Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(5): 114176, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38691454

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) carries a dismal prognosis due to therapeutic resistance. We show that PDAC cells undergo global epigenetic reprogramming to acquire chemoresistance, a process that is driven at least in part by protein arginine methyltransferase 1 (PRMT1). Genetic or pharmacological PRMT1 inhibition impairs adaptive epigenetic reprogramming and delays acquired resistance to gemcitabine and other common chemo drugs. Mechanistically, gemcitabine treatment induces translocation of PRMT1 into the nucleus, where its enzymatic activity limits the assembly of chromatin-bound MAFF/BACH1 transcriptional complexes. Cut&Tag chromatin profiling of H3K27Ac, MAFF, and BACH1 suggests a pivotal role for MAFF/BACH1 in global epigenetic response to gemcitabine, which is confirmed by genetically silencing MAFF. PRMT1 and MAFF/BACH1 signature genes identified by Cut&Tag analysis distinguish gemcitabine-resistant from gemcitabine-sensitive patient-derived xenografts of PDAC, supporting the PRMT1-MAFF/BACH1 epigenetic regulatory axis as a potential therapeutic avenue for improving the efficacy and durability of chemotherapies in patients of PDAC.


Subject(s)
Deoxycytidine , Drug Resistance, Neoplasm , Epigenesis, Genetic , Gemcitabine , Pancreatic Neoplasms , Protein-Arginine N-Methyltransferases , Repressor Proteins , Protein-Arginine N-Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases/genetics , Humans , Drug Resistance, Neoplasm/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Repressor Proteins/metabolism , Repressor Proteins/genetics , Cell Line, Tumor , Animals , Mice , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Gene Expression Regulation, Neoplastic/drug effects , Cellular Reprogramming/drug effects , Cellular Reprogramming/genetics
2.
Nat Commun ; 14(1): 1443, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36922511

ABSTRACT

Lineage plasticity has been proposed as a major source of intratumoral heterogeneity and therapeutic resistance. Here, by employing an inducible genetic engineered mouse model, we illustrate that lineage plasticity enables advanced Pancreatic Ductal Adenocarcinoma (PDAC) tumors to develop spontaneous relapse following elimination of the central oncogenic driver - Yap. Transcriptomic and immunohistochemistry analysis of a large panel of PDAC tumors reveals that within high-grade tumors, small niches of PDAC cells gradually evolve to re-activate pluripotent transcription factors (PTFs), which lessen their dependency on Yap. Comprehensive Cut&Tag analysis demonstrate that although acquisition of PTF expression is coupled with the process of epithelial-to-mesenchymal transition (EMT), PTFs form a core transcriptional regulatory circuitry (CRC) with Jun to overcome Yap dependency, which is distinct from the classic TGFb-induced EMT-TF network. A chemical-genetic screen and follow-up functional studies establish Brd4 as an epigenetic gatekeeper for the PTF-Jun CRC, and strong synergy between BET and Yap inhibitors in blocking PDAC growth.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Mice , Animals , Transcription Factors/metabolism , Nuclear Proteins/genetics , Oncogene Addiction , Neoplasm Recurrence, Local , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/pathology , Epithelial-Mesenchymal Transition/genetics , Cell Line, Tumor
3.
Nucleic Acids Res ; 49(21): 12196-12210, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34850122

ABSTRACT

The term 'super enhancers' (SE) has been widely used to describe stretches of closely localized enhancers that are occupied collectively by large numbers of transcription factors (TFs) and co-factors, and control the transcription of highly-expressed genes. Through integrated analysis of >600 DNase-seq, ChIP-seq, GRO-seq, STARR-seq, RNA-seq, Hi-C and ChIA-PET data in five human cancer cell lines, we identified a new class of autonomous SEs (aSEs) that are excluded from classic SE calls by the widely used Rank Ordering of Super-Enhancers (ROSE) method. TF footprint analysis revealed that compared to classic SEs and regular enhancers, aSEs are tightly bound by a dense array of master lineage TFs, which serve as anchors to recruit additional TFs and co-factors in trans. In addition, aSEs are preferentially enriched for Cohesins, which likely involve in stabilizing long-distance interactions between aSEs and their distal target genes. Finally, we showed that aSEs can be reliably predicted using a single DNase-seq data or combined with Mediator and/or P300 ChIP-seq. Overall, our study demonstrates that aSEs represent a unique class of functionally important enhancer elements that distally regulate the transcription of highly expressed genes.


Subject(s)
Chromatin/metabolism , Gene Expression Regulation , Transcription Factors/metabolism , Cell Line, Tumor , Enhancer Elements, Genetic , Humans
4.
Cell Death Differ ; 27(7): 2143-2157, 2020 07.
Article in English | MEDLINE | ID: mdl-31959914

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) and its evolution to inflammatory steatohepatitis (NASH) are the most common causes of chronic liver damage and transplantation that are reaching epidemic proportions due to the upraising incidence of metabolic syndrome, obesity, and diabetes. Currently, there is no approved treatment for NASH. The mitochondrial citrate carrier, Slc25a1, has been proposed to play an important role in lipid metabolism, suggesting a potential role for this protein in the pathogenesis of this disease. Here, we show that Slc25a1 inhibition with a specific inhibitor compound, CTPI-2, halts salient alterations of NASH reverting steatosis, preventing the evolution to steatohepatitis, reducing inflammatory macrophage infiltration in the liver and adipose tissue, while starkly mitigating obesity induced by a high-fat diet. These effects are differentially recapitulated by a global ablation of one copy of the Slc25a1 gene or by a liver-targeted Slc25a1 knockout, which unravel dose-dependent and tissue-specific functions of this protein. Mechanistically, through citrate-dependent activities, Slc25a1 inhibition rewires the lipogenic program, blunts signaling from peroxisome proliferator-activated receptor gamma, a key regulator of glucose and lipid metabolism, and inhibits the expression of gluconeogenic genes. The combination of these activities leads not only to inhibition of lipid anabolic processes, but also to a normalization of hyperglycemia and glucose intolerance as well. In summary, our data show for the first time that Slc25a1 serves as an important player in the pathogenesis of fatty liver disease and thus, provides a potentially exploitable and novel therapeutic target.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Glucose Intolerance/complications , Inflammation/complications , Mitochondria/metabolism , Non-alcoholic Fatty Liver Disease/complications , Acetyl Coenzyme A/metabolism , Animals , Blood Glucose/metabolism , Carrier Proteins/metabolism , Cell Polarity , Citric Acid/metabolism , Diet, High-Fat , Disease Models, Animal , Down-Regulation , Fasting/blood , Gluconeogenesis , Glucose Intolerance/blood , Hepatomegaly/blood , Hepatomegaly/complications , Hepatomegaly/diagnostic imaging , Humans , Hyperglycemia/blood , Hyperglycemia/complications , Inflammation/blood , Insulin Resistance , Interleukin-6/biosynthesis , Lipogenesis , Liver/diagnostic imaging , Liver/metabolism , Liver/pathology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/diagnostic imaging , Obesity/blood , Obesity/complications , Phenotype , Time Factors , Triglycerides/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
5.
Dev Cell ; 51(1): 113-128.e9, 2019 10 07.
Article in English | MEDLINE | ID: mdl-31447265

ABSTRACT

Employing inducible genetically engineered and orthotopic mouse models, we demonstrate a key role for transcriptional regulator Yap in maintenance of Kras-mutant pancreatic tumors. Integrated transcriptional and metabolomics analysis reveals that Yap transcribes Myc and cooperates with Myc to maintain global transcription of metabolic genes. Yap loss triggers acute metabolic stress, which causes tumor regression while inducing epigenetic reprogramming and Sox2 upregulation in a subset of pancreatic neoplastic cells. Sox2 restores Myc expression and metabolic homeostasis in Yap-deficient neoplastic ductal cells, which gradually re-differentiate into acinar-like cells, partially restoring pancreatic parenchyma in vivo. Both the short-term and long-term effects of Yap loss in inducing cell death and re-differentiation, respectively, are blunted in advanced, poorly differentiated p53-mutant pancreatic tumors. Collectively, these findings reveal a highly dynamic and interdependent metabolic, transcriptional, and epigenetic regulatory network governed by Yap, Myc, Sox2, and p53 that dictates pancreatic tumor metabolism, growth, survival, and differentiation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenocarcinoma/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Cell Cycle Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , SOXB1 Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Differentiation , Cell Lineage , Cell Proliferation , DNA Methylation , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HCT116 Cells , Homeostasis , Humans , Mice , Transcription Factors/metabolism , YAP-Signaling Proteins
6.
Trends Cancer ; 5(5): 283-296, 2019 05.
Article in English | MEDLINE | ID: mdl-31174841

ABSTRACT

Drug resistance is a major challenge in cancer treatment. Emerging evidence indicates that deregulation of YAP/TAZ signaling may be a major mechanism of intrinsic and acquired resistance to various targeted and chemotherapies. Moreover, YAP/TAZ-mediated expression of PD-L1 and multiple cytokines is pivotal for tumor immune evasion. While direct inhibitors of YAP/TAZ are still under development, FDA-approved drugs that indirectly block YAP/TAZ activation or critical downstream targets of YAP/TAZ have shown promise in the clinic in reducing therapy resistance. Finally, BET inhibitors, which reportedly block YAP/TAZ-mediated transcription, present another potential venue to overcome YAP/TAZ-induced drug resistance.


Subject(s)
Cell Cycle Proteins/metabolism , Drug Resistance, Neoplasm , Neoplasms/metabolism , Signal Transduction , Transcription Factors/metabolism , Acyltransferases , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor , Drug Resistance, Neoplasm/genetics , Humans , Immunohistochemistry , Molecular Targeted Therapy , Neoplasms/etiology , Neoplasms/pathology , Neoplasms/therapy
7.
Dev Cell ; 49(3): 425-443.e9, 2019 05 06.
Article in English | MEDLINE | ID: mdl-31063758

ABSTRACT

Merlin/NF2 is a bona fide tumor suppressor whose mutations underlie inherited tumor syndrome neurofibromatosis type 2 (NF2), as well as various sporadic cancers including kidney cancer. Multiple Merlin/NF2 effector pathways including the Hippo-YAP/TAZ pathway have been identified. However, the molecular mechanisms underpinning the growth and survival of NF2-mutant tumors remain poorly understood. Using an inducible orthotopic kidney tumor model, we demonstrate that YAP/TAZ silencing is sufficient to induce regression of pre-established NF2-deficient tumors. Mechanistically, YAP/TAZ depletion diminishes glycolysis-dependent growth and increases mitochondrial respiration and reactive oxygen species (ROS) buildup, resulting in oxidative-stress-induced cell death when challenged by nutrient stress. Furthermore, we identify lysosome-mediated cAMP-PKA/EPAC-dependent activation of RAF-MEK-ERK signaling as a resistance mechanism to YAP/TAZ inhibition. Finally, unbiased analysis of TCGA primary kidney tumor transcriptomes confirms a positive correlation of a YAP/TAZ signature with glycolysis and inverse correlations with oxidative phosphorylation and lysosomal gene expression, supporting the clinical relevance of our findings.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Neurofibromin 2/deficiency , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Glycolysis , Heterografts , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , MAP Kinase Signaling System , Mice , Mice, SCID , Neurofibromatosis 2/genetics , Neurofibromatosis 2/metabolism , Neurofibromatosis 2/pathology , Neurofibromin 2/genetics , Neurofibromin 2/metabolism , Oxidative Phosphorylation , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Trans-Activators , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins
8.
Oncogene ; 38(16): 2899-2909, 2019 04.
Article in English | MEDLINE | ID: mdl-30617303

ABSTRACT

The Hippo-Yap/Taz pathway, originally identified as a central developmental regulator of organ size, has been found perturbed in many types of human tumors, and linked to tumor growth, survival, evasion, metastasis, stemness, and drug resistance. Beside these tumor-cell-intrinsic functions, Hippo signaling also plays important immune-regulatory roles. In this review, we will summarize and discuss recent breakthroughs in our understanding of how various components of the Hippo-Yap/Taz pathway influence the tumor immune microenvironment, including their effects on the tumor secretome and immune infiltrates, their roles in regulating crosstalk between tumor cells and T cells, and finally their intrinsic functions in various types of innate and adaptive immune cells. While further research is needed to integrate and reconcile existing findings and to discern the overall effects of Hippo signaling on tumor immunity, it is clear that Hippo signaling functions as a key bridge connecting tumor cells with both the adaptive and innate immune systems. Thus, all future therapeutic development against the Hippo-Yap/Taz pathway should take into account their multi-faceted roles in regulating tumor immunity in addition to their growth-regulatory functions. Given that immune therapies have become the mainstay of cancer treatment, it is also important to pursue how to manipulate Hippo signaling to boost response or overcome resistance to existing immune therapies.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Animals , Humans
9.
Dev Cell ; 39(4): 452-465, 2016 11 21.
Article in English | MEDLINE | ID: mdl-27818180

ABSTRACT

Merlin encoded by the Nf2 gene is a bona fide tumor suppressor that has been implicated in regulation of both the Hippo-Yap and Rac1-Pak1 pathways. Using genetically engineered murine liver models, we show that co-deletion of Rac1 with Nf2 blocks tumor initiation but paradoxically exacerbates hepatomegaly induced by Nf2 loss, which can be suppressed either by treatment with pro-oxidants or by co-deletion of Yap. Our results suggest that while Yap acts as the central driver of proliferation during Nf2 tumorigenesis, Rac1 primarily functions as an inflammation switch by inducing reactive oxygen species that, on one hand, induce nuclear factor κB signaling and expression of inflammatory cytokines, and on the other activate p53 checkpoint and senescence programs dampening the cyclin D1-pRb-E2F1 pathway. Interestingly, senescence markers are associated with benign NF2 tumors but not with malignant NF2 mutant mesotheliomas, suggesting that senescence may underlie the benign nature of most NF2 tumors.


Subject(s)
Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Cycle , DNA Damage , Inflammation/pathology , Neurofibromin 2/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Biomarkers/metabolism , Cell Cycle/genetics , Cell Dedifferentiation , Cell Proliferation , Cellular Senescence , Epithelial Cells/metabolism , Epithelial Cells/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Deletion , Hepatocytes/metabolism , Hepatocytes/pathology , Hepatomegaly/metabolism , Hepatomegaly/pathology , Humans , Liver/metabolism , Liver/pathology , Meningioma/metabolism , Meningioma/pathology , Mice , Mice, Knockout , NF-kappa B/metabolism , Neurilemmoma/metabolism , Neurilemmoma/pathology , Organ Size , Phenotype , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism
10.
Sci Signal ; 7(324): ra42, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24803537

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer with poor survival rates and frequently carries oncogenic KRAS mutation. However, KRAS has thus far not been a viable therapeutic target. We found that the abundance of YAP mRNA, which encodes Yes-associated protein (YAP), a protein regulated by the Hippo pathway during tissue development and homeostasis, was increased in human PDAC tissue compared with that in normal pancreatic epithelia. In genetically engineered Kras(G12D) and Kras(G12D):Trp53(R172H) mouse models, pancreas-specific deletion of Yap halted the progression of early neoplastic lesions to PDAC without affecting normal pancreatic development and endocrine function. Although Yap was dispensable for acinar to ductal metaplasia (ADM), an initial step in the progression to PDAC, Yap was critically required for the proliferation of mutant Kras or Kras:Trp53 neoplastic pancreatic ductal cells in culture and for their growth and progression to invasive PDAC in mice. Yap functioned as a critical transcriptional switch downstream of the oncogenic KRAS-mitogen-activated protein kinase (MAPK) pathway, promoting the expression of genes encoding secretory factors that cumulatively sustained neoplastic proliferation, a tumorigenic stromal response in the tumor microenvironment, and PDAC progression in Kras and Kras:Trp53 mutant pancreas tissue. Together, our findings identified Yap as a critical oncogenic KRAS effector and a promising therapeutic target for PDAC and possibly other types of KRAS-mutant cancers.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/pathology , Genes, ras , Phosphoproteins/physiology , Adaptor Proteins, Signal Transducing/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Proliferation , Disease Progression , Genes, p53 , Humans , Mice , Mutation , Pancreatic Ducts/metabolism , Phosphoproteins/metabolism , RNA, Messenger/genetics , Transcription Factors , YAP-Signaling Proteins
11.
Sci Signal ; 6(291): ra77, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-24003254

ABSTRACT

The Hippo-Yap signaling pathway regulates a number of developmental and adult cellular processes, including cell fate determination, tissue growth, and tumorigenesis. Members of the scaffold protein angiomotin (Amot) family interact with several Hippo pathway components, including Yap (Yes-associated protein), and either stimulate or inhibit Yap activity. We used a combination of genetic, biochemical, and transcriptional approaches to assess the functional consequences of the Amot-Yap interaction in mice and in human cells. Mice with a liver-specific Amot knockout exhibited reduced hepatic "oval cell" proliferation and tumorigenesis in response to toxin-induced injury or when crossed with mice lacking the tumor suppressor Nf2. Biochemical examination of the Amot-Yap interaction revealed that the p130 splicing isoform of Amot (Amot-p130) and Yap interacted in both the cytoplasm and nucleus, which involved binding of PPxY and LPxY motifs in Amot-p130 to WW domains of Yap. In the cytoplasm, Amot-p130 prevented the phosphorylation of Yap by blocking access of the WW domains to the kinase Lats1. Within the nucleus, Amot-p130 was associated with the transcriptional complex containing Yap and Teads (TEA domain family members) and contributed to the regulation of a subset of Yap target genes, many of which are associated with tumorigenesis. These findings indicated that Amot acts as a Yap cofactor, preventing Yap phosphorylation and augmenting its activity toward a specific set of genes that facilitate tumorigenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Liver Neoplasms/metabolism , Liver/metabolism , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Neoplasm Proteins/metabolism , Phosphoproteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Motifs , Angiomotins , Animals , Cell Cycle Proteins , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Epithelial Cells/pathology , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Membrane Proteins/genetics , Mice , Mice, Knockout , Microfilament Proteins/genetics , Neoplasm Proteins/genetics , Phosphoproteins/genetics , Phosphorylation/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Transcription Factors , YAP-Signaling Proteins
12.
Cancer Cell ; 19(4): 527-40, 2011 Apr 12.
Article in English | MEDLINE | ID: mdl-21481793

ABSTRACT

The Merlin/NF2 tumor suppressor restrains cell growth and tumorigenesis by controlling contact-dependent inhibition of proliferation. We have identified a tight-junction-associated protein complex comprising Merlin, Angiomotin, Patj, and Pals1. We demonstrate that Angiomotin functions downstream of Merlin and upstream of Rich1, a small GTPase Activating Protein, as a positive regulator of Rac1. Merlin, through competitive binding to Angiomotin, releases Rich1 from the Angiomotin-inhibitory complex, allowing Rich1 to inactivate Rac1, ultimately leading to attenuation of Rac1 and Ras-MAPK pathways. Patient-derived Merlin mutants show diminished binding capacities to Angiomotin and are unable to dissociate Rich1 from Angiomotin or inhibit MAPK signaling. Depletion of Angiomotin in Nf2(-/-) Schwann cells attenuates the Ras-MAPK signaling pathway, impedes cellular proliferation in vitro and tumorigenesis in vivo.


Subject(s)
Intercellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Microfilament Proteins/physiology , Neurofibromin 2/physiology , Signal Transduction/physiology , Tight Junctions/physiology , Tumor Suppressor Proteins/physiology , Amino Acid Sequence , Angiomotins , Animals , Cell Proliferation , GTPase-Activating Proteins/physiology , Humans , MAP Kinase Signaling System , Mice , Mice, SCID , Molecular Sequence Data , Nucleoside-Phosphate Kinase/physiology , Peripheral Nerves/chemistry , Schwann Cells/chemistry , Tight Junction Proteins , rac1 GTP-Binding Protein/physiology
13.
Genes Dev ; 24(16): 1673-9, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20713513

ABSTRACT

The role of the NF2 gene as a tumor suppressor has been well established. In this issue of Genes & Development, Benhamouche and colleagues (pp. 1718-1730) demonstrate that NF2 is also involved in the regulation of organ size control in mammals. Conditional knockout of Nf2 in the mouse liver results in massive organ enlargement and eventual tumor development, which is attributed to the specific expansion of oval cells. Here we discuss these findings and the proposed molecular mechanisms involved within the context of our current understanding of the pathways regulated by NF2.


Subject(s)
ErbB Receptors/metabolism , Neurofibromatosis 2/physiopathology , Neurofibromin 2/metabolism , Organ Size/genetics , Protein Serine-Threonine Kinases/metabolism , Animals , Disease Models, Animal , Humans , Mice , Signal Transduction
14.
Biochem Pharmacol ; 80(5): 683-9, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20302846

ABSTRACT

The p21-activated kinases (PAKs), immediate downstream effectors of the small G-proteins of the Rac/cdc42 family, are critical mediators of signaling pathways regulating cellular behaviors and as such, have been implicated in pathological conditions including cancer. Recent studies have validated the requirement for PAKs in promoting tumorigenesis in breast carcinoma and neurofibromatosis. Thus, there has been considerable interest in the development of inhibitors to the PAKs, as biological markers and leads for the development of therapeutics. While initial approaches were based on screening for competitive organic inhibitors, more recent efforts have focused on the identification of allosteric inhibitors, organometallic ATP-competitive inhibitors and the use of PAK1/inhibitor crystal structures for inhibitor optimization. This has led to the identification of highly selective and potent inhibitors, which will serve as a basis for further development of inhibitors for therapeutic applications.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , p21-Activated Kinases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Drug Design , Humans , Neoplasms/enzymology , Protein Kinase Inhibitors/pharmacology
15.
J Am Chem Soc ; 130(47): 15764-5, 2008 Nov 26.
Article in English | MEDLINE | ID: mdl-18973295

ABSTRACT

A strategy for targeting protein kinases with large ATP-binding sites by using bulky and rigid octahedral ruthenium complexes as structural scaffolds is presented. A highly potent and selective GSK3 and Pim1 half-sandwich complex NP309 was successfully converted into a PAK1 inhibitor by making use of the large octahedral compounds Lambda-FL172 and Lambda-FL411 in which the cyclopentadienyl moiety of NP309 is replaced by a chloride and sterically demanding diimine ligands. A 1.65 A cocrystal structure of PAK1 with Lambda-FL172 reveals how the large coordination sphere of the ruthenium complex matches the size of the active site and serves as a yardstick to discriminate between otherwise closely related binding sites.


Subject(s)
Protein Kinases/chemistry , Protein Kinases/metabolism , Ruthenium Compounds/chemistry , Animals , Catalytic Domain , Cell Line, Tumor , Crystallography, X-Ray , Inhibitory Concentration 50 , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinases/genetics , Rats , Ruthenium Compounds/chemical synthesis , Ruthenium Compounds/pharmacology , Substrate Specificity
16.
Cancer Res ; 68(19): 7932-7, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18829550

ABSTRACT

Neurofibromatosis type 2 (NF2) is a dominantly inherited cancer disorder caused by mutations at the NF2 gene locus. Merlin, the protein product of the NF2 gene, has been shown to negatively regulate Rac1 signaling by inhibiting its downstream effector kinases, the p21-activated kinases (Pak). Given the implication of Paks in tumorigenesis, it is plausible that merlin's tumor suppressive function might be mediated, at least in part, via inhibition of the Paks. We present data indicating this is indeed the case. First, analysis of primary schwannoma samples derived from NF2 patients showed that in a significant fraction of the tumors, the activity of Pak1 was highly elevated. Second, we used shRNAs to knockdown Pak1, 2, and 3 in NIH3T3 cells expressing a dominant-negative form of merlin, NF2(BBA) (NIH3T3/NF2(BBA)), and find that simultaneous knockdown of Pak1-3 in these cells significantly reduced their growth rates in vitro and inhibited their ability to form tumors in vivo. Finally, while attempting to silence Pak1 in rat schwannoma cells, we found that these cells were unable to tolerate long-term Pak1 inhibition and rapidly moved to restore Pak1 levels by shutting down Pak1 shRNA expression through a methylation-dependent mechanism. These data suggest that inhibiting Pak could be a beneficial approach for the development of therapeutics toward NF2. In addition, the finding that the shRNA-mediated Pak1 suppression was silenced rapidly by methylation raises questions about the future application of such technologies for the treatment of diseases such as cancer.


Subject(s)
Neurilemmoma/enzymology , Neurofibromatosis 2/enzymology , p21-Activated Kinases/antagonists & inhibitors , Animals , Base Sequence , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Cells, Cultured , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Mice , Molecular Sequence Data , NIH 3T3 Cells , Neurilemmoma/genetics , Neurilemmoma/pathology , Neurofibromatosis 2/genetics , Neurofibromatosis 2/pathology , RNA, Small Interfering/pharmacology , Transplantation, Heterologous , p21-Activated Kinases/genetics , p21-Activated Kinases/physiology
17.
Genes Dev ; 21(16): 2055-68, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17699752

ABSTRACT

The development of the embryonic vascular system into a highly ordered network requires precise control over the migration and branching of endothelial cells (ECs). We have previously identified angiomotin (Amot) as a receptor for the angiogenesis inhibitor angiostatin. Furthermore, DNA vaccination targeting Amot inhibits angiogenesis and tumor growth. However, little is known regarding the role of Amot in physiological angiogenesis. We therefore investigated the role of Amot in embryonic neovascularization during zebrafish and mouse embryogenesis. Here we report that knockdown of Amot in zebrafish reduced the number of filopodia of endothelial tip cells and severely impaired the migration of intersegmental vessels. We further show that 75% of Amot knockout mice die between embryonic day 11 (E11) and E11.5 and exhibit severe vascular insufficiency in the intersomitic region as well as dilated vessels in the brain. Furthermore, using ECs differentiated from embryonic stem (ES) cells, we demonstrate that Amot-deficient cells have intact response to vascular endothelial growth factor (VEGF) in regard to differentiation and proliferation. However, the chemotactic response to VEGF was abolished in Amot-deficient cells. We provide evidence that Amot is important for endothelial polarization during migration and that Amot controls Rac1 activity in endothelial and epithelial cells. Our data demonstrate a critical role for Amot during vascular patterning and endothelial polarization.


Subject(s)
Intercellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Microfilament Proteins/physiology , Neovascularization, Physiologic/physiology , Zebrafish Proteins/physiology , Zebrafish/embryology , Zebrafish/physiology , Angiomotins , Animals , Base Sequence , Body Patterning/genetics , Body Patterning/physiology , Cell Line , Cell Movement/genetics , Cell Movement/physiology , DNA Primers/genetics , Endothelial Cells/cytology , Endothelial Cells/physiology , Female , Gene Deletion , Gene Silencing , Humans , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/deficiency , Microfilament Proteins/genetics , Neovascularization, Physiologic/genetics , Phenotype , Pregnancy , Pseudopodia/ultrastructure , Zebrafish/genetics , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , rac1 GTP-Binding Protein/metabolism
18.
Int J Biochem Cell Biol ; 38(7): 1076-83, 2006.
Article in English | MEDLINE | ID: mdl-16497536

ABSTRACT

Constitutive photomorphogenic 1 (COP1), a protein composed of a RING finger, a coiled-coil domain and seven WD40 repeats, functions as an E3 ubiquitin ligase that targets key transcription factors for ubiquitination and degradation in both higher plants and mammalian cells. While COP1 is required for light-mediated development in plants, its mammalian counterpart has been implicated in tumorigenesis. We previously showed that COP1 forms high-molecular-weight complexes in mammalian cells. Here we report our attempts in characterizing the components of the mammalian COP1 complexes by affinity purification combined with mass spectral analysis. We find that both transiently and stably expressed COP1 associates with the hetero-oligomeric TCP-1 chaperonin complex (TRiC), heat shock protein 70 (Hsp70) and BAG-family molecular chaperone regulator-2 (BAG2). In addition, stably expressed COP1 binds to major vault protein (MVP) and translocated promoter region (Tpr). The TRiC/Hsp70 complex is known to interact with and assist in the folding of a number of WD40 proteins in Saccharomyces cerevisiae. The association of WD40 protein COP1 with TRiC/Hsp70 in mammalian cells suggests that facilitating the folding of WD40 proteins may be a conserved function for TRiC/Hsp70 from yeast to mammals.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis Proteins/isolation & purification , Carrier Proteins/chemistry , Carrier Proteins/isolation & purification , Chaperonins/chemistry , Chaperonins/metabolism , Animals , Arabidopsis Proteins/metabolism , Carrier Proteins/metabolism , Chaperonin Containing TCP-1 , Chaperonins/isolation & purification , Chromatography, Affinity/methods , Humans , Metallochaperones , Molecular Sequence Data , Protein Binding , Protein Folding , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/isolation & purification , Ubiquitin-Protein Ligases
19.
Mol Cell Biol ; 25(22): 10052-9, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16260618

ABSTRACT

The band 4.1 proteins are cytoskeletal proteins, harboring a conserved FERM domain highly homologous to the N-terminal FERM domain of ezrin, radixin, moesin, and merlin. Recently, a truncated form of the 4.1B protein, termed Dal-1, was identified in a screen as down regulated in adenocarcinoma of the lung and was mapped to chromosome 18p11.3, which is lost in 38% of primary non-small cell lung carcinoma tumors. Analysis of several meningiomas has shown that Dal-1 expression was lost in 76% of the tumors. To further elucidate the function of the 4.1B/Dal-1 gene in development and tumorigenesis we generated mice deficient for this allele. The 4.1B/Dal-1 null mice develop normally and are fertile. Rates of cellular proliferation and apoptosis in brain, mammary, and lung tissues from the 4.1B/Dal-1 null mice were indistinguishable from those seen with wild-type mice. Aging studies indicate that these mice do not have a propensity to develop tumors. Analysis of fibroblasts from these mice demonstrated that the growth characteristics and kinetics of these cells were not different from those of cells from the wild-type mice. These findings indicate that the 4.1B gene is not required for normal development and that 4.1B/Dal-1 does not function as a tumor suppressor gene.


Subject(s)
Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/physiology , Genetic Predisposition to Disease , Membrane Proteins/genetics , Membrane Proteins/physiology , Neoplasms/genetics , Neoplasms/pathology , Alleles , Animals , Apoptosis , Blotting, Southern , Blotting, Western , Brain/metabolism , Cell Death , Cell Proliferation , Cells, Cultured , Cloning, Molecular , DNA Primers/chemistry , Female , Genotype , Immunohistochemistry , Kinetics , Lung/metabolism , Mammary Glands, Animal/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Models, Genetic , Mutation , Neoplasms/metabolism , Polymerase Chain Reaction
20.
Trends Cell Biol ; 15(11): 618-25, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16198569

ABSTRACT

The COP1 (constitutive photomorphogenic 1) protein, comprising RING finger, coiled-coil and WD40 domains, is conserved in both higher plants and vertebrates. In plants, COP1 acts as an E3 ubiquitin ligase to repress light signaling by targeting photoreceptors and downstream transcription factors for ubiquitylation and degradation. The activity of COP1 in plant cells correlates with its cytoplasmic and nuclear partitioning according to dark or light conditions. In addition, various signaling molecules have been shown to directly interact with COP1 and modulate its activity. Recently, scientists have begun to probe the function and regulation of COP1 in mammalian systems. Initial studies have pointed at possible roles for mammalian COP1 in tumorigenesis and the stress response through regulating the activities of p53 and c-Jun.


Subject(s)
Arabidopsis Proteins/physiology , Nuclear Proteins/physiology , Ubiquitin-Protein Ligases/physiology , Animals , Humans , Models, Biological , Morphogenesis/physiology , Morphogenesis/radiation effects , Neoplasms/etiology , Plant Development , Plants/radiation effects , Protein Transport/physiology , Proto-Oncogene Proteins c-jun/metabolism , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...