Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
1.
Mol Cell ; 81(20): 4191-4208.e8, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34686314

ABSTRACT

To survive, mammalian cells must adapt to environmental challenges. While the cellular response to mild stress has been widely studied, how cells respond to severe stress remains unclear. We show here that under severe hyperosmotic stress, cells enter a transient hibernation-like state in anticipation of recovery. We demonstrate this adaptive pausing response (APR) is a coordinated cellular response that limits ATP supply and consumption through mitochondrial fragmentation and widespread pausing of mRNA translation. This pausing is accomplished by ribosome stalling at translation initiation codons, which keeps mRNAs poised to resume translation upon recovery. We further show that recovery from severe stress involves ISR (integrated stress response) signaling that permits cell cycle progression, resumption of growth, and reversal of mitochondria fragmentation. Our findings indicate that cells can respond to severe stress via a hibernation-like mechanism that preserves vital elements of cellular function under harsh environmental conditions.


Subject(s)
Cell Proliferation , Fibroblasts/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/biosynthesis , Osmotic Pressure , Protein Biosynthesis , Ribosomes/metabolism , Adaptation, Physiological , Adenosine Triphosphate/metabolism , Animals , Codon, Initiator , Fibroblasts/pathology , HEK293 Cells , Humans , Kinetics , Mice , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Proteins/genetics , Ribosomes/genetics , Signal Transduction
2.
Mol Cell ; 68(5): 885-900.e6, 2017 Dec 07.
Article in English | MEDLINE | ID: mdl-29220654

ABSTRACT

The integrated stress response (ISR) is a homeostatic mechanism induced by endoplasmic reticulum (ER) stress. In acute/transient ER stress, decreased global protein synthesis and increased uORF mRNA translation are followed by normalization of protein synthesis. Here, we report a dramatically different response during chronic ER stress. This chronic ISR program is characterized by persistently elevated uORF mRNA translation and concurrent gene expression reprogramming, which permits simultaneous stress sensing and proteostasis. The program includes PERK-dependent switching to an eIF3-dependent translation initiation mechanism, resulting in partial, but not complete, translational recovery, which, together with transcriptional reprogramming, selectively bolsters expression of proteins with ER functions. Coordination of transcriptional and translational reprogramming prevents ER dysfunction and inhibits "foamy cell" development, thus establishing a molecular basis for understanding human diseases associated with ER dysfunction.


Subject(s)
Endoplasmic Reticulum Stress , Eukaryotic Initiation Factor-3/metabolism , Fibroblasts/metabolism , Protein Biosynthesis , RNA, Messenger/biosynthesis , Transcription, Genetic , eIF-2 Kinase/metabolism , Animals , Cellular Reprogramming , Eukaryotic Initiation Factor-3/genetics , Fibroblasts/pathology , HEK293 Cells , Humans , Mice , Open Reading Frames , Phenotype , Proteostasis , RNA Interference , RNA, Messenger/genetics , Signal Transduction , Time Factors , Transfection , eIF-2 Kinase/genetics
3.
J Physiol ; 601(3): 567-606, 2023 02.
Article in English | MEDLINE | ID: mdl-36533558

ABSTRACT

Nocturnal hypoxaemia, which is common in chronic obstructive pulmonary disease (COPD) patients, is associated with skeletal muscle loss or sarcopenia, which contributes to adverse clinical outcomes. In COPD, we have defined this as prolonged intermittent hypoxia (PIH) because the duration of hypoxia in skeletal muscle occurs through the duration of sleep followed by normoxia during the day, in contrast to recurrent brief hypoxic episodes during obstructive sleep apnoea (OSA). Adaptive cellular responses to PIH are not known. Responses to PIH induced by three cycles of 8 h hypoxia followed by 16 h normoxia were compared to those during chronic hypoxia (CH) or normoxia for 72 h in murine C2C12 and human inducible pluripotent stem cell-derived differentiated myotubes. RNA sequencing followed by downstream analyses were complemented by experimental validation of responses that included both unique and shared perturbations in ribosomal and mitochondrial function during PIH and CH. A sarcopenic phenotype characterized by decreased myotube diameter and protein synthesis, and increased phosphorylation of eIF2α (Ser51) by eIF2α kinase, and of GCN-2 (general controlled non-derepressed-2), occurred during both PIH and CH. Mitochondrial oxidative dysfunction, disrupted supercomplex assembly, lower activity of Complexes I, III, IV and V, and reduced intermediary metabolite concentrations occurred during PIH and CH. Decreased mitochondrial fission occurred during CH. Physiological relevance was established in skeletal muscle of mice with COPD that had increased phosphorylation of eIF2α, lower protein synthesis and mitochondrial oxidative dysfunction. Molecular and metabolic responses with PIH suggest an adaptive exhaustion with failure to restore homeostasis during normoxia. KEY POINTS: Sarcopenia or skeletal muscle loss is one of the most frequent complications that contributes to mortality and morbidity in patients with chronic obstructive pulmonary disease (COPD). Unlike chronic hypoxia, prolonged intermittent hypoxia is a frequent, underappreciated and clinically relevant model of hypoxia in patients with COPD. We developed a novel, in vitro myotube model of prolonged intermittent hypoxia with molecular and metabolic perturbations, mitochondrial oxidative dysfunction, and consequent sarcopenic phenotype. In vivo studies in skeletal muscle from a mouse model of COPD shared responses with our myotube model, establishing the pathophysiological relevance of our studies. These data lay the foundation for translational studies in human COPD to target prolonged, nocturnal hypoxaemia to prevent sarcopenia in these patients.


Subject(s)
Pulmonary Disease, Chronic Obstructive , Sarcopenia , Humans , Mice , Animals , Sarcopenia/metabolism , Proteostasis , Muscle, Skeletal/metabolism , Hypoxia/metabolism , Pulmonary Disease, Chronic Obstructive/complications
4.
FASEB J ; 35(11): e21990, 2021 11.
Article in English | MEDLINE | ID: mdl-34665898

ABSTRACT

Eukaryotic initiation factor 2A (eIF2A) is a 65 kDa protein that functions in minor initiation pathways, which affect the translation of only a subset of messenger ribonucleic acid (mRNAs), such as internal ribosome entry site (IRES)-containing mRNAs and/or mRNAs harboring upstream near cognate/non-AUG start codons. These non-canonical initiation events are important for regulation of protein synthesis during cellular development and/or the integrated stress response. Selective eIF2A knockdown in cellular systems significantly inhibits translation of such mRNAs, which rely on alternative initiation mechanisms for their translation. However, there exists a gap in our understanding of how eIF2A functions in mammalian systems in vivo (on the organismal level) and ex vivo (in cells). Here, using an eIF2A-knockout (KO) mouse model, we present evidence implicating eIF2A in the biology of aging, metabolic syndrome and central tolerance. We discovered that eIF2A-KO mice have reduced life span and that eIF2A plays an important role in maintenance of lipid homeostasis, the control of glucose tolerance, insulin resistance and also reduces the abundance of B lymphocytes and dendritic cells in the thymic medulla of mice. We also show the eIF2A KO affects male and female mice differently, suggesting that eIF2A may affect sex-specific pathways. Interestingly, our experiments involving pharmacological induction of endoplasmic reticulum (ER) stress with tunicamycin did not reveal any substantial difference between the response to ER stress in eIF2A-KO and wild-type mice. The identification of eIF2A function in the development of metabolic syndrome bears promise for the further identification of specific eIF2A targets responsible for these changes.


Subject(s)
Lipid Metabolism , Longevity , Metabolic Syndrome/metabolism , Protein Serine-Threonine Kinases/physiology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sex Factors
5.
Mol Cell Proteomics ; 19(5): 852-870, 2020 05.
Article in English | MEDLINE | ID: mdl-32132231

ABSTRACT

The redox-based modifications of cysteine residues in proteins regulate their function in many biological processes. The gas molecule H2S has been shown to persulfidate redox sensitive cysteine residues resulting in an H2S-modified proteome known as the sulfhydrome. Tandem Mass Tags (TMT) multiplexing strategies for large-scale proteomic analyses have become increasingly prevalent in detecting cysteine modifications. Here we developed a TMT-based proteomics approach for selectively trapping and tagging cysteine persulfides in the cellular proteomes. We revealed the natural protein sulfhydrome of two human cell lines, and identified insulin as a novel substrate in pancreatic beta cells. Moreover, we showed that under oxidative stress conditions, increased H2S can target enzymes involved in energy metabolism by switching specific cysteine modifications to persulfides. Specifically, we discovered a Redox Thiol Switch, from protein S-glutathioinylation to S-persulfidation (RTSGS). We propose that the RTSGS from S-glutathioinylation to S-persulfidation is a potential mechanism to fine tune cellular energy metabolism in response to different levels of oxidative stress.


Subject(s)
Energy Metabolism , Sulfhydryl Compounds/metabolism , Activating Transcription Factor 4/metabolism , Animals , Biological Assay , Biotin/metabolism , Cell Line , Cysteine/metabolism , Disulfides/metabolism , Glycolysis , Hepatocytes/metabolism , Humans , Hydrogen Sulfide/metabolism , Insulin-Secreting Cells/metabolism , Mass Spectrometry , Metabolic Flux Analysis , Mitochondria/metabolism , Oxidation-Reduction , Proteome/metabolism , Proteomics , Rats , Sulfides/metabolism
6.
Biochim Biophys Acta Mol Cell Res ; 1865(1): 34-47, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28986221

ABSTRACT

The ribosomal uL10 protein, formerly known as P0, is an essential element of the ribosomal GTPase-associated center responsible for the interplay with translational factors during various stages of protein synthesis. In eukaryotic cells, uL10 binds two P1/P2 protein heterodimers to form a pentameric P-stalk, described as uL10-(P1-P2)2, which represents the functional form of these proteins on translating ribosomes. Unlike most ribosomal proteins, which are incorporated into pre-ribosomal particles during early steps of ribosome biogenesis in the nucleus, P-stalk proteins are attached to the 60S subunit in the cytoplasm. Although the primary role of the P-stalk is related to the process of translation, other extraribosomal functions of its constituents have been proposed, especially for the uL10 protein; however, the list of its activities beyond the ribosome is still an open question. Here, by the combination of biochemical and advanced fluorescence microscopy techniques, we demonstrate that upon nucleolar stress induction the uL10 protein accumulates in the cytoplasm of mammalian cells as a free, ribosome-unbound protein. Importantly, using a novel approach, FRAP-AC (FRAP after photoConversion), we have shown that the ribosome-free pool of uL10 represents a population of proteins released from pre-existing ribosomes. Taken together, our data indicate that the presence of uL10 on the ribosomes is affected in stressed cells, thus it might be considered as a regulatory element responding to environmental fluctuations.


Subject(s)
Arabidopsis Proteins/metabolism , Cell Nucleolus/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Ribosomal Proteins/metabolism , Ribosomes/metabolism , Stress, Physiological/physiology , Animals , HeLa Cells , Humans , Mice , NIH 3T3 Cells , Protein Binding
7.
J Biol Chem ; 293(9): 3399-3409, 2018 03 02.
Article in English | MEDLINE | ID: mdl-29317502

ABSTRACT

Phosphoenolpyruvate carboxykinase (Pck1) is a metabolic enzyme that is integral to the gluconeogenic and glyceroneogenic pathways. However, Pck1's role in macrophage metabolism and function is unknown. Using stable isotopomer MS analysis in a mouse model with a myeloid cell-specific Pck1 deletion, we show here that this deletion increases the proinflammatory phenotype in macrophages. Incubation of LPS-stimulated bone marrow-derived macrophages (BMDM) with [U-13C]glucose revealed reduced 13C labeling of citrate and malate and increased 13C labeling of lactate in Pck1-deleted bone marrow-derived macrophages. We also found that the Pck1 deletion in the myeloid cells increases reactive oxygen species (ROS). Of note, this altered macrophage metabolism increased expression of the M1 cytokines TNFα, IL-1ß, and IL-6. We therefore conclude that Pck1 contributes to M1 polarization in macrophages. Our findings provide important insights into the factors determining the macrophage inflammatory response and indicate that Pck1 activity contributes to metabolic reprogramming and polarization in macrophages.


Subject(s)
Gene Deletion , Macrophages/enzymology , Phenotype , Phosphoenolpyruvate Carboxykinase (GTP)/deficiency , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Animals , Cell Polarity , Glucose/metabolism , Glutamine/metabolism , Inflammation/enzymology , Inflammation/genetics , Inflammation/immunology , Macrophages/cytology , Macrophages/metabolism , Mice , Palmitic Acid/metabolism , RAW 264.7 Cells
8.
J Biol Chem ; 292(32): 13143-13153, 2017 08 11.
Article in English | MEDLINE | ID: mdl-28637872

ABSTRACT

Hydrogen sulfide (H2S) regulates various physiological processes, including neuronal activity, vascular tone, inflammation, and energy metabolism. Moreover, H2S elicits cytoprotective effects against stressors in various cellular models of injury. However, the mechanism of the signaling pathways mediating the cytoprotective functions of H2S is not well understood. We previously uncovered a heme-dependent metabolic switch for transient induction of H2S production in the trans-sulfuration pathway. Here, we demonstrate that increased endogenous H2S production or its exogenous administration modulates major components of the integrated stress response promoting a metabolic state primed for stress response. We show that H2S transiently increases phosphorylation of eukaryotic translation initiation factor 2 (eIF2α) resulting in inhibition of general protein synthesis. The H2S-induced increase in eIF2α phosphorylation was mediated at least in part by inhibition of protein phosphatase-1 (PP1c) via persulfidation at Cys-127. Overexpression of a PP1c cysteine mutant (C127S-PP1c) abrogated the H2S effect on eIF2α phosphorylation. Our data support a model in which H2S exerts its cytoprotective effect on ISR signaling by inducing a transient adaptive reprogramming of global mRNA translation. Although a transient increase in endogenous H2S production provides cytoprotection, its chronic increase such as in cystathionine ß-synthase deficiency may pose a problem.


Subject(s)
Activating Transcription Factor 4/metabolism , Endoplasmic Reticulum Stress , Eukaryotic Initiation Factor-2/metabolism , Gasotransmitters/metabolism , Hydrogen Sulfide/metabolism , Protein Phosphatase 1/metabolism , Activating Transcription Factor 4/genetics , Allostasis , Amino Acid Substitution , Animals , Cell Line , Cell Survival , Cells, Cultured , Cysteine/chemistry , Eukaryotic Initiation Factor-2/genetics , Gene Expression Regulation , Humans , Mice , Mice, Knockout , Mutation , Phosphorylation/drug effects , Protein Phosphatase 1/genetics , Protein Processing, Post-Translational , Recombinant Proteins/metabolism , Signal Transduction
9.
J Biol Chem ; 292(36): 14827-14835, 2017 09 08.
Article in English | MEDLINE | ID: mdl-28726642

ABSTRACT

The breakdown of stored fat deposits into its components is a highly regulated process that maintains plasma levels of free fatty acids to supply energy to cells. Insulin-mediated transcription of Atgl, the enzyme that mediates the rate-limiting step in lipolysis, is a key point of this regulation. Under conditions such as obesity or insulin resistance, Atgl transcription is often misregulated, which can contribute to overall disease progression. The mechanisms by which Atgl is induced during adipogenesis are not fully understood. We utilized computational approaches to identify putative transcriptional regulatory elements in Atgl and then tested the effect of these elements and the transcription factors that bind to them in cultured preadipocytes and mature adipocytes. Here we report that Atgl is down-regulated by the basal transcription factor Sp1 in preadipocytes and that the magnitude of down-regulation depends on interactions between Sp1 and peroxisome proliferator-activated receptor γ (PPARγ). In mature adipocytes, when PPARγ is abundant, PPARγ abrogated transcriptional repression by Sp1 at the Atgl promoter and up-regulated Atgl mRNA expression. Targeting the PPARγ-Sp1 interaction could be a potential therapeutic strategy to restore insulin sensitivity by modulating Atgl levels in adipocytes.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis/genetics , Lipase/genetics , PPAR gamma/metabolism , Sp1 Transcription Factor/metabolism , Transcription, Genetic , 3T3-L1 Cells , Animals , Cell Line , Down-Regulation , HEK293 Cells , Humans , Lipase/metabolism , Mice , Mice, Inbred C57BL , Mutagenesis, Site-Directed
10.
Curr Opin Clin Nutr Metab Care ; 21(1): 30-36, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29035972

ABSTRACT

PURPOSE OF REVIEW: Skeletal muscle loss or sarcopenia is a frequent complication of cirrhosis that adversely affects clinical outcomes. As skeletal muscle is the largest store of proteins in the body, proteostasis or protein homeostasis is required for maintenance of muscle mass. This review will focus on disordered skeletal muscle proteostasis in liver disease. RECENT FINDINGS: Increased skeletal muscle uptake of ammonia initiates responses that result in disordered proteostasis including impaired protein synthesis and increased autophagy. The cellular response to the stress of hyperammonemia (hyperammonemic stress response, HASR) involves the coordinated action of diverse signaling pathways targeting the molecular mechanisms of regulation of protein synthesis. Transcriptional upregulation of myostatin, a TGFß superfamily member, results in impaired mTORC1 signaling. Phosphorylation of the eukaryotic translation initiation factor 2α (eIF2α) also relates to decreased global protein synthesis rates and mTORC1 signaling. Ammonia also causes mitochondrial and bioenergetic dysfunction because of cataplerosis of α-ketoglutarate. Lowering ammonia, targeting components of HASR and regulating cellular amino acid levels can potentially restore proteostasis. SUMMARY: Signaling via myostatin and eIF2α phosphorylation causes decreases in protein synthesis and mTORC1 activity with a parallel mitochondrial dysfunction and increased autophagy contributing to proteostasis perturbations during skeletal muscle hyperammonemia of liver disease.


Subject(s)
Fibrosis/physiopathology , Hyperammonemia/etiology , Liver/metabolism , Models, Biological , Muscle, Skeletal/metabolism , Proteostasis Deficiencies/etiology , Sarcopenia/etiology , Animals , Autophagy , Energy Metabolism , Fibrosis/metabolism , Humans , Liver/physiopathology , Muscle, Skeletal/physiopathology
11.
Hum Mol Genet ; 24(21): 6240-53, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26307088

ABSTRACT

The cancer epigenome exhibits global loss of DNA methylation, which contributes to genomic instability and aberrant gene expression by mechanisms that are yet to be fully elucidated. We previously discovered over 3300 long non-coding (lnc)RNAs in human cells and demonstrated that specific lncRNAs regulate gene expression via interactions with chromatin-modifying complexes. Here, we tested whether lncRNAs could also associate with DNA methyltransferases to regulate DNA methylation and gene expression. Using RIP-seq, we identified a subset of lncRNAs that interact with the DNA methyltransferase DNMT1 in a colon cancer cell line, HCT116. One lncRNA, TCONS_00023265, which we named DACOR1 (DNMT1-associated Colon Cancer Repressed lncRNA 1), shows high, tissue-specific expression in the normal colon (including colon crypts) but was repressed in a panel of colon tumors and patient-derived colon cancer cell lines. We identified the genomic occupancy sites of DACOR1, which we found to significantly overlap with known differentially methylated regions (DMRs) in colon tumors. Induction of DACOR1 in colon cancer cell lines significantly reduced their ability to form colonies in vitro, suggesting a growth suppressor function. Consistent with the observed phenotype, induction of DACOR1 led to the activation of tumor-suppressor pathways and attenuation of cancer-associated metabolic pathways. Notably, DACOR1 induction resulted in down-regulation of Cystathionine ß-synthase, which is known to lead to increased levels of S-adenosyl methionine-the key methyl donor for DNA methylation. Collectively, our results demonstrate that deregulation of DNMT1-associated lncRNAs contributes to aberrant DNA methylation and gene expression during colon tumorigenesis.


Subject(s)
Colonic Neoplasms/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , Gene Expression Regulation, Neoplastic , RNA, Long Noncoding , Cell Line, Tumor , Chromatin/metabolism , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA, Neoplasm/metabolism , Down-Regulation , Genome, Human , HCT116 Cells , Humans , Intestinal Mucosa/physiology
12.
J Biol Chem ; 290(50): 29761-8, 2015 Dec 11.
Article in English | MEDLINE | ID: mdl-26463210

ABSTRACT

In mammalian cells, mature tRNAs are cleaved by stress-activated ribonuclease angiogenin to generate 5'- and 3'-tRNA halves: a novel class of small non-coding RNAs of 30-40 nucleotides in length. The biogenesis and biological functions of tRNA halves are emerging areas of research. This review will discuss the most recent findings on: (i) the mechanism and regulation of their biogenesis, (ii) their mechanism of action (we will specifically discuss their role in the protein synthesis inhibition and the intrinsic pathway of apoptosis), and (iii) their effects on the human physiology and disease conditions.


Subject(s)
Neurodegenerative Diseases/genetics , RNA, Transfer/genetics , Stress, Physiological , Humans , Hydrolysis , Neurons/metabolism
13.
J Biol Chem ; 290(29): 17822-17837, 2015 Jul 17.
Article in English | MEDLINE | ID: mdl-26041779

ABSTRACT

Cells respond to shrinkage induced by increased extracellular osmolarity via programmed changes in gene transcription and mRNA translation. The immediate response to this stress includes the induction of expression of the neutral amino acid transporter SNAT2. Increased SNAT2-mediated uptake of neutral amino acids is an essential adaptive mechanism for restoring cell volume. In contrast, stress-induced phosphorylation of the α subunit of the translation initiation factor eIF2 (eIF2α) can promote apoptosis. Here we show that the response to mild hyperosmotic stress involves regulation of the phosphorylation of eIF2α by increased levels of GADD34, a regulatory subunit of protein phosphatase 1 (PP1). The induction of GADD34 was dependent on transcriptional control by the c-Jun-binding cAMP response element in the GADD34 gene promoter and posttranscriptional stabilization of its mRNA. This mechanism differs from the regulation of GADD34 expression by other stresses that involve activating transcription factor 4 (ATF4). ATF4 was not translated during hyperosmotic stress despite an increase in eIF2α phosphorylation. The SNAT2-mediated increase in amino acid uptake was enhanced by increased GADD34 levels in a manner involving decreased eIF2α phosphorylation. It is proposed that the induction of the SNAT2/GADD34 axis enhances cell survival by promoting the immediate adaptive response to stress.


Subject(s)
Amino Acid Transport System A/metabolism , Osmotic Pressure , Protein Phosphatase 1/metabolism , Animals , Cell Line , Cell Survival , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Mice , Phosphorylation , Promoter Regions, Genetic , Protein Phosphatase 1/genetics
14.
J Hepatol ; 65(5): 929-937, 2016 11.
Article in English | MEDLINE | ID: mdl-27318325

ABSTRACT

BACKGROUND & AIMS: Increased skeletal muscle ammonia uptake with loss of muscle mass adversely affects clinical outcomes in cirrhosis. Hyperammonemia causes reduced protein synthesis and sarcopenia but the cellular responses to impaired proteostasis and molecular mechanism of l-leucine induced adaptation to ammonia induced stress were determined. METHODS: Response to activation of amino acid deficiency sensor, GCN2, in the skeletal muscle from cirrhotic patients and the portacaval anastomosis (PCA) rat were quantified. During hyperammonemia and l-leucine supplementation, protein synthesis, phosphorylation of eIF2α, mTORC1 signaling, l-leucine transport and response to l-leucine supplementation were quantified. Adaptation to cellular stress via ATF4 and its target GADD34 were also determined. RESULTS: Activation of the eIF2α kinase GCN2 and impaired mTORC1 signaling were observed in skeletal muscle from cirrhotic patients and PCA rats. Ammonia activated GCN2 mediated eIF2α phosphorylation (eIF2α-P) and impaired mTORC1 signaling that inhibit protein synthesis in myotubes and MEFs. Adaptation to ammonia induced stress did not involve translational reprogramming by activation transcription factor 4 (ATF4) dependent induction of the eIF2α-P phosphatase subunit GADD34. Instead, ammonia increased expression of the leucine/glutamine exchanger SLC7A5, l-leucine uptake and intracellular l-leucine levels, the latter not being sufficient to rescue the inhibition of protein synthesis, due to potentially enhanced mitochondrial sequestration of l-leucine. l-leucine supplementation rescued protein synthesis inhibition caused by hyperammonemia. CONCLUSIONS: Response to hyperammonemia is reminiscent of the cellular response to amino acid starvation, but lacks the adaptive ATF4 dependent integrated stress response (ISR). Instead, hyperammonemia-induced l-leucine uptake was an adaptive response to the GCN2-mediated decreased protein synthesis. LAY SUMMARY: Sarcopenia or skeletal muscle loss is the most frequent complication in cirrhosis but there are no treatments because the cause(s) of muscle loss in liver disease are not known. Results from laboratory experiments in animals and muscle cells were validated in human patients with cirrhosis to show that ammonia plays a key role in causing muscle loss in patients with cirrhosis. We identified a novel stress response to ammonia in the muscle that decreases muscle protein content that can be reversed by supplementation with the amino acid l-leucine.


Subject(s)
Hyperammonemia , Animals , Humans , Leucine , Liver Cirrhosis , Muscle, Skeletal , Phosphorylation , Rats , Sarcopenia
15.
Gastroenterology ; 148(7): 1405-1416.e3, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25701737

ABSTRACT

BACKGROUND & AIMS: Defects in colonic epithelial barrier defenses are associated with ulcerative colitis (UC). The proteins that regulate bacterial clearance in the colonic epithelium have not been completely identified. The Drosophila chromosome-associated protein D3 (dCAP-D3) regulates responses to bacterial infection. We examined whether CAP-D3 promotes bacterial clearance in human colonic epithelium. METHODS: Clearance of Salmonella or adherent-invasive Escherichia coli LF82 was assessed by gentamycin protection assays in HT-29 and Caco-2 cells expressing small hairpin RNAs against CAP-D3. We used immunoblot assays to measure levels of CAP-D3 in colonic epithelial cells from patients with UC and healthy individuals (controls). RNA sequencing identified genes activated by CAP-D3. We analyzed the roles of CAP-D3 target genes in bacterial clearance using gentamycin protection and immunofluorescence assays and studies with pharmacologic inhibitors. RESULTS: CAP-D3 expression was reduced in colonic epithelial cells from patients with active UC. Reduced CAP-D3 expression decreased autophagy and impaired intracellular bacterial clearance by HT-29 and Caco-2 colonic epithelial cells. Lower levels of CAP-D3 increased transcription of genes encoding SLC7A5 and SLC3A2, the products of which heterodimerize to form an amino acid transporter in HT-29 cells after bacterial infection; levels of SLC7A5-SLC3A2 were increased in tissues from patients with UC compared with controls. Reduced CAP-D3 in HT-29 cells resulted in earlier recruitment of SLC7A5 to Salmonella-containing vacuoles, increased activity of mTORC1, and increased survival of bacteria. Inhibition of SLC7A5-SLC3A2 or mTORC1 activity rescued the bacterial clearance defects of CAP-D3-deficient cells. CONCLUSIONS: CAP-D3 down-regulates transcription of genes that encode amino acid transporters (SLC7A5 and SLC3A2) to promote bacterial autophagy by colon epithelial cells. Levels of CAP-D3 protein are reduced in patients with active UC; strategies to increase its levels might restore mucosal homeostasis to patients with active UC.


Subject(s)
Cell Cycle Proteins/metabolism , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Escherichia coli/physiology , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Large Neutral Amino Acid-Transporter 1/metabolism , Salmonella/physiology , Adenosine Triphosphatases , Autophagy , Caco-2 Cells , Cell Cycle Proteins/genetics , Colitis, Ulcerative/immunology , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/microbiology , Crohn Disease/immunology , Crohn Disease/metabolism , Crohn Disease/microbiology , Drosophila Proteins , Epithelial Cells/immunology , Escherichia coli/immunology , Fusion Regulatory Protein 1, Heavy Chain/genetics , Gene Expression Regulation , HT29 Cells , Humans , Immunity, Innate , Intestinal Mucosa/immunology , Large Neutral Amino Acid-Transporter 1/genetics , Mechanistic Target of Rapamycin Complex 1 , Microbial Viability , Multiprotein Complexes/metabolism , RNA Interference , Salmonella/immunology , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Time Factors , Transcription, Genetic , Transfection
16.
J Biol Chem ; 289(18): 12593-611, 2014 May 02.
Article in English | MEDLINE | ID: mdl-24648524

ABSTRACT

The accumulation of unfolded/misfolded proteins in the endoplasmic reticulum (ER) causes stress to which an unfolded protein response is activated to render cell survival or apoptosis (chronic stress). Transcriptional and translational reprogramming is tightly regulated during the unfolded protein response to ensure specific gene expression. The master regulator of this response is the PERK/eIF2α/ATF4 signaling where eIF2α is phosphorylated (eIF2α-P) by the kinase PERK. This signal leads to global translational shutdown, but it also enables translation of the transcription factor ATF4 mRNA. We showed recently that ATF4 induces an anabolic program through the up-regulation of selected amino acid transporters and aminoacyl-tRNA synthetases. Paradoxically, this anabolic program led cells to apoptosis during chronic ER stress in a manner that involved recovery from stress-induced protein synthesis inhibition. By using eIF2α-P-deficient cells as an experimental system, we identified a communicating network of signaling pathways that contribute to the inhibition of protein synthesis during chronic ER stress. This eIF2α-P-independent network includes (i) inhibition of mammalian target of rapamycin kinase protein complex 1 (mTORC1)-targeted protein phosphorylation, (ii) inhibited translation of a selective group of 5'-terminal oligopyrimidine mRNAs (encoding proteins involved in the translation machinery and translationally controlled by mTORC1 signaling), and (iii) inhibited translation of non-5'-terminal oligopyrimidine ribosomal protein mRNAs and ribosomal RNA biogenesis. We propose that the PERK/eIF2α-P/ATF4 signaling acts as a brake in the decline of protein synthesis during chronic ER stress by positively regulating signaling downstream of the mTORC1 activity. These studies advance our knowledge on the complexity of the communicating signaling pathways in controlling protein synthesis rates during chronic stress.


Subject(s)
Endoplasmic Reticulum Stress , Eukaryotic Initiation Factor-2/metabolism , Fibroblasts/metabolism , Protein Biosynthesis , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Amino Acids/metabolism , Amino Acyl-tRNA Synthetases/metabolism , Animals , Autophagy-Related Protein 5 , Blotting, Western , Calcium-Transporting ATPases/antagonists & inhibitors , Calcium-Transporting ATPases/metabolism , Cells, Cultured , Embryo, Mammalian/cytology , Eukaryotic Initiation Factor-2/genetics , Fibroblasts/cytology , Fibroblasts/drug effects , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Phosphorylation , Polyribosomes/metabolism , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Thapsigargin/pharmacology , Time Factors , eIF-2 Kinase/metabolism
17.
Nature ; 457(7231): 915-9, 2009 Feb 12.
Article in English | MEDLINE | ID: mdl-19098893

ABSTRACT

Ligand binding to structural elements in the non-coding regions of messenger RNA modulates gene expression. Ligands such as free metabolites or other small molecules directly bind and induce conformational changes in regulatory RNA elements known as riboswitches. Other types of RNA switches are activated by complexed metabolites-for example, RNA-ligated metabolites such as aminoacyl-charged transfer RNA in the T-box system, or protein-bound metabolites in the glucose- or amino-acid-stimulated terminator-anti-terminator systems. All of these switch types are found in bacteria, fungi and plants. Here we report an RNA switch in human vascular endothelial growth factor-A (VEGFA, also known as VEGF) mRNA 3' untranslated region (UTR) that integrates signals from interferon (IFN)-gamma and hypoxia to regulate VEGFA translation in myeloid cells. Analogous to riboswitches, the VEGFA 3' UTR undergoes a binary conformational change in response to environmental signals. However, the VEGFA 3' UTR switch is metabolite-independent, and the conformational change is dictated by mutually exclusive, stimulus-dependent binding of proteins, namely, the IFN-gamma-activated inhibitor of translation complex and heterogeneous nuclear ribonucleoprotein L (HNRNPL, also known as hnRNP L). We speculate that the VEGFA switch represents the founding member of a family of signal-mediated, protein-dependent RNA switches that evolved to regulate gene expression in multicellular animals in which the precise integration of disparate inputs may be more important than the rapidity of response.


Subject(s)
Gene Expression Regulation , RNA/metabolism , Stress, Physiological/physiology , Vascular Endothelial Growth Factor A/metabolism , 3' Untranslated Regions , Amino Acyl-tRNA Synthetases , Gene Silencing , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Humans , Hypoxia/metabolism , Interferon-gamma/metabolism , Myeloid Cells/metabolism , Myeloid Cells/physiology , RNA/chemistry , Signal Transduction , U937 Cells , Vascular Endothelial Growth Factor A/genetics
18.
J Biol Chem ; 288(24): 17202-13, 2013 Jun 14.
Article in English | MEDLINE | ID: mdl-23645676

ABSTRACT

Endoplasmic reticulum (ER) stress-induced responses are associated with the loss of insulin-producing ß-cells in type 2 diabetes mellitus. ß-Cell survival during ER stress is believed to depend on decreased protein synthesis rates that are mediated via phosphorylation of the translation initiation factor eIF2α. It is reported here that chronic ER stress correlated with increased islet protein synthesis and apoptosis in ß-cells in vivo. Paradoxically, chronic ER stress in ß-cells induced an anabolic transcription program to overcome translational repression by eIF2α phosphorylation. This program included expression of amino acid transporter and aminoacyl-tRNA synthetase genes downstream of the stress-induced ATF4-mediated transcription program. The anabolic response was associated with increased amino acid flux and charging of tRNAs for branched chain and aromatic amino acids (e.g. leucine and tryptophan), the levels of which are early serum indicators of diabetes. We conclude that regulation of amino acid transport in ß-cells during ER stress involves responses leading to increased protein synthesis, which can be protective during acute stress but can lead to apoptosis during chronic stress. These studies suggest that the increased expression of amino acid transporters in islets can serve as early diagnostic biomarkers for the development of diabetes.


Subject(s)
Amino Acids/metabolism , Apoptosis , Diabetes Mellitus, Type 2/metabolism , Endoplasmic Reticulum Stress , Insulin-Secreting Cells/physiology , Activating Transcription Factor 4/metabolism , Amino Acid Transport Systems/genetics , Amino Acid Transport Systems/metabolism , Amino Acyl-tRNA Synthetases/genetics , Amino Acyl-tRNA Synthetases/metabolism , Animals , Cell Survival , Diabetes Mellitus, Type 2/pathology , Eukaryotic Initiation Factor-2/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Biosynthesis , Protein Processing, Post-Translational , RNA, Transfer/metabolism , Transcriptional Activation
19.
Cell Physiol Biochem ; 33(5): 1591-606, 2014.
Article in English | MEDLINE | ID: mdl-24854847

ABSTRACT

BACKGROUND: Despite homeostatic pH regulation, systemic and cellular pH changes take place and strongly influence metabolic processes. Transcription of the glutamine transporter SNAT3 (Slc38a3) for instance is highly up-regulated in the kidney during metabolic acidosis to provide glutamine for ammonia production. METHODS: Slc38a3 promoter activity and messenger RNA stability were measured in cultured cells in response to different extracellular pH values. RESULTS: Up-regulation of SNAT3 mRNA was mediated both by the stabilization of its mRNA and by the up-regulation of gene transcription. Stabilisation of the mRNA involved a pH-response element, while enhanced transcription made use of a second pH-sensitive Sp1 binding site in addition to a constitutive Sp1 binding site. Transcriptional regulation dominated the early response to acidosis, while mRNA stability was more important for chronic adaptation. Tissue-specific expression of SNAT3, by contrast, appeared to be controlled by promoter methylation and histone modifications. CONCLUSIONS: Regulation of SNAT3 gene expression by extracellular pH involves post-transcriptional and transcriptional mechanisms, the latter being distinct from the mechanisms that control the tissue-specific expression of the gene.


Subject(s)
Acidosis/metabolism , Amino Acid Transport Systems, Neutral/biosynthesis , Amino Acid Transport Systems, Neutral/genetics , Acidosis/genetics , Animals , Cells, Cultured , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , Hydrogen-Ion Concentration , Mice , Organ Specificity/genetics , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic/genetics
20.
bioRxiv ; 2024 May 14.
Article in English | MEDLINE | ID: mdl-38798645

ABSTRACT

Vesicle-associated membrane protein-associated protein-B (VAPB) is an ER membrane bound protein. VAPB P56S causes a dominant, familial form of amyotrophic lateral sclerosis (ALS), however, the mechanism through which this mutation causes motor neuron (MN) disease remains unknown. Using inducible wild type (WT) and VAPB P56S expressing iPSC-derived MNs we show that VAPB P56S, but not WT, protein decreased neuronal firing and mitochondrial-ER contact (MERC) with an associated age-dependent decrease in mitochondrial membrane potential (MMP); all typical characteristics of MN-disease. We further show that VAPB P56S expressing iPSC-derived MNs have enhanced age-dependent sensitivity to ER stress. We identified elevated expression of the master regulator of the Integrated Stress Response (ISR) marker ATF4 and decreased protein synthesis in the VAPB P56S iPSC-derived MNs. Chemical inhibition of ISR with the compound, ISRIB, rescued all MN disease phenotype in VAPB P56S MNs. Thus, our results not only support ISR inhibition as a potential therapeutic target for ALS patients, but also provides evidence to pathogenesis.

SELECTION OF CITATIONS
SEARCH DETAIL