Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Am J Infect Control ; 51(4): 396-400, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-35870660

RESUMEN

BACKGROUND: Stabilized hypochlorous acid (HOCl) is increasingly used as a hospital disinfectant and antiseptic, yet its effect on N95 filtration facemask respirators (FFR) is unknown. These FFRs could also contribute to fomite-based transmission of nosocomial infections if worn for extended use between patient rooms. METHODS: Filtration performance of N95 FFR fabric swatches was assessed after various levels of HOCl exposure. N95 swatches were then contaminated with 108Escherichia coli or 108Staphylococci aureus and treated with HOCl solution, 70% ethyl alcohol, or normal saline. Surviving bacterial numbers were assessed by plate counts. RESULTS: The size-dependent filtration efficiency of HOCl-sprayed N95 FFR fabric ranged from 96% to 100%, showing no significant change. Flow resistance testing revealed almost no change compared to control. Submersion in HOCl, but not spraying, had an excellent bactericidal effect on contaminated swatches. DISCUSSION: The role of the outer hydrophobic layer of N95 FFRs is discussed regarding the effects of HOCl on filtration and bacterial decontamination. CONCLUSIONS: N95 material, sprayed with or briefly submerged in HOCl, maintained its filtration function. HOCl delivery by spray pump, however, would not accomplish decontamination of extended use FFRs between patient encounters. HOCl submersion of intact FFRs, contaminated with various hospital pathogens, is worth further study.


Asunto(s)
Respiradores N95 , Dispositivos de Protección Respiratoria , Humanos , Ácido Hipocloroso/farmacología , Máscaras , Descontaminación , Ventiladores Mecánicos , Filtración
2.
J Aerosol Med Pulm Drug Deliv ; 35(6): 291-295, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35960504

RESUMEN

Background: As an anti-inflammatory and antioxidant, sodium pyruvate significantly reduces inflammatory cytokines and oxygen radicals such as interleukin (IL) IL-6, IL-8, Monocyte Chemoattractant Protein-1, and hydrogen peroxide. Thus, sodium pyruvate holds promise as a treatment for many respiratory diseases, including allergic rhinitis (AR). Novel treatments for AR are needed as current medications, including steroids, often fail to treat severe symptoms. Methods: The data from five human clinical studies were analyzed to determine the effect of 20 mM sodium pyruvate nasal spray (N115) in patients with AR. Nasal inflammation scores were compared to a placebo control or a no-treatment baseline control. Three studies were open-labeled and two were appropriately blinded to both patients and clinicians using computer randomization of subjects. Results: The intranasal administration of sodium pyruvate significantly improved nasal inflammation scores in all five clinical trials of patients with AR (p < 0.0001 in all trials). Conclusions: These results give credence to the overall ability of sodium pyruvate, administered by nasal spray, to treat inflammation of the nasal airways.


Asunto(s)
Rinitis Alérgica Estacional , Rinitis Alérgica , Humanos , Administración por Inhalación , Administración Intranasal , Inflamación/tratamiento farmacológico , Rociadores Nasales , Piruvatos/uso terapéutico , Rinitis Alérgica/tratamiento farmacológico , Rinitis Alérgica Estacional/tratamiento farmacológico , Sodio/uso terapéutico
3.
Immunol Rev ; 297(1): 194-206, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32761626

RESUMEN

Infections with any pathogen can be severe and present with numerous complications caused by the pathogen or the host immune response to the invading microbe. However, coinfections, also called polymicrobial infections or secondary infections, can further exacerbate disease. Coinfections are more common than is often appreciated. In this review, we focus specifically on coinfections between viruses and other viruses, bacteria, parasites, or fungi. Importantly, innate immune signaling and innate immune cells that facilitate clearance of the initial viral infection can affect host susceptibility to coinfections. Understanding these immune imbalances may facilitate better diagnosis, prevention, and treatment of such coinfections.


Asunto(s)
Coinfección , Virosis , Virus , Bacterias , Humanos , Inmunidad Innata
4.
Virus Res ; 286: 198088, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32634445

RESUMEN

Pyruvate is the end product of glycolysis and transported into the mitochondria for use in the tricarboxylic acid (TCA) cycle. It is also a common additive in cell culture media. We discovered that inclusion of sodium pyruvate in culture media during infection of mouse bone marrow derived macrophages with influenza A virus impaired cytokine production (IL-6, IL-1ß, and TNF-α). Sodium pyruvate did not inhibit viral RNA replication. Instead, the addition of sodium pyruvate alters cellular metabolism and diminished mitochondrial reactive oxygen species (ROS) production and lowered immune signaling. Overall, sodium pyruvate affects the immune response produced by macrophages but does not inhibit virus replication.


Asunto(s)
Antiinflamatorios/farmacología , Inmunidad Innata/efectos de los fármacos , Inflamación/virología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ácido Pirúvico/farmacología , Animales , Células Cultivadas , Medios de Cultivo/química , Expresión Génica , Inflamasomas/efectos de los fármacos , Inflamasomas/metabolismo , Inflamación/inmunología , Virus de la Influenza A/efectos de los fármacos , Macrófagos/virología , Ratones , Ratones Endogámicos C57BL , Replicación Viral/efectos de los fármacos
6.
J Nat Prod ; 83(2): 344-353, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-31986046

RESUMEN

White-nose syndrome (WNS) is a devastating disease of hibernating bats caused by the fungus Pseudogymnoascus destructans. We obtained 383 fungal and bacterial isolates from the Soudan Iron Mine, an important bat hibernaculum in Minnesota, then screened this library for antifungal activity to develop biological control treatments for WNS. An extract from the fungus Oidiodendron truncatum was subjected to bioassay-guided fractionation, which led to the isolation of 14 norditerpene and three anthraquinone metabolites. Ten of these compounds were previously described in the literature, and here we present the structures of seven new norditerpene analogues. Additionally, this is the first report of 4-chlorophyscion from a natural source, previously identified as a semisynthetic product. The compounds PR 1388 and LL-Z1271α were the only inhibitors of P. destructans (MIC = 7.5 and 15 µg/mL, respectively). Compounds were tested for cytotoxicity against fibroblast cell cultures obtained from Myotis septentrionalis (northern long eared bat) and M. grisescens (gray bat) using a standard MTT viability assay. The most active antifungal compound, PR 1388, was nontoxic toward cells from both bat species (IC50 > 100 µM). We discuss the implications of these results in the context of the challenges and logistics of developing a substrate treatment or prophylactic for WNS.


Asunto(s)
Antifúngicos/química , Antifúngicos/farmacología , Ascomicetos/química , Diterpenos/química , Animales , Antifúngicos/aislamiento & purificación , Quirópteros/microbiología , Diterpenos/aislamiento & purificación , Hibernación , Minnesota
7.
PLoS One ; 14(2): e0212236, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30794604

RESUMEN

Viral-bacterial coinfections, such as with influenza A virus and Streptococcus pneumoniae (S.p.), are known to cause severe pneumonia. It is well known that the host response has an important role in disease. Interleukin-1ß (IL-1ß) is an important immune signaling cytokine responsible for inflammation and has been previously shown to contribute to disease severity in numerous infections. Other studies in mice indicate that IL-1ß levels are dramatically elevated during IAV-S.p. coinfection. However, the regulation of IL-1ß during coinfection is unknown. Here, we report the NLRP3 inflammasome is the major inflammasome regulating IL-1ß activation during coinfection. Furthermore, elevated IL-1ß mRNA expression is due to enhanced TLR2-MYD88 signaling, which increases the amount of pro-IL-1ß substrate for the inflammasome to process. Finally, NLRP3 and high IL-1ß levels were associated with increased bacterial load in the brain. Our results show the NLRP3 inflammasome is not protective during IAV-S.p. coinfection.


Asunto(s)
Coinfección/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Interleucina-1beta/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones Neumocócicas/inmunología , Transducción de Señal/inmunología , Streptococcus pneumoniae/inmunología , Receptor Toll-Like 2/inmunología , Animales , Encéfalo , Línea Celular , Embrión de Pollo , Coinfección/genética , Coinfección/patología , Interleucina-1beta/genética , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/patología , Infecciones Neumocócicas/genética , Infecciones Neumocócicas/patología , Transducción de Señal/genética , Receptor Toll-Like 2/genética
8.
Int Rev Cell Mol Biol ; 344: 139-172, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30798987

RESUMEN

The innate immune system detects the presence of pathogens based on detection of non-self. In other words, most pathogens possess intrinsic differences that can distinguish them from host cells. For example, bacteria and fungi have cell walls comprised of peptidoglycan and carbohydrates (like mannans), respectively. Germline encoded pattern recognition receptors (PRRs) of the Toll-like receptor (TLR) and C-type lectin receptor (CLR) family have the ability to detect such unique pathogen associated features. However, some TLRs and members of the RIG-I-like receptor (RLR), NOD-like receptor (NLR), or AIM2-like receptor (ALR) family can sense pathogen invasion based on pathogen nucleic acids. Nucleic acids are not unique to pathogens, thus raising the question of how such PRRs evolved to detect pathogens but not self. In this chapter, we will examine the PRRs that sense pathogen nucleic acids and subsequently activate the inflammasome signaling pathway. We will examine the selective mechanisms by which these receptors distinguish pathogens from "self" and discuss the importance of such pathways in disease development in animal models and human patients.


Asunto(s)
Autoantígenos/metabolismo , Infecciones/inmunología , Inflamasomas/metabolismo , Ácidos Nucleicos/metabolismo , Animales , Humanos , Evasión Inmune , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo
9.
Adv Microb Physiol ; 72: 65-115, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29778217

RESUMEN

Nitric oxide (NO) is a gaseous signalling molecule that plays diverse physiological functions including antimicrobial host defence. During microbial infection, NO is synthesized by inducible NO synthase (iNOS), which is expressed by host immune cells through the recognition of microbial pattern molecules. Therefore, sensing pathogens or their pattern molecules by pattern recognition receptors (PRRs), which are located at the cell surface, endosomal and phagosomal compartment, or in the cytosol, is key in inducing iNOS and eliciting antimicrobial host defence. A group of cytosolic PRRs is involved in inducing NO and other antimicrobial molecules by forming a molecular complex called the inflammasome. Assembled inflammasomes activate inflammatory caspases, such as caspase-1 and caspase-11, which in turn process proinflammatory cytokines IL-1ß and IL-18 into their mature forms and induce pyroptotic cell death. IL-1ß and IL-18 play a central role in immunity against microbial infection through activation and recruitment of immune cells, induction of inflammatory molecules, and regulation of antimicrobial mediators including NO. Interestingly, NO can also regulate inflammasome activity in an autocrine and paracrine manner. Here, we discuss molecular mechanisms of inflammasome formation and the inflammasome-mediated regulation of host defence responses during microbial infections.


Asunto(s)
Antiinfecciosos/metabolismo , Enfermedades Transmisibles/inmunología , Interacciones Huésped-Patógeno , Inflamasomas/metabolismo , Óxido Nítrico/metabolismo , Transducción de Señal , Animales , Humanos , Factores Inmunológicos/metabolismo , Piroptosis , Receptores de Reconocimiento de Patrones/metabolismo
10.
PLoS One ; 12(10): e0184629, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29028839

RESUMEN

Human Respiratory Syncytial Virus (HRSV) is a leading cause of bronchopneumonia in infants and the elderly. To date, knowledge of viral and host protein interactions within HRSV is limited and are critical areas of research. Here, we show that HRSV Matrix (M) protein interacts with the cellular adaptor protein complex 3 specifically via its medium subunit (AP-3Mu3A). This novel protein-protein interaction was first detected via yeast-two hybrid screen and was further confirmed in a mammalian system by immunofluorescence colocalization and co-immunoprecipitation. This novel interaction is further substantiated by the presence of a known tyrosine-based adaptor protein MU subunit sorting signal sequence, YXXФ: where Ф is a bulky hydrophobic residue, which is conserved across the related RSV M proteins. Analysis of point-mutated HRSV M derivatives indicated that AP-3Mu3A- mediated trafficking is contingent on the presence of the tyrosine residue within the YXXL sorting sequence at amino acids 197-200 of the M protein. AP-3Mu3A is up regulated at 24 hours post-infection in infected cells versus mock-infected HEp2 cells. Together, our data suggests that the AP-3 complex plays a critical role in the trafficking of HRSV proteins specifically matrix in epithelial cells. The results of this study add new insights and targets that may lead to the development of potential antivirals and attenuating mutations suitable for candidate vaccines in the future.


Asunto(s)
Complejo 3 de Proteína Adaptadora/metabolismo , Virus Sincitial Respiratorio Humano/metabolismo , Proteínas de la Matriz Viral/metabolismo , Complejo 3 de Proteína Adaptadora/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Secuencia Conservada , Células HeLa , Humanos , Unión Proteica , Estabilidad Proteica , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Transporte de Proteínas , Virus Sincitial Respiratorio Humano/fisiología , Regulación hacia Arriba , Proteínas de la Matriz Viral/química , Ensamble de Virus
11.
J Virol ; 91(17)2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28615208

RESUMEN

Pathogen recognition receptors are vital components of the immune system. Engagement of these receptors is important not only for instigation of innate immune responses to invading pathogens but also for initiating the adaptive immune response. Members of the NOD-like receptor (NLR) family of pathogen recognition receptors have important roles in orchestrating this response. The NLR family member NLRC5 regulates major histocompatibility complex class I (MHC-I) expression during various types of infections, but its role in immunity to influenza A virus (IAV) is not well studied. Here we show that Nlrc5-/- mice exhibit an altered CD8+ T cell response during IAV infection compared to that of wild-type (WT) mice. Nlrc5-/- mice have decreased MHC-I expression on hematopoietic cells and fewer CD8+ T cells prior to infection. NLRC5 deficiency does not affect the generation of antigen-specific CD8+ T cells following IAV infection; however, a change in epitope dominance is observed in Nlrc5-/- mice. Moreover, IAV-specific CD8+ T cells from Nlrc5-/- mice have impaired effector functions. This change in the adaptive immune response is associated with impaired viral clearance in Nlrc5-/- mice. Collectively, our results demonstrate an important role for NLRC5 in regulation of antiviral immune responses and viral clearance during IAV infection.IMPORTANCE The NOD-like receptor family member NLRC5 is known to regulate expression of MHC-I as well as other genes required for antigen processing. In addition, NLRC5 also regulates various immune signaling pathways. In this study, we investigated the role of NLRC5 during influenza virus infection and found a major role for NLRC5 in restricting virus replication and promoting viral clearance. The observed increases in viral titers in NLRC5-deficient mice correlated with impaired effector CD8+ T cell responses. Although NLRC5-deficient mice were defective at clearing the virus, they did not show an increase in morbidity or mortality following influenza virus infection because of other compensatory immune mechanisms. Therefore, our study highlights how NLRC5 regulates multiple immune effector mechanisms to promote the host defense during influenza virus infection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Células Asesinas Naturales/inmunología , Infecciones por Orthomyxoviridae/inmunología , Animales , Citocinas/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunidad Innata , Virus de la Influenza A , Péptidos y Proteínas de Señalización Intracelular/genética , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/virología , Transducción de Señal , Carga Viral
12.
FEBS J ; 284(15): 2363-2374, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28374568

RESUMEN

Inflammasomes are multiprotein complexes that form in the cytoplasm in response to cellular damage and cytosolic pathogen-associated molecules during infection. These complexes play important roles in initiating innate and adaptive immune responses to infectious disease. In addition, inflammasomes are now recognized as important mediators of sterile inflammation in various autoimmune and autoinflammatory diseases. Interestingly, microbiota and infection play critical roles in the development of 'sterile inflammation'. Herein, we highlight recent advances in our understanding of the role for inflammasomes in nucleic acid-, nucleosome-, and histone-driven sterile inflammation and discuss knowledge gaps and areas of potential future research.


Asunto(s)
Inmunidad Adaptativa , Daño del ADN , Inmunidad Innata , Inflamasomas/metabolismo , Modelos Inmunológicos , Nucleosomas/metabolismo , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/microbiología , ADN Bacteriano/metabolismo , ADN Mitocondrial/metabolismo , Histonas/metabolismo , Humanos , Infecciones/inmunología , Infecciones/metabolismo , Infecciones/microbiología , Inflamasomas/inmunología , Microbiota , Nucleosomas/inmunología , Nucleosomas/microbiología , ARN Bacteriano/metabolismo
13.
Trends Immunol ; 37(10): 703-714, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27592079

RESUMEN

Inflammasomes are cytosolic multiprotein platforms with pivotal roles in infectious diseases. Activation of inflammasomes results in proinflammatory cytokine signaling and pyroptosis. Sexually transmitted infections (STIs) are a major health problem worldwide, yet few studies have probed the impact of inflammasome signaling during these infections. Due to the dearth of appropriate infection models, our current understanding of inflammasomes in STIs is mostly drawn from results obtained in vitro, from distant infection sites, or from related microbial strains that are not sexually transmitted. Understanding how inflammasomes influence the outcome of STIs may lead to the development of novel and effective strategies to control disease and prevent transmission. Here we discuss and highlight the recent progress in this field.


Asunto(s)
Apoptosis , Inflamasomas/metabolismo , Enfermedades de Transmisión Sexual/inmunología , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Control de Infecciones , Mediadores de Inflamación/metabolismo , Enfermedades de Transmisión Sexual/transmisión , Transducción de Señal
14.
J Exp Med ; 213(10): 2081-97, 2016 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-27551156

RESUMEN

Lysosomal cathepsins regulate an exquisite range of biological functions, and their deregulation is associated with inflammatory, metabolic, and degenerative diseases in humans. In this study, we identified a key cell-intrinsic role for cathepsin B as a negative feedback regulator of lysosomal biogenesis and autophagy. Mice and macrophages lacking cathepsin B activity had increased resistance to the cytosolic bacterial pathogen Francisella novicida Genetic deletion or pharmacological inhibition of cathepsin B down-regulated mechanistic target of rapamycin activity and prevented cleavage of the lysosomal calcium channel TRPML1. These events drove transcription of lysosomal and autophagy genes via transcription factor EB, which increased lysosomal biogenesis and activation of autophagy initiation kinase ULK1 for clearance of the bacteria. Our results identified a fundamental biological function of cathepsin B in providing a checkpoint for homeostatic maintenance of lysosome populations and basic recycling functions in the cell.


Asunto(s)
Catepsina B/metabolismo , Francisella/inmunología , Infecciones por Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Gramnegativas/microbiología , Interacciones Huésped-Patógeno , Lisosomas/metabolismo , Biogénesis de Organelos , Animales , Autofagia , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Células de la Médula Ósea/patología , Regulación hacia Abajo , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Infecciones por Bacterias Gramnegativas/enzimología , Espacio Intracelular/microbiología , Activación del Canal Iónico , Macrófagos/metabolismo , Macrófagos/ultraestructura , Ratones Endogámicos C57BL , Modelos Biológicos , FN-kappa B/metabolismo , Transducción de Señal , Canales de Potencial de Receptor Transitorio/metabolismo , Regulación hacia Arriba
15.
Curr Opin Virol ; 12: 38-46, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25771504

RESUMEN

The inflammasome is a caspase-1 containing complex that activates the proinflammatory cytokines IL-1ß and IL-18 and results in the proinflammatory cell death known as pyroptosis. Numerous recent publications have highlighted the importance of inflammasome activation in the control of virus infection. Inflammasome activation during viral infection is dependent on a variety of upstream receptors including the NOD-like receptor, RIG-I-like receptor and AIM2-like receptor families. Various receptors also function in inflammasome activation in different cellular compartments, including the cytoplasm and the nucleus. The effectiveness of inflammasomes at suppressing virus replication is highlighted by the prevalence and diversity of virus encoded inflammasome inhibitors. Also, the host has a myriad of regulatory mechanisms in place to prevent unwanted inflammasome activation and overt inflammation. Finally, recent reports begin to suggest that inflammasome activation and inflammasome modulation may have important clinical applications. Herein, we highlight recent advances and discuss potential future directions toward understanding the role of inflammasomes during virus infection.


Asunto(s)
Inflamasomas/inmunología , Inflamasomas/metabolismo , Virosis/inmunología , Animales , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/inmunología , Humanos , Inflamasomas/genética , Receptores Inmunológicos , Virosis/metabolismo , Virosis/virología , Replicación Viral
16.
Nat Med ; 21(3): 263-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25686106

RESUMEN

The ketone bodies ß-hydroxybutyrate (BHB) and acetoacetate (AcAc) support mammalian survival during states of energy deficit by serving as alternative sources of ATP. BHB levels are elevated by starvation, caloric restriction, high-intensity exercise, or the low-carbohydrate ketogenic diet. Prolonged fasting reduces inflammation; however, the impact that ketones and other alternative metabolic fuels produced during energy deficits have on the innate immune response is unknown. We report that BHB, but neither AcAc nor the structurally related short-chain fatty acids butyrate and acetate, suppresses activation of the NLRP3 inflammasome in response to urate crystals, ATP and lipotoxic fatty acids. BHB did not inhibit caspase-1 activation in response to pathogens that activate the NLR family, CARD domain containing 4 (NLRC4) or absent in melanoma 2 (AIM2) inflammasome and did not affect non-canonical caspase-11, inflammasome activation. Mechanistically, BHB inhibits the NLRP3 inflammasome by preventing K(+) efflux and reducing ASC oligomerization and speck formation. The inhibitory effects of BHB on NLRP3 are not dependent on chirality or starvation-regulated mechanisms like AMP-activated protein kinase (AMPK), reactive oxygen species (ROS), autophagy or glycolytic inhibition. BHB blocks the NLRP3 inflammasome without undergoing oxidation in the TCA cycle, and independently of uncoupling protein-2 (UCP2), sirtuin-2 (SIRT2), the G protein-coupled receptor GPR109A or hydrocaboxylic acid receptor 2 (HCAR2). BHB reduces NLRP3 inflammasome-mediated interleukin (IL)-1ß and IL-18 production in human monocytes. In vivo, BHB or a ketogenic diet attenuates caspase-1 activation and IL-1ß secretion in mouse models of NLRP3-mediated diseases such as Muckle-Wells syndrome, familial cold autoinflammatory syndrome and urate crystal-induced peritonitis. Our findings suggest that the anti-inflammatory effects of caloric restriction or ketogenic diets may be linked to BHB-mediated inhibition of the NLRP3 inflammasome.


Asunto(s)
Ácido 3-Hidroxibutírico/farmacología , Proteínas Portadoras/antagonistas & inhibidores , Caspasa 1/efectos de los fármacos , Síndromes Periódicos Asociados a Criopirina , Inflamasomas/antagonistas & inhibidores , Interleucina-1beta/efectos de los fármacos , Monocitos/efectos de los fármacos , Adulto , Anciano , Animales , Dieta Cetogénica , Modelos Animales de Enfermedad , Femenino , Humanos , Inflamación , Interleucina-18 , Interleucina-1beta/metabolismo , Masculino , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR , Potasio/metabolismo
17.
PLoS Pathog ; 10(9): e1004410, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25254654

RESUMEN

Enteropathogenic and enterohemorrhagic bacterial infections in humans are a severe cause of morbidity and mortality. Although NOD-like receptors (NLRs) NOD2 and NLRP3 have important roles in the generation of protective immune responses to enteric pathogens, whether there is crosstalk among NLRs to regulate immune signaling is not known. Here, we show that mice and macrophages deficient in NOD2, or the downstream adaptor RIP2, have enhanced NLRP3- and caspases-11-dependent non-canonical inflammasome activation in a mouse model of enteropathogenic Citrobacter rodentium infection. Mechanistically, NOD2 and RIP2 regulate reactive oxygen species (ROS) production. Increased ROS in Rip2-deficient macrophages subsequently enhances c-Jun N-terminal kinase (JNK) signaling resulting in increased caspase-11 expression and activation, and more non-canonical NLRP3-dependant inflammasome activation. Intriguingly, this leads to protection of the colon epithelium for up to 10 days in Rip2-deficient mice infected with C. rodentium. Our findings designate NOD2 and RIP2 as key regulators of cellular ROS homeostasis and demonstrate for the first time that ROS regulates caspase-11 expression and non-canonical NLRP3 inflammasome activation through the JNK pathway.


Asunto(s)
Proteínas Portadoras/metabolismo , Caspasas/metabolismo , Infecciones por Enterobacteriaceae/inmunología , Inflamasomas/inmunología , Proteína Adaptadora de Señalización NOD2/fisiología , Especies Reactivas de Oxígeno/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/fisiología , Animales , Western Blotting , Caspasas Iniciadoras , Citrobacter rodentium/patogenicidad , Colitis/inmunología , Colitis/metabolismo , Colitis/microbiología , Colitis/patología , Infecciones por Enterobacteriaceae/metabolismo , Infecciones por Enterobacteriaceae/microbiología , Infecciones por Enterobacteriaceae/patología , Femenino , Inflamasomas/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor , Transducción de Señal
18.
J Virol ; 88(16): 8946-55, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24872587

RESUMEN

UNLABELLED: Nucleotide oligomerization and binding domain (NOD)-like receptors (NLRs) are important in the innate immune response to viral infection. Recent findings have implicated NLRP3, NOD2, and NLRX1 as important players in the innate antiviral response, but their roles in the generation of adaptive immunity to viruses are less clear. We demonstrate here that NOD2 is critical for both innate and adaptive immune responses necessary for controlling viral replication and survival during influenza A virus (IAV) infection. Nod2(-/-) mice have reduced beta interferon (IFN-ß) levels and fewer activated dendritic cells (DCs), and the DCs are more prone to cell death in the lungs of Nod2(-/-) mice during IAV infection. In agreement with the role for DCs in priming adaptive immunity, the generation of virus-specific CD8(+) T cells and their activation and production of IFN-γ were lower in Nod2(-/-) mice. Furthermore, Nod2(-/-) DCs, when cocultured with T cells in vitro, have a lower costimulatory capacity. Thus, Nod2(-/-) DCs are unable to efficiently prime CD8(+) T cells. These findings demonstrate that Nod2 is critical for the generation of both innate and adaptive immune responses necessary for controlling IAV infection. IMPORTANCE: The innate immune system is the host's first line of defense against invading pathogens and is also necessary for alerting and activating T and B cells to initiate the adaptive immune response. We demonstrate here that the innate immune receptor NOD2 is required for the production of antiviral type I interferons and the activation and survival of dendritic cells that, in turn, alert T cells to the presence of influenza A virus infection. In mice that are missing NOD2, interferon levels are lower, and the CD8(+) T cell response is impaired. As a result, the animals cannot control virus replication in their lungs as efficiently. This discovery helps us understand how the body naturally responds to virus infection and may help in the development of vaccines that use NOD2 to stimulate the CD8(+) T cell response, thus providing better protection against influenza A virus infection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Inmunidad Innata/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Proteína Adaptadora de Señalización NOD2/inmunología , Infecciones por Orthomyxoviridae/inmunología , Inmunidad Adaptativa/inmunología , Animales , Interferón beta/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/virología , Replicación Viral/inmunología
19.
Front Immunol ; 4: 285, 2013 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-24062750

RESUMEN

NOD-like receptors (NLRs) are a class of cytoplasmic pattern-recognition receptors. Although most NLRs play some role in immunity, their functions range from regulating antigen presentation (NLRC5, CIITA) to pathogen/damage sensing (NLRP1, NLRP3, NLRC1/2, NLRC4) to suppression or modulation of inflammation (NLRC3, NLRP6, NLRP12, NLRX1). However, NLRP2, NLRP5, and NLRP7 are also involved in non-immune pathways such as embryonic development. In this review, we highlight some of the least well-understood aspects of NLRs, including the mechanisms by which they sense pathogens or damage. NLRP3 recognizes a diverse range of stimuli and numerous publications have presented potential unifying models for NLRP3 activation, but no single mechanism proposed thus far appears to account for all possible NLRP3 activators. Additionally, NLRC3, NLRP6, and NLRP12 inhibit NF-κB activation, but whether direct ligand sensing is a requirement for this function is not known. Herein, we review the various mechanisms of sensing and activation proposed for NLRP3 and other inflammasome activators. We also discuss the role of NLRC3, NLRP6, NLRP12, and NLRX1 as inhibitors and how they are activated and function in their roles to limit inflammation. Finally, we present an overview of the emerging roles that NLRP2, NLRP5, and NLRP7 play during embryonic development and postulate on the potential pathways involved.

20.
Immunol Rev ; 255(1): 13-24, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23947344

RESUMEN

Nucleotide oligomerization and binding domain (NOD)-like receptors (NLRs) are a major constituent of the cytosolic innate immune-sensing machinery and participate in a wide array of pathways including nuclear factor κB (NF-κB), mitogen-activated protein kinase (MAPK), inflammasome, and type I interferon (IFN) signaling. NLRs have known roles in autoimmune, autoinflammatory, and infectious diseases. With respect to virus infection, NLRP3 is the most extensively studied NLR, including mechanisms of activation and inhibition. Furthermore, the importance of NLRP3 in both innate and adaptive immunity has been demonstrated. In comparison to NLRP3, the roles of other NLRs during virus infection are only just emerging. NLRC2 is an important activator of innate antiviral signaling and was recently found to mitigate inflammation during virus infection through autophagy. Finally, functions for NLRX1 in immune modulation and reactive oxygen species production require further examination and the importance of NLRC5 as a transactivator of major histocompatibility complex (MHC) class I and antigen presentation is currently developing. In this review, we discuss current knowledge pertaining to viruses and NLRs as well as areas of potential research, which will help advance the study of NLR biology during virus infection.


Asunto(s)
Receptores de Reconocimiento de Patrones/metabolismo , Virosis/inmunología , Virosis/metabolismo , Virus/inmunología , Animales , Presentación de Antígeno/inmunología , Humanos , Inflamasomas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Proteínas Adaptadoras de Señalización NOD/metabolismo , Receptores de Reconocimiento de Patrones/química , Virosis/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...