20-like kinase 1). To further analyze the role of NDR1 in apoptosis, we generated NDR1-deficient mice. Although NDR1 is activated by both intrinsic and extrinsic proapoptotic stimuli, which indicates a role for NDR1 in regulating apoptosis, NDR1-deficient T cells underwent apoptosis in a manner similar to that of wild-type cells in response to different proapoptotic stimuli. Analysis of the abundances of NDR1 and NDR2 proteins revealed that loss of NDR1 was functionally compensated for by an increase in the abundance of NDR2 protein. Despite this compensation, NDR1(-/-) and NDR1(+/-) mice were more prone to the development of T cell lymphomas than were wild-type mice. Tumor development in mice and humans was accompanied by a decrease in the overall amounts of NDR proteins in T cell lymphoma samples. Thus, reduction in the abundance of NDR1 triggered a decrease in the total amount of both isoforms. Together, our data suggest that a reduction in the abundances of the NDR proteins results in defective responses to proapoptotic stimuli, thereby facilitating the development of tumors.


Subject(s)
Lymphoma, T-Cell/etiology , Protein Serine-Threonine Kinases/physiology , Animals , Apoptosis , Humans , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/deficiency
14.
Mol Cell Biol ; 30(3): 601-12, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19933838

ABSTRACT

Protein kinase B (PKB)/Akt is considered to be a key target downstream of insulin receptor substrate 2 (IRS2) in the regulation of beta-cell mass. However, while deficiency of IRS2 in mice results in diabetes with insulin resistance and severe failure of beta-cell mass and function, only loss of the PKBbeta isoform leads to a mild metabolic phenotype with insulin resistance. Other isoforms were reported not to be required for metabolic regulation. To clarify the roles of the three PKB isoforms in the regulation of islet mass and glucose homeostasis, we assessed the metabolic and pancreatic phenotypes of Pkbalpha, Pkbbeta, and Pkbgamma-deficient mice. Our study uncovered a novel role for PKBalpha in the regulation of glucose homeostasis, whereas it confirmed that Pkbbeta(-/)(-) mice are insulin resistant with compensatory increase of islet mass. Pkbalpha(-/)(-) mice displayed an opposite phenotype with improved insulin sensitivity, lower blood glucose, and higher serum glucagon concentrations. Pkbgamma(-/)(-) mice did not show metabolic abnormalities. Additionally, our signaling analyses revealed that PKBalpha, but not PKBbeta or PKBgamma, is specifically activated by overexpression of IRS2 in beta-cells and is required for IRS2 action in the islets.


Subject(s)
Glucose/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin-Secreting Cells/enzymology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Homeostasis/physiology , Mice , Protein Isoforms/metabolism
15.
PLoS One ; 4(5): e5471, 2009.
Article in English | MEDLINE | ID: mdl-19421406

ABSTRACT

BACKGROUND: Mitochondria are central to the metabolism of cells and participate in many regulatory and signaling events. They are looked upon as dynamic tubular networks. We showed recently that the Carboxy-Terminal Modulator Protein (CTMP) is a mitochondrial protein that may be released into the cytosol under apoptotic conditions. METHODOLOGY/PRINCIPAL FINDINGS: Here we report an unexpected function of CTMP in mitochondrial homeostasis. In this study, both full length CTMP, and a CTMP mutant refractory to N-terminal cleavage and leading to an immature protein promote clustering of spherical mitochondria, suggesting a role for CTMP in the fission process. Indeed, cellular depletion of CTMP led to accumulation of swollen and interconnected mitochondria, without affecting the mitochondrial fusion process. Importantly, in vivo results support the relevance of these findings, as mitochondria from livers of adult CTMP knockout mice had a similar phenotype to cells depleted of CTMP. CONCLUSIONS/SIGNIFICANCE: Together, these results lead us to propose that CTMP has a major function in mitochondrial dynamics and could be involved in the regulation of mitochondrial functions.


Subject(s)
Carrier Proteins/physiology , Mitochondria, Liver/metabolism , Animals , Antibodies, Monoclonal/immunology , Apoptosis/physiology , Blotting, Western , Carrier Proteins/antagonists & inhibitors , Cytosol/metabolism , HeLa Cells , Humans , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Mitochondria, Liver/ultrastructure , Palmitoyl-CoA Hydrolase , RNA, Small Interfering/pharmacology
16.
J Biol Chem ; 283(44): 30025-33, 2008 Oct 31.
Article in English | MEDLINE | ID: mdl-18757368

ABSTRACT

Full activation of protein kinase B (PKB/Akt) requires phosphorylation on Thr-308 and Ser-473. It is well established that Thr-308 is phosphorylated by 3-phosphoinositide-dependent kinase-1 (PDK1). Ser-473 phosphorylation is mediated by both mammalian target of rapamycin-rictor complex (mTORC2) and DNA-dependent protein kinase (DNA-PK) depending on type of stimulus. However, the physiological role of DNA-PK in the regulation of PKB phosphorylation remains to be established. To address this, we analyzed basal, insulin-induced, and DNA damage-induced PKB Ser-473 phosphorylation in DNA-PK catalytic subunit-null DNA-PKcs(-/-) mice. Our results revealed that DNA-PK is required for DNA damage-induced phosphorylation but dispensable for insulin- and growth factor-induced PKB Ser-473 phosphorylation. Moreover, DNA-PKcs(-/-) mice showed a tissue-specific increase in basal PKB phosphorylation. In particular, persistent PKB hyperactivity in the thymus apparently contributed to spontaneous lymphomagenesis in DNA-PKcs(-/-) mice. Significantly, these tumors could be prevented by deletion of PKBalpha. These findings reveal stimulus-specific regulation of PKB activation by specific upstream kinases and provide genetic evidence of PKB deregulation in DNA-PKcs(-/-) mice.


Subject(s)
DNA-Activated Protein Kinase/metabolism , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins c-akt/metabolism , Animals , DNA Damage , Fibroblasts/metabolism , Flow Cytometry/methods , Gene Expression Profiling , Glucose Tolerance Test , Insulin/metabolism , Mice , Mice, Transgenic , Models, Genetic , Phosphorylation , Time Factors
17.
Am J Pathol ; 173(2): 451-62, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18583310

ABSTRACT

Unexplained intrauterine growth restriction of the fetus (IUGR) results from impaired placental development, frequently associated with maternal malperfusion. Some cases are complicated further by preeclampsia (PE+IUGR). Here, we provide the first evidence that placental protein synthesis inhibition and endoplasmic reticulum (ER) stress play key roles in IUGR pathophysiology. Increased phosphorylation of eukaryotic initiation factor 2alpha suggests suppression of translation initiation in IUGR placentas, with a further increase in PE+IUGR cases. Consequently, AKT levels were reduced at the protein, but not mRNA, level. Additionally, levels of other proteins in the AKT-mammalian target of rapamycin pathway were decreased, and there was associated dephosphorylation of 4E-binding protein 1 and activation of glycogen synthase kinase 3beta. Cyclin D1 and the eukaryotic initiation factor 2B epsilon subunit were also down-regulated, providing additional evidence for this placental phenotype. The central role of AKT signaling in placental growth regulation was confirmed in Akt1 null mice, which display IUGR. In addition, we demonstrated ultrastructural and molecular evidence of ER stress in human IUGR and PE+IUGR placentas, providing a potential mechanism for eukaryotic initiation factor 2alpha phosphorylation. In confirmation, induction of low-grade ER stress in trophoblast-like cell lines reduced cellular proliferation. PE+IUGR placentas showed elevated ER stress with the additional expression of the pro-apoptotic protein C/EBP-homologous protein/growth arrest and DNA damage 153. These findings may account for the increased microparticulate placental debris in the maternal circulation of these cases, leading to endothelial cell activation and impairing placental development.


Subject(s)
Endoplasmic Reticulum/physiology , Fetal Growth Retardation/metabolism , Placenta/metabolism , Pre-Eclampsia/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins , Cell Line , Cell Proliferation , Cyclin D1/metabolism , Down-Regulation , Eukaryotic Initiation Factor-2/metabolism , Female , Fetal Growth Retardation/etiology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Mice , Mice, Knockout , Phosphoproteins/metabolism , Phosphorylation , Placenta/pathology , Pregnancy , Protein Biosynthesis , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/physiology , TOR Serine-Threonine Kinases , Transcription Factor CHOP/metabolism
18.
Mol Cell ; 30(2): 203-13, 2008 Apr 25.
Article in English | MEDLINE | ID: mdl-18439899

ABSTRACT

Protein kinase B (PKB/Akt) is a well-established regulator of several essential cellular processes. Here, we report a route by which activated PKB promotes survival in response to DNA insults in vivo. PKB activation following DNA damage requires 3-phosphoinositide-dependent kinase 1 (PDK1) and DNA-dependent protein kinase (DNA-PK). Active PKB localizes in the nucleus of gamma-irradiated cells adjacent to DNA double-strand breaks, where it colocalizes and interacts with DNA-PK. Levels of active PKB inversely correlate with DNA damage-induced apoptosis. A significant portion of p53- and DNA damage-regulated genes are misregulated in cells lacking PKBalpha. PKBalpha knockout mice show impaired DNA damage-dependent induction of p21 and increased tissue apoptosis after single-dose whole-body irradiation. Our findings place PKB downstream of DNA-PK in the DNA damage response signaling cascade, where it provides a prosurvival signal, in particular by affecting transcriptional p21 regulation. Furthermore, this function is apparently restricted to the PKBalpha isoform.


Subject(s)
Apoptosis , DNA Breaks, Double-Stranded , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis/genetics , Cell Line , Cell Nucleus/enzymology , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA-Activated Protein Kinase/analysis , DNA-Activated Protein Kinase/genetics , DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , Gamma Rays , Gene Expression Regulation , Humans , Mice , Mice, Knockout , Nuclear Proteins/analysis , Nuclear Proteins/genetics , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/analysis , Proto-Oncogene Proteins c-akt/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Radiation Tolerance/genetics , Serine/genetics , Serine/metabolism , Transcription, Genetic
19.
PLoS One ; 2(10): e992, 2007 Oct 03.
Article in English | MEDLINE | ID: mdl-17912369

ABSTRACT

BACKGROUND: The thymus constitutes the primary lymphoid organ for the majority of T cells. The phosphatidyl-inositol 3 kinase (PI3K) signaling pathway is involved in lymphoid development. Defects in single components of this pathway prevent thymocytes from progressing beyond early T cell developmental stages. Protein kinase B (PKB) is the main effector of the PI3K pathway. METHODOLOGY/PRINCIPAL FINDINGS: To determine whether PKB mediates PI3K signaling in the thymus, we characterized PKB knockout thymi. Our results reveal a significant thymic hypocellularity in PKBalpha(-/-) neonates and an accumulation of early thymocyte subsets in PKBalpha(-/-) adult mice. Using thymic grafting and fetal liver cell transfer experiments, the latter finding was specifically attributed to the lack of PKBalpha within the lymphoid component of the thymus. Microarray analyses show that the absence of PKBalpha in early thymocyte subsets modifies the expression of genes known to be involved in pre-TCR signaling, in T cell activation, and in the transduction of interferon-mediated signals. CONCLUSIONS/SIGNIFICANCE: This report highlights the specific requirements of PKBalpha for thymic development and opens up new prospects as to the mechanism downstream of PKBalpha in early thymocytes.


Subject(s)
Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/physiology , Thymus Gland/embryology , Thymus Gland/pathology , Animals , Animals, Newborn , Cell Separation , Cell Transplantation/methods , Gene Deletion , Mice , Mice, Transgenic , Models, Biological , Oligonucleotide Array Sequence Analysis , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism
20.
Mol Cell Biol ; 26(21): 8042-51, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16923958

ABSTRACT

To address the issues of isoform redundancy and isoform specificity of the Akt family of protein kinases in vivo, we generated mice deficient in both Akt2 and Akt3. In these mice, only the Akt1 isoform remains to perform essential Akt functions, such as glucose homeostasis, proliferation, differentiation, and early development. Surprisingly, we found that Akt2(-/-) Akt3(-/-) and even Akt1(+/-) Akt2(-/-) Akt3(-/-) mice developed normally and survived with minimal dysfunctions, despite a dramatic reduction of total Akt levels in all tissues. A single functional allele of Akt1 appears to be sufficient for successful embryonic development and postnatal survival. This is in sharp contrast to the previously described lethal phenotypes of Akt1(-/-) Akt2(-/-) mice and Akt1(-/-) Akt3(-/-) mice. However, Akt2(-/-) Akt3(-/-) mice were glucose and insulin intolerant and exhibited an approximately 25% reduction in body weight compared to wild-type mice. In addition, we found substantial reductions in relative size and weight of the brain and testis in Akt2(-/-) Akt3(-/-) mice, demonstrating an in vivo role for both Akt2 and Akt3 in the determination of whole animal size and individual organ sizes.


Subject(s)
Embryo, Mammalian/physiology , Glucose/metabolism , Homeostasis , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Body Weight , Brain/anatomy & histology , Brain/metabolism , Cells, Cultured , Embryo, Mammalian/anatomy & histology , Female , Glucose Tolerance Test , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis , Organ Size , Phenotype , Protein Isoforms/genetics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/physiology , Testis/anatomy & histology , Testis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...