Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Am J Pathol ; 193(6): 755-768, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36868469

RESUMO

miR-194 is abundantly expressed in hepatocytes, and its depletion increases hepatic resistance to acetaminophen-induced acute injuries. In this study, the biological role of miR-194 in cholestatic liver injury was investigated by using miR-194/miR-192 cluster liver-specific knockout (LKO) mice, in which no liver injuries or metabolic disorders were predisposed. Bile duct ligation (BDL) and 1-naphthyl isothiocyanate (ANIT) were applied to LKO and matched control wild-type (WT) mice to induce hepatic cholestasis. Periportal liver damage, mortality rate, and liver injury biomarkers in LKO mice were significantly less than in WT mice after BDL and ANIT injection. Intrahepatic bile acid level was significantly lower in the LKO liver within 48 hours of BDL- and ANIT-induced cholestasis compared with WT. Western blot analysis showed that ß-catenin (CTNNB1) signaling and genes involved in cellular proliferation were activated in BDL- and ANIT-treated mice. The expression levels of cholesterol 7 alpha-hydroxylase (CYP7A1), pivotal in bile synthesis, and its upstream regulator hepatocyte nuclear factor 4α were reduced in primary LKO hepatocytes and liver tissues compared with WT. The knockdown of miR-194 using miRNA inhibitors reduced CYP7A1 expression in WT hepatocytes. In contrast, the knockdown of CTNNB1 and overexpression of miR-194, but not miR-192, in LKO hepatocytes and AML12 cells increased CYP7A1 expression. In conclusion, the results suggest that the loss of miR-194 ameliorates cholestatic liver injury and may suppress CYP7A1 expression via activation of CTNNB1 signaling.


Assuntos
Colestase , Hepatopatias , Camundongos , Animais , beta Catenina/metabolismo , Colestase/genética , Colestase/metabolismo , Hepatopatias/metabolismo , Hepatócitos/metabolismo , Ácidos e Sais Biliares/metabolismo , Colesterol/metabolismo
2.
Gut ; 71(9): 1876-1891, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-34933915

RESUMO

BACKGROUND AND OBJECTIVES: Liver fibrosis (LF) occurs following chronic liver injuries. Currently, there is no effective therapy for LF. Recently, we identified thioredoxin domain containing 5 (TXNDC5), an ER protein disulfide isomerase (PDI), as a critical mediator of cardiac and lung fibrosis. We aimed to determine if TXNDC5 also contributes to LF and its potential as a therapeutic target for LF. DESIGN: Histological and transcriptome analyses on human cirrhotic livers were performed. Col1a1-GFPTg , Alb-Cre;Rosa26-tdTomato and Tie2-Cre/ERT2;Rosa26-tdTomato mice were used to determine the cell type(s) where TXNDC5 was induced following liver injury. In vitro investigations were conducted in human hepatic stellate cells (HSCs). Col1a2-Cre/ERT2;Txndc5fl/fl (Txndc5cKO ) and Alb-Cre;Txndc5fl/fl (Txndc5Hep-cKO ) mice were generated to delete TXNDC5 in HSCs and hepatocytes, respectively. Carbon tetrachloride treatment and bile duct ligation surgery were employed to induce liver injury/fibrosis in mice. The extent of LF was quantified using histological, imaging and biochemical analyses. RESULTS: TXNDC5 was upregulated markedly in human and mouse fibrotic livers, particularly in activated HSC at the fibrotic foci. TXNDC5 was induced by transforming growth factor ß1 (TGFß1) in HSCs and it was both required and sufficient for the activation, proliferation, survival and extracellular matrix production of HSC. Mechanistically, TGFß1 induces TXNDC5 expression through increased ER stress and ATF6-mediated transcriptional regulation. In addition, TXNDC5 promotes LF by redox-dependent JNK and signal transducer and activator of transcription 3 activation in HSCs through its PDI activity, activating HSCs and making them resistant to apoptosis. HSC-specific deletion of Txndc5 reverted established LF in mice. CONCLUSIONS: ER protein TXNDC5 promotes LF through redox-dependent HSC activation, proliferation and excessive extracellular matrix production. Targeting TXNDC5, therefore, could be a potential novel therapeutic strategy to ameliorate LF.


Assuntos
Células Estreladas do Fígado , Cirrose Hepática , Animais , Tetracloreto de Carbono/efeitos adversos , Tetracloreto de Carbono/metabolismo , Fibrose , Células Estreladas do Fígado/metabolismo , Hepatócitos/metabolismo , Humanos , Fígado/metabolismo , Cirrose Hepática/patologia , Camundongos , Isomerases de Dissulfetos de Proteínas/efeitos adversos , Isomerases de Dissulfetos de Proteínas/genética , Isomerases de Dissulfetos de Proteínas/metabolismo , Tiorredoxinas/genética , Tiorredoxinas/metabolismo
3.
FASEB J ; 33(4): 5571-5584, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30640520

RESUMO

The clustered regularly interspaced short palindromic repeat (CRISPR)/Cas9 technology facilitates somatic genome editing to reveal cooperative genetic interactions at the cellular level without extensive breeding between different mutant animals. Here we propose a transgenic inducible Cas9 effector-CRISPR mutagen ( ICE CRIM) mouse model in which CRISPR/Cas9-mediated somatic mutagenesis events can occur in response to Cre expression. The well-known tumor suppressor gene, Trp53, and 2 important DNA mismatch repair genes, Mlh1 and Msh2, were selected to be our somatic mutagenesis targets. Amplicon-based sequencing was performed to validate the editing efficiency and to identify the mutant allelic series. Crossed with various Cre lines, the Trp53 ICE CRIM alleles were activated to generate targeted cancer gene somatic or germ line mutant variants. We provide experimental evidence to show that an activated ICE CRIM can mutate both targeted alleles within a cell. Simultaneous disruption of multiple genes was also achieved when there were multiple single-guide RNA expression cassettes embedded within an activated ICE CRIM. Our mouse model can be used to generate mutant pools in vivo, which enables a functional screen to be performed in situ. Our results also provide evidence to support a monoclonal origin of hematopoietic neoplasms and to indicate that DNA mismatch repair deficiency accelerates tumorigenesis in Trp53 mutant genetic background.-Fan, H.-H., Yu, I.-S., Lin, Y.-H., Wang, S.-Y., Liaw, Y.-H., Chen, P.-L., Yang, T.-L., Lin, S.-W., Chen, Y.-T. P53 ICE CRIM mouse: a tool to generate mutant allelic series in somatic cells and germ lines for cancer studies.


Assuntos
Sistemas CRISPR-Cas/genética , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Mutação/genética , Neoplasias/genética , Proteína Supressora de Tumor p53/genética , Alelos , Animais , Edição de Genes/métodos , Marcação de Genes/métodos , Células Germinativas , Camundongos , Camundongos Transgênicos/genética , Mutagênese/genética , Oncogenes/genética , RNA Guia de Cinetoplastídeos/genética
4.
Nucleic Acids Res ; 45(18): 10492-10503, 2017 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-28985359

RESUMO

Telomerase is highly expressed in cancer and embryonic stem cells (ESCs) and implicated in controlling genome integrity, cancer formation and stemness. Previous studies identified that Krüppel-like transcription factor 4 (KLF4) activates telomerase reverse transcriptase (TERT) expression and contributes to the maintenance of self-renewal in ESCs. However, little is known about how KLF4 regulates TERT expression. Here, we discover poly(ADP-ribose) polymerase 1 (PARP1) as a novel KLF4-interacting partner. Knockdown of PARP1 reduces TERT expression and telomerase activity not only in cancer cells, but also in human and mouse ESCs. Recruitment of KLF4 to TERT promoter is reduced in PARP1-suppressed cells. The poly(ADP-ribose) polymerase activity is dispensable, while the oligo(ADP-ribose) polymerase activity is required for the PARP1- and KLF4-mediated TERT activation. Repression of Parp1 in mouse ESCs decreases expression of pluripotent markers and induces differentiation. These results suggest that PARP1 recruits KLF4 to activate telomerase expression and stem cell pluripotency, indicating a positive regulatory role of the PARP1-KLF4 complex in telomerase expression in cancer and stem cells.


Assuntos
Células-Tronco Embrionárias/metabolismo , Fatores de Transcrição Kruppel-Like/fisiologia , Neoplasias/genética , Poli(ADP-Ribose) Polimerase-1/fisiologia , Telomerase/genética , Animais , Diferenciação Celular/genética , Células Cultivadas , Embrião de Mamíferos , Células-Tronco Embrionárias/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Neoplasias/patologia , Poli(ADP-Ribose) Polimerase-1/metabolismo , Telomerase/metabolismo
5.
Neurobiol Dis ; 77: 35-48, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25725421

RESUMO

Dravet syndrome (DS) is characterized by severe infant-onset myoclonic epilepsy along with delayed psychomotor development and heightened premature mortality. A primary monogenic cause is mutation of the SCN1A gene, which encodes the voltage-gated sodium channel subunit Nav1.1. The nature and timing of changes caused by SCN1A mutation in the hippocampal dentate gyrus (DG) network, a core area for gating major excitatory input to hippocampus and a classic epileptogenic zone, are not well known. In particularly, it is still not clear whether the developmental deficit of this epileptogenic neural network temporally matches with the progress of seizure development. Here, we investigated the emerging functional and structural deficits of the DG network in a novel mouse model (Scn1a(E1099X/+)) that mimics the genetic deficit of human DS. Scn1a(E1099X/+) (Het) mice, similarly to human DS patients, exhibited early spontaneous seizures and were more susceptible to hyperthermia-induced seizures starting at postnatal week (PW) 3, with seizures peaking at PW4. During the same period, the Het DG exhibited a greater reduction of Nav1.1-expressing GABAergic neurons compared to other hippocampal areas. Het DG GABAergic neurons showed altered action potential kinetics, reduced excitability, and generated fewer spontaneous inhibitory inputs into DG granule cells. The effect of reduced inhibitory input to DG granule cells was exacerbated by heightened spontaneous excitatory transmission and elevated excitatory release probability in these cells. In addition to electrophysiological deficit, we observed emerging morphological abnormalities of DG granule cells. Het granule cells exhibited progressively reduced dendritic arborization and excessive spines, which coincided with imbalanced network activity and the developmental onset of spontaneous seizures. Taken together, our results establish the existence of significant structural and functional developmental deficits of the DG network and the temporal correlation between emergence of these deficits and the onset of seizures in Het animals. Most importantly, our results uncover the developmental deficits of neural connectivity in Het mice. Such structural abnormalities likely further exacerbate network instability and compromise higher-order cognitive processing later in development, and thus highlight the multifaceted impacts of Scn1a deficiency on neural development.


Assuntos
Giro Denteado/patologia , Epilepsias Mioclônicas/genética , Epilepsias Mioclônicas/patologia , Mutação/genética , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Rede Nervosa/patologia , Convulsões/fisiopatologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Fatores Etários , Animais , Animais Recém-Nascidos , Giro Denteado/crescimento & desenvolvimento , Modelos Animais de Doenças , Glutamato Descarboxilase/metabolismo , Hipertermia Induzida/efeitos adversos , Técnicas In Vitro , Lisina/análogos & derivados , Lisina/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Modelos Moleculares , Neurônios/ultraestrutura , Convulsões/etiologia , Convulsões/genética , Ácido gama-Aminobutírico/metabolismo
6.
Dev Biol ; 374(2): 295-307, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23261931

RESUMO

The basic helix-loop-helix transcription factor Twist1 is a key regulator of craniofacial development. Twist1-null mouse embryos exhibit failure of cephalic neural tube closure and abnormal head development and die at E11.0. To dissect the function of Twist1 in the cranial mesoderm beyond mid-gestation, we used Mesp1-Cre to delete Twist1 in the anterior mesoderm, which includes the progenitors of the cranial mesoderm. Deletion of Twist1 in mesoderm cells resulted in loss and malformations of the cranial mesoderm-derived skeleton. Loss of Twist1 in the mesoderm also resulted in a failure to fully segregate the mesoderm and the neural crest cells, and the malformation of some cranial neural crest-derived tissues. The development of extraocular muscles was compromised whereas the differentiation of branchial arch muscles was not affected, indicating a differential requirement for Twist1 in these two types of craniofacial muscle. A striking effect of the loss of Twist1 was the inability of the mesodermal cells to maintain their mesenchymal characteristics, and the acquisition of an epithelial-like morphology. Our findings point to a role of Twist1 in maintaining the mesenchyme architecture and the progenitor state of the mesoderm, as well as mediating mesoderm-neural crest interactions in craniofacial development.


Assuntos
Embrião de Mamíferos/metabolismo , Mesoderma/metabolismo , Proteínas Nucleares/genética , Proteína 1 Relacionada a Twist/genética , Animais , Apoptose/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Anormalidades Craniofaciais/genética , Anormalidades Craniofaciais/metabolismo , Anormalidades Craniofaciais/patologia , Embrião de Mamíferos/embriologia , Imunofluorescência , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Mesoderma/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Anatômicos , Modelos Genéticos , Crista Neural/citologia , Crista Neural/embriologia , Crista Neural/metabolismo , Proteínas Nucleares/deficiência , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Crânio/embriologia , Crânio/metabolismo , Fatores de Tempo , Proteína 1 Relacionada a Twist/deficiência
7.
Hum Mol Genet ; 21(19): 4270-85, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22763239

RESUMO

Mutation in CUL4B, which encodes a scaffold protein of the E3 ubiquitin ligase complex, has been found in patients with X-linked mental retardation (XLMR). However, early deletion of Cul4b in mice causes prenatal lethality, which has frustrated attempts to characterize the phenotypes in vivo. In this report, we successfully rescued Cul4b mutant mice by crossing female mice in which exons 4-5 of Cul4b were flanked by loxP sequences with Sox2-Cre male mice. In Cul4b-deficient (Cul4b(Δ)/Y) mice, no CUL4B protein was detected in any of the major organs, including the brain. In the hippocampus, the levels of CUL4A, CUL4B substrates (TOP1, ß-catenin, cyclin E and WDR5) and neuronal markers (MAP2, tau-1, GAP-43, PSD95 and syn-1) were not sensitive to Cul4b deletion, whereas the number of parvalbumin (PV)-positive GABAergic interneurons was decreased in Cul4b(Δ)/Y mice, especially in the dentate gyrus (DG). Some dendritic features, including the complexity, diameter and spine density in the CA1 and DG hippocampal neurons, were also affected by Cul4b deletion. Together, the decrease in the number of PV-positive neurons and altered dendritic properties in Cul4b(Δ)/Y mice imply a reduction in inhibitory regulation and dendritic integration in the hippocampal neural circuit, which lead to increased epileptic susceptibility and spatial learning deficits. Our results identify Cul4b(Δ)/Y mice as a potential model for the non-syndromic model of XLMR that replicates the CUL4B-associated MR and is valuable for the development of a therapeutic strategy for treating MR.


Assuntos
Proteínas Culina/genética , Modelos Animais de Doenças , Deficiência Intelectual Ligada ao Cromossomo X/genética , Camundongos , Animais , Proteínas Culina/metabolismo , Feminino , Engenharia Genética , Humanos , Masculino , Deficiência Intelectual Ligada ao Cromossomo X/metabolismo , Camundongos/genética , Camundongos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout
8.
J Ovarian Res ; 17(1): 66, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38504307

RESUMO

BACKGROUND: Quiescin sulfhydryl oxidase 2 (QSOX2) is a flavin adenine dinucleotide-dependent sulfhydryl oxidase that is known to be involved in protein folding, cell growth regulation, and redox state modification through oxidative activities. Earlier studies demonstrated the tissue and cellular localization of QSOX2 in the male reproductive tract, as well as the highly-regulated mechanism of QSOX2 protein synthesis and expression through the coordinated action of testosterone and epididymal-enriched amino acid, glutamate. However, the presence and the functions of QSOX2 in female reproduction are unknown. In this study, we applied the Cre-loxP gene manipulation system to generate the heterozygous and homozygous Qsox2 knockout mice and examined its effects on ovarian function. RESULTS: We demonstrated that QSOX2 was detected in the follicle-supporting cells (granulosa and cumulus cells) of ovarian follicles of all stages but was absent in the corpus luteum, suggesting its supportive role in folliculogenesis. In comparison with reproductive organogenesis in wild-type mice, there was no difference in testicular and epididymal structure in male Qsox2 knockout; however, Qsox2 knockout disrupted the regular ovulation process in female mice as a drastic decrease in the formation of the corpus luteum was detected, and no pregnancy was achieved when mating males with homozygous Qsox2 knockout females. RNAseq analyses further revealed that Qsox2 knockout altered critical signaling pathways and genes that are responsible for maintaining ovarian functions. CONCLUSION: Our data demonstrated for the first time that Qsox2 is critical for ovarian function in mice.


Assuntos
Células da Granulosa , Oxirredutases , Tamoxifeno , Feminino , Camundongos , Masculino , Animais , Células da Granulosa/metabolismo , Tamoxifeno/farmacologia , Tamoxifeno/metabolismo , Ovário , Ovulação , Camundongos Knockout
9.
Dev Biol ; 362(2): 132-40, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22178153

RESUMO

Development of the mouse forelimb bud depends on normal Twist1 activity. Global loss of Twist1 function before limb bud formation stops limb development and loss of Twist1 throughout the mesenchyme after limb bud initiation leads to polydactyly, the ulnarization or loss of the radius and malformations and reductions of the shoulder girdle. Here we show that conditional deletion of Twist1 by Mesp1-Cre in the mesoderm that migrates into the anterior-proximal part of the forelimb bud results in the development of supernumerary digits and carpals, the acquisition of ulna-like characteristics by the radius and malformations of the humerus and scapula. The mirror-like duplications and posteriorization of pre-axial tissues are preceded by disruptions to anterior-posterior Shh, Bmp and Fgf signaling gradients and dysregulation of transcription factors that regulate anterior-posterior limb patterning.


Assuntos
Padronização Corporal/genética , Membro Anterior/anormalidades , Membro Anterior/embriologia , Morfogênese/genética , Proteínas Nucleares/metabolismo , Transdução de Sinais/genética , Proteína 1 Relacionada a Twist/metabolismo , Animais , Apoptose/fisiologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Cruzamentos Genéticos , Primers do DNA/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Imunofluorescência , Membro Anterior/metabolismo , Deleção de Genes , Genótipo , Proteínas Hedgehog/metabolismo , Hibridização In Situ , Marcação In Situ das Extremidades Cortadas , Mesoderma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Morfogênese/fisiologia , beta-Galactosidase
10.
Development ; 137(20): 3417-26, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20826535

RESUMO

Preaxial polydactyly (PPD) is a common limb-associated birth defect characterized by extra digit(s) in the anterior autopod. It often results from ectopic sonic hedgehog (Shh) expression in the anterior limb bud. Although several transcription factors are known to restrict Shh expression to the posterior limb bud, how they function together remains unclear. Here we provide evidence from mouse conditional knockout limb buds that the bHLH family transcription factor gene Twist1 is required to inhibit Shh expression in the anterior limb bud mesenchyme. More importantly, we uncovered genetic synergism between Twist1 and the ETS family transcription factor genes Etv4 and Etv5 (collectively Etv), which also inhibit Shh expression. Biochemical data suggest that this genetic interaction is a result of direct association between TWIST1 and ETV proteins. Previous studies have shown that TWIST1 functions by forming homodimers or heterodimers with other bHLH factors including HAND2, a key positive regulator of Shh expression. We found that the PPD phenotype observed in Etv mutants is suppressed by a mutation in Hand2, indicative of genetic antagonism. Furthermore, overexpression of ETV proteins influences the dimerization of these bHLH factors. Together, our data suggest that through biochemical interactions, the Shh expression regulators ETV, TWIST1 and HAND2 attain a precise balance to establish anterior-posterior patterning of the limb.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Padronização Corporal/fisiologia , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , Polidactilia/embriologia , Proteínas Proto-Oncogênicas c-ets/metabolismo , Fatores de Transcrição/metabolismo , Proteína 1 Relacionada a Twist/metabolismo , Extremidade Superior/embriologia , Animais , Western Blotting , Dimerização , Proteínas Hedgehog/metabolismo , Imunoprecipitação , Camundongos , Camundongos Knockout , Técnicas do Sistema de Duplo-Híbrido
11.
Stem Cell Res ; 69: 103068, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36933359

RESUMO

Pathogenic variants of the WFS1 gene can cause recessive-inherited Wolfram syndrome or dominant-inherited Wolfram-like syndrome with optic atrophy and hearing impairment. Using the Sendai virus delivery system, we generated induced pluripotent stem cells from the peripheral blood mononuclear cells of a female patient with the WFS1 pathogenic variant c.2051C > T (p.Ala684Val). The resulting induced pluripotent stem cells exhibited a normal karyotype and pluripotency, as confirmed using immunofluorescence staining, and differentiated into three germ layers in vivo. This cellular model provides a useful platform for investigating the pathogenic mechanisms of both blindness and deafness related to WFS1 variants.


Assuntos
Perda Auditiva , Células-Tronco Pluripotentes Induzidas , Síndrome de Wolfram , Humanos , Feminino , Células-Tronco Pluripotentes Induzidas/patologia , Leucócitos Mononucleares/patologia , Perda Auditiva/genética , Síndrome de Wolfram/genética , Síndrome de Wolfram/patologia , Mutação
12.
Biomaterials ; 285: 121471, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35490561

RESUMO

Dissemination is an ominous feature of cancer to cause poor prognosis. Formation of tumor satellites is the first step, which is closely interdependent on the factors originating from surrounding niche. Because of lacking appropriate modeling, most studies focusing on cancer-environmental interaction depend on the static pathological analyses of specimens. Using the biomimicking system capable of inducing tumor satellite formation in vitro, the niche factors were explored for their influence on cancer budding. Our results demonstrated the real-time dynamic of tumor satellite formation of head and neck squamous cell carcinoma (HNSCC) that was significantly affected by these niche factors. Hypoxia reduced the incidence and migratory distance of HNSCC tumor satellites via decreasing the nuclear localization of intracellular domain of E-cadherin (iEcad) and ß-catenin. Three-dimensional (3D) architecture and collagen were essential for mediating hypoxia effects on nuclear translocation and signaling transduction of iEcad and ß-catenin in the HNSCC cells. The findings were further confirmed by the compatible in vivo findings of the cancer specimens. This approach revealed the effects of oxygen and extracellular matrices on tumor satellites. The cellular behaviors and molecular dynamics were along a phenotypic spectrum that conferred unique change for specific cancer populations in response to the altered microenvironment. The current study provides a platform to recapitulate the process of cancer budding, and a way to investigate the mutual interaction between the cancer cells and their surrounding microenvironment.


Assuntos
Neoplasias de Cabeça e Pescoço , beta Catenina , Caderinas , Linhagem Celular Tumoral , Colágeno , Humanos , Hipóxia , Carcinoma de Células Escamosas de Cabeça e Pescoço , Microambiente Tumoral/fisiologia
13.
Stem Cell Res ; 64: 102903, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36075118

RESUMO

Pathogenic variants of OPA1 have been associated with autosomal dominant optic atrophy (DOA), leading to optic, auditory, and other sensorineural neuropathies and myopathies. Using the Sendai virus delivery system, we generated induced pluripotent stem cells from the peripheral blood mononuclear cells of a female patient with the OPA1 pathogenic variant c.1468T>C (p.Cys490Arg). The resulting induced pluripotent stem cells exhibited a normal karyotype and pluripotency, as confirmed using immunofluorescence staining, and differentiated into three germ layers in vivo. This cellular model is a useful platform for investigating the pathogenic mechanisms of both blindness and deafness related to OPA1 variants.


Assuntos
Perda Auditiva , Células-Tronco Pluripotentes Induzidas , Atrofia Óptica Autossômica Dominante , Humanos , Feminino , Células-Tronco Pluripotentes Induzidas/patologia , Leucócitos Mononucleares/patologia , Mutação , Atrofia Óptica Autossômica Dominante/genética , Atrofia Óptica Autossômica Dominante/patologia , Perda Auditiva/genética , GTP Fosfo-Hidrolases/genética
14.
Dev Biol ; 347(1): 133-46, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20732316

RESUMO

The basic helix-loop-helix transcription factor Twist1 is essential for normal limb development. Twist1(-/-) embryos die at midgestation. However, studies on early limb buds found that Twist1(-/-) mutant limb mesenchyme has an impaired response to FGF signaling from the apical ectodermal ridge, which disrupts the feedback loop between the mesenchyme and AER, and reduces and shifts anteriorly Shh expression in the zone of polarizing activity. We have combined Twist1 null, hypomorph and conditional alleles to generate a Twist1 allelic series that survives to birth. As Twist1 activity is reduced, limb skeletal defects progress from preaxial polydactyly to girdle reduction combined with hypoplasia, aplasia or mirror symmetry of all limb segments. With reduced Twist1 activity there is striking and progressive upregulation of ectopic Shh expression in the anterior of the limb, combined with an anterior shift in the posterior Shh domain, which is expressed at normal intensity, and loss of the posterior AER. Consequently limb outgrowth is initially impaired, before an ectopic anterior Shh domain expands the AER, promoting additional growth and repatterning. Reducing the dosage of FGF targets of the Etv gene family, which are known repressors of Shh expression in anterior limb mesenchyme, strongly enhances the anterior skeletal phenotype. Conversely this and other phenotypes are suppressed by reducing the dosage of the Twist1 antagonist Hand2. Our data support a model whereby multiple Twist1 activity thresholds contribute to early limb bud patterning, and suggest how particular combinations of skeletal defects result from differing amounts of Twist1 activity.


Assuntos
Extremidades/embriologia , Proteínas Nucleares/metabolismo , Proteína 1 Relacionada a Twist/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Cartilagem/embriologia , Cartilagem/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Dosagem de Genes/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/metabolismo , Botões de Extremidades/embriologia , Botões de Extremidades/metabolismo , Camundongos , Modelos Genéticos , Mutação/genética , Proteínas Nucleares/genética , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Proteína 1 Relacionada a Twist/genética
15.
Biochim Biophys Acta Mol Cell Res ; 1868(6): 119011, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33716172

RESUMO

WD repeat and HMG-box DNA binding protein 1 (Wdhd1) is the mouse ortholog of budding yeast Chromosome Transmission Fidelity 4 (CTF4), the protein product of which integrates the MCM2-7 helicase and DNA polymerase α/primase complex to initiate DNA replication. Previous work in fruit flies, Xenopus egg extracts, and human cell lines suggest that Wdhd1 is required for efficient DNA synthesis. However, rigorous in vivo functional studies on Wdhd1 in mammals are unavailable. In the present study, we have successfully generated a Wdhd1 null allele in mice through CRISPR/Cas9-mediated genome editing to investigate the role of Wdhd1 in embryogenesis in vivo. We characterized Wdhd1 expression using quantitative reverse-transcription polymerase chain reaction, and assessed embryonic cell proliferation by histology in both pre- and peri-implantation embryos. While Wdhd1 heterozygous mutant mice were grossly normal and fertile, we observed a reduction in cell proliferation by the gastrulation stage in Wdhd1 homozygous null mutant embryos which severely hampered their growth and viability. These results indicate that Wdhd1 plays a major role in cell proliferation during embryogenesis in mice.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Desenvolvimento Embrionário , Animais , Sistemas CRISPR-Cas , Técnicas de Cultura de Células , Linhagem Celular , Proliferação de Células , Fertilidade , Gastrulação , Edição de Genes , Mutação com Perda de Função , Camundongos
16.
Transplantation ; 105(4): 785-795, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32976366

RESUMO

BACKGROUND: Hepatocyte transplantation has been extensively investigated as an alternative to orthotopic liver transplantation. However, its application in routine clinical practice has been restricted because of low initial engraftment and subsequent repopulation. METHODS: Using mice as a model, we have developed a minimally invasive and nontoxic preconditioning strategy based on preadministration of antibodies against hepsin to increase donor hepatocyte retention and engraftment rate. RESULTS: Liver sinusoid diameters decreased significantly with antihepsin pretreatment, and graft cell numbers increased nearly 2-fold in the recipients' liver parenchyma for 20 days after hepatocyte transplantation. Postoperative complications such as hepatic ischemia injury or apparent immune cell accumulation were not observed in recipients. In a hemophilia B mouse model, antihepsin preconditioning enhanced the expression and clotting activity of coagulation factor IX (FIX) to nearly 2-fold that of immunoglobulin G-treated controls and maintained higher plasma FIX clotting activity relative to the prophylactic range for 50 days after hepatocyte transplantation. Antihepsin pretreatment combined with adeno-associated virus-transduced donor hepatocytes expressing human FIX-Triple, a hyperfunctional FIX variant, resulted in plasma FIX levels similar to those associated with mild hemophilia, which protected hemophilia B mice from major bleeding episodes for 50 days after transplantation. Furthermore, antihepsin pretreatment and repeated transplantation resulted in extending the therapeutic period by 30 days relative to the immunoglobulin G control. CONCLUSIONS: Thus, this antihepsin strategy improved the therapeutic effect of hepatocyte transplantation in mice with tremendous safety and minimal invasion. Taken together, we suggest that preconditioning with antihepsin may have clinical applications for liver cell therapy.


Assuntos
Anticorpos Neutralizantes/farmacologia , Sobrevivência de Enxerto , Hemofilia A/cirurgia , Hepatócitos/transplante , Transplante de Fígado , Serina Endopeptidases/imunologia , Condicionamento Pré-Transplante , Animais , Coagulação Sanguínea , Sobrevivência Celular , Modelos Animais de Doenças , Fator IX/genética , Fator IX/metabolismo , Hemofilia A/sangue , Hemofilia A/genética , Hemofilia A/imunologia , Hepatócitos/imunologia , Hepatócitos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo
17.
Stem Cell Res Ther ; 12(1): 508, 2021 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-34551822

RESUMO

BACKGROUND: The c.G6055A (p.G2019S) mutation in leucine-rich repeat kinase 2 (LRRK2) is the most prevalent genetic cause of Parkinson's disease (PD). CRISPR/Cas9-mediated genome editing by homology-directed repair (HDR) has been applied to correct the mutation but may create small insertions and deletions (indels) due to double-strand DNA breaks. Adenine base editors (ABEs) could convert targeted A·T to G·C in genomic DNA without double-strand breaks. However, the correction efficiency of ABE in LRRK2 c.G6055A (p.G2019S) mutation remains unknown yet. This study aimed to compare the mutation correction efficiencies and off-target effects between HDR and ABEs in induced pluripotent stem cells (iPSCs) carrying LRRK2 c.G6055A (p.G2019S) mutation. METHODS: A set of mutation-corrected isogenic lines by editing the LRRK2 c.G6055A (p.G2019S) mutation in a PD patient-derived iPSC line using HDR or ABE were established. The mutation correction efficacies, off-target effects, and indels between HDR and ABE were compared. Comparative transcriptomic and proteomic analyses between the LRRK2 p.G2019S iPSCs and isogenic control cells were performed to identify novel molecular targets involved in LRRK2-parkinsonism pathways. RESULTS: ABE had a higher correction rate (13/53 clones, 24.5%) than HDR (3/47 clones, 6.4%). Twenty-seven HDR clones (57.4%), but no ABE clones, had deletions, though 14 ABE clones (26.4%) had off-target mutations. The corrected isogenic iPSC-derived dopaminergic neurons exhibited reduced LRRK2 kinase activity, decreased phospho-α-synuclein expression, and mitigated neurite shrinkage and apoptosis. Comparative transcriptomic and proteomic analysis identified different gene expression patterns in energy metabolism, protein degradation, and peroxisome proliferator-activated receptor pathways between the mutant and isogenic control cells. CONCLUSIONS: The results of this study envision that ABE could directly correct the pathogenic mutation in iPSCs for reversing disease-related phenotypes in neuropathology and exploring novel pathophysiological targets in PD.


Assuntos
Células-Tronco Pluripotentes Induzidas , Doença de Parkinson , Neurônios Dopaminérgicos , Edição de Genes , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Mutação , Doença de Parkinson/genética , Doença de Parkinson/terapia , Fenótipo , Proteômica
18.
Dev Biol ; 331(2): 176-88, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19414008

RESUMO

Using a Cre-mediated conditional deletion approach, we have dissected the function of Twist1 in the morphogenesis of the craniofacial skeleton. Loss of Twist1 in neural crest cells and their derivatives impairs skeletogenic differentiation and leads to the loss of bones of the snout, upper face and skull vault. While no anatomically recognizable maxilla is formed, a malformed mandible is present. Since Twist1 is expressed in the tissues of the maxillary eminence and the mandibular arch, this finding suggests that the requirement for Twist1 is not the same in all neural crest derivatives. The effect of the loss of Twist1 function is not restricted to neural crest-derived bones, since the predominantly mesoderm-derived parietal and interparietal bones are also affected, presumably as a consequence of lost interactions with neural crest-derived tissues. In contrast, the formation of other mesodermal skeletal derivatives such as the occipital bones and most of the chondrocranium are not affected by the loss of Twist1 in the neural crest cells.


Assuntos
Morfogênese/fisiologia , Crista Neural/embriologia , Proteínas Nucleares/fisiologia , Crânio/embriologia , Proteína 1 Relacionada a Twist/fisiologia , Animais , Região Branquial/citologia , Região Branquial/embriologia , Região Branquial/fisiologia , Osso Frontal/embriologia , Osso Frontal/metabolismo , Arcada Osseodentária/embriologia , Arcada Osseodentária/metabolismo , Camundongos , Camundongos Mutantes , Osso Nasal/embriologia , Osso Nasal/metabolismo , Crista Neural/citologia , Crista Neural/fisiologia , Crânio/citologia , Crânio/fisiologia
20.
PLoS One ; 14(7): e0219221, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31260508

RESUMO

Cul4b-null (Cul4bΔ/Y) mice undergo growth arrest and degeneration during the early embryonic stages and die at E9.5. The pathogenic causes of this lethality remain incompletely characterized. However, it has been hypothesized that the loss of Cul4b function in extraembryonic tissues plays a key role. In this study, we investigated possible causes of death for Cul4b-null embryos, particularly in regard to the role of embryonic Cul4b. First, we show that the loss of embryonic Cul4b affects the growth of the inner cell mass in vitro and delays epiblast development during the gastrulation period at E6.5~E7.5 in vivo, as highlighted by the absence of the epiblastic transcription factor Brachyury from E6.5~E7.5. Additionally, at E7.5, strong and laterally expanded expression of Eomes and Fgf8 signaling was detected. Sectioning of these embryos showed disorganized primitive streak layer cells. Second, we observed that Mash2-expressing cells were present in the extraembryonic tissues of Cul4b-deficient embryos at E6.5 but were absent at E7.5. In addition, the loss of Cul4b resulted in decreased expression of cyclin proteins, which are required for the cell cycle transition from G1 to S. Taken together, these observations suggest that the embryonic expression of Cul4b is important for epiblast growth during E6.5~E7.5, and the loss of Cul4b results in either delayed growth of the epiblast or defective localization of primitive streak layer cells. As a result, the signaling activity mediated by the epiblast for subsequent ectoplacental cone development is affected, with the potential to induce growth retardation and lethality in Cul4bΔ/Y embryos.


Assuntos
Proteínas Culina/fisiologia , Gastrulação/fisiologia , Camadas Germinativas/embriologia , Linha Primitiva/embriologia , Animais , Massa Celular Interna do Blastocisto/metabolismo , Embrião de Mamíferos , Feminino , Proteínas Fetais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Heterozigoto , Masculino , Camundongos , Camundongos Knockout , Modelos Animais , Proteínas com Domínio T/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA