Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Methods Mol Biol ; 2542: 323-360, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36008676

RESUMEN

The cell wall contains mannans and glucans that are recognized by the host immune system. In this chapter, we will describe the methods to isolate mannans and glucans from the C. albicans cell wall. In addition, we describe how to determine purity, molecular size, and structure of the mannans and glucans. We also detail how to prepare the carbohydrates for in vitro, ex vivo, or in vivo use by describing endotoxin removal (depyrogenation), derivatization, and labeling and evaluation of bioactivity.


Asunto(s)
Glucanos , Mananos , Candida albicans , Pared Celular/química , Glucanos/análisis
2.
Front Cell Infect Microbiol ; 12: 939944, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36636722

RESUMEN

Genital Chlamydia is the most common bacterial sexually transmitted infection in the United States and worldwide. Previous studies indicate that the progression of chlamydial infection is influenced by various factors, including the female sex hormones estrogen and progesterone. Sex hormone levels naturally fluctuate in women throughout their menstrual cycle. Varying concentrations of estrogen and progesterone may impact the progression of chlamydial infection and the host's immune response to Chlamydia. Estrogen signals through estrogen receptors (ERs), ERα and ERß. These receptors are similar in structure and function, but are differentially expressed in tissues throughout the body, including the genital tract and on cells of the immune system. In this study, we used ovariectomized (OVT) BALB/c mice to investigate the impact of long-term administration of physiologically relevant concentrations of estrogen (E2), progesterone (P4), or a combination of E2/P4 on the progression of and immune response to C. muridarum infection. Additionally, we used ERα and ERß knockout C57/BL6 mice to determine the how ERs affect chlamydial infection and the resulting immune response. Estrogen exposure prevented C. muridarum infection in vaginally infected OVT mice exposed to E2 alone or in combination with P4, while OVT or Sham mice exposed to hormone free, P4 or depo-medroxyprogesterone acetate shed similar amounts of chlamydiae. The hormonal environment also altered T cell recruitment and IFNϵ production the genital tracts of infected OVT and Sham mice on day 10 post infection. The absence of ERα, but not ERß, in ER knockout mouse strains significantly changed the timing of C. muridarum infection. ERαKO mice shed significantly more chlamydiae at day 3 post infection and resolved the infection faster than WT or ERßKO animals. At day 9 post infection, flow cytometry showed that ERαKO mice had more T cells present and targeted RNA sequencing revealed increased expression of CD4 and FOXP3, suggesting that ERαKO mice had increased numbers of regulatory T cells compared to ERßKO and WT mice. Mock and chlamydia-infected ERαKO mice also expressed more IFNϵ early during infection. Overall, the data from these studies indicate that sex hormones and their receptors, particularly ERα and ERß, differentially affect C. muridarum infection in murine models of infection.


Asunto(s)
Infecciones por Chlamydia , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Animales , Femenino , Ratones , Infecciones por Chlamydia/microbiología , Chlamydia muridarum , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/genética , Estrógenos , Ratones Noqueados , Progesterona
3.
Pathog Dis ; 76(8)2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30321322

RESUMEN

Chlamydia trachomatis/HSV-2 vaginal co-infections are seen clinically, suggesting that these sexually transmitted pathogens may interact. We previously established an intravaginal Chlamydia muridarum/HSV-2 super-infection model and observed that chlamydial pre-infection protects mice from a subsequent lethal HSV-2 challenge. However, the mechanism of protection remains unknown. The type I interferon, IFN-ß, binds to the type I interferon receptor (IFNR), elicits a host cellular antiviral response and inhibits HSV replication in vitro and in vivo. Previous studies have demonstrated that C. muridarum infection stimulates genital tract (GT) IFN-ß production; therefore, we hypothesized that chlamydial pre-infection protects mice from HSV-2 challenge via the IFN-ß/IFNR-induced antiviral response. To test this prediction, we quantified IFN-ß levels in vaginal swab samples. Detection of IFN-ß in C. muridarum singly infected, but not in mock-infected animals, prompted the use of the super-infection model in IFNR knockout (IFNR-/-) mice. We observed that C. muridarum pre-infection reduces HSV-2-induced mortality by 40% in wild-type mice and by 60% IFNR-/- mice. Severity of HSV-2 disease symptoms and viral shedding was also similarly reduced by C. muridarum pre-infection. These data indicate that, while chlamydial infection induces GT production of IFN-ß, type I IFN-induced antiviral responses are likely not required for the observed protective effect.


Asunto(s)
Infecciones por Chlamydia/complicaciones , Herpes Genital/prevención & control , Receptor de Interferón alfa y beta/metabolismo , Sobreinfección/prevención & control , Animales , Chlamydia muridarum/inmunología , Modelos Animales de Enfermedad , Femenino , Herpesvirus Humano 2/inmunología , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Vagina/inmunología
4.
Front Microbiol ; 9: 3270, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30692972

RESUMEN

Microbial interactions represent an understudied facet of human health and disease. In this study, the interactions that occur between Chlamydia trachomatis and the opportunistic fungal pathogen, Candida albicans were investigated. Candida albicans is a common component of the oral and vaginal microbiota responsible for thrush and vaginal yeast infections. Normally, Candida exist in the body as yeast. However, disruptions to the microbiota create conditions that allow expanded growth of Candida, conversion to the hyphal form, and tissue invasion. Previous studies have shown that a myriad of outcomes can occur when Candida albicans interacts with pathogenic bacteria. To determine if C. trachomatis physically interacts with C. albicans, we incubated chlamydial elementary bodies (EB) in medium alone or with C. albicans yeast or hyphal forms for 1 h. Following incubation, the samples were formaldehyde-fixed and processed for immunofluorescence assays using anti-chlamydial MOMP or anti- chlamydial LPS antibodies. Replicate samples were replenished with culture medium and incubated at 35°C for 0-120 h prior to fixation for immunofluorescence analysis or collection for EB infectivity assays. Data from this study indicates that both C. trachomatis serovar E and C. muridarum EB bind to C. albicans yeast and hyphal forms. This interaction was not blocked by pre-incubation of EB with the Candida cell wall components, mannan or ß-glucans, suggesting that EB interact with a Candida cell wall protein or other structure. Bound EB remained attached to C. albicans for a minimum of 5 days (120 h). Infectivity assays demonstrated that EB bound to C. albicans are infectious immediately following binding (0h). However, once bound to C. albicans, EB infectivity decreased at a faster rate than EB in medium alone. At 6h post binding, 40% of EB incubated in medium alone remained infectious compared to only 16% of EB bound to C. albicans. Likewise, pre-incubation of EB with laminarin, a soluble preparation of ß-glucan, alone or in combination with other fungal cell wall components significantly decreases chlamydial infectivity in HeLa cells. These data indicate that interactions between EB and C. albicans inhibit chlamydial infectivity, possibly by physically blocking EB interactions with host cell receptors.

5.
Artículo en Inglés | MEDLINE | ID: mdl-29322031

RESUMEN

Chlamydia trachomatis infections represent the predominant cause of bacterial sexually transmitted infections. As an obligate intracellular bacterium, C. trachomatis is dependent on the host cell for survival, propagation, and transmission. Thus, factors that affect the host cell, including nutrition, cell cycle, and environmental signals, have the potential to impact chlamydial development. Previous studies have demonstrated that activation of Wnt/ß-catenin signaling benefits C. trachomatis infections in fallopian tube epithelia. In cervical epithelial cells chlamydiae sequester ß-catenin within the inclusion. These data indicate that chlamydiae interact with the Wnt signaling pathway in both the upper and lower female genital tract (FGT). However, hormonal activation of canonical and non-canonical Wnt signaling pathways is an essential component of cyclic remodeling in another prominent area of the FGT, the endometrium. Given this information, we hypothesized that Wnt signaling would impact chlamydial infection in endometrial epithelial cells. To investigate this hypothesis, we analyzed the effect of Wnt inhibition on chlamydial inclusion development and elementary body (EB) production in two endometrial cell lines, Ishikawa (IK) and Hec-1B, in nonpolarized cell culture and in a polarized endometrial epithelial (IK)/stromal (SHT-290) cell co-culture model. Inhibition of Wnt by the small molecule inhibitor (IWP2) significantly decreased inclusion size in IK and IK/SHT-290 cultures (p < 0.005) and chlamydial infectivity (p ≤ 0.01) in both IK and Hec-1B cells. Confocal and electron microscopy analysis of chlamydial inclusions revealed that Wnt inhibition caused chlamydiae to become aberrant in morphology. EB formation was also impaired in IK, Hec-1B and IK/SHT-290 cultures regardless of whether Wnt inhibition occurred throughout, in the middle (24 hpi) or late (36 hpi) during the development cycle. Overall, these data lead us to conclude that Wnt signaling in the endometrium is a key host pathway for the proper development of C. trachomatis.


