Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Pathog ; 19(8): e1011591, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37585449

RESUMO

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.


Assuntos
Carcinoma Hepatocelular , Hepatite C , Neoplasias Hepáticas , Animais , Camundongos , Hepacivirus/fisiologia , Hepatite C/metabolismo , Hepcidinas/genética , Hepcidinas/metabolismo , Ferro/metabolismo , Ativação Transcricional , Regulação para Cima
2.
Int J Mol Sci ; 23(5)2022 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-35269691

RESUMO

The rat is an important animal model for understanding gene function and developing human disease models. Knocking out a gene function in rats was difficult until recently, when a series of genome editing (GE) technologies, including zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and the type II bacterial clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated Cas9 (CRISPR/Cas9) systems were successfully applied for gene modification (as exemplified by gene-specific knockout and knock-in) in the endogenous target genes of various organisms including rats. Owing to its simple application for gene modification and its ease of use, the CRISPR/Cas9 system is now commonly used worldwide. The most important aspect of this process is the selection of the method used to deliver GE components to rat embryos. In earlier stages, the microinjection (MI) of GE components into the cytoplasm and/or nuclei of a zygote was frequently employed. However, this method is associated with the use of an expensive manipulator system, the skills required to operate it, and the egg transfer (ET) of MI-treated embryos to recipient females for further development. In vitro electroporation (EP) of zygotes is next recognized as a simple and rapid method to introduce GE components to produce GE animals. Furthermore, in vitro transduction of rat embryos with adeno-associated viruses is potentially effective for obtaining GE rats. However, these two approaches also require ET. The use of gene-engineered embryonic stem cells or spermatogonial stem cells appears to be of interest to obtain GE rats; however, the procedure itself is difficult and laborious. Genome-editing via oviductal nucleic acids delivery (GONAD) (or improved GONAD (i-GONAD)) is a novel method allowing for the in situ production of GE zygotes existing within the oviductal lumen. This can be performed by the simple intraoviductal injection of GE components and subsequent in vivo EP toward the injected oviducts and does not require ET. In this review, we describe the development of various approaches for producing GE rats together with an assessment of their technical advantages and limitations, and present new GE-related technologies and current achievements using those rats in relation to human diseases.


Assuntos
Sistemas CRISPR-Cas , Ácidos Nucleicos , Animais , Sistemas CRISPR-Cas/genética , Feminino , Edição de Genes/métodos , Genoma/genética , Humanos , Ratos , Nucleases dos Efetores Semelhantes a Ativadores de Transcrição/genética , Nucleases de Dedos de Zinco/genética
3.
Int J Mol Sci ; 23(19)2022 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-36233218

RESUMO

Specific antibodies are necessary for cellular and tissue expression, biochemical, and functional analyses of protein complexes. However, generating a specific antibody is often time-consuming and effort-intensive. The epitope tagging of an endogenous protein at an appropriate position can overcome this problem. Here, we investigated epitope tag position using AlphaFold2 protein structure prediction and developed Flag/DYKDDDDK tag knock-in CaMKIIα and CaMKIIß mice by combining CRISPR-Cas9 genome editing with electroporation (i-GONAD). With i-GONAD, it is possible to insert a small fragment of up to 200 bp into the genome of the target gene, enabling efficient and convenient tagging of a small epitope. Experiments with commercially available anti-Flag antibodies could readily detect endogenous CaMKIIα and ß proteins by Western blotting, immunoprecipitation, and immunohistochemistry. Our data demonstrated that the generation of Flag/DYKDDDDK tag knock-in mice by i-GONAD is a useful and convenient choice, especially if specific antibodies are unavailable.


Assuntos
Eletroporação , Edição de Genes , Animais , Anticorpos/metabolismo , Western Blotting , Sistemas CRISPR-Cas/genética , Epitopos/genética , Epitopos/metabolismo , Gônadas/metabolismo , Camundongos
4.
Int J Mol Sci ; 23(18)2022 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-36142589

RESUMO

Improved genome editing via oviductal nucleic acids delivery (i-GONAD) is a new technology enabling in situ genome editing of mammalian zygotes exiting the oviductal lumen, which is now available in mice, rats, and hamsters. In this method, CRISPR/Cas9 genome-editing reagents are delivered directly to the oviducts of pregnant animals (corresponding to late zygote stage). After intraoviductal instillation, electric shock to the entire oviduct was provided with a specialized electroporation (EP) device to drive the genome editing reagents into the zygotes present in the oviductal lumen. i-GONAD toward early zygotes has been recognized as difficult, because they are tightly surrounded by a cumulus cell layer, which often hampers effective transfer of nucleic acids to zygotes. However, in vivo EP three min after intraoviductal instillation of the genome-editing reagents enabled genome editing of early zygotes with an efficiency of 70%, which was in contrast with the rate of 18% when in vivo EP was performed immediately after intraoviductal instillation at Day 0.5 of pregnancy (corresponding to 13:00-13:30 p.m. on the day when vaginal plug was recognized after natural mating). We also found that addition of hyaluronidase, an enzyme capable of removing cumulus cells from a zygote, slightly enhanced the efficiency of genome editing in early zygotes. These findings suggest that cumulus cells surrounding a zygote can be a barrier for efficient generation of genome-edited mouse embryos and indicate that a three-minute interval before in vivo EP is effective for achieving i-GONAD-mediated genome editing at the early zygote stage. These results are particularly beneficial for researchers who want to perform genome editing experiments targeting early zygotes.


Assuntos
Edição de Genes , Ácidos Nucleicos , Animais , Sistemas CRISPR-Cas , Eletroporação/métodos , Feminino , Edição de Genes/métodos , Gônadas , Humanos , Hialuronoglucosaminidase/genética , Mamíferos/genética , Camundongos , Oviductos , Gravidez , Ratos , Ribonucleoproteínas/genética , Zigoto
5.
BMC Biotechnol ; 21(1): 63, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34724929

RESUMO

BACKGROUND: Improved genome-editing via oviductal nucleic acids delivery (i-GONAD) is a new technology that facilitates in situ genome-editing of mammalian zygotes exiting the oviductal lumen. The i-GONAD technology has been developed for use in mice, rats, and hamsters; however, oligonucleotide (ODN)-based knock-in (KI) is more inefficient in rats than mice. To improve the efficiency of i-GONAD in rats we examined KI efficiency using three guide RNAs (gRNA), crRNA1, crRNA2 and crRNA3. These gRNAs recognize different portions of the target locus, but also overlap each other in the target locus. We also examined the effects of commercially available KI -enhancing drugs (including SCR7, L755,507, RS-1, and HDR enhancer) on i-GONAD-mediated KI efficiency. RESULTS: The KI efficiency in rat fetuses generated after i-GONAD with crRNA2 and single-stranded ODN was significantly higher (24%) than crRNA1 (5%; p < 0.05) or crRNA3 (0%; p < 0.01). The KI efficiency of i-GONAD with triple gRNAs was 11%. These findings suggest that KI efficiency largely depends on the type of gRNA used. Furthermore, the KI efficiency drugs, SCR7, L755,507 and HDR enhancer, all of which are known to enhance KI efficiency, increased KI efficiency using the i-GONAD with crRNA1 protocol. In contrast, only L755,507 (15 µM) increased KI efficiency using the i-GONAD with crRNA2 protocol. None of them were significantly different. CONCLUSIONS: We attempted to improve the KI efficiency of i-GONAD in rats. We demonstrated that the choice of gRNA is important for determining KI efficiency and insertion and deletion rates. Some drugs (e.g. SCR7, L755,507 and HDR enhancer) that are known to increase KI efficiency in culture cells were found to be effective in i-GONAD in rats, but their effects were limited.


Assuntos
Edição de Genes , Ácidos Nucleicos , Animais , Sistemas CRISPR-Cas/genética , Eletroporação , Feminino , Gônadas , Humanos , Camundongos , Ratos
6.
Hepatology ; 66(5): 1430-1443, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28621467

RESUMO

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).


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Hepatite C/metabolismo , Cirrose Hepática/virologia , Fígado/patologia , Fator de Crescimento Transformador beta2/metabolismo , Animais , Comunicação Autócrina , Fibrose , Regulação da Expressão Gênica , Células Estreladas do Fígado/patologia , Hepatite C/complicações , Hepatite C/patologia , Cirrose Hepática/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Comunicação Parácrina , Fator de Crescimento Transformador beta1/metabolismo
7.
Zoolog Sci ; 35(2): 172-178, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29623786

RESUMO

Experimental testicular teratomas (ETTs) can be induced in 129/Sv mouse by E12.5 fetal testes transplant into adult testes. Previously, we conducted linkage analysis to explore candidate genes possibly involved in ETT development using F2 intercross fetuses derived from F1[LTXBJ × 129/Sv- + /Ter (+ /+)] hybrids. By linkage analysis on Chr 18 and Chr 19, we identified the genomic locus for experimental testicular teratoma 1 (ett1) on Chr 18. In the present study, we conducted additional mapping and linkage analysis on teratoma susceptibility and genome composition on Chr 1-17. The results revealed two new candidate loci, experimental testicular teratoma 2 (ett2) and experimental testicular teratoma 3 (ett3), on Chr 3 and 7. Interestingly, the rates of ETT generation were increased in the case of ett2 and ett3 regions replaced with LTXBJ strain. To determine whether a polymorphic gene was present, we performed exome analysis of 129/Sv- + /Ter (+ /+) and LTXBJ. This revealed the presence of SNPs in all three loci, ett1 to ett3. ett1 contains polymorphic Mc4r; ett2 contains polymorphic Polr3c, Cd160, and Pdzk1; and ett3 contains polymorphic Prmt3. We found additional loci responsible for ETT formation, namely, ett2 and ett3, and identified candidate genes in these regions by exome analysis.


Assuntos
Loci Gênicos , Genoma , Polimorfismo Genético , Teratoma/genética , Neoplasias Testiculares/genética , Animais , Masculino , Camundongos , Camundongos da Linhagem 129 , Teratoma/metabolismo , Neoplasias Testiculares/metabolismo
8.
Int Immunol ; 27(11): 567-77, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25977306

RESUMO

Analysis of the hematopoiesis of non-human primates is important to clarify the evolution of primate-specific hematopoiesis and immune regulation. However, the engraftment and development of the primate hematopoietic system are well-documented only in humans and are not clear in non-human primates. Callithrix jacchus (common marmoset, CM) is a New World monkey with a high rate of pregnancy and small size that lives in closed colonies. As stem cell factor (SCF) is an essential molecule for hematopoietic stem cell development in mice and humans, we focused on CD117, the SCF receptor, and examined whether CD117-expressing cells possess the hematopoietic stem/progenitor cell characteristics of newborn marmoset-derived hematopoietic cells that can develop into T cells and B cells. When CD117(+) cell fractions of the bone marrow were transplanted into immunodeficient NOD (non-obese diabetic)/Shi-scid, common γc-null (NOG) mice, these cells engrafted efficiently in the bone marrow and spleens of the NOG mice. The CD117(+) cells developed into myeloid lineage cells, CD20(+) B cells and CD3(+) T cells, which could express CM cytokines in vivo. The development of B cells did not precede that of T cells. The development of CD8(+) T cells was dominant in NOG mice. The engraftment was comparable for both CD117(+)CD34(+) cells and CD117(+)CD34(-) cells. These results suggest that the CD117(+) cell fraction can differentiate into all three cell lineages, and the development of marmoset immunity in the xenogeneic environment follows diverse developmental pathways compared with human immunity.


Assuntos
Diferenciação Celular , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Animais , Animais Recém-Nascidos , Antígenos de Superfície/metabolismo , Callithrix , Autorrenovação Celular , Sangue Fetal/citologia , Sobrevivência de Enxerto , Transplante de Células-Tronco Hematopoéticas , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Imunofenotipagem , Linfócitos/citologia , Linfócitos/metabolismo , Camundongos , Células Mieloides/citologia , Células Mieloides/metabolismo , Fenótipo
9.
Mamm Genome ; 25(7-8): 317-26, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24997020

RESUMO

Spontaneous testicular teratomas (STTs) composed by various kinds of tissues are derived from primordial germ cells (PGCs) in the fetal testes of the mouse. In contrast, intra-testicular grafts of the mouse strain (129/Sv-Ter (+/+)) fetal testes possessed the ability to develop the experimental testicular teratomas (ETTs), indistinguishable from the STTs at a morphological level. In this study, linkage analysis was performed for exploration of possible candidate genes involving in ETT development using F2 intercross fetuses derived from [LTXBJ × 129/Sv-Ter (+/+)] F1 hybrids. Linkage analysis with selected simple sequence length polymorphisms along chromosomes 18 and 19, which have been expected to contain ETT-susceptibility loci, demonstrated that a novel recessive candidate gene responsible for ETT development is located in 1.1 Mb region between the SSLP markers D18Mit81 and D18Mit184 on chromosome 18 in the 129/Sv-Ter (+/+) genetic background. Since this locus is different from the previously known loci (including Ter, pgct1, and Tgct1) for STT development, we named this novel gene "experimental testicular teratoma 1 (ett1)". To resolve the location of ett1 independently from other susceptibility loci, ett1 loci was introduced in a congenic strain in which the distal segment of chromosome 18 in LTXBJ strain mice had been replaced by a 1.99 Mbp genomic segment of the 129/Sv-Ter (+/+) mice. Congenic males homozygous for the ett1 loci were confirmed to have the ability to form ETTs, indicating that this locus contain the gene responsible for ETTs. We listed candidate genes included in this region, and discussed about their possible involvement in induction of ETTs.


Assuntos
Cromossomos de Mamíferos/genética , Loci Gênicos , Predisposição Genética para Doença , Teratoma/genética , Neoplasias Testiculares/genética , Animais , Feminino , Masculino , Camundongos da Linhagem 129 , Polimorfismo Genético , Teratoma/patologia , Neoplasias Testiculares/patologia , Testículo/embriologia , Testículo/patologia
10.
Sci Rep ; 13(1): 6756, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-37127675

RESUMO

Teratomas in mice, composed of different tissue types, are derived from primordial germ cells in the fetal gonads. Previously, we identified a locus responsible for experimental testicular teratoma (ETT) formation on chromosome 18, referred to as ett1. The strongest candidate sequence in the ett1 locus was found to be a missense mutation in the melanocortin 4 receptor (Mc4r), Mc4rG25S. We established a strain with a point mutation in the Mc4r gene in the ETT-nonsusceptible LT strain, called LT- Mc4rG25S, by genome editing. Surprisingly, highly developed ovarian teratomas (OTs), rather than testicular teratomas, appeared in the LT-Mc4rG25S strain. The results demonstrated that Mc4r is also one of the genes responsible for OT formation and suggested that missense mutations in Mc4r promote teratoma formation in both sexes. In this study, we performed ETT experiments in different host-graft combinations of the LT-Mc4rG25S and LT strains. Furthermore, the expression of MC4R in germ cells in the testis was demonstrated. Expression of Mc4r in testis was also confirmed by RT-PCR. The results demonstrated that MC4R is expressed in germ cells in the testis and that a point mutation in the Mc4r gene is responsible for ETT formation.


Assuntos
Teratoma , Neoplasias Testiculares , Masculino , Humanos , Feminino , Camundongos , Animais , Teratoma/metabolismo , Neoplasias Testiculares/genética , Receptor Tipo 4 de Melanocortina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA