Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 234
Filter
1.
Brain Res ; 1648(Pt B): 603-616, 2016 10 01.
Article in English | MEDLINE | ID: mdl-26923166

ABSTRACT

In neurological disorders, both acute and chronic neural stress can disrupt cellular proteostasis, resulting in the generation of pathological protein. However in most cases, neurons adapt to these proteostatic perturbations by activating a range of cellular protective and repair responses, thus maintaining cell function. These interconnected adaptive mechanisms comprise a 'proteostasis network' and include the unfolded protein response, the ubiquitin proteasome system and autophagy. Interestingly, several recent studies have shown that these adaptive responses can be stimulated by preconditioning treatments, which confer resistance to a subsequent toxic challenge - the phenomenon known as hormesis. In this review we discuss the impact of adaptive stress responses stimulated in diverse human neuropathologies including Parkinson׳s disease, Wolfram syndrome, brain ischemia, and brain cancer. Further, we examine how these responses and the molecular pathways they recruit might be exploited for therapeutic gain. This article is part of a Special Issue entitled SI:ER stress.


Subject(s)
Autophagy , Nervous System Diseases , Proteostasis Deficiencies/complications , Unfolded Protein Response/physiology , Animals , Endoplasmic Reticulum Stress/physiology , Humans , Nervous System Diseases/complications , Nervous System Diseases/metabolism , Nervous System Diseases/therapy , Proteasome Endopeptidase Complex/metabolism , Signal Transduction , Ubiquitin/metabolism
2.
J Dent Res ; 94(2): 320-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25425581

ABSTRACT

Mortality and morbidity associated with oral squamous cell carcinoma (OSCC) remain unacceptably high with disfiguring treatment options and a death rate of 1 per hour in the United States. The approval of cituximab for advanced OSCC has been the only new treatment for these patients since the 1970s, although it has not significantly increased overall survival. To address the paucity of effective new therapies, we undertook a high-throughput screen to discover small molecules and natural products that could induce endoplasmic reticulum (ER) stress and enforce a terminal unfolded protein response (UPR) in OSCC. The terpenoid cantharidin (CNT), previously used to treat various malignancies in culture-specific medical practices for over 2,000 y, emerged as a hit. CNT and its analog, cantharidic acid, potently induced protein and gene expression profiles consistent with the activation of ER stress, the UPR, and apoptosis in OSCC cells. Murine embryonic fibroblasts null for the UPR-associated transcription factors Atf4 or Chop were significantly protected from CNT, implicating a key role for the UPR in the death response. These data validate that our high-throughput screen can identify novel modulators of UPR signaling and that such compounds might provide a new therapeutic approach to treating patients with OSCC.


Subject(s)
Antineoplastic Agents/pharmacology , Cantharidin/pharmacology , Carcinoma, Squamous Cell/pathology , Endoplasmic Reticulum Stress/drug effects , Mouth Neoplasms/pathology , Unfolded Protein Response/drug effects , Activating Transcription Factor 4/genetics , Animals , Apoptosis/drug effects , Arylamine N-Acetyltransferase/antagonists & inhibitors , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , CHO Cells , Cell Death/drug effects , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cricetinae , Cricetulus , Fibroblasts/metabolism , Gene Expression Profiling , Gene Knockdown Techniques , High-Throughput Screening Assays , Humans , Leucine Zippers/genetics , Mice , RNA, Small Interfering/genetics , Transcription Factor CHOP/genetics
3.
Diabetologia ; 56(4): 911-24, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23314846

ABSTRACT

AIMS/HYPOTHESIS: Although obesity is associated with endoplasmic reticulum (ER) stress and activation of the unfolded protein response (UPR) in adipose tissue, it is not known how UPR signalling affects adipogenesis. To test whether signalling through protein kinase RNA-like ER kinase/eukaryotic initiation factor 2 alpha (PERK/eIF2α) or inositol-requiring enzyme 1 alpha/X-box binding protein 1 (IRE1α/XBP1) is required for adipogenesis, we studied the role of UPR signalling in adipocyte differentiation in vitro and in vivo in mice. METHODS: The role of UPR signalling in adipogenesis was investigated using 3T3-L1 cells and primary mouse embryonic fibroblasts (MEFs) by activation or inhibition of PERK-mediated phosphorylation of the eIF2α- and IRE1α-mediated splicing of Xbp1 mRNA. Body weight change, fat mass composition and adipocyte number and size were measured in wild-type and genetically engineered mice fed a control or high-fat diet (HFD). RESULTS: ER stress repressed adipocyte differentiation in 3T3-L1 cells. Impaired eIF2α phosphorylation enhanced adipocyte differentiation in MEFs, as well as in mice. In contrast, increased eIF2α phosphorylation reduced adipocyte differentiation in 3T3-L1 cells. Forced production of CCAAT/enhancer binding protein (C/EBP) homologous protein (CHOP), a downstream target of eIF2α phosphorylation, inhibited adipogenesis in 3T3-L1 cells. Mice with deletion of Chop (also known as Ddit3) (Chop (-/-)) gained more fat mass than wild-type mice on HFD. In addition, Chop deletion in genetically obese Lepr (db/db) mice increased body fat mass without altering adipocyte size. In contrast to the eIF2α-CHOP pathway, activation or deletion of Ire1a (also known as Ern1) did not alter adipocyte differentiation in 3T3-L1 cells. CONCLUSIONS/INTERPRETATION: These results demonstrate that eIF2α-CHOP suppresses adipogenesis and limits expansion of fat mass in vivo in mice, rendering this pathway a potential therapeutic target.


Subject(s)
Adipogenesis/genetics , Endoplasmic Reticulum Stress , Endoribonucleases/metabolism , Eukaryotic Initiation Factor-2/metabolism , Protein Serine-Threonine Kinases/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Animals , Body Composition , Cell Differentiation , Fibroblasts/cytology , Gene Deletion , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Signal Transduction , Transcription Factor CHOP/metabolism
4.
J Thromb Haemost ; 10(1): 107-15, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22044596

ABSTRACT

BACKGROUND: Heterologous expression of factor VIII (FVIII) is about two to three orders of magnitude lower than similarly sized proteins. Bioengineering strategies aimed at different structural and biochemical attributes of FVIII have been successful in enhancing its expression levels. OBJECTIVE: Disulfide bonds are vital to the proper folding, secretion and stability of most secretory proteins. In an effort to explore additional targeted bioengineering approaches, the role of disulfide bonds in FVIII secretion and function was probed in this study. METHODS AND RESULTS: Single and paired cysteine mutants were generated by substituting with serine or glycine residues and analyzed by transient transfection into COS-1 and CHO cells. Seven of the eight disulfide bonds in FVIII were found to be indispensable for proper secretion and function. However, elimination of the disulfide bond formed by C1899 and C1903 within the conserved A3 domain improved the secretion of FVIII. The addition of the C1899G/C1903G mutations to a previously described FVIII variant, 226/N6, with high secretion efficiency increased its secretion by 2.2-fold. Finally, the addition of the A1-domain mutation, F309S, in conjunction with the disulfide mutation had an additive effect, resulting in a net improvement in secretion of between 35 and 45-fold higher than wild-type FVIII in CHO cells. CONCLUSION: Such combined targeted bioengineering strategies may facilitate more efficient production of recombinant FVIII and contribute toward low-cost factor replacement therapy for hemophilia A.


Subject(s)
Disulfides/metabolism , Factor VIII/genetics , Protein Engineering/methods , Cell Line , Factor VIII/metabolism , Humans , Mutagenesis, Site-Directed , Transfection
5.
Diabetes Obes Metab ; 12 Suppl 2: 99-107, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21029306

ABSTRACT

Diabetes is an epidemic of worldwide proportions caused by ß-cell failure. Nutrient fluctuations and insulin resistance drive ß-cells to synthesize insulin beyond their capacity for protein folding and secretion and thereby activate the unfolded protein response (UPR), an adaptive signalling pathway to promote cell survival upon accumulation of unfolded protein in the endoplasmic reticulum (ER). Protein kinase-like endoplasmic reticulum kinase (PERK) signals one component of the UPR through phosphorylation of eukaryotic initiation factor 2 on the α-subunit (eIF2α) to attenuate protein synthesis, thereby reducing the biosynthetic burden. ß-Cells uniquely require PERK-mediated phosphorylation of eIF2α to preserve cell function. Unabated protein synthesis in ß-cells is sufficient to initiate a cascade of events, including oxidative stress, that are characteristic of ß-cell failure observed in type 2 diabetes. In contrast to acute adaptive UPR activation, chronic activation increases expression of the proapoptotic transcription factor CAAT/enhancer-binding protein homologous protein (CHOP). Chop deletion in insulin-resistant mice profoundly increases ß-cell mass and prevents ß-cell failure to forestall the progression of diabetes. The findings suggest an unprecedented link by which protein synthesis and/or misfolding in the ER causes oxidative stress and should encourage the development of novel strategies to treat diabetes.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Endoplasmic Reticulum/physiology , Insulin-Secreting Cells/metabolism , Unfolded Protein Response/physiology , Animals , Apoptosis/physiology , Cell Differentiation , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 2/genetics , Endoplasmic Reticulum/genetics , Gene Expression Regulation , Humans , Insulin-Secreting Cells/cytology , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/physiology , Protein Biosynthesis/physiology , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , eIF-2 Kinase/biosynthesis
6.
Cell Death Differ ; 14(2): 230-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16794605

ABSTRACT

Expanded polyglutamine 72 repeat (polyQ72) aggregates induce endoplasmic reticulum (ER) stress-mediated cell death with caspase-12 activation and vesicular formation (autophagy). We examined this relationship and the molecular mechanism of autophagy formation. Rapamycin, a stimulator of autophagy, inhibited the polyQ72-induced cell death with caspase-12 activation. PolyQ72, but not polyQ11, stimulated Atg5-Atg12-Atg16 complex-dependent microtubule-associated protein 1 (MAP1) light chain 3 (LC3) conversion from LC3-I to -II, which plays a key role in autophagy. The eucaryotic translation initiation factor 2 alpha (eIF2alpha) A/A mutation, a knock-in to replace a phosphorylatable Ser51 with Ala51, and dominant-negative PERK inhibited polyQ72-induced LC3 conversion. PolyQ72 as well as ER stress stimulators upregulated Atg12 mRNA and proteins via eIF2alpha phosphorylation. Furthermore, Atg5 deficiency as well as the eIF2alpha A/A mutation increased the number of cells showing polyQ72 aggregates and polyQ72-induced caspase-12 activation. Thus, autophagy formation is a cellular defense mechanism against polyQ72-induced ER-stress-mediated cell death by degrading polyQ72 aggregates, with PERK/eIF2alpha phosphorylation being involved in polyQ72-induced LC3 conversion.


Subject(s)
Autophagy , Endoplasmic Reticulum/metabolism , Eukaryotic Initiation Factor-2/metabolism , Microtubule-Associated Proteins/metabolism , Peptides/metabolism , eIF-2 Kinase/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Autophagy/drug effects , Autophagy-Related Protein 5 , Caspase 12/metabolism , Cell Death/drug effects , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/pathology , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Leucine/analogs & derivatives , Leucine/pharmacology , Lysosomes/drug effects , Lysosomes/enzymology , Mice , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Models, Biological , Pepstatins/pharmacology , Peptides/chemistry , Phosphorylation/drug effects , Protein Structure, Quaternary/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sirolimus/pharmacology
7.
Haemophilia ; 12 Suppl 3: 137-42, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16684009

ABSTRACT

Deficiencies of coagulation factors other than factor VIII and factor IX (afibrinogenemia, FII, FV, FV+FVIII, FVII, FX, FXI, FXIII) that cause bleeding disorders (RBDs) are inherited as autosomal recessive traits and are rare, with prevalences in the general population varying between 1 in 500,000 and 1 in 2 million for the homozygous forms. As a consequence of the rarity of these deficiencies, the type and severity of bleeding symptoms, the underlying molecular defects, and the actual management of bleeding episodes are not as well established as for hemophilia A and B. The study of the genetic basis of these disorders could represent an important tool for prevention through prenatal diagnosis. Treatment of patients with RBDs during bleeding episodes or surgery is a challenge because of the lack of experience and the paucity of data. For some deficiency factor concentrates are still non available and severe complications can occur. These complications can be minimized by assessment of risks of bleeding and thrombosis, use of haemostatic means other than blood components or no therapy at all. The RBDs pose a problem for guideline writers because there are no suitable clinical trials to supply good evidence for how these people are best treated. The lack of adequate information on clinical manifestations, treatment and genetic basis of RBDs could be improved by the collection of data in an International Database (http://www.rbdd.org), linkable to others previously published. This could be a useful tool to fill the gap between clinical data and clinical practice. This article reviews the genetic basis of RBDs, problems and complications of treatment, problems in the preparation of suitable guidelines for treatment and the future perspectives of the International Registry on RBDs.


Subject(s)
Blood Coagulation Disorders, Inherited/therapy , Blood Coagulation Disorders, Inherited/genetics , Humans , Practice Guidelines as Topic , Registries
8.
Handb Exp Pharmacol ; (172): 69-91, 2006.
Article in English | MEDLINE | ID: mdl-16610355

ABSTRACT

In all eukaryotic cells, the endoplasmic reticulum (ER) is an intracellular organelle where folding and assembly occurs for proteins destined to the extracellular space, plasma membrane, and the exo/endocytic compartments (Kaufman 1999). As a protein-folding compartment, the ER is exquisitely sensitive to alterations in homeostasis, and provides stringent quality control systems to ensure that only correctly folded proteins transit to the Golgi and unfolded or misfolded proteins are retained and ultimately degraded. A number of biochemical and physiological stimuli, such as perturbation in calcium homeostasis or redox status, elevated secretory protein synthesis, expression of misfolded proteins, sugar/glucose deprivation, altered glycosylation, and overloading of cholesterol can disrupt ER homeostasis, impose stress to the ER, and subsequently lead to accumulation of unfolded or misfolded proteins in the ER lumen. The ER has evolved highly specific signaling pathways called the unfolded protein response (UPR) to cope with the accumulation of unfolded or misfolded proteins. Elucidation of the molecular mechanisms by which accumulation of unfolded proteins in the ER transmits a signal to the cytoplasm and nucleus has led to major new insights into the diverse cellular and physiological processes that are regulated by the UPR. This chapter summarizes how cells respond to the accumulation of unfolded proteins in the cell and the relevance of these signaling pathways to human physiology and disease.


Subject(s)
Endoplasmic Reticulum/metabolism , Protein Folding , Animals , Apoptosis , Cell Differentiation , Diabetes Mellitus/etiology , Endoplasmic Reticulum/chemistry , Glucose/metabolism , Humans , Nervous System Diseases/etiology , Protein Biosynthesis , Proteins/metabolism , Transcription, Genetic
9.
Cell Death Differ ; 13(3): 374-84, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16397578

ABSTRACT

When protein folding in the endoplasmic reticulum (ER) is disrupted by alterations in homeostasis in the ER lumen, eucaryotic cells activate a series of signal transduction cascades that are collectively termed the unfolded protein response (UPR). Here we summarize our current understanding of how the UPR functions upon acute and severe stress. We discuss the mechanism of UPR receptor activation, UPR signal transduction to translational and transcriptional responses, UPR termination, and UPR signals that activate upon irreversible damage. Further, we review recent studies that have revealed that UPR provides a wide spectrum of physiological roles. Each individual UPR subpathway provides a unique and specialized role in diverse developmental and metabolic processes. This is especially observed for professional secretory cells, such as plasma cells, pancreatic beta cells, hepatocytes, and osteoblasts, where high-level secretory protein synthesis requires a highly evolved mechanism to properly fold, process, and secrete proteins. There is a growing body of data that suggest that different subpathways of the UPR are required throughout the entire life of eucaryotic organisms, from regulation of differentiation to induction of apoptosis.


Subject(s)
Apoptosis , Endoplasmic Reticulum/physiology , Animals , Mice , Protein Folding , Proteins/metabolism , Signal Transduction
10.
Nucleic Acids Res ; 32(Database issue): D560-7, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14681481

ABSTRACT

GermOnline provides information and microarray expression data for genes involved in mitosis and meiosis, gamete formation and germ line development across species. The database has been developed, and is being curated and updated, by life scientists in cooperation with bioinformaticists. Information is contributed through an online form using free text, images and the controlled vocabulary developed by the GeneOntology Consortium. Authors provide up to three references in support of their contribution. The database is governed by an international board of scientists to ensure a standardized data format and the highest quality of GermOnline's information content. Release 2.0 provides exclusive access to microarray expression data from Saccharomyces cerevisiae and Rattus norvegicus, as well as curated information on approximately 700 genes from various organisms. The locus report pages include links to external databases that contain relevant annotation, microarray expression and proteome data. Conversely, the Saccharomyces Genome Database (SGD), S.cerevisiae GeneDB and Swiss-Prot link to the budding yeast section of GermOnline from their respective locus pages. GermOnline, a fully operational prototype subject-oriented knowledgebase designed for community annotation and array data visualization, is accessible at http://www.germonline.org. The target audience includes researchers who work on mitotic cell division, meiosis, gametogenesis, germ line development, human reproductive health and comparative genomics.


Subject(s)
Cell Differentiation/genetics , Databases, Genetic , Gene Expression Profiling , Germ Cells/cytology , Germ Cells/metabolism , Animals , Computational Biology , Genomics , Humans , Information Storage and Retrieval , Internet , Meiosis/genetics , Mitosis/genetics , Oligonucleotide Array Sequence Analysis , Proteins/metabolism , Proteome , Proteomics , Rats
12.
J Thromb Haemost ; 1(11): 2360-7, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14629470

ABSTRACT

Combined deficiency of both coagulation factors (F)V and VIII is a rare autosomal recessive bleeding disorder caused by null expression of LMAN1 (previously termed ERGIC-53) in a majority of affected individuals. Previously, a requirement for a functional LMAN1 cycling pathway between the ER and Golgi was demonstrated for efficient secretion of FV and FVIII (Moussalli et al. J Biol Chem 1999; 274: 32569), however, the molecular nature of the interaction between LMAN1 and its cargo was not characterized. Using coimmunoprecipitation of LMAN1 and FVIII from transfected HeLa and COS-1 cells, we demonstrate an interaction between LMAN1 and FVIII in vivo. The interaction was mediated via high mannose-containing asparagine-linked oligosaccharides that are densely situated within the B domain of FVIII, as well as protein-protein interactions. These results are interpreted based on the recent determination of the crystal structure of the carbohydrate recognition domain of LMAN1.


Subject(s)
Factor VIII/metabolism , Mannose-Binding Lectins/metabolism , Membrane Proteins/metabolism , Molecular Chaperones/metabolism , Factor V Deficiency , HeLa Cells , Hemophilia A , Humans , Mannose-Binding Lectins/deficiency , Mannose-Binding Lectins/physiology , Membrane Proteins/deficiency , Membrane Proteins/physiology , Molecular Chaperones/physiology , Oligosaccharides , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Protein Transport , Transfection
14.
J Biol Chem ; 276(38): 35867-74, 2001 Sep 21.
Article in English | MEDLINE | ID: mdl-11447214

ABSTRACT

Severe hyperhomocysteinemia is associated with endothelial cell injury that may contribute to an increased incidence of thromboembolic disease. In this study, homocysteine induced programmed cell death in human umbilical vein endothelial cells as measured by TdT-mediated dUTP nick end labeling assay, DNA ladder formation, induction of caspase 3-like activity, and cleavage of procaspase 3. Homocysteine-induced cell death was specific to homocysteine, was not mediated by oxidative stress, and was mimicked by inducers of the unfolded protein response (UPR), a signal transduction pathway activated by the accumulation of unfolded proteins in the lumen of the endoplasmic reticulum. Dominant negative forms of the endoplasmic reticulum-resident protein kinases IRE1alpha and -beta, which function as signal transducers of the UPR, prevented the activation of glucose-regulated protein 78/immunoglobulin chain-binding protein and C/EBP homologous protein/growth arrest and DNA damage-inducible protein 153 in response to homocysteine. Furthermore, overexpression of the point mutants of IRE1 with defective RNase more effectively suppressed the cell death than the kinase-defective mutant. These results indicate that homocysteine induces apoptosis in human umbilical vein endothelial cells by activation of the UPR and is signaled through IRE1. The studies implicate that the UPR may cause endothelial cell injury associated with severe hyperhomocysteinemia.


Subject(s)
Apoptosis/drug effects , Endothelium, Vascular/drug effects , Heat-Shock Proteins , Homocysteine/pharmacology , Muscle Proteins/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Carrier Proteins/metabolism , Caspase 3 , Caspases/metabolism , Cells, Cultured , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Enzyme Activation , Humans , Molecular Chaperones/metabolism , Protein Folding , Reactive Oxygen Species , Transcription Factor CHOP , Transcription Factors/metabolism
15.
Am J Pathol ; 159(1): 273-83, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11438474

ABSTRACT

Viral infection is one environmental factor that has been implicated as a precipitating event that may initiate beta-cell damage during the development of diabetes. This study examines the mechanisms by which the viral replicative intermediate, double-stranded (ds) RNA impairs beta-cell function and induces beta-cell death. The synthetic dsRNA molecule polyinosinic-polycytidylic acid (poly IC) stimulates beta-cell DNA damage and apoptosis without impairing islet secretory function. In contrast, the combination of poly IC and interferon (IFN)-gamma stimulates DNA damage, apoptosis, and necrosis of islet cells, and this damage is associated with the inhibition of glucose-stimulated insulin secretion. Nitric oxide mediates the inhibitory and destructive actions of poly IC + IFN-gamma on insulin secretion and islet cell necrosis. Inhibitors of nitric oxide synthase, aminoguanidine, and N(G)-monomethyl-L-arginine, attenuate poly IC + IFN-gamma-induced DNA damage to levels observed in response to poly IC alone, prevent islet cell necrosis, and prevent the inhibitory actions on glucose-stimulated insulin secretion. N(G)-monomethyl-L-arginine fails to prevent poly IC- and poly IC + IFN-gamma-induced islet cell apoptosis. PKR, the dsRNA-dependent protein kinase that mediates the antiviral response in infected cells, is required for poly IC- and poly IC + IFN-gamma-induced islet cell apoptosis, but not nitric oxide-mediated islet cell necrosis. Alone, poly IC fails to stimulate DNA damage in islets isolated from PKR-deficient mice; however, nitric oxide-dependent DNA damage induced by the combination of poly IC + IFN-gamma is not attenuated by the genetic absence of PKR. These findings indicate that dsRNA stimulates PKR-dependent islet cell apoptosis, an event that is associated with normal islet secretory function. In contrast, poly IC + IFN-gamma-induced inhibition of glucose-stimulated insulin secretion and islet cell necrosis are events that are mediated by islet production of nitric oxide. These findings suggest that at least one IFN-gamma-induced antiviral response (islet cell necrosis) is mediated through a PKR-independent pathway.


Subject(s)
Interferon-gamma/pharmacology , Islets of Langerhans/physiology , Poly I-C/pharmacology , RNA, Double-Stranded/pharmacology , Animals , Apoptosis/physiology , Cell Death/physiology , Cells, Cultured , DNA Damage/physiology , Drug Synergism , Glucose/pharmacology , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Islets of Langerhans/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Knockout/genetics , Microscopy, Electron , Necrosis , Nitric Oxide/physiology , Protein Kinases/physiology , Rats , Rats, Sprague-Dawley , eIF-2 Kinase/physiology
16.
Mol Cell ; 7(6): 1165-76, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11430820

ABSTRACT

The accumulation of unfolded protein in the endoplasmic reticulum (ER) attenuates protein synthesis initiation through phosphorylation of the alpha subunit of eukaryotic translation initiation factor 2 (eIF2alpha) at Ser51. Subsequently, transcription of genes encoding adaptive functions including the glucose-regulated proteins is induced. We show that eIF2alpha phosphorylation is required for translation attenuation, transcriptional induction, and survival in response to ER stress. Mice with a homozygous mutation at the eIF2alpha phosphorylation site (Ser51Ala) died within 18 hr after birth due to hypoglycemia associated with defective gluconeogenesis. In addition, homozygous mutant embryos and neonates displayed a deficiency in pancreatic beta cells. The results demonstrate that regulation of translation through eIF2alpha phosphorylation is essential for the ER stress response and in vivo glucose homeostasis.


Subject(s)
DNA-Binding Proteins/genetics , Glucose/metabolism , Heat-Shock Proteins , Homeostasis/physiology , Hypoglycemia/metabolism , Protein Biosynthesis/physiology , Transcription Factors/genetics , Activating Transcription Factors , Animals , Animals, Newborn , Blood Proteins/genetics , Blood Proteins/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Survival/physiology , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Gene Expression/physiology , Germ-Line Mutation , Gluconeogenesis/physiology , Homozygote , Hypoglycemia/genetics , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Mice , Mice, Mutant Strains , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Mutagenesis/physiology , Phosphorylation , Protein Folding , RNA, Messenger/analysis , Transcription Factor CHOP , Transcription Factors/chemistry , Transcription Factors/metabolism , Transcriptional Activation/physiology
17.
Blood ; 97(3): 685-91, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11157485

ABSTRACT

Thrombin-activated factor VIII (FVIIIa) is a heterotrimer with the A2 subunit (amino acid residues 373-740) in a weak ionic interaction with the A1 and A3-C1-C2 subunits. Dissociation of the A2 subunit correlates with inactivation of FVIIIa. Patients with hemophilia A have been described whose plasmas display a discrepancy between their FVIII activities, where the 1-stage activity assay displays greater activity than the 2-stage activity assay. The molecular basis for one of these mutations, (ARG)531(HIS), is an increased rate of A2 subunit dissociation. Examination of a homology model of the A domains of FVIII predicted (ARG)531 to lie at the interface of the A1 and A2 subunits and stabilize their interaction. Indeed, patients with mutations either directly contacting (ARG)531 ((ALA)284(GLU), (ALA)284(PRO)) or closely adjacent to the A1-A2 interface in the tightly packed hydrophobic core ((SER)289(LEU)) have the same phenotype of 1-stage/2-stage discrepancy. The (ALA)284(GLU) and (SER)289(LEU) mutations in FVIII were produced by transfection of COS-1 monkey cells. Compared to FVIII wild-type both mutants had reduced specific activity by 1-stage clotting activity and at least a 2-fold lower activity by 2-stage analysis (COAMATIC), similar to the reported clinical data. Analysis of immunoaffinity purified (ALA)284(GLU) and (SER)289(LEU) proteins in an optical biosensor demonstrated that A2 dissociation was 3-fold faster for both FVIIIa mutants compared to FVIIIa wild-type. Therefore, these mutations within the A1 subunit of FVIIIa introduce a similar destabilization of the FVIIIa heterotrimer compared to the (ARG)531(HIS) mutation within the A2 subunit and support that these residues stabilize the A domain interface of FVIIIa.


Subject(s)
Factor VIIIa/genetics , Hemophilia A/blood , Hemophilia A/genetics , Animals , Biosensing Techniques , Blood Coagulation , COS Cells , Chromogenic Compounds/chemistry , Factor VIIIa/metabolism , Humans , Mutation, Missense , Phenotype , Protein Conformation , Protein Structure, Tertiary , Recombinant Proteins/blood , Thrombin/pharmacology , Transfection
18.
Nutrition ; 17(1): 59-61, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11165893
19.
Cell ; 107(7): 893-903, 2001 Dec 28.
Article in English | MEDLINE | ID: mdl-11779465

ABSTRACT

The unfolded protein response (UPR) is a transcriptional and translational intracellular signaling pathway activated by the accumulation of unfolded proteins in the lumen of the endoplasmic reticulum (ER). We have used C. elegans as a genetic model system to dissect UPR signaling in a multicellular organism. C. elegans requires ire-1-mediated splicing of xbp-1 mRNA for UPR gene transcription and survival upon ER stress. In addition, ire-1/xbp-1 acts with pek-1, a protein kinase that mediates translation attenuation, in complementary pathways that are essential for worm development and survival. We propose that UPR transcriptional activation by ire-1 as well as translational attenuation by pek-1 maintain ER homeostasis. The results demonstrate that the UPR and ER homeostasis are essential for metazoan development.


Subject(s)
Caenorhabditis elegans/physiology , Cell Cycle Proteins , MAP Kinase Kinase 1 , Protein Folding , Protein Serine-Threonine Kinases , Saccharomyces cerevisiae Proteins , Signal Transduction , Transcriptional Activation/physiology , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans Proteins , Fungal Proteins/genetics , Membrane Glycoproteins/genetics , Mitogen-Activated Protein Kinase Kinases/genetics , Molecular Sequence Data , Mutation , Repressor Proteins/genetics , Transcription Factors/genetics
20.
Mol Biotechnol ; 16(2): 151-60, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11131974

ABSTRACT

The expression of cloned genes in mammalian cells is a basic tool for understanding gene expression, protein structure, and function, and biological regulatory mechanisms. The level of protein expression from heterologous genes introduced into mammalian cells depends upon multiple factors including DNA copy number, efficiency of transportation, mRNA processing, mRNA transport, mRNA stability, and translational efficiency, and protein processing, transport, and stability. Different genes exhibit different rate limiting steps for efficient expression. Multiple strategies are available to obtain high level expression in mammalian cells. This article reviews vector design for expression of foreign genes in mammalian cells.


Subject(s)
Genetic Vectors , Transfection/methods , Transformation, Genetic , Animals , Cell Line , Escherichia coli/metabolism , HeLa Cells , Humans , Protein Biosynthesis , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL