Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
Proc Natl Acad Sci U S A ; 121(14): e2320013121, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38547060

ABSTRACT

Dephosphorylation of pSer51 of the α subunit of translation initiation factor 2 (eIF2αP) terminates signaling in the integrated stress response (ISR). A trimeric mammalian holophosphatase comprised of a protein phosphatase 1 (PP1) catalytic subunit, the conserved C-terminally located ~70 amino acid core of a substrate-specific regulatory subunit (PPP1R15A/GADD34 or PPP1R15B/CReP) and G-actin (an essential cofactor) efficiently dephosphorylate eIF2αP in vitro. Unlike their viral or invertebrate counterparts, with whom they share the conserved 70 residue core, the mammalian PPP1R15s are large proteins of more than 600 residues. Genetic and cellular observations point to a functional role for regions outside the conserved core of mammalian PPP1R15A in dephosphorylating its natural substrate, the eIF2 trimer. We have combined deep learning technology, all-atom molecular dynamics simulations, X-ray crystallography, and biochemistry to uncover binding of the γ subunit of eIF2 to a short helical peptide repeated four times in the functionally important N terminus of human PPP1R15A that extends past its conserved core. Binding entails insertion of Phe and Trp residues that project from one face of an α-helix formed by the conserved repeats of PPP1R15A into a hydrophobic groove exposed on the surface of eIF2γ in the eIF2 trimer. Replacing these conserved Phe and Trp residues with Ala compromises PPP1R15A function in cells and in vitro. These findings suggest mechanisms by which contacts between a distant subunit of eIF2 and elements of PPP1R15A distant to the holophosphatase active site contribute to dephosphorylation of eIF2αP by the core PPP1R15 holophosphatase and to efficient termination of the ISR in mammals.


Subject(s)
Eukaryotic Initiation Factor-2 , Protein Processing, Post-Translational , Animals , Humans , Actins/metabolism , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Phosphorylation , Protein Phosphatase 1/metabolism
2.
EMBO J ; 43(5): 719-753, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38177498

ABSTRACT

Effector mechanisms of the unfolded protein response (UPR) in the endoplasmic reticulum (ER) are well-characterised, but how ER proteostasis is sensed is less well understood. Here, we exploited the beta isoform of the UPR transducer IRE1, that is specific to mucin-producing cells in order to gauge the relative regulatory roles of activating ligands and repressing chaperones of the specialised ER of goblet cells. Replacement of the stress-sensing luminal domain of endogenous IRE1α in CHO cells (normally expressing neither mucin nor IRE1ß) with the luminal domain of IRE1ß deregulated basal IRE1 activity. The mucin-specific chaperone AGR2 repressed IRE1 activity in cells expressing the domain-swapped IRE1ß/α chimera, but had no effect on IRE1α. Introduction of the goblet cell-specific client MUC2 reversed AGR2-mediated repression of the IRE1ß/α chimera. In vitro, AGR2 actively de-stabilised the IRE1ß luminal domain dimer and formed a reversible complex with the inactive monomer. These features of the IRE1ß-AGR2 couple suggest that active repression of IRE1ß by a specialised mucin chaperone subordinates IRE1 activity to a proteostatic challenge unique to goblet cells, a challenge that is otherwise poorly recognised by the pervasive UPR transducers.


Subject(s)
Endoribonucleases , Goblet Cells , Mucins , Animals , Cricetinae , Humans , Cricetulus , Goblet Cells/metabolism , Molecular Chaperones/genetics , Mucins/genetics , Mucoproteins/genetics , Oncogene Proteins , Protein Serine-Threonine Kinases/genetics , CHO Cells
3.
EMBO Mol Med ; 15(3): e16491, 2023 03 08.
Article in English | MEDLINE | ID: mdl-36704923

ABSTRACT

Dysfunction of the endoplasmic reticulum (ER) in insulin-producing beta cells results in cell loss and diabetes mellitus. Here we report on five individuals from three different consanguineous families with infancy-onset diabetes mellitus and severe neurodevelopmental delay caused by a homozygous p.(Arg371Ser) mutation in FICD. The FICD gene encodes a bifunctional Fic domain-containing enzyme that regulates the ER Hsp70 chaperone, BiP, via catalysis of two antagonistic reactions: inhibitory AMPylation and stimulatory deAMPylation of BiP. Arg371 is a conserved residue in the Fic domain active site. The FICDR371S mutation partially compromises BiP AMPylation in vitro but eliminates all detectable deAMPylation activity. Overexpression of FICDR371S or knock-in of the mutation at the FICD locus of stressed CHO cells results in inappropriately elevated levels of AMPylated BiP and compromised secretion. These findings, guided by human genetics, highlight the destructive consequences of de-regulated BiP AMPylation and raise the prospect of tuning FICD's antagonistic activities towards therapeutic ends.


Subject(s)
Diabetes Mellitus , Endoplasmic Reticulum Chaperone BiP , Animals , Cricetinae , Humans , Infant , Protein Processing, Post-Translational , Cricetulus , Adenosine Monophosphate
4.
Nat Struct Mol Biol ; 28(10): 835-846, 2021 10.
Article in English | MEDLINE | ID: mdl-34625748

ABSTRACT

Many regulatory PPP1R subunits join few catalytic PP1c subunits to mediate phosphoserine and phosphothreonine dephosphorylation in metazoans. Regulatory subunits engage the surface of PP1c, locally affecting flexible access of the phosphopeptide to the active site. However, catalytic efficiency of holophosphatases towards their phosphoprotein substrates remains unexplained. Here we present a cryo-EM structure of the tripartite PP1c-PPP1R15A-G-actin holophosphatase that terminates signaling in the mammalian integrated stress response (ISR) in the pre-dephosphorylation complex with its substrate, translation initiation factor 2α (eIF2α). G-actin, whose essential role in eIF2α dephosphorylation is supported crystallographically, biochemically and genetically, aligns the catalytic and regulatory subunits, creating a composite surface that engages the N-terminal domain of eIF2α to position the distant phosphoserine-51 at the active site. Substrate residues that mediate affinity for the holophosphatase also make critical contacts with eIF2α kinases. Thus, a convergent process of higher-order substrate recognition specifies functionally antagonistic phosphorylation and dephosphorylation in the ISR.


Subject(s)
Protein Phosphatase 1/chemistry , Protein Phosphatase 1/metabolism , Stress, Physiological/physiology , eIF-2 Kinase/metabolism , Actins/chemistry , Actins/metabolism , Animals , CHO Cells , Catalytic Domain , Cricetulus , Cryoelectron Microscopy , Crystallography, X-Ray , Humans , Models, Molecular , Phosphorylation , Phosphoserine/metabolism , Protein Phosphatase 1/genetics , Reproducibility of Results , eIF-2 Kinase/genetics
5.
Cell Rep ; 35(7): 109144, 2021 05 18.
Article in English | MEDLINE | ID: mdl-34010647

ABSTRACT

Circulating polymers of α1-antitrypsin (α1AT) are neutrophil chemo-attractants and contribute to inflammation, yet cellular factors affecting their secretion remain obscure. We report on a genome-wide CRISPR-Cas9 screen for genes affecting trafficking of polymerogenic α1ATH334D. A CRISPR enrichment approach based on recovery of single guide RNA (sgRNA) sequences from phenotypically selected fixed cells reveals that cells with high-polymer content are enriched in sgRNAs targeting genes involved in "cargo loading into COPII-coated vesicles," where "COPII" is coat protein II, including the cargo receptors lectin mannose binding1 (LMAN1) and surfeit protein locus 4 (SURF4). LMAN1- and SURF4-disrupted cells display a secretion defect extending beyond α1AT monomers to polymers. Polymer secretion is especially dependent on SURF4 and correlates with a SURF4-α1ATH334D physical interaction and with their co-localization at the endoplasmic reticulum (ER). These findings indicate that ER cargo receptors co-ordinate progression of α1AT out of the ER and modulate the accumulation of polymeric α1AT not only by controlling the concentration of precursor monomers but also by promoting secretion of polymers.


Subject(s)
Endoplasmic Reticulum/metabolism , Polymers/metabolism , alpha 1-Antitrypsin/metabolism , Humans
6.
Mol Cell ; 81(1): 88-103.e6, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33220178

ABSTRACT

The small molecule ISRIB antagonizes the activation of the integrated stress response (ISR) by phosphorylated translation initiation factor 2, eIF2(αP). ISRIB and eIF2(αP) bind distinct sites in their common target, eIF2B, a guanine nucleotide exchange factor for eIF2. We have found that ISRIB-mediated acceleration of eIF2B's nucleotide exchange activity in vitro is observed preferentially in the presence of eIF2(αP) and is attenuated by mutations that desensitize eIF2B to the inhibitory effect of eIF2(αP). ISRIB's efficacy as an ISR inhibitor in cells also depends on presence of eIF2(αP). Cryoelectron microscopy (cryo-EM) showed that engagement of both eIF2B regulatory sites by two eIF2(αP) molecules remodels both the ISRIB-binding pocket and the pockets that would engage eIF2α during active nucleotide exchange, thereby discouraging both binding events. In vitro, eIF2(αP) and ISRIB reciprocally opposed each other's binding to eIF2B. These findings point to antagonistic allostery in ISRIB action on eIF2B, culminating in inhibition of the ISR.


Subject(s)
Acetamides/chemistry , Cyclohexylamines/chemistry , Eukaryotic Initiation Factor-2B/chemistry , Eukaryotic Initiation Factor-2/chemistry , Allosteric Regulation , Animals , Binding Sites , CHO Cells , Cricetulus , Cryoelectron Microscopy , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-2B/genetics , Eukaryotic Initiation Factor-2B/metabolism , HeLa Cells , Humans , Phosphorylation
8.
Nature ; 578(7795): 444-448, 2020 02.
Article in English | MEDLINE | ID: mdl-31875646

ABSTRACT

Metformin, the world's most prescribed anti-diabetic drug, is also effective in preventing type 2 diabetes in people at high risk1,2. More than 60% of this effect is attributable to the ability of metformin to lower body weight in a sustained manner3. The molecular mechanisms by which metformin lowers body weight are unknown. Here we show-in two independent randomized controlled clinical trials-that metformin increases circulating levels of the peptide hormone growth/differentiation factor 15 (GDF15), which has been shown to reduce food intake and lower body weight through a brain-stem-restricted receptor. In wild-type mice, oral metformin increased circulating GDF15, with GDF15 expression increasing predominantly in the distal intestine and the kidney. Metformin prevented weight gain in response to a high-fat diet in wild-type mice but not in mice lacking GDF15 or its receptor GDNF family receptor α-like (GFRAL). In obese mice on a high-fat diet, the effects of metformin to reduce body weight were reversed by a GFRAL-antagonist antibody. Metformin had effects on both energy intake and energy expenditure that were dependent on GDF15, but retained its ability to lower circulating glucose levels in the absence of GDF15 activity. In summary, metformin elevates circulating levels of GDF15, which is necessary to obtain its beneficial effects on energy balance and body weight, major contributors to its action as a chemopreventive agent.


Subject(s)
Body Weight/drug effects , Energy Metabolism/drug effects , Growth Differentiation Factor 15/metabolism , Metformin/pharmacology , Administration, Oral , Adult , Aged , Animals , Blood Glucose/analysis , Blood Glucose/metabolism , Diet, High-Fat , Double-Blind Method , Energy Intake/drug effects , Enterocytes/cytology , Enterocytes/drug effects , Female , Glial Cell Line-Derived Neurotrophic Factor Receptors/antagonists & inhibitors , Glial Cell Line-Derived Neurotrophic Factor Receptors/deficiency , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Growth Differentiation Factor 15/blood , Growth Differentiation Factor 15/deficiency , Growth Differentiation Factor 15/genetics , Homeostasis/drug effects , Humans , Intestines/cytology , Intestines/drug effects , Male , Metformin/administration & dosage , Mice , Mice, Obese , Middle Aged , Weight Loss/drug effects
9.
Elife ; 82019 11 21.
Article in English | MEDLINE | ID: mdl-31749445

ABSTRACT

The eukaryotic translation initiation factor 2α (eIF2α) kinase GCN2 is activated by amino acid starvation to elicit a rectifying physiological program known as the Integrated Stress Response (ISR). A role for uncharged tRNAs as activating ligands of yeast GCN2 is supported experimentally. However, mouse GCN2 activation has recently been observed in circumstances associated with ribosome stalling with no global increase in uncharged tRNAs. We report on a mammalian CHO cell-based CRISPR-Cas9 mutagenesis screen for genes that contribute to ISR activation by amino acid starvation. Disruption of genes encoding components of the ribosome P-stalk, uL10 and P1, selectively attenuated GCN2-mediated ISR activation by amino acid starvation or interference with tRNA charging without affecting the endoplasmic reticulum unfolded protein stress-induced ISR, mediated by the related eIF2α kinase PERK. Wildtype ribosomes isolated from CHO cells, but not those with P-stalk lesions, stimulated GCN2-dependent eIF2α phosphorylation in vitro. These observations support a model whereby lack of a cognate charged tRNA exposes a latent capacity of the ribosome P-stalk to activate GCN2 in cells and help explain the emerging link between ribosome stalling and ISR activation.


Subject(s)
Amino Acids/metabolism , Protein Serine-Threonine Kinases/metabolism , Ribosomes/metabolism , Starvation/metabolism , Animals , CHO Cells , CRISPR-Cas Systems , Cricetulus , Endoplasmic Reticulum/metabolism , Gene Expression Regulation, Enzymologic , HeLa Cells , Humans , Kinetics , Ligands , Mice , Models, Molecular , Mutagenesis , Phosphorylation , Protein Binding , Protein Conformation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Unfolding , RNA, Transfer/metabolism , Ribosomes/chemistry , Signal Transduction , Transcriptome , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
10.
Cell Metab ; 29(3): 707-718.e8, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30639358

ABSTRACT

GDF15 is an established biomarker of cellular stress. The fact that it signals via a specific hindbrain receptor, GFRAL, and that mice lacking GDF15 manifest diet-induced obesity suggest that GDF15 may play a physiological role in energy balance. We performed experiments in humans, mice, and cells to determine if and how nutritional perturbations modify GDF15 expression. Circulating GDF15 levels manifest very modest changes in response to moderate caloric surpluses or deficits in mice or humans, differentiating it from classical intestinally derived satiety hormones and leptin. However, GDF15 levels do increase following sustained high-fat feeding or dietary amino acid imbalance in mice. We demonstrate that GDF15 expression is regulated by the integrated stress response and is induced in selected tissues in mice in these settings. Finally, we show that pharmacological GDF15 administration to mice can trigger conditioned taste aversion, suggesting that GDF15 may induce an aversive response to nutritional stress.


Subject(s)
Energy Intake/physiology , Growth Differentiation Factor 15/metabolism , Adult , Animals , Cell Line , Diet, High-Fat/methods , Growth Differentiation Factor 15/pharmacology , Humans , Mice , Mice, Inbred C57BL , Middle Aged , Young Adult
12.
Science ; 359(6383): 1533-1536, 2018 03 30.
Article in English | MEDLINE | ID: mdl-29599245

ABSTRACT

The integrated stress response (ISR) is a conserved translational and transcriptional program affecting metabolism, memory, and immunity. The ISR is mediated by stress-induced phosphorylation of eukaryotic translation initiation factor 2α (eIF2α) that attenuates the guanine nucleotide exchange factor eIF2B. A chemical inhibitor of the ISR, ISRIB, reverses the attenuation of eIF2B by phosphorylated eIF2α, protecting mice from neurodegeneration and traumatic brain injury. We describe a 4.1-angstrom-resolution cryo-electron microscopy structure of human eIF2B with an ISRIB molecule bound at the interface between the ß and δ regulatory subunits. Mutagenesis of residues lining this pocket altered the hierarchical cellular response to ISRIB analogs in vivo and ISRIB binding in vitro. Our findings point to a site in eIF2B that can be exploited by ISRIB to regulate translation.


Subject(s)
Acetamides/chemistry , Cyclohexylamines/chemistry , Eukaryotic Initiation Factor-2B/chemistry , Acetamides/pharmacology , Animals , Cryoelectron Microscopy , Cyclohexylamines/pharmacology , Eukaryotic Initiation Factor-2B/genetics , HeLa Cells , Humans , Mice , Mutagenesis , Phosphorylation , Protein Binding , Protein Biosynthesis/drug effects , Protein Conformation , Stress, Physiological/drug effects
13.
Cell ; 171(7): 1625-1637.e13, 2017 Dec 14.
Article in English | MEDLINE | ID: mdl-29198525

ABSTRACT

When unfolded proteins accumulate in the endoplasmic reticulum (ER), the unfolded protein response (UPR) increases ER-protein-folding capacity to restore protein-folding homeostasis. Unfolded proteins activate UPR signaling across the ER membrane to the nucleus by promoting oligomerization of IRE1, a conserved transmembrane ER stress receptor. However, the coupling of ER stress to IRE1 oligomerization and activation has remained obscure. Here, we report that the ER luminal co-chaperone ERdj4/DNAJB9 is a selective IRE1 repressor that promotes a complex between the luminal Hsp70 BiP and the luminal stress-sensing domain of IRE1α (IRE1LD). In vitro, ERdj4 is required for complex formation between BiP and IRE1LD. ERdj4 associates with IRE1LD and recruits BiP through the stimulation of ATP hydrolysis, forcibly disrupting IRE1 dimers. Unfolded proteins compete for BiP and restore IRE1LD to its default, dimeric, and active state. These observations establish BiP and its J domain co-chaperones as key regulators of the UPR.


Subject(s)
Endoribonucleases/metabolism , HSP40 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Membrane Proteins/metabolism , Molecular Chaperones/metabolism , Protein Serine-Threonine Kinases/metabolism , Unfolded Protein Response , Animals , Cricetinae , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Humans , Protein Folding
14.
J Exp Med ; 214(2): 401-422, 2017 02.
Article in English | MEDLINE | ID: mdl-28082357

ABSTRACT

ATG16L1T300A, a major risk polymorphism in Crohn's disease (CD), causes impaired autophagy, but it has remained unclear how this predisposes to CD. In this study, we report that mice with Atg16l1 deletion in intestinal epithelial cells (IECs) spontaneously develop transmural ileitis phenocopying ileal CD in an age-dependent manner, driven by the endoplasmic reticulum (ER) stress sensor IRE1α. IRE1α accumulates in Paneth cells of Atg16l1ΔIEC mice, and humans homozygous for ATG16L1T300A exhibit a corresponding increase of IRE1α in intestinal epithelial crypts. In contrast to a protective role of the IRE1ß isoform, hyperactivated IRE1α also drives a similar ileitis developing earlier in life in Atg16l1;Xbp1ΔIEC mice, in which ER stress is induced by deletion of the unfolded protein response transcription factor XBP1. The selective autophagy receptor optineurin interacts with IRE1α, and optineurin deficiency amplifies IRE1α levels during ER stress. Furthermore, although dysbiosis of the ileal microbiota is present in Atg16l1;Xbp1ΔIEC mice as predicted from impaired Paneth cell antimicrobial function, such structural alteration of the microbiota does not trigger ileitis but, rather, aggravates dextran sodium sulfate-induced colitis. Hence, we conclude that defective autophagy in IECs may predispose to CD ileitis via impaired clearance of IRE1α aggregates during ER stress at this site.


Subject(s)
Autophagy-Related Proteins/physiology , Crohn Disease/etiology , Endoribonucleases/physiology , Ileitis/etiology , Protein Serine-Threonine Kinases/physiology , Age Factors , Animals , Autophagy , Endoplasmic Reticulum Stress , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Microbiota
15.
Oncotarget ; 7(39): 64124-64135, 2016 09 27.
Article in English | MEDLINE | ID: mdl-27802424

ABSTRACT

In response to endoplasmic reticulum (ER) stress, activation of pancreatic ER kinase (PERK) coordinates an adaptive program known as the integrated stress response (ISR) by phosphorylating translation initiation factor 2α (eIF2α). Phosphorylated eIF2α is quickly dephosphorylated by the protein phosphatase 1 and growth arrest and DNA damage 34 (GADD34) complex. Data indicate that the ISR can either promote or suppress tumor development. Our previous studies showed that the ISR is activated in medulloblastoma in both human patients and animal models, and that the decreased ISR via PERK heterozygous deficiency attenuates medulloblastoma formation in Patched1 heterozygous deficient (Ptch1+/-) mice by enhancing apoptosis of pre-malignant granule cell precursors (GCPs) during cell transformation. We showed here that GADD34 heterozygous mutation moderately enhanced the ISR and noticeably increased the incidence of medulloblastoma in adult Ptch1+/- mice. Surprisingly, GADD34 homozygous mutation strongly enhanced the ISR, but significantly decreased the incidence of medulloblastoma in adult Ptch1+/- mice. Intriguingly, GADD34 homozygous mutation significantly enhanced pre-malignant GCP apoptosis in cerebellar hyperplastic lesions and reduced the lesion numbers in young Ptch1+/- mice. Nevertheless, neither GADD34 heterozygous mutation nor GADD34 homozygous mutation had a significant effect on medulloblastoma cells in adult Ptch1+/- mice. Collectively, these data imply the dual role of the ISR, promoting and inhibiting, in medulloblastoma tumorigenesis by regulating apoptosis of pre-malignant GCPs during the course of malignant transformation.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cerebellar Neoplasms/enzymology , Endoplasmic Reticulum Stress , Eukaryotic Initiation Factor-2/metabolism , Medulloblastoma/enzymology , Protein Phosphatase 1/metabolism , eIF-2 Kinase/metabolism , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Enzyme Activation , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Homozygote , Humans , Medulloblastoma/genetics , Medulloblastoma/pathology , Mice, Inbred C57BL , Mice, Knockout , Mutation , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Neovascularization, Pathologic , Patched-1 Receptor/deficiency , Patched-1 Receptor/genetics , Phenotype , Phosphorylation , Protein Phosphatase 1/deficiency , Protein Phosphatase 1/genetics , Signal Transduction , Time Factors
16.
PLoS One ; 11(11): e0166278, 2016.
Article in English | MEDLINE | ID: mdl-27812215

ABSTRACT

The eukaryotic translation initiation factor eIF2B promotes mRNA translation as a guanine nucleotide exchange factor (GEF) for translation initiation factor 2 (eIF2). Endoplasmic reticulum (ER) stress-mediated activation of the kinase PERK and the resultant phosphorylation of eIF2's alpha subunit (eIF2α) attenuates eIF2B GEF activity thereby inducing an integrated stress response (ISR) that defends against protein misfolding in the ER. Mutations in all five subunits of human eIF2B cause an inherited leukoencephalopathy with vanishing white matter (VWM), but the role of the ISR in its pathogenesis remains unclear. Using CRISPR-Cas9 genome editing we introduced the most severe known VWM mutation, EIF2B4A391D, into CHO cells. Compared to isogenic wildtype cells, GEF activity of cells with the VWM mutation was impaired and the mutant cells experienced modest enhancement of the ISR. However, despite their enhanced ISR, imposed by the intrinsic defect in eIF2B, disrupting the inhibitory effect of phosphorylated eIF2α on GEF by a contravening EIF2S1/eIF2αS51A mutation that functions upstream of eIF2B, selectively enfeebled both EIF2B4A391D and the related severe VWM EIF2B4R483W cells. The basis for paradoxical dependence of cells with the VWM mutations on an intact eIF2α genotype remains unclear, as both translation rates and survival from stressors that normally activate the ISR were not reproducibly affected by the VWM mutations. Nonetheless, our findings support an additional layer of complexity in the development of VWM, beyond a hyperactive ISR.


Subject(s)
Endoplasmic Reticulum Stress/genetics , Eukaryotic Initiation Factor-2B/genetics , Mutation , White Matter/cytology , Amino Acid Sequence , Animals , Base Sequence , CHO Cells , Cell Line , Cricetinae , Cricetulus , Eukaryotic Initiation Factor-2B/chemistry , Humans , Recombination, Genetic , Unfolded Protein Response/genetics , White Matter/metabolism
17.
Am J Pathol ; 186(7): 1939-1951, 2016 07.
Article in English | MEDLINE | ID: mdl-27181404

ABSTRACT

Evidence suggests that activation of pancreatic endoplasmic reticulum kinase (PERK) signaling in response to endoplasmic reticulum stress negatively or positively influences cell transformation by regulating apoptosis. Patched1 heterozygous deficient (Ptch1(+/-)) mice reproduce human Gorlin's syndrome and are regarded as the best animal model to study tumorigenesis of the sonic hedgehog subgroup of medulloblastomas. It is believed that medulloblastomas in Ptch1(+/-) mice results from the transformation of granule cell precursors (GCPs) in the developing cerebellum. Here, we determined the role of PERK signaling on medulloblastoma tumorigenesis by assessing its effects on premalignant GCPs and tumor cells. We found that PERK signaling was activated in both premalignant GCPs in young Ptch1(+/-) mice and medulloblastoma cells in adult mice. We demonstrated that PERK haploinsufficiency reduced the incidence of medulloblastomas in Ptch1(+/-) mice. Interestingly, PERK haploinsufficiency enhanced apoptosis of premalignant GCPs in young Ptch1(+/-) mice but had no significant effect on medulloblastoma cells in adult mice. Moreover, we showed that the PERK pathway was activated in medulloblastomas in humans. These results suggest that PERK signaling promotes medulloblastoma tumorigenesis by attenuating apoptosis of premalignant GCPs during the course of malignant transformation.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cerebellar Neoplasms/pathology , Medulloblastoma/pathology , Neural Stem Cells/pathology , eIF-2 Kinase/metabolism , Adult , Animals , Apoptosis , Blotting, Western , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Transformation, Neoplastic/pathology , Cerebellar Neoplasms/enzymology , Child , Child, Preschool , Disease Models, Animal , Enzyme Activation/physiology , Female , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Infant , Male , Medulloblastoma/enzymology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Mutant Strains , Neurons/pathology , Real-Time Polymerase Chain Reaction
18.
FASEB J ; 30(2): 798-812, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26487695

ABSTRACT

The eukaryotic translation initiation factor 2α (eIF2α) phosphorylation-dependent integrated stress response (ISR), a component of the unfolded protein response, has long been known to regulate intermediary metabolism, but the details are poorly worked out. We report that profiling of mRNAs of transgenic mice harboring a ligand-activated skeletal muscle-specific derivative of the eIF2α protein kinase R-like ER kinase revealed the expected up-regulation of genes involved in amino acid biosynthesis and transport but also uncovered the induced expression and secretion of a myokine, fibroblast growth factor 21 (FGF21), that stimulates energy consumption and prevents obesity. The link between the ISR and FGF21 expression was further reinforced by the identification of a small-molecule ISR activator that promoted Fgf21 expression in cell-based screens and by implication of the ISR-inducible activating transcription factor 4 in the process. Our findings establish that eIF2α phosphorylation regulates not only cell-autonomous proteostasis and amino acid metabolism, but also affects non-cell-autonomous metabolic regulation by induced expression of a potent myokine.


Subject(s)
Amino Acids/metabolism , Energy Metabolism/physiology , Eukaryotic Initiation Factor-2/metabolism , Fibroblast Growth Factors/biosynthesis , Gene Expression Regulation/physiology , Muscle, Skeletal/metabolism , Unfolded Protein Response/physiology , Amino Acids/genetics , Animals , Eukaryotic Initiation Factor-2/genetics , Fibroblast Growth Factors/genetics , Humans , Mice , Mice, Transgenic , Muscle, Skeletal/cytology , Phosphorylation/genetics
19.
Elife ; 4: e08961, 2015 Oct 16.
Article in English | MEDLINE | ID: mdl-26473973

ABSTRACT

DnaK/Hsp70 chaperones form oligomers of poorly understood structure and functional significance. Site-specific proteolysis and crosslinking were used to probe the architecture of oligomers formed by the endoplasmic reticulum (ER) Hsp70, BiP. These were found to consist of adjacent protomers engaging the interdomain linker of one molecule in the substrate binding site of another, attenuating the chaperone function of oligomeric BiP. Native gel electrophoresis revealed a rapidly-modulated reciprocal relationship between the burden of unfolded proteins and BiP oligomers and slower equilibration between oligomers and inactive, covalently-modified BiP. Lumenal ER calcium depletion caused rapid oligomerization of mammalian BiP and a coincidental diminution in substrate binding, pointing to the relative inertness of the oligomers. Thus, equilibration between inactive oligomers and active monomeric BiP is poised to buffer fluctuations in ER unfolded protein load on a rapid timescale attainable neither by inter-conversion of active and covalently-modified BiP nor by the conventional unfolded protein response.


Subject(s)
Heat-Shock Proteins/metabolism , Protein Multimerization , Animals , Cricetinae , Electrophoresis , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum Chaperone BiP , Protein Binding , Protein Subunits/chemistry , Protein Subunits/metabolism
20.
Diabetes ; 64(11): 3951-62, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26159176

ABSTRACT

Dysregulated endoplasmic reticulum stress and phosphorylation of eukaryotic translation initiation factor 2α (eIF2α) are associated with pancreatic ß-cell failure and diabetes. Here, we report the first homozygous mutation in the PPP1R15B gene (also known as constitutive repressor of eIF2α phosphorylation [CReP]) encoding the regulatory subunit of an eIF2α-specific phosphatase in two siblings affected by a novel syndrome of diabetes of youth with short stature, intellectual disability, and microcephaly. The R658C mutation in PPP1R15B affects a conserved amino acid within the domain important for protein phosphatase 1 (PP1) binding. The R658C mutation decreases PP1 binding and eIF2α dephosphorylation and results in ß-cell apoptosis. Our findings support the concept that dysregulated eIF2α phosphorylation, whether decreased by mutation of the kinase (EIF2AK3) in Wolcott-Rallison syndrome or increased by mutation of the phosphatase (PPP1R15B), is deleterious to ß-cells and other secretory tissues, resulting in diabetes associated with multisystem abnormalities.


Subject(s)
Diabetes Mellitus/genetics , Growth Disorders/genetics , Microcephaly/genetics , Mutation, Missense , Protein Phosphatase 1/genetics , Adolescent , Adult , Female , Humans , Male , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL