Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Virol ; 93(21)2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31391268

RESUMEN

Cleavage of influenza virus hemagglutinin (HA) by host cell proteases is essential for virus infectivity and spread. We previously demonstrated in vitro that the transmembrane protease TMPRSS2 cleaves influenza A virus (IAV) and influenza B virus (IBV) HA possessing a monobasic cleavage site. Subsequent studies revealed that TMPRSS2 is crucial for the activation and pathogenesis of H1N1pdm and H7N9 IAV in mice. In contrast, activation of H3N2 IAV and IBV was found to be independent of TMPRSS2 expression and supported by an as-yet-undetermined protease(s). Here, we investigated the role of TMPRSS2 in proteolytic activation of IAV and IBV in three human airway cell culture systems: primary human bronchial epithelial cells (HBEC), primary type II alveolar epithelial cells (AECII), and Calu-3 cells. Knockdown of TMPRSS2 expression was performed using a previously described antisense peptide-conjugated phosphorodiamidate morpholino oligomer, T-ex5, that interferes with splicing of TMPRSS2 pre-mRNA, resulting in the expression of enzymatically inactive TMPRSS2. T-ex5 treatment produced efficient knockdown of active TMPRSS2 in all three airway cell culture models and prevented proteolytic activation and multiplication of H7N9 IAV in Calu-3 cells and H1N1pdm, H7N9, and H3N2 IAV in HBEC and AECII. T-ex5 treatment also inhibited the activation and spread of IBV in AECII but did not affect IBV activation in HBEC and Calu-3 cells. This study identifies TMPRSS2 as the major HA-activating protease of IAV in human airway cells and IBV in type II pneumocytes and as a potential target for the development of novel drugs to treat influenza infections.IMPORTANCE Influenza A viruses (IAV) and influenza B viruses (IBV) cause significant morbidity and mortality during seasonal outbreaks. Cleavage of the viral surface glycoprotein hemagglutinin (HA) by host proteases is a prerequisite for membrane fusion and essential for virus infectivity. Inhibition of relevant proteases provides a promising therapeutic approach that may avoid the development of drug resistance. HA of most influenza viruses is cleaved at a monobasic cleavage site, and a number of proteases have been shown to cleave HA in vitro This study demonstrates that the transmembrane protease TMPRSS2 is the major HA-activating protease of IAV in primary human bronchial cells and of both IAV and IBV in primary human type II pneumocytes. It further reveals that human and murine airway cells can differ in their HA-cleaving protease repertoires. Our data will help drive the development of potent and selective protease inhibitors as novel drugs for influenza treatment.


Asunto(s)
Virus de la Influenza A/fisiología , Virus de la Influenza B/fisiología , Gripe Humana/virología , Serina Endopeptidasas/metabolismo , Animales , Bronquios/citología , Células Cultivadas , Células Epiteliales/virología , Técnicas de Silenciamiento del Gen , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Interacciones Huésped-Patógeno , Humanos , Gripe Humana/enzimología , Gripe Humana/metabolismo , Ratones , Infecciones por Orthomyxoviridae/enzimología , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , Alveolos Pulmonares/citología , Serina Endopeptidasas/genética , Regulación hacia Arriba , Replicación Viral
2.
Infect Agent Cancer ; 13: 4, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29375654

RESUMEN

BACKGROUND: Cervical cancer (CC) is the leading cause of morbidity and mortality from cancer in Nepalese women. Nearly all cases of CC are caused by infection with certain genotypes of human papillomavirus (HPV). Data on HPV genotype distribution in Nepalese CC patients is sparse. We aimed to determine the distribution of HPV genotypes in biopsies of CC tissue from Nepalese women. METHODS: This study examined 248 archived paraffin-embedded tissue specimens from CC cases from patients of B.P. Koirala Memorial Cancer Hospital, Bharatpur, Chitwan, Nepal. DNA was extracted from the biopsies and HPV detection performed by PCR. HPV genotyping was then carried out by a reverse line hybridization technique capable of identifying 36 distinct HPV genotypes. RESULTS: Most of the samples were from tumors that had been designated by hospital pathologists as squamous cell carcinoma (77.6%). 165 of the 248 samples contained DNA of sufficient quality for rigorous PCR testing. All the analyzable specimens were positive for HPV. The most common HPV genotypes, in decreasing order of frequency were 16, 18, 45, 33, 52, 56 and 31; most were found as single infections (94.5%). Together, HPV types 16, 18, and 45 were found in 92% of the tumor samples. CONCLUSION: This study strengthens the knowledge-base of HPV genotype distribution in CC cases in Nepal. Hopefully, this information will be useful to the medical community and public health policy-makers in generating improved HPV-surveillance, -prevention and -treatment strategies in Nepal.

3.
Nat Microbiol ; 2: 17022, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-28248290

RESUMEN

Retinoic acid-inducible gene I (RIG-I) receptor recognizes 5'-triphosphorylated RNA and triggers a signalling cascade that results in the induction of type-I interferon (IFN)-dependent responses. Its precise regulation represents a pivotal balance between antiviral defences and autoimmunity. To elucidate the cellular cofactors that regulate RIG-I signalling, we performed two global RNA interference analyses to identify both positive and negative regulatory nodes operating on the signalling pathway during virus infection. These factors were integrated with experimentally and computationally derived interactome data to build a RIG-I protein interaction network. Our analysis revealed diverse cellular processes, including the unfolded protein response, Wnt signalling and RNA metabolism, as critical cellular components governing innate responses to non-self RNA species. Importantly, we identified K-Homology Splicing Regulatory Protein (KHSRP) as a negative regulator of this pathway. We find that KHSRP associates with the regulatory domain of RIG-I to maintain the receptor in an inactive state and attenuate its sensing of viral RNA (vRNA). Consistent with increased RIG-I antiviral signalling in the absence of KHSRP, viral replication is reduced when KHSRP expression is knocked down both in vitro and in vivo. Taken together, these data indicate that KHSRP functions as a checkpoint regulator of the innate immune response to pathogen challenge.


Asunto(s)
Proteína 58 DEAD Box/antagonistas & inhibidores , ARN Viral/inmunología , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Transactivadores/metabolismo , Células HEK293 , Humanos , Inmunidad Innata , Subtipo H1N1 del Virus de la Influenza A/inmunología , Unión Proteica , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas , Receptores Inmunológicos
4.
Antiviral Res ; 120: 134-9, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26086884

RESUMEN

Hepatitis E virus (HEV) infection is a cause of hepatitis in humans worldwide and has been associated with a case-fatality rate of up to 30% in pregnant women. Recently, persistent and chronic HEV infections have been recognized as a serious clinical problem, especially in immunocompromised individuals. To date, there are no FDA-approved HEV-specific antiviral drugs. In this study, we evaluated antisense peptide-conjugated morpholino oligomers (PPMO) designed against HEV genomic sequences as potential HEV-specific antiviral compounds. Two genetically-distinct strains of human HEV, genotype 1 Sar55 and genotype 3 Kernow-C1, isolated from patients with acute and chronic hepatitis, respectively, were used to evaluate inhibition of viral replication by PPMO in liver cells. The anti-HEV PPMO produced a significant reduction in the levels of HEV RNA and capsid protein, indicating effective inhibition of HEV replication. PPMO HP1, which targets a highly conserved sequence in the start site region of ORF1, was also effective against the genotype 3 Kernow-C1 strain in stably-infected HepG2/C3A liver cells. The antiviral activity observed was specific, dose-responsive and potent, suggesting that further exploration of PPMO HP1 as a potential HEV-specific antiviral agent is warranted.


Asunto(s)
Antivirales/farmacología , Portadores de Fármacos/metabolismo , Virus de la Hepatitis E/efectos de los fármacos , Morfolinos/farmacología , Oligonucleótidos Antisentido/farmacología , Péptidos/metabolismo , Replicación Viral/efectos de los fármacos , Proteínas de la Cápside/análisis , Línea Celular , Virus de la Hepatitis E/fisiología , Hepatocitos/virología , Humanos , Pruebas de Sensibilidad Microbiana , ARN Viral/análisis , Carga Viral
5.
J Virol ; 87(17): 9411-9, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23824813

RESUMEN

Dengue virus has emerged as a global health threat to over one-third of humankind. As a positive-strand RNA virus, dengue virus relies on the host cell metabolism for its translation, replication, and egress. Therefore, a better understanding of the host cell metabolic pathways required for dengue virus infection offers the opportunity to develop new approaches for therapeutic intervention. In a recently described screen of known drugs and bioactive molecules, we observed that methotrexate and floxuridine inhibited dengue virus infections at low micromolar concentrations. Here, we demonstrate that all serotypes of dengue virus, as well as West Nile virus, are highly sensitive to both methotrexate and floxuridine, whereas other RNA viruses (Sindbis virus and vesicular stomatitis virus) are not. Interestingly, flavivirus replication was restored by folinic acid, a thymidine precursor, in the presence of methotrexate and by thymidine in the presence of floxuridine, suggesting an unexpected role for thymidine in flavivirus replication. Since thymidine is not incorporated into RNA genomes, it is likely that increased thymidine production is indirectly involved in flavivirus replication. A possible mechanism is suggested by the finding that p53 inhibition restored dengue virus replication in the presence of floxuridine, consistent with thymidine-less stress triggering p53-mediated antiflavivirus effects in infected cells. Our data reveal thymidine synthesis pathways as new and unexpected therapeutic targets for antiflaviviral drug development.


Asunto(s)
Antivirales/farmacología , Virus del Dengue/efectos de los fármacos , Virus del Dengue/metabolismo , Flavivirus/efectos de los fármacos , Flavivirus/metabolismo , Timidina/biosíntesis , Animales , Línea Celular , Chlorocebus aethiops , Virus ADN/efectos de los fármacos , Virus del Dengue/fisiología , Modelos Animales de Enfermedad , Flavivirus/fisiología , Infecciones por Flavivirus/tratamiento farmacológico , Floxuridina/farmacología , Células HEK293 , Células HeLa , Humanos , Leucovorina/farmacología , Metotrexato/farmacología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Virus ARN/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Células Vero , Replicación Viral/efectos de los fármacos , Virus del Nilo Occidental/efectos de los fármacos , Virus del Nilo Occidental/metabolismo , Virus del Nilo Occidental/fisiología
6.
Antimicrob Agents Chemother ; 55(10): 4631-8, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21825302

RESUMEN

Members of the Arenaviridae family are a threat to public health and can cause meningitis and hemorrhagic fever, and yet treatment options remain limited by a lack of effective antivirals. In this study, we found that peptide-conjugated phosphorodiamidate morpholino oligomers (PPMO) complementary to viral genomic RNA were effective in reducing arenavirus replication in cell cultures and in vivo. PPMO complementary to the Junín virus genome were designed to interfere with viral RNA synthesis or translation or both. However, only PPMO designed to potentially interfere with translation were effective in reducing virus replication. PPMO complementary to sequences that are highly conserved across the arenaviruses and located at the 5' termini of both genomic segments were effective against Junín virus, Tacaribe virus, Pichinde virus, and lymphocytic choriomeningitis virus (LCMV)-infected cell cultures and suppressed viral titers in the livers of LCMV-infected mice. These results suggest that arenavirus 5' genomic termini represent promising targets for pan-arenavirus antiviral therapeutic development.


Asunto(s)
Antivirales/farmacología , Arenavirus/efectos de los fármacos , Morfolinos/farmacología , Péptidos/farmacología , Animales , Infecciones por Arenaviridae/tratamiento farmacológico , Infecciones por Arenaviridae/virología , Arenavirus/genética , Arenavirus/crecimiento & desarrollo , Arenavirus del Nuevo Mundo/efectos de los fármacos , Línea Celular , Chlorocebus aethiops , Genoma Viral , Virus Junin/efectos de los fármacos , Virus de la Coriomeningitis Linfocítica/efectos de los fármacos , Ratones , Pruebas de Sensibilidad Microbiana , Virus Pichinde/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , ARN Viral/genética , Células Vero , Replicación Viral/efectos de los fármacos
7.
J Virol ; 85(4): 1554-62, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21123387

RESUMEN

Influenza A viruses constitute a major and ongoing global public health concern. Current antiviral strategies target viral gene products; however, the emergence of drug-resistant viruses highlights the need for novel antiviral approaches. Cleavage of the influenza virus hemagglutinin (HA) by host cell proteases is crucial for viral infectivity and therefore presents a potential drug target. Peptide-conjugated phosphorodiamidate morpholino oligomers (PPMO) are single-stranded-DNA-like antisense agents that readily enter cells and can act as antisense agents by sterically blocking cRNA. Here, we evaluated the effect of PPMO targeted to regions of the pre-mRNA or mRNA of the HA-cleaving protease TMPRSS2 on proteolytic activation and spread of influenza viruses in human Calu-3 airway epithelial cells. We found that treatment of cells with a PPMO (T-ex5) designed to interfere with TMPRSS2 pre-mRNA splicing resulted in TMPRSS2 mRNA lacking exon 5 and consequently the expression of a truncated and enzymatically inactive form of TMPRSS2. Altered splicing of TMPRSS2 mRNA by the T-ex5 PPMO prevented HA cleavage in different human seasonal and pandemic influenza A viruses and suppressed viral titers by 2 to 3 log(10) units, strongly suggesting that TMPRSS2 is responsible for HA cleavage in Calu-3 airway cells. The data indicate that PPMO provide a useful reagent for investigating HA-activating proteases and may represent a promising strategy for the development of novel therapeutics to address influenza infections.


Asunto(s)
Células Epiteliales/virología , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/patogenicidad , Metaloendopeptidasas/metabolismo , Morfolinas/farmacología , Serina Endopeptidasas/metabolismo , Animales , Bronquios/citología , Línea Celular , Células Cultivadas , Embrión de Pollo , Perros , Humanos , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/enzimología , Subtipo H3N2 del Virus de la Influenza A/efectos de los fármacos , Subtipo H3N2 del Virus de la Influenza A/enzimología , Subtipo H7N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H7N1 del Virus de la Influenza A/enzimología , Virus de la Influenza A/enzimología , Morfolinos , Precursores del ARN/genética , Precursores del ARN/metabolismo , Empalme del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo
8.
Antivir Ther ; 15(8): 1141-9, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21149921

RESUMEN

BACKGROUND: Genital herpes, caused by herpes simplex virus type-2 (HSV-2), is a recurrent, lifelong disease affecting tens of millions of people in the USA alone. HSV-2 can be treated therapeutically with acyclovir (ACV) and its derivatives; however, no treatment can prevent HSV reactivation. Novel topical anti-HSV microbicides are much needed to reduce HSV-2 transmission and to treat primary or reactivated infections, especially for ACV-resistant strains. Peptide-conjugated phosphorodiamidate morpholino oligomers (PPMOs) are single-stranded DNA analogues that enter cells readily and can reduce target gene expression through steric blockage of complementary messenger RNA (mRNA). METHODS: We investigated the antiviral activities of PPMOs targeted to the translation start-site regions of the mRNA for two HSV-2 immediate early genes, immediate early protein (ICP)0 and ICP27, and two early genes, unique long gene (UL)30 and UL39. RESULTS: In cell cultures, PPMOs targeting ICP0 or ICP27 mRNA were found to be highly effective against two strains of HSV-2, one of which was ACV-resistant. In vivo, daily topical applications of up to 1 mM ICP27 PPMO caused no gross or microscopic damage to the genital tract of uninfected BALB/c mice or cotton rats. Cotton rats receiving topical application of ICP27 PPMO 24 h after HSV-2 inoculation showed a reduction in genital lesions and a 37.5% reduction in mortality at 14 days post-infection. Mice receiving topical application of 100 µM of an ICP27 and ICP0 PPMO combination before HSV-2 inoculation had no detectable viral replication in the genital tract at 3-5 days post-infection. CONCLUSIONS: These results demonstrate that topically applied PPMOs hold promise as candidate antiviral microbicides against HSV-2 genital infection.


Asunto(s)
Herpes Genital/tratamiento farmacológico , Herpesvirus Humano 2/efectos de los fármacos , Morfolinas/farmacología , Replicación Viral/efectos de los fármacos , Aciclovir/farmacología , Aciclovir/uso terapéutico , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Chlorocebus aethiops , Modelos Animales de Enfermedad , Farmacorresistencia Viral , Femenino , Herpes Genital/virología , Herpesvirus Humano 2/genética , Herpesvirus Humano 2/fisiología , Proteínas Inmediatas-Precoces/efectos de los fármacos , Proteínas Inmediatas-Precoces/genética , Ratones , Ratones Endogámicos BALB C , Morfolinas/síntesis química , Morfolinas/uso terapéutico , Morfolinos , Péptidos/metabolismo , Prevención Secundaria , Sigmodontinae , Células Vero , Proteínas Virales/efectos de los fármacos , Proteínas Virales/genética , Activación Viral/efectos de los fármacos
9.
J Antimicrob Chemother ; 65(5): 953-61, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20299495

RESUMEN

BACKGROUND: Japanese encephalitis virus (JEV) has a significant impact on public health throughout Asia, and there is a pressing need for development of new therapeutics against it. METHODS: Peptide-conjugated phosphorodiamidate morpholino oligomers (PPMOs) are antisense agents that enter cells readily and interfere with gene expression. Four PPMOs, targeting various locations in the JEV genome, were evaluated for antiviral activity against JEV in cultured cells and the mouse model of JEV infection. RESULTS: A PPMO (P10882) targeting the JEV 3' cyclization sequence (3'CSI) had significant antiviral activity in Vero (epithelial), Neuro2A (neuronal) and J774E (macrophage) cells at concentrations that were not cytotoxic. P10882 added before infection suppressed JEV replication to an undetectable level in Vero cells and produced a 93% and 66% reduction in titre in J774E and Neuro2A cells, respectively, when measured at 24 h post-infection. In uninfected cells, fluorescein-labelled PPMOs entered J774E cells most efficiently, followed by Vero and Neuro2A cells. The antiviral effect of P10882 was also demonstrated in vivo, where 60%-80% of 1-week-old mice treated intracerebrally with a 20 mg/kg dose of P10882 every 12 h for 5 days were protected from a lethal dose of JEV and showed an undetectable level of virus in brain tissue at 2 days post-infection. CONCLUSIONS: P10882, which targets sequence that is highly conserved across members of the JEV serocomplex, was previously shown to be effective in a mouse model of West Nile disease, and represents a candidate antiviral agent against members of the JEV serocomplex.


Asunto(s)
Antivirales/farmacología , Virus de la Encefalitis Japonesa (Especie)/efectos de los fármacos , Oligonucleótidos/farmacología , Péptidos/farmacología , Animales , Antivirales/uso terapéutico , Línea Celular , Chlorocebus aethiops , Encefalitis Japonesa/tratamiento farmacológico , Células Epiteliales/virología , Macrófagos/virología , Ratones , Ratones Endogámicos BALB C , Neuronas/virología , Oligonucleótidos/uso terapéutico , Péptidos/uso terapéutico , Análisis de Supervivencia
10.
Virus Res ; 150(1-2): 138-42, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20206215

RESUMEN

A significant consequence of equine arteritis virus (EAV) infection of horses is persistence of the virus in a variable percentage of infected stallions. We recently established an in vitro model of EAV persistence in cell culture for the purpose of furthering our understanding of EAV biology in general and viral persistence in the stallion in particular. In this study we investigated whether persistently infected HeLa cells could be cured of EAV infection by treatment with an antisense peptide-conjugated phosphorodiamidate morpholino oligomer (PPMO) designed to target the 5'-terminal region of the EAV genome. We found that persistently infected HeLa cells passaged three times in the presence of 5-10 microM EAV-specific PPMO produced no detectable virus. The PPMO-cured HeLa cells were free of infectious virus, viral antigen and EAV RNA as measured by plaque assay, indirect immunofluorescence assay and RT-PCR, respectively. Furthermore, when re-challenged with EAV at several passages after discontinuation of PPMO treatments, PPMO-cured HeLa cells were found to be refractory to re-infection and to the re-establishment of viral persistence. While these findings demonstrate that PPMO can be used to eliminate persistent EAV infection in cell culture, the efficacy of PPMO against EAV in vivo remains to be addressed.


Asunto(s)
Antivirales/farmacología , Equartevirus/efectos de los fármacos , Equartevirus/crecimiento & desarrollo , Morfolinas/farmacología , Oligonucleótidos Antisentido/farmacología , Péptidos/farmacología , Animales , Antivirales/química , Células HeLa , Caballos , Humanos , Morfolinas/química , Morfolinos , Oligonucleótidos Antisentido/química , Péptidos/química
11.
Virus Res ; 140(1-2): 49-56, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19059443

RESUMEN

Measles virus (MeV) is a highly contagious human pathogen. Despite the success of measles vaccination programs, measles is still responsible for an estimated 245,000 deaths each year. There are currently no antiviral compounds available for the treatment of measles. Peptide-conjugated phosphorodiamidate morpholino oligomers (PPMO) are antisense compounds that enter cells readily and can interfere with mRNA function by steric blocking. A panel of PPMO was designed to target various sequences of MeV RNA that are known to be important for viral replication. Five PPMO, targeting MeV genomic RNA or mRNA, inhibited the replication of MeV, in a dose-responsive and sequence-specific manner in cultured cells. One of the highly active PPMO (PPMO 454), targeting a conserved sequence in the translation start site of the mRNA coding for the nucleocapsid protein, inhibited multiple genotypes of MeV. This report provides evidence that PPMO treatment represents a promising approach for developing antiviral agents against measles and other paramyxoviruses.


Asunto(s)
Antivirales/farmacología , Virus del Sarampión/efectos de los fármacos , Morfolinas/farmacología , Péptidos/farmacología , Replicación Viral/efectos de los fármacos , Animales , Células CHO , Chlorocebus aethiops , Cricetinae , Cricetulus , Virus del Sarampión/fisiología , Morfolinos , ARN Mensajero/efectos de los fármacos , ARN Viral/efectos de los fármacos , Células Vero
12.
Curr Pharm Des ; 14(25): 2619-34, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18991679

RESUMEN

RNA virus infections cause immense human disease burdens globally, and few effective antiviral drugs are available for their treatment. Peptide-conjugated phosphorodiamidate morpholino oligomers (PPMO) are nuclease resistant and water-soluble single-stranded-DNA-analogues that can enter cells readily and act as steric-blocking antisense agents through stable duplex formation with complementary RNA. Recently there have been a number of publications documenting sequence-specific and dose-dependent inhibition of non-retroviral RNA virus infections by PPMO in both cell culture and murine experimental systems. PPMO have suppressed viral titers by several orders of magnitude in cell cultures, and have reduced viral replication in and/or increased survivorship of mice experimentally infected with poliovirus, coxsackievirus B3, dengue virus, West Nile virus, Venezuelan Equine encephalitis virus, respiratory syncytial virus, Ebola virus and influenza A virus. Along with evaluating PPMO efficacy and toxicity, these studies also explored PPMO mechanism of action, pharmacologic properties and the generation and characterization of resistant virus. Effective PPMO target sites in viral RNA have included regions of highly conserved sequence thought to be important in the pre-initiation or initiation of translation, or in long-range RNA-RNA interactions involved in viral RNA synthesis. These studies provide guidance for the design of steric-blocking antisense agents against RNA viruses, insights into viral molecular biology and novel strategies for the development of antiviral therapeutics. The purpose of this review is to summarize notable findings from the reports documenting antiviral activity by PPMO, with a focus on the specific regions of viral RNA that provided the most effective targets for PPMO-based inhibition of viral replication.


Asunto(s)
Morfolinas/química , Morfolinas/uso terapéutico , Péptidos/química , Péptidos/uso terapéutico , Infecciones por Virus ARN/tratamiento farmacológico , Replicación Viral/efectos de los fármacos , Animales , Antivirales/química , Antivirales/farmacología , Antivirales/uso terapéutico , Humanos , Morfolinas/farmacología , Morfolinos , Péptidos/farmacología , Infecciones por Virus ARN/metabolismo , Replicación Viral/fisiología
13.
Mol Ther ; 16(6): 1120-8, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18443602

RESUMEN

Respiratory syncytial virus (RSV) is a major cause of lower respiratory tract infection in infants, young children, and high-risk adults. Currently, there is no vaccine to prevent RSV infection, and the available therapeutic agents are of limited utility. Peptide-conjugated phosphorodiamidate morpholino oligomers (PPMOs) are a class of antisense agents that can enter cells readily and interfere with viral protein expression through steric blocking of complementary RNA. Two antisense PPMOs, designed to target sequence that includes the 5'-terminal region and translation start-site region of RSV L mRNA, were tested for anti-RSV activity in cultures of two human-airway cell lines. Both PPMOs showed minimal cytotoxicity and one of them, (AUG-2), reduced viral titers by >2.0 log(10). Intranasal (i.n.) treatment of BALB/c mice with AUG-2 PPMO before the RSV inoculation produced a reduction in viral titer of 1.2 log(10) in lung tissue at day 5 postinfection (p.i.), and attenuated pulmonary inflammation at day 7 postinfection. These data show that the AUG-2 PPMO possesses potent anti-RSV activity and is worthy of further investigation as a candidate for potential therapeutic application.


Asunto(s)
Técnicas de Transferencia de Gen , Terapia Genética/métodos , Infecciones por Virus Sincitial Respiratorio/metabolismo , Animales , Secuencia de Bases , Línea Celular Tumoral , Células Cultivadas , Codón , Femenino , Humanos , Pulmón/metabolismo , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , ARN Complementario/metabolismo , ARN Mensajero/metabolismo
14.
J Gen Virol ; 89(Pt 4): 939-948, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18343835

RESUMEN

Peptide-conjugated phosphorodiamidate morpholino oligomers (PPMO) are single-stranded nucleic acid-analogue antisense agents that enter cells readily and can reduce gene expression by steric blocking of complementary RNA (cRNA) sequences. Here, we tested a panel of PPMO designed to target conserved sequences in the RNA genome segments encoding polymerase subunits of a highly pathogenic mouse-adapted influenza A virus (SC35M; H7N7). Three PPMO, targeting the translation start site region of PB1 or NP mRNA or the 3'-terminal region of NP viral RNA (vRNA), potently inhibited virus replication in MDCK cells. Primer extension assays showed that treatment with any of the effective PPMO led to markedly reduced levels of mRNA, cRNA and vRNA. Initially, the potential toxicity of a range of intranasally administered PPMO doses was evaluated, by measuring their effect on body weight of uninfected mice. Subsequently, a non-toxic dosing regimen was used to investigate the effect of various PPMO on SC35M infection in a mouse model. Mice administered intranasal treatment of PPMO targeting the PB1-AUG region or NP vRNA, at 3 mug per dose, given once 3 h before and once 2 days after intranasal infection with 10xLD(50) of SC35M, showed a 2 log(10) reduction of viral titre in the lungs and 50 % survival for the 16 day duration of the experiment, whereas the NP-AUG-targeted PPMO treatment resulted in 30 % survival of an otherwise lethal infection. These data suggest that PPMO provide a useful reagent to investigate influenza virus molecular biology and may constitute a therapeutic strategy against highly pathogenic influenza viruses.


Asunto(s)
Antiinfecciosos/uso terapéutico , Subtipo H7N7 del Virus de la Influenza A , Virus de la Influenza A , Morfolinas/uso terapéutico , Nucleoproteínas/efectos de los fármacos , Infecciones por Orthomyxoviridae/prevención & control , Péptidos/uso terapéutico , Proteínas de Unión al ARN/efectos de los fármacos , Proteínas del Núcleo Viral/efectos de los fármacos , Proteínas Virales/efectos de los fármacos , Administración Intranasal , Animales , Antiinfecciosos/administración & dosificación , Antiinfecciosos/síntesis química , Antiinfecciosos/toxicidad , Línea Celular , Perros , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Evaluación Preclínica de Medicamentos , Marcación de Gen , Genes Virales/genética , Virus de la Influenza A/genética , Ratones , Ratones Endogámicos BALB C , Morfolinas/administración & dosificación , Morfolinas/síntesis química , Morfolinas/toxicidad , Morfolinos , Proteínas de la Nucleocápside , Nucleoproteínas/genética , Péptidos/administración & dosificación , Péptidos/síntesis química , Proteínas de Unión al ARN/genética , Proteínas del Núcleo Viral/genética , Proteínas Virales/genética , Proteínas Virales/toxicidad
15.
Mol Cancer Ther ; 7(3): 712-20, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18347156

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV), also known as human herpesvirus 8, is associated with several malignant disorders, including Kaposi's sarcoma, primary effusion lymphoma (PEL), and multicentric Castleman's disease. An early lytic gene of KSHV encodes viral interleukin-6 (vIL-6), a viral homologue of the proinflammatory cytokine and an autocrine/paracrine growth factor human IL-6. In this study, we examined the effects of suppressing vIL-6 expression in PEL cells with antisense peptide-conjugated phosphorodiamidate morpholino oligomers (PPMO). PPMO are ssDNA-analogues that have a modified backbone and enter cells readily. Treatment of PEL cells with a PPMO designed against vIL-6 mRNA led to a marked reduction in the proportion of vIL-6-positive cells detected by immunofluorescence assay. Analysis by Western blot confirmed a specific reduction in the vIL-6 protein level and showed that the reduction was dependent on the dose of vIL-6 PPMO. PEL cells treated with the vIL-6 PPMO exhibited reduced levels of cellular growth, IL-6 expression and KSHV DNA, and an elevated level of p21 protein. Treatment of PEL cells with a combination of two vIL-6 PPMO compounds targeting different sequences in the vIL-6 mRNA led to an inhibitory effect that was greater than that achieved with either PPMO alone. These results show that PPMO targeting vIL-6 mRNA can potently reduce vIL-6 protein translation and indicate that further exploration of these compounds in an animal model for potential clinical application is warranted.


Asunto(s)
Herpesvirus Humano 8/fisiología , Interleucina-6/antagonistas & inhibidores , Polímeros/farmacología , Western Blotting , Línea Celular Tumoral , Técnica del Anticuerpo Fluorescente , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , ARN Mensajero/genética , Replicación Viral
16.
Arch Virol ; 153(5): 929-37, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18369525

RESUMEN

New methods to combat influenza A virus (FLUAV) in humans and animals are needed. The H3N8 subtype virus was the cause of the pandemic of 1890 and has recently undergone cross-species transmission from horses to dogs in the USA. In 2007 H3N8 spread to Australia, a continent previously devoid of equine influenza. Here, we show that antisense-peptide-conjugated phosphorodiamidate morpholino oligomers (PPMOs), delivered by intranasal administration, are able to inhibit the replication of FLUAV A/Eq/Miami/1/63 (H3N8) in mice by over 95% compared to controls. Monitoring of body weight and immune cell infiltrates in the lungs of noninfected mice indicated that PPMO treatment was not toxic at a concentration shown to be effectively antiviral in vivo. In addition, we detected a naturally occurring mutation within the PPMO target site of a viral gene that may be the cause of resistance to one of the two antisense PPMO sequences tested. These data indicate that PPMOs targeting highly conserved regions of FLUAV are promising novel therapeutic candidates.


Asunto(s)
Subtipo H3N8 del Virus de la Influenza A/efectos de los fármacos , Subtipo H3N8 del Virus de la Influenza A/genética , Vacunas contra la Influenza/farmacología , Oligodesoxirribonucleótidos Antisentido/genética , Oligodesoxirribonucleótidos Antisentido/farmacología , Infecciones por Orthomyxoviridae/prevención & control , Administración Intranasal , Animales , Secuencia de Bases , Femenino , Genes Virales , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/genética , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Proteínas de la Nucleocápside , Nucleoproteínas/genética , Oligodesoxirribonucleótidos Antisentido/administración & dosificación , Infecciones por Orthomyxoviridae/virología , ARN Viral/genética , Proteínas de Unión al ARN/genética , Proteínas del Núcleo Viral/genética , Proteínas Virales/genética
17.
Antimicrob Agents Chemother ; 52(6): 1970-81, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18347107

RESUMEN

Members of the genera Enterovirus and Rhinovirus (family Picornaviridae) cause a wide range of human diseases. An established vaccine is available only for poliovirus, and no effective therapy is available for the treatment of infections caused by any pathogenic picornavirus. Peptide-conjugated phosphorodiamidate morpholino oligomers (PPMO) are single-stranded DNA-like antisense agents that readily enter cells. A panel of PPMO was tested for their antiviral activities against various picornaviruses. PPMO targeting conserved internal ribosome entry site (IRES) sequence were highly active against human rhinovirus type 14, coxsackievirus type B2, and poliovirus type 1 (PV1), reducing PV1 titers by up to 6 log(10) in cell cultures. Comparative sequence analysis led us to design a PPMO (EnteroX) targeting 22 nucleotides of IRES sequence that are perfectly conserved across greater than 99% of all human enteroviruses and rhinoviruses. EnteroX reduced PV1 replication in cell culture to an extent similar to that of other IRES-specific PPMO. Resistant PV1 arose in cell cultures after 12 passages in the presence of EnteroX and were found to have two mutations within the EnteroX target sequence. Nevertheless, cPVR transgenic mice treated once daily by intraperitoneal (i.p.) injection with EnteroX before and/or after i.p. infection with 3 x 10(8) PFU (three times the 50% lethal dose) of PV1 had an approximately 80% higher rate of survival than the controls. The viral titer in tissues taken at day 5 postinfection showed that animals in the EnteroX-treated group averaged over 3, 4, and 5 log(10) less virus in the small intestine, spinal cord, and brain, respectively, than the amount in the control animals. These results suggest that EnteroX may have broad therapeutic potential against entero- and rhinoviruses.


Asunto(s)
Antivirales/farmacología , Enterovirus/efectos de los fármacos , Morfolinas/farmacología , Péptidos/farmacología , Rhinovirus/efectos de los fármacos , Animales , Antivirales/uso terapéutico , Secuencia de Bases , Secuencia Conservada , Enterovirus/clasificación , Enterovirus/genética , Enterovirus/patogenicidad , Células HeLa , Humanos , Ratones , Datos de Secuencia Molecular , Morfolinas/química , Morfolinas/uso terapéutico , Morfolinos , Péptidos/química , Péptidos/genética , Péptidos/uso terapéutico , Poliomielitis/tratamiento farmacológico , Poliomielitis/mortalidad , Poliovirus/efectos de los fármacos , Poliovirus/patogenicidad , Rhinovirus/clasificación , Rhinovirus/genética , Rhinovirus/patogenicidad , Ribosomas/metabolismo , Replicación Viral
18.
Antiviral Res ; 77(2): 95-107, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17959259

RESUMEN

Porcine reproductive and respiratory syndrome (PRRS) has been devastating the global swine industry for more than a decade, and current strategies to control PRRS are inadequate. In this study we characterized the inhibition of PRRS virus (PRRSV) replication by antisense phosphorodiamidate morpholino oligomers (PMO). Of 12 peptide-conjugated PMO (PPMO), four were found to be highly effective at inhibiting PRRSV replication in cell culture in a dose-dependant and sequence-specific manner. PPMO 5UP2 and 5HP are complementary to sequence in the 5' end of the PRRSV genome, and 6P1 and 7P1 to sequence in the translation initiation regions of ORF6 and ORF7, respectively. Treatment of cells with 5UP2 or 5HP caused a 4.5log(10) reduction in PRRSV yield, compared to a control PPMO. Combination of 6P1 and 7P1 led to higher level reduction than 6P1 or 7P1 alone. 5UP2, 5HP, and a combination of 6P1 and 7P1 inhibited PRRSV replication in porcine alveolar macrophages and protected the cells from PRRSV-induced cytopathic effect. Northern blot and real-time RT-PCR results demonstrated that the effective PPMO led to a reduction of PRRSV RNA level. 5UP2 and 5HP inhibited virus replication of 10 other strains of PRRSV. Results from this study suggest potential applications of PPMO for PRRS control.


Asunto(s)
Antivirales/farmacología , Morfolinas/farmacología , Péptidos/farmacología , Virus del Síndrome Respiratorio y Reproductivo Porcino/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Secuencia de Bases , Línea Celular , Supervivencia Celular/efectos de los fármacos , Sistema Libre de Células , Técnica del Anticuerpo Fluorescente Indirecta , Genes Reporteros , Humanos , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/virología , Ratones , Morfolinas/síntesis química , Morfolinos , Oligonucleótidos Antisentido/farmacología , Reacción en Cadena de la Polimerasa , Biosíntesis de Proteínas/efectos de los fármacos , ARN Viral/biosíntesis , Alineación de Secuencia , Porcinos
19.
Adv Drug Deliv Rev ; 60(4-5): 517-29, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18037527

RESUMEN

Charge neutral steric block oligonucleotide analogues, such as peptide nucleic acids (PNA) or phosphorodiamidate morpholino oligomers (PMO), have promising biological and pharmacological properties for antisense applications, such as for example in mRNA splicing redirection. However, cellular uptake of free oligomers is poor and the utility of conjugates of PNA or PMO to cell penetrating peptides (CPP), such as Tat or Penetratin, is limited by endosomal sequestration. Two new families of arginine-rich CPPs named (R-Ahx-R)(4) AhxB and R(6)Pen allow efficient nuclear delivery of splice correcting PNA and PMO at micromolar concentrations in the absence of endosomolytic agents. The in vivo efficacy of (R-Ahx-R)(4) AhxB PMO conjugates has been demonstrated in mouse models of Duchenne muscular dystrophy and in various viral infections.


Asunto(s)
Oligonucleótidos/química , Péptidos/química , Animales , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular , Sistemas de Liberación de Medicamentos/métodos , Humanos , Estructura Molecular , Ácidos Nucleicos de Péptidos/administración & dosificación , Ácidos Nucleicos de Péptidos/química , Ácidos Nucleicos de Péptidos/farmacocinética , Péptidos/administración & dosificación , Péptidos/farmacocinética
20.
Antimicrob Agents Chemother ; 51(7): 2470-82, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17485503

RESUMEN

We characterize in vitro resistance to and demonstrate the in vivo efficacy of two antisense phosphorodiamidate morpholino oligomers (PMOs) against West Nile virus (WNV). Both PMOs were conjugated with an Arg-rich peptide. One peptide-conjugated PMO (PPMO) binds to the 5' terminus of the viral genome (5'-end PPMO); the other targets an essential 3' RNA element required for genome cyclization (3' conserved sequence I [3' CSI] PPMO). The 3' CSI PPMO displayed a broad spectrum of antiflavivirus activity, suppressing WNV, Japanese encephalitis virus, and St. Louis encephalitis virus, as demonstrated by reductions in viral titers of 3 to 5 logs in cell cultures, likely due to the absolute conservation of the 3' CSI PPMO-targeted sequences among these viruses. The selection and sequencing of PPMO-resistant WNV showed that the 5'-end-PPMO-resistant viruses contained two to three mismatches within the PPMO-binding site whereas the 3' CSI PPMO-resistant viruses accumulated mutations outside the PPMO-targeted region. The mutagenesis of a WNV infectious clone demonstrated that the mismatches within the PPMO-binding site were responsible for the 5'-end PPMO resistance. In contrast, a U insertion or a G deletion located within the 3'-terminal stem-loop of the viral genome was the determinant of the 3' CSI PPMO resistance. In a mouse model, both the 5'-end and 3' CSI PPMOs (administered at 100 or 200 microg/day) partially protected mice from WNV disease, with minimal to no PPMO-mediated toxicity. A higher treatment dose (300 microg/day) caused toxicity. Unconjugated PMOs (3 mg/day) showed neither efficacy nor toxicity, suggesting the importance of the peptide conjugate for efficacy. The results suggest that a modification of the peptide conjugate composition to reduce its toxicity yet maintain its ability to effectively deliver PMO into cells may improve PMO-mediated therapy.


Asunto(s)
Antivirales/farmacología , Morfolinas/farmacología , Selección Genética , Replicación Viral/efectos de los fármacos , Virus del Nilo Occidental/efectos de los fármacos , Animales , Arginina/química , Secuencia de Bases , Línea Celular , Chlorocebus aethiops , Codón , Secuencia Conservada , Cricetinae , Relación Dosis-Respuesta a Droga , Femenino , Técnicas In Vitro , Inyecciones Intraperitoneales , Inyecciones Subcutáneas , Riñón/citología , Cinética , Ratones , Ratones Endogámicos C3H , Datos de Secuencia Molecular , Morfolinas/administración & dosificación , Morfolinas/síntesis química , Morfolinas/química , Morfolinas/uso terapéutico , Morfolinos , Mutación , Conformación de Ácido Nucleico , Sistemas de Lectura Abierta , Péptidos/química , ARN Viral/genética , Células Vero , Ensayo de Placa Viral , Virus del Nilo Occidental/genética , Virus del Nilo Occidental/crecimiento & desarrollo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA