Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Food Chem ; 438: 137920, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38000156

RESUMO

The digestion products of Penaeus vannamei still had sensitizing and eliciting capacity; however, the underlying mechanism has not been identified. This study analyzed the structural changes of shrimp proteins during digestion, predicted the linearmimotopepeptides and first validated the allergenicity of immunodominantepitopes with binding ability. The results showed that the shrimp proteins were gradually degraded into small peptides during digestion, which might lead to the destruction of linear epitopes. However, these peptides carried IgE epitopes that still trigger allergic reactions. Eighteen digestion-resistant epitopes were predicted by multiple immunoinformatics tools and digestomics. Five epitopes contained more critical amino acids and had strong molecular docking (P1: DSGVGIYAPDAEA, P2: EGELKGTYYPLTGM, P3: GRQGDPHGKFDLPPGV, P4: IFAWPHKDNNGIE, P5: KSTESSVTVPDVPSIHD), and these epitopes were identified as novel IgE binding immunodominantepitopes in Penaeus vannamei. These findings provide novel insight into allergenic epitopes, which might serve as key targets for reducing the allergenicity in shrimp.


Assuntos
Penaeidae , Animais , Sequência de Aminoácidos , Epitopos Imunodominantes , Alérgenos/química , Simulação de Acoplamento Molecular , Imunoglobulina E , Peptídeos , Epitopos/química , Digestão , Tropomiosina/química
3.
Nat Genet ; 51(4): 728-738, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30778223

RESUMO

Characterizing the stem cells responsible for lung repair and regeneration is important for the treatment of pulmonary diseases. Recently, a unique cell population located at the bronchioalveolar-duct junctions has been proposed to comprise endogenous stem cells for lung regeneration. However, the role of bronchioalveolar stem cells (BASCs) in vivo remains debated, and the contribution of such cells to lung regeneration is not known. Here we generated a genetic lineage-tracing system that uses dual recombinases (Cre and Dre) to specifically track BASCs in vivo. Fate-mapping and clonal analysis showed that BASCs became activated and responded distinctly to different lung injuries, and differentiated into multiple cell lineages including club cells, ciliated cells, and alveolar type 1 and type 2 cells for lung regeneration. This study provides in vivo genetic evidence that BASCs are bona fide lung epithelial stem cells with deployment of multipotency and self-renewal during lung repair and regeneration.


Assuntos
Bronquíolos/fisiologia , Líquido da Lavagem Broncoalveolar/citologia , Pulmão/fisiologia , Células-Tronco Multipotentes/fisiologia , Regeneração/genética , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Células Cultivadas , Células Epiteliais/fisiologia , Genótipo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
4.
Cell Rep ; 25(5): 1241-1254.e5, 2018 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-30380415

RESUMO

Identification of cellular surface markers that distinguish tumorous from normal vasculature is important for the development of tumor vessel-targeted therapy. Here, we show that Apj, a G protein-coupled receptor, is highly enriched in tumor endothelial cells but absent from most endothelial cells of adult tissues in homeostasis. By genetic targeting using Apj-CreER and Apj-DTRGFP-Luciferase, we demonstrated that hypoxia-VEGF signaling drives expansion of Apj+ tumor vessels and that targeting of these vessels, genetically and pharmacologically, remarkably inhibits tumor angiogenesis and restricts tumor growth. These in vivo findings implicate Apj+ vessels as a key driver of pathological angiogenesis and identify Apj+ endothelial cells as an important therapeutic target for the anti-angiogenic treatment of tumors.


Assuntos
Receptores de Apelina/metabolismo , Vasos Sanguíneos/patologia , Terapia de Alvo Molecular , Neoplasias/patologia , Envelhecimento/metabolismo , Animais , Vasos Sanguíneos/embriologia , Vasos Sanguíneos/metabolismo , Hipóxia Celular , Proliferação de Células , Células Endoteliais/metabolismo , Feminino , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Masculino , Camundongos Endogâmicos C57BL , Neoplasias/irrigação sanguínea , Neovascularização Patológica , Transdução de Sinais , Hipóxia Tumoral , Fator A de Crescimento do Endotélio Vascular/metabolismo
5.
Circ Res ; 123(1): 86-99, 2018 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-29764841

RESUMO

RATIONALE: Organs of the body require vascular networks to supply oxygen and nutrients and maintain physiological function. The blood vessels of different organs are structurally and functionally heterogeneous in nature. To more precisely dissect their distinct in vivo function in individual organs, without potential interference from off-site targets, it is necessary to genetically target them in an organ-specific manner. OBJECTIVE: The objective of this study was to generate a genetic system that targets vascular endothelial cells in an organ- or tissue-specific manner and to exemplify the potential application of intersectional genetics for precise, target-specific gene manipulation in vivo. METHODS AND RESULTS: We took advantage of 2 orthogonal recombination systems, Dre-rox and Cre-loxP, to create a genetic targeting system based on intersectional genetics. Using this approach, Cre activity was only detectable in cells that had expressed both Dre and Cre. Applying this new system, we generated a coronary endothelial cell-specific Cre (CoEC-Cre) and a brain endothelial cell-specific Cre (BEC-Cre). Through lineage tracing, gene knockout and overexpression experiments, we demonstrated that CoEC-Cre and BEC-Cre efficiently and specifically target blood vessels in the heart and brain, respectively. By deletion of vascular endothelial growth factor receptor 2 using BEC-Cre, we showed that vascular endothelial growth factor signaling regulates angiogenesis in the central nervous system and also controls the integrity of the blood-brain barrier. CONCLUSIONS: We provide 2 examples to illustrate the use of intersectional genetics for more precise gene targeting in vivo, namely manipulation of genes in blood vessels of the heart and brain. More broadly, this system provides a valuable strategy for tissue-specific gene manipulation that can be widely applied to other fields of biomedical research.


Assuntos
Vasos Sanguíneos , Encéfalo/irrigação sanguínea , Vasos Coronários , Marcação de Genes/métodos , Animais , Barreira Hematoencefálica , Hipóxia Celular , Células Endoteliais , Técnicas de Inativação de Genes , Hibridização In Situ/métodos , Camundongos , Neovascularização Fisiológica , Especificidade de Órgãos , Receptores de Fatores de Crescimento do Endotélio Vascular/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia
6.
Biomed Pharmacother ; 104: 223-228, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29775889

RESUMO

Emerging evidence has indicated that long non-coding RNAs (lncRNAs) play critical roles in tumor development and progression. Recent studies reported that lncRNA MIR22HG could play important roles in hepatocellular carcinoma and lung cancer progression. However, the expression and underlying mechanism of MIR22HG in endometrial cancer (EC) remain unclear. In the present study, qRT-PCR showed that MIR22HG expression was significantly downregulated in EC tissues. In vitro function assays showed that increased MIR22HG expression significantly inhibited EC cells proliferation, induced EC cells apoptosis, and arrested EC cells in G0/G1 phase. Furthermore, miR-141-3p was identified and confirmed to be the target of MIR22HG. Subsequently, DAPK1 was confirmed to be regulated by MIR22HG and miR-141-3p, and could play a positive role in inhibiting EC cells proliferation. Collectively, these data demonstrated that lncRNA MIR22HG could act as a tumor suppressor and inhibited EC cells proliferation through regulating miR-141-3p/DAPK1 axis, which provides a new therapeutic target for EC treatment.


Assuntos
Carcinoma/genética , Proliferação de Células/genética , Proteínas Quinases Associadas com Morte Celular/genética , Neoplasias do Endométrio/genética , MicroRNAs/genética , Apoptose/genética , Carcinoma/patologia , Pontos de Checagem do Ciclo Celular/genética , Linhagem Celular Tumoral , Regulação para Baixo/genética , Neoplasias do Endométrio/patologia , Feminino , Fase G1/genética , Regulação Neoplásica da Expressão Gênica/genética , Genes Supressores de Tumor/fisiologia , Humanos , Fase de Repouso do Ciclo Celular/genética
7.
Nat Commun ; 8(1): 87, 2017 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-28729659

RESUMO

Noncompaction cardiomyopathy is characterized by the presence of extensive trabeculations, which could lead to heart failure and malignant arrhythmias. How trabeculations resolve to form compact myocardium is poorly understood. Elucidation of this process is critical to understanding the pathophysiology of noncompaction disease. Here we use genetic lineage tracing to mark the Nppa+ or Hey2+ cardiomyocytes as trabecular and compact components of the ventricular wall. We find that Nppa+ and Hey2+ cardiomyocytes, respectively, from the endocardial and epicardial zones of the ventricular wall postnatally. Interposed between these two postnatal layers is a hybrid zone, which is composed of cells derived from both the Nppa+ and Hey2+ populations. Inhibition of the fetal Hey2+ cell contribution to the hybrid zone results in persistence of excessive trabeculations in postnatal heart. Our findings indicate that the expansion of Hey2+ fetal compact component, and its contribution to the hybrid myocardial zone, are essential for normal formation of the ventricular walls.Fetal trabecular muscles in the heart undergo a poorly described morphogenetic process that results into a solidified compact myocardium after birth. Tian et al. show that cardiomyocytes in the fetal compact layer also contribute to this process, forming a hybrid myocardial zone that is composed of cells derived from both trabecular and compact layers.


Assuntos
Cardiomiopatias/embriologia , Ventrículos do Coração/embriologia , Animais , Animais Recém-Nascidos , Fator Natriurético Atrial , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Cardiomiopatias/congênito , Cardiomiopatias/metabolismo , Linhagem da Célula , Coração/embriologia , Coração/crescimento & desenvolvimento , Cardiopatias Congênitas/embriologia , Cardiopatias Congênitas/metabolismo , Ventrículos do Coração/crescimento & desenvolvimento , Ventrículos do Coração/metabolismo , Ventrículos do Coração/patologia , Camundongos , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Peptídeo Natriurético Tipo C/metabolismo , Organogênese , Precursores de Proteínas/metabolismo , Proteínas Repressoras/metabolismo
8.
J Biol Chem ; 292(21): 8594-8604, 2017 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-28377509

RESUMO

The liver possesses a remarkable capacity to regenerate after damage. There is a heated debate on the origin of new hepatocytes after injuries in adult liver. Hepatic stem/progenitor cells have been proposed to produce functional hepatocytes after injury. Recent studies have argued against this model and suggested that pre-existing hepatocytes, rather than stem cells, contribute new hepatocytes. This hepatocyte-to-hepatocyte model is mainly based on labeling of hepatocytes with Cre-recombinase delivered by the adeno-associated virus. However, the impact of virus infection on cell fate determination, consistency of infection efficiency, and duration of Cre-virus in hepatocytes remain confounding factors that interfere with the data interpretation. Here, we generated a new genetic tool Alb-DreER to label almost all hepatocytes (>99.5%) and track their contribution to different cell lineages in the liver. By "pulse-and-chase" strategy, we found that pre-existing hepatocytes labeled by Alb-DreER contribute to almost all hepatocytes during normal homeostasis and after liver injury. Virtually all hepatocytes in the injured liver are descendants of pre-existing hepatocytes through self-expansion. We concluded that stem cell differentiation is unlikely to be responsible for the generation of a substantial number of new hepatocytes in adult liver. Our study also provides a new mouse tool for more precise in vivo genetic study of hepatocytes in the field.


Assuntos
Diferenciação Celular , Hepatócitos , Regeneração Hepática , Fígado , Células-Tronco , Animais , Rastreamento de Células/métodos , Hepatócitos/metabolismo , Hepatócitos/patologia , Integrases/biossíntese , Integrases/genética , Fígado/lesões , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Transgênicos , Células-Tronco/metabolismo , Células-Tronco/patologia
9.
Circ Res ; 118(12): 1880-93, 2016 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-27056912

RESUMO

RATIONALE: There is persistent uncertainty regarding the developmental origins of coronary vessels, with 2 principal sources suggested as ventricular endocardium or sinus venosus (SV). These 2 proposed origins implicate fundamentally distinct mechanisms of vessel formation. Resolution of this controversy is critical for deciphering the programs that result in the formation of coronary vessels and has implications for research on therapeutic angiogenesis. OBJECTIVE: To resolve the controversy over the developmental origin of coronary vessels. METHODS AND RESULTS: We first generated nuclear factor of activated T cells (Nfatc1)-Cre and Nfatc1-Dre lineage tracers for endocardium labeling. We found that Nfatc1 recombinases also label a significant portion of SV endothelial cells in addition to endocardium. Therefore, restricted endocardial lineage tracing requires a specific marker that distinguishes endocardium from SV. By single-cell gene expression analysis, we identified a novel endocardial gene natriuretic peptide receptor 3 (Npr3). Npr3 is expressed in the entirety of the endocardium but not in the SV. Genetic lineage tracing based on Npr3-CreER showed that endocardium contributes to a minority of coronary vessels in the free walls of embryonic heart. Intersectional genetic lineage tracing experiments demonstrated that endocardium minimally contributes to coronary endothelium in the embryonic ventricular free walls. CONCLUSIONS: Our study suggested that SV, but not endocardium, is the major origin for coronary endothelium in the embryonic ventricular free walls. This work thus resolves the recent controversy over the developmental origin of coronary endothelium, providing the basis for studying coronary vessel formation and regeneration after injury.


Assuntos
Linhagem da Célula , Vasos Coronários/embriologia , Endocárdio/embriologia , Endotélio Vascular/metabolismo , Ventrículos do Coração/embriologia , Animais , Vasos Coronários/citologia , Vasos Coronários/metabolismo , Endocárdio/citologia , Endocárdio/metabolismo , Endotélio Vascular/citologia , Ventrículos do Coração/citologia , Ventrículos do Coração/metabolismo , Camundongos , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Receptores do Fator Natriurético Atrial/genética , Receptores do Fator Natriurético Atrial/metabolismo
10.
Nat Genet ; 48(5): 537-43, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27019112

RESUMO

The hepatic vasculature is essential for liver development, homeostasis and regeneration, yet the developmental program of hepatic vessel formation and the embryonic origin of the liver vasculature remain unknown. Here we show in mouse that endocardial cells form a primitive vascular plexus surrounding the liver bud and subsequently contribute to a substantial portion of the liver vasculature. Using intersectional genetics, we demonstrate that the endocardium of the sinus venosus is a source for the hepatic plexus. Inhibition of endocardial angiogenesis results in reduced endocardial contribution to the liver vasculature and defects in liver organogenesis. We conclude that a substantial portion of liver vessels derives from the endocardium and shares a common developmental origin with coronary arteries.


Assuntos
Linhagem da Célula , Endocárdio/embriologia , Fígado/embriologia , Animais , Vasos Coronários/embriologia , Endocárdio/citologia , Endocárdio/metabolismo , Átrios do Coração/embriologia , Átrios do Coração/metabolismo , Ventrículos do Coração/embriologia , Ventrículos do Coração/metabolismo , Fígado/irrigação sanguínea , Circulação Hepática , Camundongos , Morfogênese , Fatores de Transcrição NFATC/metabolismo , Neovascularização Fisiológica
11.
Cell Res ; 26(1): 119-30, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26634606

RESUMO

Cardiac cells marked by c-Kit or Kit, dubbed cardiac stem cells (CSCs), are in clinical trials to investigate their ability to stimulate cardiac regeneration and repair. These studies were initially motivated by the purported cardiogenic activity of these cells. Recent lineage tracing studies using Kit promoter to drive expression of the inducible Cre recombinase showed that these CSCs had highly limited cardiogenic activity, inadequate to support efficient cardiac repair. Here we reassess the lineage tracing data by investigating the identity of cells immediately after Cre labeling. Our instant lineage tracing approach identifies Kit-expressing cardiomyocytes, which are labeled immediately after tamoxifen induction. In combination with long-term lineage tracing experiments, these data reveal that the large majority of long-term labeled cardiomyocytes are pre-existing Kit-expressing cardiomyocytes rather than cardiomyocytes formed de novo from CSCs. This study presents a new interpretation for the contribution of Kit(+) cells to cardiomyocytes and shows that Kit genetic lineage tracing over-estimates the cardiogenic activity of Kit(+) CSCs.


Assuntos
Coração/fisiologia , Miócitos Cardíacos/citologia , Proteínas Proto-Oncogênicas c-kit/análise , Regeneração , Células-Tronco/citologia , Animais , Linhagem da Célula , Células Cultivadas , Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/citologia , Miócitos Cardíacos/metabolismo , Proteínas Proto-Oncogênicas c-kit/genética , Células-Tronco/metabolismo
12.
Nat Med ; 21(8): 866-8, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26168292

RESUMO

Unraveling the fate specification of resident stem cells during lung regeneration is of clinical importance. It has been reported that c-kit(+) progenitor cells resident in the human lung regenerate epithelial lineages upon transplantation into injured mouse lung. Here we test the lineage potential of c-kit(+) cells by inducible genetic lineage tracing. We find that c-kit(+) cells do not contribute to lung epithelium during homeostasis and repair, and instead maintain a vascular endothelial cell fate. These findings call attention to the clinical application of c-kit(+) stem cells as lung epithelial progenitors for the treatment of pulmonary disease.


Assuntos
Células Endoteliais/fisiologia , Células Epiteliais/fisiologia , Pulmão/citologia , Proteínas Proto-Oncogênicas c-kit/análise , Animais , Linhagem da Célula , Feminino , Homeostase , Masculino , Camundongos , Células-Tronco/fisiologia
13.
Nat Commun ; 6: 6020, 2015 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-25597280

RESUMO

Under pathophysiological conditions in adults, endothelial cells (ECs) sprout from pre-existing blood vessels to form new ones by a process termed angiogenesis. During embryonic development, Apelin (APLN) is robustly expressed in vascular ECs. In adult mice, however, APLN expression in the vasculature is significantly reduced. Here we show that APLN expression is reactivated in adult ECs after ischaemia insults. In models of both injury ischaemia and tumor angiogenesis, we find that Apln-CreER genetically labels sprouting but not quiescent vasculature. By leveraging this specific activity, we demonstrate that abolishment of the VEGF-VEGFR2 signalling pathway as well as ablation of sprouting ECs diminished tumour vascularization and growth without compromising vascular homeostasis in other organs. Collectively, we show that Apln-CreER distinguishes sprouting vessels from stabilized vessels in multiple pathological settings. The Apln-CreER line described here will greatly aid future mechanistic studies in both vascular developmental biology and adult vascular diseases.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neovascularização Fisiológica/fisiologia , Adipocinas , Animais , Apelina , Linhagem Celular Tumoral , Células Endoteliais/metabolismo , Feminino , Membro Posterior , Peptídeos e Proteínas de Sinalização Intercelular/genética , Isquemia , Masculino , Camundongos , Neovascularização Fisiológica/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA