Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
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
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.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
EMBO Rep ; 15(5): 557-65, 2014 May.
Article in English | MEDLINE | ID: mdl-24668264

ABSTRACT

Several autophagy proteins contain an LC3-interacting region (LIR) responsible for their interaction with Atg8 homolog proteins. Here, we show that ALFY binds selectively to LC3C and the GABARAPs through a LIR in its WD40 domain. Binding of ALFY to GABARAP is indispensable for its recruitment to LC3B-positive structures and, thus, for the clearance of certain p62 structures by autophagy. In addition, the crystal structure of the GABARAP-ALFY-LIR peptide complex identifies three conserved residues in the GABARAPs that are responsible for binding to ALFY. Interestingly, introduction of these residues in LC3B is sufficient to enable its interaction with ALFY, indicating that residues outside the LIR-binding hydrophobic pockets confer specificity to the interactions with Atg8 homolog proteins.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autophagy , Membrane Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Apoptosis Regulatory Proteins , Autophagy-Related Protein 8 Family , Autophagy-Related Proteins , Cell Line, Tumor , HeLa Cells , Humans , Hydrophobic and Hydrophilic Interactions , Membrane Proteins/ultrastructure , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/ultrastructure , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Transcription Factors/ultrastructure
10.
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
11.
Nucleic Acids Res ; 39(2): e7, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21051352

ABSTRACT

Tissue-/cancer-specific promoters for use in adenovirus vectors (AdVs) are valuable for elucidating specific gene functions and for use in gene therapy. However, low activity, non-specific expression and size limitations in the vector are always problems. Here, we developed a 'double-unit' AdV containing the Cre gene under the control of an α-fetoprotein promoter near the right end of its genome and bearing a compact 'excisional-expression' unit consisting of a target cDNA 'upstream' of a potent promoter between two loxPs near the left end of its genome. When Cre was expressed, the expression unit was excised as a circular molecule and strongly expressed. Undesired leak expression of Cre during virus preparation was completely suppressed by a dominant-negative Cre and a short-hairpin RNA against Cre. Using this novel construct, a very strict specificity was maintained while achieving a 40- to 90-fold higher expression level, compared with that attainable using a direct specific promoter. Therefore, the 'double-unit' AdV enabled us to produce a tissue-/cancer-specific promoter in an AdV with a high expression level and strict specificity.


Subject(s)
Adenoviridae/genetics , Genetic Vectors , Promoter Regions, Genetic , Cell Line , Cell Line, Tumor , Escherichia coli/genetics , Humans , Integrases/genetics , Integrases/metabolism , Mutation , RNA, Small Interfering/metabolism , alpha-Fetoproteins/genetics
12.
Uirusu ; 63(2): 155-64, 2013.
Article in Japanese | MEDLINE | ID: mdl-25366050

ABSTRACT

First-generation adenovirus vectors (FG-AdVs) are widely used because transduction efficiency of the vectors is very high. However, severe immune responses especially to the liver have been a serious problem of this vector. We succeeded to identify a viral protein that cause the immune responses and reported ''low-inflammatory AdVs'' that mostly solve this problem. However, to develop the ultimate form of this vector, it is necessary to remove virus-associated RNA (VA RNA) genes from the AdV vector genome. VA RNAs are transcribed by polymerase III; they are not essential for viral growth but have important roles to make appropriate circumstances for this virus. Large amount of VA RNAs are required in the late phase to support viral growth. Hence it is difficult to establish 293 cell lines that can support replication of AdVs lacking VA RNA genes (VA-deleted AdVs) supplying sufficient amount of VA RNA in trans. Recently we have developed a method for efficient production of VA-deleted AdVs and succeeded to obtain a high titer of VA-deleted AdVs. Then we construct VA-deleted AdVs expressing shRNA that knockdown the replication of hepatitis C virus (HCV). In fact, VA-deleted AdVs expressing these shRNAs suppressed HCV replication more effectively than conventional FG-AdV. Therefore, we showed that VA RNAs expressed from FG-AdVs probably compete with shRNA in the maturation pathway and reduce the effect of shRNAs. We think that VA-deleted AdV may substitute for current FG-AdVs and become a standard AdV.


Subject(s)
Adenoviridae/genetics , Genetic Vectors , Adenoviridae/immunology , Adenoviridae/physiology , Cells, Cultured , Genetic Vectors/immunology , Hepacivirus/physiology , Humans , RNA Polymerase III/physiology , RNA, Small Interfering , RNA, Viral/genetics , RNA, Viral/physiology , Transcription, Genetic , Virus Replication/genetics
13.
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
14.
J Biol Chem ; 286(28): 25256-64, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21596743

ABSTRACT

Chronic infection of Helicobacter pylori in the stomach mucosa with translocation of the bacterial cytotoxin-associated gene A (CagA) effector protein via the cag-Type IV secretion system (TFSS) into host epithelial cells are major risk factors for gastritis, gastric ulcers, and cancer. The blood group antigen-binding adhesin BabA mediates the adherence of H. pylori to ABO/Lewis b (Le(b)) blood group antigens in the gastric pit region of the human stomach mucosa. Here, we show both in vitro and in vivo that BabA-mediated binding of H. pylori to Le(b) on the epithelial surface augments TFSS-dependent H. pylori pathogenicity by triggering the production of proinflammatory cytokines and precancer-related factors. We successfully generated Le(b)-positive cell lineages by transfecting Le(b)-negative cells with several glycosyltransferase genes. Using these established cell lines, we found increased mRNA levels of proinflammatory cytokines (CCL5 and IL-8) as well as precancer-related factors (CDX2 and MUC2) after the infection of Le(b)-positive cells with WT H. pylori but not with babA or TFSS deletion mutants. This increased mRNA expression was abrogated when Le(b)-negative cells were infected with WT H. pylori. Thus, H. pylori can exploit BabA-Le(b) binding to trigger TFSS-dependent host cell signaling to induce the transcription of genes that enhance inflammation, development of intestinal metaplasia, and associated precancerous transformations.


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Adhesion/physiology , Bacterial Secretion Systems/physiology , Helicobacter Infections/metabolism , Helicobacter pylori/physiology , Helicobacter pylori/pathogenicity , Adhesins, Bacterial/genetics , Animals , CHO Cells , Chemokine CCL5/biosynthesis , Chemokine CCL5/genetics , Cricetinae , Cricetulus , Dogs , Gastric Mucosa/immunology , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Gene Deletion , Helicobacter Infections/genetics , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Interleukin-8/biosynthesis , Interleukin-8/genetics , Lewis Blood Group Antigens/genetics , Lewis Blood Group Antigens/metabolism , Metaplasia/genetics , Metaplasia/metabolism , Metaplasia/microbiology , Metaplasia/pathology , Mucin-2/biosynthesis , Mucin-2/genetics , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Precancerous Conditions/microbiology , Precancerous Conditions/pathology , Signal Transduction/genetics
15.
Biochem Biophys Res Commun ; 417(3): 945-50, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22202173

ABSTRACT

Both transfection and adenovirus vectors are commonly used in studies measuring gene expression. However, the real DNA copy number that is actually transduced into target cells cannot be measured using quantitative PCR because attached DNA present on the cell surface is difficult to distinguish from successfully transduced DNA. Here, we used Cre/loxP system to show that most of the transfected DNA was in fact attached to the cell surface; in contrast, most of the viral vector DNA used to infect the target cells was present inside the cells after the cells were washed according to the conventional infection protocol. We applied this characteristic to adenoviral vector titration. Current methods of vector titration using the growth of 293 cells are influenced by the effect of the expressed gene product as well as the cell conditions and culture techniques. The titration method proposed here indicates the copy numbers introduced to the target cells using a control vector that is infected in parallel (relative vector titer: rVT). Moreover, the new titration method is simple and reliable and may replace the current titration methods of viral vectors.


Subject(s)
Adenoviridae/genetics , DNA, Viral/analysis , Genetic Vectors/analysis , Genome, Viral , Polymerase Chain Reaction/methods , Base Sequence , Cell Membrane/virology , DNA, Viral/genetics , Genetic Vectors/genetics , HEK293 Cells , Humans , Molecular Sequence Data , Transduction, Genetic
16.
Genes Cells ; 16(7): 765-77, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21707874

ABSTRACT

Cre and FLP recombinases mediate not only specific deletions and insertions, but also the recombinase-mediated cassette exchange (RMCE) reaction, which is used in cell biotechnology including ES cells and mouse genetics. However, comparison of efficiencies for Cre and FLP in RMCE has not been made. We here examined the detailed process of RMCE with Cre and FLP in vitro using mutant loxP 2272 and three mutant FRTs (FRT G, FRT H, and FRT F3) and then quantitatively compared the RMCE reactions in vitro. Interestingly, in the in vitro reactions, the RMCE efficiency of Cre reached a plateau level of approximately 5% and did not proceed further, whereas that of FLPe reached approximately 12-13%, showing that FLPe reached a higher level of efficiency than Cre possibly when they were supplied at a very high concentration. Moreover, we quantitatively compared the production efficiency of E1-deleted adenovirus vector using the RMCE method with Cre or FLP. The results showed that FLPe was again found more efficient than Cre in RMCE reaction. Thus, although Cre is considered more active than, or similar to, FLPe, it may not be necessarily true for RMCE reaction. Possible reasons explaining these results are discussed.


Subject(s)
Adenoviridae/genetics , DNA Nucleotidyltransferases/metabolism , Genetic Vectors/biosynthesis , Integrases/metabolism , Recombination, Genetic/genetics , Adenoviridae/growth & development , Animals , Cell Line , DNA Nucleotidyltransferases/genetics , Genetic Vectors/genetics , HEK293 Cells , Haplorhini , Humans , Integrases/genetics , Mutagenesis, Insertional , Polymerase Chain Reaction
17.
Microbiol Immunol ; 56(7): 447-55, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22734445

ABSTRACT

First-generation AdV enables efficient gene transduction, although its immunogenicity is an important problem in vivo. Helper-dependent AdV (HD-AdV) is one possible solution to this problem. The construction of HD-AdV requires a helper virus, in which the viral packaging domain is flanked by two inserted loxP to hamper its packaging in Cre-expressing 293 cells. Here, we constructed 19L viruses containing loxP at 191 nt from the left end of the genome upstream of the packaging domain, 15L viruses bearing loxP at 143 nt, and a control ΔL virus lacking loxP at these positions. The 19L position is used worldwide, and the 15L position has been reported to result in a lower titer than that of 19L. When the titers were compared for six pairs of 19L and 15L AdV, the 19L AdV produced titers similar to, or sometimes lower than, the 15L and ΔL AdV, unlike the results of previous reports. We next chose one pair of 15L and 19L AdV that produced titers similar to that of ΔL and a competitor AdV lacking loxP for use in a competition assay. When a small amount of the competitor AdV was co-infected, both the 15L and the 19L AdV, but not ΔL, gradually became minority components during subsequent viral passages. Therefore, the loxP insertions at 143 nt and 191 nt decreased the viral packaging efficiency.


Subject(s)
Adenoviridae Infections/virology , Adenoviridae/genetics , Genetic Vectors/genetics , Mutagenesis, Insertional , Virus Assembly , Adenoviridae/chemistry , Adenoviridae/physiology , Base Sequence , Cell Line , Genetic Vectors/physiology , Helper Viruses/genetics , Helper Viruses/physiology , Humans , Molecular Sequence Data , Virus Integration
18.
Am J Respir Cell Mol Biol ; 45(3): 489-97, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21131445

ABSTRACT

Pulmonary fibrosis has high rates of mortality and morbidity, but there is no established therapy at present. We demonstrate here that bleomycin-induced pulmonary fibrosis in mice is ameliorated by intratracheal administration of keratinocyte growth factor (KGF)-expressing adenovirus vector. Progressive pulmonary fibrosis was created by continuous subcutaneous administration of 120 mg/kg of bleomycin subcutaneously using an osmotic pump twice from Day 1 to 7 and Day 29 to 35. The mice initially exhibited subpleural fibrosis and then exhibited advanced fibrosis in the parenchyma of the lungs. These histopathological changes were accompanied by reduced lung compliance (0.041 ± 0.011 versus 0.097 ± 0.004; P < 0.001), reduced messenger expression of surfactant proteins, and reduced KGF messenger expression in the lungs at 4 weeks compared with naive group. Intratracheal instillation of Ad-KGF at 1 week after the first administration of bleomycin increased KGF mRNA expression in the lungs compared with the fibrosis-induced mice that received saline alone. The phenotype was associated with alveolar epithelial cell proliferation, increased pulmonary compliance (0.062 ± 0.005 versus 0.041 ± 0.011; P = 0.023), and decreased mortality (survival rate on Day 56: 68.8% versus 0%; P = 0.002), compared with mice receiving only the saline vehicle. These observations suggest the therapeutic utility of a KGF-expressing adenoviral vector for pulmonary fibrosis.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Bleomycin/pharmacology , Fibroblast Growth Factor 7/metabolism , Pulmonary Fibrosis/metabolism , Adenoviridae/metabolism , Animals , Cell Proliferation , Genetic Therapy/methods , Immunohistochemistry/methods , Lung/immunology , Male , Mice , Mice, Inbred C57BL , Phenotype , Pulmonary Surfactant-Associated Protein D/metabolism , RNA, Messenger/metabolism , Surface-Active Agents/metabolism , Transforming Growth Factor beta1/metabolism
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL