Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Hum Genet ; 107(2): 173-174, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32773128
2.
Am J Hum Genet ; 102(2): 197-198, 2018 Feb 01.
Article in English | MEDLINE | ID: mdl-29407281
3.
Semin Oncol ; 33(4): 457-65, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16890800

ABSTRACT

A large body of work spanning the past decade has identified the molecular chaperone heat shock protein 90 (Hsp90) as a critical modulator of an extensive network of cellular signaling pathways. Many of the processes overseen by Hsp90 are deregulated in tumor cells, including cell cycle control, gene transcription, and apoptotic signaling. Hsp90 inhibition offers the potential of accomplishing what most molecularly targeted anticancer therapies do not--the simultaneous disruption of multiple signaling events critical to tumor cell growth and survival. Indeed, small molecule inhibitors of Hsp90 function are actively being evaluated in the clinic as anticancer agents. In this review, we highlight the current understanding of Hsp90 biology as it relates to cancer and discuss the discovery, development, and clinical status of Hsp90 inhibitors as anticancer drugs.


Subject(s)
Antineoplastic Agents/therapeutic use , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Signal Transduction/drug effects , Clinical Trials as Topic , HSP90 Heat-Shock Proteins/metabolism , Humans , Neoplasms/metabolism , Signal Transduction/physiology
4.
Int J Biochem Cell Biol ; 38(3): 317-32, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16290097

ABSTRACT

In the broadest sense, cellular stress describes conditions wherein cells encounter and react to a 'non-normal' state. Perturbations may originate through both extracellular and intracellular means. Whereas transient levels of stress are expected to occur on a regular basis, a series of checks and balances ensures that cells are well equipped to maintain a homeostatic state. In the case of supra-physiological stress signaling, cellular challenges are more severe, and programmed cell death may be the best option for the organism. The ability of a cell, and by extension, an organism, to adequately manage cellular stress is fundamental--a question of life or death. The endoplasmic reticulum (ER) is exquisitely poised to sense and respond to cellular stresses including those that result from metabolic and/or protein folding imbalances. In response to stress originating from within the ER, the PERK and Ire1 protein kinases, along with other proximal signaling molecules, initiate a program of transcriptional and translational regulation termed the unfolded protein response. A consequence of ER stress is the accumulation of reactive oxygen species that promotes a state of oxidative stress. PERK signaling, via activation of the Nrf2 and ATF4 transcription factors, coordinates the convergence of ER stress with oxidative stress signaling. Here we discuss progress regarding the signaling pathways involved in these cellular stresses and the implications of the intersection between the two signaling pathways.


Subject(s)
Endoplasmic Reticulum/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Signal Transduction/physiology , eIF-2 Kinase/metabolism , Activating Transcription Factor 6/metabolism , Animals , Caspase 12 , Caspases/metabolism , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism
5.
Mol Biol Cell ; 16(12): 5493-501, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16176978

ABSTRACT

Exposure of cells to endoplasmic reticulum (ER) stress leads to activation of PKR-like ER kinase (PERK), eukaryotic translation initiation factor 2alpha (eIF2alpha) phosphorylation, repression of cyclin D1 translation, and subsequent cell cycle arrest in G1 phase. However, whether PERK is solely responsible for regulating cyclin D1 accumulation after unfolded protein response pathway (UPR) activation has not been assessed. Herein, we demonstrate that repression of cyclin D1 translation after UPR activation occurs independently of PERK, but it remains dependent on eIF2alpha phosphorylation. Although phosphorylation of eIF2alpha in PERK-/- fibroblasts is attenuated in comparison with wild-type fibroblasts, it is not eliminated. The residual eIF2alpha phosphorylation correlates with the kinetics of cyclin D1 loss, suggesting that another eIF2alpha kinase functions in the absence of PERK. In cells harboring targeted deletion of both PERK and GCN2, cyclin D1 loss is attenuated, suggesting GCN2 functions as the redundant kinase. Consistent with these results, cyclin D1 translation is also stabilized in cells expressing a nonphosphorylatable allele of eIF2alpha; in contrast, repression of global protein translation still occurs in these cells, highlighting a high degree of specificity in transcripts targeted for translation inhibition by phosphorylated eIF2alpha. Our results demonstrate that PERK and GCN2 function to cooperatively regulate eIF2alpha phosphorylation and cyclin D1 translation after UPR activation.


Subject(s)
Cell Cycle/physiology , Eukaryotic Initiation Factor-2/metabolism , Protein Kinases/metabolism , eIF-2 Kinase/metabolism , Animals , Cell Line , Cyclin D1/genetics , Cyclin D1/metabolism , Endoplasmic Reticulum/physiology , Flow Cytometry , Mice , Mice, Knockout , Mice, Transgenic , Phosphorylation , Protein Biosynthesis , Protein Denaturation , Protein Serine-Threonine Kinases , Reverse Transcriptase Polymerase Chain Reaction , eIF-2 Kinase/deficiency , eIF-2 Kinase/genetics
6.
Mol Cell Biol ; 24(19): 8477-86, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15367669

ABSTRACT

The Nrf2 transcription factor promotes survival following cellular insults that trigger oxidative damage. Nrf2 activity is opposed by the BTB/POZ domain protein Keap1. Keap1 is proposed to regulate Nrf2 activity strictly through its capacity to inhibit Nrf2 nuclear import. Recent work suggests that inhibition of Nrf2 may also depend upon ubiquitin-mediated proteolysis. To address the contribution of Keap1-dependent sequestration versus Nrf2 proteolysis, we identified the E3 ligase that regulates Nrf2 ubiquitination. We demonstrate that Keap1 is not solely a cytosolic anchor; rather, Keap1 is an adaptor that bridges Nrf2 to Cul3. We demonstrate that Cul3-Keap1 complexes regulate Nrf2 polyubiquitination both in vitro and in vivo. Inhibition of either Keap1 or Cul3 increases Nrf2 nuclear accumulation, leading to promiscuous activation of Nrf2-dependent gene expression. Our data demonstrate that Keap1 restrains Nrf2 activity via its capacity to target Nrf2 to a cytoplasmic Cul3-based E3 ligase and suggest a model in which Keap1 coordinately regulates both Nrf2 accumulation and access to target genes.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/metabolism , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , Oxidative Stress , Trans-Activators/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Endoplasmic Reticulum/metabolism , Kelch-Like ECH-Associated Protein 1 , Mice , NF-E2-Related Factor 2 , Time Factors
7.
J Biol Chem ; 279(19): 20108-17, 2004 May 07.
Article in English | MEDLINE | ID: mdl-14978030

ABSTRACT

The accumulation of unfolded proteins elicits a cellular response that triggers both pro-survival and pro-apoptotic signaling events. PERK-dependent activation of NF-E2-related factor-2 (Nrf2) is critical for survival signaling during this response; however, the mechanism whereby Nrf2 confers a protective advantage to stressed cells remains to be defined. We now demonstrate that Nrf2 activation contributes to the maintenance of glutathione levels, which in turn functions as a buffer for the accumulation of reactive oxygen species during the unfolded protein response. The deleterious effects of Nrf2 or PERK deficiencies could be attenuated by the restoration of cellular glutathione levels or Nrf2 activity. In addition, the inhibition of reactive oxygen species production attenuated apoptotic induction following endoplasmic reticulum stress. Our data suggest that perturbations in cellular redox status sensitize cells to the harmful effects of endoplasmic reticulum stress, but that other factors are essential for apoptotic commitment.


Subject(s)
DNA-Binding Proteins/metabolism , Endoplasmic Reticulum/metabolism , Oxidation-Reduction , Trans-Activators/metabolism , eIF-2 Kinase/metabolism , Animals , Apoptosis , Blotting, Northern , Blotting, Southern , Cell Survival , Cells, Cultured , Culture Techniques , Electrophoresis, Polyacrylamide Gel , Fibroblasts/metabolism , Glucose/metabolism , Glutathione/metabolism , Immunoblotting , Mice , Microscopy, Fluorescence , NF-E2-Related Factor 2 , Oxidative Stress , Plasmids/metabolism , Precipitin Tests , Protein Folding , Reactive Oxygen Species , Signal Transduction , Subcellular Fractions , Time Factors , Transfection
8.
Mol Cell Biol ; 23(20): 7198-209, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14517290

ABSTRACT

Activation of PERK following the accumulation of unfolded proteins in the endoplasmic reticulum (ER) promotes translation inhibition and cell cycle arrest. PERK function is essential for cell survival following exposure of cells to ER stress, but the mechanisms whereby PERK signaling promotes cell survival are not thoroughly understood. We have identified the Nrf2 transcription factor as a novel PERK substrate. In unstressed cells, Nrf2 is maintained in the cytoplasm via association with Keap1. PERK-dependent phosphorylation triggers dissociation of Nrf2/Keap1 complexes and inhibits reassociation of Nrf2/Keap1 complexes in vitro. Activation of PERK via agents that trigger the unfolded protein response is both necessary and sufficient for dissociation of cytoplasmic Nrf2/Keap1 and subsequent Nrf2 nuclear import. Finally, we demonstrate that cells harboring a targeted deletion of Nrf2 exhibit increased cell death relative to wild-type counterparts following exposure to ER stress. Our data demonstrate that Nrf2 is a critical effector of PERK-mediated cell survival.


Subject(s)
DNA-Binding Proteins/metabolism , Trans-Activators/metabolism , eIF-2 Kinase/metabolism , Active Transport, Cell Nucleus , Animals , Annexin A5/pharmacology , Apoptosis , Blotting, Northern , Cell Cycle , Cell Nucleus/metabolism , Cell Survival , Cytoplasm/metabolism , Endoplasmic Reticulum/metabolism , Genes, Reporter , Glutathione Transferase/metabolism , Immunoblotting , Mice , Microscopy, Fluorescence , Models, Biological , NF-E2-Related Factor 2 , NIH 3T3 Cells , Phosphorylation , Plasmids/metabolism , Precipitin Tests , Protein Binding , Protein Biosynthesis , Protein Folding , Protein Transport , Signal Transduction , Subcellular Fractions , Time Factors , Transcription, Genetic , Two-Hybrid System Techniques
9.
J Biol Chem ; 278(35): 32692-701, 2003 Aug 29.
Article in English | MEDLINE | ID: mdl-12788914

ABSTRACT

Hsp90 complexes contain a class of co-chaperones characterized by a tetratricopeptide repeat (TPR) domain, which mediates binding to a carboxyl-terminal EEVD region in Hsp90. Among Hsp90 TPR co-chaperones in Saccharomyces cerevisiae, only Cns1 is essential. The amino terminus of Cns1, which harbors the TPR domain, is sufficient for viability when overexpressed. In a screen for temperature-sensitive alleles of CNS1, we identified mutations resulting in substitutions of conserved residues in the TPR domain. Mutations in CNS1 disrupt in vitro and in vivo interaction with Hsp90 and reduce Hsp90 function, indicating that Cns1 is a bona fide co-chaperone. Genetic interactions between CNS1 and another Hsp90 co-chaperone, CPR7, suggest that the two co-chaperones share an essential role in the cell. Although both the TPR and the isomerase domains of the cyclophilin Cpr7 are required for viability of cns1 mutant cells, this requirement does not depend on the catalytic function of the isomerase domain. Instead, hydrophilic residues on the surface of this domain appear to be important for the common Cns1.Cpr7 function. Although both co-chaperones interact with Hsp90 primarily through the carboxyl terminus (EEVD), Cns1 and Cpr7 are mostly found in complexes distinct from Hsp90. EEVD is required for normal growth in cns1 mutant cells, demonstrating for the first time in vivo requirement for this conserved region of Hsp90. Overall, our findings reveal a considerable degree of complexity in the interactions not only between Hsp90 and its co-chaperones, but also among the co-chaperones themselves.


Subject(s)
Carrier Proteins/metabolism , Cyclophilins , HSP90 Heat-Shock Proteins/chemistry , Molecular Chaperones/metabolism , Peptidylprolyl Isomerase/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/metabolism , Blotting, Western , Cell Division , Cell Survival , Centrifugation , Chromatography, Gel , Peptidyl-Prolyl Isomerase F , Escherichia coli/metabolism , Genotype , Glutathione Transferase/metabolism , HSP90 Heat-Shock Proteins/metabolism , Mutation , Phenotype , Plasmids/metabolism , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Temperature , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...