Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Account Res ; 26(5): 288-310, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31155934

RESUMO

Academic medical centers rarely require all of their research faculty and staff to participate in educational programs on the responsible conduct of research (RCR). There is also little published evidence of RCR programs addressing high-profile, internal cases of misconduct as a way of promoting deliberation and learning. In the wake of major research misconduct, Duke University School of Medicine (DUSoM) expanded its RCR education activities to include all DUSoM faculty and staff engaged in research. The program included formal deliberation of the Translational Omics misconduct case, which occurred at Duke. Over 5,000 DUSoM faculty and staff participated in the first phase of this new program, with a 100% completion rate. The article reports on the program's development, challenges and successes, and future directions. This experience at Duke University illustrates that, although challenging and resource intensive, engagement with RCR activities can be integrated into programs for all research faculty and staff. Formal, participatory deliberation of recent cases of internal misconduct can add a novel dimension of reflection and openness to RCR educational activities.


Assuntos
Currículo , Faculdades de Medicina , Má Conduta Científica , North Carolina , Desenvolvimento de Programas
2.
Elife ; 82019 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-30693866

RESUMO

Mycobacterium tuberculosis is the leading worldwide cause of death due to a single infectious agent. Existing anti-tuberculous therapies require long treatments and are complicated by multi-drug-resistant strains. Host-directed therapies have been proposed as an orthogonal approach, but few have moved into clinical trials. Here, we use the zebrafish-Mycobacterium marinum infection model as a whole-animal screening platform to identify FDA-approved, host-directed compounds. We identify multiple compounds that modulate host immunity to limit mycobacterial disease, including the inexpensive, safe, and widely used drug clemastine. We find that clemastine alters macrophage calcium transients through potentiation of the purinergic receptor P2RX7. Host-directed drug activity in zebrafish larvae depends on both P2RX7 and inflammasome signaling. Thus, targeted activation of a P2RX7 axis provides a novel strategy for enhanced control of mycobacterial infections. Using a novel explant model, we find that clemastine is also effective within the complex granulomas that are the hallmark of mycobacterial infection.


Assuntos
Antituberculosos/farmacologia , Clemastina/farmacologia , Granuloma/tratamento farmacológico , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Receptores Purinérgicos P2X7/genética , Proteínas de Peixe-Zebra/genética , Animais , Antialérgicos/farmacologia , Cálcio/imunologia , Cálcio/metabolismo , Modelos Animais de Doenças , Reposicionamento de Medicamentos , Regulação da Expressão Gênica , Granuloma/genética , Granuloma/imunologia , Granuloma/microbiologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata/efeitos dos fármacos , Inflamassomos , Larva/efeitos dos fármacos , Larva/genética , Larva/imunologia , Larva/microbiologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/microbiologia , Infecções por Mycobacterium não Tuberculosas/genética , Infecções por Mycobacterium não Tuberculosas/imunologia , Infecções por Mycobacterium não Tuberculosas/microbiologia , Mycobacterium marinum/crescimento & desenvolvimento , Mycobacterium marinum/imunologia , Mycobacterium marinum/patogenicidade , Mycobacterium tuberculosis/patogenicidade , Receptores Purinérgicos P2X7/imunologia , Transdução de Sinais , Técnicas de Cultura de Tecidos , Tuberculose Pulmonar/tratamento farmacológico , Tuberculose Pulmonar/microbiologia , Peixe-Zebra/genética , Peixe-Zebra/imunologia , Peixe-Zebra/microbiologia , Proteínas de Peixe-Zebra/agonistas , Proteínas de Peixe-Zebra/imunologia
3.
J Infect Dis ; 215(5): 813-817, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-27496976

RESUMO

Pathogenic mycobacteria trigger formation of organized granulomas. As granulomas mature, they induce angiogenesis and vascular permeability. Here, in a striking parallel to tumor pro-angiogenic signaling, we identify angiopoietin-2 (ANG-2) induction as an important component of vascular dysfunction during mycobacterial infection. Mycobacterial infection in humans and zebrafish results in robust induction of ANG-2 expression from macrophages and stromal cells. Using a small-molecule inhibitor closely related to one currently in clinical trials, we link ANG-2/TIE2 signaling to vascular permeability during mycobacterial infection. Targeting granuloma-induced vascular permeability via vascular endothelial-protein tyrosine phosphatase inhibition limits mycobacterial growth, suggesting a new strategy for host-directed therapies against tuberculosis.


Assuntos
Angiopoietina-2/metabolismo , Permeabilidade Capilar , Infecções por Mycobacterium/patologia , Mycobacterium/crescimento & desenvolvimento , Angiopoietina-2/genética , Animais , Animais Geneticamente Modificados , Modelos Animais de Doenças , Regulação da Expressão Gênica , Granuloma/microbiologia , Interações Hospedeiro-Patógeno , Humanos , Larva , Macrófagos/efeitos dos fármacos , Macrófagos/microbiologia , Mycobacterium/efeitos dos fármacos , Receptor TIE-2/metabolismo , Transdução de Sinais , Tuberculose/microbiologia , Peixe-Zebra
4.
Immunity ; 45(4): 861-876, 2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27760340

RESUMO

Mycobacterium tuberculosis infection in humans triggers formation of granulomas, which are tightly organized immune cell aggregates that are the central structure of tuberculosis. Infected and uninfected macrophages interdigitate, assuming an altered, flattened appearance. Although pathologists have described these changes for over a century, the molecular and cellular programs underlying this transition are unclear. Here, using the zebrafish-Mycobacterium marinum model, we found that mycobacterial granuloma formation is accompanied by macrophage induction of canonical epithelial molecules and structures. We identified fundamental macrophage reprogramming events that parallel E-cadherin-dependent mesenchymal-epithelial transitions. Macrophage-specific disruption of E-cadherin function resulted in disordered granuloma formation, enhanced immune cell access, decreased bacterial burden, and increased host survival, suggesting that the granuloma can also serve a bacteria-protective role. Granuloma macrophages in humans with tuberculosis were similarly transformed. Thus, during mycobacterial infection, granuloma macrophages are broadly reprogrammed by epithelial modules, and this reprogramming alters the trajectory of infection and the associated immune response.


Assuntos
Epitélio/imunologia , Macrófagos/imunologia , Mycobacterium marinum/imunologia , Animais , Caderinas/imunologia , Epitélio/microbiologia , Granuloma/imunologia , Granuloma/microbiologia , Macrófagos/microbiologia , Mycobacterium tuberculosis/imunologia , Peixe-Zebra
6.
Cell Rep ; 13(10): 2107-17, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26673320

RESUMO

Calcium signaling has long been associated with key events of immunity, including chemotaxis, phagocytosis, and activation. However, imaging and manipulation of calcium flux in motile immune cells in live animals remain challenging. Using light-sheet microscopy for in vivo calcium imaging in zebrafish, we observe characteristic patterns of calcium flux triggered by distinct events, including phagocytosis of pathogenic bacteria and migration of neutrophils toward inflammatory stimuli. In contrast to findings from ex vivo studies, we observe enriched calcium influx at the leading edge of migrating neutrophils. To directly manipulate calcium dynamics in vivo, we have developed transgenic lines with cell-specific expression of the mammalian TRPV1 channel, enabling ligand-gated, reversible, and spatiotemporal control of calcium influx. We find that controlled calcium influx can function to help define the neutrophil's leading edge. Cell-specific TRPV1 expression may have broad utility for precise control of calcium dynamics in other immune cell types and organisms.


Assuntos
Sinalização do Cálcio/fisiologia , Microscopia de Fluorescência/métodos , Neutrófilos/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Animais Geneticamente Modificados , Cálcio/metabolismo , Quimiotaxia/fisiologia , Ratos , Canais de Cátion TRPV/genética , Peixe-Zebra
7.
PLoS One ; 10(10): e0138949, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26445458

RESUMO

Transgenic labeling of innate immune cell lineages within the larval zebrafish allows for real-time, in vivo analyses of microbial pathogenesis within a vertebrate host. To date, labeling of zebrafish macrophages has been relatively limited, with the most specific expression coming from the mpeg1 promoter. However, mpeg1 transcription at both endogenous and transgenic loci becomes attenuated in the presence of intracellular pathogens, including Salmonella typhimurium and Mycobacterium marinum. Here, we describe mfap4 as a macrophage-specific promoter capable of producing transgenic lines in which transgene expression within larval macrophages remains stable throughout several days of infection. Additionally, we have developed a novel macrophage-specific Cre transgenic line under the control of mfap4, enabling macrophage-specific expression using existing floxed transgenic lines. These tools enrich the repertoire of transgenic lines and promoters available for studying zebrafish macrophage dynamics during infection and inflammation and add flexibility to the design of future macrophage-specific transgenic lines.


Assuntos
Interações Hospedeiro-Patógeno/genética , Macrófagos/microbiologia , Infecções por Mycobacterium/genética , Regiões Promotoras Genéticas/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Peixe-Zebra/microbiologia , Animais , Animais Geneticamente Modificados/genética , Animais Geneticamente Modificados/microbiologia , Linhagem da Célula/genética , Modelos Animais de Doenças , Imunidade Inata/genética , Larva/genética , Larva/microbiologia , Infecções por Mycobacterium/microbiologia , Mycobacterium marinum/patogenicidade , Salmonella typhimurium/patogenicidade , Transgenes/genética
8.
Proc Natl Acad Sci U S A ; 112(9): 2770-5, 2015 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-25730872

RESUMO

The intestinal epithelium forms a barrier protecting the organism from microbes and other proinflammatory stimuli. The integrity of this barrier and the proper response to infection requires precise regulation of powerful immune homing signals such as tumor necrosis factor (TNF). Dysregulation of TNF leads to inflammatory bowel diseases (IBD), but the mechanism controlling the expression of this potent cytokine and the events that trigger the onset of chronic inflammation are unknown. Here, we show that loss of function of the epigenetic regulator ubiquitin-like protein containing PHD and RING finger domains 1 (uhrf1) in zebrafish leads to a reduction in tnfa promoter methylation and the induction of tnfa expression in intestinal epithelial cells (IECs). The increase in IEC tnfa levels is microbe-dependent and results in IEC shedding and apoptosis, immune cell recruitment, and barrier dysfunction, consistent with chronic inflammation. Importantly, tnfa knockdown in uhrf1 mutants restores IEC morphology, reduces cell shedding, and improves barrier function. We propose that loss of epigenetic repression and TNF induction in the intestinal epithelium can lead to IBD onset.


Assuntos
Metilação de DNA , Epigênese Genética/fisiologia , Doenças Inflamatórias Intestinais/metabolismo , Mucosa Intestinal/embriologia , Peixe-Zebra/embriologia , Animais , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Inflamação/genética , Inflamação/mortalidade , Inflamação/patologia , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/patologia , Mucosa Intestinal/patologia , Transativadores/genética , Transativadores/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
9.
Nature ; 517(7536): 612-5, 2015 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-25470057

RESUMO

Pathogenic mycobacteria induce the formation of complex cellular aggregates called granulomas that are the hallmark of tuberculosis. Here we examine the development and consequences of vascularization of the tuberculous granuloma in the zebrafish-Mycobacterium marinum infection model, which is characterized by organized granulomas with necrotic cores that bear striking resemblance to those of human tuberculosis. Using intravital microscopy in the transparent larval zebrafish, we show that granuloma formation is intimately associated with angiogenesis. The initiation of angiogenesis in turn coincides with the generation of local hypoxia and transcriptional induction of the canonical pro-angiogenic molecule Vegfaa. Pharmacological inhibition of the Vegf pathway suppresses granuloma-associated angiogenesis, reduces infection burden and limits dissemination. Moreover, anti-angiogenic therapies synergize with the first-line anti-tubercular antibiotic rifampicin, as well as with the antibiotic metronidazole, which targets hypoxic bacterial populations. Our data indicate that mycobacteria induce granuloma-associated angiogenesis, which promotes mycobacterial growth and increases spread of infection to new tissue sites. We propose the use of anti-angiogenic agents, now being used in cancer regimens, as a host-targeting tuberculosis therapy, particularly in extensively drug-resistant disease for which current antibiotic regimens are largely ineffective.


Assuntos
Inibidores da Angiogênese/farmacologia , Infecções por Mycobacterium não Tuberculosas/microbiologia , Mycobacterium marinum/efeitos dos fármacos , Mycobacterium marinum/crescimento & desenvolvimento , Neovascularização Patológica/microbiologia , Transdução de Sinais/efeitos dos fármacos , Peixe-Zebra/microbiologia , Inibidores da Angiogênese/uso terapêutico , Animais , Antibióticos Antituberculose/farmacologia , Carga Bacteriana/efeitos dos fármacos , Modelos Animais de Doenças , Sinergismo Farmacológico , Granuloma/tratamento farmacológico , Granuloma/metabolismo , Granuloma/microbiologia , Granuloma/patologia , Hipóxia/metabolismo , Hipóxia/microbiologia , Hipóxia/patologia , Larva/efeitos dos fármacos , Larva/microbiologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Macrófagos/patologia , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Infecções por Mycobacterium não Tuberculosas/metabolismo , Infecções por Mycobacterium não Tuberculosas/patologia , Mycobacterium marinum/patogenicidade , Neovascularização Patológica/tratamento farmacológico , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Tuberculose/tratamento farmacológico , Tuberculose/microbiologia , Tuberculose/patologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/metabolismo , Peixe-Zebra/crescimento & desenvolvimento
10.
PLoS One ; 4(10): e7618, 2009 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-19888420

RESUMO

Fragile X Syndrome is caused by the silencing of the Fragile X Mental Retardation gene (FMR1). Regulating dosage of FMR1 levels is critical for proper development and function of the nervous system and germ line, but the pathways responsible for maintaining normal expression levels are less clearly defined. Loss of Drosophila Fragile X protein (dFMR1) causes several behavioral and developmental defects in the fly, many of which are analogous to those seen in Fragile X patients. Over-expression of dFMR1 also causes specific neuronal and behavioral abnormalities. We have found that Argonaute2 (Ago2), the core component of the small interfering RNA (siRNA) pathway, regulates dfmr1 expression. Previously, the relationship between dFMR1 and Ago2 was defined by their physical interaction and co-regulation of downstream targets. We have found that Ago2 and dFMR1 are also connected through a regulatory relationship. Ago2 mediated repression of dFMR1 prevents axon growth and branching defects of the Drosophila neuromuscular junction (NMJ). Consequently, the neurogenesis defects in larvae mutant for both dfmr1 and Ago2 mirror those in dfmr1 null mutants. The Ago2 null phenotype at the NMJ is rescued in animals carrying an Ago2 genomic rescue construct. However, animals carrying a mutant Ago2 allele that produces Ago2 with significantly reduced endoribonuclease catalytic activity are normal with respect to the NMJ phenotypes examined. dFMR1 regulation by Ago2 is also observed in the germ line causing a multiple oocyte in a single egg chamber mutant phenotype. We have identified Ago2 as a regulator of dfmr1 expression and have clarified an important developmental role for Ago2 in the nervous system and germ line that requires dfmr1 function.


Assuntos
Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/genética , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Oogênese/fisiologia , Complexo de Inativação Induzido por RNA/fisiologia , Animais , Proteínas Argonautas , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Feminino , Proteína do X Frágil da Deficiência Intelectual/genética , Regulação da Expressão Gênica , Inativação Gênica , Microscopia de Fluorescência/métodos , Modelos Biológicos , Neurônios Motores/metabolismo , Sistema Nervoso/metabolismo , Neurônios/metabolismo , Fenótipo , RNA Interferente Pequeno/metabolismo
11.
Virology ; 386(1): 183-91, 2009 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-19201438

RESUMO

SV40 transforms cells through the action of two oncoproteins, large T antigen and small t antigen. Small t antigen targets phosphatase PP2A, while large T antigen stimulates cell proliferation and survival by action on multiple proteins, including the tumor suppressors Rb and p53. Large T antigen also binds components of the transcription initiation complex and several transcription factors. We examined global gene expression in SV40-transformed mouse embryo fibroblasts, and in enterocytes obtained from transgenic mice. SV40 transformation alters the expression of approximately 800 cellular genes in both systems. Much of this regulation is observed in both MEFs and enterocytes and is consistent with T antigen action on the Rb-E2F pathway. However, the regulation of many genes is cell-type specific, suggesting that unique signaling pathways are activated in different cell types upon transformation, and that the consequences of SV40 transformation depends on the type of cell targeted.


Assuntos
Antígenos Transformantes de Poliomavirus/fisiologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Vírus 40 dos Símios/fisiologia , Animais , Células Cultivadas , Enterócitos/virologia , Fibroblastos/virologia , Camundongos , Camundongos Transgênicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA