Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 132(20)2022 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-36006736

RESUMO

Invasive bacterial infections remain a major cause of human morbidity. Group B streptococcus (GBS) are Gram-positive bacteria that cause invasive infections in humans. Here, we show that factor XIIIA-deficient (FXIIIA-deficient) female mice exhibited significantly increased susceptibility to GBS infections. Additionally, female WT mice had increased levels of FXIIIA and were more resistant to GBS infection compared with isogenic male mice. We observed that administration of exogenous FXIIIA to male mice increased host resistance to GBS infection. Conversely, administration of a FXIIIA transglutaminase inhibitor to female mice decreased host resistance to GBS infection. Interestingly, male gonadectomized mice exhibited decreased sensitivity to GBS infection, suggesting a role for gonadal androgens in host susceptibility. FXIIIA promoted GBS entrapment within fibrin clots by crosslinking fibronectin with ScpB, a fibronectin-binding GBS surface protein. Thus, ScpB-deficient GBS exhibited decreased entrapment within fibrin clots in vitro and increased dissemination during systemic infections. Finally, using mice in which FXIIIA expression was depleted in mast cells, we observed that mast cell-derived FXIIIA contributes to host defense against GBS infection. Our studies provide insights into the effects of sexual dimorphism and mast cells on FXIIIA expression and its interactions with GBS adhesins that mediate bacterial dissemination and pathogenesis.


Assuntos
Fator XIIIa , Infecções Estreptocócicas , Androgênios/metabolismo , Animais , Fator XIIIa/metabolismo , Feminino , Fibrina/metabolismo , Fibronectinas/genética , Fibronectinas/metabolismo , Humanos , Masculino , Mastócitos/metabolismo , Camundongos , Infecções Estreptocócicas/genética , Streptococcus agalactiae/metabolismo , Transglutaminases/metabolismo
2.
Methods Mol Biol ; 2159: 129-140, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32529368

RESUMO

Dynamin-related proteins on both the mitochondrial outer and inner membranes mediate membrane fusion. Mitochondrial fusion is regulated in many different physiological contexts including cell cycle progression, differentiation pathways, stress responses, and cell death. Mitochondrial fusion is opposed by mitochondrial division and requires movement of mitochondria on microtubules. We developed a cell-free reconstituted mitochondrial fusion assay to circumvent the complexity of the pathways impinging on the activity of the mitochondrial fusion machinery in vivo. This allows for quantification of mitochondrial fusion in defined conditions and in the absence of other processes such as mitochondrial division or transport. The impact of proteins or small molecules on mitochondria fusion can also be assessed. Here we describe the cell-free mitochondrial fusion assay using mitochondria isolated from mouse embryonic fibroblasts.


Assuntos
Microscopia de Fluorescência , Mitocôndrias/metabolismo , Dinâmica Mitocondrial , Animais , Biomarcadores , Fracionamento Celular , Fibroblastos/metabolismo , Imunofluorescência , Fusão de Membrana , Camundongos , Proteínas Mitocondriais/metabolismo , Imagem Molecular
3.
Endocr Relat Cancer ; 22(2): 239-48, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25800038

RESUMO

The ACI rat model of 17ß-estradiol (E2)-induced mammary cancer is highly relevant for use in establishing the endocrine, genetic, and environmental bases of breast cancer etiology and identifying novel agents and strategies for preventing breast cancer. E2 treatment rapidly induces mammary cancer in female ACI rats and simultaneously induces pituitary lactotroph hyperplasia and adenoma. The pituitary tumors can result in undesired morbidity, which compromises long-term studies focused on mammary cancer etiology and prevention. We have defined the genetic bases of susceptibility to E2-induced mammary cancers and pituitary tumors and have utilized the knowledge gained in these studies to develop a novel inbred rat strain, designated ACWi, that retains the high degree of susceptibility to E2-induced mammary cancer exhibited by ACI rats, but lacks the treatment-related morbidity associated with pituitary lactotroph hyperplasia/adenoma. When treated with E2, female ACWi rats developed palpable mammary cancer at a median latency of 116 days, an incidence of 100% by 161 days and exhibited an average of 15.6 mammary tumors per rat following 196 days of treatment. These parameters did not differ from those observed for contemporaneously treated ACI rats. None of the E2-treated ACWi rats were killed before the intended experimental end point due to any treatment-related morbidity other than mammary cancer burden, whereas 20% of contemporaneously treated ACI rats exhibited treatment-related morbidity that necessitated premature killing. The ACWi rat strain is well suited for use by those in the research community, focusing on breast cancer etiology and prevention.


Assuntos
Neoplasias Mamárias Experimentais , Ratos Endogâmicos , Animais , Estradiol , Feminino , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/patologia , Fenótipo , Hipófise/patologia
4.
PLoS One ; 10(2): e0118147, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25693193

RESUMO

Congenital anomalies of the kidney and urogenital tract (CAKUT) occur in approximately 0.5% of live births and represent the most frequent cause of end-stage renal disease in neonates and children. The genetic basis of CAKUT is not well defined. To understand more fully the genetic basis of one type of CAKUT, unilateral renal agenesis (URA), we are studying inbred ACI rats, which spontaneously exhibit URA and associated urogenital anomalies at an incidence of approximately 10%. URA is inherited as an incompletely dominant trait with incomplete penetrance in crosses between ACI and Brown Norway (BN) rats and a single responsible genetic locus, designated Renag1, was previously mapped to rat chromosome 14 (RNO14). The goals of this study were to fine map Renag1, identify the causal genetic variant responsible for URA, confirm that the Renag1 variant is the sole determinant of URA in the ACI rat, and define the embryologic basis of URA in this rat model. Data presented herein localize Renag1 to a 379 kilobase (kb) interval that contains a single protein coding gene, Kit (v-kit Hardy-Zukerman 4 feline sarcoma viral oncogene homolog); identify an endogenous retrovirus-derived long terminal repeat located within Kit intron 1 as the probable causal variant; demonstrate aberrant development of the nephric duct in the anticipated number of ACI rat embryos; and demonstrate expression of Kit and Kit ligand (Kitlg) in the nephric duct. Congenic rats that harbor ACI alleles at Renag1 on the BN genetic background exhibit the same spectrum of urogenital anomalies as ACI rats, indicating that Renag1 is necessary and sufficient to elicit URA and associated urogenital anomalies. These data reveal the first genetic link between Kit and URA and illustrate the value of the ACI rat as a model for defining the mechanisms and cell types in which Kit functions during urogenital development.


Assuntos
Cromossomos de Mamíferos/genética , Anormalidades Congênitas/genética , Anormalidades Congênitas/patologia , Nefropatias/congênito , Rim/anormalidades , Mapeamento Físico do Cromossomo/métodos , Proteínas Proto-Oncogênicas c-kit/genética , Animais , Modelos Animais de Doenças , Estudos de Associação Genética , Loci Gênicos , Predisposição Genética para Doença , Variação Genética , Rim/patologia , Nefropatias/genética , Nefropatias/patologia , Ratos , Ratos Endogâmicos ACI , Fator de Células-Tronco/genética
5.
G3 (Bethesda) ; 4(8): 1385-94, 2014 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-24875630

RESUMO

When treated with 17ß-estradiol, female ACI rats (Rattus norvegicus) rapidly develop mammary cancers that share multiple phenotypes with luminal breast cancers. Seven distinct quantitative trait loci that harbor genetic determinants of susceptibility to 17ß-estradiol-induced mammary cancer have been mapped in reciprocal intercrosses between susceptible ACI rats and resistant Brown Norway (BN) rats. A panel of unique congenic rat strains has now been generated and characterized to confirm the existence of these quantitative trait loci, designated Emca3 through Emca9, and to quantify their individual effects on susceptibility to 17ß-estradiol-induced mammary cancer. Each congenic strain carries BN alleles spanning an individual Emca locus, introgressed onto the ACI genetic background. Data presented herein indicate that BN alleles at Emca3, Emca4, Emca5, Emca6, and Emca9 reduce susceptibility to 17ß-estradiol-induced mammary cancer, whereas BN alleles at Emca7 increase susceptibility, thereby confirming the previous interval mapping data. All of these Emca loci are orthologous to regions of the human genome that have been demonstrated in genome-wide association studies to harbor genetic variants that influence breast cancer risk. Moreover, four of the Emca loci are orthologous to loci in humans that have been associated with mammographic breast density, a biomarker of breast cancer risk. This study further establishes the relevance of the ACI and derived congenic rat models of 17ß-estradiol-induced mammary cancer for defining the genetic bases of breast cancer susceptibility and elucidating the mechanisms through which 17ß-estradiol contributes to breast cancer development.


Assuntos
Predisposição Genética para Doença , Neoplasias Mamárias Animais/genética , Locos de Características Quantitativas , Animais , Animais Congênicos , Estradiol , Estrogênios , Feminino , Humanos , Neoplasias Mamárias Animais/induzido quimicamente , Fenótipo , Ratos Endogâmicos BN , Risco
6.
Mamm Genome ; 25(5-6): 244-52, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24448715

RESUMO

Estrogens control many aspects of pituitary gland biology, including regulation of lactotroph homeostasis and synthesis and secretion of prolactin. In rat models, these actions are strain specific and heritable, and multiple quantitative trait loci (QTL) have been mapped that impact the responsiveness of the lactotroph to estrogens. One such QTL, Ept7, was mapped to RNO7 in female progeny generated in an intercross between BN rats, in which the lactotroph population is insensitive to estrogens, and ACI rats, which develop lactotroph hyperplasia/adenoma and associated hyperprolactinemia in response to estrogen treatment. The primary objective of this study was to confirm the existence of Ept7 and to quantify the impact of this QTL on responsiveness of the pituitary gland of female and male rats to 17ß-estradiol (E2) and diethylstilbestrol (DES), respectively. Secondary objectives were to determine if Ept7 influences the responsiveness of the male reproductive tract to DES and to identify other discernible phenotypes influenced by Ept7. To achieve these objectives, a congenic rat strain that harbors BN alleles across the Ept7 interval on the genetic background of the ACI strain was generated and characterized to define the effect of administered estrogens on the anterior pituitary gland and male reproductive tissues. Data presented herein indicate Ept7 exerts a marked effect on development of lactotroph hyperplasia in response to estrogen treatment, but does not affect atrophy of the male reproductive tissues in response to hormone treatment. Ept7 was also observed to exert gender specific effects on body weight in young adult rats.


Assuntos
Peso Corporal , Estrogênios/metabolismo , Hipófise/metabolismo , Ratos/genética , Alelos , Animais , Feminino , Masculino , Locos de Características Quantitativas , Ratos/crescimento & desenvolvimento , Ratos/metabolismo , Ratos Endogâmicos ACI , Ratos Endogâmicos BN
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...