Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cancers (Basel) ; 14(19)2022 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-36230482

RESUMO

Modeling human genetic diseases and cancer in lab animals has been greatly aided by the emergence of genetic engineering tools such as TALENs and CRISPR/Cas9. We have previously demonstrated the ease with which genetically engineered Xenopus models (GEXM) can be generated via injection of early embryos with Cas9 recombinant protein loaded with sgRNAs targeting single or multiple tumor suppressor genes. What has been lacking so far is the possibility to propagate and characterize the induced cancers via transplantation. Here, we describe the generation of a rag2 knockout line in Xenopus tropicalis that is deficient in functional T and B cells. This line was validated by means of allografting experiments with primary tp53-/- and apc+/-/tp53-/- donor tumors. In addition, we optimized available protocols for the sub-lethal irradiation of wild-type X. tropicalis froglets. Irradiated animals also allowed the stable, albeit transient, engraftment of transplanted X. tropicalis tumor cells. The novel rag2-/- line and the irradiated wild-type froglets will further expand the experimental toolbox in the diploid amphibian X. tropicalis and help to establish it as a versatile and relevant model for exploring human cancer.

2.
J Allergy Clin Immunol ; 149(4): 1413-1427.e2, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34506849

RESUMO

BACKGROUND: The most common endotype of asthma is type 2-high asthma, which is sometimes driven by adaptive allergen-specific TH2 lymphocytes that react to allergens presented by dendritic cells (DCs), or sometimes by an innate immune response dominated by type 2 innate lymphocytes (ILC2s). Understanding the underlying pathophysiology of asthma is essential to improve patient-tailored therapy. The STE20 kinase thousand-and-one kinase 3 (TAOK3) controls key features in the biology of DCs and lymphocytes, but to our knowledge, its potential usefulness as a target for asthma therapy has not yet been addressed. OBJECTIVE: We examined if and how loss of Taok3 affects the development of house dust mite (HDM)-driven allergic asthma in an in vivo mouse model. METHODS: Wild-type Taok3+/+ and gene-deficient Taok3-/- mice were sensitized and challenged with HDM, and bronchoalveolar lavage fluid composition, mediastinal lymph node cytokine production, lung histology, and bronchial hyperreactivity measured. Conditional Taok3fl/fl mice were crossed to tissue- and cell-specific specific deletor Cre mice to understand how Taok3 acted on asthma susceptibility. Kinase-dead (KD) Taok3KD mice were generated to probe for the druggability of this pathway. Activation of HDM-specific T cells was measured in adoptively transferred HDM-specific T-cell receptor-transgenic CD4+ T cells. ILC2 biology was assessed by in vivo and in vitro IL-33 stimulation assays in Taok3-/- and Taok3+/+, Taok3KD, and Red5-Cre Taok3fl/fl mice. RESULTS: Taok3-/- mice failed to mount salient features of asthma, including airway eosinophilia, TH2 cytokine production, IgE secretion, airway goblet cell metaplasia, and bronchial hyperreactivity compared to controls. This was due to intrinsic loss of Taok3 in hematopoietic and not epithelial cells. Loss of Taok3 resulted in hampered HDM-induced lung DC migration to the draining lymph nodes and defective priming of HDM-specific TH2 cells. Strikingly, HDM and IL-33-induced ILC2 proliferation and function were also severely affected in Taok3-deficient and Taok3KD mice. CONCLUSIONS: Absence of Taok3 or loss of its kinase activity protects from HDM-driven allergic asthma as a result of defects in both adaptive DC-mediated TH2 activation and innate ILC2 function. This identifies Taok3 as an interesting drug target, justifying further testing as a new treatment for type 2-high asthma.


Assuntos
Asma , Hiper-Reatividade Brônquica , Alérgenos , Animais , Hiper-Reatividade Brônquica/patologia , Citocinas , Dermatophagoides pteronyssinus , Modelos Animais de Doenças , Humanos , Imunidade Inata , Interleucina-33 , Pulmão , Linfócitos , Camundongos , Proteínas Serina-Treonina Quinases , Pyroglyphidae , Células Th2
3.
Cardiovasc Res ; 116(2): 416-428, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30924866

RESUMO

AIMS: Therapies to prevent vein graft disease, a major problem in cardiovascular and lower extremity bypass surgeries, are currently lacking. Short-term preoperative protein restriction holds promise as an effective preconditioning method against surgical stress in rodent models, but whether it can improve vein graft patency after bypass surgery is undetermined. Here, we hypothesized that short-term protein restriction would limit vein graft disease via up-regulation of cystathionine γ-lyase and increased endogenous production of the cytoprotective gaseous signalling molecule hydrogen sulfide. METHODS AND RESULTS: Low-density lipoprotein receptor knockout mice were preconditioned for 1 week on a high-fat high-cholesterol (HFHC) diet with or without protein prior to left common carotid interposition vein graft surgery with caval veins from donor mice on corresponding diets. Both groups were returned to a complete HFHC diet post-operatively, and vein grafts analysed 4 or 28 days later. A novel global transgenic cystathionine γ-lyase overexpressing mouse model was also employed to study effects of genetic overexpression on graft patency. Protein restriction decreased vein graft intimal/media+adventitia area and thickness ratios and intimal smooth muscle cell infiltration 28 days post-operatively, and neutrophil transmigration 4 days post-operatively. Protein restriction increased cystathionine γ-lyase protein expression in aortic and caval vein endothelial cells (ECs) and frequency of lung EC producing hydrogen sulfide. The cystathionine γ-lyase inhibitor propargylglycine abrogated protein restriction-mediated protection from graft failure and the increase in hydrogen sulfide-producing ECs, while cystathionine γ-lyase transgenic mice displayed increased hydrogen sulfide production capacity and were protected from vein graft disease independent of diet. CONCLUSION: One week of protein restriction attenuates vein graft disease via increased cystathionine γ-lyase expression and hydrogen sulfide production, and decreased early inflammation. Dietary or pharmacological interventions to increase cystathionine γ-lyase or hydrogen sulfide may thus serve as new and practical strategies to improve vein graft durability.


Assuntos
Cistationina gama-Liase/biossíntese , Dieta com Restrição de Proteínas , Oclusão de Enxerto Vascular/prevenção & controle , Veia Cava Inferior/transplante , Animais , Artéria Carótida Primitiva/cirurgia , Colesterol na Dieta , Cistationina gama-Liase/genética , Dieta Hiperlipídica , Modelos Animais de Doenças , Indução Enzimática , Oclusão de Enxerto Vascular/enzimologia , Oclusão de Enxerto Vascular/patologia , Oclusão de Enxerto Vascular/fisiopatologia , Sulfeto de Hidrogênio/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neointima , Estado Nutricional , Receptores de LDL/deficiência , Receptores de LDL/genética , Fatores de Tempo , Grau de Desobstrução Vascular , Veia Cava Inferior/enzimologia , Veia Cava Inferior/patologia , Veia Cava Inferior/fisiopatologia
4.
Immunity ; 51(4): 638-654.e9, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31561945

RESUMO

Macrophages are strongly adapted to their tissue of residence. Yet, little is known about the cell-cell interactions that imprint the tissue-specific identities of macrophages in their respective niches. Using conditional depletion of liver Kupffer cells, we traced the developmental stages of monocytes differentiating into Kupffer cells and mapped the cellular interactions imprinting the Kupffer cell identity. Kupffer cell loss induced tumor necrosis factor (TNF)- and interleukin-1 (IL-1) receptor-dependent activation of stellate cells and endothelial cells, resulting in the transient production of chemokines and adhesion molecules orchestrating monocyte engraftment. Engrafted circulating monocytes transmigrated into the perisinusoidal space and acquired the liver-associated transcription factors inhibitor of DNA 3 (ID3) and liver X receptor-α (LXR-α). Coordinated interactions with hepatocytes induced ID3 expression, whereas endothelial cells and stellate cells induced LXR-α via a synergistic NOTCH-BMP pathway. This study shows that the Kupffer cell niche is composed of stellate cells, hepatocytes, and endothelial cells that together imprint the liver-specific macrophage identity.


Assuntos
Células Endoteliais/fisiologia , Células Estreladas do Fígado/fisiologia , Hepatócitos/fisiologia , Células de Kupffer/fisiologia , Fígado/citologia , Macrófagos/fisiologia , Monócitos/fisiologia , Animais , Comunicação Celular , Diferenciação Celular , Células Cultivadas , Microambiente Celular , Feminino , Regulação da Expressão Gênica , Proteínas Inibidoras de Diferenciação/genética , Proteínas Inibidoras de Diferenciação/metabolismo , Receptores X do Fígado/genética , Receptores X do Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores Notch/metabolismo
5.
Immunity ; 49(2): 312-325.e5, 2018 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-30076102

RESUMO

Heterogeneity between different macrophage populations has become a defining feature of this lineage. However, the conserved factors defining macrophages remain largely unknown. The transcription factor ZEB2 is best described for its role in epithelial to mesenchymal transition; however, its role within the immune system is only now being elucidated. We show here that Zeb2 expression is a conserved feature of macrophages. Using Clec4f-cre, Itgax-cre, and Fcgr1-cre mice to target five different macrophage populations, we found that loss of ZEB2 resulted in macrophage disappearance from the tissues, coupled with their subsequent replenishment from bone-marrow precursors in open niches. Mechanistically, we found that ZEB2 functioned to maintain the tissue-specific identities of macrophages. In Kupffer cells, ZEB2 achieved this by regulating expression of the transcription factor LXRα, removal of which recapitulated the loss of Kupffer cell identity and disappearance. Thus, ZEB2 expression is required in macrophages to preserve their tissue-specific identities.


Assuntos
Células de Kupffer/citologia , Receptores X do Fígado/genética , Homeobox 2 de Ligação a E-box com Dedos de Zinco/genética , Animais , Linhagem da Célula/imunologia , Transição Epitelial-Mesenquimal , Feminino , Regulação Neoplásica da Expressão Gênica , Células de Kupffer/imunologia , Fígado/citologia , Receptores X do Fígado/metabolismo , Pulmão/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
6.
Sci Rep ; 7(1): 13646, 2017 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-29057962

RESUMO

TNF has as detrimental role in multiple sclerosis (MS), however, anti-TNF medication is not working. Selective TNF/TNFR1 inhibition whilst sparing TNFR2 signaling reduces the pro-inflammatory effects of TNF but preserves the important neuroprotective signals via TNFR2. We previously reported the generation of a Nanobody-based selective inhibitor of human TNFR1, TROS that will be tested in experimental autoimmune encephalomyelitis (EAE). We specifically antagonized TNF/TNFR1 signaling using TROS in a murine model of MS, namely MOG35-55-induced EAE. Because TROS does not cross-react with mouse TNFR1, we generated mice expressing human TNFR1 in a mouse TNFR1-knockout background (hTNFR1 Tg), and we determined biodistribution of 99mTc-TROS and effectiveness of TROS in EAE in those mice. Biodistribution analysis demonstrated that intraperitoneally injected TROS is retained more in organs of hTNFR1 Tg mice compared to wild type mice. TROS was also detected in the cerebrospinal fluid (CSF) of hTNFR1 Tg mice. Prophylactic TROS administration significantly delayed disease onset and ameliorated its symptoms. Moreover, treatment initiated early after disease onset prevented further disease development. TROS reduced spinal cord inflammation and neuroinflammation, and preserved myelin and neurons. Collectively, our data illustrate that TNFR1 is a promising therapeutic target in MS.


Assuntos
Encefalomielite Autoimune Experimental/prevenção & controle , Fatores Imunológicos/farmacologia , Fármacos Neuroprotetores/farmacologia , Receptores Tipo I de Fatores de Necrose Tumoral/antagonistas & inibidores , Anticorpos de Domínio Único/farmacologia , Animais , Encefalomielite Autoimune Experimental/patologia , Humanos , Fatores Imunológicos/farmacocinética , Masculino , Camundongos Transgênicos , Glicoproteína Mielina-Oligodendrócito , Fármacos Neuroprotetores/farmacocinética , Fragmentos de Peptídeos , Receptores Tipo I de Fatores de Necrose Tumoral/deficiência , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/patologia , Tecnécio , Fator de Necrose Tumoral alfa/metabolismo , Imagem Corporal Total
7.
Dev Cell ; 31(6): 722-33, 2014 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-25499914

RESUMO

Cellular senescence suppresses cancer by halting the growth of premalignant cells, yet the accumulation of senescent cells is thought to drive age-related pathology through a senescence-associated secretory phenotype (SASP), the function of which is unclear. To understand the physiological role(s) of the complex senescent phenotype, we generated a mouse model in which senescent cells can be visualized and eliminated in living animals. We show that senescent fibroblasts and endothelial cells appear very early in response to a cutaneous wound, where they accelerate wound closure by inducing myofibroblast differentiation through the secretion of platelet-derived growth factor AA (PDGF-AA). In two mouse models, topical treatment of senescence-free wounds with recombinant PDGF-AA rescued the delayed wound closure and lack of myofibroblast differentiation. These findings define a beneficial role for the SASP in tissue repair and help to explain why the SASP evolved.


Assuntos
Senescência Celular , Células Endoteliais/citologia , Mesoderma/citologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Cicatrização , Animais , Apoptose , Diferenciação Celular , Feminino , Fibroblastos/metabolismo , Luminescência , Masculino , Camundongos , Camundongos Transgênicos , Miofibroblastos/metabolismo , Fator de Crescimento Derivado de Plaquetas/química , Transgenes
8.
PLoS Biol ; 4(10): e322, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17020410

RESUMO

Although compound heterozygosity, or the presence of two different mutant alleles of the same gene, is common in human recessive disease, its potential to impact disease outcome has not been well documented. This is most likely because of the inherent difficulty in distinguishing specific biallelic effects from differences in environment or genetic background. We addressed the potential of different recessive alleles to contribute to the enigmatic pleiotropy associated with XPD recessive disorders in compound heterozygous mouse models. Alterations in this essential helicase, with functions in both DNA repair and basal transcription, result in diverse pathologies ranging from elevated UV sensitivity and cancer predisposition to accelerated segmental progeria. We report a variety of biallelic effects on organismal phenotype attributable to combinations of recessive Xpd alleles, including the following: (i) the ability of homozygous lethal Xpd alleles to ameliorate a variety of disease symptoms when their essential basal transcription function is supplied by a different disease-causing allele, (ii) differential developmental and tissue-specific functions of distinct Xpd allele products, and (iii) interallelic complementation, a phenomenon rarely reported at clinically relevant loci in mammals. Our data suggest a re-evaluation of the contribution of "null" alleles to XPD disorders and highlight the potential of combinations of recessive alleles to affect both normal and pathological phenotypic plasticity in mammals.


Assuntos
Alelos , Transtornos do Crescimento/genética , Doenças do Cabelo/genética , Homozigoto , Ictiose/genética , Progéria/genética , Proteína Grupo D do Xeroderma Pigmentoso/genética , Animais , Dano ao DNA , Genes Letais , Genes Recessivos , Transtornos do Crescimento/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Progéria/metabolismo , Fator de Transcrição TFIIH/genética , Fator de Transcrição TFIIH/metabolismo , Transcrição Gênica , Raios Ultravioleta
9.
Cancer Cell ; 10(2): 121-32, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16904611

RESUMO

Inborn defects in nucleotide excision DNA repair (NER) can paradoxically result in elevated cancer incidence (xeroderma pigmentosum [XP]) or segmental progeria without cancer predisposition (Cockayne syndrome [CS] and trichothiodystrophy [TTD]). We report generation of a knockin mouse model for the combined disorder XPCS with a G602D-encoding mutation in the Xpd helicase gene. XPCS mice are the most skin cancer-prone NER model to date, and we postulate an unusual NER dysfunction that is likely responsible for this susceptibility. XPCS mice also displayed symptoms of segmental progeria, including cachexia and progressive loss of germinal epithelium. Like CS fibroblasts, XPCS and TTD fibroblasts from human and mouse showed evidence of defective repair of oxidative DNA lesions that may underlie these segmental progeroid symptoms.


Assuntos
Síndrome de Cockayne/patologia , Progéria/patologia , Neoplasias Cutâneas/patologia , Proteína Grupo D do Xeroderma Pigmentoso/metabolismo , Xeroderma Pigmentoso/patologia , Animais , Carcinoma de Células Escamosas/etiologia , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Transformada , Síndrome de Cockayne/complicações , Síndrome de Cockayne/metabolismo , Reparo do DNA , Modelos Animais de Doenças , Suscetibilidade a Doenças , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Masculino , Camundongos , Camundongos Mutantes , Mutação , Papiloma/etiologia , Papiloma/metabolismo , Papiloma/patologia , Fenótipo , Progéria/complicações , Progéria/metabolismo , Neoplasias Cutâneas/etiologia , Neoplasias Cutâneas/metabolismo , Xeroderma Pigmentoso/complicações , Xeroderma Pigmentoso/metabolismo , Proteína Grupo D do Xeroderma Pigmentoso/genética
10.
J Virol ; 77(22): 12132-9, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14581550

RESUMO

The presence of porcine endogenous retroviruses presents a potential risk of transmission of infectious diseases (xenozoonosis) if tissues and organs from genetically modified pigs are to be used in xenotransplantation. Here, we report that intracellular expression of a llama single-domain antibody against p15, the matrix domain protein of the porcine endogenous retrovirus Gag polyprotein, blocks retrovirus production, providing the possibility of eliminating the risk of infection in xenotransplantation.


Assuntos
Anticorpos Antivirais/fisiologia , Retrovirus Endógenos/fisiologia , Produtos do Gene gag/antagonistas & inibidores , Infecções por Retroviridae/prevenção & controle , Suínos/virologia , Transplante Heterólogo/efeitos adversos , Proteínas da Matriz Viral/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Afinidade de Anticorpos , Mapeamento de Epitopos , Produtos do Gene gag/imunologia , Humanos , Soros Imunes/imunologia , Cadeias Pesadas de Imunoglobulinas/fisiologia , Região Variável de Imunoglobulina/fisiologia , Dados de Sequência Molecular , Proteínas da Matriz Viral/imunologia , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA