Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Vaccines (Basel) ; 9(5)2021 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-34070152

RESUMO

BACKGROUND: Persistent transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has given rise to a COVID-19 pandemic. Several vaccines, conceived in 2020, that evoke protective spike antibody responses are being deployed in mass public health vaccination programs. Recent data suggests, however, that as sequence variation in the spike genome accumulates, some vaccines may lose efficacy. METHODS: Using a macaque model of SARS-CoV-2 infection, we tested the efficacy of a peptide-based vaccine targeting MHC class I epitopes on the SARS-CoV-2 nucleocapsid protein. We administered biodegradable microspheres with synthetic peptides and adjuvants to rhesus macaques. Unvaccinated control and vaccinated macaques were challenged with 1 × 108 TCID50 units of SARS-CoV-2, followed by assessment of clinical symptoms and viral load, chest radiographs, and sampling of peripheral blood and bronchoalveolar lavage (BAL) fluid for downstream analysis. RESULTS: Vaccinated animals were free of pneumonia-like infiltrates characteristic of SARS-CoV-2 infection and presented with lower viral loads relative to controls. Gene expression in cells collected from BAL samples of vaccinated macaques revealed a unique signature associated with enhanced development of adaptive immune responses relative to control macaques. CONCLUSIONS: We demonstrate that a room temperature stable peptide vaccine based on known immunogenic HLA class I bound CTL epitopes from the nucleocapsid protein can provide protection against SARS-CoV-2 infection in nonhuman primates.

2.
Sci Rep ; 11(1): 699, 2021 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-33436708

RESUMO

Developing compact ion accelerators using intense lasers is a very active area of research, motivated by a strong applicative potential in science, industry and healthcare. However, proposed applications in medical therapy, as well as in nuclear and particle physics demand a strict control of ion energy, as well as of the angular and spectral distribution of ion beam, beyond the intrinsic limitations of the several acceleration mechanisms explored so far. Here we report on the production of highly collimated ([Formula: see text] half angle divergence), high-charge (10s of pC) and quasi-monoenergetic proton beams up to [Formula: see text] 50 MeV, using a recently developed method based on helical coil targetry. In this concept, ions accelerated from a laser-irradiated foil are post-accelerated and conditioned in a helical structure positioned at the rear of the foil. The pencil beam of protons was produced by guided post-acceleration at a rate of [Formula: see text] 2 GeV/m, without sacrificing the excellent beam emittance of the laser-driven proton beams. 3D particle tracing simulations indicate the possibility of sustaining high acceleration gradients over extended helical coil lengths, thus maximising the gain from such miniature accelerating modules.

3.
Viruses ; 12(2)2020 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-32033253

RESUMO

Currently, no rabies virus-specific antiviral drugs are available. Ranpirnase has strong antitumor and antiviral properties associated with its ribonuclease activity. TMR-001, a proprietary bulk drug substance solution of ranpirnase, was evaluated against rabies virus in three cell types: mouse neuroblastoma, BSR (baby hamster kidney cells), and bat primary fibroblast cells. When TMR-001 was added to cell monolayers 24 h preinfection, rabies virus release was inhibited for all cell types at three time points postinfection. TMR-001 treatment simultaneous with infection and 24 h postinfection effectively inhibited rabies virus release in the supernatant and cell-to-cell spread with 50% inhibitory concentrations of 0.2-2 nM and 20-600 nM, respectively. TMR-001 was administered at 0.1 mg/kg via intraperitoneal, intramuscular, or intravenous routes to Syrian hamsters beginning 24 h before a lethal rabies virus challenge and continuing once per day for up to 10 days. TMR-001 at this dose, formulation, and route of delivery did not prevent rabies virus transit from the periphery to the central nervous system in this model (n = 32). Further aspects of local controlled delivery of other active formulations or dose concentrations of TMR-001 or ribonuclease analogues should be investigated for this class of drugs as a rabies antiviral therapeutic.


Assuntos
Antivirais/farmacologia , Vírus da Raiva/efeitos dos fármacos , Ribonucleases/farmacologia , Liberação de Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Animais , Linhagem Celular , Células Cultivadas , Quirópteros , Cricetinae , Feminino , Fibroblastos/virologia , Mesocricetus , Camundongos , Raiva/prevenção & controle , Vírus da Raiva/fisiologia , Ribonucleases/administração & dosagem
4.
AIDS Res Hum Retroviruses ; 34(10): 838-848, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29936861

RESUMO

Ranpirnase (RNP) is a low molecular weight type III endoribonuclease, which demonstrates broad antiviral and antitumor properties. We sought to characterize the antiviral activity of RNP against HIV-1 and to determine whether RNP modulates local inflammatory changes associated with HIV infection in the colorectal explant model. Colorectal explants were incubated for 2 h with HIV-1BaL, in the presence of increasing concentrations of RNP (0-60 µg/mL). After washing, explants were cultured for 14 days, with supernatant collected at days 3, 7, 10, and 14. All samples were assayed for HIV-1 p24. Additionally, 30 soluble inflammatory biomarkers were assayed in the day 3 supernatant sample. Other biopsies were stimulated with lipopolysaccharides (LPS) (10 µg/mL) in the presence of RNP and soluble biomarkers assayed at day 3. RNP inhibited productive infection of the colorectal explants with HIV-1BaL and induced a dose-dependent decrease in 15/30 biomarkers. Affected biomarkers included IP-10, MDC, MIP-1α, MIP-1ß, TARC, IL12-p40, IL-15, IL-17, IL-1α, IL-7, IFNγ, IL12-p70, IL-1ß, IL-4, IL-5, and TNF-ß. Similarly, RNP dose-dependent inhibition was demonstrated in 7/30 biomarkers after LPS stimulation, all of which overlapped with HIV-1BaL-induced biomarker changes. The ability of RNP to inhibit both colorectal explant HIV-1BaL infection and inflammatory changes associated with HIV-1 infection makes RPN a promising agent for topical rectal pre-exposure prophylaxis.


Assuntos
Fármacos Anti-HIV/farmacologia , Infecções por HIV/prevenção & controle , Mediadores da Inflamação/metabolismo , Ribonucleases/farmacologia , Biomarcadores/metabolismo , Colo/patologia , Colo/virologia , Relação Dose-Resposta a Droga , Infecções por HIV/metabolismo , Infecções por HIV/patologia , HIV-1/fisiologia , Voluntários Saudáveis , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Mucosa Intestinal/virologia , Modelos Biológicos , Cultura Primária de Células , Reto/patologia , Reto/virologia
5.
Antivir Ther ; 22(3): 247-255, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28121292

RESUMO

BACKGROUND: Human papillomaviruses (HPV), the causative agents of anogenital warts, are the most prevalent sexually transmitted infectious agents, and wart treatment poses a persistent challenge. We assessed the safety and efficacy of treating HPV with ranpirnase, an endoribonuclease from the northern leopard frog that has been used extensively in Phase III oncology trials. METHODS: As initial verification of ranpirnase antiviral activity, we assessed its ability to eliminate papillomaviruses in cultured cells. To further assess its feasibility for treating anogenital warts in humans, we performed a Phase I study. Forty-two male volunteers with genital/perianal warts were treated topically with three different formulations of 1 mg/ml ranpirnase. Patients were monitored for 8 weeks or until healing. Four patients with HIV were treated in accordance with the compassionate programme but were not evaluated. RESULTS: In cultured cells, ranpirnase showed specific activity against HPV-11 with low toxicity (selectivity index >88). The broad applicability of ranpirnase for treating papillomaviruses was verified using the cottontail rabbit papillomavirus. In the clinical study, eight participants were lost-to-follow-up or discontinued due to protocol violation or non-compliance. Among 30 evaluable participants, topical ranpirnase was moderately well-tolerated, with discontinuation by 5 (16.7%) due to adverse reactions. Clinical healing was achieved by 25 participants (83.3%) and 50% improvement by the 5 discontinued participants (16.7%). The median time to clinical healing was 30 days. CONCLUSIONS: This study provides the first in vitro and clinical evidence of the antiviral efficacy of ranpirnase against HPV and supports assessment of ranpirnase in expanded clinical studies.


Assuntos
Condiloma Acuminado/tratamento farmacológico , Condiloma Acuminado/virologia , Papillomaviridae/efeitos dos fármacos , Infecções por Papillomavirus/tratamento farmacológico , Infecções por Papillomavirus/virologia , Ribonucleases/uso terapêutico , Administração Tópica , Adulto , Animais , Linhagem Celular , Células Cultivadas , Terapia Combinada , Condiloma Acuminado/patologia , Relação Dose-Resposta a Droga , Humanos , Kappapapillomavirus/efeitos dos fármacos , Kappapapillomavirus/genética , Masculino , Camundongos , Pessoa de Meia-Idade , Papillomaviridae/classificação , Papillomaviridae/genética , Infecções por Papillomavirus/patologia , Coelhos , Ribonucleases/farmacologia , Resultado do Tratamento , Adulto Jovem
6.
Stem Cells Dev ; 25(22): 1691-1697, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27627457

RESUMO

The explosive spread of the Zika virus (ZIKV) through South and Central America has been linked to an increase in congenital birth defects, specifically microcephaly. Representative rodent models for investigating infections include direct central nervous system (CNS) injections late in pregnancy and transplacental transmission in immunodeficient mice. Microcephaly in humans may be the result of infection occurring early in pregnancy, therefore recapitulating that the human course of ZIKV infection should include normal embryo exposed to ZIKV during the first trimester. In ovo development of the chicken embryo closely mirrors human fetal neurodevelopment and, as a comparative model, could provide key insights into both temporal and pathophysiological effects of ZIKV. Chick embryos were directly infected early and throughout incubation with ZIKV isolated from a Mexican mosquito in January 2016. High doses of virus caused embryonic lethality. In a subset of lower dosed embryos, replicating ZIKV was present in various organs, including the CNS, throughout development. Surviving ZIKV-infected embryos presented a microcephaly-like phenotype. Chick embryos were longitudinally monitored by magnetic resonance imaging that documented CNS structural malformations, including enlarged ventricles (30% increase) and stunted cortical growth (decreased telencephalon by 18%, brain stem by 32%, and total brain volume by 18%), on both embryonic day 15 (E15) and E20 of development. ZIKV-induced microcephaly was observed with inoculations of as few as 2-20 viral particles. The chick embryo model presented ZIKV embryonic lethal effects and progressive CNS damage similar to microcephaly.


Assuntos
Microcefalia/patologia , Microcefalia/virologia , Infecção por Zika virus/patologia , Zika virus/fisiologia , Animais , Encéfalo/embriologia , Encéfalo/patologia , Encéfalo/virologia , Embrião de Galinha , Imageamento por Ressonância Magnética
7.
Antiviral Res ; 132: 210-8, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27350309

RESUMO

The recent epidemic of Ebola has intensified the need for the development of novel antiviral therapeutics that prolong and improve survival against deadly viral diseases. We sought to determine whether ranpirnase, an endoribonuclease from Rana pipiens with a demonstrated human safety profile in phase III oncology trials, can reduce titers of Ebola virus (EBOV) in infected cells, protect mice against mouse-adapted EBOV challenge, and reduce virus levels in infected mice. Our results demonstrate that 0.50 µg/ml ranpirnase is potently effective at reducing EBOV Zaire Kikwit infection in cultured Vero E6 cells (Selectivity Index 47.8-70.2). In a prophylactic study, a single intravenous dose of 0.1 mg/kg ranpirnase protected 70% of mice from progressive infection. Additionally, in a post-exposure prophylactic study, 100% of female mice survived infection after intraperitoneal administration of 0.1 mg/kg ranpirnase for ten days beginning 1 h post challenge. Most of the male counterparts were sacrificed due to weight loss by Study Day 8 or 9; however, the Clinical Activity/Behavior scores of these mice remained low and no significant microscopic pathologies could be detected in the kidneys, livers or spleens. Furthermore, live virus could not be detected in the sera of ranpirnase-treated mice by Study Day 8 or in the kidneys, livers or spleens by Study Day 12, and viral RNA levels declined exponentially by Study Day 12. Because ranpirnase is exceptionally stable and has a long track record of safe intravenous administration to humans, this drug provides a promising new candidate for clinical consideration in the treatment of Ebola virus disease alone or in combination with other therapeutics.


Assuntos
Antivirais/farmacologia , Ebolavirus/efeitos dos fármacos , Doença pelo Vírus Ebola/virologia , Ribonucleases/farmacologia , Animais , Linhagem Celular , Chlorocebus aethiops , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ebolavirus/fisiologia , Feminino , Doença pelo Vírus Ebola/tratamento farmacológico , Doença pelo Vírus Ebola/mortalidade , Humanos , Camundongos , RNA Viral , Células Vero , Carga Viral , Replicação Viral/efeitos dos fármacos
8.
Virology ; 454-455: 60-6, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24725932

RESUMO

Previously, we showed that ADAM10 is necessary for HIV-1 replication in primary human macrophages and immortalized cell lines. Silencing ADAM10 expression interrupted the HIV-1 life cycle prior to nuclear translocation of viral cDNA. Furthermore, our data indicated that HIV-1 replication depends on the expression of ADAM15 and γ-secretase, which proteolytically processes ADAM10. Silencing ADAM15 or γ-secretase expression inhibits HIV-1 replication between reverse transcription and nuclear entry. Here, we show that ADAM10 expression also supports replication in CD4(+) T lymphocytes. The intracellular domain (ICD) of ADAM10 associates with the HIV-1 pre-integration complex (PIC) in the cytoplasm and immunoprecipitates and co-localizes with HIV-1 integrase, a key component of PIC. Taken together, our data support a model whereby ADAM15/γ-secretase processing of ADAM10 releases the ICD, which then incorporates into HIV-1 PIC to facilitate nuclear trafficking. Thus, these studies suggest ADAM10 as a novel therapeutic target for inhibiting HIV-1 prior to nuclear entry.


Assuntos
Proteínas ADAM/metabolismo , Transporte Ativo do Núcleo Celular , Secretases da Proteína Precursora do Amiloide/metabolismo , HIV-1/fisiologia , Interações Hospedeiro-Patógeno , Substâncias Macromoleculares/metabolismo , Proteínas de Membrana/metabolismo , Integração Viral , Proteína ADAM10 , Células Cultivadas , DNA Viral/metabolismo , Humanos , Imunoprecipitação , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Virais/metabolismo
9.
AIDS Res Hum Retroviruses ; 28(10): 1329-39, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22404213

RESUMO

Cellular proteins are essential for human immunodeficiency virus type 1 (HIV-1) replication and may serve as viable new targets for treating infection. Using gene trap insertional mutagenesis, a high-throughput approach based on random inactivation of cellular genes, candidate genes were found that limit virus replication when mutated. Disrupted genes (N=87) conferring resistance to lytic infection with several viruses were queried for an affect on HIV-1 replication by utilizing small interfering RNA (siRNA) screens in TZM-bl cells. Several genes regulating diverse pathways were found to be required for HIV-1 replication, including DHX8, DNAJA1, GTF2E1, GTF2E2, HAP1, KALRN, UBA3, UBE2E3, and VMP1. Candidate genes were independently tested in primary human macrophages, toxicity assays, and/or Tat-dependent ß-galactosidase reporter assays. Bioinformatics analyses indicated that several host factors present in this study participate in canonical pathways and functional processes implicated in prior genome-wide studies. However, the genes presented in this study did not share identity with those found previously. Novel antiviral targets identified in this study should open new avenues for mechanistic investigation.


Assuntos
HIV-1/isolamento & purificação , Mutagênese Insercional/genética , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Ativação Viral/genética , Replicação Viral/genética , Linhagem Celular , Células Cultivadas , Feminino , Regulação da Expressão Gênica/genética , HIV-1/fisiologia , Humanos , Masculino , Programas de Rastreamento , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , RNA Viral/genética , Ativação Viral/fisiologia , Replicação Viral/fisiologia
10.
Antivir Chem Chemother ; 22(5): 205-15, 2012 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-22374988

RESUMO

BACKGROUND: Host genes serving potential roles in virus replication may be exploited as novel antiviral targets. METHODS: Small interfering RNA (siRNA)-mediated knockdown of host gene expression was used to validate candidate genes in screens against six unrelated viruses, most importantly influenza. A mouse model of influenza A virus infection was used to evaluate the efficacy of a candidate FDA-approved drug identified in the screening effort. RESULTS: Several genes in the PI3K-AKT-mTOR pathway were found to support broad-spectrum viral replication in vitro by RNA interference. This led to the discovery that everolimus, an mTOR inhibitor, showed in vitro antiviral activity against cowpox, dengue type 2, influenza A, rhino- and respiratory syncytial viruses. In a lethal mouse infection model of influenza A (H1N1 and H5N1) virus infection, everolimus treatment (1 mg/kg/day) significantly delayed death but could not prevent mortality. Fourteen days of treatment was more beneficial in delaying the time to death than treatment for seven days. Pathological findings in everolimus-treated mice showed reduced lung haemorrhage and lung weights in response to infection. CONCLUSIONS: These results provide proof of concept that cellular targets can be identified by gene knockout methods, and highlight the importance of the PI3K-AKT-mTOR pathway in supporting viral infections.


Assuntos
Antivirais/farmacologia , Vírus da Influenza A Subtipo H1N1/fisiologia , Virus da Influenza A Subtipo H5N1/fisiologia , Mutagênese Insercional , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Serina-Treonina Quinases TOR/antagonistas & inibidores , Replicação Viral/efeitos dos fármacos , Animais , Linhagem Celular , Everolimo , Feminino , Humanos , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Virus da Influenza A Subtipo H5N1/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Testes de Sensibilidade Microbiana , Oseltamivir/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribavirina/farmacologia , Sirolimo/análogos & derivados , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo
11.
Retrovirology ; 8: 32, 2011 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-21569301

RESUMO

BACKGROUND: Gene trap insertional mutagenesis was used as a high-throughput approach to discover cellular genes participating in viral infection by screening libraries of cells selected for survival from lytic infection with a variety of viruses. Cells harboring a disrupted ADAM10 (A Disintegrin and Metalloprotease 10) allele survived reovirus infection, and subsequently ADAM10 was shown by RNA interference to be important for replication of HIV-1. RESULTS: Silencing ADAM10 expression with small interfering RNA (siRNA) 48 hours before infection significantly inhibited HIV-1 replication in primary human monocyte-derived macrophages and in CD4⁺ cell lines. In agreement, ADAM10 over-expression significantly increased HIV-1 replication. ADAM10 down-regulation did not inhibit viral reverse transcription, indicating that viral entry and uncoating are also independent of ADAM10 expression. Integration of HIV-1 cDNA was reduced in ADAM10 down-regulated cells; however, concomitant 2-LTR circle formation was not detected, suggesting that HIV-1 does not enter the nucleus. Further, ADAM10 silencing inhibited downstream reporter gene expression and viral protein translation. Interestingly, we found that while the metalloprotease domain of ADAM10 is not required for HIV-1 replication, ADAM15 and γ-secretase (which proteolytically release the extracellular and intracellular domains of ADAM10 from the plasma membrane, respectively) do support productive infection. CONCLUSIONS: We propose that ADAM10 facilitates replication at the level of nuclear trafficking. Collectively, our data support a model whereby ADAM10 is cleaved by ADAM15 and γ-secretase and that the ADAM10 intracellular domain directly facilitates HIV-1 nuclear trafficking. Thus, ADAM10 represents a novel cellular target class for development of antiretroviral drugs.


Assuntos
Proteínas ADAM/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , HIV-1/patogenicidade , Interações Hospedeiro-Patógeno , Proteínas de Membrana/metabolismo , Replicação Viral , Proteína ADAM10 , Transporte Ativo do Núcleo Celular , Células Cultivadas , HIV-1/fisiologia , Humanos , Macrófagos/virologia , Modelos Biológicos , Mutagênese Insercional , Integração Viral
12.
Virology ; 379(2): 191-6, 2008 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-18682304

RESUMO

HIV infection typically involves interaction of Env with CD4 and a chemokine coreceptor, either CCR5 or CXCR4. Other cellular factors supporting HIV replication have also been characterized. We previously demonstrated a role for CD63 in early HIV infection events in macrophages via inhibition by anti-CD63 antibody pretreatment. To confirm the requirement for CD63 in HIV replication, we decreased CD63 expression using CD63-specific short interfering RNAs (siRNA), and showed inhibition of HIV replication in macrophages. Surprisingly, pretreatment with CD63 siRNA not only silenced CD63 expression by 90%, but also inhibited HIV-1 replication in a cultured cell line (U373-MAGI) which had been previously shown to be insensitive to CD63 monoclonal antibody inhibition. Although the anti-CD63 antibody was previously shown to inhibit early HIV infection events only in macrophages, we now show a potential role for CD63 in later HIV replication events in macrophages and cell lines. Further delineation of the role of CD63 in HIV replication may lead to development of novel therapeutic compounds.


Assuntos
Antígenos CD/fisiologia , HIV-1/fisiologia , Glicoproteínas da Membrana de Plaquetas/fisiologia , Replicação Viral/fisiologia , Antígenos CD/genética , Linhagem Celular , Células Cultivadas , Regulação para Baixo , HIV-1/imunologia , Humanos , Macrófagos/imunologia , Macrófagos/virologia , Glicoproteínas da Membrana de Plaquetas/antagonistas & inibidores , Glicoproteínas da Membrana de Plaquetas/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Tetraspanina 30 , Transfecção , Replicação Viral/imunologia
13.
J Virol ; 81(9): 4881-5, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17301152

RESUMO

The mechanisms of cellular entry of dengue and West Nile viruses are not well characterized. We show that both these viruses enter HeLa cells by clathrin-dependent endocytosis and require vacuolar acidic pH. Inhibition of the GTPase Rab 5 or 7, which regulates transport to early or late endosomes, respectively, demonstrated that Rab 5 was essential for survival of both dengue and West Nile virus. These data broaden our understanding of the pathways required for productive dengue and West Nile virus infection and may facilitate new strategies for combating disease.


Assuntos
Vírus da Dengue/metabolismo , Internalização do Vírus , Vírus do Nilo Ocidental/metabolismo , Proteínas rab5 de Ligação ao GTP/metabolismo , Endocitose/fisiologia , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Concentração de Íons de Hidrogênio , Interferência de RNA , Vacúolos/química , Vacúolos/metabolismo , Proteínas rab5 de Ligação ao GTP/genética
14.
J Vet Med Educ ; 34(4): 485-91, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18287477

RESUMO

This article describes the ECVP/ESVP Summer School in Veterinary Pathology, a new annual two-week European training facility established by the European College of Veterinary Pathologists (ECVP) in collaboration with the European Society of Veterinary Pathology (ESVP). The aim of the Summer Schools is to provide Europe-wide, harmonized, top-standard theoretical and practical post-graduate training for veterinarians specializing in veterinary pathology. In particular, it aims to support trainees in veterinary pathology in their individual preparation for the ECVP certifying examination. Ultimately, it aims to provide young pathologists with the skills and knowledge necessary to participate in international, high-quality research and the tools for applying international standards to their own research and for independent study for the ECVP certifying examination, even if they do not work in comparable academic environments and do not have the same level of local support and training. The ECVP/ESVP Summer Schools take place in European countries, with local organization from a university department of veterinary pathology. Each event comprises modules provided by internationally recognized specialists in their specific fields of expertise on different organ systems, diseases of specific species, specific techniques, and specific topics relevant to pathology, forming a cycle of four events to cover all major topics. Every two years a mock exam is organized as a tool to monitor individual progress in preparing for the ECVP certifying examination.


Assuntos
Competência Clínica , Educação em Veterinária/normas , Cooperação Internacional , Patologia Veterinária/educação , Medicina Veterinária/normas , Animais , Certificação , Europa (Continente) , Humanos , Pesquisa/educação
15.
Traffic ; 7(4): 379-93, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16536737

RESUMO

Receptor-mediated internalization to the endoplasmic reticulum (ER) and subsequent retro-translocation to the cytosol are essential sequential processes required for the intoxication of mammalian cells by Pseudomonas exotoxin A (PEx). The toxin binds the alpha2-macroglobulin receptor/low-density lipoprotein receptor-related protein. Here, we show that in HeLa cells, PEx recruits a proportion of this receptor to detergent-resistant microdomains (DRMs). Uptake of receptor-bound PEx involves transport steps both directly from early endosomes to the trans-Golgi network (TGN) independently of Rab9 function and from late endosomes to the TGN in a Rab9-dependent manner. Furthermore, treatments that simultaneously perturb both Arf1-dependent and Rab6-dependent retrograde pathways show that PEx can use multiple routes to reach the ER. The Rab6-dependent route has only been described previously for cargo with lipid-sorting signals. These findings suggest that partial localization of PEx within DRM permits a choice of trafficking routes consistent with a model that DRM-associated toxins reach the ER on a lipid-dependent sorting pathway whilst non-DRM-associated PEx exploits the previously characterized KDEL receptor-mediated uptake pathway. Thus, unexpectedly, an ER-directed toxin with a proteinaceous receptor shows promiscuity in its intracellular trafficking pathways, exploiting routes controlled by both lipid- and protein-sorting signals.


Assuntos
ADP Ribose Transferases/metabolismo , Toxinas Bacterianas/metabolismo , Retículo Endoplasmático/metabolismo , Exotoxinas/metabolismo , Fatores de Virulência/metabolismo , Células 3T3 , Animais , Chlorocebus aethiops , Complexo de Golgi/metabolismo , Células HeLa , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Microdomínios da Membrana/metabolismo , Camundongos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores de Peptídeos/metabolismo , Transdução de Sinais/fisiologia , Sintaxina 16/genética , Sintaxina 16/metabolismo , Células Vero , Proteínas rab de Ligação ao GTP/metabolismo , Exotoxina A de Pseudomonas aeruginosa
16.
J Virol ; 79(18): 11742-51, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16140752

RESUMO

Rab proteins and their effectors facilitate vesicular transport by tethering donor vesicles to their respective target membranes. By using gene trap insertional mutagenesis, we identified Rab9, which mediates late-endosome-to-trans-Golgi-network trafficking, among several candidate host genes whose disruption allowed the survival of Marburg virus-infected cells, suggesting that Rab9 is utilized in Marburg replication. Although Rab9 has not been implicated in human immunodeficiency virus (HIV) replication, previous reports suggested that the late endosome is an initiation site for HIV assembly and that TIP47-dependent trafficking out of the late endosome to the trans-Golgi network facilitates the sorting of HIV Env into virions budding at the plasma membrane. We examined the role of Rab9 in the life cycles of HIV and several unrelated viruses, using small interfering RNA (siRNA) to silence Rab9 expression before viral infection. Silencing Rab9 expression dramatically inhibited HIV replication, as did silencing the host genes encoding TIP47, p40, and PIKfyve, which also facilitate late-endosome-to-trans-Golgi vesicular transport. In addition, silencing studies revealed that HIV replication was dependent on the expression of Rab11A, which mediates trans-Golgi-to-plasma-membrane transport, and that increased HIV Gag was sequestered in a CD63+ endocytic compartment in a cell line stably expressing Rab9 siRNA. Replication of the enveloped Ebola, Marburg, and measles viruses was inhibited with Rab9 siRNA, although the non-enveloped reovirus was insensitive to Rab9 silencing. These results suggest that Rab9 is an important cellular target for inhibiting diverse viruses and help to define a late-endosome-to-plasma-membrane vesicular transport pathway important in viral assembly.


Assuntos
Filoviridae/fisiologia , HIV-1/fisiologia , Vírus do Sarampo/fisiologia , Replicação Viral/fisiologia , Proteínas rab de Ligação ao GTP/fisiologia , Animais , Linhagem Celular , Chlorocebus aethiops , Cães , Ebolavirus/genética , Ebolavirus/fisiologia , Filoviridae/genética , Produtos do Gene gag/metabolismo , HIV-1/genética , Humanos , Marburgvirus/genética , Marburgvirus/fisiologia , Vírus do Sarampo/genética , Modelos Biológicos , Mutagênese , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Viral/genética , RNA Viral/metabolismo , Ratos , Células Vero , Replicação Viral/genética , Proteínas rab de Ligação ao GTP/genética
17.
Clin Infect Dis ; 40(10): 1481-91, 2005 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15844071

RESUMO

BACKGROUND: The occurrence of acquired rifamycin resistance despite use of directly observed therapy for tuberculosis is associated with advanced human immunodeficiency virus (HIV) disease and highly intermittent administration of antituberculosis drugs. Beyond these associations, the pathogenesis of acquired rifamycin resistance is unknown. METHODS: We performed a pharmacokinetic substudy of patients in a trial of treatment with twice-weekly rifabutin and isoniazid. RESULTS: A total of 102 (60%) of 169 patients in the treatment trial participated in the pharmacokinetic substudy, including 7 of 8 patients in whom tuberculosis treatment failure or relapse occurred in association with acquired rifamycin-resistant mycobacteria (hereafter, "ARR failure or relapse"). The median rifabutin area under the concentration-time curve (AUC(0-24)) was lower for patients with than for patients without ARR failure or relapse (3.3 vs. 5.2 microg*h/mL; P = .06, by the Mann-Whitney exact test). In a multivariate analysis adjusted for CD4+ T cell count, the mean rifabutin AUC(0-24) was significantly lower for patients with ARR failure or relapse than for other patients (3.0 microg*h/mL [95% confidence interval {CI}, 1.9-4.5] vs. 5.2 microg*h/mL [95% CI, 4.6-5.8]; P = .02, by analysis of covariance). The median isoniazid AUC(0-12) was not significantly associated with ARR failure or relapse (20.6 vs. 28.0 microg*h/mL; P = .24, by the Mann-Whitney exact test). However, in a multivariate logistic regression model that adjusted for the rifabutin AUC(0-24), a lower isoniazid AUC(0-12) was associated with ARR failure or relapse (OR, 10.5; 95% CI, 1.1-100; P = .04). CONCLUSIONS: Lower plasma concentrations of rifabutin and, perhaps, isoniazid were associated with ARR failure or relapse in patients with tuberculosis and HIV infection treated with twice-weekly therapy.


Assuntos
Farmacorresistência Bacteriana , Infecções por HIV/complicações , Isoniazida/farmacocinética , Rifabutina/farmacocinética , Rifamicinas/farmacologia , Tuberculose/complicações , Tuberculose/tratamento farmacológico , Adulto , Antituberculosos/farmacocinética , Antituberculosos/uso terapêutico , Área Sob a Curva , Contagem de Linfócito CD4 , Terapia Diretamente Observada , Feminino , Humanos , Isoniazida/sangue , Isoniazida/uso terapêutico , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Mycobacterium tuberculosis/efeitos dos fármacos , Recidiva , Rifabutina/sangue , Rifabutina/uso terapêutico , Fatores de Risco , Falha de Tratamento
18.
AIDS ; 18(9): 1281-9, 2004 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-15362660

RESUMO

OBJECTIVE: When children acquire HIV infection from their mothers (with whom they share at least 50% of their HLA alleles), they acquire virus with a history of encounter with maternal HLA-mediated immune responses. We investigated whether maternal HLA selection pressures on the virus would adversely influence clinical outcomes of HIV-infected children. METHODS: We tested whether time to AIDS diagnosis or death, among a cohort of 59 HIV-infected children in New York City followed from birth for up to 12 years, was associated with maternally- or paternally-inherited child HLA class I alleles, and with HLA similarity between mother and child. RESULTS: HIV-infected children with an HLA allele usually associated with slow disease experienced a slower progression to AIDS or death only if the allele was paternally inherited. If the allele was present in the mother, no association was observed. Children who were homozygous or who shared both alleles with their mothers at more than one HLA class I locus were more likely to progress to AIDS or death than other children (relative hazard, 3.46; 95% confidence interval, 1.24-9.71). CONCLUSION: Genetic similarity between mother and child may compromise the child's capacity to control HIV replication when the virus is acquired from the mother. HLA-mediated selective pressures on the virus in a transmitting mother-infant pair may undermine future HLA-mediated viral control in the child.


Assuntos
Pai , Genes MHC Classe I , Infecções por HIV/genética , Mães , Síndrome da Imunodeficiência Adquirida/genética , Síndrome da Imunodeficiência Adquirida/mortalidade , Alelos , Criança , Pré-Escolar , Progressão da Doença , Feminino , Infecções por HIV/mortalidade , Infecções por HIV/transmissão , Homozigoto , Humanos , Lactente , Recém-Nascido , Transmissão Vertical de Doenças Infecciosas , Masculino , Prognóstico , Modelos de Riscos Proporcionais , Taxa de Sobrevida , Carga Viral
19.
J Acquir Immune Defic Syndr ; 37(2): 1318-23, 2004 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15385741

RESUMO

The loss of viral control by the host may be due to the evolution of viruses with mutations that limit presentation by human leukocyte antigen (HLA) to cytotoxic T cells. The authors hypothesized that the consequence of such evolution might be that persons with common HLA class I alleles would be less able to control viremia, on average, than would those with rare alleles. HLA class I typing was completed for 128 injection drug users who seroconverted in a prospective cohort study in Bangkok, Thailand. Logistic regression was used to model viral load (greater than or equal to the median) at 9 and 12 months after seroconversion with an HLA score that profiled the relative prevalence of each individual's alleles. At 12 months after seroconversion, injection drug users with the most common HLA alleles (highest quartile HLA score) had an almost 4-fold increased risk for higher viral load (> or = 32,055 copies/mL) than injection drug users with less common HLA alleles (adjusted odds ratio, 3.92; 95% confidence interval, 1.3-11.8). These findings support the importance of frequency-dependent effects of host genes on HIV type 1 evolution in different populations and suggest that HLA-driven viral evolution critically influences control of viremia in early HIV type 1 infection.


Assuntos
Genes MHC Classe I/genética , Anticorpos Anti-HIV/sangue , Infecções por HIV/genética , HIV-1/fisiologia , Antígenos HLA-B/genética , Abuso de Substâncias por Via Intravenosa/complicações , Alelos , Infecções por HIV/virologia , Soropositividade para HIV , HIV-1/genética , HIV-1/imunologia , Haplótipos , Humanos , Tailândia , Carga Viral
20.
J Biol Chem ; 279(38): 40204-8, 2004 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-15263003

RESUMO

Rab GTPases and their effectors facilitate vesicular transport by tethering donor vesicles to their respective target membranes. Rab9 mediates late endosome to trans-Golgi transport and has recently been found to be a key cellular component for human immunodeficiency virus-1, Ebola, Marburg, and measles virus replication, suggesting that it may be a novel target in the development of broad spectrum antiviral drugs. As part of our structure-based drug design program, we have determined the crystal structure of a C-terminally truncated human Rab9 (residues 1-177) to 1.25-A resolution. The overall structure shows a characteristic nucleotide binding fold consisting of a six-stranded beta-sheet surrounded by five alpha-helices with a tightly bound GDP molecule in the active site. Structure-based sequence alignment of Rab9 with other Rab proteins reveals that its active site consists of residues highly conserved in the Rab GTPase family, implying a common catalytic mechanism. However, Rab9 contains seven regions that are significantly different in conformation from other Rab proteins. Some of those regions coincide with putative effector-binding sites and switch I and switch II regions identified by structure/sequence alignments. The Rab9 structure at near atomic resolution provides an excellent model for structure-based antiviral drug design.


Assuntos
Proteínas rab de Ligação ao GTP/química , Sequência de Aminoácidos , Sítios de Ligação , Cristalografia , Guanosina Difosfato/metabolismo , Humanos , Ligação de Hidrogênio , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas rab de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA