Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Med Chem ; 67(7): 5259-5271, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38530741

ABSTRACT

A series of activators of GCN2 (general control nonderepressible 2) kinase have been developed, leading to HC-7366, which has entered the clinic as an antitumor therapy. Optimization resulted in improved permeability compared to that of the original indazole hinge binding scaffold, while maintaining potency at GCN2 and selectivity over PERK (protein kinase RNA-like endoplasmic reticulum kinase). The improved ADME properties of this series led to robust in vivo compound exposure in both rats and mice, allowing HC-7366 to be dosed in xenograft models, demonstrating that activation of the GCN2 pathway by this compound leads to tumor growth inhibition.


Subject(s)
Protein Serine-Threonine Kinases , eIF-2 Kinase , Humans , Mice , Rats , Animals , Protein Serine-Threonine Kinases/metabolism , eIF-2 Kinase/metabolism , Mice, Inbred C57BL , RNA , Endoplasmic Reticulum/metabolism
2.
J Biol Chem ; 299(4): 104595, 2023 04.
Article in English | MEDLINE | ID: mdl-36898579

ABSTRACT

The integrated stress response (ISR) is an important mechanism by which cells confer protection against environmental stresses. Central to the ISR is a collection of related protein kinases that monitor stress conditions, such as Gcn2 (EIF2AK4) that recognizes nutrient limitations, inducing phosphorylation of eukaryotic translation initiation factor 2 (eIF2). Gcn2 phosphorylation of eIF2 lowers bulk protein synthesis, conserving energy and nutrients, coincident with preferential translation of stress-adaptive gene transcripts, such as that encoding the Atf4 transcriptional regulator. While Gcn2 is central for cell protection to nutrient stress and its depletion in humans leads to pulmonary disorders, Gcn2 can also contribute to the progression of cancers and facilitate neurological disorders during chronic stress. Consequently, specific ATP-competitive inhibitors of Gcn2 protein kinase have been developed. In this study, we report that one such Gcn2 inhibitor, Gcn2iB, can activate Gcn2, and we probe the mechanism by which this activation occurs. Low concentrations of Gcn2iB increase Gcn2 phosphorylation of eIF2 and enhance Atf4 expression and activity. Of importance, Gcn2iB can activate Gcn2 mutants devoid of functional regulatory domains or with certain kinase domain substitutions derived from Gcn2-deficient human patients. Other ATP-competitive inhibitors can also activate Gcn2, although there are differences in their mechanisms of activation. These results provide a cautionary note about the pharmacodynamics of eIF2 kinase inhibitors in therapeutic applications. Compounds designed to be kinase inhibitors that instead directly activate Gcn2, even loss of function variants, may provide tools to alleviate deficiencies in Gcn2 and other regulators of the ISR.


Subject(s)
Eukaryotic Initiation Factor-2 , Protein Serine-Threonine Kinases , Humans , Adenosine Triphosphate/metabolism , Enzyme Activation/drug effects , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
4.
Nat Cell Biol ; 21(7): 889-899, 2019 07.
Article in English | MEDLINE | ID: mdl-31263264

ABSTRACT

The c-Myc oncogene drives malignant progression and induces robust anabolic and proliferative programmes leading to intrinsic stress. The mechanisms enabling adaptation to MYC-induced stress are not fully understood. Here we reveal an essential role for activating transcription factor 4 (ATF4) in survival following MYC activation. MYC upregulates ATF4 by activating general control nonderepressible 2 (GCN2) kinase through uncharged transfer RNAs. Subsequently, ATF4 co-occupies promoter regions of over 30 MYC-target genes, primarily those regulating amino acid and protein synthesis, including eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1), a negative regulator of translation. 4E-BP1 relieves MYC-induced proteotoxic stress and is essential to balance protein synthesis. 4E-BP1 activity is negatively regulated by mammalian target of rapamycin complex 1 (mTORC1)-dependent phosphorylation and inhibition of mTORC1 signalling rescues ATF4-deficient cells from MYC-induced endoplasmic reticulum stress. Acute deletion of ATF4 significantly delays MYC-driven tumour progression and increases survival in mouse models. Our results establish ATF4 as a cellular rheostat of MYC activity, which ensures that enhanced translation rates are compatible with survival and tumour progression.


Subject(s)
Activating Transcription Factor 4/genetics , Genes, myc/genetics , Transcriptional Activation/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins , Endoplasmic Reticulum Stress/genetics , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice, Transgenic , Phosphoproteins/genetics , Phosphorylation , Protein Biosynthesis/physiology , TOR Serine-Threonine Kinases/metabolism
5.
Cancer Discov ; 9(3): 396-415, 2019 03.
Article in English | MEDLINE | ID: mdl-30563872

ABSTRACT

Resistance to BRAF and MEK inhibitors (BRAFi + MEKi) in BRAF-mutant tumors occurs through heterogeneous mechanisms, including ERK reactivation and autophagy. Little is known about the mechanisms by which ERK reactivation or autophagy is induced by BRAFi + MEKi. Here, we report that in BRAF-mutant melanoma cells, BRAFi + MEKi induced SEC61-dependent endoplasmic reticulum (ER) translocation of the MAPK pathway via GRP78 and KSR2. Inhibition of ER translocation prevented ERK reactivation and autophagy. Following ER translocation, ERK exited the ER and was rephosphorylated by PERK. Reactivated ERK phosphorylated ATF4, which activated cytoprotective autophagy. Upregulation of GRP78 and phosphorylation of ATF4 were detected in tumors of patients resistant to BRAFi + MEKi. ER translocation of the MAPK pathway was demonstrated in therapy-resistant patient-derived xenografts. Expression of a dominant-negative ATF4 mutant conferred sensitivity to BRAFi + MEKi in vivo. This mechanism reconciles two major targeted therapy resistance pathways and identifies druggable targets, whose inhibition would likely enhance the response to BRAFi + MEKi. SIGNIFICANCE: ERK reactivation and autophagy are considered distinct resistance pathways to BRAF + MEK inhibition (BRAFi + MEKi) in BRAF V600E cancers. Here, we report BRAFi + MEKi-induced ER translocation of the MAPK pathway is necessary for ERK reactivation, which drives autophagy. The ER translocation mechanism is a major druggable driver of resistance to targeted therapy.This article is highlighted in the In This Issue feature, p. 305.


Subject(s)
Endoplasmic Reticulum/metabolism , MAP Kinase Signaling System , Melanoma/drug therapy , Melanoma/metabolism , Mutation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Animals , Autophagy , Cell Line, Tumor , Drug Resistance, Neoplasm , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/metabolism , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , Male , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Protein Transport , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Cell Death Dis ; 9(11): 1108, 2018 10 31.
Article in English | MEDLINE | ID: mdl-30382078

ABSTRACT

Terminal differentiation opposes proliferation in the vast majority of tissue types. As a result, loss of lineage differentiation is a hallmark of aggressive cancers, including soft tissue sarcomas (STS). Consistent with these observations, undifferentiated pleomorphic sarcoma (UPS), an STS subtype devoid of lineage markers, is among the most lethal sarcomas in adults. Though tissue-specific features are lost in these mesenchymal tumors they are most commonly diagnosed in skeletal muscle, and are thought to develop from transformed muscle progenitor cells. We have found that a combination of HDAC (Vorinostat) and BET bromodomain (JQ1) inhibition partially restores differentiation to skeletal muscle UPS cells and tissues, enforcing a myoblast-like identity. Importantly, differentiation is partially contingent upon downregulation of the Hippo pathway transcriptional effector Yes-associated protein 1 (YAP1) and nuclear factor (NF)-κB. Previously, we observed that Vorinostat/JQ1 inactivates YAP1 and restores oscillation of NF-κB in differentiating myoblasts. These effects correlate with reduced tumorigenesis, and enhanced differentiation. However, the mechanisms by which the Hippo/NF-κB axis impact differentiation remained unknown. Here, we report that YAP1 and NF-κB activity suppress circadian clock function, inhibiting differentiation and promoting proliferation. In most tissues, clock activation is antagonized by the unfolded protein response (UPR). However, skeletal muscle differentiation requires both Clock and UPR activity, suggesting the molecular link between them is unique in muscle. In skeletal muscle-derived UPS, we observed that YAP1 suppresses PERK and ATF6-mediated UPR target expression as well as clock genes. These pathways govern metabolic processes, including autophagy, and their disruption shifts metabolism toward cancer cell-associated glycolysis and hyper-proliferation. Treatment with Vorinostat/JQ1 inhibited glycolysis/MTOR signaling, activated the clock, and upregulated the UPR and autophagy via inhibition of YAP1/NF-κB. These findings support the use of epigenetic modulators to treat human UPS. In addition, we identify specific autophagy, UPR, and muscle differentiation-associated genes as potential biomarkers of treatment efficacy and differentiation.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Autophagy/genetics , Cell Cycle Proteins/genetics , Gene Expression Regulation, Neoplastic , Muscle Neoplasms/genetics , NF-kappa B/genetics , Sarcoma/genetics , Activating Transcription Factor 6/genetics , Activating Transcription Factor 6/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autophagy/drug effects , Azepines/pharmacology , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Line , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Circadian Clocks/drug effects , Circadian Clocks/genetics , Humans , Mice , Mice, Transgenic , Muscle Neoplasms/drug therapy , Muscle Neoplasms/metabolism , Muscle Neoplasms/pathology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myoblasts/drug effects , Myoblasts/metabolism , Myoblasts/pathology , NF-kappa B/metabolism , Sarcoma/drug therapy , Sarcoma/metabolism , Sarcoma/pathology , Signal Transduction , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/pathology , Triazoles/pharmacology , Unfolded Protein Response/drug effects , Vorinostat/pharmacology , YAP-Signaling Proteins , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
7.
Cell ; 174(1): 72-87.e32, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29861175

ABSTRACT

Recent reports indicate that hypoxia influences the circadian clock through the transcriptional activities of hypoxia-inducible factors (HIFs) at clock genes. Unexpectedly, we uncover a profound disruption of the circadian clock and diurnal transcriptome when hypoxic cells are permitted to acidify to recapitulate the tumor microenvironment. Buffering against acidification or inhibiting lactic acid production fully rescues circadian oscillation. Acidification of several human and murine cell lines, as well as primary murine T cells, suppresses mechanistic target of rapamycin complex 1 (mTORC1) signaling, a key regulator of translation in response to metabolic status. We find that acid drives peripheral redistribution of normally perinuclear lysosomes away from perinuclear RHEB, thereby inhibiting the activity of lysosome-bound mTOR. Restoring mTORC1 signaling and the translation it governs rescues clock oscillation. Our findings thus reveal a model in which acid produced during the cellular metabolic response to hypoxia suppresses the circadian clock through diminished translation of clock constituents.


Subject(s)
Cell Hypoxia , Circadian Clocks , Mechanistic Target of Rapamycin Complex 1/metabolism , Adaptor Proteins, Signal Transducing , Amino Acids, Dicarboxylic/pharmacology , Animals , CLOCK Proteins/metabolism , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins , Cells, Cultured , Circadian Clocks/drug effects , Culture Media/chemistry , Eukaryotic Initiation Factors , Hydrogen-Ion Concentration , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mice , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Ras Homolog Enriched in Brain Protein/metabolism , Signal Transduction/drug effects , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Transcriptome/drug effects , Tuberous Sclerosis Complex 2 Protein/deficiency , Tuberous Sclerosis Complex 2 Protein/genetics
8.
Sci Transl Med ; 10(439)2018 05 02.
Article in English | MEDLINE | ID: mdl-29720449

ABSTRACT

Oncogenic lesions up-regulate bioenergetically demanding cellular processes, such as protein synthesis, to drive cancer cell growth and continued proliferation. However, the hijacking of these key processes by oncogenic pathways imposes onerous cell stress that must be mitigated by adaptive responses for cell survival. The mechanism by which these adaptive responses are established, their functional consequences for tumor development, and their implications for therapeutic interventions remain largely unknown. Using murine and humanized models of prostate cancer (PCa), we show that one of the three branches of the unfolded protein response is selectively activated in advanced PCa. This adaptive response activates the phosphorylation of the eukaryotic initiation factor 2-α (P-eIF2α) to reset global protein synthesis to a level that fosters aggressive tumor development and is a marker of poor patient survival upon the acquisition of multiple oncogenic lesions. Using patient-derived xenograft models and an inhibitor of P-eIF2α activity, ISRIB, our data show that targeting this adaptive brake for protein synthesis selectively triggers cytotoxicity against aggressive metastatic PCa, a disease for which presently there is no cure.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Prostatic Neoplasms/metabolism , Animals , Antineoplastic Agents/therapeutic use , Humans , Male , Mice , Prostatic Neoplasms/drug therapy , Unfolded Protein Response/drug effects , Unfolded Protein Response/physiology
9.
Nat Cell Biol ; 20(1): 104-115, 2018 01.
Article in English | MEDLINE | ID: mdl-29230015

ABSTRACT

The unfolded protein response (UPR) is a stress-activated signalling pathway that regulates cell proliferation, metabolism and survival. The circadian clock coordinates metabolism and signal transduction with light/dark cycles. We explore how UPR signalling interfaces with the circadian clock. UPR activation induces a 10 h phase shift in circadian oscillations through induction of miR-211, a PERK-inducible microRNA that transiently suppresses both Bmal1 and Clock, core circadian regulators. Molecular investigation reveals that miR-211 directly regulates Bmal1 and Clock via distinct mechanisms. Suppression of Bmal1 and Clock has the anticipated impact on expression of select circadian genes, but we also find that repression of Bmal1 is essential for UPR-dependent inhibition of protein synthesis and cell adaptation to stresses that disrupt endoplasmic reticulum homeostasis. Our data demonstrate that c-Myc-dependent activation of the UPR inhibits Bmal1 in Burkitt's lymphoma, thereby suppressing both circadian oscillation and ongoing protein synthesis to facilitate tumour progression.


Subject(s)
Bone Neoplasms/genetics , Circadian Clocks/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Osteosarcoma/genetics , eIF-2 Kinase/genetics , ARNTL Transcription Factors/antagonists & inhibitors , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , CLOCK Proteins/antagonists & inhibitors , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Cell Line, Tumor , Cell Survival , Heterografts , Humans , Light Signal Transduction , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/metabolism , Osteoblasts/metabolism , Osteoblasts/pathology , Osteosarcoma/metabolism , Osteosarcoma/pathology , Photoperiod , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Unfolded Protein Response , eIF-2 Kinase/metabolism
10.
Semin Cancer Biol ; 33: 3-15, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25920797

ABSTRACT

A variety of cell intrinsic or extrinsic stresses evoke perturbations in the folding environment of the endoplasmic reticulum (ER), collectively known as ER stress. Adaptation to stress and re-establishment of ER homeostasis is achieved by activation of an integrated signal transduction pathway called the unfolded protein response (UPR). Both ER stress and UPR activation have been implicated in a variety of human cancers. Although at early stages or physiological conditions of ER stress, the UPR generally promotes survival, when the stress becomes more stringent or prolonged, its role can switch to a pro-cell death one. Here, we discuss historical and recent evidence supporting an involvement of the UPR in malignancy, describe the main mechanisms by which tumor cells overcome ER stress to promote their survival, tumor progression and metastasis and discuss the current state of efforts to develop therapeutic approaches of targeting the UPR.


Subject(s)
Neoplasms/metabolism , Neoplasms/pathology , Unfolded Protein Response , Adaptation, Physiological , Animals , Apoptosis , Autophagy , Cell Lineage , Cellular Senescence , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Homeostasis , Humans , Hypoxia , Mice , Mice, Transgenic , Neoplasm Metastasis , Neoplasms/therapy , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , ras Proteins/metabolism
11.
Cell Rep ; 6(6): 1046-1058, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24613355

ABSTRACT

The ability to interconvert terminally differentiated cells could serve as a powerful tool for cell-based treatment of degenerative diseases, including diabetes mellitus. To determine which, if any, adult tissues are competent to activate an islet ß cell program, we performed an in vivo screen by expressing three ß cell "reprogramming factors" in a wide spectrum of tissues. We report that transient intestinal expression of these factors-Pdx1, MafA, and Ngn3 (PMN)-promotes rapid conversion of intestinal crypt cells into endocrine cells, which coalesce into "neoislets" below the crypt base. Neoislet cells express insulin and show ultrastructural features of ß cells. Importantly, intestinal neoislets are glucose-responsive and able to ameliorate hyperglycemia in diabetic mice. Moreover, PMN expression in human intestinal "organoids" stimulates the conversion of intestinal epithelial cells into ß-like cells. Our results thus demonstrate that the intestine is an accessible and abundant source of functional insulin-producing cells.


Subject(s)
Insulin-Secreting Cells/cytology , Insulin/biosynthesis , Intestines/cytology , Islets of Langerhans/cytology , Animals , Cell Differentiation/physiology , Humans , Insulin-Secreting Cells/metabolism , Intestinal Mucosa/metabolism , Islets of Langerhans/metabolism , Mice , Mice, Transgenic
12.
Autophagy ; 9(4): 612-4, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23328692

ABSTRACT

Stress in the tumor microenvironment in the form of hypoxia and low glucose/amino acid levels activates the evolutionarily conserved cellular adaptation program called the unfolded protein response (UPR) promoting cell survival in such conditions. Our recent studies showed that cell autonomous stress such as activation of the proto-oncogene MYC/c-Myc, can also trigger the UPR and induce endoplasmic reticulum (ER) stress-mediated autophagy. Amelioration of ER stress or autophagy enhances cancer cell death in vitro and attenuates tumor growth in vivo. Here we will discuss the role of the UPR and autophagy in MYC-induced transformation. Our findings demonstrate that the EIF2AK3/PERK-EIF2S1/eIF2α-ATF4 arm of the UPR promotes tumorigenesis by activating autophagy and enhancing tumor formation. Therefore, the UPR is an attractive target in MYC-driven cancers.


Subject(s)
Autophagy , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Proto-Oncogene Proteins c-myc/metabolism , eIF-2 Kinase/metabolism , Animals , Cell Line, Tumor , Endoplasmic Reticulum Stress , Humans , Mice , Models, Biological , Proto-Oncogene Mas , Signal Transduction , Unfolded Protein Response
13.
J Clin Invest ; 122(12): 4621-34, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23143306

ABSTRACT

The proto-oncogene c-Myc paradoxically activates both proliferation and apoptosis. In the pathogenic state, c-Myc-induced apoptosis is bypassed via a critical, yet poorly understood escape mechanism that promotes cellular transformation and tumorigenesis. The accumulation of unfolded proteins in the ER initiates a cellular stress program termed the unfolded protein response (UPR) to support cell survival. Analysis of spontaneous mouse and human lymphomas demonstrated significantly higher levels of UPR activation compared with normal tissues. Using multiple genetic models, we demonstrated that c-Myc and N-Myc activated the PERK/eIF2α/ATF4 arm of the UPR, leading to increased cell survival via the induction of cytoprotective autophagy. Inhibition of PERK significantly reduced Myc-induced autophagy, colony formation, and tumor formation. Moreover, pharmacologic or genetic inhibition of autophagy resulted in increased Myc-dependent apoptosis. Mechanistically, we demonstrated an important link between Myc-dependent increases in protein synthesis and UPR activation. Specifically, by employing a mouse minute (L24+/-) mutant, which resulted in wild-type levels of protein synthesis and attenuation of Myc-induced lymphomagenesis, we showed that Myc-induced UPR activation was reversed. Our findings establish a role for UPR as an enhancer of c-Myc-induced transformation and suggest that UPR inhibition may be particularly effective against malignancies characterized by c-Myc overexpression.


Subject(s)
Autophagy , Burkitt Lymphoma/metabolism , Cell Transformation, Neoplastic/metabolism , Proto-Oncogene Proteins c-myc/physiology , Animals , Apoptosis , Burkitt Lymphoma/pathology , Calcium Signaling , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cluster Analysis , Endoplasmic Reticulum Stress , Gene Knockout Techniques , Heterozygote , Humans , Mice , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Transcriptome , Unfolded Protein Response , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
14.
Mol Biol Cell ; 21(24): 4373-86, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20980617

ABSTRACT

Deletion of the paralogs ZDS1 and ZDS2 in the budding yeast Saccharomyces cerevisiae causes a mis-regulation of polarized cell growth. Here we show a function for these genes as regulators of the Swe1p (Wee1p) kinase-dependent G2/M checkpoint. We identified a conserved domain in the C-terminus of Zds2p consisting of amino acids 813-912 (hereafter referred to as ZH4 for Zds homology 4) that is required for regulation of Swe1p-dependent polarized bud growth. ZH4 is shown by protein affinity assays to be necessary and sufficient for interaction with Cdc55p, a regulatory subunit of protein phosphatase 2A (PP2A). We hypothesized that the Zds proteins are in a pathway that negatively regulates the Swe1p-dependent G2/M checkpoint via Cdc55p. Supporting this model, deletion of CDC55 rescues the aberrant bud morphology of a zds1Δzds2Δ strain. We also show that expression of ZDS1 or ZDS2 from a strong galactose-inducible promoter can induce mitosis even when the Swe1p-dependent G2/M checkpoint is activated by mis-organization of the actin cytoskeleton. This negative regulation requires the CDC55 gene. Together these data indicate that the Cdc55p/Zds2p module has a function in the regulation of the Swe1p-dependent G2/M checkpoint.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Protein Phosphatase 2/metabolism , Protein-Tyrosine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Cell Cycle Proteins/genetics , Conserved Sequence , G2 Phase , Gene Deletion , Mitosis , Mutation, Missense , Phosphorylation , Protein Interaction Mapping , Protein Phosphatase 2/genetics , Protein-Tyrosine Kinases/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Temperature , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...