Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Biol Res Commun ; 10(4): 165-170, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35097138

RESUMEN

We investigated the association between p16 expression and histopathologic parameters including size, neural and vascular invasion, and lymph node involvement in breast cancer. 58 specimens from patients with different grades of breast cancer were included. Hematoxylin and eosin and immunohistochemistry staining for p16 was performed. 5 patients (8.6%) had grade I, 23 (39.7%) had grade II, and 30 (51.7%) had grade III breast cancer. Assessment of the tumor size showed that 5 (8.6%) tumors had a size of ≤2cm, 29 (50%) were between 2-5 cm and 24 (41.4%) had a size of ≥5cm. Moreover, 45 (77.6%) of the included patients had axillary lymph node involvement. Investigation of association between p16 positivity with pathological parameters in three groups with positivity to p16 (1-25%, 26-75%, >75%) showed that there was no association between p16 positivity and other parameters including histologic score (p=0.44), tumor size (p=0.77), neural invasion (p=0.79), perivascular invasion (p=0.98) and the number of involved LNs (p=0.49). From the group including eight patients with >75% p16 positivity, seven (87.5%) were found with neural invasion and two (25%) with perivascular invasion. P16 positivity was not associated with size, neural and vascular invasion, and LN involvement in breast cancer.

2.
Front Immunol ; 10: 2075, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31552032

RESUMEN

Tissue-resident memory T cells (TRM) are critical to host defense at mucosal tissue sites. However, the parenteral route of immunization as the most commonly used immunization route in practice is not effective in inducing mucosal TRM cells particularly in the lung. While various respiratory mucosal (RM)-pull strategies are exploited to mobilize parenteral vaccine-primed T cells into the lung, whether such RM-pull strategies can all or differentially induce Ag-specific TRM cells in the lung remains unclear. Here, we have addressed this issue by using a parenteral TB vaccine-primed and RM-pull model. We show that both Ag-independent and Ag-dependent RM-pull strategies are able to mobilize Ag-specific CD8 T cells into the lung. However, only the RM-pull strategy with cognate antigens can induce robust Ag-specific CD8 TRM cells in the lung. We also show that the cognate Ag-based RM-pull strategy is the most effective in inducing TRM cells when carried out during the memory phase, as opposed to the effector phase, of T cell responses to parenteral prime vaccination. We further find that cognate Ag-induced CD4 T cells play an important role in the development of CD8 TRM cells in the lung. Our study holds implications in developing effective vaccine strategies against respiratory pathogens.


Asunto(s)
Antígenos Bacterianos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Interacciones Huésped-Patógeno/inmunología , Memoria Inmunológica , Membrana Mucosa/inmunología , Vacunas contra la Tuberculosis/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Citocinas/metabolismo , Femenino , Inmunización , Mediadores de Inflamación/metabolismo , Activación de Linfocitos/inmunología , Ratones , Membrana Mucosa/metabolismo , Mycobacterium tuberculosis/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
3.
Cell ; 175(6): 1634-1650.e17, 2018 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-30433869

RESUMEN

Innate immune memory is an emerging area of research. However, innate immune memory at major mucosal sites remains poorly understood. Here, we show that respiratory viral infection induces long-lasting memory alveolar macrophages (AMs). Memory AMs are programed to express high MHC II, a defense-ready gene signature, and increased glycolytic metabolism, and produce, upon re-stimulation, neutrophil chemokines. Using a multitude of approaches, we reveal that the priming, but not maintenance, of memory AMs requires the help from effector CD8 T cells. T cells jump-start this process via IFN-γ production. We further find that formation and maintenance of memory AMs are independent of monocytes or bone marrow progenitors. Finally, we demonstrate that memory AMs are poised for robust trained immunity against bacterial infection in the lung via rapid induction of chemokines and neutrophilia. Our study thus establishes a new paradigm of immunological memory formation whereby adaptive T-lymphocytes render innate memory of mucosal-associated macrophages.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunidad Innata , Pulmón/inmunología , Macrófagos Alveolares/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Linfocitos T CD8-positivos/citología , Memoria Inmunológica , Pulmón/citología , Macrófagos Alveolares/citología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Monocitos/citología , Monocitos/inmunología , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología , Linfocitos T Colaboradores-Inductores/citología
4.
J Immunol ; 199(7): 2555-2569, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28827285

RESUMEN

Although most novel tuberculosis (TB) vaccines are designed for delivery via the muscle or skin for enhanced protection in the lung, it has remained poorly understood whether systemic vaccine-induced memory T cells can readily home to the lung mucosa prior to and shortly after pathogen exposure. We have investigated this issue by using a model of parenteral TB immunization and intravascular immunostaining. We find that systemically induced memory T cells are restricted to the blood vessels in the lung, unable to populate either the lung parenchymal tissue or the airway under homeostatic conditions. We further find that after pulmonary TB infection, it still takes many days before such T cells can enter the lung parenchymal tissue and airway. We have identified the acquisition of CXCR3 expression by circulating T cells to be critical for their entry to these lung mucosal compartments. Our findings offer new insights into mucosal T cell biology and have important implications in vaccine strategies against pulmonary TB and other intracellular infections in the lung.


Asunto(s)
Pulmón/inmunología , Mycobacterium tuberculosis/inmunología , Receptores CXCR3/metabolismo , Vacunas contra la Tuberculosis/inmunología , Tuberculosis Pulmonar/inmunología , Traslado Adoptivo , Animales , Antígenos Bacterianos/inmunología , Linfocitos T CD8-positivos/inmunología , Movimiento Celular , Inmunización , Memoria Inmunológica , Leucocitos/inmunología , Pulmón/citología , Pulmón/microbiología , Ratones , Receptores CXCR3/genética , Receptores CXCR3/inmunología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/microbiología , Transducción de Señal , Vacunas contra la Tuberculosis/administración & dosificación , Tuberculosis Pulmonar/microbiología
5.
Sci Rep ; 7(1): 9525, 2017 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-28842633

RESUMEN

Lung resident memory T cells (TRM) characterized by selective expression of mucosal integrins VLA-1 (α1ß1) and CD103 (αEß7) are generated following primary respiratory viral infections. Despite recent progress, the generation of lung TRM and the role of mucosal integrins following viral vector respiratory mucosal immunization still remains poorly understood. Here by using a replication-defective viral vector tuberculosis vaccine, we show that lung Ag-specific CD8 T cells express both VLA-1 and CD103 following respiratory mucosal immunization. However, VLA-1 and CD103 are acquired in differential tissue sites with the former acquired during T cell priming in the draining lymph nodes and the latter acquired after T cells entered the lung. Once in the lung, Ag-specific CD8 T cells continue to express VLA-1 at high levels through the effector/expansion, contraction, and memory phases of T cell responses. Using a functional VLA-1 blocking mAb, we show that VLA-1 is not required for trafficking of these cells to the lung, but it negatively regulates them in the contraction phase. Furthermore, VLA-1 plays a negligible role in the maintenance of these cells in the lung. Our study provides new information on vaccine-inducible lung TRM and shall help develop effective viral vector respiratory mucosal tuberculosis vaccination strategies.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Expresión Génica , Memoria Inmunológica , Integrina alfa1beta1/genética , Mucosa Respiratoria/inmunología , Vacunas contra la Tuberculosis/inmunología , Tuberculosis/etiología , Animales , Linfocitos T CD8-positivos/metabolismo , Epítopos de Linfocito T/inmunología , Femenino , Vectores Genéticos/genética , Inmunofenotipificación , Integrina alfa1beta1/metabolismo , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ratones , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/microbiología , Tuberculosis/metabolismo , Tuberculosis/prevención & control
6.
J Infect Dis ; 216(1): 135-145, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28531291

RESUMEN

Background: The translation of preclinically promising novel tuberculosis vaccines to ultimate human applications has been challenged by the lack of animal models with an immune system equivalent to the human immune system in its genetic diversity and level of susceptibility to tuberculosis. Methods: We have developed a humanized mice (Hu-mice) tuberculosis model system to investigate the clinical relevance of a novel virus-vectored (VV) tuberculosis vaccine administered via respiratory mucosal or parenteral route. Results: We find that VV vaccine activates T cells in Hu-mice as it does in human vaccinees. The respiratory mucosal route for delivery of VV vaccine in Hu-mice, but not the parenteral route, significantly reduces the humanlike lung tuberculosis outcomes in a human T-cell-dependent manner. Conclusions: Our results suggest that the Hu-mouse can be used to predict the protective efficacy of novel tuberculosis vaccines/strategies before they proceed to large, expensive human trials. This new vaccine testing system will facilitate the global pace of clinical tuberculosis vaccine development.


Asunto(s)
Vacuna BCG/administración & dosificación , Inmunidad Mucosa , Mucosa Respiratoria/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Antígenos Virales/sangre , Antígenos Virales/inmunología , Vacuna BCG/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Vectores Genéticos/inmunología , Humanos , Inmunización , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/microbiología , Ratones , Ratones Noqueados , Mycobacterium tuberculosis/inmunología , Tuberculosis Pulmonar/prevención & control
7.
Vaccine ; 35(22): 2916-2924, 2017 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-28438408

RESUMEN

Cold chain-free vaccine technologies are needed to ensure effective vaccine delivery and coverage, particularly in resource-poor countries. However, the immunogenicity and thermostability of spray dried live viral vector-based vaccines such as recombinant adenoviral-vectored vaccines remain to be investigated. To address this issue, we have spray dried human adenoviral (AdHu5)- and chimpanzee adenoviral (AdCh68)-vectored tuberculosis vaccines in a mannitol and dextran matrix. Spray dried powders containing these two vaccines display the morphologic and chemical properties desired for long-term thermostability and vaccination. Upon reconstitution, they effectively transfected the cells in vitro with relatively small losses in viral infectivity related to the spray drying process. Following in vivo vaccination, AdHu5- and AdCh68-vectored vaccines were as immunogenic as the conventional fresh, cryopreserved liquid vaccine samples. Of importance, even after cold chain-free storage, at ambient temperatures and relatively low humidity for 30 and 90days, the vaccines retained their in vivo immunogenicity, while the liquid vaccine samples stored under the same conditions lost their immune-activating capability almost entirely. Our results support further development of our spray drying technologies for generating thermally stable adenoviral-vectored and other viral-vectored vaccines.


Asunto(s)
Adenoviridae/genética , Inmunogenicidad Vacunal , Vacunas contra la Tuberculosis/inmunología , Potencia de la Vacuna , Vacunas Sintéticas/inmunología , Adenovirus de los Simios , Animales , Desecación , Almacenaje de Medicamentos , Humanos , Manitol , Pan troglodytes , Polvos , Temperatura , Trehalosa , Vacunas contra la Tuberculosis/administración & dosificación , Vacunas contra la Tuberculosis/química , Vacunas Sintéticas/química
8.
Eur Respir Rev ; 24(136): 356-60, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26028646

RESUMEN

Despite the use of bacille Calmette-Guérin (BCG) for almost a century, pulmonary tuberculosis (TB) continues to be a serious global health concern. Therefore, there has been a pressing need for the development of new booster vaccines to enhance existing BCG-induced immunity. Protection following mucosal intranasal immunisation with AdHu5Ag85A is associated with the localisation of antigen-specific T-cells to the lung airway. However, parenteral intramuscular immunisation is unable to provide protection despite the apparent presence of antigen-specific T-cells in the lung interstitium. Recent advances in intravascular staining have allowed us to reassess the previously established T-cell distribution profile and its relationship with the observed differential protection. Respiratory mucosal immunisation empowers T-cells to home to both the lung interstitium and the airway lumen, whereas intramuscular immunisation-activated T-cells are largely trapped within the pulmonary vasculature, unable to populate the lung interstitium and airway. Given the mounting evidence supporting the safety and enhanced efficacy of respiratory mucosal immunisation over the traditional parenteral immunisation route, a greater effort should be made to clinically develop respiratory mucosal-deliverable TB vaccines.


Asunto(s)
Quimiotaxis de Leucocito , Inmunidad Mucosa , Inmunización , Pulmón/inmunología , Mycobacterium tuberculosis/inmunología , Linfocitos T/inmunología , Vacunas contra la Tuberculosis/uso terapéutico , Tuberculosis Pulmonar/prevención & control , Administración por Inhalación , Animales , Interacciones Huésped-Patógeno , Humanos , Mediadores de Inflamación/inmunología , Pulmón/microbiología , Mycobacterium tuberculosis/patogenicidad , Linfocitos T/microbiología , Resultado del Tratamiento , Vacunas contra la Tuberculosis/administración & dosificación , Vacunas contra la Tuberculosis/inmunología , Tuberculosis Pulmonar/diagnóstico , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/microbiología
9.
Can J Vet Res ; 78(4): 274-82, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25355996

RESUMEN

Macrophages function as phagocytes and antigen-presenting cells in the body. As has been demonstrated in mammals, administration of clodronate [dichloromethylene bisphosphonate (Cl2MBP)] encapsulated liposomes results in depletion of macrophages. Although this compound has been used in chickens, its effectiveness in depleting macrophages has yet to be fully determined. Here, we show that a single administration of clodronate liposomes to chickens results in a significant depletion of macrophages within the spleen and lungs of chickens up to 4 d post-treatment. This finding suggests that, in order to obtain depletion of macrophages in chickens for greater than 5 d, it is necessary to administer clodronate liposomes 4 d apart. The study also showed that 2 treatments of clodronate liposomes at 4-day intervals resulted in the depletion of macrophages for up to 10 d. The findings of the present study will encourage more precise studies to be done on the potential roles of macrophages in immune responses and in the pathogenesis of microbial infections in chickens.


Les macrophages agissent comme phagocytes et cellules présentatrices d'antigènes dans l'organisme. Tel que démontré chez les mammifères, l'administration de liposomes encapsulés de clodronate [biphosphanate de dichlorométhylène (Cl2MBP)] cause une déplétion des macrophages. Bien que ce composé ait été utilisé chez les poulets, son efficacité à causer une déplétion des macrophages reste encore à être entièrement déterminée. Nous démontrons ici que l'administration d'une dose unique de liposomes de clodronate à des poulets a causé une déplétion significative des macrophages dans la rate et les poumons de poulets jusqu'à 4 j post-traitement. Cette trouvaille suggère qu'afin d'obtenir une déplétion des macrophages chez les poulets pour plus de 5 j, il est nécessaire d'administrer des liposomes de clodronate à un intervalle de 4 j. Cette étude a aussi démontré que deux traitements de liposomes de clodronate à 4 j d'intervalle a causé une déplétion des macrophages pour une durée allant jusqu'à 10 j. Les présentes trouvailles encourageront la mise en place d'études plus précises sur les rôles potentiels des macrophages dans la réponse immunitaire et dans la pathogénèse des infections microbiennes chez les poulets.(Traduit par Docteur Serge Messier).


Asunto(s)
Pollos/inmunología , Ácido Clodrónico/farmacología , Pulmón/inmunología , Macrófagos/inmunología , Bazo/inmunología , Animales , Ácido Clodrónico/administración & dosificación , Citometría de Flujo/veterinaria , Inmunohistoquímica/veterinaria , Liposomas/administración & dosificación , Pulmón/citología , Macrófagos/efectos de los fármacos , Bazo/citología
10.
Virology ; 450-451: 114-21, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24503073

RESUMEN

Infectious bronchitis virus (IBV) replicates in the epithelial cells of trachea and lungs of chicken, however the mechanism of generation of innate immune response against IBV infection in these tissues has not been fully characterized. Our objective was to study innate responses induced early following IBV infection in chickens. Initiation of the transcription of selected innate immune genes such as TLR3, TLR7, MyD88, IL-1ß and IFN-ß, as well as recruitment of macrophages, were evident following an initial down regulation of some of the observed genes (TLR3, IL-1ß, and IFN-γ) in trachea and lung. This initial down-regulation followed by the induction of innate immune response to IBV infection appears to be inadequate for the control of IBV genome accumulation and consequent histopathological changes in these tissues. Potential induction of innate immunity before infection occurs may be necessary to reduce the consequences since vaccine induced immunity is slow to develop.


Asunto(s)
Infecciones por Coronavirus/veterinaria , Inmunidad Innata , Virus de la Bronquitis Infecciosa/fisiología , Enfermedades de las Aves de Corral/inmunología , Sistema Respiratorio/inmunología , Animales , Proteínas Aviares/genética , Proteínas Aviares/inmunología , Pollos , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Virus de la Bronquitis Infecciosa/inmunología , Enfermedades de las Aves de Corral/genética , Enfermedades de las Aves de Corral/virología , Sistema Respiratorio/virología , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/inmunología , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/inmunología
11.
Vet Immunol Immunopathol ; 155(4): 270-5, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24034933

RESUMEN

LPS is one of the pathogen associated molecular patterns that activates Toll-like receptor 4 (TLR4) signaling pathway eliciting antiviral host responses in mammals although information on such responses in avian species is scarce. Our objectives were to characterize the LPS induced innate responses particularly the expression of LPS receptors (TLR4, CD14) in avian macrophages and observe whether TLR4 mediated induction of NO can elicit antiviral response against infectious laryngotracheitis virus (ILTV) replication. We found that LPS was capable of inducing the expression of TLR4, CD14 and NO production but not the type 1 interferons in an avian macrophage cell line, MQ-NCSU. We also showed that TLR4 mediated NO production can lead to antiviral response against ILTV replication when MQ-NCSU cells were treated with LPS and the resultant supernatant was then transferred to ILTV replicating cells to assess antiviral activity. Antiviral activity of NO was blocked by a selective inhibitor, S-methylisothiourea sulfate that inhibits inducible NO synthase. This observation confirms that the antiviral activity is positively correlated with NO production. The data show that LPS can be a potential innate immune stimulant that can be used against ILTV infection in chickens that require further evaluation in vivo.


Asunto(s)
Pollos , Infecciones por Herpesviridae/veterinaria , Herpesvirus Gallináceo 1/inmunología , Óxido Nítrico/inmunología , Enfermedades de las Aves de Corral/virología , Transducción de Señal/inmunología , Receptor Toll-Like 4/inmunología , Animales , Línea Celular , Inhibidores Enzimáticos/farmacología , Citometría de Flujo , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Isotiuronio/análogos & derivados , Isotiuronio/farmacología , Lipopolisacáridos/inmunología , Macrófagos , Óxido Nítrico/antagonistas & inhibidores , Enfermedades de las Aves de Corral/inmunología , Replicación Viral/inmunología
12.
J Basic Microbiol ; 50 Suppl 1: S68-73, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20967787

RESUMEN

We evaluated the ability of randomly amplified polymorphic DNA (RAPD) to type Epidermophyton floccosum isolates recovered from patients with dermatophytosis originating from different regions of Iran. A total of 13 clinical isolates of E. floccosum obtained from Iranian patients were analyzed by RAPD with 7 arbitrary primers (OPN16, OPD18' OPU15, OPX19, R28, OPA04 and OPAA17). Among the applied primers, OPN16 produced banding patterns from all the isolates. In addition, some of the isolates had very close relation. The phenon line which represented the mean similarities was at the value of 73%. At this level, 4 groups were characterized. Two isolates of a patient had different molecular patterns, suggesting infection transmission from different sources in the case of a single patient. RAPD-PCR provided a rapid and practical tool for identification of E. floccosum isolates, which was independent of morphological characteristics, and enhanced laboratory diagnosis of dermatophytosis.


Asunto(s)
Dermatomicosis/microbiología , Epidermophyton/clasificación , Tipificación Molecular/métodos , Técnica del ADN Polimorfo Amplificado Aleatorio , ADN de Hongos/genética , Dermatomicosis/epidemiología , Epidermophyton/genética , Humanos , Irán/epidemiología , Técnicas de Tipificación Micológica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...