Asunto(s)
Infecciones por Chlamydia/microbiología , Chlamydia trachomatis/fisiología , Endometrio/microbiología , Células Epiteliales/microbiología , Interacciones Huésped-Patógeno , Vía de Señalización Wnt , Línea Celular , Técnicas de Cocultivo , Femenino , Humanos , Cuerpos de Inclusión/microbiología , Microscopía Confocal , Microscopía Electrónica , Modelos Biológicos
6.
PLoS One ; 11(8): e0160511, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27486990

RESUMEN

Chlamydia trachomatis is the most common bacterial sexually transmitted pathogen, but more than 70% of patients fail to seek treatment due to the asymptomatic nature of these infections. Women suffer from numerous complications from chronic chlamydial infections, which include pelvic inflammatory disease and infertility. We previously demonstrated in culture that host cell nectin-1 knockdown significantly reduced chlamydial titers and inclusion size. Here, we sought to determine whether nectin-1 was required for chlamydial development in vivo by intravaginally infecting nectin-1-/- mice with Chlamydia muridarum and monitoring chlamydial shedding by chlamydial titer assay. We observed a significant reduction in chlamydial shedding in female nectin-1-/- mice compared to nectin-1+/+ control mice, an observation that was confirmed by PCR. Immunohistochemical staining in mouse cervical tissue confirmed that there are fewer chlamydial inclusions in Chlamydia-infected nectin-1-/- mice. Notably, anorectal chlamydial infections are becoming a substantial health burden, though little is known regarding the pathogenesis of these infections. We therefore established a novel male murine model of rectal chlamydial infection, which we used to determine whether nectin-1 is required for anorectal chlamydial infection in male mice. In contrast to the data from vaginal infection, no difference in rectal chlamydial shedding was observed when male nectin-1+/+ and nectin-1-/- mice were compared. Through the use of these two models, we have demonstrated that nectin-1 promotes chlamydial infection in the female genital tract but does not appear to contribute to rectal infection in male mice. These models could be used to further characterize tissue and sex related differences in chlamydial infection.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Infecciones por Chlamydia/genética , Enfermedades de los Genitales Femeninos/genética , Enfermedades del Recto/genética , Infecciones del Sistema Genital/genética , Animales , Moléculas de Adhesión Celular/genética , Chlamydia muridarum/crecimiento & desarrollo , Chlamydia muridarum/patogenicidad , Chlamydia trachomatis/crecimiento & desarrollo , Chlamydia trachomatis/patogenicidad , Femenino , Predisposición Genética a la Enfermedad , Enfermedades de los Genitales Femeninos/microbiología , Células HeLa , Interacciones Huésped-Patógeno/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nectinas , Enfermedades del Recto/microbiología
7.
PLoS One ; 11(1): e0146186, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26726882

RESUMEN

Chlamydia trachomatis and Herpes Simplex Virus-2 (HSV-2) genital tract co-infections have been reported in humans and studied in vitro but the clinical consequences are unknown. Limited epidemiologic evidence suggests that these co-infections could be more severe than single infections of either pathogen, but the host-pathogen interactions during co-infection remain uncharacterized. To determine whether disease progression and/or pathogen shedding differs between singly-infected and super-infected animals, we developed an in vivo super-infection model in which female BALB/c mice were vaginally infected with Chlamydia muridarum (Cm) followed later by HSV-2. Pre-infection with Chlamydia 3 or 9 days prior to HSV-2 super-infection conferred significant protection from HSV-2-induced neurologic disease and significantly reduced viral recovery compared to HSV-2 singly-infected controls. Neither protection from mortality nor reduced viral recovery were observed when mice were i) super-infected with HSV-2 on day 27 post Cm; ii) infected with UV-irradiated Cm and super-infected with HSV-2; or iii) azithromycin-treated prior to HSV-2 super-infection. Therefore, protection from HSV-2-induced disease requires active infection with viable chlamydiae and is not observed after chlamydial shedding ceases, either naturally or due to antibiotic treatment. Thus, Chlamydia-induced protection is transient and requires the continued presence of chlamydiae or their components. These data demonstrate that chlamydial pre-infection can alter progression of subsequent HSV-2 infection, with implications for HSV-2 transmission from co-infected humans.


Asunto(s)
Infecciones por Chlamydia/complicaciones , Chlamydia trachomatis/fisiología , Herpes Genital/prevención & control , Herpesvirus Humano 2/fisiología , Interacciones Huésped-Patógeno , Sobreinfección , Vaginosis Bacteriana/complicaciones , Animales , Azitromicina/uso terapéutico , Infecciones por Chlamydia/tratamiento farmacológico , Infecciones por Chlamydia/microbiología , Infecciones por Chlamydia/virología , Chlamydia trachomatis/aislamiento & purificación , Chlamydia trachomatis/efectos de la radiación , Coinfección , Progresión de la Enfermedad , Femenino , Herpes Genital/complicaciones , Herpes Genital/microbiología , Herpes Genital/virología , Herpesvirus Humano 2/aislamiento & purificación , Ratones , Ratones Endogámicos BALB C , Paraplejía/etiología , Paraplejía/virología , Factores de Tiempo , Vaginosis Bacteriana/tratamiento farmacológico , Vaginosis Bacteriana/microbiología , Vaginosis Bacteriana/virología , Carga Viral
8.
JPEN J Parenter Enteral Nutr ; 40(6): 877-82, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-25940610

RESUMEN

BACKGROUND: There is no consensus whether resting energy expenditure (REE) following orthotopic liver transplantation (OLT) is altered. METHODS: The objectives of this investigation were to describe changes in measured REE (mREE) using indirect calorimetry in 25 OLT patients on days 5, 10, and 15 after baseline (within 72 hours following OLT) and compare mREE changes with those calculated with 2 predicted equations for energy expenditure (pREE): the Harris-Benedict and Schofield equations. RESULTS: Patients were 57 ± 5.4 years of age, 44% were male, 36% were black, and 72% had liver disease of viral etiology. Measured REE (at baseline and days 5, 10, and 15, per kcal/d: 1832 ± 952, 1565 ± 383, 1538 ± 345, 1578 ± 418) and kcal per kilogram of body weight (22.7 ± 12.8, 18.4 ± 4, 18.7 ± 3.8, 21 ± 6.5) did not change over time. In contrast, changes in pREE based on either the Harris-Benedict (P < .001) or Schofield (P = .006) equation using measured weights at each corresponding time point and lowest body weight during the study to estimate dry weight were significant. CONCLUSIONS: Wide ranges in both mREE and mREE expressed per kilogram of body weight at each study time point were observed in contrast to pREE, which declined by day 15. The observed differences in mREE over time suggest indirect calorimetry is indicated if available following OLT. Additional research is warranted to determine the most appropriate predictive equation with suitable stress factors to use when indirect calorimetry is not available.


Asunto(s)
Metabolismo Energético/fisiología , Hepatopatías/cirugía , Trasplante de Hígado/métodos , Calorimetría Indirecta , Enfermedad Hepática en Estado Terminal/cirugía , Femenino , Humanos , Masculino , Persona de Mediana Edad , Descanso
9.
Pathog Dis ; 73(4)2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25724891

RESUMEN

Studies indicate that estrogen enhances Chlamydia trachomatis serovar E infection in genital epithelial cells. Hormones have direct and indirect effects on endometrial epithelial cells. Estrogen and progesterone exposure induces endometrial stromal cells to release effectors that subsequently regulate growth and maturation of uterine epithelial cells. Estrogen enhances C. trachomatis infection by aiding entry and intracellular development in endometrial epithelial cell (Ishikawa, IK)/SHT-290 stromal cell co-culture. Enhanced chlamydial infection was mediated by direct estrogen-stimulated signaling events in epithelial cells and indirectly via estrogen-induced stromal cell effectors. The current study investigates the effects of hormones on chlamydial development using culture conditions representative of the menstrual cycle. Chlamydia trachomatis-infected IK or IK/SHT-290 cultures were exposed to 10(-8) M estrogen (E2), 10(-7) M progesterone (P4) or a combination of both hormones (10(-8) M E2 followed by 10(-9) M E2/10(-7) M P4). Chlamydial infectivity and progeny production were significantly decreased (30-66%) in cultures exposed to progesterone or estrogen/progesterone combination compared to estrogen alone. Thus, progesterone antagonized the positive effects of estrogen on chlamydial infection. These data indicate the susceptibility of endometrial epithelial cells to C. trachomatis infection during the menstrual cycle is altered by phase specific actions of sex hormones in the genital tract.


Asunto(s)
Chlamydia trachomatis/efectos de los fármacos , Chlamydia trachomatis/crecimiento & desarrollo , Endocitosis , Células Epiteliales/efectos de los fármacos , Células Epiteliales/microbiología , Estrógenos/metabolismo , Progesterona/metabolismo , Línea Celular , Chlamydia trachomatis/clasificación , Técnicas de Cocultivo , Femenino , Humanos , Serogrupo
10.
Artículo en Inglés | MEDLINE | ID: mdl-25414835

RESUMEN

Interaction of Herpes Simplex Virus (HSV) glycoprotein D (gD) with the host cell surface during Chlamydia trachomatis/HSV co-infection stimulates chlamydiae to become persistent. During viral entry, gD interacts with one of 4 host co-receptors: HVEM (herpes virus entry mediator), nectin-1, nectin-2 and 3-O-sulfated heparan sulfate. HVEM and nectin-1 are high-affinity entry receptors for both HSV-1 and HSV-2. Nectin-2 mediates HSV-2 entry but is inactive for HSV-1, while 3-O-sulfated heparan sulfate facilitates HSV-1, but not HSV-2, entry. Western blot and RT-PCR analyses demonstrate that HeLa and HEC-1B cells express nectin-1 and nectin-2, but not HVEM. Because both HSV-1 and HSV-2 trigger persistence, these data suggest that nectin-1 is the most likely co-receptor involved. Co-infections with nectin-1 specific HSV-1 mutants stimulate chlamydial persistence, as evidenced by aberrant body (AB) formation and decreased production of elementary bodies (EBs). These data indicate that nectin-1 is involved in viral-induced chlamydial persistence. However, inhibition of signal transduction molecules associated with HSV attachment and entry does not rescue EB production during C. trachomatis/HSV-2 co-infection. HSV attachment also does not activate Cdc42 in HeLa cells, as would be expected with viral stimulated activation of nectin-1 signaling. Additionally, immunofluorescence assays confirm that HSV infection decreases nectin-1 expression. Together, these observations suggest that gD binding-induced loss of nectin-1 signaling negatively influences chlamydial growth. Chlamydial infection studies in nectin-1 knockdown (NKD) HeLa cell lines support this hypothesis. In NKD cells, chlamydial inclusions are smaller in size, contain ABs, and produce significantly fewer infectious EBs compared to C. trachomatis infection in control HeLa cells. Overall, the current study indicates that the actions of host molecule, nectin-1, are required for successful C. trachomatis development.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Infecciones por Chlamydia/metabolismo , Infecciones por Chlamydia/microbiología , Chlamydia trachomatis/clasificación , Chlamydia trachomatis/fisiología , Interacciones Huésped-Patógeno , Animales , Moléculas de Adhesión Celular/genética , Línea Celular , Infecciones por Chlamydia/genética , Coinfección , Cricetinae , Expresión Génica , Técnicas de Inactivación de Genes , Células HeLa , Herpesvirus Humano 1 , Humanos , Cuerpos de Inclusión Viral , Nectinas , Estrés Oxidativo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/genética , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Simplexvirus/fisiología , Acoplamiento Viral , Internalización del Virus
11.
Artículo en Inglés | MEDLINE | ID: mdl-24783061

RESUMEN

Chlamydia trachomatis, the most common bacterial sexually transmitted disease agent worldwide, enters a viable, non-dividing and non-infectious state (historically termed persistence and more recently referred to as the chlamydial stress response) when exposed to penicillin G in culture. Notably, penicillin G-exposed chlamydiae can reenter the normal developmental cycle upon drug removal and are resistant to azithromycin-mediated killing. Because penicillin G is less frequently prescribed than other ß-lactams, the clinical relevance of penicillin G-induced chlamydial persistence/stress has been questioned. The goal of this study was to determine whether more commonly used penicillins also induce C. trachomatis serovar E persistence/stress. All penicillins tested, as well as clavulanic acid, induced formation of aberrant, enlarged reticulate bodies (RB) (called aberrant bodies or AB) characteristic of persistent/stressed chlamydiae. Exposure to the penicillins and clavulanic acid also reduced chlamydial infectivity by >95%. None of the drugs tested significantly reduced chlamydial unprocessed 16S rRNA or genomic DNA accumulation, indicating that the organisms were viable, though non-infectious. Finally, recovery assays demonstrated that chlamydiae rendered essentially non-infectious by exposure to ampicillin, amoxicillin, carbenicillin, piperacillin, penicillin V, and clavulanic acid recovered infectivity after antibiotic removal. These data definitively demonstrate that several commonly used penicillins induce C. trachomatis persistence/stress at clinically relevant concentrations.


Asunto(s)
Antibacterianos/farmacología , Chlamydia trachomatis/efectos de los fármacos , Chlamydia trachomatis/fisiología , Estrés Fisiológico/efectos de los fármacos , beta-Lactamas/farmacología , Línea Celular , Células Cultivadas , Infecciones por Chlamydia/microbiología , Chlamydia trachomatis/ultraestructura , ADN Bacteriano/efectos de los fármacos , Humanos , Pruebas de Sensibilidad Microbiana , Penicilinas/farmacología , ARN Ribosómico 16S/efectos de los fármacos , ARN Ribosómico 16S/genética
12.
Microbiology (Reading) ; 154(Pt 5): 1290-1299, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18451037

RESUMEN

Nectin-1, a member of the immunoglobulin superfamily, is a Ca(2+)-independent cell adhesion protein implicated in the organization of E-cadherin-based adherens junctions (AJs) and claudin-based tight junctions (TJs) in epithelial cells. Nectin-1 also regulates cell-cell adhesion and cell polarization in a Cdc42- and Rac-dependent manner. Western blot analyses demonstrated that accumulation of host nectin-1 is decreased by 85 % at 48 hours post-infection (h.p.i.) in Chlamydia trachomatis serovar E-infected HeLa cells. Time-course experiments demonstrated that this decrease was sustained to 60 h.p.i. Nectin-1 downregulation in C. trachomatis-infected cells was prevented by both chloramphenicol exposure and prior inactivation of the chlamydiae with UV light, demonstrating that active C. trachomatis replication was required. Penicillin G-exposure studies demonstrated that nectin-1 accumulation was also altered during persistent infection. Finally, RT-PCR analyses indicated that chlamydial infection did not alter accumulation of any nectin-1 transcripts, demonstrating that nectin-1 accumulation is reduced at a post-transcriptional level. Intesrestingly, N-cadherin-dependent cell-cell junctions can be disrupted by C. trachomatis infection, as reported by Prozialeck et al. (2002). Because interaction of nectin molecules on adjacent cells is essential for AJ formation, these data suggest that C. trachomatis may disrupt AJs, at least in part, by diminishing nectin-1 accumulation. Notably, release of chlamydiae-infected epithelial cells has been observed both in vitro from polarized monolayers and in vivo from tissues, suggesting that chlamydia-modulated downregulation of adhesion molecules and the subsequent disruption of host cell adherence may be involved in chlamydial dissemination or pathogenesis.


Asunto(s)
Moléculas de Adhesión Celular/biosíntesis , Chlamydia trachomatis/fisiología , Regulación hacia Abajo , Células Epiteliales/microbiología , Uniones Adherentes/microbiología , Uniones Adherentes/patología , Perfilación de la Expresión Génica , Células HeLa , Humanos , Nectinas , ARN Mensajero/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo
13.
Microbiology (Reading) ; 154(Pt 3): 971-978, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18310043

RESUMEN

Several inducers of chlamydial persistence have been described, including interferon-gamma (IFN-gamma), IFN-alpha, IFN-beta, and tumour necrosis factor-alpha (TNF-alpha) exposure, and iron, amino acid or glucose deprivation. A tissue-culture model of Chlamydia trachomatis/herpes simplex virus type-2 (HSV-2) co-infection indicates that viral co-infection stimulates the formation of persistent chlamydiae. This study was designed to ascertain whether co-infection-induced persistence is mediated by a previously characterized mechanism. Luminex assays indicate that IFN-gamma, IFN-alpha, and TNF-alpha are not released from co-infected cells. Semiquantitative RT-PCR studies demonstrate that IFN-beta, IFN-gamma, indoleamine 2,3-dioxygenase, lymphotoxin-alpha and inducible nitric oxide synthase are not expressed during co-infection. These data indicate that viral-induced persistence is not stimulated by any persistence-associated cytokine. Supplementation of co-infected cells with excess amino acids, iron-saturated holotransferrin, glucose or a combination of amino acids and iron does not restore chlamydial infectivity, demonstrating that HSV-2-induced persistence is not mediated by depletion of these nutrients. Finally, inclusions within co-infected cells continue to enlarge and incorporate C(6)-NBD-ceramide, indicating that HSV-2 co-infection does not inhibit vesicular transport to the developing inclusion. Collectively these data demonstrate that co-infection-induced persistence is not mediated by any currently characterized persistence inducer or anti-chlamydial pathway. Previous studies indicate that HSV-2 attachment and/or entry into the host cell is sufficient for stimulating chlamydial persistence, suggesting that viral attachment and/or entry may trigger a novel host pathway which restricts chlamydial development.


Asunto(s)
Chlamydia trachomatis/crecimiento & desarrollo , Herpesvirus Humano 2/crecimiento & desarrollo , Aminoácidos/metabolismo , Línea Celular , Chlamydia trachomatis/patogenicidad , Perfilación de la Expresión Génica , Humanos , Cuerpos de Inclusión/microbiología , Indolamina-Pirrol 2,3,-Dioxigenasa/biosíntesis , Interferón-alfa/metabolismo , Interferón beta/biosíntesis , Interferón gamma/metabolismo , Hierro/metabolismo , Linfotoxina-alfa/biosíntesis , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Necrosis Tumoral alfa/metabolismo , Virulencia/efectos de los fármacos
14.
J Vasc Res ; 44(4): 283-91, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17406120

RESUMEN

BACKGROUND: Infusion of exogenous vascular endothelial growth factor (VEGF) into adult brain at doses above 60 ng/day induces dramatic angiogenesis accompanied by vascular leak and inflammation. Blood vessels formed by this treatment are dilated and tortuous, exhibiting a pathological morphology. Pathological VEGF-induced angiogenesis is preceded by vascular leak and inflammation, which have been proposed to mediate subsequent angiogenesis. METHODS: To test this hypothesis, we infused VEGF into the brains of adult rats to induce pathological angiogenesis. Some of these rats were treated with dexamethasone, a potent anti-inflammatory glucocorticoid, to inhibit inflammation and edema. RESULTS: We demonstrate that inhibition of inflammation by treatment with dexamethasone significantly attenuated VEGF-induced pathological angiogenesis. To present converging evidence that inflammation may be important in this angiogenic process, we also demonstrate that mice genetically deficient in the inflammatory mediator intercellular adhesion molecule-1 have attenuated VEGF-induced angiogenesis. These same mice showed normal amounts of physiological angiogenesis in response to enriched environments, however, suggesting that a generalized reduction in vascular plasticity could not account for their poor angiogenic response to VEGF. CONCLUSIONS: Taken together, the data from these experiments suggest that the inflammation which occurs before or during VEGF-induced pathological brain angiogenesis plays a contributory role in the pathological angiogenic process.


Asunto(s)
Circulación Cerebrovascular/efectos de los fármacos , Dexametasona/farmacología , Glucocorticoides/farmacología , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/inmunología , Neovascularización Patológica/tratamiento farmacológico , Factores de Edad , Animales , Encéfalo/irrigación sanguínea , Encéfalo/inmunología , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Masculino , Ratones , Ratones Noqueados , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/inmunología , Ratas , Ratas Sprague-Dawley , Venas Umbilicales/citología , Factor A de Crecimiento Endotelial Vascular/farmacología
15.
Cell Microbiol ; 9(3): 725-37, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17140408

RESUMEN

Epidemiological studies have demonstrated that co-infections of herpes simplex virus type 2 (HSV-2) and Chlamydia trachomatis occur in vivo. Data from a tissue culture model of C. trachomatis/HSV-2 co-infection indicate that viral co-infection stimulates the formation of persistent chlamydiae. Transmission electron microscopic (TEM) analyses demonstrated that in both HeLa and HEC-1B cells, co-infection caused developing chlamydiae to exhibit swollen, aberrantly shaped reticulate bodies (RBs), characteristically observed in persistence. Additionally, HSV-2 co-infection suppressed production of infectious chlamydial elementary bodies (EBs) in both host cell types. Co-infection with HSV type 1 (HSV-1) produced similar morphologic alterations and abrogated infectious EB production. These data indicate that virus-induced chlamydial persistence was neither host cell- nor virus strain-specific. Purification of crude HSV-2 stocks demonstrated that viral particles were required for coinfection-induced chlamydial persistence to occur. Finally, co-infection with either UV-inactivated, replication-incompetent virus or replication-competent HSV-2 in the presence of cyclohexamide reduced chlamydial infectivity without altering chlamydial genomic DNA accumulation. These data demonstrate that productive viral replication is not required for the induction of chlamydial persistence and suggest that HSV attachment and entry can provide the necessary stimulus to alter C. trachomatis development.


Asunto(s)
Chlamydia trachomatis/efectos de los fármacos , Cicloheximida/farmacología , Herpesvirus Humano 2/efectos de los fármacos , Animales , Línea Celular , Chlamydia trachomatis/fisiología , Chlorocebus aethiops , Células Epiteliales/microbiología , Células Epiteliales/ultraestructura , Células Epiteliales/virología , Células HeLa , Herpesvirus Humano 2/fisiología , Humanos , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Inhibidores de la Síntesis de la Proteína/farmacología , Factores de Tiempo , Células Vero
16.
Nat Med ; 11(2): 199-205, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15654325

RESUMEN

Genetic ablation of Inppl1, which encodes SHIP2 (SH2-domain containing inositol 5-phosphatase 2), was previously reported to induce severe insulin sensitivity, leading to early postnatal death. In the previous study, the targeting construct left the first eighteen exons encoding Inppl1 intact, generating a Inppl1(EX19-28-/-) mouse, and apparently also deleted a second gene, Phox2a. We report a new SHIP2 knockout (Inppl1(-/-)) targeted to the translation-initiating ATG, which is null for Inppl1 mRNA and protein. Inppl1(-/-) mice are viable, have normal glucose and insulin levels, and normal insulin and glucose tolerances. The Inppl1(-/-) mice are, however, highly resistant to weight gain when placed on a high-fat diet. These results suggest that inhibition of SHIP2 would be useful in the effort to ameliorate diet-induced obesity, but call into question a dominant role of SHIP2 in modulating glucose homeostasis.


Asunto(s)
Grasas de la Dieta/metabolismo , Obesidad/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Animales , Análisis Químico de la Sangre , Peso Corporal , Exones , Femenino , Eliminación de Gen , Genes Reporteros , Glucosa/metabolismo , Homeostasis , Inositol Polifosfato 5-Fosfatasas , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas , Monoéster Fosfórico Hidrolasas/genética , Transducción de Señal , Distribución Tisular
17.
Exp Neurol ; 187(2): 388-402, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15144865

RESUMEN

Vascular endothelial growth factor (VEGF) has been shown to induce angiogenesis when infused continuously into adult rat brain tissue. In addition, VEGF has been shown to enhance permeability in brain vasculature. Adult rats were continuously infused with mouse VEGF into neocortex for up to 7 days. We studied the development of VEGF-induced vasculature in rat neocortex and evaluated the temporal expression of a wide variety of markers for inflammation and vascular leak in relation to the angiogenic response using immunohistochemistry and Western blot analysis. We report here that VEGF-mediated inflammation in brain is characterized by upregulation of ICAM-1 and the chemokine MIP-1alpha, as well as a preferential extravasation of monocytes. VEGF causes a dramatic breakdown of the blood-brain barrier, which is characterized by decreased investment of the vasculature with astroglial endfeet. Perivascular cells, in contrast, increase around the newly formed cerebrovasculature. In addition, breakdown of the blood-brain barrier, leukocyte extravasation, and extracellular matrix deposition occur before vascular proliferation. Furthermore, administration of low doses of VEGF induces permeability and inflammation without appreciable vascular proliferation.


Asunto(s)
Encéfalo/irrigación sanguínea , Encéfalo/efectos de los fármacos , Encefalitis/inducido químicamente , Mediadores de Inflamación/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/patología , Barrera Hematoencefálica/efectos de los fármacos , Western Blotting , Encéfalo/patología , Permeabilidad Capilar/efectos de los fármacos , División Celular/efectos de los fármacos , Quimiocinas/genética , Relación Dosis-Respuesta a Droga , Vías de Administración de Medicamentos , Encefalitis/metabolismo , Encefalitis/patología , Hibridación in Situ , Mediadores de Inflamación/administración & dosificación , Molécula 1 de Adhesión Intercelular/metabolismo , Antígeno Ki-67/metabolismo , Masculino , Neocórtex/irrigación sanguínea , Neocórtex/efectos de los fármacos , Neocórtex/patología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Factor A de Crecimiento Endotelial Vascular/administración & dosificación
18.
Toxicol Appl Pharmacol ; 196(1): 68-79, 2004 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15050409

RESUMEN

Mitochondrial DNA-deficient (rho(0)) cells were generated following a 26-day incubation of MOLT-4 lymphoblastoid T cells in ethidium bromide (3,8-diamino-5-ethyl-6-phenylphenanthridinium bromide). The absence of mitochondrial DNA (mtDNA) in the resultant MOLT-4 rho(0) cells was confirmed by Southern analysis and quantitative polymerase chain reaction (PCR). MOLT-4 rho(0) cells proliferated more slowly than parental cells (wild type) and produced significantly more lactate (approximately fourfold increase; P < 0.001) with concomitantly reduced oxygen consumption (12.3% vs. 100%; P < 0.001) compared with the wild type. MOLT-4 rho(0) cells also showed reduced cytochrome c oxidase activity and a reduced cytochrome c oxidase/citrate synthase activity ratio compared to parental wild-type MOLT-4 cells (P < 10(-11)). Electron microscopy showed elongated mitochondria with parallel cristae in MOLT-4 cells although the mitochondria in MOLT-4 rho(0) cells appeared enlarged, some were vacuolated with either an absent or a grossly distorted cristae pattern. Vital staining with 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazolyl-carbocyanine iodide (JC-1) was used to image mitochondria in intact cells and study mitochondrial transmembrane potential (Deltapsi(m)). Flow cytometry using JC-1 indicated that MOLT-4 rho(0) had a lower Deltapsi(m) than MOLT-4. Sodium fluoride (an inhibitor of the glycolytic pathway) at a concentration of 20 mM further reduced the Deltapsi(m) in MOLT-4-rho(0) cells. This data suggested that a glycolytic pathway product, possibly ATP, was required for the maintenance of Deltapsi(m) in MOLT-4 rho(0) cells.


Asunto(s)
ADN Mitocondrial/metabolismo , Etidio/toxicidad , Mitocondrias/ultraestructura , Southern Blotting , División Celular/efectos de los fármacos , Línea Celular Tumoral , Respiración de la Célula/efectos de los fármacos , Citrato (si)-Sintasa/metabolismo , ADN Mitocondrial/genética , Complejo IV de Transporte de Electrones/metabolismo , Humanos , Leucemia de Células T/patología , Potenciales de la Membrana/efectos de los fármacos , Microscopía Electrónica , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mutación , Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...