Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 837
Filtrar
1.
Int J Mol Sci ; 23(5)2022 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-35269795

RESUMEN

COVID-19, resulting from the SARS-CoV-2 virus, is a major pandemic that the world is fighting. SARS-CoV-2 primarily causes lung infection by attaching to the ACE2 receptor on the alveolar epithelial cells. However, the ACE2 receptor is also present in intestinal epithelial cells, suggesting a link between nutrition, virulence and clinical outcomes of COVID-19. Respiratory viral infections perturb the gut microbiota. The gut microbiota is shaped by our diet; therefore, a healthy gut is important for optimal metabolism, immunology and protection of the host. Malnutrition causes diverse changes in the immune system by repressing immune responses and enhancing viral vulnerability. Thus, improving gut health with a high-quality, nutrient-filled diet will improve immunity against infections and diseases. This review emphasizes the significance of dietary choices and its subsequent effects on the immune system, which may potentially impact SARS-CoV-2 vulnerability.


Asunto(s)
COVID-19/inmunología , Conducta Alimentaria , Sistema Inmunológico/inmunología , Desnutrición/inmunología , SARS-CoV-2/inmunología , COVID-19/epidemiología , COVID-19/virología , Microbioma Gastrointestinal/inmunología , Estado de Salud , Humanos , Modelos Inmunológicos , Estado Nutricional , Pandemias , SARS-CoV-2/patogenicidad , Virulencia/inmunología
2.
Cell Rep ; 38(1): 110183, 2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34986357

RESUMEN

Candida albicans is a commensal of the urogenital tract and the predominant cause of vulvovaginal candidiasis (VVC). Factors that increase circulatory estrogen levels such as pregnancy, the use of oral contraceptives, and hormone replacement therapy predispose women to VVC, but the reasons for this are largely unknown. Here, we investigate how adaptation of C. albicans to estrogen impacts the fungal host-pathogen interaction. Estrogen promotes fungal virulence by enabling C. albicans to avoid the actions of the innate immune system. Estrogen-induced innate immune evasion is mediated via inhibition of opsonophagocytosis through enhanced acquisition of the human complement regulatory protein, Factor H, on the fungal cell surface. Estrogen-induced accumulation of Factor H is dependent on the fungal cell surface protein Gpd2. The discovery of this hormone-sensing pathway might pave the way in explaining gender biases associated with fungal infections and may provide an alternative approach to improving women's health.


Asunto(s)
Candida albicans/inmunología , Candidiasis Vulvovaginal/patología , Vía Alternativa del Complemento/inmunología , Estrógenos/metabolismo , Evasión Inmune/inmunología , Fagocitosis/inmunología , Candida albicans/patogenicidad , Factor H de Complemento/metabolismo , Femenino , Glicerol-3-Fosfato Deshidrogenasa (NAD+)/metabolismo , Humanos , Inmunidad Innata/inmunología , Progesterona/metabolismo , Virulencia/inmunología
3.
Int J Mol Sci ; 23(2)2022 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-35055041

RESUMEN

Preterm infants are at increased risk for invasive neonatal bacterial infections. S. epidermidis, a ubiquitous skin commensal, is a major cause of late-onset neonatal sepsis, particularly in high-resource settings. The vulnerability of preterm infants to serious bacterial infections is commonly attributed to their distinct and developing immune system. While developmentally immature immune defences play a large role in facilitating bacterial invasion, this fails to explain why only a subset of infants develop infections with low-virulence organisms when exposed to similar risk factors in the neonatal ICU. Experimental research has explored potential virulence mechanisms contributing to the pathogenic shift of commensal S. epidermidis strains. Furthermore, comparative genomics studies have yielded insights into the emergence and spread of nosocomial S. epidermidis strains, and their genetic and functional characteristics implicated in invasive disease in neonates. These studies have highlighted the multifactorial nature of S. epidermidis traits relating to pathogenicity and commensalism. In this review, we discuss the known host and pathogen drivers of S. epidermidis virulence in neonatal sepsis and provide future perspectives to close the gap in our understanding of S. epidermidis as a cause of neonatal morbidity and mortality.


Asunto(s)
Interacciones Huésped-Patógeno , Sepsis Neonatal/microbiología , Infecciones Estafilocócicas/microbiología , Staphylococcus epidermidis/fisiología , Factores de Edad , Toxinas Bacterianas/genética , Biopelículas , Susceptibilidad a Enfermedades/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos , Tolerancia Inmunológica , Inmunidad Innata , Recién Nacido , Sepsis Neonatal/diagnóstico , Sepsis Neonatal/prevención & control , Sepsis Neonatal/terapia , Factores de Riesgo , Infecciones Estafilocócicas/diagnóstico , Infecciones Estafilocócicas/prevención & control , Infecciones Estafilocócicas/terapia , Virulencia/genética , Virulencia/inmunología , Factores de Virulencia/genética , Factores de Virulencia/inmunología
4.
PLoS Pathog ; 18(1): e1010270, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35089988

RESUMEN

ASFV is a large DNA virus that is highly pathogenic in domestic pigs. How this virus is sensed by the innate immune system as well as why it is so virulent remains enigmatic. In this study, we show that the ASFV genome contains AT-rich regions that are recognized by the DNA-directed RNA polymerase III (Pol-III), leading to viral RNA sensor RIG-I-mediated innate immune responses. We further show that ASFV protein I267L inhibits RNA Pol-III-RIG-I-mediated innate antiviral responses. I267L interacts with the E3 ubiquitin ligase Riplet, disrupts Riplet-RIG-I interaction and impairs Riplet-mediated K63-polyubiquitination and activation of RIG-I. I267L-deficient ASFV induces higher levels of interferon-ß, and displays compromised replication both in primary macrophages and pigs compared with wild-type ASFV. Furthermore, I267L-deficiency attenuates the virulence and pathogenesis of ASFV in pigs. These findings suggest that ASFV I267L is an important virulence factor by impairing innate immune responses mediated by the RNA Pol-III-RIG-I axis.


Asunto(s)
Virus de la Fiebre Porcina Africana/patogenicidad , Inmunidad Innata/inmunología , Factores de Virulencia/inmunología , Virulencia/inmunología , Fiebre Porcina Africana/inmunología , Virus de la Fiebre Porcina Africana/inmunología , Animales , ARN Polimerasa III/inmunología , Receptores de Superficie Celular/inmunología , Porcinos
5.
Front Immunol ; 12: 761981, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34858414

RESUMEN

In inflammatory bowel disease (IBD), intestinal mucosa cell and intestinal epithelial cell are severely damaged, and then their susceptibility to bacteria increases, so many commensal bacteria become pathogenic. The pathogenic commensal bacteria can stimulate a series of compensatory immune responses in the intestine. However, the immune response prevents the intestinal tract from restoring homeostasis, which in turn produces an indispensable inflammatory response. On the contrary, in IBD, the fierce inflammatory response contributes to the development of IBD. However, the effect of commensal bacteria on inflammation in IBD has not been clearly studied. Therefore, we further summarize the changes brought about by the changes of commensal bacteria to the inflammation of the intestines and their mutual influence. This article reviews the protective mechanism of commensal bacteria in healthy people and the mechanism of commensal bacteria and immune response to the destruction of the intestinal barrier when IBD occurs. The treatment and prevention of IBD are also briefly summarized.


Asunto(s)
Bacterias/inmunología , Inmunidad Innata/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Mucosa Intestinal/inmunología , Intestinos/inmunología , Bacterias/patogenicidad , Citocinas/inmunología , Citocinas/metabolismo , Homeostasis/inmunología , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/microbiología , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/microbiología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Intestinos/metabolismo , Intestinos/microbiología , Modelos Inmunológicos , Simbiosis/inmunología , Virulencia/inmunología
6.
Plant Commun ; 2(6): 100236, 2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34778749

RESUMEN

Plant nucleotide-binding leucine-rich repeat (NLR) receptors mediate immune responses by directly or indirectly sensing pathogen-derived effectors. Despite significant advances in the understanding of NLR-mediated immunity, the mechanisms by which pathogens evolve to suppress NLR activation triggered by cognate effectors and gain virulence remain largely unknown. The agronomically important immune receptor RB recognizes the ubiquitous and highly conserved IPI-O RXLR family members (e.g., IPI-O1) from Phytophthora infestans, and this process is suppressed by the rarely present and homologous effector IPI-O4. Here, we report that self-association of RB via the coiled-coil (CC) domain is required for RB activation and is differentially affected by avirulence and virulence effectors. IPI-O1 moderately reduces the self-association of RB CC, potentially leading to changes in the conformation and equilibrium of RB, whereas IPI-O4 dramatically impairs CC self-association to prevent RB activation. We also found that IPI-O1 associates with itself, whereas IPI-O4 does not. Notably, IPI-O4 interacts with IPI-O1 and disrupts its self-association, therefore probably blocking its avirulence function. Furthermore, IPI-O4 enhances the interaction between RB CC and IPI-O1, possibly sequestering RB and IPI-O1 and subsequently blocking their interactions with signaling components. Taken together, these findings considerably extend our understanding of the underlying mechanisms by which emerging virulent pathogens suppress the NLR-mediated recognition of cognate effectors.


Asunto(s)
Resistencia a la Enfermedad/inmunología , Interacciones Huésped-Patógeno/inmunología , Proteínas NLR/genética , Nicotiana/genética , Nicotiana/inmunología , Phytophthora infestans/patogenicidad , Enfermedades de las Plantas/inmunología , Virulencia/inmunología , Resistencia a la Enfermedad/genética , Interacciones Huésped-Patógeno/genética , Proteínas NLR/metabolismo , Enfermedades de las Plantas/genética , Plantas Modificadas Genéticamente , Nicotiana/microbiología , Virulencia/genética
7.
Front Immunol ; 12: 745315, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34671358

RESUMEN

Peste des petits ruminants (PPR) is an acute transboundary infectious viral disease of small ruminants, mainly sheep and goats. Host susceptibility varies considerably depending on the PPR virus (PPRV) strain, the host species and breed. The effect of strains with different levels of virulence on the modulation of the immune system has not been thoroughly compared in an experimental setting so far. In this study, we used a multi-omics approach to investigate the host cellular factors involved in different infection phenotypes. Peripheral blood mononuclear cells (PBMCs) from Saanen goats were activated with a T-cell mitogen and infected with PPRV strains of different virulence: Morocco 2008 (high virulence), Ivory Coast 1989 (low virulence) and Nigeria 75/1 (live attenuated vaccine strain). Our results showed that the highly virulent strain replicated better than the other two in PBMCs and rapidly induced cell death and a stronger inhibition of lymphocyte proliferation. However, all the strains affected lymphocyte proliferation and induced upregulation of key antiviral genes and proteins, meaning a classical antiviral response is orchestrated regardless of the virulence of the PPRV strain. On the other hand, the highly virulent strain induced stronger inflammatory responses and activated more genes related to lymphocyte migration and recruitment, and inflammatory processes. Both transcriptomic and proteomic approaches were successful in detecting viral and antiviral effectors under all conditions. The present work identified key immunological factors related to PPRV virulence in vitro.


Asunto(s)
Cabras/inmunología , Leucocitos Mononucleares/inmunología , Peste de los Pequeños Rumiantes/inmunología , Virus de la Peste de los Pequeños Rumiantes/patogenicidad , Virulencia/inmunología , Animales , Perfilación de la Expresión Génica , Cabras/virología , Peste de los Pequeños Rumiantes/virología , Virus de la Peste de los Pequeños Rumiantes/inmunología , Proteómica
8.
Proc Natl Acad Sci U S A ; 118(43)2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34686605

RESUMEN

Self-amplifying RNA replicons are promising platforms for vaccine generation. Their defects in one or more essential functions for viral replication, particle assembly, or dissemination make them highly safe as vaccines. We previously showed that the deletion of the envelope (E) gene from the Middle East respiratory syndrome coronavirus (MERS-CoV) produces a replication-competent propagation-defective RNA replicon (MERS-CoV-ΔE). Evaluation of this replicon in mice expressing human dipeptidyl peptidase 4, the virus receptor, showed that the single deletion of the E gene generated an attenuated mutant. The combined deletion of the E gene with accessory open reading frames (ORFs) 3, 4a, 4b, and 5 resulted in a highly attenuated propagation-defective RNA replicon (MERS-CoV-Δ[3,4a,4b,5,E]). This RNA replicon induced sterilizing immunity in mice after challenge with a lethal dose of a virulent MERS-CoV, as no histopathological damage or infectious virus was detected in the lungs of challenged mice. The four mutants lacking the E gene were genetically stable, did not recombine with the E gene provided in trans during their passage in cell culture, and showed a propagation-defective phenotype in vivo. In addition, immunization with MERS-CoV-Δ[3,4a,4b,5,E] induced significant levels of neutralizing antibodies, indicating that MERS-CoV RNA replicons are highly safe and promising vaccine candidates.


Asunto(s)
Infecciones por Coronavirus/prevención & control , Coronavirus del Síndrome Respiratorio de Oriente Medio/genética , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunología , ARN Viral/administración & dosificación , Replicón , Vacunas Virales/administración & dosificación , Animales , Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Antivirales/biosíntesis , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Virus Defectuosos/genética , Virus Defectuosos/inmunología , Femenino , Eliminación de Gen , Genes env , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Coronavirus del Síndrome Respiratorio de Oriente Medio/patogenicidad , ARN Viral/genética , ARN Viral/inmunología , Vacunas de ADN , Vacunas de Partículas Similares a Virus/administración & dosificación , Vacunas de Partículas Similares a Virus/genética , Vacunas de Partículas Similares a Virus/inmunología , Vacunas Virales/genética , Vacunas Virales/inmunología , Virulencia/genética , Virulencia/inmunología
9.
Sci Rep ; 11(1): 18982, 2021 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-34556711

RESUMEN

In the years 2006-2011, 617 Proteus spp. strains isolated mostly from urine and wounds or other clinical sources were collected in Lódz, Poland, to determine the offensive O serotypes frequently occurring among patients. P. mirabilis exhibited the most intensive swarming growth and was dominating species (86.9%), followed by P. genomospecies, P. vulgaris, and P. penneri. Ninety four per cent strains were recognized as S (smooth) forms. Serological studies (involving ELISA-enzyme-linked immunosorbent assay and Western blotting using native and adsorbed rabbit antisera) enabled classification of 80% S isolates into respective Proteus O serogroups among the 83 ones, described so far. The remaining strains seemed to be serologically unique. Despite the observed big serological variety of Proteus spp. isolates, we found the O78 serogroup recently described in Poland as dominating and identified other widespread serotypes: O3, O6, O10, O11, O27, O28, and O30 reported earlier as predominating also in other countries; O77 and O79 detected lately in Poland; O16, O18, O20, and O50. No unique structural feature of the prevalent O serotypes has been indicated. However, the prevalence of some O serogroups indicates that particular serotypes may be in some ways beneficial to the strains producing these kinds of O antigen.


Asunto(s)
Antígenos O/inmunología , Infecciones por Proteus/microbiología , Proteus/inmunología , Humanos , Polonia , Proteus/aislamiento & purificación , Proteus/patogenicidad , Infecciones por Proteus/sangre , Infecciones por Proteus/diagnóstico , Infecciones por Proteus/inmunología , Serogrupo , Serotipificación/métodos , Virulencia/inmunología
10.
Front Immunol ; 12: 702359, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34276695

RESUMEN

It was previously shown that secretion of PE-PGRS and PPE-MPTR proteins is abolished in clinical M. tuberculosis isolates with a deletion in the ppe38-71 operon, which is associated with increased virulence. Here we investigate the proteins dependent on PPE38 for their secretion and their role in the innate immune response using temporal proteomics and protein turnover analysis in a macrophage infection model. A decreased pro-inflammatory response was observed in macrophages infected with PPE38-deficient M. tuberculosis CDC1551 as compared to wild type bacteria. We could show that dampening of the pro-inflammatory response is associated with activation of a RelB/p50 pathway, while the canonical inflammatory pathway is active during infection with wild type M. tuberculosis CDC1551. These results indicate a molecular mechanism by which M. tuberculosis PE/PPE proteins controlled by PPE38 have an effect on modulating macrophage responses through NF-kB signalling.


Asunto(s)
Antígenos Bacterianos/inmunología , Macrófagos/inmunología , FN-kappa B/inmunología , Tuberculosis/inmunología , Factores de Virulencia/inmunología , Humanos , Inflamación/inmunología , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/patogenicidad , Transducción de Señal/inmunología , Células THP-1 , Virulencia/inmunología
11.
Biomolecules ; 11(6)2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34203937

RESUMEN

Bacterial secretory systems are essential for virulence in human pathogens. The systems have become a target of alternative antibacterial strategies based on small molecules and antibodies. Strategies to use components of the systems to design prophylactics have been less publicized despite vaccines being the preferred solution to dealing with bacterial infections. In the current review, strategies to design vaccines against selected pathogens are presented and connected to the biology of the system. The examples are given for Y. pestis, S. enterica, B. anthracis, S. flexneri, and other human pathogens, and discussed in terms of effectiveness and long-term protection.


Asunto(s)
Bacterias , Infecciones Bacterianas , Proteínas Bacterianas/inmunología , Vacunas Bacterianas , Bacterias/inmunología , Bacterias/patogenicidad , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/prevención & control , Sistemas de Secreción Bacterianos , Vacunas Bacterianas/inmunología , Vacunas Bacterianas/uso terapéutico , Humanos , Virulencia/inmunología
12.
Infect Immun ; 89(10): e0027021, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34227837

RESUMEN

Footrot is a polymicrobial infectious disease in sheep causing severe lameness, leading to one of the industry's largest welfare problems. The complex etiology of footrot makes in situ or in vitro investigations difficult. Computational methods offer a solution to understanding the bacteria involved and how they may interact with the host, ultimately providing a way to identify targets for future hypothesis-driven investigative work. Here, we present the first combined global analysis of bacterial community transcripts together with the host immune response in healthy and diseased ovine feet during a natural polymicrobial infection state using metatranscriptomics. The intratissue and surface bacterial populations and the most abundant bacterial transcriptomes were analyzed, demonstrating that footrot-affected skin has reduced diversity and increased abundances of not only the causative bacterium Dichelobacter nodosus but also other species such as Mycoplasma fermentans and Porphyromonas asaccharolytica. Host transcriptomics reveals the suppression of biological processes related to skin barrier function, vascular functions, and immunosurveillance in unhealthy interdigital skin, supported by histological findings that type I collagen (associated with scar tissue formation) is significantly increased in footrot-affected interdigital skin compared to outwardly healthy skin. Finally, we provide some interesting indications of host and pathogen interactions associated with virulence genes and the host spliceosome, which could lead to the identification of future therapeutic targets.


Asunto(s)
Bacterias/inmunología , Panadizo Interdigital/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad/inmunología , Ovinos/inmunología , Animales , Colágeno Tipo I/inmunología , Panadizo Interdigital/microbiología , Ovinos/microbiología , Enfermedades de las Ovejas/inmunología , Enfermedades de las Ovejas/microbiología , Piel/inmunología , Piel/microbiología , Transcriptoma/inmunología , Virulencia/inmunología
13.
Front Immunol ; 12: 677730, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34305907

RESUMEN

Ichthyophthirius multifiliis is a major pathogen that causes a high mortality rate in trout farms. However, systemic responses to the pathogen and its interactions with multiple organs during the course of infection have not been well described. In this study, dual-organ transcriptomic responses in the liver and head kidney and hemato-serological indexes were profiled under I. multifiliis infection and recovery to investigate systemic immuno-physiological characteristics. Several strategies for massive transcriptomic interpretation, such as differentially expressed genes (DEGs), Poisson linear discriminant (PLDA), and weighted gene co-expression network analysis (WGCNA) models were used to investigate the featured genes/pathways while minimizing the disadvantages of individual methods. During the course of infection, 6,097 and 2,931 DEGs were identified in the head kidney and liver, respectively. Markers of protein processing in the endoplasmic reticulum, oxidative phosphorylation, and the proteasome were highly expressed. Likewise, simultaneous ferroptosis and cellular reconstruction was observed, which is strongly linked to multiple organ dysfunction. In contrast, pathways relevant to cellular replication were up-regulated in only the head kidney, while endocytosis- and phagosome-related pathways were notably expressed in the liver. Moreover, interestingly, most immune-relevant pathways (e.g., leukocyte trans-endothelial migration, Fc gamma R-mediated phagocytosis) were highly activated in the liver, but the same pathways in the head kidney were down-regulated. These conflicting results from different organs suggest that interpretation of co-expression among organs is crucial for profiling of systemic responses during infection. The dual-organ transcriptomics approaches presented in this study will greatly contribute to our understanding of multi-organ interactions under I. multifiliis infection from a broader perspective.


Asunto(s)
Infecciones por Cilióforos/genética , Enfermedades de los Peces/genética , Interacciones Huésped-Patógeno/genética , Hymenostomatida/patogenicidad , Aprendizaje Automático , Oncorhynchus mykiss/genética , Oncorhynchus mykiss/parasitología , Transcriptoma , Animales , Infecciones por Cilióforos/inmunología , Infecciones por Cilióforos/parasitología , Enfermedades de los Peces/inmunología , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Branquias/inmunología , Riñón Cefálico/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata/genética , Hígado/inmunología , Oncorhynchus mykiss/inmunología , RNA-Seq/métodos , Transducción de Señal/genética , Transducción de Señal/inmunología , Virulencia/genética , Virulencia/inmunología , Factores de Virulencia
14.
PLoS Pathog ; 17(7): e1009733, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34310655

RESUMEN

Inflammatory factors and type I interferons (IFNs) are key components of host antiviral innate immune responses, which can be released from the pathogen-infected macrophages. African swine fever virus (ASFV) has developed various strategies to evade host antiviral innate immune responses, including alteration of inflammatory responses and IFNs production. However, the molecular mechanism underlying inhibition of inflammatory responses and IFNs production by ASFV-encoded proteins has not been fully understood. Here we report that ASFV infection only induced low levels of IL-1ß and type I IFNs in porcine alveolar macrophages (PAMs), even in the presence of strong inducers such as LPS and poly(dA:dT). Through further exploration, we found that several members of the multigene family 360 (MGF360) and MGF505 strongly inhibited IL-1ß maturation and IFN-ß promoter activation. Among them, pMGF505-7R had the strongest inhibitory effect. To verify the function of pMGF505-7R in vivo, a recombinant ASFV with deletion of the MGF505-7R gene (ASFV-Δ7R) was constructed and assessed. As we expected, ASFV-Δ7R infection induced higher levels of IL-1ß and IFN-ß compared with its parental ASFV HLJ/18 strain. ASFV infection-induced IL-1ß production was then found to be dependent on TLRs/NF-κB signaling pathway and NLRP3 inflammasome. Furthermore, we demonstrated that pMGF505-7R interacted with IKKα in the IKK complex to inhibit NF-κB activation and bound to NLRP3 to inhibit inflammasome formation, leading to decreased IL-1ß production. Moreover, we found that pMGF505-7R interacted with and inhibited the nuclear translocation of IRF3 to block type I IFN production. Importantly, the virulence of ASFV-Δ7R is reduced in piglets compared with its parental ASFV HLJ/18 strain, which may due to induction of higher IL-1ß and type I IFN production in vivo. Our findings provide a new clue to understand the functions of ASFV-encoded pMGF505-7R and its role in viral infection-induced pathogenesis, which might help design antiviral agents or live attenuated vaccines to control ASF.


Asunto(s)
Virus de la Fiebre Porcina Africana/patogenicidad , Fiebre Porcina Africana/inmunología , Evasión Inmune/inmunología , Macrófagos Alveolares/inmunología , Proteínas Virales/inmunología , Virus de la Fiebre Porcina Africana/inmunología , Animales , Inmunidad Innata , Interferón Tipo I/biosíntesis , Interleucina-1beta/biosíntesis , Familia de Multigenes , Porcinos , Virulencia/inmunología
15.
Infect Immun ; 89(10): e0020121, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34097460

RESUMEN

Endophthalmitis is a devastating infection that can cause blindness. Over half of Bacillus endophthalmitis cases result in significant loss of useful vision. Bacillus produces many virulence factors that may contribute to retinal damage and robust inflammation. We analyzed Bacillus immune inhibitor A (InhA) metalloproteases in the context of this disease, hypothesizing that InhAs contribute to Bacillus intraocular virulence and inflammation. We analyzed phenotypes and infectivity of wild-type (WT), InhA1-deficient (ΔinhA1), InhA2-deficient (ΔinhA2), or InhA1, A2, and A3-deficient (ΔinhA1-3) Bacillus thuringiensis. In vitro analysis of growth, proteolysis, and cytotoxicity were compared. WT and InhA mutants were similarly cytotoxic to retinal cells. The ΔinhA1 and ΔinhA2 mutants entered log-phase growth earlier than WT B. thuringiensis. Proteolysis by the ΔinhA1-3 mutant was decreased, but this strain grew similar to WT in vitro. Experimental endophthalmitis was initiated by intravitreally infecting C57BL/6J mice with 200 CFU of WT B. thuringiensis or InhA mutants. Eyes were analyzed for intraocular Bacillus and myeloperoxidase concentrations, retinal function loss, and gross histological changes. Eyes infected with the ΔinhA1 or ΔinhA2 mutant strains contained greater numbers of bacteria than eyes infected with WT throughout the infection course. Eyes infected with single mutants had inflammation and retinal function loss similar to eyes infected with the WT strain. Eyes infected with the ΔinhA1-3 mutant cleared the infection. Quantitative real-time PCR (qRT-PCR) results suggested that there may be compensatory expression of the other InhAs in the single InhA mutant. These results indicate that together, the InhA metalloproteases contribute to the severity of infection and inflammation in Bacillus endophthalmitis.


Asunto(s)
Bacillus thuringiensis/inmunología , Endoftalmitis/inmunología , Metaloendopeptidasas/inmunología , Metaloproteasas/inmunología , Virulencia/inmunología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Endoftalmitis/microbiología , Infecciones Bacterianas del Ojo/inmunología , Infecciones Bacterianas del Ojo/microbiología , Humanos , Inflamación/inmunología , Inflamación/microbiología , Ratones , Ratones Endogámicos C57BL , Retina/inmunología , Retina/microbiología
16.
Front Biosci (Landmark Ed) ; 26(5): 51-75, 2021 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-34027650

RESUMEN

In 2020, a novel strain of coronavirus (COVID-19) has led to a significant morbidity and mortality worldwide. As of the date of this writing, a total of 116 M cases has been diagnosed worldwide leading to 2.5 M deaths. The number of mortalities is directly correlated with the rise of innate immune cells (especially macrophages) in the lungs that secrete inflammatory cytokines (IL-1ß and IL-6) leading to the development of "Cytokine Storm Syndrome" (CSS), multi-organ-failure and death. Given that currently the treatment of this condition is rare and release of effective vaccine might be months away, here, we review the plants and their pharmacologically active-compounds as potential phytopharmaceuticals for the virus induced inflammatory response. Experimental validation of the effectiveness of these natural compounds to prevent or reduce the cytokine storm might be beneficial as an adjunct treatment of SARS-CoV-2.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Síndrome de Liberación de Citoquinas/prevención & control , Fitoterapia/métodos , Extractos Vegetales/uso terapéutico , Plantas Medicinales/química , SARS-CoV-2/efectos de los fármacos , COVID-19/inmunología , COVID-19/virología , Síndrome de Liberación de Citoquinas/inmunología , Citocinas/inmunología , Citocinas/metabolismo , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Plantas Medicinales/clasificación , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad , Virulencia/efectos de los fármacos , Virulencia/inmunología
17.
Biochem Soc Trans ; 49(3): 1287-1297, 2021 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-34003245

RESUMEN

Interferon (IFN)-induced guanosine triphosphate hydrolysing enzymes (GTPases) have been identified as cornerstones of IFN-mediated cell-autonomous defence. Upon IFN stimulation, these GTPases are highly expressed in various host cells, where they orchestrate anti-microbial activities against a diverse range of pathogens such as bacteria, protozoan and viruses. IFN-induced GTPases have been shown to interact with various host pathways and proteins mediating pathogen control via inflammasome activation, destabilising pathogen compartments and membranes, orchestrating destruction via autophagy and the production of reactive oxygen species as well as inhibiting pathogen mobility. In this mini-review, we provide an update on how the IFN-induced GTPases target pathogens and mediate host defence, emphasising findings on protection against bacterial pathogens.


Asunto(s)
Bacterias/inmunología , Infecciones Bacterianas/inmunología , GTP Fosfohidrolasas/inmunología , Inmunidad Innata/inmunología , Interferones/inmunología , Animales , Bacterias/patogenicidad , Infecciones Bacterianas/metabolismo , Infecciones Bacterianas/microbiología , GTP Fosfohidrolasas/metabolismo , Interacciones Huésped-Patógeno/inmunología , Humanos , Interferones/metabolismo , Transducción de Señal/inmunología , Virulencia/inmunología
18.
Nat Commun ; 12(1): 2790, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33986286

RESUMEN

SARS-CoV-2 is of zoonotic origin and contains a PRRA polybasic cleavage motif which is considered critical for efficient infection and transmission in humans. We previously reported on a panel of attenuated SARS-CoV-2 variants with deletions at the S1/S2 junction of the spike protein. Here, we characterize pathogenicity, immunogenicity, and protective ability of a further cell-adapted SARS-CoV-2 variant, Ca-DelMut, in in vitro and in vivo systems. Ca-DelMut replicates more efficiently than wild type or parental virus in Vero E6 cells, but causes no apparent disease in hamsters, despite replicating in respiratory tissues. Unlike wild type virus, Ca-DelMut causes no obvious pathological changes and does not induce elevation of proinflammatory cytokines, but still triggers a strong neutralizing antibody and T cell response in hamsters and mice. Ca-DelMut immunized hamsters challenged with wild type SARS-CoV-2 are fully protected, with little sign of virus replication in the upper or lower respiratory tract, demonstrating sterilizing immunity.


Asunto(s)
COVID-19/diagnóstico , Mutación , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/genética , Replicación Viral/genética , Animales , COVID-19/inmunología , COVID-19/virología , Línea Celular Tumoral , Chlorocebus aethiops , Cricetinae , Citocinas/inmunología , Citocinas/metabolismo , Femenino , Interacciones Huésped-Patógeno , Humanos , Masculino , Mesocricetus , Ratones Endogámicos BALB C , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad , Linfocitos T/inmunología , Linfocitos T/metabolismo , Células Vero , Virulencia/genética , Virulencia/inmunología
20.
PLoS Pathog ; 17(4): e1009073, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33914852

RESUMEN

Bacteriophages (or phages) play major roles in the evolution of bacterial pathogens via horizontal gene transfer. Multiple phages are often integrated in a host chromosome as prophages, not only carrying various novel virulence-related genetic determinants into host bacteria but also providing various possibilities for prophage-prophage interactions in bacterial cells. In particular, Escherichia coli strains such as Shiga toxin (Stx)-producing E. coli (STEC) and enteropathogenic E. coli (EPEC) strains have acquired more than 10 prophages (up to 21 prophages), many of which encode type III secretion system (T3SS) effector gene clusters. In these strains, some prophages are present at a single locus in tandem, which is usually interpreted as the integration of phages that use the same attachment (att) sequence. Here, we present phages integrating into T3SS effector gene cluster-associated loci in prophages, which are widely distributed in STEC and EPEC. Some of the phages integrated into prophages are Stx-encoding phages (Stx phages) and have induced the duplication of Stx phages in a single cell. The identified attB sequences in prophage genomes are apparently derived from host chromosomes. In addition, two or three different attB sequences are present in some prophages, which results in the generation of prophage clusters in various complex configurations. These phages integrating into prophages represent a medically and biologically important type of inter-phage interaction that promotes the accumulation of T3SS effector genes in STEC and EPEC, the duplication of Stx phages in STEC, and the conversion of EPEC to STEC and that may be distributed in other types of E. coli strains as well as other prophage-rich bacterial species.


Asunto(s)
Infecciones por Escherichia coli/microbiología , Transferencia de Gen Horizontal/genética , Profagos/genética , Toxina Shiga II/farmacología , Toxina Shiga/genética , Bacteriófagos/genética , Escherichia coli/metabolismo , Transferencia de Gen Horizontal/inmunología , Profagos/patogenicidad , Toxina Shiga II/genética , Virulencia/inmunología , Factores de Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...