Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
J Antibiot (Tokyo) ; 77(2): 85-92, 2024 02.
Article in English | MEDLINE | ID: mdl-38008738

ABSTRACT

Hepatitis B virus (HBV) causes chronic hepatitis in humans, and current antiviral therapies rarely treat viral infections. To improve the treatment efficacy, novel therapeutic agents, especially those with different mechanisms of action, need to be developed for use in combination with the current antivirals. Here, we isolated new anti-HBV compounds, named catenulopyrizomicins A-C, from the fermentation broth of rare actinomycete Catenuloplanes sp. MM782L-181F7. Structural analysis revealed that these compounds contained a structure that is composed of thiazolyl pyridine moiety. The catenulopyrizomicins reduced the amount of intracellular viral DNA in HepG2.2.15 cells with EC50 values ranging from 1.94 to 2.63 µM with small but notable selectivity. Mechanistic studies indicated that catenulopyrizomicin promotes the release of immature virion particles that fail to be enveloped through alterations in membrane permeability.


Subject(s)
Actinobacteria , Humans , Actinobacteria/genetics , Virus Replication , Hepatitis B virus , Hep G2 Cells , Antiviral Agents/pharmacology , DNA, Viral/genetics , DNA, Viral/pharmacology
2.
Cancer Sci ; 114(12): 4558-4570, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37776195

ABSTRACT

Dual-specificity tyrosine-regulated kinase 2 (DYRK2) is a protein kinase that phosphorylates p53-Ser46 and induces apoptosis in response to DNA damage. However, the relationship between DYRK2 expression and chemosensitivity after DNA damage in colorectal cancer has not been well investigated. The aim of the present study was to examine whether DYRK2 could be a novel marker for predicting chemosensitivity after 5-fluorouracil- and oxaliplatin-induced DNA damage in colorectal cancer. Here we showed that DYRK2 knockout decreased the chemosensitivity to 5-fluorouracil and oxaliplatin in p53 wild-type colorectal cancer cells, whereas the chemosensitivity remained unchanged in p53-deficient/mutated colorectal cancer cells. In addition, no significant differences in chemosensitivity to 5-fluorouracil and oxaliplatin between scramble and siDYRK2 p53(-/-) colorectal cancer cells were observed. Conversely, the combination of adenovirus-mediated overexpression of DYRK2 with 5-fluorouracil or oxaliplatin enhanced apoptosis and chemosensitivity through p53-Ser46 phosphorylation in p53 wild-type colorectal cancer cells. Furthermore, DYRK2 knockout decreased chemosensitivity to 5-fluorouracil and oxaliplatin in p53 wild-type xenograft mouse models. Taken together, these findings demonstrated that DYRK2 expression was associated with chemosensitivity to 5-fluorouracil and oxaliplatin in p53 wild-type colorectal cancer, suggesting the importance of evaluating the p53 status and DYRK2 expression as a novel marker in therapeutic strategies for colorectal cancer.


Subject(s)
Colorectal Neoplasms , Tumor Suppressor Protein p53 , Humans , Animals , Mice , Oxaliplatin/pharmacology , Oxaliplatin/therapeutic use , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Apoptosis/genetics , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , DNA Damage
3.
PLoS Pathog ; 19(8): e1011591, 2023 08.
Article in English | MEDLINE | ID: mdl-37585449

ABSTRACT

Hepatitis C virus (HCV) is a pathogen characterized not only by its persistent infection leading to the development of cirrhosis and hepatocellular carcinoma (HCC), but also by metabolic disorders such as lipid and iron dysregulation. Elevated iron load is commonly observed in the livers of patients with chronic hepatitis C, and hepatic iron overload is a highly profibrogenic and carcinogenic factor that increases the risk of HCC. However, the underlying mechanisms of elevated iron accumulation in HCV-infected livers remain to be fully elucidated. Here, we observed iron accumulation in cells and liver tissues under HCV infection and in mice expressing viral proteins from recombinant adenoviruses. We established two molecular mechanisms that contribute to increased iron load in cells caused by HCV infection. One is the transcriptional induction of hepcidin, the key hormone for modulating iron homeostasis. The transcription factor cAMP-responsive element-binding protein hepatocyte specific (CREBH), which was activated by HCV infection, not only directly recognizes the hepcidin promoter but also induces bone morphogenetic protein 6 (BMP6) expression, resulting in an activated BMP-SMAD pathway that enhances hepcidin promoter activity. The other is post-translational regulation of the iron-exporting membrane protein ferroportin 1 (FPN1), which is cleaved between residues Cys284 and Ala285 in the intracytoplasmic loop region of the central portion mediated by HCV NS3-4A serine protease. We propose that host transcriptional activation triggered by endoplasmic reticulum stress and FPN1 cleavage by viral protease work in concert to impair iron efflux, leading to iron accumulation in HCV-infected cells.


Subject(s)
Carcinoma, Hepatocellular , Hepatitis C , Liver Neoplasms , Animals , Mice , Hepacivirus/physiology , Hepatitis C/metabolism , Hepcidins/genetics , Hepcidins/metabolism , Iron/metabolism , Transcriptional Activation , Up-Regulation
4.
Mol Brain ; 16(1): 62, 2023 07 26.
Article in English | MEDLINE | ID: mdl-37496071

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a disease that affects motor neurons and has a poor prognosis. We focused on TAR DNA-binding protein 43 kDa (TDP-43), which is a common component of neuronal inclusions in many ALS patients. To analyze the contribution of TDP-43 mutations to ALS in human cells, we first introduced TDP-43 mutations into healthy human iPSCs using CRISPR/Cas9 gene editing technology, induced the differentiation of these cells into motor and sensory neurons, and analyzed factors that are assumed to be altered in or associated with ALS (cell morphology, TDP-43 localization and aggregate formation, cell death, TDP-43 splicing function, etc.). We aimed to clarify the pathological alterations caused solely by TDP-43 mutation, i.e., the changes in human iPSC-derived neurons with TDP-43 mutation compared with those with the same genetic background except TDP-43 mutation. Oxidative stress induced by hydrogen peroxide administration caused the death of TDP-43 mutant-expressing motor neurons but not in sensory neurons, indicating the specific vulnerability of human iPSC-derived motor neurons with TDP-43 mutation to oxidative stress. In our model, we observed aggregate formation in a small fraction of TDP-43 mutant-expressing motor neurons, suggesting that aggregate formation seems to be related to ALS pathology but not the direct cause of cell death. This study provides basic knowledge for elucidating the pathogenesis of ALS and developing treatments for the disease.


Subject(s)
Amyotrophic Lateral Sclerosis , Induced Pluripotent Stem Cells , Humans , Amyotrophic Lateral Sclerosis/pathology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Induced Pluripotent Stem Cells/metabolism , Motor Neurons/pathology , Mutation/genetics , Oxidative Stress
5.
Cancer Sci ; 113(3): 960-970, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34932844

ABSTRACT

Colorectal cancer is one of the most common gastrointestinal tumors with good outcomes; however, with distant metastasis, the outcomes are poor. Novel treatment methods are urgently needed. Our in vitro studies indicate that dual-specificity tyrosine-regulated kinase 2 (DYRK2) functions as a tumor suppressor in colorectal cancer by regulating cell survival, proliferation, and apoptosis induction. In addition, DYRK2 expression is decreased in tumor tissues compared to nontumor tissues in colorectal cancer, indicating a correlation with clinical prognosis. In this context, we devised a novel therapeutic strategy to overexpress DYRK2 in tumors by adenovirus-mediated gene transfer. The present study shows that overexpression of DYRK2 in colon cancer cell lines by adenovirus inhibits cell proliferation and induces apoptosis in vitro. Furthermore, in mouse subcutaneous xenograft and liver metastasis models, enforced expression of DYRK2 by direct or intravenous injection of adenovirus to the tumor significantly inhibits tumor growth. Taken together, these findings show that adenovirus-based overexpression of DYRK2 could be a novel gene therapy for liver metastasis of colorectal cancer.


Subject(s)
Adenoviridae/genetics , Colorectal Neoplasms/therapy , Genetic Therapy/methods , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Genetic Vectors , Humans , Liver Neoplasms/genetics , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Mice , Tumor Suppressor Proteins/genetics , Xenograft Model Antitumor Assays , Dyrk Kinases
6.
Sci Rep ; 11(1): 24369, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34934130

ABSTRACT

Genome editing techniques such as CRISPR/Cas9 have both become common gene engineering technologies and have been applied to gene therapy. However, the problems of increasing the efficiency of genome editing and reducing off-target effects that induce double-stranded breaks at unexpected sites in the genome remain. In this study, we developed a novel Cas9 transduction system, Exci-Cas9, using an adenovirus vector (AdV). Cas9 was expressed on a circular molecule excised by the site-specific recombinase Cre and succeeded in shortening the expression period compared to AdV, which expresses the gene of interest for at least 6 months. As an example, we chose hepatitis B, which currently has more than 200 million carriers in the world and frequently progresses to liver cirrhosis or hepatocellular carcinoma. The efficiencies of hepatitis B virus genome disruption by Exci-Cas9 and Cas9 expression by AdV directly (Avec) were the same, about 80-90%. Furthermore, Exci-Cas9 enabled cell- or tissue-specific genome editing by expressing Cre from a cell- or tissue-specific promoter. We believe that Exci-Cas9 developed in this study is useful not only for resolving the persistent expression of Cas9, which has been a problem in genome editing, but also for eliminating long-term DNA viruses such as human papilloma virus.


Subject(s)
Adenoviridae/genetics , CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems , Gene Editing , Hepatitis B virus/genetics , Hepatocytes/metabolism , Integrases/metabolism , CRISPR-Associated Protein 9/genetics , Genetic Vectors , HEK293 Cells , HeLa Cells , Hep G2 Cells , Hepatitis B/chemically induced , Hepatitis B/genetics , Hepatocytes/virology , Humans , Integrases/genetics
7.
Virus Res ; 306: 198565, 2021 12.
Article in English | MEDLINE | ID: mdl-34555437

ABSTRACT

Hepatitis B virus (HBV) is the causative agent of chronic liver disease and is correlated with the development of subsequent hepatic cirrhosis and hepatocellular carcinoma. Current antiviral therapy using nucleos(t)ide analogs is effective in suppressing viral replication and interrupting disease progression, but HBV is rarely cured completely. Thus, there remains an unmet need for the development of novel anti-HBV drugs. Here, we report the identification of N-(4-Nitrophenyl)-1-phenylethanone hydrazone (ANPH) as a novel structural class of selective inhibitors targeting the replication of the HBV genome using adenovirus vector-mediated HBV genome transduction. ANPH inhibited viral genome replication in HepG2.2.15 cells by inducing the formation of empty capsids devoid of pregenomic RNA without affecting its transcription and translation. Biochemical assays using a truncated core protein consisting of the assembly domain showed that ANPH accelerates the formation of morphologically intact capsids. Taken together, we propose that ANPH might provide a new structural scaffold to design a new anti-HBV drug in medicinal chemistry as well as chemical probes for HBV core protein functions in the future.


Subject(s)
Hepatitis B , Liver Neoplasms , Acetophenones , Antiviral Agents/therapeutic use , Capsid/metabolism , Capsid Proteins/genetics , Capsid Proteins/metabolism , Hepatitis B virus , Humans , Virus Assembly , Virus Replication
8.
Cancer Sci ; 112(6): 2335-2348, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33931930

ABSTRACT

Lysosomal degradation plays a crucial role in the metabolism of biological macromolecules supplied by autophagy. The regulation of the autophagy-lysosome system, which contributes to intracellular homeostasis, chemoresistance, and tumor progression, has recently been revealed as a promising therapeutic approach for pancreatic cancer (PC). However, the details of lysosomal catabolic function in PC cells have not been fully elucidated. In this study, we show evidence that suppression of acid alpha-glucosidase (GAA), one of the lysosomal enzymes, improves chemosensitivity and exerts apoptotic effects on PC cells through the disturbance of expression of the transcription factor EB. The levels of lysosomal enzyme were elevated by gemcitabine in PC cells. In particular, the levels of GAA were responsive to gemcitabine in a dose-dependent and time-dependent manner. Small interfering RNA against the GAA gene (siGAA) suppressed cell proliferation and promoted apoptosis in gemcitabine-treated PC cells. In untreated PC cells, we observed accumulation of depolarized mitochondria. Gene therapy using adenoviral vectors carrying shRNA against the GAA gene increased the number of apoptotic cells and decreased the tumor growth in xenograft model mice. These results indicate that GAA is one of the key targets to improve the efficacy of gemcitabine and develop novel therapies for PC.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm/drug effects , Pancreatic Neoplasms/drug therapy , RNA, Small Interfering/administration & dosage , alpha-Glucosidases/genetics , Animals , Autophagy , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Lysosomes/drug effects , Lysosomes/enzymology , Male , Mice , Mice, Nude , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/genetics , RNA, Small Interfering/pharmacology , Time Factors , Up-Regulation , Xenograft Model Antitumor Assays , Gemcitabine
9.
RNA Biol ; 17(3): 403-416, 2020 03.
Article in English | MEDLINE | ID: mdl-31924127

ABSTRACT

Shortening of mRNA poly(A) tails (deadenylation) to trigger their decay is mediated mainly by the CCR4-NOT deadenylase complex. While four catalytic subunits (CNOT6, 6L 7, and 8) have been identified in the mammalian CCR4-NOT complex, their individual biological roles are not fully understood. In this study, we addressed the contribution of CNOT7/8 to viability of primary mouse embryonic fibroblasts (MEFs). We found that MEFs lacking CNOT7/8 expression [Cnot7/8-double knockout (dKO) MEFs] undergo cell death, whereas MEFs lacking CNOT6/6L expression (Cnot6/6l-dKO MEFs) remain viable. Co-immunoprecipitation analyses showed that CNOT6/6L are also absent from the CCR4-NOT complex in Cnot7/8-dKO MEFs. In contrast, either CNOT7 or CNOT8 still interacts with other subunits in the CCR4-NOT complex in Cnot6/6l-dKO MEFs. Exogenous expression of a CNOT7 mutant lacking catalytic activity in Cnot7/8-dKO MEFs cannot recover cell viability, even though CNOT6/6L exists to some extent in the CCR4-NOT complex, confirming that CNOT7/8 is essential for viability. Bulk poly(A) tail analysis revealed that mRNAs with longer poly(A) tails are more numerous in Cnot7/8-dKO MEFs than in Cnot6/6l-dKO MEFs. Consistent with elongated poly(A) tails, more mRNAs are upregulated and stabilized in Cnot7/8-dKO MEFs than in Cnot6/6l-dKO MEFs. Importantly, Cnot6/6l-dKO mice are viable and grow normally to adulthood. Taken together, the CNOT7/8 catalytic subunits are essential for deadenylation, which is necessary to maintain cell viability, whereas CNOT6/6L are not.


Subject(s)
Exoribonucleases/metabolism , RNA, Messenger/metabolism , Receptors, CCR4/metabolism , Repressor Proteins/metabolism , Animals , Cell Survival/genetics , Exoribonucleases/genetics , Female , Fibroblasts/cytology , Fibroblasts/physiology , Male , Mice, Knockout , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Poly A/genetics , Poly A/metabolism , Protein Subunits , RNA Stability , RNA, Messenger/genetics , Receptors, CCR4/genetics , Repressor Proteins/genetics
10.
Nat Commun ; 10(1): 1567, 2019 04 05.
Article in English | MEDLINE | ID: mdl-30952864

ABSTRACT

Selective autophagy ensures the removal of specific soluble proteins, protein aggregates, damaged mitochondria, and invasive bacteria from cells. Defective autophagy has been directly linked to metabolic disorders. However how selective autophagy regulates metabolism remains largely uncharacterized. Here we show that a deficiency in selective autophagy is associated with suppression of lipid oxidation. Hepatic loss of Atg7 or Atg5 significantly impairs the production of ketone bodies upon fasting, due to decreased expression of enzymes involved in ß-oxidation following suppression of transactivation by PPARα. Mechanistically, nuclear receptor co-repressor 1 (NCoR1), which interacts with PPARα to suppress its transactivation, binds to the autophagosomal GABARAP family proteins and is degraded by autophagy. Consequently, loss of autophagy causes accumulation of NCoR1, suppressing PPARα activity and resulting in impaired lipid oxidation. These results suggest that autophagy contributes to PPARα activation upon fasting by promoting degradation of NCoR1 and thus regulates ß-oxidation and ketone bodies production.


Subject(s)
Autophagy , Lipid Metabolism , Nuclear Receptor Co-Repressor 1/metabolism , Animals , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Autophagy-Related Protein 5/physiology , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Autophagy-Related Protein 7/physiology , Fasting , Ketone Bodies/metabolism , Liver/metabolism , Mice , Nuclear Receptor Co-Repressor 1/physiology , Oxidation-Reduction , PPAR alpha
11.
Cancer Lett ; 451: 100-109, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30851422

ABSTRACT

Liver cancer is highly aggressive and globally exhibits a poor prognosis. Therefore, the identification of novel molecules that can become targets for future therapies is urgently required. We have reported that dual-specificity tyrosine-regulated kinase 2 (DYRK2) functions as a tumor suppressor by regulating cell survival, differentiation, proliferation and apoptosis. However, the research into its clinical application as a molecular target has remained to be explored. Here we showed that DYRK2 knockdown enhanced tumor growth of liver cancer cells. Conversely and more importantly, adenovirus-mediated overexpression of DYRK2 resulted in inhibition of cell proliferation and tumor growth, and induction of apoptosis both in vitro and in vivo. Furthermore, we found that liver cancer patients with low DYRK2 expression had a significantly shorter overall survival. Given the findings that DYRK2 regulates proliferation and apoptosis of cancer cells, DYRK2 expression could be a promising predictive marker of the prognosis in liver cancer. Stabilized or forced expression of DYRK2 may become thus a potential target for novel gene therapy against liver cancer.


Subject(s)
Apoptosis , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Adenoviridae/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Heterografts , Humans , Mice , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Survival Analysis , Dyrk Kinases
12.
Leuk Res ; 74: 57-63, 2018 11.
Article in English | MEDLINE | ID: mdl-30300821

ABSTRACT

Novel epigenetic therapies targeting bromodomain and extra-terminal (BET) family proteins have shown therapeutic efficacy in diverse hematologic malignancies and solid cancers. However, the mechanism of resistance remains poorly understood. In the present study, we evaluated the mechanism of resistance to the BET inhibitor I-BET151 and its signaling pathway to overcome resistance in U937 cells. Treatment with 10 µM I-BET151 significantly induced growth inhibition, apoptosis, and cell cycle modulation, including increases in sub-G1 and G1 phases and decreases in S and G2/M phases, in U937 cells. However, no significant changes in these factors were detected in I-BET151-resistant U937 (U937R) cells. Combined treatment with I-BET151 and IKK inhibitor VII synergistically induced apoptosis in U937 and U937R cells. Increased expression of bromodomain-containing protein (BRD) 2, BRD4, and nuclear NF-κBp65 proteins was detected in U937R cells. IKK inhibitor VII inhibited the activation of NF-κBp65 protein in the nuclear fraction of U937R cells. These findings suggest that resistance to I-BET151 in U937R cells is related to constitutive activation of the NF-κB signaling pathway via increased expression of both BRD2 and BRD4. Targeting the NF-κB signaling pathway may be an effective therapeutic strategy to enhance or restore the sensitivity to I-BET151 in U937 cells.


Subject(s)
Cell Cycle/drug effects , Drug Resistance, Neoplasm/drug effects , Hematologic Neoplasms/metabolism , Heterocyclic Compounds, 4 or More Rings/pharmacology , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Transcription Factor RelA/metabolism , Transcription Factors/metabolism , Cell Cycle/genetics , Cell Cycle Proteins , Drug Resistance, Neoplasm/genetics , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Humans , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , Transcription Factor RelA/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , U937 Cells
13.
Sci Rep ; 8(1): 14215, 2018 09 21.
Article in English | MEDLINE | ID: mdl-30242188

ABSTRACT

Multiple-system atrophy (MSA) is a neurodegenerative disease characterized by autonomic failure with various combinations of parkinsonism, cerebellar ataxia, and pyramidal dysfunction. We previously reported that functionally impaired variants of COQ2, which encodes an essential enzyme in the biosynthetic pathway of coenzyme Q10, are associated with MSA. Here, we report functional deficiencies in mitochondrial respiration and the antioxidative system in induced pluripotent stem cell (iPSC)-derived neurons from an MSA patient with compound heterozygous COQ2 mutations. The functional deficiencies were rescued by site-specific CRISPR/Cas9-mediated gene corrections. We also report an increase in apoptosis of iPSC-derived neurons from MSA patients. Coenzyme Q10 reduced apoptosis of neurons from the MSA patient with compound heterozygous COQ2 mutations. Our results reveal that cellular dysfunctions attributable to decreased coenzyme Q10 levels are related to neuronal death in MSA, particularly in patients with COQ2 variants, and may contribute to the development of therapy using coenzyme Q10 supplementation.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Multiple System Atrophy/metabolism , Multiple System Atrophy/pathology , Neurons/metabolism , Ubiquinone/analogs & derivatives , Adult , Alkyl and Aryl Transferases/metabolism , Amino Acid Sequence , Atrophy/metabolism , Atrophy/pathology , Base Sequence , Female , Humans , Induced Pluripotent Stem Cells/pathology , Male , Middle Aged , Mitochondria/metabolism , Mutation/genetics , Neurons/pathology , Ubiquinone/metabolism
14.
Hepatology ; 66(5): 1430-1443, 2017 11.
Article in English | MEDLINE | ID: mdl-28621467

ABSTRACT

Mechanisms of hepatic fibrogenesis induced by hepatitis C virus (HCV), one of the leading causes of liver fibrosis, are not fully understood. We studied transcriptional up-regulation of transforming growth factor ß (TGF-ß), especially TGF-ß2, which is mediated by activation of liver-enriched transcription factor cAMP-responsive element-binding protein, hepatocyte specific (CREBH) triggered by HCV infection and its functional significance for induction of profibrogenic phenotypes by interaction of HCV-infected cells with hepatic stellate cells (HSCs). Compared to TGF-ß1, expression of TGF-ß2 mRNA was induced faster and to a higher level upon HCV infection. Serum TGF-ß2 levels in hepatitis C patients were higher compared to those in healthy individuals and were positively correlated with hepatic fibrosis stages F0-F2. TGF-ß2 promoter activity was decreased and increased, respectively, by silencing and overexpression of CREBH. CREBH recognition sites were identified in the TGF-ß2 promoter. CREBH binding to the promoter and its increase in cells expressing HCV Core-NS2 were shown by gel mobility shift and chromatin immunoprecipitation, respectively. The active form of CREBH was detectable in HCV-infected chimeric mice with human livers and cells expressing HCV proteins. Involvement of CREBH in HCV-induced fibrogenic response was further demonstrated in the CREBH null-mutant mouse model. Fibrogenic phenotypes were assessed using co-cultures of HCV-infected cells and HSCs. Expressions of fibrogenic factors and TGF-ß1 increasing in the co-cultures was prevented by TGF-ß2- or CREBH silencing. CONCLUSION: CREBH was identified as a key positive regulator of TGF-ß2 transcription in HCV-infected cells. TGF-ß2 released from infected cells potentially contributes to cross-induction of TGF-ß in an autocrine manner through its own signaling pathway, leading to an increase in fibrogenic responses in adjacent HSCs. (Hepatology 2017;66:1430-1443).


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Hepatitis C/metabolism , Liver Cirrhosis/virology , Liver/pathology , Transforming Growth Factor beta2/metabolism , Animals , Autocrine Communication , Fibrosis , Gene Expression Regulation , Hepatic Stellate Cells/pathology , Hepatitis C/complications , Hepatitis C/pathology , Liver Cirrhosis/metabolism , Male , Mice, Inbred C57BL , Paracrine Communication , Transforming Growth Factor beta1/metabolism
15.
Microbiol Immunol ; 61(7): 264-271, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28543309

ABSTRACT

Pulmonary emphysema impairs quality of life and increases mortality. It has previously been shown that administration of adenovirus vector expressing murine keratinocyte growth factor (KGF) before elastase instillation prevents pulmonary emphysema in mice. We therefore hypothesized that therapeutic administration of KGF would restore damage to lungs caused by elastase instillation and thus improve pulmonary function in an animal model. KGF expressing adenovirus vector, which prevented bleomycin-induced pulmonary fibrosis in a previous study, was constructed. Adenovirus vector (1.0 × 109 plaque-forming units) was administered intratracheally one week after administration of elastase into mouse lungs. One week after administration of KGF-vector, exercise tolerance testing and blood gas analysis were performed, after which the lungs were removed under deep anesthesia. KGF-positive pneumocytes were more numerous, surfactant protein secretion in the airspace greater and mean linear intercept of lungs shorter in animals that had received KGF than in control animals. Unexpectedly, however, arterial blood oxygenation was worse in the KGF group and maximum running speed, an indicator of exercise capacity, had not improved after KGF in mice with elastase-induced emphysema, indicating that KGF-expressing adenovirus vector impaired pulmonary function in these mice. Notably, vector lacking KGF-expression unit did not induce such impairment, implying that the KGF expression unit itself may cause the damage to alveolar cells. Possible involvement of the CAG promoter used for KGF expression in impairing pulmonary function is discussed.


Subject(s)
Adenoviridae/genetics , Emphysema/therapy , Fibroblast Growth Factor 7/biosynthesis , Fibroblast Growth Factor 7/genetics , Adenoviridae/metabolism , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/pathology , Animals , Bleomycin/pharmacology , DNA, Viral/genetics , Disease Models, Animal , Emphysema/chemically induced , Emphysema/physiopathology , Fibroblast Growth Factor 7/administration & dosage , Genetic Therapy , Genetic Vectors/genetics , Genetic Vectors/metabolism , Male , Mice , Mice, Inbred BALB C , Pancreatic Elastase , Promoter Regions, Genetic , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/virology , Pulmonary Surfactant-Associated Protein D/metabolism
16.
Sci Rep ; 7: 41851, 2017 02 03.
Article in English | MEDLINE | ID: mdl-28157182

ABSTRACT

The complicated replication mechanisms of hepatitis B virus (HBV) have impeded HBV studies and anti-HBV therapy development as well. Herein we report efficient genome replication of HBV applying adenovirus vectors (AdVs) showing high transduction efficiency. Even in primary hepatocytes derived from humanized mice the transduction efficiencies using AdVs were 450-fold higher compared than those using plasmids. By using an expression unit consisting of the CMV promoter, 1.03-copy HBV genome and foreign poly(A) signal, we successfully generated an improved AdV (HBV103-AdV) that efficiently provided 58 times more pregenomic RNA than previously reported AdVs. The HBV103-AdV-mediated HBV replication was easily and precisely detected using quantitative real-time PCR in primary hepatocytes as well as in HepG2 cells. Notably, when the AdV containing replication-defective HBV genome of 1.14 copy was transduced, we observed that HBV DNA-containing circular molecules (pseudo-ccc DNA) were produced, which were probably generated through homologous recombination. However, the replication-defective HBV103-AdV hardly yielded the pseudo-ccc, probably because the repeated sequences are vey short. Additionally, the efficacies of entecavir and lamivudine were quantitatively evaluated using this system at only 4 days postinfection with HBV103-AdVs. Therefore, this system offers high production of HBV genome replication and thus could become used widely.


Subject(s)
Hepatitis B virus/metabolism , Transfection/methods , Virus Replication , Adenoviridae/genetics , Cytomegalovirus/genetics , Genetic Vectors/genetics , Genome, Viral , HEK293 Cells , Hep G2 Cells , Hepatitis B virus/physiology , Humans , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism
17.
Exp Anim ; 65(3): 231-44, 2016 Jul 29.
Article in English | MEDLINE | ID: mdl-26923756

ABSTRACT

Temporal genetic modification of mice using the ligand-inducible Cre/loxP system is an important technique that allows the bypass of embryonic lethal phenotypes and access to adult phenotypes. In this study, we generated a tamoxifen-inducible Cre-driver mouse strain for the purpose of widespread and temporal Cre recombination. The new line, named CM32, expresses the GFPneo-fusion gene in a wide variety of tissues before FLP recombination and tamoxifen-inducible Cre after FLP recombination. Using FLP-recombined CM32 mice (CM32Δ mice) and Cre reporter mouse lines, we evaluated the efficiency of Cre recombination with and without tamoxifen administration to adult mice, and found tamoxifen-dependent induction of Cre recombination in a variety of adult tissues. In addition, we demonstrated that conditional activation of an oncogene could be achieved in adults using CM32Δ mice. CM32Δ;T26 mice, which harbored a Cre recombination-driven, SV40 large T antigen-expressing transgene, were viable and fertile. No overt phenotype was found in the mice up to 3 months after birth. Although they displayed pineoblastomas (pinealoblastomas) and/or thymic enlargement due to background Cre recombination by 6 months after birth, they developed epidermal hyperplasia when administered tamoxifen. Collectively, our results suggest that the CM32Δ transgenic mouse line can be applied to the assessment of adult phenotypes in mice with loxP-flanked transgenes.


Subject(s)
Genetic Techniques , Integrases/genetics , Mice, Transgenic/genetics , Recombination, Genetic , Tamoxifen , Transgenes , Animals , Antigens, Polyomavirus Transforming/genetics , Gene Fusion , Mice, Inbred C57BL , Mice, Inbred ICR , Oncogenes/genetics , Phenotype , Transcriptional Activation
18.
J Gen Virol ; 97(4): 887-892, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26861864

ABSTRACT

Hepatitis C virus (HCV) core plays a key role in viral particle formation and is involved in viral pathogenesis. Here, constructs for single-domain intrabodies consisting of variable regions derived from mouse mAbs against HCV core were established. Expressed single-domain intrabodies were shown to bind to HCV core, and inhibit the growth of cell culture-produced HCV derived from JFH-1 (genotype 2a) and a TH (genotype 1b)/JFH-1 chimera. Adenovirus vectors expressing intrabodies were also capable of reducing HCV propagation. Intrabody expression did not affect viral entry or genome replication of single-round infectious trans-complemented HCV particles. However, intrabody expression reduced intracellular and extracellular infectious titres in CD81-defective Huh7-25 cells transfected with the HCV genome, suggesting that these intrabodies impair HCV assembly. Furthermore, intrabody expression suppressed HCV core-induced NFκB promoter activity. These intrabodies may therefore serve as tools for elucidating the role of core in HCV pathogenesis.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Hepacivirus/genetics , Hepatocytes/immunology , Single-Domain Antibodies/immunology , Viral Core Proteins/genetics , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Viral/biosynthesis , Cell Line, Tumor , Epitope Mapping , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation , Genotype , HEK293 Cells , Hepacivirus/immunology , Hepatocytes/virology , Host-Pathogen Interactions , Humans , Hybridomas/immunology , Immunization , Mice , NF-kappa B/genetics , NF-kappa B/immunology , Plasmids/chemistry , Plasmids/immunology , Promoter Regions, Genetic , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Single-Domain Antibodies/biosynthesis , Transfection , Viral Core Proteins/immunology , Virus Assembly/genetics
19.
Sci Rep ; 5: 14779, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26437789

ABSTRACT

The CCR4-NOT complex is conserved in eukaryotes and is involved in mRNA metabolism, though its molecular physiological roles remain to be established. We show here that CNOT3-depleted mouse embryonic fibroblasts (MEFs) undergo cell death. Levels of other complex subunits are decreased in CNOT3-depleted MEFs. The death phenotype is rescued by introduction of wild-type (WT), but not mutated CNOT3, and is not suppressed by the pan-caspase inhibitor, zVAD-fluoromethylketone. Gene expression profiling reveals that mRNAs encoding cell death-related proteins, including receptor-interacting protein kinase 1 (RIPK1) and RIPK3, are stabilized in CNOT3-depleted MEFs. Some of these mRNAs bind to CNOT3, and in the absence of CNOT3 their poly(A) tails are elongated. Inhibition of RIPK1-RIPK3 signaling by a short-hairpin RNA or a necroptosis inhibitor, necrostatin-1, confers viability upon CNOT3-depleted MEFs. Therefore, we conclude that CNOT3 targets specific mRNAs to prevent cells from being disposed to necroptotic death.


Subject(s)
Fibroblasts/metabolism , RNA, Messenger/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Transcription Factors/genetics , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Caspase Inhibitors/pharmacology , Cell Death/drug effects , Cell Survival/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Embryo, Mammalian , Fibroblasts/cytology , Fibroblasts/drug effects , Gene Expression Regulation , Imidazoles/pharmacology , Indoles/pharmacology , Leupeptins/pharmacology , Mice , Primary Cell Culture , Protein Binding , RNA Stability , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Transcription Factors/metabolism
20.
PLoS One ; 9(9): e108627, 2014.
Article in English | MEDLINE | ID: mdl-25275311

ABSTRACT

Non-coding small RNAs are involved in many physiological responses including viral life cycles. Adenovirus-encoding small RNAs, known as virus-associated RNAs (VA RNAs), are transcribed throughout the replication process in the host cells, and their transcript levels depend on the copy numbers of the viral genome. Therefore, VA RNAs are abundant in infected cells after genome replication, i.e. during the late phase of viral infection. Their function during the late phase is the inhibition of interferon-inducible protein kinase R (PKR) activity to prevent antiviral responses; recently, mivaRNAs, the microRNAs processed from VA RNAs, have been reported to inhibit cellular gene expression. Although VA RNA transcription starts during the early phase, little is known about its function. The reason may be because much smaller amount of VA RNAs are transcribed during the early phase than the late phase. In this study, we applied replication-deficient adenovirus vectors (AdVs) and novel AdVs lacking VA RNA genes to analyze the expression changes in cellular genes mediated by VA RNAs using microarray analysis. AdVs are suitable to examine the function of VA RNAs during the early phase, since they constitutively express VA RNAs but do not replicate except in 293 cells. We found that the expression level of hepatoma-derived growth factor (HDGF) significantly decreased in response to the VA RNAs under replication-deficient condition, and this suppression was also observed during the early phase under replication-competent conditions. The suppression was independent of mivaRNA-induced downregulation, suggesting that the function of VA RNAs during the early phase differs from that during the late phase. Notably, overexpression of HDGF inhibited AdV growth. This is the first report to show the function, in part, of VA RNAs during the early phase that may be contribute to efficient viral growth.


Subject(s)
Adenoviridae/physiology , Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins/genetics , RNA, Viral/metabolism , Virus Replication , Adenoviridae/growth & development , Cell Line, Tumor , Down-Regulation/genetics , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Poly(A)-Binding Proteins/genetics , Poly(A)-Binding Proteins/metabolism , T-Cell Intracellular Antigen-1 , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL