Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 182
Filter
1.
Neural Plast ; 2021: 4090441, 2021.
Article in English | MEDLINE | ID: mdl-34471408

ABSTRACT

Our previous study showed that growth arrest- and DNA damage-inducible gene 153 (GAD153/CHOP) plays an important role in intermittent hypoxia- (IH-) induced apoptosis and impaired synaptic plasticity. This study is aimed at determining which signaling pathway is activated to induce CHOP and the role of this protein in mitochondria-dependent apoptosis induced by IH. In the in vivo study, mice were placed in IH chambers for 8 h daily over a period of 2 weeks; the IH chambers had oxygen (O2) concentrations that oscillated between 10% and 21%, cycling every 90 s. In the in vitro study, PC12 cells were exposed to 21% O2 (normoxia) or 8 IH cycles (25 min at 21% O2 and 35 min at 0.1% O2 for each cycle). After 2 weeks of IH treatment, we observed that the expression levels of phosphorylated protein kinase-like endoplasmic reticulum kinase (p-PERK), activating transcription factor 4 (ATF-4) and phosphorylated eukaryotic initiation factor 2 alpha (p-elf2α), were increased, but the levels of activating transcription factor 6 (ATF-6) and inositol-requiring enzyme 1 (IRE-1) were not increased. GSK2606414, a specific chemical inhibitor of the PERK pathway, reduced the expression of p-PERK, ATF-4, p-elf2α, and CHOP and rescued ER structure. In addition, Bax and Bak accumulated in the mitochondria after IH treatment, which induced cytochrome c release and initiated apoptosis. These effects were prevented by GSK2606414 and CHOP shRNA. Finally, the impaired long-term potentiation and long-term spatial memory in the IH group were rescued by GSK2606414. Together, the data from the in vitro and in vivo experiments indicate that IH-induced apoptosis and impaired synaptic plasticity were mediated by the PERK-ATF-4-CHOP pathway. Suppressing PERK-ATF-4-CHOP signaling pathway attenuated mitochondria-dependent apoptosis by reducing the expression of Bax and Bak in mitochondria, which may serve as novel adjunct therapeutic strategy for ameliorating obstructive sleep apnea- (OSA-) induced neurocognitive impairment.


Subject(s)
Cognitive Dysfunction/metabolism , Hypoxia/metabolism , Neurons/metabolism , Transcription Factor CHOP/biosynthesis , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Adenine/therapeutic use , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cognitive Dysfunction/drug therapy , Hypoxia/drug therapy , Indoles/pharmacology , Indoles/therapeutic use , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , PC12 Cells , Rats , Transcription Factor CHOP/antagonists & inhibitors
2.
Sci Rep ; 11(1): 11893, 2021 06 04.
Article in English | MEDLINE | ID: mdl-34088951

ABSTRACT

The imipramine ONC201 has antiproliferative effects in several cancer cell types and activates integrated stress response pathway associated with the induction of Damage Inducible Transcript 3 (DDIT3, also known as C/EBP homologous protein or CHOP). We investigated the signaling pathways through which ONC201/CHOP crosstalk is regulated in ONC201-treated nonmetastatic and metastatic cancer cell lines (Dukes' type B colorectal adenocarcinoma nonmetastatic SW480 and metastatic LS-174T cells, respectively). Cell proliferation and apoptosis were evaluated by MTT assays and flow cytometry, gene expression was assessed by Affymetrix microarray, signaling pathway perturbations were assessed in silico, and key regulatory proteins were validated by Western blotting. Unlike LS-174T cells, SW480 cells were resistant to ONC201 treatment; Gene Ontology analysis of differentially expressed genes showed that cellular responsiveness to ONC201 treatment also differed substantially. In both ONC201-treated cell lines, CHOP expression was upregulated; however, its upstream regulatory mechanisms were perturbed. Although, PERK, ATF6 and IRE1 ER-stress pathways upregulated CHOP in both cell types, the Bak/Bax pathway regulated CHOP only LS-174T cells. Additionally, CHOP RNA splicing profiles varied between cell lines; these were further modified by ONC201 treatment. In conclusion, we delineated the signaling mechanisms by which CHOP expression is regulated in ONC201-treated non-metastatic and metastatic colorectal cell lines. The observed differences could be related to cellular plasticity and metabolic reprogramming, nevertheless, detailed mechanistic studies are required for further validations.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Imidazoles/pharmacology , Oligonucleotide Array Sequence Analysis , Pyridines/pharmacology , Pyrimidines/pharmacology , Transcription Factor CHOP/biosynthesis , Alternative Splicing , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/genetics , Computational Biology , Humans , Neoplasm Metastasis , Polymerase Chain Reaction , RNA, Messenger/metabolism , Signal Transduction , Tetrazolium Salts , Thiazoles , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Tumor Microenvironment , Up-Regulation
3.
Mod Pathol ; 34(7): 1367-1372, 2021 07.
Article in English | MEDLINE | ID: mdl-33731886

ABSTRACT

Myxoid liposarcoma (MLPS) is a malignant adipocytic neoplasm with predilection for the extremities. MLPS is genetically defined by a t(12;16) translocation leading to FUS-DDIT3 (95%) or more rarely t(12;22) leading to EWSR1-DDIT3. Low-grade MLPS is characterized by bland spindle cells within a myxoid matrix containing delicate "chicken-wire" vasculature, whereas high-grade ("round cell") MLPS may be indistinguishable from other round cell sarcomas. In many cases, cytogenetic or molecular genetic techniques are applied to confirm the diagnosis. A recent study documented the utility of DDIT3 immunohistochemistry (IHC) in the differential diagnosis of adipocytic and myxoid soft tissue tumors. The purpose of this study was to evaluate DDIT3 IHC as a surrogate for molecular testing in high-grade MLPS. IHC was performed using a mouse monoclonal antibody directed against the N-terminus of DDIT3 on whole tissue sections from 50 high-grade MLPS cases and 319 histologic mimics used as controls (170 on whole tissue sections and 149 on a tissue microarray). Histologic mimics included Ewing sarcoma, CIC-rearranged sarcoma, sarcomas with BCOR genetic alterations, poorly differentiated synovial sarcoma, alveolar and embryonal rhabdomyosarcomas, mesenchymal chondrosarcoma, desmoplastic small round cell tumor, and neuroblastoma. Nuclear staining in >5% of cells was considered positive. By IHC, 48 (96%) high-grade MLPS showed strong diffuse nuclear staining for DDIT3. Of the controls, 2% of cases were positive, with no more than 25% nuclear staining. An additional 19% of control cases displayed less than 5% nuclear staining. Overall, DDIT3 IHC showed 96% sensitivity and 98% specificity for high-grade MLPS; strong, diffuse staining is also 96% sensitive but is 100% specific. IHC using an antibody directed against the N-terminus of DDIT3 is highly sensitive and specific for high-grade MLPS among histologic mimics and could replace molecular genetic testing in many cases, although limited labeling may be seen in a range of other tumor types.


Subject(s)
Biomarkers, Tumor/analysis , Immunohistochemistry/methods , Liposarcoma, Myxoid/diagnosis , Sarcoma/diagnosis , Transcription Factor CHOP/analysis , Cell Nucleus/metabolism , Diagnosis, Differential , Humans , Retrospective Studies , Sensitivity and Specificity , Soft Tissue Neoplasms/diagnosis , Transcription Factor CHOP/biosynthesis
4.
J Chem Neuroanat ; 114: 101942, 2021 07.
Article in English | MEDLINE | ID: mdl-33675952

ABSTRACT

Methamphetamine (Meth) is a neuro-stimulator substrate which might lead to neural cell death and the activation of several interconnected cellular pathways as well. However, the precise molecular mechanisms underlying Meth-induced neural cell death remained unclear yet. The current study aimed to assess the specific relationship between long-term Meth exposure and several endoplasmic reticulum stress, autophagy, and apoptosis associated markers including C/EBP homologous protein (CHOP), Tribbles homolog 3(Trib3), Nuclear protein 1(NUPR1), and Beclin-1 expression in postmortem human striatum. Therefore, the effects of long-term Meth exposure on autophagy and apoptosis in the striatum of postmortem users were evaluated and molecular, immunehistochemical, and histological examinations were performed on 10 control and 10 Meth-addicted brains. The level of CHOP, Trib3, NUPR1, and Beclin-1, Microtubule-associated proteins 1A/1B light chain 3B(LC3), Caspase 3, and Autophagy protein 5 (ATG5) were measured by using qPCR and immunohistochemistry. Stereological neural cell counting, Hematoxylin and Eosin, Nissl and Tunel staining were also performed. Based on our findings, the expression level of CHOP, Trib3, NUPR1, and Beclin-1 in the striatum of Meth group were significantly higher than the control group. Besides, the neuronal cell death was substantially increased in the striatum based on data obtained from the Tunel assay and the stereological analysis. Long-term presence of Meth in the brain can induce ER stress and overexpression of NUPR1 which is associated with the upregulation of CHOP, a pro-apoptotic transcription factor. Moreover, an increase in Trib3 expression is implicated in CHOP-dependent autophagic cell death during Meth-induced ER stress accompanied by an increase in neuronal cell death in the striatum of the postmortem human brains. Beclin 1 expression was also upregulated which may due to the activation of autophagic mechanisms upon prolonged Meth exposure.


Subject(s)
Amphetamine-Related Disorders/complications , Autophagosomes/drug effects , Basic Helix-Loop-Helix Transcription Factors/drug effects , Corpus Striatum/drug effects , Neoplasm Proteins/drug effects , Transcription Factor CHOP/drug effects , Adult , Apoptosis/drug effects , Autopsy , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Central Nervous System Stimulants/adverse effects , Humans , Male , Methamphetamine/adverse effects , Neoplasm Proteins/biosynthesis , Transcription Factor CHOP/biosynthesis
5.
Am J Surg Pathol ; 45(2): 230-239, 2021 02 01.
Article in English | MEDLINE | ID: mdl-32815829

ABSTRACT

Myxoid liposarcoma is a malignant adipogenic neoplasm characterized by prominent arborizing capillaries, occasional lipoblasts, and primitive-appearing spindle cells in a myxoid background. A recurrent translocation in myxoid liposarcoma results in an oncoprotein consisting of full-length DDIT3 (CHOP) fused to an N-terminal segment of either FUS (TLS) or, less often, EWSR1. Here, we explore the diagnostic significance of DDIT3 expression in myxoid liposarcoma using a mouse monoclonal antibody recognizing an epitope in the N-terminal region. Studying a total of 300 tumors, we find diffuse, moderate-to-strong nuclear-localized anti-DDIT3 immunoreactivity in all 46 cases of myxoid liposarcoma representing 36 unique tumors, including 6 cases with high-grade (round cell) morphology. DDIT3 immunohistochemistry also highlighted a distinctive vasculocentric growth pattern in 7 myxoid liposarcomas treated with neoadjuvant radiation. In contrast, the vast majority of other examined lipomatous and myxoid neoplasms exhibited no DDIT3 expression; limited, weak immunoreactivity in <10% of cells was infrequently observed in dedifferentiated liposarcoma (6/39, 15%), solitary fibrous tumor (3/12, 25%), pleomorphic liposarcoma (1/15, 7%), and high-grade myxofibrosarcoma (2/17, 12%). Although this minimal DDIT3 expression did not correlate with DDIT3 amplification or myxoid liposarcoma-like morphology in dedifferentiated liposarcoma, there was evidence among sarcomas (excluding myxoid liposarcoma) of a relationship between expression and exposure to neoadjuvant radiation or cytotoxic chemotherapy. The constellation of findings indicates that DDIT3 immunohistochemistry may have utility in the evaluation of myxoid and lipomatous neoplasms to support the diagnosis of myxoid liposarcoma.


Subject(s)
Biomarkers, Tumor/analysis , Liposarcoma, Myxoid/diagnosis , Soft Tissue Neoplasms/diagnosis , Transcription Factor CHOP/analysis , Humans , Immunohistochemistry , Transcription Factor CHOP/biosynthesis
6.
Life Sci ; 254: 117778, 2020 Aug 01.
Article in English | MEDLINE | ID: mdl-32407850

ABSTRACT

Long non-coding RNA (LncRNA) involved in types of physiological insults and diseases via regulating the responses of complex molecular, including cerebral ischemia-reperfusion (I/R) injury. LncRNA SNHG16 played a potential role in ketamine-induced neurotoxicity. In this study, we utilized an in vitro cell model of I/R to examine the specific function and mechanism of LncRNA SNHG16 in oxygen-glucose deprivation and reperfusion (OGD/R) induced SH-SY5Y cells. After in vitro treatment of OGD/R, the lower the SH-SY5Y cell survival, the higher cell the apoptosis and increased caspase-3 activity was observed. Also, OGD/R induced endoplasmic reticulum stress (ERS) through increasing GRP78 and CHOP expressions and down-regulated LncRNA SNHG16 in SH-SY5Y cells. Conversely, LncRNA SNHG16 overexpression promoted OGD/R induced SH-SY5Y cell survival, suppressed its apoptosis, and caspase-3 activity. GRP78 and CHOP expressions were significantly suppressed in LncRNA SNHG16 overexpressing cells. MiR-106b-5p expression was increased and LIMK1 expression was down-regulated in OGD/R induced SH-SY5Y cells, and these effects were reversed by LncRNA SNHG16 overexpression, respectively. Moreover, LIMK1 is a direct target of MiR-106b-5p, and knockdown of LIMK1 reversed the effects of LncRNA SNHG16 on OGD/R-induced SH-SY5Y cells biology. Altogether, these results confirmed an important neuroprotection role of LncRNA SNHG16 in OGD/R induced SH-SY5Y cells injury, and miR-106b-5p/LIMK1 signal axis was involved in the action of LncRNA SNHG16.


Subject(s)
Cell Survival/physiology , Lim Kinases/physiology , MicroRNAs/physiology , RNA, Long Noncoding/biosynthesis , RNA, Long Noncoding/physiology , Reperfusion Injury/metabolism , Apoptosis/physiology , Caspase 3/metabolism , Cells, Cultured , Down-Regulation , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Gene Knockdown Techniques , Heat-Shock Proteins/biosynthesis , Humans , Lim Kinases/genetics , Lim Kinases/metabolism , MicroRNAs/metabolism , Signal Transduction/physiology , Transcription Factor CHOP/biosynthesis
7.
Biomed Res Int ; 2020: 9029868, 2020.
Article in English | MEDLINE | ID: mdl-33457422

ABSTRACT

Endoplasmic reticulum stress (ERS) is strongly associated with acute kidney injury (AKI) to chronic kidney disease (CKD) transition. Huaier extract (HE) protects against kidney injury; albeit, the underlying mechanism is unknown. We hypothesized that HE reduces kidney injury by inhibiting ERS. In this study, using an AKI-CKD mouse model of ischemia-reperfusion injury (IRI), we evaluated the effect of HE on AKI-CKD transition. We also explored the underlying molecular mechanisms in this animal model and in the HK-2 human kidney cell line. The results showed that HE treatment improved the renal function, demonstrated by a significant decrease in serum creatinine levels after IRI. HE appreciably reduced the degree of kidney injury and fibrosis and restored the expression of the microRNA miR-1271 after IRI. Furthermore, HE reduced the expression of ERS markers glucose-regulated protein 78 (GRP78) and C/EBP homologous protein (CHOP) and inhibited apoptosis in the IRI group. This in vivo effect was supported by in vitro results in which HE inhibited apoptosis and decreased the expression of CHOP and GRP78 induced by ERS. We demonstrated that CHOP is a target of miR-1271. In conclusion, HE reduces kidney injury, probably by inhibiting apoptosis and decreasing the expression of GRP78 and CHOP via miR-1271 upregulation.


Subject(s)
Acute Kidney Injury/drug therapy , Apoptosis , Complex Mixtures/pharmacology , Endoplasmic Reticulum Stress , Gene Expression Regulation , Kidney Failure, Chronic/drug therapy , MicroRNAs/genetics , Up-Regulation , Animals , Cell Line , Disease Progression , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/biosynthesis , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/biosynthesis , Trametes , Transcription Factor CHOP/biosynthesis
8.
Invest Ophthalmol Vis Sci ; 60(6): 1953-1966, 2019 05 01.
Article in English | MEDLINE | ID: mdl-31060051

ABSTRACT

Purpose: Increased endoplasmic reticulum (ER) stress is one of the earliest subcellular changes in neuro-ophthalmic diseases. In this study, we investigated the expression of key molecules in the ER stress pathways following nonarteritic anterior ischemic optic neuropathy (AION), the most common acute optic neuropathy in adults over 50, and assessed the impact of chemical chaperon 4-phenylbutyric acid (4-PBA) in vivo. Methods: We induced AION using photochemical thrombosis in adult mice and performed histologic analyses of key molecules in the ER stress pathway in the retina and optic nerve. We also assessed the effects of daily intraperitoneal injections of 4-PBA after AION. Results: In the retina at baseline, there was low proapoptotic transcriptional regulator C/EBP homologous protein (CHOP) and high prosurvival chaperon glucose-regulated protein 78 (GRP78) expression in retinal ganglion cells (RGCs). One day after AION, there was significantly increased CHOP and reduced GRP78 expressions in the ganglion cell layer. In the optic nerve at baseline, there was little CHOP and high GRP78 expression. One day after AION, there was significantly increased CHOP and no change in GRP78 expression. Treatment immediately after AION using daily intraperitoneal injection of chemical chaperone 4-PBA for 19 days significantly rescued Brn3A+ RGCs and Olig2+ optic nerve oligodendrocytes. Conclusions: We showed for the first time that acute AION resulted in increased ER stress and differential expression of ER stress markers CHOP and GRP78 in the retina and optic nerve. Rescue of RGCs and oligodendrocytes with 4-PBA provides support for ER stress reduction as possible treatment for AION.


Subject(s)
Endoplasmic Reticulum Stress , Oligodendroglia/pathology , Optic Disk/pathology , Optic Neuropathy, Ischemic/pathology , Phenylbutyrates/pharmacology , Retinal Ganglion Cells/pathology , Animals , Disease Models, Animal , Endoplasmic Reticulum Chaperone BiP , Female , Gene Expression Regulation , Heat-Shock Proteins/biosynthesis , Heat-Shock Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Molecular Chaperones , Oligodendroglia/metabolism , Optic Disk/metabolism , Optic Neuropathy, Ischemic/drug therapy , Optic Neuropathy, Ischemic/genetics , Retinal Ganglion Cells/metabolism , Transcription Factor CHOP/biosynthesis , Transcription Factor CHOP/genetics
9.
Mol Oncol ; 13(5): 1150-1165, 2019 05.
Article in English | MEDLINE | ID: mdl-30854784

ABSTRACT

FAM175B is a reported regulator of p53 and suppresses tumorigenesis in numerous types of cancer, but very little is known about its function in esophageal squamous cell carcinomas (ESCCs), almost 70% of which exhibit mutations in p53. Here, we report that FAM175B expression is downregulated in high-grade intraepithelial neoplasia (t = 2.44, P = 0.031) and ESCC (t = 5.664, P < 0.001) tissues relative to that in adjacent normal esophageal tissues. Exogenous expression of FAM175B in ESCC cells resulted in a decrease in proliferation rate, inhibition of colony formation, and an increase in apoptosis rate. Knockdown of FAM175B produced the opposite results. Furthermore, confocal microscopy and coimmunoprecipitation assay showed that Activating transcription factor 4 (ATF4) colocalized and interacted with FAM175B. Ubiquitination assays revealed that FAM175B inhibited ubiquitin-dependent ATF4 degradation and elevated ATF4 protein level. Finally, luciferase reporter experiments further clarified that FAM175B promoted CHOP expression in an ATF4-dependent manner. Accordingly, the proapoptotic activity of FAM175B was significantly rescued by treatment with si-ATF4 and the CHOP inhibitor 4-PBA. In summary, FAM175B inhibited ATF4 ubiquitination and promoted ESCC cell apoptosis in a p53-independent manner. FAM175B expression loss may be an early diagnostic biomarker in ESCC patients.


Subject(s)
Activating Transcription Factor 4/metabolism , Apoptosis , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/metabolism , Gene Expression Regulation, Neoplastic , Nuclear Matrix-Associated Proteins/biosynthesis , Ubiquitin-Specific Proteases/biosynthesis , Ubiquitination , Activating Transcription Factor 4/genetics , Cell Line, Tumor , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology , Humans , Nuclear Matrix-Associated Proteins/genetics , Transcription Factor CHOP/biosynthesis , Transcription Factor CHOP/genetics , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Ubiquitin-Specific Proteases/genetics
10.
Cell Signal ; 59: 163-170, 2019 07.
Article in English | MEDLINE | ID: mdl-30826455

ABSTRACT

Regulator of G protein signaling 2 (RGS2) is upregulated by multiple forms of stress and can augment translational attenuation associated with the phosphorylation of the initiation factor eIF2, a hallmark of several stress-induced coping mechanisms. Under stress-induced translational inhibition, key factors, such as ATF4, are selectively expressed via alternative translation mechanisms. These factors are known to regulate molecular switches that control cell fate by regulating pro-survival and pro-apoptotic signals. The molecular mechanisms that balance these opposing responses to stresses are unclear. The present results suggest that RGS2 may be an important regulatory component in the cellular stress response through its translational control abilities. Previously, we have shown that RGS2 can interact with the translation initiation factor, eIF2B, and inhibit de novo protein synthesis. Here, we demonstrate that the expression of either full length RGS2 or its eIF2B-interacting domain (RGS2eb) significantly increases levels of ATF4 and CHOP, both of which are linked to stress-induced apoptosis. Furthermore, we show that these effects are translationally regulated and independent of eIF2 phosphorylation. The present results thus point to a novel function of RGS2 in the stress response directly related to its ability to reduce global protein synthesis.


Subject(s)
Activating Transcription Factor 4/biosynthesis , Peptide Chain Initiation, Translational , RGS Proteins/physiology , Stress, Physiological/physiology , Transcription Factor CHOP/biosynthesis , Activating Transcription Factor 4/genetics , Animals , Apoptosis , Eukaryotic Initiation Factor-2B/chemistry , Mice , NIH 3T3 Cells , Protein Domains , RGS Proteins/genetics , Transcription Factor CHOP/genetics
11.
Ann Biomed Eng ; 47(4): 1106-1115, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30671754

ABSTRACT

miR-214 has been recently found to be significantly downregulated in calcified human aortic valves (AVs). ER stress, especially the ATF4-mediated pathway, has also been shown to be significantly upregulated in calcific AV disease. Since elevated cyclic stretch is one of the major mechanical stimuli for AV calcification and ATF4 is a validated target of miR-214, we investigated the effect of cyclic stretch on miR-214 expression as well as those of ATF4 and two downstream genes (CHOP and BCL2L1). Porcine aortic valve (PAV) leaflets were cyclically stretched at 15% for 48 h in regular medium and for 1 week in osteogenic medium to simulate the early remodeling and late calcification stages of stretch-induced AV disease, respectively. For both stages, 10% cyclic stretch served as the physiological counterpart. RT-qPCR revealed that miR-214 expression was significantly downregulated during the late calcification stage, whereas the mRNA expression of ATF4 and BCL2L1 was upregulated and downregulated, respectively, during both early remodeling and late calcification stages. When PAV leaflets were statically transfected with miR-214 mimic in osteogenic medium for 2 weeks, calcification was significantly reduced compared to the control mimic case. This implies that miR-214 may have a protective role in stretch-induced calcific AV disease.


Subject(s)
Aortic Valve Stenosis/metabolism , Aortic Valve/metabolism , Aortic Valve/pathology , Calcinosis/metabolism , Gene Expression Regulation , MicroRNAs/metabolism , Up-Regulation , Activating Transcription Factor 4/biosynthesis , Animals , Aortic Valve Stenosis/pathology , Calcinosis/pathology , Swine , Transcription Factor CHOP/biosynthesis , bcl-X Protein/biosynthesis
12.
Biomed Pharmacother ; 111: 468-475, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30594786

ABSTRACT

Kaempferol is a flavonoid compound that has many functions, such as anti-inflammation and antioxidation. Acute liver failure (ALF) is a life-threatening illness accompanied by serious inflammation and extensive hepatocyte apoptosis. The aim of this study was to examine the therapeutic potential of kaempferol and its mechanism in ALF. In a murine ALF model induced by d-galactosamine (d-GalN, 700 mg/kg) / lipopolysaccharide (LPS, 10 µg/kg), mice were pretreated with kaempferol at 2 h before d-GalN/LPS administration and then sacrificed 6 h after d-GalN/LPS injection. Lethality, liver damage, endoplasmic reticulum(ER) stress, hepatocyte viability and apoptosis were evaluated. Whether pretreatment of kaempferol protected hepatocytes from ER stress-induced apoptosis was detected in vitro. Pretreatment of kaempferol decreased lethality, prolonged the survival time and significantly protected against liver injury, which was indicated by decreased transaminase levels and the well-preserved liver structure. The protective effect of kaempferol on the ALF mouse model was achieved by inhibiting hepatocyte apoptosis. Moreover, pretreatment of kaempferol increased the expression of glucose-regulated/binding immunoglobulin protein 78 (Grp78), decreased the expression of C/EBP-homologous protein (CHOP), and protected hepatocytes from ER stress-induced apoptosis in vitro. Our results showed that pretreatment of Grp78 siRNA partially negated the hepatic protection from kaempferol and reversed the inhibition of CHOP protein expression in d-GalN/LPS-induced ALF mice. In conclusion, kaempferol inhibits hepatocyte apoptosis to protect mice from liver failure by regulating the ER stress-Grp78-CHOP signaling pathway. Therefore, kaempferol may be used to treat ALF.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Galactosamine/toxicity , Heat-Shock Proteins/biosynthesis , Kaempferols/therapeutic use , Liver Failure, Acute/metabolism , Transcription Factor CHOP/biosynthesis , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cells, Cultured , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/physiology , Heat-Shock Proteins/agonists , Hepatocytes/drug effects , Hepatocytes/metabolism , Kaempferols/pharmacology , Lipopolysaccharides/toxicity , Liver Failure, Acute/chemically induced , Liver Failure, Acute/prevention & control , Male , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Signal Transduction/physiology , Transcription Factor CHOP/antagonists & inhibitors
13.
Pathol Res Pract ; 214(8): 1105-1109, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29910063

ABSTRACT

BACKGROUND: Few studies have reported the clinical and prognostic significance of C/EBP homologous protein (CHOP) in advanced gastric cancer (GC). Therefore, the present study investigated the expression of CHOP in advanced GC patients to determine its potential prognostic role. METHODS: The levels of CHOP in 95 patients with advanced GC and adjacent non-cancerous tissues were evaluated by qRT-PCR, western blot and immunohistochemistry. Furthermore, the association of CHOP expression with clinicopathological parameters and prognosis of advanced GC patients was analyzed. RESULTS: The levels of CHOP were down-regulated in advanced GC compared with non-cancerous tissues (P<0.01). In addition, high CHOP expression more frequently occurred in advanced GC tissues with depth of invasion of T1-2 (P < 0.01), lower clinical stage (TNM Ⅰ-Ⅱ stage) (P<0.05) and without lymph node metastasis (P<0.05). No significant difference was observed between the expression of CHOP and age, gender, tumor size, lesion site and differentiation (P>0.05). The Kaplan-Meier survival analyses showed that the overall survival rate of advanced GC patients with positive CHOP expression was significantly higher than that of patients with negative CHOP expression (P<0.01). Univariate and multivariate Cox proportional hazards models revealed that low CHOP expression (OR = 0.314, 95%CI: 0.176~0.794, P = 0.003) was an independent factor for poor overall survival in advanced GC patients. CONCLUSION: Low expression of CHOP predicts the poor prognosis of advanced GC patients, and CHOP may be a prognostic biomarker for patients with advanced GC.


Subject(s)
Biomarkers, Tumor/analysis , Stomach Neoplasms/pathology , Transcription Factor CHOP/biosynthesis , Adult , Aged , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Proportional Hazards Models , Stomach Neoplasms/mortality
14.
Eur Rev Med Pharmacol Sci ; 22(7): 2160-2168, 2018 04.
Article in English | MEDLINE | ID: mdl-29687876

ABSTRACT

OBJECTIVE: GRP78 and CHOP play essential roles in endoplasmic reticulum stress (ERS) of the vascular smooth muscle cells. We aim to investigate the effect of enalapril maleate and folic acid tablet on the expressions of GRP78 and CHOP and vascular remodeling in a homocysteine (HCY)-treated hypertensive rat model. MATERIALS AND METHODS: The hypertensive rat model was established with the technique of coarctation in the abdominal aorta, and the blood pressure of the rat was measured with the non-destructive tail-cuff method two weeks after operation. Thirty-six rats with hypertension were randomly divided into 3 groups (n=12 in each group). The control group received common diet and double distilled water, methionine group received 30 g/L methionine diet and double distilled water, while enalapril maleate and folic acid tablet group received 30 g/L methionine diet and 0.2 mg.kg-1.d-1 solution of enalapril maleate and folic acid tablet. Samples were collected at week 4 and week 8 for analysis. The plasma homocysteine was measured by homocysteine detector; MAP was detected through carotid artery incubation and aortic media thickness was determined by image analyses software. The expression of GRP78 and CHOP in the vascular smooth muscle cells were identified by immunohistochemistry and Western blot. RESULTS: Compared with the control group, the concentration of HCY in the serum of rats in methionine group was increased significantly after 4 weeks (p < 0.01), and even more significant after 8 weeks (p < 0.01). Compared with that of methionine group, the level of HCY in enalapril maleate and folic acid tablet group rats was significantly decreased (p < 0.01). The level of MAP in methionine group was increased significantly after 8 weeks compared with that of control group (p < 0.05). However, the MAP in enalapril maleate and folic acid tablet group was decreased significantly compared with that of methionine group. Compared with control group, the media thickness of vascular smooth muscle of rats in the methionine group was increased significantly (p < 0.05) while was statistically reduced in the enalapril maleate and folic acid tablet group (p < 0.05). The expressions of GRP78 and CHOP in methionine group were significantly elevated compared to that of control in a time dependent manner (p < 0.05), which were remarkably down regulated in enalapril maleate and folic acid tablet group compared with that in methionine group. CONCLUSIONS: The administration of enalapril maleate and folic acid tablet can maintain the normal state of cells via the alleviation of ERS and vascular damages, reduction of HCY and the thickness of arterial media as well as the improvement of vascular remodeling.


Subject(s)
Enalapril/administration & dosage , Folic Acid/administration & dosage , Heat-Shock Proteins/biosynthesis , Homocysteine/blood , Hypertension/blood , Transcription Factor CHOP/biosynthesis , Animals , Gene Expression , Heat-Shock Proteins/genetics , Homocysteine/antagonists & inhibitors , Hypertension/drug therapy , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Rats , Rats, Sprague-Dawley , Transcription Factor CHOP/genetics , Vascular Remodeling/drug effects , Vascular Remodeling/physiology
15.
Pharmazie ; 73(3): 156-160, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29544563

ABSTRACT

Previous studies have shown sorafenib to function as a multitargeted tyrosine kinase inhibitor in different tumors. However, whether sorafenib improves renal cell carcinoma (RCC) through activating transcription factor 4 (ATF4) has never been explored. In the current study, we showed that sorafenib could suppress RCC cell viability in a time- and dose-dependent manner. Furthermore, sorafenib is demonstrated to enhance the mRNA and protein levels of ATF4. Meanwhile, overexpression of ATF4 was demonstrated to induce ACHN cell cycle arrest and cell apoptosis. Moreover, treatment with sorafenib could enhance the expression of CCAAT/enhancer-binding protein-homologous protein (CHOP) and p53 upregulated modulator of apoptosis (PUMA), thereby leading to ACHN cell apoptosis. More importantly, silencing of ATF4 could largely abolish sorafenib-induced upregulation of CHOP and PUMA in ACHN cells. Meanwhile, sorafenib-induced cell apoptosis may be dependent on the activation of ATF4 since knockdown of ATF4 partially reversed sorafenib-induced ACHN cell apoptosis. In summary, the present study demonstrates that sorafenib activates ATF4-CHOP-PUMA pathway in RCC cells, resulting in enhanced ER stress-related cell apoptosis.


Subject(s)
Activating Transcription Factor 4/drug effects , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Sorafenib/pharmacology , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Gene Knockdown Techniques , Humans , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/drug effects , Reactive Oxygen Species/metabolism , Transcription Factor CHOP/biosynthesis , Transcription Factor CHOP/drug effects , Transfection , Up-Regulation/drug effects
16.
J Biol Chem ; 293(13): 4735-4751, 2018 03 30.
Article in English | MEDLINE | ID: mdl-29378845

ABSTRACT

Estrogen receptor α (ERα) action plays an important role in pancreatic ß-cell function and survival; thus, it is considered a potential therapeutic target for the treatment of type 2 diabetes in women. However, the mechanisms underlying the protective effects of ERα remain unclear. Because ERα regulates mitochondrial metabolism in other cell types, we hypothesized that ERα may act to preserve insulin secretion and promote ß-cell survival by regulating mitochondrial-endoplasmic reticulum (EndoRetic) function. We tested this hypothesis using pancreatic islet-specific ERα knockout (PERαKO) mice and Min6 ß-cells in culture with Esr1 knockdown (KD). We found that Esr1-KD promoted reactive oxygen species production that associated with reduced fission/fusion dynamics and impaired mitophagy. Electron microscopy showed mitochondrial enlargement and a pro-fusion phenotype. Mitochondrial cristae and endoplasmic reticulum were dilated in Esr1-KD compared with ERα replete Min6 ß-cells. Increased expression of Oma1 and Chop was paralleled by increased oxygen consumption and apoptosis susceptibility in ERα-KD cells. In contrast, ERα overexpression and ligand activation reduced both Chop and Oma1 expression, likely by ERα binding to consensus estrogen-response element sites in the Oma1 and Chop promoters. Together, our findings suggest that ERα promotes ß-cell survival and insulin secretion through maintenance of mitochondrial fission/fusion-mitophagy dynamics and EndoRetic function, in part by Oma1 and Chop repression.


Subject(s)
Apoptosis , Endoplasmic Reticulum Stress , Estrogen Receptor alpha/metabolism , Insulin-Secreting Cells/metabolism , Mitochondria/metabolism , Mitophagy , Animals , Cell Survival , Estrogen Receptor alpha/genetics , Female , Insulin/genetics , Insulin/metabolism , Metalloproteases/biosynthesis , Metalloproteases/genetics , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/genetics , Reactive Oxygen Species/metabolism , Transcription Factor CHOP/biosynthesis , Transcription Factor CHOP/genetics
17.
Biotechnol Lett ; 40(1): 205-213, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28986701

ABSTRACT

OBJECTIVE: To evaluate the effects of mechano growth factor E peptide (MGF) on the invasive properties of melanoma cells. RESULTS: Melanoma cells (GLL19) were treated with 10, 20 and 30 ng MGF/ml for 24 h. Their invasive properties were investigated by transwell assay. Cytoskeleton reorganization was assessed via staining with phalloidin-FITC; lysyl oxidase (LOX) family gene expression was tested by qRT-PCR, and western blotting was used to detect expression of the matrix metalloproteinases (MMPs) and endoplasmic reticulum (ER) stress. MGF decreased the invasive capabilities of melanoma cells and induced changes in cytoskeleton distribution. MGF also down-regulated the expression of MMPs and up-regulated the expression of the cell apoptosis-related protein CHOP by inducing ER stress. CONCLUSIONS: MGF can decrease the invasive properties of melanoma cells and induce ER stress, promoting cell apoptosis. Thus, MGF represents a novel strategy for the potential treatment of patients presenting with cutaneous melanoma.


Subject(s)
Biological Factors/metabolism , Cell Movement , Endoplasmic Reticulum Stress/drug effects , Insulin-Like Growth Factor I/metabolism , Melanocytes/drug effects , Transcription Factor CHOP/biosynthesis , Cell Line, Tumor , Cell Proliferation , Cytoskeleton/metabolism , Humans , Melanoma
18.
Biochem J ; 474(24): 4065-4074, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29089376

ABSTRACT

The aim of the present study was to investigate the effects and molecular mechanisms of GPR4 (G-protein-coupled receptor 4) in cell apoptosis and renal ischemia-reperfusion (IR) injury in vivo and in vitro GPR4-/- mice and wild-type (WT) mice underwent renal IR or sham procedures. For hypoxia/reoxygenation (HR), human umbilical vein endothelial cells (HUVECs) were subjected to 4 h of hypoxia, followed by 6 h of reoxygenation. Renal histological changes were observed by periodic acid-Schiff staining and myeloperoxidase activity. Apoptosis was detected by TUNEL staining. GPR4, C/EBP-homologous protein (CHOP) and cleaved caspase-3 protein expressions were detected by western blot. Both GPR4 and CHOP were up-regulated after renal IR in mice. GPR4-knockout mice had significantly less renal damage and decreased TUNEL-positive cells than WT controls after IR. Bone marrow chimeras demonstrated that it was due to the GPR4 inactivation in renal parenchymal cells. Moreover, GPR4 was mainly expressed in endothelial cells after renal IR. GPR4 knockdown markedly inhibited CHOP expression and cell apoptosis in the HUVECs after HR treatment. GPR4 blockade attenuated renal injury after IR and reduced the cell apoptosis through the suppression of CHOP expression.


Subject(s)
Apoptosis , Kidney Diseases/metabolism , Kidney/metabolism , Receptors, G-Protein-Coupled/deficiency , Reperfusion Injury/metabolism , Transcription Factor CHOP/biosynthesis , Animals , Caspase 3/biosynthesis , Caspase 3/genetics , Gene Expression Regulation , Gene Knockout Techniques , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Kidney/pathology , Kidney Diseases/genetics , Kidney Diseases/pathology , Male , Mice , Mice, Knockout , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Transcription Factor CHOP/genetics
19.
Biosci Rep ; 37(4)2017 Aug 31.
Article in English | MEDLINE | ID: mdl-28667101

ABSTRACT

C/EBP-homologous protein (CHOP) is a ubiquitously expressed stress-inducible transcription factor robustly induced by maladaptive endoplasmic reticulum (ER) stresses in a wide variety of cells. Here, we examined a novel function of Sigma 1 receptor (Sigmar1) in regulating CHOP expression under ER stress in cardiomyocytes. We also defined Sigmar1-dependent activation of the adaptive ER-stress pathway in regulating CHOP expression. We used adenovirus-mediated Sigmar1 overexpression as well as Sigmar1 knockdown by siRNA in neonatal rat ventricular cardiomyocytes (NRCs); to induce ER stress, cardiomyocytes were treated with tunicamycin. Sigmar1-siRNA knockdown significantly increased the expression of CHOP and significantly induced cellular toxicity by sustained activation of ER stress in cardiomyocytes. Sigmar1 overexpression decreased the expression of CHOP and significantly decreased cellular toxicity in cells. Using biochemical and immunocytochemical experiments, we also defined the specific ER-stress pathway associated with Sigmar1-dependent regulation of CHOP expression and cellular toxicity. We found that Sigmar1 overexpression significantly increased inositol requiring kinase 1α (IRE1α) phosphorylation and increased spliced X-box-binding proteins (XBP1s) expression as well as nuclear localization. In contrast, Sigmar1 knockdown significantly decreased IRE1α phosphorylation and decreased XBP1s expression as well as nuclear transport. Taken together, these results indicate that Sigmar1-dependent activation of IRE1α-XBP1s ER-stress response pathways are associated with inhibition of CHOP expression and suppression of cellular toxicity. Hence, Sigmar1 is an essential component of the adaptive ER-stress response pathways eliciting cellular protection in cardiomyocytes.


Subject(s)
Endoplasmic Reticulum Stress , Gene Expression Regulation , Myocytes, Cardiac/metabolism , Receptors, sigma/metabolism , Transcription Factor CHOP/biosynthesis , Animals , Endoribonucleases/genetics , Endoribonucleases/metabolism , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Myocytes, Cardiac/cytology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rats, Sprague-Dawley , Receptors, sigma/genetics , Transcription Factor CHOP/genetics , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism , Sigma-1 Receptor
20.
PLoS Genet ; 13(7): e1006849, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28678786

ABSTRACT

The unfolded protein response (UPR) in the endoplasmic reticulum (ER) and the cytoplasmic heat stress response are two major stress response systems necessary for maintaining proteostasis for cellular health. Failure of either of these systems, such as in sustained UPR activation or in insufficient heat shock response activation, can lead to the development of neurodegeneration. Alleviation of ER stress and enhancement of heat shock response through heat shock factor 1 (HSF1) activation have previously been considered as attractive potential therapeutic targets for Alzheimer's disease (AD)-a prevalent and devastating tauopathy. Understanding the interplay of the two aforementioned systems and their cooperative role in AD remain elusive. Here we report studies in human brain and tau pathogenic mouse models (rTg4510, PS19, and rTg21221), identifying HSF1 degradation and UPR activation as precursors of aberrant tau pathogenesis. We demonstrate that chemical ER stress inducers caused autophagy-lysosomal HSF1 degradation, resulting in tau hyperphosphorylation in rat primary neurons. In addition, permanent HSF1 loss reversely causes chronic UPR activation, leading to aberrant tau phosphorylation and aggregation in the hippocampus of aged HSF1 heterozygous knock-out mice. The deleterious interplay of UPR activation and HSF1 loss is exacerbated in N2a cells stably overexpressing a pro-aggregation mutant TauRD ΔK280 (N2a-TauRD ΔK280). We provide evidence of how these two stress response systems are intrinsically interweaved by showing that the gene encoding C/EBP-homologous protein (CHOP) activation in the UPR apoptotic pathway facilitates HSF1 degradation, which likely further contributes to prolonged UPR via ER chaperone HSP70 a5 (BiP/GRP78) suppression. Upregulating HSF1 relieves the tau toxicity in N2a-TauRD ΔK280 by reducing CHOP and increasing HSP70 a5 (BiP/GRP78). Our work reveals how the bidirectional crosstalk between the two stress response systems promotes early tau pathology and identifies HSF1 being one likely key player in both systems.


Subject(s)
Alzheimer Disease/genetics , DNA-Binding Proteins/genetics , Heat-Shock Proteins/genetics , Transcription Factor CHOP/genetics , Transcription Factors/genetics , Unfolded Protein Response/genetics , tau Proteins/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Autophagy/genetics , DNA-Binding Proteins/biosynthesis , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation , Heat Shock Transcription Factors , Heat-Shock Response/genetics , Hippocampus/metabolism , Hippocampus/pathology , Humans , Mice , Mice, Knockout , Neurons/metabolism , Neurons/pathology , Phosphorylation , Protein Aggregation, Pathological/genetics , Proteolysis , Rats , Tauopathies/genetics , Tauopathies/metabolism , Tauopathies/pathology , Transcription Factor CHOP/biosynthesis , Transcription Factors/biosynthesis , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...