Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 36(6): 933-46, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22749352

RESUMEN

The mitochondrial protein MAVS (also known as IPS-1, VISA, and CARDIF) interacts with RIG-I-like receptors (RLRs) to induce type I interferon (IFN-I). NLRX1 is a mitochondrial nucleotide-binding, leucine-rich repeats (NLR)-containing protein that attenuates MAVS-RLR signaling. Using Nlrx1(-/-) cells, we confirmed that NLRX1 attenuated IFN-I production, but additionally promoted autophagy during viral infection. This dual function of NLRX1 paralleled the previously described functions of the autophagy-related proteins Atg5-Atg12, but NLRX1 did not associate with Atg5-Atg12. High-throughput quantitative mass spectrometry and endogenous protein-protein interaction revealed an NLRX1-interacting partner, mitochondrial Tu translation elongation factor (TUFM). TUFM interacted with Atg5-Atg12 and Atg16L1 and has similar functions as NLRX1 by inhibiting RLR-induced IFN-I but promoting autophagy. In the absence of NLRX1, increased IFN-I and decreased autophagy provide an advantage for host defense against vesicular stomatitis virus. This study establishes a link between an NLR protein and the viral-induced autophagic machinery via an intermediary partner, TUFM.


Asunto(s)
Autofagia/fisiología , Interferón Tipo I/biosíntesis , Proteínas Mitocondriales/fisiología , Factor Tu de Elongación Peptídica/fisiología , Proteínas Adaptadoras Transductoras de Señales/fisiología , Secuencia de Aminoácidos , Animales , Proteína 12 Relacionada con la Autofagia , Proteína 5 Relacionada con la Autofagia , Proteínas Relacionadas con la Autofagia , Proteínas Portadoras/fisiología , Citocinas/biosíntesis , Citocinas/genética , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/fisiología , Fibroblastos/metabolismo , Regulación de la Expresión Génica/inmunología , Células HEK293 , Humanos , Interferón Tipo I/genética , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/inmunología , Ratones , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/fisiología , Proteínas Mitocondriales/química , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/genética , Datos de Secuencia Molecular , Complejos Multiproteicos/fisiología , Factor Tu de Elongación Peptídica/química , Mapeo de Interacción de Proteínas , Proteínas/fisiología , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/fisiología , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Organismos Libres de Patógenos Específicos , Vesiculovirus/fisiología
2.
Immunity ; 30(4): 556-65, 2009 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-19362020

RESUMEN

The nucleotide-binding domain and leucine-rich-repeat-containing (NLR) family of pattern-recognition molecules mediate host immunity to various pathogenic stimuli. However, in vivo evidence for the involvement of NLR proteins in viral sensing has not been widely investigated and remains controversial. As a test of the physiologic role of the NLR molecule NLRP3 during RNA viral infection, we explored the in vivo role of NLRP3 inflammasome components during influenza virus infection. Mice lacking Nlrp3, Pycard, or caspase-1, but not Nlrc4, exhibited dramatically increased mortality and a reduced immune response after exposure to the influenza virus. Utilizing analogs of dsRNA (poly(I:C)) and ssRNA (ssRNA40), we demonstrated that an NLRP3-mediated response could be activated by RNA species. Mechanistically, NLRP3 inflammasome activation by the influenza virus was dependent on lysosomal maturation and reactive oxygen species (ROS). Inhibition of ROS induction eliminated IL-1beta production in animals during influenza infection. Together, these data place the NLRP3 inflammasome as an essential component in host defense against influenza infection through the sensing of viral RNA.


Asunto(s)
Proteínas Portadoras/fisiología , Exosomas/inmunología , Inmunidad Innata , Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/inmunología , ARN Viral , Virosis/inmunología , Animales , Proteínas Portadoras/genética , Línea Celular , Humanos , Subtipo H1N1 del Virus de la Influenza A , Subtipo H3N2 del Virus de la Influenza A , Gripe Humana/inmunología , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR
3.
Toxicol Pathol ; 46(6): 706-718, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30045675

RESUMEN

Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death worldwide; however, the mutational properties of HCC-associated carcinogens remain largely uncharacterized. We hypothesized that mechanisms underlying chemical-induced HCC can be characterized by evaluating the mutational spectra of these tumors. To test this hypothesis, we performed exome sequencing of B6C3F1/N HCCs that arose either spontaneously in vehicle controls ( n = 3) or due to chronic exposure to gingko biloba extract (GBE; n = 4) or methyleugenol (MEG; n = 3). Most archived tumor samples are available as formalin-fixed paraffin-embedded (FFPE) blocks, rather than fresh-frozen (FF) samples; hence, exome sequencing from paired FF and FFPE samples was compared. FF and FFPE samples showed 63% to 70% mutation concordance. Multiple known (e.g., Ctnnb1T41A, BrafV637E) and novel (e.g., Erbb4C559S, Card10A700V, and Klf11P358L) mutations in cancer-related genes were identified. The overall mutational burden was greater for MEG than for GBE or spontaneous HCC samples. To characterize the mutagenic mechanisms, we analyzed the mutational spectra in the HCCs according to their trinucleotide motifs. The MEG tumors clustered closest to Catalogue of Somatic Mutations in Cancer signatures 4 and 24, which are, respectively, associated with benzo(a)pyrene- and aflatoxin-induced HCCs in humans. These results establish a novel approach for classifying liver carcinogens and understanding the mechanisms of hepatocellular carcinogenesis.


Asunto(s)
Carcinógenos/toxicidad , Exoma/genética , Perfilación de la Expresión Génica , Neoplasias Hepáticas Experimentales/genética , Hígado/efectos de los fármacos , Mutación , Análisis de Secuencia de ADN/métodos , Animales , Criopreservación , ADN de Neoplasias/genética , Eugenol/análogos & derivados , Eugenol/toxicidad , Femenino , Formaldehído/química , Ginkgo biloba , Hígado/patología , Neoplasias Hepáticas Experimentales/inducido químicamente , Neoplasias Hepáticas Experimentales/patología , Masculino , Ratones Endogámicos , Adhesión en Parafina , Extractos Vegetales/toxicidad , Reproducibilidad de los Resultados , Fijación del Tejido
4.
J Biol Chem ; 289(31): 21716-26, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24939850

RESUMEN

The induction of inflammatory cytokines such as IL-1ß is associated with the progression of human immunodeficiency virus, type 1 (HIV-1) disease or AIDS. Unlike most inflammatory cytokines that are regulated by NF-κB at the transcriptional level, production of mature IL-1ß also depends on inflammasome activation. The mechanism by which HIV-1 induces pro-IL-1ß expression and activates inflammasomes to cleave pro-IL-1ß into its bioactive form is not clearly defined. We report here that HIV-1 infection in human monocytes efficiently induced IL-1ß expression and inflammasome activation. Toll-like receptor 8 (TLR8) was required for inducing pro-IL-1ß expression, whereas the NLRP3 inflammasome was required for IL-1ß maturation and release. Furthermore, the lysosomal protease cathepsin B and HIV-1 induced production of reactive oxygen species were critical for HIV-induced inflammasome activation and IL-1ß production. HIV-1 entry, reverse transcription, and integration were all required for both pro-IL-1ß expression and inflammasome activation. Finally, we show that HIV-1-derived RNA was sufficient to induce both pro-IL-1ß expression and inflammasome activation. We conclude that HIV-1 infection induced the expression of pro-IL-1ß via TLR8-mediated mechanisms and activated caspase-1 through the NLRP3 inflammasome to cleave pro-IL-1ß into bioactive IL-1ß. These findings help to elucidate mechanisms of HIV-1 disease progression and identify novel targets for treating HIV-1 induced inflammation and immune activation.


Asunto(s)
Proteínas Portadoras/metabolismo , Infecciones por VIH/metabolismo , Inflamasomas/metabolismo , Interleucina-1beta/biosíntesis , Monocitos/metabolismo , Receptor Toll-Like 8/fisiología , Catepsina B/biosíntesis , Técnicas de Silenciamiento del Gen , VIH-1/genética , Humanos , Proteína con Dominio Pirina 3 de la Familia NLR , ARN Viral/genética , Especies Reactivas de Oxígeno/metabolismo , Receptor Toll-Like 8/genética
5.
J Biol Chem ; 287(39): 32791-9, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22843689

RESUMEN

The interleukin (IL)-1ß-processing inflammasome has recently been identified as a target for pathogenic evasion of the inflammatory response by a number of bacteria and viruses. We postulated that the periodontal pathogen, Porphyromonas gingivalis may suppress the inflammasome as a mechanism for its low immunogenicity and pathogenic synergy with other, more highly immunogenic periodontal bacteria. Our results show that P. gingivalis lacks signaling capability for the activation of the inflammasome in mouse macrophages. Furthermore, P. gingivalis can suppress inflammasome activation by another periodontal bacterium, Fusobacterium nucleatum. This repression affects IL-1ß processing, as well as other inflammasome-mediated processes, including IL-18 processing and cell death, in both human and mouse macrophages. F. nucleatum activates IL-1ß processing through the Nlrp3 inflammasome; however, P. gingivalis repression is not mediated through reduced levels of inflammasome components. P. gingivalis can repress Nlrp3 inflammasome activation by Escherichia coli, and by danger-associated molecular patterns and pattern-associated molecular patterns that mediate activation through endocytosis. However, P. gingivalis does not suppress Nlrp3 inflammasome activation by ATP or nigericin. This suggests that P. gingivalis may preferentially suppress endocytic pathways toward inflammasome activation. To directly test whether P. gingivalis infection affects endocytosis, we assessed the uptake of fluorescent particles in the presence or absence of P. gingivalis. Our results show that P. gingivalis limits both the number of cells taking up beads and the number of beads taken up for bead-positive cells. These results provide a novel mechanism of pathogen-mediated inflammasome inhibition through the suppression of endocytosis.


Asunto(s)
Infecciones por Bacteroidaceae/inmunología , Endocitosis/inmunología , Inflamasomas/inmunología , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Porphyromonas gingivalis/inmunología , Animales , Proteínas Portadoras/inmunología , Células Cultivadas , Técnicas de Cocultivo , Escherichia coli/inmunología , Fusobacterium/inmunología , Humanos , Macrófagos/microbiología , Macrófagos/patología , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR
6.
J Immunol ; 186(3): 1333-7, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21191067

RESUMEN

The nucleotide-binding domain leucine-rich repeat-containing proteins, NLRs, are intracellular sensors of pathogen-associated molecular patterns and damage-associated molecular patterns. A subgroup of NLRs can form inflammasome complexes, which facilitate the maturation of procaspase 1 to caspase 1, leading to IL-1ß and IL-18 cleavage and secretion. NLRC5 is predominantly expressed in hematopoietic cells and has not been studied for inflammasome function. RNA interference-mediated knockdown of NLRC5 nearly eliminated caspase 1, IL-1ß, and IL-18 processing in response to bacterial infection, pathogen-associated molecular patterns, and damage-associated molecular patterns. This was confirmed in primary human monocytic cells. NLRC5, together with procaspase 1, pro-IL-1ß, and the inflammasome adaptor ASC, reconstituted inflammasome activity that showed cooperativity with NLRP3. The range of pathogens that activate NLRC5 inflammasome overlaps with those that activate NLRP3. Furthermore, NLRC5 biochemically associates with NLRP3 in a nucleotide-binding domain-dependent but leucine-rich repeat-inhibitory fashion. These results invoke a model in which NLRC5 interacts with NLRP3 to cooperatively activate the inflammasome.


Asunto(s)
Inflamasomas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/fisiología , Animales , Proteínas Portadoras/fisiología , Línea Celular Transformada , Línea Celular Tumoral , Escherichia coli/inmunología , Escherichia coli/patogenicidad , Regulación de la Expresión Génica/inmunología , Humanos , Inmunidad Innata , Inflamasomas/biosíntesis , Inflamasomas/genética , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Leucina/metabolismo , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR , Estructura Terciaria de Proteína/genética , Shigella flexneri/inmunología , Shigella flexneri/patogenicidad , Staphylococcus aureus/inmunología , Staphylococcus aureus/patogenicidad
7.
J Biol Chem ; 286(22): 19605-16, 2011 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-21487011

RESUMEN

ASC/PYCARD is a common adaptor for a diverse set of inflammasomes that activate caspase-1, most prominently the NLR-based inflammasome. Mounting evidence indicates that ASC and these NLRs also elicit non-overlapping functions, but the molecular basis for this difference is unclear. To address this, we performed microarray and network analysis of ASC shRNA knockdown cells. In pathogen-infected cells, an ASC-dependent interactome is centered on the mitogen-activated protein kinase (MAPK) ERK and on multiple chemokines. ASC did not affect the expression of MAPK but affected its phosphorylation by pathogens and Toll-like receptor agonists via suppression of the dual-specificity phosphatase, DUSP10/MKP5. Chemokine induction, DUSP function, and MAPK phosphorylation were independent of caspase-1 and IL-1ß. MAPK activation by pathogen was abrogated in Asc(-/-) but not Nlrp3(-/-), Nlrc4(-/-), or Casp1(-/-) macrophages. These results demonstrate a function for ASC that is distinct from the inflammasome in modulating MAPK activity and chemokine expression and further identify DUSP10 as a novel ASC target.


Asunto(s)
Quimiocinas/biosíntesis , Proteínas del Citoesqueleto/metabolismo , Fosfatasas de Especificidad Dual/metabolismo , Inflamasomas/metabolismo , Macrófagos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Proteínas Adaptadoras de Señalización CARD , Línea Celular , Quimiocinas/genética , Proteínas del Citoesqueleto/genética , Fosfatasas de Especificidad Dual/genética , Activación Enzimática/fisiología , Técnicas de Silenciamiento del Gen , Humanos , Inflamasomas/genética , Macrófagos/citología , Ratones , Ratones Noqueados , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/genética
8.
J Immunol ; 185(9): 5476-85, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20921527

RESUMEN

Francisella tularensis is a facultative intracellular pathogen and potential biothreat agent. Evasion of the immune response contributes to the extraordinary virulence of this organism although the mechanism is unclear. Whereas wild-type strains induced low levels of cytokines, an F. tularensis ripA deletion mutant (LVSΔripA) provoked significant release of IL-1ß, IL-18, and TNF-α by resting macrophages. IL-1ß and IL-18 secretion was dependent on inflammasome components pyrin-caspase recruitment domain/apoptotic speck-containing protein with a caspase recruitment domain and caspase-1, and the TLR/IL-1R signaling molecule MyD88 was required for inflammatory cytokine synthesis. Complementation of LVSΔripA with a plasmid encoding ripA restored immune evasion. Similar findings were observed in a human monocytic line. The presence of ripA nearly eliminated activation of MAPKs including ERK1/2, JNK, and p38, and pharmacologic inhibitors of these three MAPKs reduced cytokine induction by LVSΔripA. Animals infected with LVSΔripA mounted a stronger IL-1ß and TNF-α response than that of mice infected with wild-type live vaccine strain. This analysis revealed novel immune evasive mechanisms of F. tularensis.


Asunto(s)
Francisella tularensis/patogenicidad , Genes Bacterianos/inmunología , Inflamación/genética , Macrófagos/inmunología , Proteínas Quinasas Activadas por Mitógenos/genética , Transducción de Señal/genética , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Western Blotting , Citocinas/biosíntesis , Ensayo de Inmunoadsorción Enzimática , Femenino , Francisella tularensis/genética , Francisella tularensis/inmunología , Genes Bacterianos/genética , Humanos , Evasión Inmune/genética , Evasión Inmune/inmunología , Inflamación/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/inmunología , Transducción de Señal/inmunología , Tularemia/genética , Tularemia/inmunología
9.
J Immunol ; 183(3): 2008-15, 2009 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-19587006

RESUMEN

Bacterial infection elicits a range of beneficial as well as detrimental host inflammatory responses. Key among these responses are macrophage/monocyte necrosis, release of the proinflammatory factor high-mobility group box 1 protein (HMGB1), and induction of the cytokine IL-1. Although the control of IL-1beta has been well studied, processes that control macrophage cell death and HMGB1 release in animals are poorly understood. This study uses Klebsiella pneumonia as a model organism because it elicits all three responses in vivo. The regulation of these responses is studied in the context of the inflammasome components NLRP3 and ASC, which are important for caspase-1 activation and IL-1beta release. Using a pulmonary infection model that reflects human infection, we show that K. pneumonia-induced mouse macrophage necrosis, HMGB1, and IL-1beta release are dependent on NLRP3 and ASC. K. pneumoniae infection of mice lacking Nlrp3 results in decreased lung inflammation and reduced survival relative to control, indicating the overall protective role of this gene. Macrophage/monocyte necrosis and HMGB1 release are controlled independently of caspase-1, suggesting that the former two responses are separable from inflammasome-associated functions. These results provide critical in vivo validation that the physiologic role of NLRP3 and ASC is not limited to inflammasome formation.


Asunto(s)
Proteínas Portadoras/fisiología , Caspasa 1/metabolismo , Proteínas del Citoesqueleto/fisiología , Proteína HMGB1/metabolismo , Neumonía/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Proteínas Adaptadoras de Señalización CARD , Modelos Animales de Enfermedad , Inflamación/metabolismo , Inflamación/microbiología , Interleucina-1beta/metabolismo , Klebsiella , Macrófagos/microbiología , Macrófagos/patología , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Necrosis , Neumonía/microbiología , Neumonía/patología
10.
J Immunol ; 182(4): 2395-404, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19201894

RESUMEN

Periodontal disease is a chronic inflammatory disorder that leads to the destruction of tooth-supporting tissue and affects 10-20 million people in the U.S. alone. The oral pathogen Porphyromonas gingivalis causes inflammatory host response leading to periodontal and other secondary inflammatory diseases. To identify molecular components that control host response to P. gingivalis in humans, roles for the NLR (NBD-LRR) protein, NLRP3 (cryopyrin, NALP3), and its adaptor apoptotic speck protein containing a C-terminal caspase recruitment domain (ASC) were studied. P. gingivalis strain A7436 induces cell death in THP1 monocytic cells and in human primary peripheral blood macrophages. This process is ASC and NLRP3 dependent and can be replicated by P. gingivalis LPS and Escherichia coli. P. gingivalis-induced cell death is caspase and IL-1 independent and exhibits morphological features consistent with necrosis including loss of membrane integrity and release of cellular content. Intriguingly, P. gingivalis-induced cell death is accompanied by the formation of ASC aggregation specks, a process not previously described during microbial infection. ASC specks are observed in P. gingivalis-infected primary human mononuclear cells and are dependent on NLRP3. This work shows that P. gingivalis causes ASC- and NLRP3-dependent necrosis, accompanied by ASC speck formation.


Asunto(s)
Infecciones por Bacteroidaceae/metabolismo , Proteínas Portadoras/metabolismo , Proteínas del Citoesqueleto/metabolismo , Macrófagos/microbiología , Monocitos/microbiología , Necrosis/metabolismo , Infecciones por Bacteroidaceae/inmunología , Infecciones por Bacteroidaceae/patología , Western Blotting , Proteínas Adaptadoras de Señalización CARD , Proteínas Portadoras/inmunología , Proteínas del Citoesqueleto/inmunología , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Microscopía Electrónica de Transmisión , Monocitos/inmunología , Monocitos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Necrosis/inmunología , Necrosis/microbiología , Porphyromonas gingivalis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Front Public Health ; 8: 582205, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33330323

RESUMEN

Background: Given the worldwide spread of the 2019 Novel Coronavirus (COVID-19), there is an urgent need to identify risk and protective factors and expose areas of insufficient understanding. Emerging tools, such as the Rapid Evidence Map (rEM), are being developed to systematically characterize large collections of scientific literature. We sought to generate an rEM of risk and protective factors to comprehensively inform areas that impact COVID-19 outcomes for different sub-populations in order to better protect the public. Methods: We developed a protocol that includes a study goal, study questions, a PECO statement, and a process for screening literature by combining semi-automated machine learning with the expertise of our review team. We applied this protocol to reports within the COVID-19 Open Research Dataset (CORD-19) that were published in early 2020. SWIFT-Active Screener was used to prioritize records according to pre-defined inclusion criteria. Relevant studies were categorized by risk and protective status; susceptibility category (Behavioral, Physiological, Demographic, and Environmental); and affected sub-populations. Using tagged studies, we created an rEM for COVID-19 susceptibility that reveals: (1) current lines of evidence; (2) knowledge gaps; and (3) areas that may benefit from systematic review. Results: We imported 4,330 titles and abstracts from CORD-19. After screening 3,521 of these to achieve 99% estimated recall, 217 relevant studies were identified. Most included studies concerned the impact of underlying comorbidities (Physiological); age and gender (Demographic); and social factors (Environmental) on COVID-19 outcomes. Among the relevant studies, older males with comorbidities were commonly reported to have the poorest outcomes. We noted a paucity of COVID-19 studies among children and susceptible sub-groups, including pregnant women, racial minorities, refugees/migrants, and healthcare workers, with few studies examining protective factors. Conclusion: Using rEM analysis, we synthesized the recent body of evidence related to COVID-19 risk and protective factors. The results provide a comprehensive tool for rapidly elucidating COVID-19 susceptibility patterns and identifying resource-rich/resource-poor areas of research that may benefit from future investigation as the pandemic evolves.


Asunto(s)
Investigación Biomédica/estadística & datos numéricos , COVID-19/epidemiología , Interpretación Estadística de Datos , Pandemias/estadística & datos numéricos , Factores Protectores , Informe de Investigación , Humanos , Factores de Riesgo
12.
Antivir Ther ; 22(3): 247-255, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28121292

RESUMEN

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.


Asunto(s)
Condiloma Acuminado/tratamiento farmacológico , Condiloma Acuminado/virología , Papillomaviridae/efectos de los fármacos , Infecciones por Papillomavirus/tratamiento farmacológico , Infecciones por Papillomavirus/virología , Ribonucleasas/uso terapéutico , Administración Tópica , Adulto , Animales , Línea Celular , Células Cultivadas , Terapia Combinada , Condiloma Acuminado/patología , Relación Dosis-Respuesta a Droga , Humanos , Kappapapillomavirus/efectos de los fármacos , Kappapapillomavirus/genética , Masculino , Ratones , Persona de Mediana Edad , Papillomaviridae/clasificación , Papillomaviridae/genética , Infecciones por Papillomavirus/patología , Conejos , Ribonucleasas/farmacología , Resultado del Tratamiento , Adulto Joven
13.
BMC Biotechnol ; 6: 7, 2006 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-16433925

RESUMEN

BACKGROUND: RNA interference (RNAi) technology is a powerful methodology recently developed for the specific knockdown of targeted genes. RNAi is most commonly achieved either transiently by transfection of small interfering (si) RNA oligonucleotides, or stably using short hairpin (sh) RNA expressed from a DNA vector or virus. Much controversy has surrounded the development of rules for the design of effective siRNA oligonucleotides; and whether these rules apply to shRNA is not well characterized. RESULTS: To determine whether published algorithms for siRNA oligonucleotide design apply to shRNA, we constructed 27 shRNAs from 11 human genes expressed stably using retroviral vectors. We demonstrate an efficient method for preparing wild-type and mutant control shRNA vectors simultaneously using oligonucleotide hybrids. We show that sequencing through shRNA vectors can be problematic due to the intrinsic secondary structure of the hairpin, and we determine a strategy for effective sequencing by using a combination of modified BigDye chemistries and DNA relaxing agents. The efficacy of knockdown for the 27 shRNA vectors was evaluated against six published algorithms for siRNA oligonucleotide design. Our results show that none of the scoring algorithms can explain a significant percentage of variance in shRNA knockdown efficacy as assessed by linear regression analysis or ROC curve analysis. Application of a modification based on the stability of the 6 central bases of each shRNA provides fair-to-good predictions of knockdown efficacy for three of the algorithms. Analysis of an independent set of data from 38 shRNAs pooled from previous publications confirms these findings. CONCLUSION: The use of mixed oligonucleotide pairs provides a time and cost efficient method of producing wild type and mutant control shRNA vectors. The addition to sequencing reactions of a combination of mixed dITP/dGTP chemistries and DNA relaxing agents enables read through the intrinsic secondary structure of problematic shRNA vectors. Six published algorithms for siRNA oligonucleotide design that were tested in this study show little or no efficacy at predicting shRNA knockdown outcome. However, application of a modification based on the central shRNA stability should provide a useful improvement to the design of effective shRNA vectors.


Asunto(s)
Algoritmos , Silenciador del Gen , Marcación de Gen/métodos , Sondas ARN/genética , ARN Interferente Pequeño/genética , Análisis de Secuencia de ARN/métodos , Diseño de Fármacos , Vectores Genéticos/genética , Alineación de Secuencia/métodos
14.
Cancer Res ; 63(16): 5095-104, 2003 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-12941840

RESUMEN

A combination of paclitaxel (Taxol) and mitogen-activated protein/extracellular signal-regulated kinase kinase (MEK/Erk) inhibitor represents a rational new approach to chemotherapy. We performed Affymetrix microarray analysis to understand the global effects of this combination in lung carcinoma. Genes involved in cell cycle control, apoptosis, adhesion, proliferation, invasion, and metastasis were modulated. We observed similar patterns of gene modulation in ovarian and melanoma cell lines, indicating the general applicability of these findings. Functional genomic analysis identified two genes as new targets of drug-induced tumor apoptosis. The MGSA/Gro1 gene, important in melanoma growth, was induced by paclitaxel and reduced by MEK inhibition. Blockage of paclitaxel-induced melanoma growth stimulatory activity significantly reduced melanoma growth. Additionally, the expression of topoisomerase III beta, which exhibited a clear pattern of gene reduction by a combination of the two drugs, was significantly increased (5.7-fold) in primary lung cancers but not adjacent tissues. These findings provide potential new biomarkers and gene targets for the development of improved cancer treatment.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Butadienos/administración & dosificación , Quimiocinas CXC , Inhibidores Enzimáticos/administración & dosificación , Perfilación de la Expresión Génica , Neoplasias Pulmonares/tratamiento farmacológico , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Nitrilos/administración & dosificación , Paclitaxel/administración & dosificación , Quimiocina CXCL1 , Quimiocinas/biosíntesis , Factores Quimiotácticos/biosíntesis , Dactinomicina/farmacología , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Melanoma/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Ováricas/genética
15.
Antiviral Res ; 132: 210-8, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27350309

RESUMEN

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.


Asunto(s)
Antivirales/farmacología , Ebolavirus/efectos de los fármacos , Fiebre Hemorrágica Ebola/virología , Ribonucleasas/farmacología , Animales , Línea Celular , Chlorocebus aethiops , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ebolavirus/fisiología , Femenino , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Fiebre Hemorrágica Ebola/mortalidad , Humanos , Ratones , ARN Viral , Células Vero , Carga Viral , Replicación Viral/efectos de los fármacos
16.
Clin Cancer Res ; 8(7): 2091-9, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12114408

RESUMEN

Paclitaxel (Taxol) activates a number of signal transduction pathways that lead to apoptosis. In contrast, paclitaxel also activates cell survival pathways, such as the Raf-mitogen-activated protein kinase kinase (MEK)-extracellular signal-regulated kinase (ERK) pathway. Previously, we have shown that inhibition of MEK combined with paclitaxel treatment causes an impressive enhancement of apoptosis in various tumor cell lines. Here, we find that the combination of paclitaxel with a MEK inhibitor leads to a dramatic inactivation of the antiapoptotic Akt (protein kinase B) kinase. The decrease in Akt is not reflected at the protein or mRNA level but rather attributed to kinase inactivation. To confirm that inactivation of Akt is significant, a constitutively active Akt mutant was introduced and shown to reverse tumor cell apoptosis. Further analysis upstream of Akt shows that treatment with the combination of paclitaxel and MEK inhibitor down-regulates PI3K activity more than either agent alone. The direct pharmacological inhibition of phosphatidylinositol 3-kinase (PI3K) similarly enhances paclitaxel-induced tumor apoptosis in a dose-dependent manner. Our results suggest the combination of paclitaxel and MEK inhibitor leads to down-regulation of the PI3K-Akt signaling in addition to the proapoptotic effects of paclitaxel and MEK inhibitor alone. Overall, these findings render the combined use of paclitaxel with MEK inhibitors, or paclitaxel with PI3K inhibitors, as a promising new strategy for cancer chemotherapy.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Paclitaxel/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Células Tumorales Cultivadas/patología , Northern Blotting , Butadienos/farmacología , Cromonas/farmacología , Quimioterapia Combinada , Factor de Crecimiento Epidérmico/farmacología , Humanos , Immunoblotting , Morfolinas/farmacología , Neurregulina-1/farmacología , Nitrilos/farmacología , Fosforilación , Plásmidos , Proteínas Proto-Oncogénicas c-akt , ARN Mensajero/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/enzimología
17.
Cell Rep ; 3(3): 678-88, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23434512

RESUMEN

MyD88, the intracellular adaptor of most TLRs, mediates either proinflammatory or immunosuppressive signaling that contributes to chronic inflammation-associated diseases. Although gene-specific chromatin modifications regulate inflammation, the role of MyD88 signaling in establishing such epigenetic landscapes under different inflammatory states remains elusive. Using quantitative proteomics to enumerate the inflammation-phenotypic constituents of the MyD88 interactome, we found that in endotoxin-tolerant macrophages, protein phosphatase 2A catalytic subunit α (PP2Ac) enhances its association with MyD88 and is constitutively activated. Knockdown of PP2Ac prevents suppression of proinflammatory genes and resistance to apoptosis. Through site-specific dephosphorylation, constitutively active PP2Ac disrupts the signal-promoting TLR4-MyD88 complex and broadly suppresses the activities of multiple proinflammatory/proapoptotic pathways as well, shifting proinflammatory MyD88 signaling to a prosurvival mode. Constitutively active PP2Ac translocated with MyD88 into the nuclei of tolerant macrophages establishes the immunosuppressive pattern of chromatin modifications and represses chromatin remodeling to selectively silence proinflammatory genes, coordinating the MyD88-dependent inflammation control at both signaling and epigenetic levels under endotoxin-tolerant conditions.


Asunto(s)
Epigénesis Genética/inmunología , Tolerancia Inmunológica/genética , Lipopolisacáridos/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Proteína Fosfatasa 2/metabolismo , Transporte Activo de Núcleo Celular , Animales , Apoptosis , Núcleo Celular/metabolismo , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Células HEK293 , Humanos , Macrófagos/inmunología , Ratones , Ratones Transgénicos , Factor 88 de Diferenciación Mieloide/genética , Fenotipo , Fosforilación , Unión Proteica , Proteína Fosfatasa 2/genética , Proteoma/metabolismo , Transducción de Señal , Receptor Toll-Like 4/metabolismo
18.
J Leukoc Biol ; 92(5): 1069-81, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22892107

RESUMEN

The Staphylococcus aureus pore-forming toxin PVL is most likely causative for life-threatening necrotizing infections, which are characterized by massive tissue inflammation and necrosis. Whereas the cytotoxic action of PVL on human neutrophils is already well established, the PVL effects on other sensitive cell types, such as monocytes and macrophages, are less clear. In this study, we used different types of human leukocytes (neutrophils, monocytes, macrophages, lymphocytes) to investigate cell-specific binding of PVL subunits and subsequent proinflammatory and cytotoxic effects. In all PVL-sensitive cells, we identified the binding of the subunit LukS-PV as the critical factor for PVL-induced cytotoxicity, which was followed by binding of LukF-PV. LukS-PV binds to monocytes, macrophages, and neutrophils but not to lymphocytes. Additionally, we showed that PVL binding to monocytes and macrophages leads to release of caspase-1-dependent proinflammatory cytokines IL-1ß and IL-18. PVL activates the NLRP3 inflammasome, a signaling complex of myeloid cells that is involved in caspase-1-dependent IL-1ß processing in response to pathogens and endogenous danger signals. Specific inhibition of this pathway at several steps significantly reduced inflammasome activation and subsequent pyronecrosis. Furthermore, we found that PAMPs and DAMPs derived from dying neutrophils can dramatically enhance this response by up-regulating pro-IL-1ß in monocytes/macrophages. This study analyzes a specific host signaling pathway that mediates PVL-induced inflammation and cytotoxicity, which has high relevance for CA-MRSA-associated and PVL-mediated pathogenic processes, such as necrotizing infections.


Asunto(s)
Toxinas Bacterianas/inmunología , Proteínas Portadoras/inmunología , Exotoxinas/inmunología , Inflamasomas/inmunología , Inflamación/inmunología , Leucocidinas/inmunología , Fagocitos/inmunología , Animales , Toxinas Bacterianas/metabolismo , Western Blotting , Exotoxinas/metabolismo , Humanos , Leucocidinas/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/inmunología , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/inmunología , Staphylococcus aureus/metabolismo , Transfección
19.
Curr Opin Immunol ; 23(5): 591-7, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21839623

RESUMEN

The inflammasome-forming NLRs are well characterized members of a protein complex mediating the activation of caspase-1 and the cleavage of pro-IL-1ß and pro-IL-18 into their active, secreted forms. New data suggest that components of the inflammasome cascade may have roles in influencing inflammasome-independent pathways of cytokine production. These influences on other immune cytokine pathways are complemented by data suggesting that non-inflammasome cytokines can influence the activation of the inflammasome, either directly or by influencing transcription of inflammasome components. The crosstalk between these cytokine cascades may lead to increased abilities for the cell to respond to diverse pathogen threats.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Inmunidad Innata , Inflamasomas/inmunología , Inflamación/inmunología , Interleucina-18/inmunología , Interleucina-1beta/inmunología , FN-kappa B/inmunología , Transducción de Señal/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Caspasa 1/genética , Caspasa 1/inmunología , Caspasa 1/metabolismo , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/inmunología , ARN Helicasas DEAD-box/metabolismo , Redes Reguladoras de Genes/inmunología , Humanos , Inflamasomas/genética , Inflamasomas/metabolismo , Inflamación/genética , Inflamación/metabolismo , Interleucina-18/genética , Interleucina-18/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , FN-kappa B/genética , FN-kappa B/metabolismo
20.
Science ; 331(6015): 330-4, 2011 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-21252346

RESUMEN

The NLR (nucleotide binding and oligomerization, leucine-rich repeat) family of proteins senses microbial infections and activates the inflammasome, a multiprotein complex that promotes microbial clearance. Kaposi's sarcoma-associated herpesvirus (KSHV) is linked to several human malignancies. We found that KSHV Orf63 is a viral homolog of human NLRP1. Orf63 blocked NLRP1-dependent innate immune responses, including caspase-1 activation and processing of interleukins IL-1ß and IL-18. KSHV Orf63 interacted with NLRP1, NLRP3, and NOD2. Inhibition of Orf63 expression resulted in increased expression of IL-1ß during the KSHV life cycle. Furthermore, inhibition of NLRP1 was necessary for efficient reactivation and generation of progeny virus. The viral homolog subverts the function of cellular NLRs, which suggests that modulation of NLR-mediated innate immunity is important for the lifelong persistence of herpesviruses.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Herpesvirus Humano 8/fisiología , Evasión Inmune , Inmunidad Innata , Inflamasomas/antagonistas & inhibidores , Proteínas Virales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Apoptosis , Proteínas Reguladoras de la Apoptosis/química , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Portadoras/metabolismo , Caspasa 1/metabolismo , Inhibidores de Caspasas , Línea Celular , Línea Celular Tumoral , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/inmunología , Humanos , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Datos de Secuencia Molecular , Monocitos/virología , Proteína con Dominio Pirina 3 de la Familia NLR , Proteínas NLR , Proteína Adaptadora de Señalización NOD2/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transfección , Proteínas Virales/química , Proteínas Virales/genética , Activación Viral , Latencia del Virus , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA