Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
Adv Exp Med Biol ; 1294: 151-166, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33079368

RESUMEN

Mouse models are invaluable resources for studying the pathogenesis and preclinical evaluation of therapeutics and vaccines against many human pathogens. Infections caused by group A streptococcus (GAS, Streptococcus pyogenes) are heterogeneous ranging from mild pharyngitis to severe invasive necrotizing fasciitis, a subgroup of necrotizing soft-tissue infections (NSTIs). While several strains of mice including BALB/c, C3H/HeN, CBA/J, and C57BL/10 offered significant insights, the human specificity and the interindividual variations on susceptibility or resistance to GAS infections limit their ability to mirror responses as seen in humans. In this chapter, we discuss the advanced recombinant inbred (ARI) BXD mouse model that mimics the genetic diversity as seen in humans and underpins the feasibility to map multiple genes (genetic loci) modulating GAS NSTI. GAS produces a myriad of virulence factors, including superantigens (SAg). Superantigens are potent immune toxins that activate T cells by cross-linking T cell receptors with human leukocyte antigen class-II (HLA-II) molecules expressed on antigen-presenting cells. This leads to a pro-inflammatory cytokine storm and the subsequent multiple organ damage and shock. Inbred mice are innately refractive to SAg-mediated responses. In this chapter, we discuss the versatility of the HLA-II transgenic mouse model that allowed the biological validation of known genetic associations to GAS NSTI. The combined utility of ARI-BXD and HLA-II mice as complementary approaches that offer clinically translatable insights into pathomechanisms driven by complex traits and host genetic context and novel means to evaluate the in vivo efficiency of therapies to improve outcomes of GAS NSTI are also discussed.


Asunto(s)
Modelos Animales de Enfermedad , Infecciones Estreptocócicas/genética , Infecciones Estreptocócicas/microbiología , Streptococcus/clasificación , Streptococcus/patogenicidad , Animales , Ratones , Ratones Endogámicos , Necrosis , Infecciones Estreptocócicas/patología
2.
Infect Immun ; 86(12)2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30224551

RESUMEN

Invasive group A streptococcus (GAS) infections include necrotizing soft tissue infections (NSTI) and streptococcal toxic shock syndrome (STSS). We have previously shown that host HLA class II allelic variations determine the risk for necrotizing fasciitis (NF), a dominant subgroup of NSTI, and STSS by modulating responses to GAS superantigens (SAgs). SAgs are pivotal mediators of uncontrolled T-cell activation, triggering a proinflammatory cytokine storm in the host. FoxP3-expressing CD4+ CD25+ T regulatory cells (Tregs) comprise phenotypically and functionally heterogeneous subsets with a profound ability to suppress inflammatory responses. Specifically, activated Tregs, which express glycoprotein A repetitions predominant (GARP) and display latent transforming growth factor ß1 (TGF-ß1) complexes (latency-associated peptide [LAP]), exhibit strong immunosuppressive functions. The significance of Tregs that may participate in suppressing inflammatory responses during NSTI is unknown. Here, we phenotypically characterized FoxP3/GARP/LAP-expressing Tregs in GAS-infected or SAg (SmeZ)-stimulated splenocytes from transgenic (tg) mice expressing human HLA-II DRB1*15 (DR15 allele associated with nonsevere NF/STSS-protective responses) or DRB1*0402/DQB1*0302 (DR4/DQ8 alleles associated with neutral risk for combined NF/STSS). We demonstrated both in vivo and in vitro that the neutral-risk allele upregulates expression of CD4+ CD25+ activated effector T cells, with a significantly lower frequency of Foxp3+/GARP+ LAP- but higher frequency of Foxp3- LAP+ Tregs than seen with the protective allele. Additional in vitro studies revealed that the presentation of SmeZ by the neutral-risk allele significantly increases proliferation and expression of effector cytokines gamma interferon (IFN-γ) and interleukin-2 (IL-2) and upregulates CD4+ CD25+ T cell receptors (TCRs) carrying specific Vß 11 chain (TCRVß11+) T cells and Th1 transcription factor Tbx21 mRNA levels. Our data suggest that neutral-risk alleles may drive Th1 differentiation while attenuating the induction of Tregs associated with suppressive function.


Asunto(s)
Factores de Transcripción Forkhead/genética , Proteínas de la Membrana/genética , Infecciones de los Tejidos Blandos/inmunología , Infecciones Estreptocócicas/inmunología , Linfocitos T Reguladores/inmunología , Factor 3 Asociado a Receptor de TNF/genética , Alelos , Animales , Citocinas/inmunología , Modelos Animales de Enfermedad , Cadenas HLA-DRB1/genética , Humanos , Activación de Linfocitos , Ratones , Ratones Transgénicos , Infecciones de los Tejidos Blandos/microbiología , Proteínas de Dominio T Box/genética , Células TH1/inmunología
3.
Sci Rep ; 6: 31360, 2016 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-27511873

RESUMEN

Methicillin-resistant Staphylococcus aureus (MRSA) is a major cause of skin and soft tissue infections. One of the highly successful and rapidly disseminating clones is MRSA ST22 commonly associated with skin tropism. Here we show that a naturally occurring single amino acid substitution (tyrosine to cysteine) at position 223 of AgrC determines starkly different ST22 S. aureus virulence phenotypes, e.g. cytotoxic or colonizing, as evident in both in vitro and in vivo skin infections. Y223C amino acid substitution destabilizes AgrC-AgrA interaction leading to a colonizing phenotype characterized by upregulation of bacterial surface proteins. The colonizing phenotype strains cause less severe skin tissue damage, show decreased susceptibility towards the antimicrobial LL-37 and induce autophagy. In contrast, cytotoxic strains with tyrosine at position 223 of AgrC cause infections characterized by inflammasome activation and severe skin tissue pathology. Taken together, the study demonstrates how a single amino acid substitution in the histidine kinase receptor AgrC of ST22 strains determines virulence properties and infection outcome.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/farmacología , Proteínas Bacterianas/genética , Staphylococcus aureus Resistente a Meticilina/fisiología , Mutación Puntual , Proteínas Quinasas/genética , Infecciones Cutáneas Estafilocócicas/microbiología , Sustitución de Aminoácidos , Animales , Proteínas Bacterianas/metabolismo , Línea Celular , Cisteína/genética , Modelos Animales de Enfermedad , Humanos , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/genética , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Ratones , Fenotipo , Unión Proteica , Proteínas Quinasas/metabolismo , Tirosina/genética , Tropismo Viral , Catelicidinas
4.
PLoS Pathog ; 12(7): e1005732, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27399650

RESUMEN

Host genetic variations play an important role in several pathogenic diseases, and we have previously provided strong evidences that these genetic variations contribute significantly to differences in susceptibility and clinical outcomes of invasive Group A Streptococcus (GAS) infections, including sepsis and necrotizing soft tissue infections (NSTIs). Our initial studies with conventional mouse strains revealed that host genetic variations and sex differences play an important role in orchestrating the severity, susceptibility and outcomes of NSTIs. To understand the complex genetic architecture of NSTIs, we utilized an unbiased, forward systems genetics approach in an advanced recombinant inbred (ARI) panel of mouse strains (BXD). Through this approach, we uncovered interactions between host genetics, and other non-genetic cofactors including sex, age and body weight in determining susceptibility to NSTIs. We mapped three NSTIs-associated phenotypic traits (i.e., survival, percent weight change, and lesion size) to underlying host genetic variations by using the WebQTL tool, and identified four NSTIs-associated quantitative genetic loci (QTL) for survival on mouse chromosome (Chr) 2, for weight change on Chr 7, and for lesion size on Chr 6 and 18 respectively. These QTL harbor several polymorphic genes. Identification of multiple QTL highlighted the complexity of the host-pathogen interactions involved in NSTI pathogenesis. We then analyzed and rank-ordered host candidate genes in these QTL by using the QTLminer tool and then developed a list of 375 candidate genes on the basis of annotation data and biological relevance to NSTIs. Further differential expression analyses revealed 125 genes to be significantly differentially regulated in susceptible strains compared to their uninfected controls. Several of these genes are involved in innate immunity, inflammatory response, cell growth, development and proliferation, and apoptosis. Additional network analyses using ingenuity pathway analysis (IPA) of these 125 genes revealed interleukin-1 beta network as key network involved in modulating the differential susceptibility to GAS NSTIs.


Asunto(s)
Fascitis Necrotizante/genética , Predisposición Genética a la Enfermedad/genética , Infecciones de los Tejidos Blandos/genética , Infecciones de los Tejidos Blandos/microbiología , Infecciones Estreptocócicas/genética , Streptococcus pyogenes , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Redes Reguladoras de Genes , Humanos , Masculino , Ratones , Fenotipo , Sitios de Carácter Cuantitativo , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Riesgo
5.
Infect Immun ; 84(2): 416-24, 2016 02.
Artículo en Inglés | MEDLINE | ID: mdl-26573737

RESUMEN

Host genetic variations play an important role in several pathogenic diseases, and we previously provided strong evidence that these genetic variations contribute significantly to differences in susceptibility and clinical outcomes of invasive group A Streptococcus (GAS) patients, including sepsis and necrotizing soft tissue infections (NSTIs). The goal of the present study was to investigate how genetic variations and sex differences among four commonly used mouse strains contribute to variation in severity, manifestations, and outcomes of NSTIs. DBA/2J mice were more susceptible to NSTIs than C57BL/6J, BALB/c, and CD-1 mice, as exhibited by significantly greater bacteremia, excessive dissemination to the spleen, and significantly higher mortality. Differences in the sex of the mice also contributed to differences in disease severity and outcomes: DBA/2J female mice were relatively resistant compared to their male counterparts. However, DBA/2J mice exhibited minimal weight loss and developed smaller lesions than did the aforementioned strains. Moreover, at 48 h after infection, compared with C57BL/6J mice, DBA/2J mice had increased bacteremia, excessive dissemination to the spleen, and excessive concentrations of inflammatory cytokines and chemokines. These results indicate that variations in the host genetic context as well as sex play a dominant role in determining the severity of and susceptibility to GAS NSTIs.


Asunto(s)
Susceptibilidad a Enfermedades , Variación Genética , Caracteres Sexuales , Infecciones de los Tejidos Blandos/genética , Infecciones Estreptocócicas/genética , Streptococcus pyogenes/patogenicidad , Animales , Carga Bacteriana , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Interacciones Huésped-Patógeno , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Necrosis , Infecciones de los Tejidos Blandos/microbiología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/crecimiento & desarrollo
6.
BMC Genomics ; 16: 947, 2015 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-26573818

RESUMEN

BACKGROUND: Shiga toxin (Stx)-producing E. coli (STEC) are responsible for foodborne outbreaks that can result in severe human disease. During an outbreak, differential disease outcomes are observed after infection with the same STEC strain. One question of particular interest is why some infected people resolve infection after hemorrhagic colitis whereas others progress to the hemolytic uremic syndrome (HUS). Host age and infection dose have been implicated; however, these parameters do not appear to fully account for all of the observed variation in disease severity. Therefore, we hypothesized that additional host genetic factors may play a role in progression to HUS. METHODS AND RESULTS: To mimic the genetic diversity in the human response to infection by STEC, we measured the capacity of an O157:H7 outbreak isolate to colonize mouse strains from the advanced recombinant inbred (ARI) BXD panel. We first infected the BXD parental strains C57BL/6 J (B6) and DBA/2 J (D2) with either 86-24 (Stx2a+) or TUV86-2, an Stx2a-negative isogenic mutant. Colonization levels were determined in an intact commensal flora (ICF) infection model. We found a significant difference in colonization levels between the parental B6 and D2 strains after infection with TUV86-2 but not with 86-24. This observation suggested that a host factor that may be masked by Stx2a affects O157:H7 colonization in some genetic backgrounds. We then determined the TUV86-2 colonization levels of 24 BXD strains in the ICF model. We identified several quantitative trait loci (QTL) associated with variation in colonization by correlation analyses. We found a highly significant QTL on proximal chromosome 9 (12.5-26.7 Mb) that strongly predicts variation in colonization levels and accounts for 15-20 % of variance. Linkage, polymorphism and co-citation analyses of the mapped region revealed 36 candidate genes within the QTL, and we identified five genes that are most likely responsible for the differential colonization. CONCLUSIONS: The identification of the QTL on chromosome 9 supports our hypothesis that individual genetic makeup affects the level of colonization after infection with STEC O157:H7.


Asunto(s)
Mapeo Cromosómico , ADN Recombinante/genética , Escherichia coli O157/fisiología , Interacciones Huésped-Patógeno , Sitios de Carácter Cuantitativo/genética , Animales , Escherichia coli O157/metabolismo , Femenino , Perfilación de la Expresión Génica , Ontología de Genes , Redes Reguladoras de Genes , Ligamiento Genético , Variación Genética , Síndrome Hemolítico-Urémico/genética , Síndrome Hemolítico-Urémico/microbiología , Humanos , Ratones , Ratones Endogámicos DBA , Toxina Shiga/metabolismo , Especificidad de la Especie , Factores de Tiempo
7.
PLoS Pathog ; 10(5): e1004155, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24875883

RESUMEN

Establishing the genetic determinants of niche adaptation by microbial pathogens to specific hosts is important for the management and control of infectious disease. Streptococcus pyogenes is a globally prominent human-specific bacterial pathogen that secretes superantigens (SAgs) as 'trademark' virulence factors. SAgs function to force the activation of T lymphocytes through direct binding to lateral surfaces of T cell receptors and class II major histocompatibility complex (MHC-II) molecules. S. pyogenes invariably encodes multiple SAgs, often within putative mobile genetic elements, and although SAgs are documented virulence factors for diseases such as scarlet fever and the streptococcal toxic shock syndrome (STSS), how these exotoxins contribute to the fitness and evolution of S. pyogenes is unknown. Here we show that acute infection in the nasopharynx is dependent upon both bacterial SAgs and host MHC-II molecules. S. pyogenes was rapidly cleared from the nasal cavity of wild-type C57BL/6 (B6) mice, whereas infection was enhanced up to ∼10,000-fold in B6 mice that express human MHC-II. This phenotype required the SpeA superantigen, and vaccination with an MHC -II binding mutant toxoid of SpeA dramatically inhibited infection. Our findings indicate that streptococcal SAgs are critical for the establishment of nasopharyngeal infection, thus providing an explanation as to why S. pyogenes produces these potent toxins. This work also highlights that SAg redundancy exists to avoid host anti-SAg humoral immune responses and to potentially overcome host MHC-II polymorphisms.


Asunto(s)
Proteínas Bacterianas/metabolismo , Exotoxinas/metabolismo , Antígenos de Histocompatibilidad Clase II/inmunología , Proteínas de la Membrana/metabolismo , Infecciones Estreptocócicas/inmunología , Streptococcus pyogenes/inmunología , Superantígenos/inmunología , Enfermedad Aguda , Animales , Proteínas Bacterianas/inmunología , Exotoxinas/inmunología , Humanos , Proteínas de la Membrana/inmunología , Ratones Endogámicos C57BL , Nasofaringe/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Infecciones Estreptocócicas/prevención & control , Streptococcus pyogenes/genética , Superantígenos/genética , Linfocitos T/inmunología
8.
Infect Immun ; 82(7): 2992-3001, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24799625

RESUMEN

Streptococcal cysteine protease (SpeB), the major secreted protease produced by group A streptococcus (GAS), cleaves both host and bacterial proteins and contributes importantly to the pathogenesis of invasive GAS infections. Modulation of SpeB expression and/or its activity during invasive GAS infections has been shown to affect bacterial virulence and infection severity. Expression of SpeB is regulated by the GAS CovR-CovS two-component regulatory system, and we demonstrated that bacteria with mutations in the CovR-CovS two-component regulatory system are selected for during localized GAS infections and that these bacteria lack SpeB expression and exhibit a hypervirulent phenotype. Additionally, in a separate study, we showed that expression of SpeB can also be modulated by human transferrin- and/or lactoferrin-mediated iron chelation. Accordingly, the goal of this study was to investigate the possible roles of iron and other metals in modulating SpeB expression and/or activity in a manner that would potentiate bacterial virulence. Here, we report that the divalent metals zinc and copper inhibit SpeB activity at the posttranslational level. Utilizing online metal-binding site prediction servers, we identified two putative metal-binding sites in SpeB, one of which involves the catalytic-dyad residues (47)Cys and (195)His. Based on our findings, we propose that zinc and/or copper availability in the bacterial microenvironment can modulate the proteolytic activity of SpeB in a manner that preserves the integrity of several other virulence factors essential for bacterial survival and dissemination within the host and thereby may exacerbate the severity of invasive GAS infections.


Asunto(s)
Cobre/farmacología , Proteasas de Cisteína/metabolismo , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Streptococcus pyogenes/enzimología , Zinc/farmacología , Proteasas de Cisteína/genética , Relación Dosis-Respuesta a Droga , Regulación Bacteriana de la Expresión Génica/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Metales/farmacología , Ácido Pentético/administración & dosificación , Ácido Pentético/farmacología , Proteómica , Streptococcus pyogenes/metabolismo , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
10.
PLoS One ; 9(3): e92342, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24651695

RESUMEN

Both viral and host genetics affect the outcome of herpes simplex virus type 1 (HSV-1) infection in humans and experimental models. Little is known about specific host gene variants and molecular networks that influence herpetic disease progression, severity, and episodic reactivation. To identify such host gene variants we have initiated a forward genetic analysis using the expanded family of BXD strains, all derived from crosses between C57BL/6J and DBA/2J strains of mice. One parent is highly resistant and one highly susceptible to HSV-1. Both strains have also been fully sequenced, greatly facilitating the search for genetic modifiers that contribute to differences in HSV-1 infection. We monitored diverse disease phenotypes following infection with HSV-1 strain 17syn+ including percent mortality (herpes simplex encephalitis, HSE), body weight loss, severity of herpetic stromal keratitis (HSK), spleen weight, serum neutralizing antibody titers, and viral titers in tear films in BXD strains. A significant quantitative trait locus (QTL) on chromosome (Chr) 16 was found to associate with both percent mortality and HSK severity. Importantly, this QTL maps close to a human QTL and the gene proposed to be associated with the frequency of recurrent herpetic labialis (cold sores). This suggests that a single host locus may influence these seemingly diverse HSV-1 pathogenic phenotypes by as yet unknown mechanisms. Additional suggestive QTLs for percent mortality were identified--one on Chr X that is epistatically associated with that on Chr 16. As would be anticipated the Chr 16 QTL also modulated weight loss, reaching significance in females. A second significant QTL for maximum weight loss in male and female mice was mapped to Chr 12. To our knowledge this is the first report of a host genetic locus that modulates the severity of both herpetic disease in the nervous system and herpetic stromal keratitis.


Asunto(s)
Sustancia Propia/patología , Sustancia Propia/virología , Genes , Interacciones Huésped-Patógeno/genética , Queratitis Herpética/genética , Queratitis Herpética/virología , Simplexvirus/patogenicidad , Animales , Emparejamiento Base/genética , Cromosomas de los Mamíferos/genética , Femenino , Humanos , Masculino , Ratones , Fenotipo , Sitios de Carácter Cuantitativo , Virulencia/genética
11.
Methods Mol Biol ; 1028: 47-74, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23740113

RESUMEN

Nanotechnology involves the creation and manipulation of materials at nanoscale levels (1-100 nm) to create products that exhibit novel properties. While this motivation has driven nanoscience and technology in physics and engineering, it is not the main reason that nanoparticles are useful for systemic applications in the human body. The application of nanotechnology to medicine, known as nanomedicine, concerns the use of precisely engineered materials at this length scale to develop novel therapeutic and diagnostic modalities. A number of nanotherapeutic formulations are already approved for medical use and more are in the approval pipeline currently. This chapter is intended to provide an overview of the toxicity of these therapeutic nanoparticles and to summarize the current state of the field. We begin with background on the sources of exposure to nanoparticles, followed by reviewing different forms of nanosized therapeutic tools as quantum dots, nanoshells, nanocapsules, echogenic bubble, and "nanoshuttles." Moreover, cytotoxic effects of nanoparticles on cell membrane, mitochondrial function, prooxidant/antioxidant status, enzyme leakage, DNA, and other biochemical endpoints were elucidated. We highlight the need for caution during the use and disposal of such manufactured nanomaterials to prevent unintended environmental impacts. Moreover, different strategies which could be used to minimize or eliminate nanotoxicity were also discussed in detail. Understanding of how to tune size and surface properties to provide safety will permit the creation of new, more effective nanomedicines for systemic use.


Asunto(s)
Portadores de Fármacos/toxicidad , Nanopartículas del Metal/toxicidad , Animales , Membrana Celular/efectos de los fármacos , Membrana Celular/fisiología , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/uso terapéutico , Composición de Medicamentos , Humanos , Nanopartículas del Metal/uso terapéutico , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Nanomedicina , Oxidación-Reducción , Tamaño de la Partícula , Propiedades de Superficie
12.
Toxicol Sci ; 134(1): 49-63, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23640863

RESUMEN

Noninvasive vaginal infections by Staphylococcus aureus strains producing the superantigen TSST-1 can cause menstrual toxic shock syndrome (mTSS). With the objective of exploring the basis for differential susceptibility to mTSS, the relative responsiveness to TSST-1 of healthy women has been investigated. Peripheral blood mononuclear cells from healthy donors were incubated with purified TSST-1 or with the T-cell mitogen phytohemmaglutinin (PHA), and proliferation was measured. The concentrations of TSST-1 and PHA required to elicit a response equivalent to 15% of the maximal achievable response (EC15) were determined. Although with PHA, EC15 values were comparable between donors, subjects could be classified as being of high, medium, or low sensitivity based on responsiveness to TSST-1. Sensitivity to TSST-1-induced proliferation was associated with increased production of the cytokines interleukin-2 and interferon-γ. When the entire T lymphocyte population was considered, there were no differences between sensitivity groups with respect to the frequency of cells known to be responsive to TSST-1 (those bearing CD3(+) Vß2(+)). However, there was an association between sensitivity to TSST-1 and certain HLA-class II haplotypes. Thus, the frequencies of DR7DQ2, DR14DQ5, DR4DQ8, and DR8DQ4 haplotypes were greater among those with high sensitivity, a finding confirmed by analysis of responses to immortalized homozygous B cell lines. Collectively, the results reveal that factors other than neutralizing antibody and the frequency of Vß2(+) T lymphocytes determine immunological responsiveness to TSST-1. Differential responsiveness of lymphocytes to TSST-1 may form the basis of interindividual variations in susceptibility to mTSS.


Asunto(s)
Toxinas Bacterianas/inmunología , Toxinas Bacterianas/toxicidad , Enterotoxinas/inmunología , Enterotoxinas/toxicidad , Choque Séptico/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Superantígenos/inmunología , Superantígenos/toxicidad , Adolescente , Adulto , Anticuerpos Antibacterianos/sangre , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Estudios de Cohortes , Umbral Diferencial , Relación Dosis-Respuesta Inmunológica , Femenino , Antígenos HLA-D/genética , Antígenos HLA-D/inmunología , Haplotipos , Humanos , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Interleucina-2/biosíntesis , Interleucina-2/inmunología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Menstruación , Mitógenos/inmunología , Mitógenos/farmacología , Fitohemaglutininas/inmunología , Fitohemaglutininas/farmacología , Choque Séptico/microbiología , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/aislamiento & purificación , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Adulto Joven
13.
RNA ; 19(6): 767-77, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23604636

RESUMEN

Due to structural flexibility, RNase sensitivity, and serum instability, RNA nanoparticles with concrete shapes for in vivo application remain challenging to construct. Here we report the construction of 14 RNA nanoparticles with solid shapes for targeting cancers specifically. These RNA nanoparticles were resistant to RNase degradation, stable in serum for >36 h, and stable in vivo after systemic injection. By applying RNA nanotechnology and exemplifying with these 14 RNA nanoparticles, we have established the technology and developed "toolkits" utilizing a variety of principles to construct RNA architectures with diverse shapes and angles. The structure elements of phi29 motor pRNA were utilized for fabrication of dimers, twins, trimers, triplets, tetramers, quadruplets, pentamers, hexamers, heptamers, and other higher-order oligomers, as well as branched diverse architectures via hand-in-hand, foot-to-foot, and arm-on-arm interactions. These novel RNA nanostructures harbor resourceful functionalities for numerous applications in nanotechnology and medicine. It was found that all incorporated functional modules, such as siRNA, ribozymes, aptamers, and other functionalities, folded correctly and functioned independently within the nanoparticles. The incorporation of all functionalities was achieved prior, but not subsequent, to the assembly of the RNA nanoparticles, thus ensuring the production of homogeneous therapeutic nanoparticles. More importantly, upon systemic injection, these RNA nanoparticles targeted cancer exclusively in vivo without accumulation in normal organs and tissues. These findings open a new territory for cancer targeting and treatment. The versatility and diversity in structure and function derived from one biological RNA molecule implies immense potential concealed within the RNA nanotechnology field.


Asunto(s)
Sistemas de Liberación de Medicamentos , Nanopartículas/administración & dosificación , Nanotecnología/métodos , Neoplasias Experimentales/terapia , ARN/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Secuencia de Bases , Línea Celular Tumoral , Silenciador del Gen , Terapia Genética/métodos , Secuencias Invertidas Repetidas , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Nanopartículas/química , Electroforesis en Gel de Poliacrilamida Nativa , Neoplasias Experimentales/genética , Motivos de Nucleótidos , ARN/química , Pliegue del ARN , Estabilidad del ARN , Relación Estructura-Actividad
14.
FASEB J ; 27(7): 2633-43, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23531597

RESUMEN

In Western countries, invasive infections caused by M1T1 serotype group A Streptococcus (GAS) are epidemiologically linked to mutations in the control of virulence regulatory 2-component operon (covRS). In indigenous communities and developing countries, severe GAS disease is associated with genetically diverse non-M1T1 GAS serotypes. Hypervirulent M1T1 covRS mutant strains arise through selection by human polymorphonuclear cells for increased expression of GAS virulence factors such as the DNase Sda1, which promotes neutrophil resistance. The GAS bacteremia isolate NS88.2 (emm 98.1) is a covS mutant that exhibits a hypervirulent phenotype and neutrophil resistance yet lacks the phage-encoded Sda1. Here, we have employed a comprehensive systems biology (genomic, transcriptomic, and proteomic) approach to identify NS88.2 virulence determinants that enhance neutrophil resistance in the non-M1T1 GAS genetic background. Using this approach, we have identified streptococcal collagen-like protein A and general stress protein 24 proteins as NS88.2 determinants that contribute to survival in whole blood and neutrophil resistance in non-M1T1 GAS. This study has revealed new factors that contribute to GAS pathogenicity that may play important roles in resisting innate immune defenses and the development of human invasive infections.


Asunto(s)
Proteínas Bacterianas/inmunología , Infecciones Estreptocócicas/inmunología , Streptococcus pyogenes/inmunología , Animales , Adhesión Bacteriana/genética , Adhesión Bacteriana/inmunología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Línea Celular , Electroforesis en Gel Bidimensional , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica , Genoma Bacteriano/genética , Genómica/métodos , Interacciones Huésped-Patógeno/inmunología , Humanos , Ratones , Viabilidad Microbiana/genética , Viabilidad Microbiana/inmunología , Mutación , Activación Neutrófila/inmunología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteoma/genética , Proteoma/metabolismo , Proteómica/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Infecciones Estreptocócicas/sangre , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidad , Virulencia/genética , Virulencia/inmunología
15.
Nat Rev Cardiol ; 10(3): 171-7, 2013 03.
Artículo en Inglés | MEDLINE | ID: mdl-23319102

RESUMEN

Rheumatic fever is one of the most-neglected ailments, and its pathogenesis remains poorly understood. The major thrust of research has been directed towards cross-reactivity between streptococcal M protein and myocardial α-helical coiled-coil proteins. M protein has also been the focus of vaccine development. The characteristic pathological findings suggest that the primary site of rheumatic-fever-related damage is subendothelial and perivascular connective tissue matrix and overlying endothelium. Over the past 5 years, a streptococcal M protein N-terminus domain has been shown to bind to the CB3 region in collagen type IV. This binding seems to initiate an antibody response to the collagen and result in ground substance inflammation. These antibodies do not cross-react with M proteins, and we believe that no failure of immune system and, possibly, no molecular mimicry occur in rheumatic fever. This alternative hypothesis shares similarity with collagen involvement in both Goodpasture syndrome and Alport syndrome.


Asunto(s)
Autoinmunidad , Miocarditis/microbiología , Fiebre Reumática/microbiología , Streptococcus pyogenes/patogenicidad , Animales , Antígenos Bacterianos/inmunología , Autoanticuerpos/biosíntesis , Proteínas de la Membrana Bacteriana Externa/inmunología , Proteínas Portadoras/inmunología , Niño , Preescolar , Colágeno Tipo IV/inmunología , Endotelio Vascular/inmunología , Endotelio Vascular/microbiología , Endotelio Vascular/patología , Femenino , Humanos , Miocarditis/inmunología , Miocarditis/patología , Miocardio/inmunología , Miocardio/patología , Unión Proteica , Fiebre Reumática/inmunología , Fiebre Reumática/patología , Streptococcus pyogenes/inmunología
16.
Viruses ; 4(12): 3468-93, 2012 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-23207275

RESUMEN

Countering aerosolized filovirus infection is a major priority of biodefense research. Aerosol models of filovirus infection have been developed in knock-out mice, guinea pigs and non-human primates; however, filovirus infection of immunocompetent mice by the aerosol route has not been reported. A murine model of aerosolized filovirus infection in mice should be useful for screening vaccine candidates and therapies. In this study, various strains of wild-type and immunocompromised mice were exposed to aerosolized wild-type (WT) or mouse-adapted (MA) Ebola virus (EBOV). Upon exposure to aerosolized WT-EBOV, BALB/c, C57BL/6 (B6), and DBA/2 (D2) mice were unaffected, but 100% of severe combined immunodeficiency (SCID) and 90% of signal transducers and activators of transcription (Stat1) knock-out (KO) mice became moribund between 7-9 days post-exposure (dpe). Exposure to MA-EBOV caused 15% body weight loss in BALB/c, but all mice recovered. In contrast, 10-30% lethality was observed in B6 and D2 mice exposed to aerosolized MA-EBOV, and 100% of SCID, Stat1KO, interferon (IFN)-γ KO and Perforin KO mice became moribund between 7-14 dpe. In order to identify wild-type, inbred, mouse strains in which exposure to aerosolized MA-EBOV is uniformly lethal, 60 BXD (C57BL/6 crossed with DBA2) recombinant inbred (RI) and advanced RI (ARI) mouse strains were exposed to aerosolized MA-EBOV, and monitored for disease severity. A complete spectrum of disease severity was observed. All BXD strains lost weight but many recovered. However, infection was uniformly lethal within 7 to 12 days post-exposure in five BXD strains. Aerosol exposure of these five BXD strains to 10-fold less MA-EBOV resulted in lethality ranging from 0% in two strains to 90-100% lethality in two strains. Analysis of post-mortem tissue from BXD strains that became moribund and were euthanized at the lower dose of MA-EBOV, showed liver damage in all mice as well as lung lesions in two of the three strains. The two BXD strains that exhibited 90-100% mortality, even at a low dose of airborne MA-EBOV will be useful mouse models for testing vaccines and therapies. Additionally, since disease susceptibility is affected by complex genetic traits, a systems genetics approach was used to identify preliminary gene loci modulating disease severity among the panel BXD strains. Preliminary quantitative trait loci (QTLs) were identified that are likely to harbor genes involved in modulating differential susceptibility to Ebola infection.


Asunto(s)
Aerosoles , Transmisión de Enfermedad Infecciosa , Ebolavirus/patogenicidad , Fiebre Hemorrágica Ebola/transmisión , Animales , Modelos Animales de Enfermedad , Fiebre Hemorrágica Ebola/mortalidad , Fiebre Hemorrágica Ebola/patología , Huésped Inmunocomprometido , Hígado/patología , Pulmón/patología , Ratones , Ratones Noqueados , Ratones SCID , Análisis de Supervivencia
17.
FASEB J ; 26(11): 4675-84, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22878963

RESUMEN

The past 50 years has witnessed the emergence of new viral and bacterial pathogens with global effect on human health. The hyperinvasive group A Streptococcus (GAS) M1T1 clone, first detected in the mid-1980s in the United States, has since disseminated worldwide and remains a major cause of severe invasive human infections. Although much is understood regarding the capacity of this pathogen to cause disease, much less is known of the precise evolutionary events selecting for its emergence. We used high-throughput technologies to sequence a World Health Organization strain collection of serotype M1 GAS and reconstructed its phylogeny based on the analysis of core genome single-nucleotide polymorphisms. We demonstrate that acquisition of a 36-kb genome segment from serotype M12 GAS and the bacteriophage-encoded DNase Sda1 led to increased virulence of the M1T1 precursor and occurred relatively early in the molecular evolutionary history of this strain. The more recent acquisition of the phage-encoded superantigen SpeA is likely to have provided selection advantage for the global dissemination of the M1T1 clone. This study provides an exemplar for the evolution and emergence of virulent clones from microbial populations existing commensally or causing only superficial infection.


Asunto(s)
Evolución Biológica , Pandemias , Infecciones Estreptocócicas/epidemiología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Células Epiteliales/microbiología , Exotoxinas/genética , Exotoxinas/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Genoma Bacteriano , Salud Global , Interacciones Huésped-Patógeno , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Neutrófilos/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Fagocitosis , Filogenia , Streptococcus pyogenes/clasificación , Streptococcus pyogenes/genética , Streptococcus pyogenes/patogenicidad , Transcriptoma , Virulencia
18.
PLoS One ; 7(7): e41157, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22815951

RESUMEN

Staphylococcus aureus produces superantigens (SAgs) that bind and cross-link T cells and APCs, leading to activation and proliferation of immune cells. SAgs bind to variable regions of the ß-chains of T cell receptors (Vß-TCRs), and each SAg binds a unique subset of Vß-TCRs. This binding leads to massive cytokine production and can result in toxic shock syndrome (TSS). The most abundantly produced staphylococcal SAgs and the most common causes of staphylococcal TSS are TSS toxin-1 (TSST-1), and staphylococcal enterotoxins B and C (SEB and SEC, respectively). There are several characterized variants of humans SECs, designated SEC1-4, but only one variant of SEB has been described. Sequencing the seb genes from over 20 S. aureus isolates show there are at least five different alleles of seb, encoding forms of SEB with predicted amino acid substitutions outside of the predicted immune-cell binding regions of the SAgs. Examination of purified, variant SEBs indicates that these amino acid substitutions cause differences in proliferation of rabbit splenocytes in vitro. Additionally, the SEBs varied in lethality in a rabbit model of TSS. The SEBs were diverse in their abilities to cause proliferation of human peripheral blood mononuclear cells, and differed in their activation of subsets of T cells. A soluble, high-affinity Vß-TCR, designed to neutralize the previously characterized variant of SEB (SEB1), was able to neutralize the variant SEBs, indicating that this high-affinity peptide may be useful in treating a variety of SEB-mediated illnesses.


Asunto(s)
Enterotoxinas/metabolismo , Staphylococcus aureus/metabolismo , Secuencia de Aminoácidos , Animales , Proliferación Celular , Clonación Molecular , Genes Codificadores de la Cadena beta de los Receptores de Linfocito T/genética , Variación Genética , Humanos , Leucocitos Mononucleares/metabolismo , Linfocitos/citología , Modelos Moleculares , Conformación Molecular , Datos de Secuencia Molecular , Péptidos/química , Estructura Terciaria de Proteína , Conejos , Homología de Secuencia de Aminoácido , Superantígenos/metabolismo
19.
PLoS One ; 7(4): e35218, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22511985

RESUMEN

Streptococcus pyogenes is an important human pathogen, which has recently gained recognition as an intracellular microorganism during the course of severe invasive infections such as necrotizing fasciitis. Although the surface anchored M protein has been identified as a pivotal factor affecting phagosomal maturation and S. pyogenes survival within macrophages, the overall transcriptional profile required for the pathogen to adapt and persist intracellularly is as of yet unknown. To address this, the gene expression profile of S. pyogenes within human macrophages was determined and compared to that of extracellular bacteria using customized microarrays and real-time qRT-PCR. In order to model the early phase of infection involving adaptation to the intracellular compartment, samples were collected 2h post-infection. Microarray analysis revealed that the expression of 145 streptococcal genes was significantly altered in the intracellular environment. The majority of differentially regulated genes were associated with metabolic and energy-dependent processes. Key up-regulated genes in early phase intracellular bacteria were ihk and irr, encoding a two-component gene regulatory system (TCS). Comparison of gene expression of selected genes at 2h and 6h post-infection revealed a dramatic shift in response regulators over time with a down-regulation of ihk/irr genes concurring with an up-regulation of the covR/S TCS. In re-infection assays, intracellular bacteria from the 6h time point exhibited significantly greater survival within macrophages than did bacteria collected at the 2h time point. An isogenic S. pyogenes mutant deficient in ihk/irr displayed significantly reduced bacterial counts when compared to wild-type bacteria following infection of macrophages. The findings illustrate how gene expression of S. pyogenes during the intracellular life cycle is fine-tuned by temporal expression of specific two-component systems.


Asunto(s)
Macrófagos/microbiología , Streptococcus pyogenes/genética , Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Humanos , Streptococcus pyogenes/patogenicidad , Transcriptoma , Virulencia
20.
J Neuroinflammation ; 9: 74, 2012 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-22531301

RESUMEN

BACKGROUND: Recent clinical observations suggest that certain gut and dietary factors may transiently worsen symptoms in autism. Propionic acid (PA) is a short chain fatty acid and an important intermediate of cellular metabolism. Although PA has several beneficial biological effects, its accumulation is neurotoxic. METHODS: Two groups of young Western albino male rats weighing about 45 to 60 grams (approximately 21 days old) were used in the present study. The first group consisted of oral buffered PA-treated rats that were given a neurotoxic dose of 250 mg/kg body weight/day for three days, n = eight; the second group of rats were given only phosphate buffered saline and used as a control. Biochemical parameters representing oxidative stress, energy metabolism, neuroinflammation, neurotransmission, and apoptosis were investigated in brain homogenates of both groups. RESULTS: Biochemical analyses of brain homogenates from PA-treated rats showed an increase in oxidative stress markers (for example, lipid peroxidation), coupled with a decrease in glutathione (GSH) and glutathione peroxidase (GPX) and catalase activities. Impaired energy metabolism was ascertained through the decrease of lactate dehydrogenase and activation of creatine kinase (CK). Elevated IL-6, TNFα, IFNγ and heat shock protein 70 (HSP70) confirmed the neuroinflammatory effect of PA. Moreover, elevation of caspase3 and DNA fragmentation proved the pro-apoptotic and neurotoxic effect of PA to rat pups CONCLUSION: By comparing the results obtained with those from animal models of autism or with clinical data on the biochemical profile of autistic patients, this study showed that the neurotoxicity of PA as an environmental factor could play a central role in the etiology of autistic biochemical features.


Asunto(s)
Trastorno Autístico/inducido químicamente , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Propionatos/toxicidad , Animales , Trastorno Autístico/fisiopatología , Encéfalo/efectos de los fármacos , Masculino , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Propionatos/metabolismo , Distribución Aleatoria , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...