Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ACS Infect Dis ; 10(5): 1780-1792, 2024 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-38651692

RESUMO

The recent COVID-19 pandemic underscored the limitations of currently available direct-acting antiviral treatments against acute respiratory RNA-viral infections and stimulated major research initiatives targeting anticoronavirus agents. Two novel nsp5 protease (MPro) inhibitors have been approved, nirmatrelvir and ensitrelvir, along with two existing nucleos(t)ide analogues repurposed as nsp12 polymerase inhibitors, remdesivir and molnupiravir, but a need still exists for therapies with improved potency and systemic exposure with oral dosing, better metabolic stability, and reduced resistance and toxicity risks. Herein, we summarize our research toward identifying nsp12 inhibitors that led to nucleoside analogues 10e and 10n, which showed favorable pan-coronavirus activity in cell-infection screens, were metabolized to active triphosphate nucleotides in cell-incubation studies, and demonstrated target (nsp12) engagement in biochemical assays.


Assuntos
Antivirais , Tratamento Farmacológico da COVID-19 , Nucleosídeos , SARS-CoV-2 , Antivirais/farmacologia , Antivirais/química , SARS-CoV-2/efeitos dos fármacos , Humanos , Nucleosídeos/farmacologia , Nucleosídeos/química , Animais , Descoberta de Drogas , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas não Estruturais Virais/metabolismo , Chlorocebus aethiops , Células Vero , COVID-19/virologia , RNA-Polimerase RNA-Dependente de Coronavírus
2.
Viruses ; 16(3)2024 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-38543689

RESUMO

HBV RNA destabilizers are a class of small-molecule compounds that target the noncanonical poly(A) RNA polymerases PAPD5 and PAPD7, resulting in HBV RNA degradation and the suppression of viral proteins including the hepatitis B surface antigen (HBsAg). AB-161 is a next-generation HBV RNA destabilizer with potent antiviral activity, inhibiting HBsAg expressed from cccDNA and integrated HBV DNA in HBV cell-based models. AB-161 exhibits broad HBV genotype coverage, maintains activity against variants resistant to nucleoside analogs, and shows additive effects on HBV replication when combined with other classes of HBV inhibitors. In AAV-HBV-transduced mice, the dose-dependent reduction of HBsAg correlated with concentrations of AB-161 in the liver reaching above its effective concentration mediating 90% inhibition (EC90), compared to concentrations in plasma which were substantially below its EC90, indicating that high liver exposure drives antiviral activities. In preclinical 13-week safety studies, minor non-adverse delays in sensory nerve conductance velocity were noted in the high-dose groups in rats and dogs. However, all nerve conduction metrics remained within physiologically normal ranges, with no neurobehavioral or histopathological findings. Despite the improved neurotoxicity profile, microscopic findings associated with male reproductive toxicity were detected in dogs, which subsequently led to the discontinuation of AB-161's clinical development.


Assuntos
Complexos de Coordenação , Vírus da Hepatite B , Hepatite B Crônica , Naftalenossulfonatos , Masculino , Camundongos , Ratos , Animais , Cães , Vírus da Hepatite B/fisiologia , Antígenos de Superfície da Hepatite B/genética , RNA Viral , RNA Mensageiro , Antivirais/farmacologia , Antivirais/uso terapêutico , DNA Viral/genética , Hepatite B Crônica/tratamento farmacológico , DNA Circular
3.
Bioorg Med Chem Lett ; 94: 129456, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37633618

RESUMO

Disruption of the HBV capsid assembly process through small-molecule interaction with HBV core protein is a validated target for the suppression of hepatitis B viral replication and the development of new antivirals. Through combination of key structural features associated with two distinct series of capsid assembly modulators, a novel aminochroman-based chemotype was identified. Optimization of anti-HBV potency through generation of SAR in addition to further core modifications provided a series of related functionalized aminoindanes. Key compounds demonstrated excellent cellular potency in addition to favorable ADME and pharmacokinetic profiles and were shown to be highly efficacious in a mouse model of HBV replication. Aminoindane derivative AB-506 was subsequently advanced into clinical development.


Assuntos
Antivirais , Proteínas do Capsídeo , Capsídeo , Animais , Camundongos , Antivirais/farmacologia , Modelos Animais de Doenças , Relação Estrutura-Atividade , Vírus da Hepatite B/efeitos dos fármacos , Vírus da Hepatite B/metabolismo
4.
Hepatol Commun ; 7(4)2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36930867

RESUMO

BACKGROUND: HBV pregenomic RNA (pgRNA) is a circulating biomarker for covalently closed circular DNA activity in HBV-infected individuals and has been studied for treatment efficacy, disease staging, and off-therapy outcomes; however, data on the stability are scarce. Increasing HBV pgRNA assay sensitivity may improve its predictive value and provide additional insights at low viral levels. METHODS: Modifications to a fully automated first (v1) generation HBV pgRNA assay improved sensitivity up to 15-fold over the previous assay. Flexible sample input volumes yielded lower limits of quantitation of 10 and 22 copies/mL for 0.6 and 0.2 mL assays, respectively. Results are standardized to secondary standards that are traceable to the WHO HBV DNA standard, and internal and external controls are included. RESULTS: Comparison between v1 and modified v2 assays showed increased sensitivity from 152 copies/mL with v1 to 10 (0.6 mL) and 22 (0.2 mL) copies/mL with v2, respectively. Quantitated v2 results were indistinguishable from v1, indicating that comparisons can be made to previous studies. Single timepoint treatment-naive blood donors or longitudinal draws from patients with chronic hepatitis B on AB-729, an investigational siRNA therapy, showed improved detection and quantifiable pgRNA with v2 compared with v1. Stability testing demonstrated excellent HBV pgRNA plasma stability after 3 freeze-thaw cycles, for at least 7 days at 25-37 °C and at least 30 days at 4°C, with ≤0.25 Log U/mL decrease. CONCLUSION: HBV pgRNA v2 assays with increased sensitivity and flexible input volumes demonstrated increased detection and quantitation of low viral titer samples. Highly sensitive HBV pgRNA assays may be useful in refining predictive treatment outcomes based on this marker. HBV pgRNA was stable under multiple conditions, which increases the reliability of this marker.


Assuntos
Vírus da Hepatite B , Hepatite B , Humanos , Vírus da Hepatite B/genética , Reprodutibilidade dos Testes , RNA Viral/genética
5.
Hepatol Commun ; 6(12): 3457-3472, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36194181

RESUMO

AB-506 is a potent, pan-genotypic small molecule capsid inhibitor that inhibits hepatitis B virus (HBV) pregenomic RNA encapsidation. We assessed the safety, pharmacokinetics, and antiviral activity of AB-506 in two randomized, double-blinded Phase 1 studies in healthy subjects (HS) and subjects with chronic HBV infection (CHB). Single ascending and multiple doses of AB-506 or placebo (30-1000 mg or 400 mg daily for 10 days) were assessed in HS. AB-506 or placebo was assessed at either 160 mg or 400 mg daily for 28 days in subjects with CHB. A second follow-up study examined AB-506 or placebo at 400 mg daily for 28 days in 14 Caucasian and 14 East-Asian HS. Twenty-eight days of AB-506 at 160 mg and 400 mg produced mean HBV-DNA declines from baseline of 2.1 log10 IU/ml and 2.8 log10 IU/ml, respectively. Four subjects with CHB (all Asian) had Grade 4 alanine aminotransferase (ALT) elevations (2 at each dose) as HBV DNA was declining; three events led to treatment discontinuation. In the second follow-up study, 2 Asian HS had serious transaminitis events leading to treatment and study termination. No subjects had bilirubin elevations or signs of hepatic decompensation. Conclusion: AB-506 demonstrated mean HBV-DNA declines of >2 log10 ; however, transient but severe ALT flares were observed in 4 Asian subjects with CHB. In the follow-up study in HS, 2 additional Asian HS had Grade 4 flares, suggesting that AB-506 hepatotoxicity contributed to the ALT elevations. The AB-506 development program was terminated because of these findings.


Assuntos
Antivirais , Hepatite B , Humanos , Antivirais/efeitos adversos , Capsídeo , Proteínas do Capsídeo , DNA Viral , Seguimentos , Voluntários Saudáveis , Hepatite B/tratamento farmacológico , Antígenos E da Hepatite B , Vírus da Hepatite B/genética
6.
RSC Med Chem ; 13(3): 343-349, 2022 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-35434625

RESUMO

Disruption of the HBV viral life cycle with small molecules that prevent the encapsidation of pregenomic RNA and viral polymerase through binding to HBV core protein is a clinically validated approach to inhibiting HBV viral replication. Herein we report the further optimisation of clinical candidate AB-506 through core modification with a focus on increasing oral exposure and oral half-life. Maintenance of high levels of anti-HBV cellular potency in conjunction with improvements in pharmacokinetic properties led to multi-log10 reductions in serum HBV DNA following low, once-daily oral dosing for key analogues in a preclinical animal model of HBV replication.

7.
Mol Ther ; 29(10): 2910-2919, 2021 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-34091052

RESUMO

N-Acetylgalactosamine (GalNAc) conjugated short interfering RNAs (siRNAs) are a leading RNA interference (RNAi) platform allowing targeted inhibition of disease-causing genes in hepatocytes. More than a decade of development has recently resulted in the first approvals for this class of drugs. While substantial effort has been made to improve nucleic acid modification patterns for better payload stability and efficacy, relatively little attention has been given to the GalNAc targeting ligand. In addition, the lack of an intrinsic endosomal release mechanism has limited potency. Here, we report a stepwise analysis of the structure activity relationships (SAR) of the components comprising these targeting ligands. We show that there is relatively little difference in biological performance between bi-, tri-, and tetravalent ligand structures while identifying other features that affect their biological activity more significantly. Further, we demonstrate that subcutaneous co-administration of a GalNAc-functionalized, pH responsive endosomal release agent markedly improved the activity and duration of effect for siRNA conjugates, without compromising tolerability, in non-human primates. These findings could address a significant bottleneck for future siRNA ligand conjugate development.


Assuntos
Acetilgalactosamina/química , Receptor de Asialoglicoproteína/metabolismo , RNA Interferente Pequeno/administração & dosagem , Animais , Feminino , Células Hep G2 , Humanos , Injeções Subcutâneas , Ligantes , Lipossomos , Masculino , Camundongos , Nanopartículas , Primatas , RNA Interferente Pequeno/química , Relação Estrutura-Atividade
8.
Nat Commun ; 12(1): 1222, 2021 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-33619272

RESUMO

Programmed death-ligand 1 is a glycoprotein expressed on antigen presenting cells, hepatocytes, and tumors which upon interaction with programmed death-1, results in inhibition of antigen-specific T cell responses. Here, we report a mechanism of inhibiting programmed death-ligand 1 through small molecule-induced dimerization and internalization. This represents a mechanism of checkpoint inhibition, which differentiates from anti-programmed death-ligand 1 antibodies which function through molecular disruption of the programmed death 1 interaction. Testing of programmed death ligand 1 small molecule inhibition in a humanized mouse model of colorectal cancer results in a significant reduction in tumor size and promotes T cell proliferation. In addition, antigen-specific T and B cell responses from patients with chronic hepatitis B infection are significantly elevated upon programmed death ligand 1 small molecule inhibitor treatment. Taken together, these data identify a mechanism of small molecule-induced programmed death ligand 1 internalization with potential therapeutic implications in oncology and chronic viral infections.


Assuntos
Antígeno B7-H1/metabolismo , Endocitose , Inibidores de Checkpoint Imunológico/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Antineoplásicos/farmacologia , Antivirais/farmacologia , Células CHO , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/patologia , Cricetulus , Modelos Animais de Doenças , Feminino , Vírus da Hepatite B/efeitos dos fármacos , Humanos , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1/metabolismo , Multimerização Proteica/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/química
9.
ACS Infect Dis ; 5(5): 738-749, 2019 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-30408957

RESUMO

Hepatitis delta virus (HDV) infects 10-20 million individuals worldwide and causes severe fulminant hepatitis with high likelihood of cirrhosis and hepatocellular carcinoma. HDV infection cannot occur in the absence of the surface antigen (HBsAg) of the hepatitis B virus. RNA interference is an effective mechanism by which to inhibit viral transcripts, and siRNA therapeutics sharing this mechanism have begun to demonstrate clinical efficacy. Here we assessed the outcome of HBV-targeting siRNA intervention against HDV and compared it to a direct anti-HDV siRNA approach in dually infected humanized mice. Treatment with ARB-1740, a clinical stage HBV-targeting siRNA agent delivered using lipid nanoparticle (LNP) technology, effectively reduced HBV viremia by 2.3 log10 and serum HBsAg by 2.6 log10, leading to 1.6 log10 reduction of HDV viremia. In contrast, HDV-targeting siRNA inhibited HDV in both blood and liver compartments without affecting HBV and PEGylated interferon-alpha reduced HBV viremia by 2.0 log10 but had no effect on HDV viremia under these study conditions. These results illustrate the inhibitory effects of siRNAs against these two viral infections and suggest that ARB-1740 may be of therapeutic benefit for hepatitis delta patients, a subpopulation with high unmet medical need.


Assuntos
Antivirais/uso terapêutico , Hepatite D/tratamento farmacológico , Vírus Delta da Hepatite/efeitos dos fármacos , Interferência de RNA , Animais , Animais Geneticamente Modificados , Modelos Animais de Doenças , Feminino , Vírus da Hepatite B/efeitos dos fármacos , Humanos , Camundongos
10.
ACS Infect Dis ; 5(5): 725-737, 2019 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-30403127

RESUMO

Current approved nucleoside analogue treatments for chronic hepatitis B virus (HBV) infection are effective at controlling viral titer but are not curative and have minimal impact on the production of viral proteins such as surface antigen (HBsAg), the HBV envelope protein believed to play a role in maintaining the immune tolerant state required for viral persistence. Novel agents are needed to effect HBV cure, and reduction of HBV antigenemia may potentiate activation of effective and long-lasting host immune control. ARB-1740 is a clinical stage RNA interference agent composed of three siRNAs delivered using lipid nanoparticle technology. In a number of cell and animal models of HBV, ARB-1740 caused HBV RNA reduction, leading to inhibition of multiple elements of the viral life cycle including HBsAg, HBeAg, and HBcAg viral proteins as well as replication marker HBV DNA. ARB-1740 demonstrated pan-genotypic activity in vitro and in vivo, targeting three distinct highly conserved regions of the HBV genome, and effectively inhibited replication of nucleoside analogue-resistant HBV variants. Combination of ARB-1740 with a capsid inhibitor and pegylated interferon-alpha led to greater liver HBsAg reduction which correlated with more robust induction of innate immune responses in a human chimeric mouse model of HBV. The preclinical profile of ARB-1740 demonstrates the promise of RNA interference and HBV antigen reduction in treatment strategies driving toward a cure for HBV.


Assuntos
Antivirais/uso terapêutico , Vírus da Hepatite B/efeitos dos fármacos , Hepatite B Crônica/tratamento farmacológico , Interferência de RNA , RNA Interferente Pequeno/uso terapêutico , Animais , Genoma Viral , Humanos , Imunidade Inata , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nanopartículas/administração & dosagem , Nanopartículas/química , RNA Interferente Pequeno/química , Replicação Viral/efeitos dos fármacos
11.
J Clin Invest ; 127(12): 4437-4448, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29106386

RESUMO

Ebolaviruses and marburgviruses belong to the family Filoviridae and cause high lethality in infected patients. There are currently no licensed filovirus vaccines or antiviral therapies. The development of broad-spectrum therapies against members of the Marburgvirus genus, including Marburg virus (MARV) and Ravn virus (RAVV), is difficult because of substantial sequence variability. RNAi therapeutics offer a potential solution, as identification of conserved target nucleotide sequences may confer activity across marburgvirus variants. Here, we assessed the therapeutic efficacy of lipid nanoparticle (LNP) delivery of a single nucleoprotein-targeting (NP-targeting) siRNA in nonhuman primates at advanced stages of MARV or RAVV disease to mimic cases in which patients begin treatment for fulminant disease. Sixteen rhesus monkeys were lethally infected with MARV or RAVV and treated with NP siRNA-LNP, with MARV-infected animals beginning treatment four or five days after infection and RAVV-infected animals starting treatment three or six days after infection. While all untreated animals succumbed to disease, NP siRNA-LNP treatment conferred 100% survival of RAVV-infected macaques, even when treatment began just 1 day prior to the death of the control animals. In MARV-infected animals, day-4 treatment initiation resulted in 100% survival, and day-5 treatment resulted in 50% survival. These results identify a single siRNA therapeutic that provides broad-spectrum protection against both MARV and RAVV.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Doença do Vírus de Marburg/tratamento farmacológico , Marburgvirus , Nanopartículas/uso terapêutico , RNA Interferente Pequeno/farmacologia , Animais , Macaca mulatta , Doença do Vírus de Marburg/metabolismo , Doença do Vírus de Marburg/patologia , Nanopartículas/química , RNA Interferente Pequeno/química
12.
Nat Microbiol ; 1(10): 16142, 2016 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-27670117

RESUMO

Although significant progress has been made in developing therapeutics against Zaire ebolavirus, these therapies do not protect against other Ebola species such as Sudan ebolavirus (SUDV). Here, we describe an RNA interference therapeutic comprising siRNA targeting the SUDV VP35 gene encapsulated in lipid nanoparticle (LNP) technology with increased potency beyond formulations used in TKM-Ebola clinical trials. Twenty-five rhesus monkeys were challenged with a lethal dose of SUDV. Twenty animals received siRNA-LNP beginning at 1, 2, 3, 4 or 5 days post-challenge. VP35-targeting siRNA-LNP treatment resulted in up to 100% survival, even when initiated when fever, viraemia and disease signs were evident. Treatment effectively controlled viral replication, mediating up to 4 log10 reductions after dosing. Mirroring clinical findings, a correlation between high viral loads and fatal outcome was observed, emphasizing the importance of stratifying efficacy according to viral load. In summary, strong survival benefit and rapid control of SUDV replication by VP35-targeting LNP confirm its therapeutic potential in combatting this lethal disease.


Assuntos
Doença pelo Vírus Ebola/terapia , Lipídeos , Interferência de RNA , RNA Interferente Pequeno/uso terapêutico , Animais , Anticorpos Antivirais , Modelos Animais de Doenças , Composição de Medicamentos , Ebolavirus/isolamento & purificação , Ebolavirus/patogenicidade , Doença pelo Vírus Ebola/virologia , Células Hep G2 , Humanos , Macaca mulatta , Nanopartículas/administração & dosagem , Nanopartículas/química , RNA Interferente Pequeno/genética , Sudão , Carga Viral/efeitos dos fármacos , Proteínas Virais Reguladoras e Acessórias/genética , Proteínas Virais Reguladoras e Acessórias/metabolismo , Viremia/terapia , Replicação Viral
13.
J Infect Dis ; 214(suppl 3): S367-S374, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27571900

RESUMO

BACKGROUND: Convalescent serum and blood were used to treat patients during outbreaks of Zaire ebolavirus (ZEBOV) infection in 1976 and 1995, with inconclusive results. During the recent 2013-2016 West African epidemic, serum/plasma from survivors of ZEBOV infection was used to treat patients in the affected countries and several repatriated patients. The effectiveness of this strategy remains unknown. METHODS: Nine rhesus monkeys were experimentally infected with ZEBOV-Makona. Beginning on day 3 after exposure (at the onset of viremia), 4 animals were treated with homologous ZEBOV-Makona convalescent macaque sera, 3 animals were treated in parallel with heterologous Sudan ebolavirus (SEBOV) convalescent macaque sera, and 2 animals served as positive controls and were not treated. Surviving animals received additional treatments on days 6 and 9. RESULTS: Both untreated control animals died on postinfection day 9. All 4 ZEBOV-Makona-infected macaques treated with homologous ZEBOV-Makona convalescent sera died on days 8-9. One macaque treated with heterologous SEBOV convalescent sera survived, while the other animals treated with the heterologous SEBOV sera died on days 7 and 9. CONCLUSIONS: The findings suggest that convalescent sera alone is not sufficient for providing 100% protection against lethal ZEBOV infection when administered at the onset of viremia.


Assuntos
Ebolavirus/imunologia , Doença pelo Vírus Ebola/prevenção & controle , Imunização Passiva , Animais , Convalescença , Feminino , Doença pelo Vírus Ebola/virologia , Humanos , Macaca mulatta , Masculino , Soro/imunologia , Viremia
14.
Nature ; 521(7552): 362-5, 2015 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-25901685

RESUMO

The current outbreak of Ebola virus in West Africa is unprecedented, causing more cases and fatalities than all previous outbreaks combined, and has yet to be controlled. Several post-exposure interventions have been employed under compassionate use to treat patients repatriated to Europe and the United States. However, the in vivo efficacy of these interventions against the new outbreak strain of Ebola virus is unknown. Here we show that lipid-nanoparticle-encapsulated short interfering RNAs (siRNAs) rapidly adapted to target the Makona outbreak strain of Ebola virus are able to protect 100% of rhesus monkeys against lethal challenge when treatment was initiated at 3 days after exposure while animals were viraemic and clinically ill. Although all infected animals showed evidence of advanced disease including abnormal haematology, blood chemistry and coagulopathy, siRNA-treated animals had milder clinical features and fully recovered, while the untreated control animals succumbed to the disease. These results represent the first, to our knowledge, successful demonstration of therapeutic anti-Ebola virus efficacy against the new outbreak strain in nonhuman primates and highlight the rapid development of lipid-nanoparticle-delivered siRNA as a countermeasure against this highly lethal human disease.


Assuntos
Ebolavirus/efeitos dos fármacos , Ebolavirus/genética , Doença pelo Vírus Ebola/terapia , Doença pelo Vírus Ebola/virologia , Nanopartículas/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/uso terapêutico , Animais , Sequência de Bases , Modelos Animais de Doenças , Ebolavirus/classificação , Feminino , Doença pelo Vírus Ebola/patologia , Doença pelo Vírus Ebola/prevenção & controle , Humanos , Macaca mulatta/virologia , Masculino , RNA Interferente Pequeno/farmacologia , Análise de Sobrevida , Fatores de Tempo , Resultado do Tratamento , Carga Viral/efeitos dos fármacos
15.
Sci Transl Med ; 6(250): 250ra116, 2014 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-25143366

RESUMO

Marburg virus (MARV) and the closely related filovirus Ebola virus cause severe and often fatal hemorrhagic fever (HF) in humans and nonhuman primates with mortality rates up to 90%. There are no vaccines or drugs approved for human use, and no postexposure treatment has completely protected nonhuman primates against MARV-Angola, the strain associated with the highest rate of mortality in naturally occurring human outbreaks. Studies performed with other MARV strains assessed candidate treatments at times shortly after virus exposure, before signs of disease are detectable. We assessed the efficacy of lipid nanoparticle (LNP) delivery of anti-MARV nucleoprotein (NP)-targeting small interfering RNA (siRNA) at several time points after virus exposure, including after the onset of detectable disease in a uniformly lethal nonhuman primate model of MARV-Angola HF. Twenty-one rhesus monkeys were challenged with a lethal dose of MARV-Angola. Sixteen of these animals were treated with LNP containing anti-MARV NP siRNA beginning at 30 to 45 min, 1 day, 2 days, or 3 days after virus challenge. All 16 macaques that received LNP-encapsulated anti-MARV NP siRNA survived infection, whereas the untreated or mock-treated control subjects succumbed to disease between days 7 and 9 after infection. These results represent the successful demonstration of therapeutic anti-MARV-Angola efficacy in nonhuman primates and highlight the substantial impact of an LNP-delivered siRNA therapeutic as a countermeasure against this highly lethal human disease.


Assuntos
Lipídeos/uso terapêutico , Macaca mulatta/virologia , Doença do Vírus de Marburg/virologia , Marburgvirus/fisiologia , Nanopartículas/uso terapêutico , RNA Interferente Pequeno/uso terapêutico , Animais , Antígenos Virais/imunologia , Humanos , Macaca mulatta/imunologia , Doença do Vírus de Marburg/patologia , Doença do Vírus de Marburg/terapia , Marburgvirus/imunologia , Nanopartículas/química , RNA Viral/metabolismo , Análise de Sobrevida , Resultado do Tratamento , Viremia/patologia
16.
Future Microbiol ; 8(3): 403-11, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23464375

RESUMO

AIM: Infections associated with medical devices are an important cause of morbidity and mortality. Microorganisms are responsible for catheter infections that may then result in the local or systemic dissemination of the microorganism into the bloodstream. The aim of this study was to evaluate the antimicrobial activity of silver nanoparticles (AgNPs) embedded in polyurethane plastics, commonly used for catheter fabrication. MATERIALS & METHODS: AgNPs in the range of 25-30 nm were synthesized and embedded in polyurethane plastics at different concentrations. The antimicrobial activities of these plastics were tested against the three pathogenic microorganisms, Escherichia coli, Staphylococcus epidermidis and Candida albicans, frequently associated with catheter infections. The cytotoxicity of the plastics was evaluated on human-derived macrophages using propidium iodide and the secretion of the pro- and anti-inflammatory cytokines IL-6, IL-10 and TNF-a was measured using ELISA. RESULTS: A significant reduction of 6- to 7-log in the number of bacteria was measured, while a reduction of 90% was measured in the case of C. albicans. Neither cytotoxic effect on macrophages nor immunological response was observed. CONCLUSION: Plastics embedded with AgNPs have great potential to limit microbial colonization of implanted medical devices.


Assuntos
Anti-Infecciosos/farmacologia , Nanopartículas/toxicidade , Poliuretanos/farmacologia , Poliuretanos/toxicidade , Prata/farmacologia , Prata/toxicidade , Apoptose , Candida albicans/efeitos dos fármacos , Linhagem Celular , Ensaio de Imunoadsorção Enzimática , Escherichia coli/efeitos dos fármacos , Humanos , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Macrófagos/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Staphylococcus epidermidis/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
17.
Cell Microbiol ; 15(5): 795-808, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23167250

RESUMO

Using a genetic screen in yeast we found that Mycobacterium tuberculosis PE-PGRS62 was capable of disrupting yeast vacuolar protein sorting, suggesting effects on endosomal trafficking. To study the impact of PE-PGRS62 on macrophage function, we infected murine macrophages with Mycobacterium smegmatis expressing PE-PGRS62. Infected cells displayed phagosome maturation arrest. Phagosomes acquired Rab5, but displayed a significant defect in Rab7 and LAMP-1 acquisition. Macrophages infected with M. smegmatis expressing PE-PGRS62 also expressed two- to threefold less iNOS protein when compared with cells infected with wild-type bacteria. Consistent with this, cells infected with a Mycobacterium marinum transposon mutant for the PE-PGRS62 orthologue showed greater iNOS protein expression when compared to cells infected with wild-type organisms. Complementation restored the ability of the mutant to inhibit iNOS expression. No differences in iNOS transcript levels were observed, suggesting that PE-PGRS62 effects on iNOS expression occurred post-transcriptionally. Marked differences in colony morphology were also seen in M. smegmatis expressing PE-PGRS62 and in the M. marinum transposon mutant, suggesting that PE-PGRS62 may affect cell wall composition. These findings suggest that PE-PGRS62 supports virulence via inhibition of phagosome maturation and iNOS expression, and these phenotypes may be linked to effects on bacterial cell wall composition.


Assuntos
Proteínas de Bactérias/isolamento & purificação , Macrófagos/metabolismo , Mycobacterium tuberculosis/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Animais , Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica , Humanos , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , Mycobacterium smegmatis/metabolismo , Mycobacterium smegmatis/patogenicidade , Mycobacterium tuberculosis/patogenicidade , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Fagossomos/metabolismo , Fagossomos/microbiologia
18.
PLoS One ; 7(8): e43668, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22928013

RESUMO

Of the various phosphatidylinositol 3- kinases (PI3Ks), only the class III enzyme Vps34 has been shown to regulate phagosome maturation. During studies of phagosome maturation in THP-1 cells deficient in class IA PI3K p110α, we discovered that this PI3K isoform is required for vacuole maturation to progress beyond acquisition of Rab7 leading to delivery of lysosomal markers. Bead phagosomes from THP-1 cells acquired p110α and contained PI3P and PI(3,4,5)P3; however, p110α and PI(3,4,5)P3 levels in phagosomes from p110α knockdown cells were decreased. Phagosomes from p110α knock down cells showed normal acquisition of both Rab5 and EEA-1, but were markedly deficient in the lysosomal markers LAMP-1 and LAMP-2, and the lysosomal hydrolase, ß-galactosidase. Phagosomes from p110α deficient cells also displayed impaired fusion with Texas Red dextran-loaded lysosomes. Despite lacking lysosomal components, phagosomes from p110α deficient cells recruited normal levels of Rab7, Rab-interacting lysosomal protein (RILP) and homotypic vacuole fusion and protein sorting (HOPs) components Vps41 and Vps16. The latter observations demonstrated that phagosomal Rab7 was active and capable of recruiting effectors involved in membrane fusion. Nevertheless, active Rab7 was not sufficient to bring about the delivery of lysosomal proteins to the maturing vacuole, which is shown for the first time to be dependent on a class I PI3K.


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Fagossomos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Linhagem Celular Tumoral , Classe Ia de Fosfatidilinositol 3-Quinase/deficiência , Classe Ia de Fosfatidilinositol 3-Quinase/genética , Técnicas de Silenciamento de Genes , Humanos , Proteína 1 de Membrana Associada ao Lisossomo/metabolismo , Lisossomos/metabolismo , Fagossomos/enzimologia , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas de Transporte Vesicular/metabolismo , beta-Galactosidase/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
19.
J Leukoc Biol ; 92(3): 553-66, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22569897

RESUMO

Phagosome maturation is a highly organized and sequential process that results in the formation of a microbicidal phagolysosome. This results in crucial contributions to innate and adaptive immunity through pathogen clearance and antigen presentation. Thus, it is important to understand the regulatory networks that control the extent and nature of phagosome maturation. PI3Ks are lipid kinases that catalyze the phosphorylation of the 3' position of the inositol ring. This enzyme family is divided into three classes based on structure and substrate preferences. Previously, only the class III PI3K, hVps34, was thought to contribute to phagosome maturation. Recent evidence, however, suggests important contributions by class I PI3Ks in bringing about the diverse phagosome maturation phenotypes. Class I PI3Ks have also been implicated in the activation of Rab GTPases that function in maturation, such as Rab14. In addition, recent studies have illuminated the overlap between phagosome maturation and autophagy, which itself is regulated by multiple classes of PI3K. Taken together, a picture of phagosome maturation is emerging in which multiple classes of PI3Ks are involved in modulating maturation phenotypes. This review summarizes the known contributions of PI3Ks to phagosome maturation. Special emphasis is placed on the impact of PI3Ks on different maturation outcomes stemming from the engagement of diverse phagocytic receptors and on Rab and Ca(2+) signaling cascades.


Assuntos
Fagocitose/fisiologia , Fagossomos/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Animais , Humanos , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...