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










Publication year range
1.
Sci Rep ; 9(1): 2903, 2019 02 27.
Article in English | MEDLINE | ID: mdl-30814564

ABSTRACT

Phosphorylation of the translation initiation factor eIF2α within the mediobasal hypothalamus is known to suppress food intake, but the role of the eIF2α phosphatases in regulating body weight is poorly understood. Mice deficient in active PPP1R15A, a stress-inducible eIF2α phosphatase, are healthy and more resistant to endoplasmic reticulum stress than wild type controls. We report that when female Ppp1r15a mutant mice are fed a high fat diet they gain less weight than wild type littermates owing to reduced food intake. This results in healthy leaner Ppp1r15a mutant animals with reduced hepatic steatosis and improved insulin sensitivity, albeit with a possible modest defect in insulin secretion. By contrast, no weight differences are observed between wild type and Ppp1r15a deficient mice fed a standard diet. We conclude that female mice lacking the C-terminal PP1-binding domain of PPP1R15A show reduced dietary intake and preserved glucose tolerance. Our data indicate that this results in reduced weight gain and protection from diet-induced obesity.


Subject(s)
Hypothalamus/metabolism , Obesity/prevention & control , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Weight Gain/physiology , Animals , Diet, High-Fat , Eating , Endoplasmic Reticulum Stress , Female , Humans , Insulin Resistance , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation
3.
PLoS Pathog ; 11(6): e1004946, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26083346

ABSTRACT

Pseudomonas aeruginosa infection can be disastrous in chronic lung diseases such as cystic fibrosis and chronic obstructive pulmonary disease. Its toxic effects are largely mediated by secreted virulence factors including pyocyanin, elastase and alkaline protease (AprA). Efficient functioning of the endoplasmic reticulum (ER) is crucial for cell survival and appropriate immune responses, while an excess of unfolded proteins within the ER leads to "ER stress" and activation of the "unfolded protein response" (UPR). Bacterial infection and Toll-like receptor activation trigger the UPR most likely due to the increased demand for protein folding of inflammatory mediators. In this study, we show that cell-free conditioned medium of the PAO1 strain of P. aeruginosa, containing secreted virulence factors, induces ER stress in primary bronchial epithelial cells as evidenced by splicing of XBP1 mRNA and induction of CHOP, GRP78 and GADD34 expression. Most aspects of the ER stress response were dependent on TAK1 and p38 MAPK, except for the induction of GADD34 mRNA. Using various mutant strains and purified virulence factors, we identified pyocyanin and AprA as inducers of ER stress. However, the induction of GADD34 was mediated by an ER stress-independent integrated stress response (ISR) which was at least partly dependent on the iron-sensing eIF2α kinase HRI. Our data strongly suggest that this increased GADD34 expression served to protect against Pseudomonas-induced, iron-sensitive cell cytotoxicity. In summary, virulence factors from P. aeruginosa induce ER stress in airway epithelial cells and also trigger the ISR to improve cell survival of the host.


Subject(s)
Epithelial Cells/metabolism , Protein Phosphatase 1/metabolism , Pseudomonas Infections/metabolism , Unfolded Protein Response/physiology , Virulence Factors/metabolism , Blotting, Western , Cells, Cultured , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/physiology , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/microbiology , Epithelial Cells/pathology , Humans , Pseudomonas Infections/pathology , Pseudomonas aeruginosa , Respiratory Mucosa/metabolism , Respiratory Mucosa/microbiology , Respiratory Mucosa/pathology , Reverse Transcriptase Polymerase Chain Reaction
4.
Elife ; 42015 Mar 16.
Article in English | MEDLINE | ID: mdl-25774599

ABSTRACT

Four stress-sensing kinases phosphorylate the alpha subunit of eukaryotic translation initiation factor 2 (eIF2α) to activate the integrated stress response (ISR). In animals, the ISR is antagonised by selective eIF2α phosphatases comprising a catalytic protein phosphatase 1 (PP1) subunit in complex with a PPP1R15-type regulatory subunit. An unbiased search for additional conserved components of the PPP1R15-PP1 phosphatase identified monomeric G-actin. Like PP1, G-actin associated with the functional core of PPP1R15 family members and G-actin depletion, by the marine toxin jasplakinolide, destabilised the endogenous PPP1R15A-PP1 complex. The abundance of the ternary PPP1R15-PP1-G-actin complex was responsive to global changes in the polymeric status of actin, as was its eIF2α-directed phosphatase activity, while localised G-actin depletion at sites enriched for PPP1R15 enhanced eIF2α phosphorylation and the downstream ISR. G-actin's role as a stabilizer of the PPP1R15-containing holophosphatase provides a mechanism for integrating signals regulating actin dynamics with stresses that trigger the ISR.


Subject(s)
Actins/metabolism , Eukaryotic Initiation Factor-2/metabolism , Stress, Physiological , Amino Acid Sequence , Animals , Conserved Sequence , Depsipeptides/pharmacology , Drosophila melanogaster , HEK293 Cells , Humans , Mice , Molecular Sequence Data , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Phosphatase 1/chemistry , Stress, Physiological/drug effects
5.
J Cell Sci ; 126(Pt 6): 1406-15, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23418347

ABSTRACT

Phosphorylation of eukaryotic translation initiation factor 2 alpha (eIF2α) by the kinase GCN2 attenuates protein synthesis during amino acid starvation in yeast, whereas in mammals a family of related eIF2α kinases regulate translation in response to a variety of stresses. Unlike single-celled eukaryotes, mammals also possess two specific eIF2α phosphatases, PPP1R15a and PPP1R15b, whose combined deletion leads to a poorly understood early embryonic lethality. We report the characterisation of the first non-mammalian eIF2α phosphatase and the use of Drosophila to dissect its role during development. The Drosophila protein demonstrates features of both mammalian proteins, including limited sequence homology and association with the endoplasmic reticulum. Of note, although this protein is not transcriptionally regulated, its expression is controlled by the presence of upstream open reading frames in its 5'UTR, enabling induction in response to eIF2α phosphorylation. Moreover, we show that its expression is necessary for embryonic and larval development and that this is to oppose the inhibitory effects of GCN2 on anabolic growth.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Eukaryotic Initiation Factor-2/metabolism , Protein Kinases/metabolism , Protein Phosphatase 1/metabolism , 5' Untranslated Regions/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , COS Cells , Chlorocebus aethiops , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Embryo, Nonmammalian , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Eukaryotic Initiation Factor-2/genetics , HEK293 Cells , Humans , Molecular Sequence Data , Phosphorylation/genetics , Protein Kinases/genetics , Protein Phosphatase 1/genetics , RNA Processing, Post-Transcriptional/genetics , Sequence Homology, Amino Acid
6.
J Biol Chem ; 288(11): 7606-7617, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23341460

ABSTRACT

Cell cycle checkpoints ensure that proliferation occurs only under permissive conditions, but their role in linking nutrient availability to cell division is incompletely understood. Protein folding within the endoplasmic reticulum (ER) is exquisitely sensitive to energy supply and amino acid sources because deficiencies impair luminal protein folding and consequently trigger ER stress signaling. Following ER stress, many cell types arrest within the G(1) phase, although recent studies have identified a novel ER stress G(2) checkpoint. Here, we report that ER stress affects cell cycle progression via two classes of signal: an early inhibition of protein synthesis leading to G(2) delay involving CHK1 and a later induction of G(1) arrest associated both with the induction of p53 target genes and loss of cyclin D(1). We show that substitution of p53/47 for p53 impairs the ER stress G(1) checkpoint, attenuates the recovery of protein translation, and impairs induction of NOXA, a mediator of cell death. We propose that cell cycle regulation in response to ER stress comprises redundant pathways invoked sequentially first to impair G(2) progression prior to ultimate G(1) arrest.


Subject(s)
Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Genes, p53 , Tumor Suppressor Protein p53/genetics , Animals , Cell Cycle , Cell Line , Cell Line, Tumor , Cell Proliferation , Cell Separation , Drosophila melanogaster , Flow Cytometry , HEK293 Cells , HeLa Cells , Humans , Plasmids/metabolism , Protein Biosynthesis , Protein Phosphatase 1/metabolism , RNA Interference , Tumor Suppressor Protein p53/metabolism
7.
J Invest Dermatol ; 133(6): 1572-81, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23337888

ABSTRACT

Deregulated Ras signaling initiates and maintains melanocyte neoplasia. The Rho-like GTPase Rac has been implicated in Ras-induced neoplastic transformation. Moreover, a recurrent UV-induced mutation activating RAC1 has recently been detected in human melanoma. Here, a role for Rac in melanoma initiation and progression was investigated in human melanomas and zebrafish models of melanocyte neoplasia. Immunohistochemical analysis revealed RAC expression and activity restricted to melanocytes at the junction of the epidermis and dermis in benign neoplasms. Malignant melanocytes displayed elevated RAC activity that extended into the suprabasal epidermis, deeper into the dermis, and was maintained in metastases. Previously, we have used zebrafish transgenic models to demonstrate that deregulated Ras/Raf/mitogen-activated protein kinase signaling can initiate melanocyte neoplasia. Expression of a constitutively active RAC1 mutant (V12RAC1) was not sufficient to initiate melanocyte neoplasia in this organism. Furthermore, we did not detect an additive effect when combined with V600EBRAF, nor could V12RAC1 substitute for suppressed Pi3k signaling to restore melanoma progression. However, coexpression of V12RAC1 and oncogenic RAS accelerated tumor nodule formation. Immunohistochemical analysis revealed that the Rac activator Tiam1 (T-cell lymphoma invasion and metastasis 1) is overexpressed in melanoma tumor nodules in both zebrafish and humans. Thus, our data suggest that Rac contributes to the progression of melanoma and that Tiam1 may activate Rac in nodular presentations.


Subject(s)
Melanocytes/pathology , Melanoma/pathology , Signal Transduction/physiology , Skin Neoplasms/pathology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , Animals , Animals, Genetically Modified , Cell Line, Tumor , Disease Progression , Fibroblasts/cytology , Fibroblasts/physiology , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , Humans , Melanocytes/physiology , Melanoma/metabolism , Melanoma/physiopathology , Mice , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/physiopathology , Skin Neoplasms/metabolism , Skin Neoplasms/physiopathology , T-Lymphoma Invasion and Metastasis-inducing Protein 1 , Zebrafish
8.
Prog Mol Biol Transl Sci ; 106: 189-221, 2012.
Article in English | MEDLINE | ID: mdl-22340719

ABSTRACT

The integrated stress response (ISR) is an evolutionarily conserved homeostatic program activated by specific pathological states. These include amino acid deprivation, viral infection, iron deficiency, and the misfolding of proteins within the endoplasmic reticulum (ER), the so-called ER stress. Although apparently disparate, each of these stresses induces phosphorylation of a translation initiation factor, eIF2α, to attenuate new protein translation while simultaneously triggering a transcriptional program. This is achieved by four homologous stress-sensing kinases: GCN2, PKR, HRI, and PERK. In addition to these kinases, mammals possess two specific eIF2α phosphatases, GADD34 and CReP, which play crucial roles in the recovery of protein synthesis following the initial insult. They are not only important in embryonic development but also appear to play important roles in disease, particularly cancer. In this chapter, we discuss each of the eIF2α kinases, in turn, with particular emphasis on their regulation and the new insights provided by recent structural studies. We also discuss the potential for developing novel drug therapies that target the ISR.


Subject(s)
Phosphoproteins/physiology , Protein Processing, Post-Translational , Stress, Physiological/physiology , Amino Acids/metabolism , Animals , Endoplasmic Reticulum Stress/physiology , Eukaryotic Initiation Factor-2/physiology , Evolution, Molecular , Heme/metabolism , Humans , Mice , Mice, Knockout , Models, Molecular , Phosphorylation/drug effects , Protein Biosynthesis/drug effects , Protein Biosynthesis/physiology , Protein Conformation , Protein Phosphatase 1/physiology , Protein Processing, Post-Translational/drug effects , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/physiology , Signal Transduction/physiology , Stress, Physiological/drug effects , Structure-Activity Relationship , Transcription Factor CHOP/deficiency , Transcription Factor CHOP/physiology , Transcription, Genetic/physiology , Virus Diseases/enzymology , eIF-2 Kinase/chemistry , eIF-2 Kinase/physiology
9.
Methods Enzymol ; 501: 421-66, 2011.
Article in English | MEDLINE | ID: mdl-22078544

ABSTRACT

The serpinopathies result from point mutations in members of the serine protease inhibitor or serpin superfamily. They are characterized by the formation of ordered polymers that are retained within the cell of synthesis. This causes disease by a "toxic gain of function" from the accumulated protein and a "loss of function" as a result of the deficiency of inhibitors that control important proteolytic cascades. The serpinopathies are exemplified by the Z (Glu342Lys) mutant of α1-antitrypsin that results in the retention of ordered polymers within the endoplasmic reticulum of hepatocytes. These polymers form the intracellular inclusions that are associated with neonatal hepatitis, cirrhosis, and hepatocellular carcinoma. A second example results from mutations in the neurone-specific serpin-neuroserpin to form ordered polymers that are retained as inclusions within subcortical neurones as Collins' bodies. These inclusions underlie the autosomal dominant dementia familial encephalopathy with neuroserpin inclusion bodies or FENIB. There are different pathways to polymer formation in vitro but not all form polymers that are relevant in vivo. It is therefore essential that protein-based structural studies are interpreted in the context of human samples and cell and animal models of disease. We describe here the biochemical techniques, monoclonal antibodies, cell biology, animal models, and stem cell technology that are useful to characterize the serpin polymers that form in vivo.


Subject(s)
Biophysics/methods , Epilepsies, Myoclonic/metabolism , Heredodegenerative Disorders, Nervous System/metabolism , Image Processing, Computer-Assisted/methods , Lung/metabolism , Neuropeptides/metabolism , Point Mutation , Serpins/metabolism , alpha 1-Antitrypsin/metabolism , Animals , Cell Culture Techniques , Cell Line , Epilepsies, Myoclonic/genetics , Epilepsies, Myoclonic/pathology , Heredodegenerative Disorders, Nervous System/genetics , Heredodegenerative Disorders, Nervous System/pathology , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Lung/pathology , Mice , Mice, Transgenic , Microscopy, Electron , Neuropeptides/chemistry , Neuropeptides/genetics , Neutrophils/cytology , Neutrophils/metabolism , Peptide Fragments , Polymerization , Protein Binding , Protein Conformation , Proteolysis , Serpins/chemistry , Serpins/genetics , Transfection , alpha 1-Antitrypsin/chemistry , alpha 1-Antitrypsin/genetics , Neuroserpin
10.
Diabetes Metab Res Rev ; 26(8): 611-21, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20922715

ABSTRACT

Endoplasmic reticulum (ER) stress is an integral part of life for all professional secretory cells, but it has been studied to greatest depth in the pancreatic ß-cell. This reflects both the crucial role played by ER stress in the pathogenesis of diabetes and also the exquisite vulnerability of these cells to ER dysfunction. The adaptive cellular response to ER stress, the unfolded protein response, comprises mechanisms to both regulate new protein translation and a transcriptional program to allow adaptation to the stress. The core of this response is a triad of stress-sensing proteins: protein kinase R-like endoplasmic reticulum kinase (PERK), inositol-requiring enzyme 1 (IRE1) and activating transcription factor 6. All three regulate portions of the transcriptional unfolded protein response, while PERK also attenuates protein synthesis during ER stress and IRE1 interacts directly with the c-Jun amino-terminal kinase stress kinase pathway. In this review we shall discuss these processes in detail, with emphasis given to their impact on diabetes and how recent findings indicate that ER stress may be responsible for the loss of ß-cell mass in the disease.


Subject(s)
Activating Transcription Factor 6/physiology , Diabetes Mellitus/physiopathology , Endoplasmic Reticulum/physiology , Endoribonucleases/physiology , Islets of Langerhans/physiology , Protein Serine-Threonine Kinases/physiology , Stress, Physiological/physiology , Animals , DNA-Binding Proteins/physiology , Diabetes Mellitus, Type 1/physiopathology , Epiphyses/abnormalities , Epiphyses/physiopathology , Glycoproteins/physiology , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/physiology , Mice , Osteochondrodysplasias/physiopathology , Oxidoreductases , Regulatory Factor X Transcription Factors , Transcription Factors/physiology , Unfolded Protein Response
SELECTION OF CITATIONS
SEARCH DETAIL
...