Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 19(7): e1011059, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37399208

RESUMO

Transmitted/founder (TF) simian-human immunodeficiency viruses (SHIVs) express HIV-1 envelopes modified at position 375 to efficiently infect rhesus macaques while preserving authentic HIV-1 Env biology. SHIV.C.CH505 is an extensively characterized virus encoding the TF HIV-1 Env CH505 mutated at position 375 shown to recapitulate key features of HIV-1 immunobiology, including CCR5-tropism, a tier 2 neutralization profile, reproducible early viral kinetics, and authentic immune responses. SHIV.C.CH505 is used frequently in nonhuman primate studies of HIV, but viral loads after months of infection are variable and typically lower than those in people living with HIV. We hypothesized that additional mutations besides Δ375 might further enhance virus fitness without compromising essential components of CH505 Env biology. From sequence analysis of SHIV.C.CH505-infected macaques across multiple experiments, we identified a signature of envelope mutations associated with higher viremia. We then used short-term in vivo mutational selection and competition to identify a minimally adapted SHIV.C.CH505 with just five amino acid changes that substantially improve virus replication fitness in macaques. Next, we validated the performance of the adapted SHIV in vitro and in vivo and identified the mechanistic contributions of selected mutations. In vitro, the adapted SHIV shows improved virus entry, enhanced replication on primary rhesus cells, and preserved neutralization profiles. In vivo, the minimally adapted virus rapidly outcompetes the parental SHIV with an estimated growth advantage of 0.14 days-1 and persists through suppressive antiretroviral therapy to rebound at treatment interruption. Here, we report the successful generation of a well-characterized, minimally adapted virus, termed SHIV.C.CH505.v2, with enhanced replication fitness and preserved native Env properties that can serve as a new reagent for NHP studies of HIV-1 transmission, pathogenesis, and cure.


Assuntos
Infecções por HIV , Soropositividade para HIV , HIV-1 , Síndrome de Imunodeficiência Adquirida dos Símios , Vírus da Imunodeficiência Símia , Animais , Humanos , Macaca mulatta/metabolismo , Produtos do Gene env do Vírus da Imunodeficiência Humana , Replicação Viral/fisiologia
2.
PLoS One ; 17(4): e0266703, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35404944

RESUMO

In 2019 a newly identified coronavirus, designated as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has spread rapidly from the epicenter in Wuhan (China) to more than 150 countries around the world, causing the Coronavirus disease 2019 (COVID-19) pandemic. In this study, we describe an extraction-less method based on reverse transcriptase loop-mediated isothermal amplification (RT-LAMP) intended for the rapid qualitative detection of nucleic acid from SARS-CoV-2 in upper respiratory specimens, including oropharyngeal and nasopharyngeal swabs, anterior nasal and mid-turbinate nasal swabs, nasopharyngeal washes/aspirates or nasal aspirates as well as bronchoalveolar lavage (BAL) from individuals suspected of COVID-19 by their healthcare provider. The assay's performance was evaluated and compared to an RT quantitative PCR-based assay (FDA-approved). With high sensitivity, specificity, and bypassing the need for RNA extraction, the RT-LAMP Rapid Detection assay is a valuable and fast test for an accurate and rapid RNA detection of the SARS-CoV-2 virus and potentially other pathogens. Additionally, the versatility of this test allows its application in virtually every laboratory setting and remote location where access to expensive laboratory equipment is a limiting factor for testing during pandemic crises.


Assuntos
COVID-19 , SARS-CoV-2 , COVID-19/diagnóstico , Análise Custo-Benefício , Humanos , Técnicas de Diagnóstico Molecular/métodos , Técnicas de Amplificação de Ácido Nucleico/métodos , RNA Viral/genética , SARS-CoV-2/genética , Sensibilidade e Especificidade
3.
Gene Expr Patterns ; 23-24: 70-79, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28363632

RESUMO

Activins and inhibins, members of the TGF-ß superfamily, are growth and differentiation factors involved in the regulation of several biological processes, including reproduction, development, and fertility. Previous studies have shown that the activin-ßA subunit plays a pivotal role in prostate development. Activin-A inhibits branching morphogenesis in the developing prostate, and its expression is associated with increased apoptosis in the adult prostate. Follistatin, a structurally unrelated protein to activins, is an antagonist of activin-A. A balance between endogenous activin-A and follistatin is required to maintain prostatic branching morphogenesis. Deregulation of this balance leads to branching inhibition or excessive branching and increased maturation of the stroma surrounding the differentiating epithelial ducts. Recent work identified another member of the TGF-ß superfamily, the activin-ßC subunit, as a novel antagonist of activin-A. Over-expression of activin-C (ßC-ßC) alters prostate homeostasis, by interfering with the activin-A signaling. The current study characterized the spatiotemporal localization of activin-A, activin-C and follistatin in the adult and developing mouse prostate using immunohistochemical analysis. Results showed activin-C and follistatin are differentially expressed during prostate development and suggested that the antagonistic property of follistatin is secondary to the action of activin-C. In conclusion, the present study provides evidence to support a role of activin-C in prostate development and provides new insights in the spatiotemporal localization of activins and their antagonists during mouse prostate development.


Assuntos
Folistatina/análise , Folistatina/metabolismo , Subunidades beta de Inibinas/análise , Subunidades beta de Inibinas/metabolismo , Próstata/crescimento & desenvolvimento , Animais , Apoptose , Diferenciação Celular , Subunidades beta de Inibinas/antagonistas & inibidores , Masculino , Camundongos , Próstata/citologia , Próstata/metabolismo
4.
J Cachexia Sarcopenia Muscle ; 6(4): 365-80, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26673867

RESUMO

BACKGROUND: Cancer-associated cachexia and muscle wasting are considered key determinants of cancer-related death and reduction in the quality of life of cancer patients. A crucial link has been established between activin signaling and skeletal muscle atrophy-hypertrophy. We previously showed that activin-ßC, a novel activin-A antagonist, is a tumor modulator that abolishes the cancer-associated cachexia in a mouse genetic model of gonadal tumorigenesis, in which the normal balance of inhibin/activin signalling is disrupted by a targeted mutation in the Inha gene (inhibin α-KO mouse). This study aimed to identify the molecular mechanism by which activin-ßC increases survival and abolishes cancer-associated cachexia in α-KO mice. We hypothesized that overexpression of activin-ßC modulates the cachexia phenotype by antagonizing the activin signaling pathway and repressing muscle wasting via the ubiquitin-proteasome and the autophagic-lysosomal degradation pathways. METHODS: Male and female ActC++, α-KO, and α-KO/ActC++ mice and WT littermate controls were studied. Western blot analysis for the specific E3 ubiquitin ligases, atrogin-1 and MuRF1, markers of the autophagic-lysosomal pathway, Beclin-1, p62, and LC3A/B, effectors Smad-2, Smad-3 and myostatin was performed in the gastrocnemius of age-matched mice. Histopathology of the gastrocnemius and survival analysis were also conducted in animals from the same breeding cohort. Serum levels of activin-A, inflammatory cytokines, hormonal profile, and bone density were also assessed. RESULTS: Increased levels of atrogin-1, MuRF-1, Beclin-1, p62, LC3A/B-I, Smad-2 and serum levels of activin-A were noted in the α-KO mice. These mice developed gonadal cancers followed by severe weight loss, and reduced survival. Overexpression of activin- ßC antagonized the activin signaling cascade, attenuating the ubiquitin-proteasome and the autophagic-lysosomal degradation pathways, and reduced serum levels of activin-A. α-KO/ActC++ mice displayed a less aggressive cachectic phenotype, reduced tumor weight, and prolonged survival. CONCLUSION: Our findings show for the first time a specific effect of activin-ßC on muscle wasting and transcription factors involved in muscle protein degradation. The study indicates that activin-ßC may be a novel therapy to abrogate cancer-associated weight loss and prolong survival.

5.
Cytokine Growth Factor Rev ; 26(4): 463-70, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26058376

RESUMO

Transforming growth factor-ß (TGF-ß) superfamily signaling pathway and its ligands are essential regulators of cellular processes such as proliferation, differentiation, migration, and survival. Alteration of this pathway results in uncontrolled proliferation and cancer progression. This review focuses on a specific member of the TGF-ß superfamily: activin-ßC. After its initial discovery, activin-ßC has been considered non-biologically relevant. Therefore, for years several experimental designs have ignored the potential contribution of this molecule to the final biological outcome. Here we focus on recent advances in the activin field, with a particular emphasis on activin-ßC, its antagonistic mechanism, and the physiological relevance of activin-ßC actions in reproductive and cancer biology. Covering a novel and previously unexplored function of activin-ßC on cancer associated weight loss and muscle metabolism, this review suggests an imminent need to re-evaluate the function of activin-ßC in biological systems and advances the understanding of how activin-ßC antagonizes the activin signaling pathway. Thus, challenging activin biologists to consider the impact of activin-ßC when interpreting their work.


Assuntos
Subunidades beta de Inibinas/biossíntese , Neoplasias/metabolismo , Animais , Diferenciação Celular/fisiologia , Humanos , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
6.
Mol Cell Endocrinol ; 409: 41-50, 2015 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-25869398

RESUMO

Activins and inhibins are involved in the regulation of several biological processes, including reproduction, development and fertility. Deregulation of the inhibin/activin signaling pathway has been implicated in the progression of reproductive and adrenal cancers. Deletion of the inhibin α-subunit results in up-regulation of the circulating levels of activins and this leads to the development of sex-cord stromal tumors followed by a cancer associated-cachexia in mice. When gonadectomy is performed, development of adrenocortical carcinomas is observed. We previously showed that overexpression of activin-ßC modulates the development of sex-cord stromal tumors and reduces cancer-cachexia in the inhibin-deficient mice by antagonizing the activin signaling pathway. The adrenal cortex and gonads share in common a large subset of genes, consistent with their common embryonic lineage. Additionally, it has been shown that adrenocortical carcinomas adopt an altered cellular identity resembling the ovary. Therefore, a study to assess the impact of overexpression of activin-ßC on the onset of adrenocortical carcinoma in gonadectomized inhibin-deficient mice was warranted. Within the current study we evaluated markers of apoptosis, proliferation, tumor burden, survival analysis and serum levels of activin-A in gonadectomized mice versus sham operated controls. Results showed that overexpression of activin-ßC modulated the development of reproductive tumors but had no effect on adrenal tumorigenesis. Our data reinforces the importance of activin-ßC in reproductive biology and suggest that activin-ßC is a tumor modulator with gonadal specificity.


Assuntos
Neoplasias das Glândulas Suprarrenais/patologia , Subunidades beta de Inibinas/deficiência , Subunidades beta de Inibinas/metabolismo , Neoplasias de Tecido Gonadal/patologia , Neoplasias das Glândulas Suprarrenais/metabolismo , Animais , Feminino , Gônadas/cirurgia , Masculino , Camundongos , Camundongos Knockout , Neoplasias de Tecido Gonadal/metabolismo , Análise de Sobrevida
7.
Mol Hum Reprod ; 20(12): 1223-37, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25180271

RESUMO

The biological function of the inhibin-α subunit (INHA) in gonadal tumorigenesis is different in humans compared with mouse. The INHA subunit is up-regulated in most human ovarian and testicular cancers but knock-out studies in mice showed the INHA subunit is a tumour suppressor with gonadal and adrenal specificity. The INHA subunit is a component of the inhibin/activin signalling pathway, which includes activin receptors ActRIIA/IIB and intracellular Smads-2/3. To resolve the incongruity in function in humans versus mouse, we re-evaluated the inhibin/activin pathway in human gonadal and adrenal cancers using contemporary protein and mRNA expression data for multiple pathway components rather than INHA alone. We used an INHA antibody raised against the N-terminal domain to compare immunoreactivity with the more commonly used antibody raised against the C-terminal domain. This study also described, for the first time, a comprehensive protein expression profile of activin-ßC in reproductive and adrenal cancers, and its effect on a human granulosa cell line, providing evidence for a role in ovarian, testis and adrenal tumour biology. Our data show reduced INHA expression at both protein and mRNA levels, and increased activin signalling in human testicular, ovarian and malignant versus benign forms of adrenal cancer. We also found that activin-C acts as an activin-A antagonist by binding to activin receptor subunits IIA and IIB and modulating the canonical Smad pathway. In conclusion, analysis of the inhibin/activin signalling pathway helps to explain discrepancies arising from studies of only one hormone or subunit and suggests that altered expression of the inhibin and activin subunits is associated with reproductive and adrenal cancer biology.


Assuntos
Ativinas/metabolismo , Neoplasias das Glândulas Suprarrenais/metabolismo , Inibinas/metabolismo , Neoplasias Ovarianas/metabolismo , Transdução de Sinais , Neoplasias Testiculares/metabolismo , Receptores de Activinas Tipo II/metabolismo , Ativinas/genética , Neoplasias das Glândulas Suprarrenais/genética , Neoplasias das Glândulas Suprarrenais/patologia , Linhagem Celular Tumoral , Bases de Dados Genéticas , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Subunidades beta de Inibinas/metabolismo , Inibinas/genética , Masculino , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , Proteínas Smad Reguladas por Receptor/metabolismo , Neoplasias Testiculares/genética , Neoplasias Testiculares/patologia
8.
Cytokine Growth Factor Rev ; 24(5): 477-84, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23787160

RESUMO

Members of the transforming growth factor ß (TGF-ß) family regulate fundamental physiological process, such as cell growth, differentiation and apoptosis. As a result, defects in this pathway have been linked to uncontrolled proliferation and cancer progression. Here we explore the signal transduction mechanism of TGF-ß focusing on therapeutic intervention in human diseases. Like TGF-ß, another member of the TGF-ß superfamily, activin has been proven to play an important role in maintenance of tissue homeostasis and dysregulation leads to disease. Several studies showed elevated levels of activin are responsible for the development of gonadal tumours and a cachexia-like weight loss syndrome. Discussing the recent advances in approaches developed to antagonise the activin pathway and the encouraging results obtained in animal models, this review presents a therapeutic rationale for targeting the activin pathway in conditions such as cachexia, neuromuscular and/or musculoskeletal disorders.


Assuntos
Ativinas/antagonistas & inibidores , Antineoplásicos/uso terapêutico , Caquexia/tratamento farmacológico , Doenças Musculoesqueléticas/tratamento farmacológico , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias de Tecido Gonadal/tratamento farmacológico , Doenças Neuromusculares/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Ativinas/genética , Ativinas/metabolismo , Animais , Caquexia/genética , Caquexia/metabolismo , Proliferação de Células/efeitos dos fármacos , Humanos , Doenças Musculoesqueléticas/genética , Doenças Musculoesqueléticas/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias de Tecido Gonadal/genética , Neoplasias de Tecido Gonadal/metabolismo , Doenças Neuromusculares/genética , Doenças Neuromusculares/metabolismo , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
9.
J Pathol ; 229(4): 599-607, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23180294

RESUMO

Activins are involved in the regulation of a diverse range of physiological processes including development, reproduction, and fertility, and have been implicated in the progression of cancers. Bioactivity is regulated by the inhibin α-subunit and by an activin-binding protein, follistatin. The activin-ß(C) subunit was not considered functionally significant in this regard due to an absence of phenotype in knockout mice. However, activin-ß(C) forms heterodimers with activin-ß(A) and activin-C antagonizes activin-A in vitro. Thus, it is proposed that overexpression, rather than loss of activin-ß(C) , regulates activin-A bioactivity. In order to prove biological efficacy, inhibin α-subunit knockout mice (α-KO) were crossed with mice overexpressing activin-ß(C) (ActC++). Deletion of inhibin leads to Sertoli and granulosa cell tumours, increased activin-A, and cancer-associated cachexia. Therefore, cachexia and reproductive tumour development should be modulated in α-KO/ActC++ mice, where excessive activin-A is the underlying cause. Accordingly, a reduction in activin-A, no significant weight loss, and reduced incidence of reproductive tumours were evident in α-KO/ActC++ mice. Overexpression of activin-ß(C) antagonized the activin signalling cascade; thus, the tumourigenic effects of activin-A were abrogated. This study provides proof of the biological relevance of activin-ß(C) . Being a regulator of activin-A, it is able to abolish cachexia and modulate reproductive tumour development in α-KO mice.


Assuntos
Ativinas/metabolismo , Caquexia/genética , Subunidades beta de Inibinas/metabolismo , Neoplasias Ovarianas/genética , Neoplasias Testiculares/genética , Ativinas/sangue , Ativinas/genética , Animais , Apoptose , Linhagem Celular , Proliferação de Células , Dimerização , Progressão da Doença , Feminino , Folistatina/metabolismo , Expressão Gênica , Humanos , Subunidades beta de Inibinas/genética , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Fosforilação , Deleção de Sequência , Proteína Smad2/metabolismo , Neoplasias Testiculares/metabolismo , Neoplasias Testiculares/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...