Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
JHEP Rep ; 4(4): 100446, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35284810

RESUMO

Background & Aims: The truncating mutations in tight junction protein 2 (TJP2) cause progressive cholestasis, liver failure, and hepatocyte carcinogenesis. Due to the lack of effective model systems, there are no targeted medications for the liver pathology with TJP2 deficiency. We leveraged the technologies of patient-specific induced pluripotent stem cells (iPSC) and CRISPR genome-editing, and we aim to establish a disease model which recapitulates phenotypes of patients with TJP2 deficiency. Methods: We differentiated iPSC to hepatocyte-like cells (iHep) on the Transwell membrane in a polarized monolayer. Immunofluorescent staining of polarity markers was detected by a confocal microscope. The epithelial barrier function and bile acid transport of bile canaliculi were quantified between the two chambers of Transwell. The morphology of bile canaliculi was measured in iHep cultured in the Matrigel sandwich system using a fluorescent probe and live-confocal imaging. Results: The iHep differentiated from iPSC with TJP2 mutations exhibited intracellular inclusions of disrupted apical membrane structures, distorted canalicular networks, altered distribution of apical and basolateral markers/transporters. The directional bile acid transport of bile canaliculi was compromised in the mutant hepatocytes, resembling the disease phenotypes observed in the liver of patients. Conclusions: Our iPSC-derived in vitro hepatocyte system revealed canalicular membrane disruption in TJP2 deficient hepatocytes and demonstrated the ability to model cholestatic disease with TJP2 deficiency to serve as a platform for further pathophysiologic study and drug discovery. Lay summary: We investigated a genetic liver disease, progressive familial intrahepatic cholestasis (PFIC), which causes severe liver disease in newborns and infants due to a lack of gene called TJP2. By using cutting-edge stem cell technology and genome editing methods, we established a novel disease modeling system in cell culture experiments. Our experiments demonstrated that the lack of TJP2 induced abnormal cell polarity and disrupted bile acid transport. These findings will lead to the subsequent investigation to further understand disease mechanisms and develop an effective treatment.

2.
J Cell Sci ; 132(21)2019 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-31601613

RESUMO

Timely and precise control of Aurora B kinase, the chromosomal passenger complex (CPC) catalytic subunit, is essential for accurate chromosome segregation and cytokinesis. Post-translational modifications of CPC subunits are directly involved in controlling Aurora B activity. Here, we identified a highly conserved acidic STD-rich motif of INCENP that is phosphorylated during mitosis in vivo and by Plk1 in vitro and is involved in controlling Aurora B activity. By using an INCENP conditional-knockout cell line, we show that impairing the phosphorylation status of this region disrupts chromosome congression and induces cytokinesis failure. In contrast, mimicking constitutive phosphorylation not only rescues cytokinesis but also induces ectopic furrows and contractile ring formation in a Plk1- and ROCK1-dependent manner independent of cell cycle and microtubule status. Our experiments identify the phospho-regulation of the INCENP STD motif as a novel mechanism that is key for chromosome alignment and cytokinesis.This article has an associated First Person interview with the first author of the paper.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/fisiologia , Microtúbulos/metabolismo , Mutação/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Cromossomos/metabolismo , Citocinese/fisiologia , Humanos , Mitose/fisiologia , Quinases Associadas a rho/metabolismo , Quinase 1 Polo-Like
3.
J Cell Sci ; 131(4)2018 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-29361541

RESUMO

The requirement for condensin in chromosome formation in somatic cells remains unclear, as imperfectly condensed chromosomes do form in cells depleted of condensin by conventional methodologies. In order to dissect the roles of condensin at different stages of vertebrate mitosis, we have established a versatile cellular system that combines auxin-mediated rapid degradation with chemical genetics to obtain near-synchronous mitotic entry of chicken DT40 cells in the presence and absence of condensin. We analyzed the outcome by live- and fixed-cell microscopy methods, including serial block face scanning electron microscopy with digital reconstruction. Following rapid depletion of condensin, chromosomal defects were much more obvious than those seen after a slow depletion of condensin. The total mitotic chromatin volume was similar to that in control cells, but a single mass of mitotic chromosomes was clustered at one side of a bent mitotic spindle. Cultures arrest at prometaphase, eventually exiting mitosis without segregating chromosomes. Experiments where the auxin concentration was titrated showed that different condensin levels are required for anaphase chromosome segregation and formation of a normal chromosome architecture.This article has an associated First Person interview with the first author of the paper.


Assuntos
Adenosina Trifosfatases/genética , Cromatina/ultraestrutura , Cromossomos/ultraestrutura , Proteínas de Ligação a DNA/genética , Mitose/genética , Complexos Multiproteicos/genética , Adenosina Trifosfatases/metabolismo , Animais , Galinhas , Cromatina/genética , Cromatina/metabolismo , Aberrações Cromossômicas , Segregação de Cromossomos/genética , Cromossomos/genética , Cromossomos/metabolismo , Proteínas de Ligação a DNA/metabolismo , Ácidos Indolacéticos/farmacologia , Microscopia Eletrônica de Varredura , Complexos Multiproteicos/metabolismo , Proteólise/efeitos dos fármacos
5.
J Autoimmun ; 77: 76-88, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27894837

RESUMO

It is widely accepted that central and effector memory CD4+ T cells originate from naïve T cells after they have encountered their cognate antigen in the setting of appropriate co-stimulation. However, if this were true the diversity of T cell receptor (TCR) sequences within the naïve T cell compartment should be far greater than that of the memory T cell compartment, which is not supported by TCR sequencing data. Here we demonstrate that aged mice with far fewer naïve T cells, respond to the model antigen, hen eggwhite lysozyme (HEL), by utilizing the same TCR sequence as their younger counterparts. CD4+ T cell repertoire analysis of highly purified T cell populations from naive animals revealed that the HEL-specific clones displayed effector and central "memory" cell surface phenotypes even prior to having encountered their cognate antigen. Furthermore, HEL-inexperienced CD4+ T cells were found to reside within the naïve, regulatory, central memory, and effector memory T cell populations at similar frequencies and the majority of the CD4+ T cells within the regulatory and memory populations were unexpanded. These findings support a new paradigm for CD4+ T cell maturation in which a specific clone can undergo a differentiation process to exhibit a "memory" or regulatory phenotype without having undergone a clonal expansion event. It also demonstrates that a foreign-specific T cell is just as likely to reside within the regulatory T cell compartment as it would the naïve compartment, arguing against the specificity of the regulatory T cell compartment being skewed towards self-reactive T cell clones. Finally, we demonstrate that the same set of foreign and autoreactive CD4+ T cell clones are repetitively generated throughout adulthood. The latter observation argues against T cell-depleting strategies or autologous stem cell transplantation as therapies for autoimmunity-as the immune system has the ability to regenerate pathogenic clones.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Memória Imunológica , Subpopulações de Linfócitos T/imunologia , Fatores Etários , Animais , Antígenos/imunologia , Autoimunidade , Linfócitos T CD4-Positivos/metabolismo , Galinhas , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Proteínas do Ovo/imunologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/terapia , Feminino , Transplante de Células-Tronco Hematopoéticas , Imunofenotipagem , Contagem de Linfócitos , Depleção Linfocítica , Camundongos , Fenótipo , Especificidade do Receptor de Antígeno de Linfócitos T/genética , Especificidade do Receptor de Antígeno de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
6.
Mol Cell Proteomics ; 15(8): 2802-18, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27231315

RESUMO

Packaging of DNA into condensed chromosomes during mitosis is essential for the faithful segregation of the genome into daughter nuclei. Although the structure and composition of mitotic chromosomes have been studied for over 30 years, these aspects are yet to be fully elucidated. Here, we used stable isotope labeling with amino acids in cell culture to compare the proteomes of mitotic chromosomes isolated from cell lines harboring conditional knockouts of members of the condensin (SMC2, CAP-H, CAP-D3), cohesin (Scc1/Rad21), and SMC5/6 (SMC5) complexes. Our analysis revealed that these complexes associate with chromosomes independently of each other, with the SMC5/6 complex showing no significant dependence on any other chromosomal proteins during mitosis. To identify subtle relationships between chromosomal proteins, we employed a nano Random Forest (nanoRF) approach to detect protein complexes and the relationships between them. Our nanoRF results suggested that as few as 113 of 5058 detected chromosomal proteins are functionally linked to chromosome structure and segregation. Furthermore, nanoRF data revealed 23 proteins that were not previously suspected to have functional interactions with complexes playing important roles in mitosis. Subsequent small-interfering-RNA-based validation and localization tracking by green fluorescent protein-tagging highlighted novel candidates that might play significant roles in mitotic progression.


Assuntos
Proteínas de Ciclo Celular/genética , Cromossomos/genética , Mitose , Proteômica/métodos , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Animais , Técnicas de Cultura de Células , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Galinhas , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Cromossomos/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Marcação por Isótopo , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Coesinas
7.
Drug Des Devel Ther ; 10: 43-52, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26730180

RESUMO

The treatment of melanoma has improved markedly over the last several years with the advent of more targeted therapies. Unfortunately, complex compensation mechanisms, such as those of the mitogen-activated protein kinase (MAPK) pathway, have limited the clinical benefit of these treatments. Recently, a better understanding of melanoma resistance mechanisms has given way to intelligently designed multidrug regimes. Herein, we review the extensive pathways of BRAF inhibitor (vemurafenib and dabrafenib) resistance. We also review the advantages of dual therapy, including the addition of an MEK inhibitor (cobimetinib or trametinib), which has proven to increase progression-free survival when compared to BRAF inhibitor monotherapy. Finally, this review touches on future treatment strategies that are being developed for advanced melanoma, including the possibility of triple therapy with immune checkpoint inhibitors and the work on optimizing sequential therapy.


Assuntos
Melanoma/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Neoplasias Cutâneas/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Humanos , Melanoma/patologia , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Terapia de Alvo Molecular , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia , Neoplasias Cutâneas/patologia
8.
Mol Cell Biol ; 35(20): 3566-78, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26240282

RESUMO

Aurora kinase B, one of the three members of the mammalian Aurora kinase family, is the catalytic component of the chromosomal passenger complex, an essential regulator of chromosome segregation in mitosis. Aurora B is overexpressed in human tumors although whether this kinase may function as an oncogene in vivo is not established. Here, we report a new mouse model in which expression of the endogenous Aurkb locus can be induced in vitro and in vivo. Overexpression of Aurora B in cultured cells induces defective chromosome segregation and aneuploidy. Long-term overexpression of Aurora B in vivo results in aneuploidy and the development of multiple spontaneous tumors in adult mice, including a high incidence of lymphomas. Overexpression of Aurora B also results in a reduced DNA damage response and decreased levels of the p53 target p21(Cip1) in vitro and in vivo, in line with an inverse correlation between Aurora B and p21(Cip1) expression in human leukemias. Thus, overexpression of Aurora B may contribute to tumor formation not only by inducing chromosomal instability but also by suppressing the function of the cell cycle inhibitor p21(Cip1).


Assuntos
Aneuploidia , Aurora Quinase B/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/genética , Animais , Carcinogênese/genética , Carcinogênese/metabolismo , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Fibroblastos/metabolismo , Expressão Gênica , Inativação Gênica , Camundongos Endogâmicos C57BL , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética
9.
J Biol Chem ; 289(45): 31617-23, 2014 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-25248749

RESUMO

Caspase-activated DNase (CAD) is a major apoptotic nuclease, responsible for DNA fragmentation and chromatin condensation during apoptosis. CAD is normally activated in apoptosis as a result of caspase cleavage of its inhibitory chaperone ICAD. Other aspects of CAD regulation are poorly understood. In particular, it has been unclear whether direct CAD activation in non-apoptotic living cells can trigger cell death. Taking advantage of the auxin-inducible degron (AID) system, we have developed a suicide system with which ICAD is rapidly degraded in living cells in response to the plant hormone auxin. Our studies demonstrate that rapid ICAD depletion is sufficient to activate CAD and induce cell death in DT40 and yeast cells. In the vertebrate cells, ectopic CAD activation triggered caspase activation and subsequent hallmarks of caspase-dependent apoptotic changes, including phosphatidylserine exposure and nuclear fragmentation. These observations not only suggest that CAD activation drives apoptosis through a positive feedback loop, but also identify a unique suicide system that can be used for controlling gene-modified organisms.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Caspases/metabolismo , Desoxirribonucleases/metabolismo , Regulação Enzimológica da Expressão Gênica , Ácidos Indolacéticos/metabolismo , Animais , Anexina A5/metabolismo , Apoptose , Morte Celular , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Galinhas , Fragmentação do DNA , Ativação Enzimática , Técnica Indireta de Fluorescência para Anticorpo , Técnicas de Inativação de Genes , Fosfatidilserinas/metabolismo , Saccharomyces cerevisiae/enzimologia
10.
J Cell Biol ; 199(5): 755-70, 2012 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-23166350

RESUMO

Mitotic chromosome formation involves a relatively minor condensation of the chromatin volume coupled with a dramatic reorganization into the characteristic "X" shape. Here we report results of a detailed morphological analysis, which revealed that chromokinesin KIF4 cooperated in a parallel pathway with condensin complexes to promote the lateral compaction of chromatid arms. In this analysis, KIF4 and condensin were mutually dependent for their dynamic localization on the chromatid axes. Depletion of either caused sister chromatids to expand and compromised the "intrinsic structure" of the chromosomes (defined in an in vitro assay), with loss of condensin showing stronger effects. Simultaneous depletion of KIF4 and condensin caused complete loss of chromosome morphology. In these experiments, topoisomerase IIα contributed to shaping mitotic chromosomes by promoting the shortening of the chromatid axes and apparently acting in opposition to the actions of KIF4 and condensins. These three proteins are major determinants in shaping the characteristic mitotic chromosome morphology.


Assuntos
Adenosina Trifosfatases/metabolismo , Antígenos de Neoplasias/metabolismo , Cromossomos/metabolismo , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/metabolismo , Cinesinas/metabolismo , Mitose , Complexos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo , Adenosina Trifosfatases/genética , Animais , Galinhas , Cromátides/metabolismo , Proteínas de Ligação a DNA/genética , Cinesinas/genética , Complexos Multiproteicos/genética , Mutação , Proteínas Nucleares/genética , Células Tumorais Cultivadas
11.
Biochem Biophys Res Commun ; 420(1): 114-8, 2012 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-22405769

RESUMO

Tumor necrosis factor-α (TNF-α) is a potent proinflammatory cytokine produced predominantly by activated macrophages, and plays a central role in the protective immunity against intracellular pathogens and the pathogenesis of autoimmune and inflammatory diseases. While both the soluble and transmembrane forms of TNF-α (sTNF-α and tmTNF-α) are biologically functional, the latter but not the former acts as a receptor besides as a ligand, and transmit a retrograde signal in a cell-to-cell contact manner. The production of TNF-α by macrophages under Th2-type (allergic) inflammatory conditions has been ill defined, compared to that under Th1-type inflammatory conditions. Here we examined the effect of representative Th2-inducing cytokines IL-4 and IL-33 on the TNF-α expression in macrophages. IL-4 induced the production of neither sTNF-α nor tmTNF-α while IL-33 promoted the production of sTNF-α with no detectable tmTNF-α. Notably, the combination of IL-4 and IL-33 elicited the tmTNF-α expression on macrophages, in addition to the enhanced production of sTNF-α and IL-6. The IL-4/IL-33-elicited tmTNF-α expression was not observed in IL-6-deficient macrophages, suggesting the involvement of macrophage-derived IL-6 in the tmTNF-α expression. Indeed, the stimulation of macrophages with the combination of IL-4 and IL-6 induced the tmTNF-α expression with no detectable production of sTNF-α. Thus, IL-4 and IL-33 synergistically elicit the tmTNF-α expression on macrophages through the autocrine action of IL-6.


Assuntos
Comunicação Autócrina , Interleucina-4/fisiologia , Interleucina-6/metabolismo , Interleucinas/fisiologia , Macrófagos/imunologia , Células Th2/imunologia , Fator de Necrose Tumoral alfa/biossíntese , Animais , Membrana Celular/imunologia , Interleucina-33 , Interleucina-4/farmacologia , Interleucina-6/genética , Interleucinas/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
12.
Exp Anim ; 54(5): 421-8, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16365519

RESUMO

Hematopoietic stem cells in bone marrow can be mobilized into peripheral blood by cytokine administration. Cytokine-mobilized peripheral blood stem cells are of great use in clinical applications. We previously established a modified procedure for the collection of cytokine-mobilized peripheral blood cells from rhesus monkeys (Macaca mulata) using a commercially available apparatus originally developed for human subjects. In this study, we examined the efficacy and safety of this method with even smaller macaques, cynomolgus monkeys (Macaca fascicularis), which are equivalent to human newborns in body weight (mean = 3.3 kg). Using the manufacturer's unmodified protocol (n=6), one monkey died of cardiac failure and three developed severe anemia. In contrast, using our modified procedure (n=6), no such complication was observed in any animal. In addition, the harvested nuclear cell, mononuclear cell and CD34(+) cell counts were significantly higher with the modified method. The modified method should allow safe and efficient collection of cytokine-mobilized peripheral blood cells from non-human primates as small as human newborns in a non-invasive manner.


Assuntos
Células da Medula Óssea/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos/farmacologia , Mobilização de Células-Tronco Hematopoéticas/métodos , Leucaférese/métodos , Macaca fascicularis/fisiologia , Fator de Células-Tronco/farmacologia , Anemia/induzido quimicamente , Animais , Contagem de Células Sanguíneas , Feminino , Células-Tronco Hematopoéticas , Leucaférese/instrumentação , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA