Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 139
Filter
Add more filters

Publication year range
1.
Nucleic Acids Res ; 49(4): 2085-2101, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33544858

ABSTRACT

Antibiotic resistance is a serious problem which may be caused by bacterial dormancy. It has been suggested that bacterial toxin-antitoxin systems induce dormancy. We analyzed the genome-wide role of Staphylococcus aureus endoribonuclease toxin MazF using RNA-Seq, Ribo-Seq and quantitative proteomics. We characterized changes in transcriptome, translatome and proteome caused by MazF, and proposed that MazF decreases translation directly by cleaving mRNAs, and indirectly, by decreasing translation factors and by promoting ribosome hibernation. Important pathways affected during the early stage of MazF induction were identified: MazF increases cell wall thickness and decreases cell division; MazF activates SsrA-system which rescues stalled ribosomes, appearing as a result of MazF mRNA cleavage. These pathways may be promising targets for new antibacterial drugs that prevent bacteria dormancy. Finally, we described the overall impact of MazF on S. aureus cell physiology, and propose one of the mechanisms by which MazF might regulate cellular changes leading to dormancy.


Subject(s)
Bacterial Toxins/metabolism , Endoribonucleases/physiology , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Bacterial Toxins/biosynthesis , Cell Division/genetics , Cell Wall/genetics , Cell Wall/metabolism , Endoribonucleases/biosynthesis , Endoribonucleases/metabolism , Protein Biosynthesis , Proteome , Staphylococcus aureus/enzymology , Transcriptome
2.
Protein Expr Purif ; 190: 105987, 2022 02.
Article in English | MEDLINE | ID: mdl-34637916

ABSTRACT

Combinations of ribonucleases (RNases) are commonly used to digest RNA into oligoribonucleotide fragments prior to liquid chromatography-mass spectrometry (LC-MS) analysis. The distribution of the RNase target sequences or nucleobase sites within an RNA molecule is critical for achieving a high mapping coverage. Cusativin and MC1 are nucleotide-specific endoribonucleases encoded in the cucumber and bitter melon genomes, respectively. Their high specificity for cytidine (Cusativin) and uridine (MC1) make them ideal molecular biology tools for RNA modification mapping. However, heterogenous recombinant expression of either enzyme has been challenging because of their high toxicity to expression hosts and the requirement of posttranslational modifications. Here, we present two highly efficient and time-saving protocols that overcome these hurdles and enhance the expression and purification of these RNases. We first purified MC1 and Cusativin from bacteria by expressing and shuttling both enzymes to the periplasm as MBP-fusion proteins in T7 Express lysY/IqE. coli strain at low temperature. The RNases were enriched using amylose affinity chromatography, followed by a subsequent purification via a C-terminal 6xHIS tag. This fast, two-step purification allows for the purification of highly active recombinant RNases significantly surpassing yields reported in previous studies. In addition, we expressed and purified a Cusativin-CBD fusion enzyme in P. pastoris using chitin magnetic beads. Both Cusativin variants exhibited a similar sequence preference, suggesting that neither posttranslational modifications nor the epitope-tags have a substantial effect on the sequence specificity of the enzyme.


Subject(s)
Endoribonucleases , Escherichia coli , Gene Expression , Ribonucleases , Endoribonucleases/biosynthesis , Endoribonucleases/chemistry , Endoribonucleases/genetics , Endoribonucleases/isolation & purification , Escherichia coli/genetics , Escherichia coli/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Ribonucleases/biosynthesis , Ribonucleases/chemistry , Ribonucleases/genetics , Ribonucleases/isolation & purification
3.
Nucleic Acids Res ; 46(13): 6726-6745, 2018 07 27.
Article in English | MEDLINE | ID: mdl-29893914

ABSTRACT

Expression of CRISPR-Cas systems is a prerequisite for their defensive role against invading genetic elements. Yet, much remains unknown about how this crucial step is regulated. We describe a new mechanism controlling CRISPR-cas expression, which requires an extracytoplasmic function (ECF) σ factor (DdvS), its membrane-bound anti-σ (DdvA) and a global regulatory complex (CarD-CarG). Transcriptomic analyses revealed that the DdvS/CarD/CarG-dependent regulon comprises a type III-B CRISPR-Cas system in Myxococcus xanthus. We mapped four DdvS-driven CarD/CarG-dependent promoters, with one lying immediately upstream of the cas cluster. Consistent with direct action, DdvS and CarD-CarG localize at these promoters in vivo. The cas genes are transcribed as a polycistronic mRNA that reads through the leader into the CRISPR array, a putative σA-dependent promoter in the leader having negligible activity in vivo. Consequently, expression of the entire CRISPR-Cas system and mature CRISPR-RNA (crRNA) production is DdvS/CarD/CarG-dependent. DdvA likely uses its large C-terminal domain to sense and transduce the extracytoplasmic signal triggering CRISPR-cas expression, which we show is not starvation-induced multicellular development. An ECF-σ/anti-σ pair and a global regulatory complex provide an effective mechanism to coordinate signal-sensing with production of precursor crRNA, its processing Cas6 endoribonuclease and other Cas proteins for mature crRNA biogenesis and interference.


Subject(s)
CRISPR-Associated Proteins/genetics , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Endoribonucleases/genetics , Gene Expression Regulation, Bacterial , Myxococcus xanthus/genetics , Sigma Factor/metabolism , Bacterial Proteins/metabolism , CRISPR-Associated Proteins/metabolism , Endoribonucleases/biosynthesis , Endoribonucleases/metabolism , Myxococcus xanthus/metabolism , Operon , Promoter Regions, Genetic , RNA, Bacterial/metabolism , Regulon , Trans-Activators/metabolism , Transcription, Genetic
4.
J Cell Physiol ; 234(12): 23667-23674, 2019 12.
Article in English | MEDLINE | ID: mdl-31188482

ABSTRACT

Up to date, the mechanism of gastric cancer (GC) development is poorly understood. This study was to demonstrate the effects of LINC00339 on GC progression. Here, we found that LINC00339 was overexpressed expressed in GC tissues and predicted poor outcome. By CCK8, colony formation and Transwell assays, we showed LINC00339 knockdown suppressed GC cell proliferation, migration, and invasion in vitro. Flow cytometry analysis (FACS) indicated that LINC00339 knockdown induced tumor cell apoptosis. Besides, we utilized the xenograft assay and found that LINC00339 depletion led to decreased tumor growth in vivo. Mechanistically, miR-377-3p was found to be inhibited by LINC00339. And LINC00339 suppressed miR-377-3p to upregulate DCP1A, which consequently promoted GC progression. In conclusion, LINC00339 promotes gastric cancer progression by elevating DCP1A expression via inhibiting miR-377-3p.


Subject(s)
Endoribonucleases/biosynthesis , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Stomach Neoplasms/pathology , Trans-Activators/biosynthesis , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Disease Progression , Humans , Neoplasm Invasiveness/genetics , Neoplasm Transplantation , Oncogenes/genetics , RNA Interference , RNA, Small Interfering/genetics , Stomach Neoplasms/genetics , Transplantation, Heterologous
5.
Alcohol Clin Exp Res ; 43(1): 69-78, 2019 01.
Article in English | MEDLINE | ID: mdl-30403409

ABSTRACT

BACKGROUND: Ethanol (EtOH) exposure during pregnancy may result in fetal alcohol spectrum disorders (FASD). One of the most deleterious consequences of EtOH exposure is neuronal loss in the developing brain. Previously, we showed that EtOH exposure induced neuroapoptosis in the brain of postnatal day 4 (PD4) mice but not PD12 mice. This differential susceptibility may result from an insufficient cellular stress response system such as unfolded protein response (also known as endoplasmic reticulum [ER] stress) in PD4 mice. In this study, we compared the effect of EtOH on ER stress in PD4 and PD12 mice and determined whether the inhibition of ER stress could protect the developing brain against EtOH damage. METHODS: We used a third-trimester equivalent mouse model of FASD. PD4 and PD12 C57BL/6 mice were subcutaneously injected with saline (control), EtOH, EtOH plus 4-phenylbutyric acid (4-PBA), a chemical chaperone known as ER stress inhibitor, and 4-PBA alone. The expression of apoptosis marker, ER stress markers, and markers for glial cell activation was examined in the cerebral cortex. RESULTS: EtOH induced neuroapoptosis and increased the expression of ER stress markers, such as activating transcription factor 6, 78-kDa glucose-regulated protein, inositol-requiring enzyme 1α, mesencephalic astrocyte-derived neurotrophic factor, and caspase-12 in PD4 but not PD12 mice. EtOH exposure also activated microglia and astrocytes. Interestingly, treatment with 4-PBA attenuated EtOH-induced neuroapoptosis. Moreover, 4-PBA inhibited the expression of the aforementioned ER stress markers and EtOH-induced glial activation in PD4 mice. CONCLUSIONS: ER stress plays an important role in EtOH-induced damage to the developing brain. Inhibition of ER stress is neuroprotective and may provide a new therapeutic strategy for treating FASD.


Subject(s)
Aging/metabolism , Apoptosis/drug effects , Biomarkers/metabolism , Endoplasmic Reticulum Stress/drug effects , Ethanol/antagonists & inhibitors , Phenylbutyrates/pharmacology , Activating Transcription Factor 6/biosynthesis , Animals , Astrocytes/metabolism , Caspase 12/biosynthesis , Cerebral Cortex/metabolism , Endoplasmic Reticulum Chaperone BiP , Endoribonucleases/biosynthesis , Ethanol/adverse effects , Female , Heat-Shock Proteins/biosynthesis , Male , Mice , Microglia/metabolism , Nerve Growth Factors/biosynthesis , Neuroprotective Agents/pharmacology , Protein Serine-Threonine Kinases/biosynthesis
6.
PLoS Genet ; 12(10): e1006395, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27768707

ABSTRACT

In eukaryotes, Dom34 upregulates translation by securing levels of activatable ribosomal subunits. We found that in the yeast Saccharomyces cerevisiae and the human fungal pathogen Candida albicans, Dom34 interacts genetically with Pmt1, a major isoform of protein O-mannosyltransferase. In C. albicans, lack of Dom34 exacerbated defective phenotypes of pmt1 mutants, while they were ameliorated by Dom34 overproduction that enhanced Pmt1 protein but not PMT1 transcript levels. Translational effects of Dom34 required the 5'-UTR of the PMT1 transcript, which bound recombinant Dom34 directly at a CA/AC-rich sequence and regulated in vitro translation. Polysomal profiling revealed that Dom34 stimulates general translation moderately, but that it is especially required for translation of transcripts encoding Pmt isoforms 1, 4 and 6. Because defective protein N- or O-glycosylation upregulates transcription of PMT genes, it appears that Dom34-mediated specific translational upregulation of the PMT transcripts optimizes cellular responses to glycostress. Its translational function as an RNA binding protein acting at the 5'-UTR of specific transcripts adds another facet to the known ribosome-releasing functions of Dom34 at the 3'-UTR of transcripts.


Subject(s)
Candida albicans/genetics , Cell Cycle Proteins/genetics , Endoribonucleases/genetics , Mannosyltransferases/genetics , Protein Biosynthesis/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Candida albicans/growth & development , Cell Cycle Proteins/biosynthesis , Endoribonucleases/biosynthesis , Glycosylation , Humans , Oligonucleotides/genetics , Phenotype , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Ribosomes/genetics , Ribosomes/metabolism , Saccharomyces cerevisiae Proteins/biosynthesis
7.
J Neuroinflammation ; 15(1): 32, 2018 Feb 02.
Article in English | MEDLINE | ID: mdl-29394934

ABSTRACT

BACKGROUND: The endoplasmic reticulum (ER) is responsible for the control of correct protein folding and protein function which is crucial for cell survival. However, under pathological conditions, such as hypoxia-ischemia (HI), there is an accumulation of unfolded proteins thereby triggering the unfolded protein response (UPR) and causing ER stress which is associated with activation of several stress sensor signaling pathways, one of them being the inositol requiring enzyme-1 alpha (IRE1α) signaling pathway. The UPR is regarded as a potential contributor to neuronal cell death and inflammation after HI. In the present study, we sought to investigate whether microRNA-17 (miR-17), a potential IRE1α ribonuclease (RNase) substrate, arbitrates downregulation of thioredoxin-interacting protein (TXNIP) and consequent NLRP3 inflammasome activation in the immature brain after HI injury and whether inhibition of IRE1α may attenuate inflammation via miR-17/TXNIP regulation. METHODS: Postnatal day 10 rat pups (n = 287) were subjected to unilateral carotid artery ligation followed by 2.5 h of hypoxia (8% O2). STF-083010, an IRE1α RNase inhibitor, was intranasally delivered at 1 h post-HI or followed by an additional one administration per day for 2 days. MiR-17-5p mimic or anti-miR-17-5p inhibitor was injected intracerebroventricularly at 48 h before HI. Infarct volume and body weight were used to evaluate the short-term effects while brain weight, gross and microscopic brain tissue morphologies, and neurobehavioral tests were conducted for the long-term evaluation. Western blots, immunofluorescence staining, reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR), and co-immunoprecipitation (Co-IP) were used for mechanism studies. RESULTS: Endogenous phosphorylated IRE1α expression was significantly increased after HI. Intranasal administration of STF-083010 alleviated brain injury and improved neurological behavior. MiR-17-5p expression was reduced after HI, and this decrease was attenuated by STF-083010 treatment. MiR-17-5p mimic administration ameliorated TXNIP expression, NLRP3 inflammasome activation, caspase-1 cleavage, and IL-1ß production, as well as brain infarct volume. Conversely, anti-miR-17-5p inhibitor reversed IRE1α inhibition-induced decrease in TXNIP expression and inflammasome activation, as well as exacerbated brain injury after HI. CONCLUSIONS: IRE1a-induced UPR pathway may contribute to inflammatory activation and brain injury following neonatal HI. IRE1a activation, through decay of miR-17-5p, elevated TXNIP expression to activate NLRP3 inflammasome and aggravated brain damage.


Subject(s)
Carrier Proteins/metabolism , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/biosynthesis , Hypoxia-Ischemia, Brain/metabolism , MicroRNAs/metabolism , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/biosynthesis , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/biosynthesis , Administration, Intranasal , Animals , Animals, Newborn , Carrier Proteins/antagonists & inhibitors , Cell Cycle Proteins , Hypoxia-Ischemia, Brain/drug therapy , Inflammasomes/antagonists & inhibitors , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Sulfonamides/administration & dosage , Thiophenes/administration & dosage
8.
Rheumatology (Oxford) ; 57(6): 1021-1032, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29534223

ABSTRACT

Objectives: Labial salivary glands (LSGs) of SS patients show alterations related to endoplasmic reticulum stress. Glandular dysfunction could be partly the consequence of an altered inositol-requiring enzyme 1α (IRE1α)/X box-binding protein 1 (XBP-1) signalling pathway of the unfolded protein response, which then regulates genes involved in biogenesis of the secretory machinery. This study aimed to determine the expression, promoter methylation and localization of the IRE1α/XBP-1 pathway components in LSGs of SS patients and also their expression induced by IFN-γ in vitro. Methods: IRE1α, XBP-1 and glucose-regulated protein 78 (GRP78) mRNA and protein levels were measured by qPCR and western blot, respectively, in LSGs of SS patients (n = 47) and control subjects (n = 37). Methylation of promoters was evaluated by methylation-sensitive high resolution melting, localization was analysed by immunofluorescence and induction of the IRE1α/XBP-1 pathway components by IFN-γ was evaluated in 3D acini. Results: A significant decrease of IRE1α, XBP-1u, XBP-1s, total XBP-1 and GRP78 mRNAs was observed in LSGs of SS patients, which was correlated with increased methylation levels of their respective promoters, and consistently the protein levels for IRE1α, XBP-1s and GRP78 were observed to decrease. IFN-γ decreased the mRNA and protein levels of XBP-1s, IRE1α and GRP78, and increased methylation of their promoters. Significant correlations were also found between IRE1α/XBP-1 pathway components and clinical parameters. Conclusion: Decreased mRNA levels for IRE1α, XBP-1 and GRP78 can be partially explained by hypermethylation of their promoters and is consistent with chronic endoplasmic reticulum stress, which may explain the glandular dysfunction observed in LSGs of SS patients. Additionally, glandular stress signals, including IFN-γ, could modulate the expression of the IRE1α/XBP-1 pathway components.


Subject(s)
Endoribonucleases/genetics , Gene Expression Regulation , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics , Salivary Glands/physiopathology , Sjogren's Syndrome/genetics , X-Box Binding Protein 1/genetics , Adult , Aged , Blotting, Western , DNA Methylation , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Endoribonucleases/biosynthesis , Female , Humans , Immunohistochemistry , Male , Middle Aged , Polymerase Chain Reaction , Protein Serine-Threonine Kinases/biosynthesis , Salivary Glands/metabolism , Signal Transduction , Sjogren's Syndrome/metabolism , Sjogren's Syndrome/pathology , X-Box Binding Protein 1/biosynthesis , Young Adult
9.
J Virol ; 90(6): 3160-72, 2016 Jan 06.
Article in English | MEDLINE | ID: mdl-26739051

ABSTRACT

UNLABELLED: The oligoadenylate synthetase (OAS)-RNase L pathway is a potent interferon (IFN)-induced antiviral activity. Upon sensing double-stranded RNA, OAS produces 2',5'-oligoadenylates (2-5A), which activate RNase L. Murine coronavirus (mouse hepatitis virus [MHV]) nonstructural protein 2 (ns2) is a 2',5'-phosphodiesterase (PDE) that cleaves 2-5A, thereby antagonizing RNase L activation. PDE activity is required for robust replication in myeloid cells, as a mutant of MHV (ns2(H126R)) encoding an inactive PDE fails to antagonize RNase L activation and replicates poorly in bone marrow-derived macrophages (BMM), while ns2(H126R) replicates to high titer in several types of nonmyeloid cells, as well as in IFN receptor-deficient (Ifnar1(-/-)) BMM. We reported previously that myeloid cells express significantly higher basal levels of OAS transcripts than nonmyeloid cells. Here, we investigated the contributions of Oas gene expression, basal IFN signaling, and virus-induced IFN to RNase L activation. Infection with ns2(H126R) activated RNase L in Ifih1(-/-) BMM to a similar extent as in wild-type (WT) BMM, despite the lack of IFN induction in the absence of MDA5 expression. However, ns2(H126R) failed to induce RNase L activation in BMM treated with IFNAR1-blocking antibody, as well as in Ifnar1(-/-) BMM, both expressing low basal levels of Oas genes. Thus, activation of RNase L does not require virus-induced IFN but rather correlates with adequate levels of basal Oas gene expression, maintained by basal IFN signaling. Finally, overexpression of RNase L is not sufficient to compensate for inadequate basal OAS levels. IMPORTANCE: The oligoadenylate synthetase (OAS)-RNase L pathway is a potent antiviral activity. Activation of RNase L during murine coronavirus (mouse hepatitis virus [MHV]) infection of myeloid cells correlates with high basal Oas gene expression and is independent of virus-induced interferon secretion. Thus, our data suggest that cells with high basal Oas gene expression levels can activate RNase L and thereby inhibit virus replication early in infection upon exposure to viral double-stranded RNA (dsRNA) before the induction of interferon and prior to transcription of interferon-stimulated antiviral genes. These findings challenge the notion that activation of the OAS-RNase L pathway requires virus to induce type I IFN, which in turn upregulates OAS gene expression, as well as to provide dsRNA to activate OAS. Our data further suggest that myeloid cells may serve as sentinels to restrict viral replication, thus protecting other cell types from infection.


Subject(s)
2',5'-Oligoadenylate Synthetase/metabolism , Endoribonucleases/biosynthesis , Gene Expression , Host-Pathogen Interactions , Murine hepatitis virus/physiology , Myeloid Cells/enzymology , Myeloid Cells/virology , Animals , Cells, Cultured , Mice , Mice, Knockout
10.
Tumour Biol ; 39(6): 1010428317705517, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28618969

ABSTRACT

Pancreatic cancer is one of the most aggressive and difficult to treat cancers. Experimental and clinical evidence suggests that high basal state autophagy in pancreatic tumors could induce resistance to chemotherapy. Recently, we have demonstrated that penfluridol suppresses pancreatic tumor growth by autophagy-mediated apoptosis both in vitro and in vivo; however, the mechanism of autophagy induction by penfluridol was not clear. Several studies have established that endoplasmic reticulum stress could lead to autophagy and inhibit tumor progression. In this study, we demonstrated that penfluridol induced endoplasmic reticulum stress in BxPC-3, AsPC-1, and Panc-1 pancreatic cancer cell lines as indicated by upregulation of endoplasmic reticulum stress markers such as binding protein (BIP), C/EBP homologous protein (CHOP) and inositol requiring 1α (IRE1α) after treatment with penfluridol in a concentration-dependent manner. Inhibiting endoplasmic reticulum stress by pretreatment with pharmacological inhibitors such as sodium phenylbutyrate and mithramycin or by silencing CHOP using CHOP small interfering RNA, blocked penfluridol-induced autophagy. These results clearly indicate that penfluridol-induced endoplasmic reticulum stress lead to autophagy in our model. Western blot analysis of subcutaneously implanted AsPC-1 and BxPC-3 tumors as well as orthotopically implanted Panc-1 tumors demonstrated upregulation of BIP, CHOP, and IRE1α expression in the tumor lysates from penfluridol-treated mice as compared to tumors from control mice. Altogether, our study establishes that penfluridol-induced endoplasmic reticulum stress leads to autophagy resulting in reduced pancreatic tumor growth. Our study opens a new therapeutic target for advanced chemotherapies against pancreatic cancer.


Subject(s)
Endoribonucleases/biosynthesis , Heat-Shock Proteins/genetics , Pancreatic Neoplasms/drug therapy , Penfluridol/administration & dosage , Protein Serine-Threonine Kinases/biosynthesis , Transcription Factor CHOP/biosynthesis , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/genetics , Gene Expression Regulation, Neoplastic , Heat-Shock Proteins/biosynthesis , Humans , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , Transcription Factor CHOP/antagonists & inhibitors , Transcription Factor CHOP/genetics , Xenograft Model Antitumor Assays
11.
J Biol Chem ; 290(38): 23282-90, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26240153

ABSTRACT

Aberrant deposition of fat including free fatty acids in the liver often causes damage to hepatocytes, namely lipotoxicity, which is a key pathogenic event in the development and progression of fatty liver diseases. This study demonstrates a pivotal role of sphingosine kinase 1 (SphK1) in protecting hepatocytes from lipotoxicity. Exposure of primary murine hepatocytes to palmitate resulted in dose-dependent cell death, which was enhanced significantly in Sphk1-deficient cells. In keeping with this, expression of dominant-negative mutant SphK1 also markedly promoted palmitate-induced cell death. In contrast, overexpression of wild-type SphK1 profoundly protected hepatocytes from lipotoxicity. Mechanistically, the protective effect of SphK1 is attributable to suppression of ER stress-mediated pro-apoptotic pathways, as reflected in the inhibition of IRE1α activation, XBP1 splicing, JNK phosphorylation, and CHOP induction. Of note, SphK1 inhibited the IRE1α pathway by reducing IRE1α expression at the transcriptional level. Moreover, S1P mimicked the effect of SphK1, suppressing IRE1α expression in a receptor-dependent manner. Furthermore, enforced overexpression of IRE1α significantly blocked the protective effect of SphK1 against lipotoxicity. Therefore, this study provides new insights into the role of SphK1 in hepatocyte survival and uncovers a novel mechanism for protection against ER stress-mediated cell death.


Subject(s)
Down-Regulation/drug effects , Endoribonucleases/biosynthesis , Enzyme Inhibitors/adverse effects , Hepatocytes/metabolism , Palmitic Acid/adverse effects , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Serine-Threonine Kinases/biosynthesis , Animals , Cell Survival/drug effects , Cell Survival/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Down-Regulation/genetics , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Endoribonucleases/genetics , Enzyme Inhibitors/pharmacology , Hepatocytes/pathology , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Palmitic Acid/pharmacology , Phosphorylation/drug effects , Phosphorylation/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Serine-Threonine Kinases/genetics , RNA Splicing/drug effects , RNA Splicing/genetics , Regulatory Factor X Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , X-Box Binding Protein 1
12.
Cell Tissue Res ; 364(2): 443-51, 2016 05.
Article in English | MEDLINE | ID: mdl-26662055

ABSTRACT

Tob1 is a member of the BTG/TOB family of proteins with established antiproliferative function. In Danio rerio and Xenopus laevis, the Tob1 gene is expressed from the one-cell stage through to early gastrula stages, followed in later development by discrete expression in many tissues including the notochord and somites. In both mouse and human, Tob1 is expressed in many adult tissues including the testis and ovary; however, the specific cell types are unknown. We examine Tob1 gene expression in mouse in developing germ cells and in sorted male germ cells (gonocytes, spermatogonia, pachytene spermatocytes and round spermatids) by reverse transcription and droplet digital polymerase chain reaction (RT-ddPCR) and in adult ovary and testis by immunofluorescence with anti-Tob1 protein staining. By RT-ddPCR, Tob1 expression was low in developing male germ cells but was highly expressed in round spermatids. In developing female germ cells undergoing entry into meiosis, it increased 10-fold. Tob1 was also highly expressed in round spermatids and in oocytes in all stages of folliculogenesis. Notably, a marker for P-bodies, Dcp-2, was also highly expressed in round spermatids and all oocyte stages examined. The cytoplasmic presence of Tob1 protein in round spermatids and oocytes and the association of Tob1 protein with Dcp2 in both cell types suggest that Tob1 protein plays a role in post-transcriptional mechanisms.


Subject(s)
Carrier Proteins/biosynthesis , Embryonic Germ Cells/metabolism , Endoribonucleases/biosynthesis , Gene Expression Regulation, Developmental , Oocytes/metabolism , Spermatids/metabolism , Spermatocytes/metabolism , Spermatogonia/metabolism , Animals , Biomarkers/metabolism , Female , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oogenesis/physiology , Ovary/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spermatogenesis/physiology , Testis/metabolism
13.
PLoS Pathog ; 9(8): e1003544, 2013.
Article in English | MEDLINE | ID: mdl-23950715

ABSTRACT

During viral infection, a massive demand for viral glycoproteins can overwhelm the capacity of the protein folding and quality control machinery, leading to an accumulation of unfolded proteins in the endoplasmic reticulum (ER). To restore ER homeostasis, cells initiate the unfolded protein response (UPR) by activating three ER-to-nucleus signaling pathways, of which the inositol-requiring enzyme 1 (IRE1)-dependent pathway is the most conserved. To reduce ER stress, the UPR decreases protein synthesis, increases degradation of unfolded proteins, and upregulates chaperone expression to enhance protein folding. Cytomegaloviruses, as other viral pathogens, modulate the UPR to their own advantage. However, the molecular mechanisms and the viral proteins responsible for UPR modulation remained to be identified. In this study, we investigated the modulation of IRE1 signaling by murine cytomegalovirus (MCMV) and found that IRE1-mediated mRNA splicing and expression of the X-box binding protein 1 (XBP1) is repressed in infected cells. By affinity purification, we identified the viral M50 protein as an IRE1-interacting protein. M50 expression in transfected or MCMV-infected cells induced a substantial downregulation of IRE1 protein levels. The N-terminal conserved region of M50 was found to be required for interaction with and downregulation of IRE1. Moreover, UL50, the human cytomegalovirus (HCMV) homolog of M50, affected IRE1 in the same way. Thus we concluded that IRE1 downregulation represents a previously undescribed viral strategy to curb the UPR.


Subject(s)
Cytomegalovirus Infections/metabolism , Cytomegalovirus/metabolism , Endoribonucleases/biosynthesis , Membrane Proteins/biosynthesis , Muromegalovirus/metabolism , Protein Serine-Threonine Kinases/biosynthesis , Unfolded Protein Response , Animals , Cell Line, Transformed , Cytomegalovirus/genetics , Cytomegalovirus Infections/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Down-Regulation/genetics , Endoribonucleases/genetics , Humans , Membrane Proteins/genetics , Mice , Muromegalovirus/genetics , NIH 3T3 Cells , Protein Serine-Threonine Kinases/genetics , Regulatory Factor X Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Viral Structural Proteins/genetics , Viral Structural Proteins/metabolism , X-Box Binding Protein 1
14.
Genet Mol Res ; 14(2): 5994-6002, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-26125799

ABSTRACT

We evaluated the effects of down-regulated heme oxygenase (HO)-1 expression on the proliferation of the acute myelocytic leukemia Kasumi-1 cell line by using the HO-1 inhibitor zinc protoporphyrin IX (ZnPPIX) in combination with daunorubicin (DNR), and evaluated the mechanism. The proliferation rates of cells treated with 10 mg/mL DNR and 10 mM ZnPPIX individually or in combination for different time periods were detected using the MTT assay. The apoptotic outcomes of the blank control, ZnPPIX, DNR, and ZnPPIX groups in combination with the DNR group were detected by flow cytometry. The expression of HO-1, activating transcription factor 4, CCAAT-enhancer-binding protein homologous protein, and inositol-requiring enzyme-α mRNA and proteins were detected by fluorescent quantitative real-time polymerase chain reaction and western blotting, respectively. Combined administration inhibited the cells most potently and time-dependently, decreased the expression of HO-1, and significantly increased the expression of activating transcription factor 4, CCAAT-enhancer-binding protein homologous protein, and inositol-requiring enzyme-α expression levels. The cell apoptotic rates in the blank control, DNR, ZnPPIX, and combined administration groups were 8.32 ± 0.53, 39.16 ± 1.46, 10.46 ± 0.88, and 56.26 ± 2.24%, respectively. Inhibiting HO-1 expression can enhance the damaging effects of DNR on Kasumi-1 cells, providing experimental evidence for the improvement of therapeutic effects on acute myelocytic leukemia in clinical practice.


Subject(s)
Activating Transcription Factor 4/biosynthesis , Endoribonucleases/biosynthesis , Heme Oxygenase-1/biosynthesis , Leukemia, Myeloid, Acute/genetics , Protein Serine-Threonine Kinases/biosynthesis , Transcription Factor CHOP/biosynthesis , Activating Transcription Factor 4/genetics , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Endoribonucleases/genetics , Enzyme Inhibitors/administration & dosage , Gene Expression Regulation, Leukemic/drug effects , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/genetics , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/biosynthesis , Signal Transduction/drug effects , Transcription Factor CHOP/genetics
15.
J Biol Chem ; 288(46): 33272-82, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24100031

ABSTRACT

The inositol-requiring enzyme 1α (IRE1α) is a serine-threonine kinase that plays crucial roles in activating the unfolded protein response. Studies suggest that IRE1α is activated during thymic T cell development and in effector CD8(+) T cells. However, its role in regulating T helper cell differentiation remains unknown. We find that IRE1α is up-regulated and activated upon CD4(+) T cell activation and plays an important role in promoting cytokine IL-4 production. CD4(+) T cells from IRE1α KO mice have reduced IL-4 protein expression, and this impaired IL-4 production is not due to the altered expression of Th2 lineage-specific transcription factors, such as GATA3. Instead, IL-4 mRNA stability is reduced in IRE1α KO T cells. Furthermore, treatment of T cells with an IRE1α-specific inhibitor, 4µ8C, leads to a block in IL-4, IL-5, and IL-13 production, confirming the role of IRE1α in the regulation of IL-4. This study identifies a regulatory function for IRE1α in the promotion of IL-4 in T cells.


Subject(s)
Cell Differentiation/immunology , Endoribonucleases/immunology , Interleukin-4/immunology , Protein Serine-Threonine Kinases/immunology , Th2 Cells/immunology , Up-Regulation/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Endoribonucleases/biosynthesis , Endoribonucleases/genetics , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/immunology , GATA3 Transcription Factor/metabolism , Interleukin-13/biosynthesis , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-4/biosynthesis , Interleukin-4/genetics , Interleukin-5/biosynthesis , Interleukin-5/genetics , Interleukin-5/immunology , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , RNA Stability/immunology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/immunology , Th2 Cells/cytology , Th2 Cells/metabolism , Up-Regulation/genetics
16.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 68(Pt 11): 1346-50, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23143246

ABSTRACT

PIWI-interacting RNAs (piRNAs) bind PIWI proteins and silence transposons to maintain the genomic integrity of germ cells. Zucchini (Zuc), a phospholipase D superfamily member, is conserved among animals and is implicated in piRNA biogenesis. However, the underlying mechanism by which Zuc participates in piRNA biogenesis remains elusive. Drosophila melanogaster Zuc (DmZuc) was expressed in Escherichia coli, purified and crystallized. X-ray diffraction data were collected to 1.75 Šresolution. The crystal belonged to space group P2(1), with unit-cell parameters a=55.0, b=71.2, c=56.3 Å, ß=107.9°.


Subject(s)
Drosophila Proteins/chemistry , Drosophila melanogaster/enzymology , Endoribonucleases/chemistry , Amino Acid Sequence , Animals , Chromatography, Affinity , Conserved Sequence , Crystallization , Crystallography, X-Ray , Drosophila Proteins/biosynthesis , Drosophila Proteins/isolation & purification , Endoribonucleases/biosynthesis , Endoribonucleases/isolation & purification , Escherichia coli , Molecular Sequence Data , Protein Sorting Signals , Protein Structure, Secondary , Sequence Alignment
17.
Biochem J ; 435(1): 103-11, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21208191

ABSTRACT

tRNase Z is the endonuclease that is involved in tRNA 3'-end maturation by removal of the 3'-trailer sequences from tRNA precursors. Most eukaryotes examined to date, including the budding yeast Saccharomyces cerevisiae and humans, have a single long form of tRNase Z (tRNase ZL). In contrast, the fission yeast Schizosaccharomyces pombe contains two candidate tRNase ZLs encoded by the essential genes sptrz1+ and sptrz2+. In the present study, we have expressed recombinant SpTrz1p and SpTrz2p in S. pombe. Both recombinant proteins possess precursor tRNA 3'-endonucleolytic activity in vitro. SpTrz1p localizes to the nucleus and has a simian virus 40 NLS (nuclear localization signal)-like NLS at its N-terminus, which contains four consecutive arginine and lysine residues between residues 208 and 211 that are critical for the NLS function. In contrast, SpTrz2p is a mitochondrial protein with an N-terminal MTS (mitochondrial-targeting signal). High-level overexpression of sptrz1+ has no detectable phenotypes. In contrast, strong overexpression of sptrz2+ is lethal in wild-type cells and results in morphological abnormalities, including swollen and round cells, demonstrating that the correct expression level of sptrz2+ is critical. The present study provides evidence for partitioning of tRNase Z function between two different proteins in S. pombe, although we cannot rule out specialized functions for each protein.


Subject(s)
Endoribonucleases/genetics , Endoribonucleases/metabolism , Genes, Fungal , RNA Precursors/metabolism , RNA, Transfer/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/metabolism , Cell Nucleus/enzymology , Cell Nucleus/metabolism , Endoribonucleases/biosynthesis , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Microbial Viability , Mitochondria/enzymology , Mitochondria/metabolism , Mutant Proteins/biosynthesis , Mutant Proteins/metabolism , Nuclear Localization Signals/metabolism , Protein Sorting Signals , Protein Transport , RNA Processing, Post-Transcriptional , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Saccharomyces cerevisiae Proteins/biosynthesis , Schizosaccharomyces/cytology , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/biosynthesis , Schizosaccharomyces pombe Proteins/genetics
18.
J Neurosci ; 30(11): 4048-61, 2010 Mar 17.
Article in English | MEDLINE | ID: mdl-20237275

ABSTRACT

MicroRNAs (miRNAs), small 19-25 nucleotide RNAs that influence gene expression through posttranscriptional regulation of mRNA translation and degradation, have recently emerged as important regulators of neural development. Using conditional knock-out of Dicer, an RNase III enzyme required for miRNA maturation, previous studies have demonstrated an essential role for miRNAs in mouse cortical, inner ear, and olfactory development. However, a previous study (Damiani et al., 2008) using a Chx10cre mouse to delete Dicer in retinal progenitors reported no defects in the retina before the second postnatal week, suggesting that miRNAs are not required for mouse retinal development. In an effort to further study the role of miRNAs during retinal development and resolve this apparent conflict, we conditionally knocked out Dicer using a different (alphaPax6cre) line of transgenic mice. In contrast to the previous study, we demonstrate an essential role for miRNAs during mouse retinal development. In the absence of Dicer in the embryonic retina, production of early generated cell types (ganglion and horizontal cells) is increased, and markers of late progenitors are not expressed. This phenotype persists into postnatal retina, in which we find the Dicer-deficient progenitors fail to generate late-born cell types such as rods and Müller glia but continue to generate ganglion cells. We further characterize the dynamic expression of miRNAs during retinal progenitor differentiation and provide a comprehensive profile of miRNAs expressed during retinal development. We conclude that Dicer is necessary for the developmental change in competence of the retinal progenitor cells.


Subject(s)
DEAD-box RNA Helicases/biosynthesis , Endoribonucleases/biosynthesis , Retina/embryology , Retina/metabolism , Stem Cells/metabolism , Animals , Animals, Newborn , Cell Differentiation/physiology , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/genetics , Endoribonucleases/deficiency , Endoribonucleases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , MicroRNAs/biosynthesis , Neurogenesis/physiology , Retina/growth & development , Ribonuclease III , Stem Cells/cytology
19.
J Neurosci ; 30(44): 14835-42, 2010 Nov 03.
Article in English | MEDLINE | ID: mdl-21048142

ABSTRACT

Dicer-dependent noncoding RNAs, including microRNAs (miRNAs), play an important role in a modulation of translation of mRNA transcripts necessary for differentiation in many cell types. In vivo experiments using cell type-specific Dicer1 gene inactivation in neurons showed its essential role for neuronal development and survival. However, little is known about the consequences of a loss of miRNAs in adult, fully differentiated neurons. To address this question, we used an inducible variant of the Cre recombinase (tamoxifen-inducible CreERT2) under control of Camk2a gene regulatory elements. After induction of Dicer1 gene deletion in adult mouse forebrain, we observed a progressive loss of a whole set of brain-specific miRNAs. Animals were tested in a battery of both aversively and appetitively motivated cognitive tasks, such as Morris water maze, IntelliCage system, or trace fear conditioning. Compatible with rather long half-life of miRNAs in hippocampal neurons, we observed an enhancement of memory strength of mutant mice 12 weeks after the Dicer1 gene mutation, before the onset of neurodegenerative process. In acute brain slices, immediately after high-frequency stimulation of the Schaffer collaterals, the efficacy at CA3-to-CA1 synapses was higher in mutant than in control mice, whereas long-term potentiation was comparable between genotypes. This phenotype was reflected at the subcellular and molecular level by the elongated filopodia-like shaped dendritic spines and an increased translation of synaptic plasticity-related proteins, such as BDNF and MMP-9 in mutant animals. The presented work shows miRNAs as key players in the learning and memory process of mammals.


Subject(s)
DEAD-box RNA Helicases/deficiency , Endoribonucleases/deficiency , Gene Deletion , Hippocampus/metabolism , Learning/physiology , Memory/physiology , MicroRNAs/genetics , Animals , DEAD-box RNA Helicases/biosynthesis , DEAD-box RNA Helicases/genetics , Dendritic Spines/metabolism , Dendritic Spines/ultrastructure , Electric Stimulation/methods , Endoribonucleases/biosynthesis , Endoribonucleases/genetics , Hippocampus/ultrastructure , Long-Term Potentiation/genetics , Mice , Mice, Knockout , Mice, Transgenic , MicroRNAs/antagonists & inhibitors , MicroRNAs/biosynthesis , Organ Culture Techniques , Ribonuclease III , Synapses/metabolism , Synapses/ultrastructure
20.
Proc Natl Acad Sci U S A ; 105(52): 20816-21, 2008 Dec 30.
Article in English | MEDLINE | ID: mdl-19075243

ABSTRACT

Type I IFNs were discovered as the primary antiviral cytokines and are now known to serve critical functions in host defense against bacterial pathogens. Accordingly, established mediators of IFN antiviral activity may mediate previously unrecognized antibacterial functions. RNase-L is the terminal component of an RNA decay pathway that is an important mediator of IFN-induced antiviral activity. Here, we identify a role for RNase-L in the host antibacterial response. RNase-L(-/-) mice exhibited a dramatic increase in mortality after challenge with Bacillus anthracis and Escherichia coli; this increased susceptibility was due to a compromised immune response resulting in increased bacterial load. Investigation of the mechanisms of RNase-L antibacterial activity indicated that RNase-L is required for the optimal induction of proinflammatory cytokines that play essential roles in host defense from bacterial pathogens. RNase-L also regulated the expression of the endolysosomal protease, cathepsin-E, and endosome-associated activities, that function to eliminate internalized bacteria and may contribute to RNase-L antimicrobial action. Our results reveal a unique role for RNase-L in the antibacterial response that is mediated through multiple mechanisms. As a regulator of fundamental components of the innate immune response, RNase-L represents a viable therapeutic target to augment host defense against diverse microbial pathogens.


Subject(s)
Anthrax/enzymology , Bacillus anthracis , Endoribonucleases/biosynthesis , Escherichia coli Infections/enzymology , Escherichia coli , Interferon Type I/biosynthesis , Animals , Anthrax/genetics , Anthrax/immunology , Bacillus anthracis/immunology , Cathepsin E/biosynthesis , Cathepsin E/genetics , Cathepsin E/immunology , Endoribonucleases/genetics , Endoribonucleases/immunology , Endosomes/enzymology , Endosomes/genetics , Endosomes/immunology , Escherichia coli/immunology , Escherichia coli Infections/genetics , Escherichia coli Infections/immunology , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Enzymologic/immunology , Interferon Type I/genetics , Interferon Type I/immunology , Mice , Mice, Knockout , RNA Stability/genetics , RNA Stability/immunology
SELECTION OF CITATIONS
SEARCH DETAIL