Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunol Rev ; 298(1): 254-263, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33037700

RESUMEN

Unique Vγ2Vδ2 (Vγ9Vδ2) T cells existing only in human and non-human primates, account for the majority of circulating γδ T cells in human adults. Vγ2Vδ2 T cells are the sole γδ T-cell subpopulation capable of recognizing the microbial (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate (HMBPP) produced by selected pathogens during infections. Recent seminal studies in non-human primate models have demonstrated that the unique HMBPP-specific Vγ2Vδ2 T cells are fast-acting, multi-functional, and protective during infections. This article reviews the recent seminal observations of Vγ2Vδ2 T cells in protective mechanisms against tuberculosis and other infections.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Animales , Receptores de Antígenos de Linfocitos T gamma-delta , Linfocitos T
2.
Proc Natl Acad Sci U S A ; 116(13): 6371-6378, 2019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30850538

RESUMEN

Tuberculosis (TB) remains a leading killer among infectious diseases, and a better TB vaccine is urgently needed. The critical components and mechanisms of vaccine-induced protection against Mycobacterium tuberculosis (Mtb) remain incompletely defined. Our previous studies demonstrate that Vγ2Vδ2 T cells specific for (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate (HMBPP) phosphoantigen are unique in primates as multifunctional effectors of immune protection against TB infection. Here, we selectively immunized Vγ2Vδ2 T cells and assessed the effect on infection in a rhesus TB model. A single respiratory vaccination of macaques with an HMBPP-producing attenuated Listeria monocytogenes (Lm ΔactA prfA*) caused prolonged expansion of HMBPP-specific Vγ2Vδ2 T cells in circulating and pulmonary compartments. This did not occur in animals similarly immunized with an Lm ΔgcpE strain, which did not produce HMBPP. Lm ΔactA prfA* vaccination elicited increases in Th1-like Vγ2Vδ2 T cells in the airway, and induced containment of TB infection after pulmonary challenge. The selective immunization of Vγ2Vδ2 T cells reduced lung pathology and mycobacterial dissemination to extrapulmonary organs. Vaccine effects coincided with the fast-acting memory-like response of Th1-like Vγ2Vδ2 T cells and tissue-resident Vγ2Vδ2 effector T cells that produced both IFN-γ and perforin and inhibited intracellular Mtb growth. Furthermore, selective immunization of Vγ2Vδ2 T cells enabled CD4+ and CD8+ T cells to mount earlier pulmonary Th1 responses to TB challenge. Our findings show that selective immunization of Vγ2Vδ2 T cells can elicit fast-acting and durable memory-like responses that amplify responses of other T cell subsets, and provide an approach to creating more effective TB vaccines.


Asunto(s)
Inmunización , Activación de Linfocitos/efectos de los fármacos , Vacunas contra la Tuberculosis/inmunología , Tuberculosis/inmunología , Tuberculosis/prevención & control , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Femenino , Memoria Inmunológica/inmunología , Interferón gamma/metabolismo , Listeria monocytogenes/genética , Listeria monocytogenes/inmunología , Pulmón/inmunología , Pulmón/patología , Macaca mulatta/inmunología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Mycobacterium tuberculosis/efectos de los fármacos , Organofosfatos , Factores de Terminación de Péptidos/genética , Factores de Terminación de Péptidos/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Subgrupos de Linfocitos T/inmunología , Tuberculosis/patología , Vacunas contra la Tuberculosis/farmacología , Vacunas Atenuadas/inmunología
3.
J Immunol ; 200(7): 2405-2417, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29453279

RESUMEN

The ability of Mycobacterium tuberculosis to block host antimicrobial responses in infected cells provides a key mechanism for disease pathogenesis. The immune system has evolved to overcome this blockade to restrict the infection, but it is not clear whether two key innate cytokines (IL-12/IL-18) involved in host defense can enhance antimycobacterial mechanisms. In this study, we demonstrated that the combination of IL-12 and IL-18 triggered an antimicrobial response against mycobacteria in infected macrophages (THP-1 and human primary monocyte-derived macrophages) and pulmonary epithelial A549 cells. The inhibition of intracellular bacterial growth required p38-MAPK and STAT4 pathways, the vitamin D receptor, the vitamin D receptor-derived antimicrobial peptide cathelicidin, and autophagy, but not caspase-mediated apoptosis. Finally, the ability of IL-12+IL-18 to activate an innate antimicrobial response in human primary macrophages was dependent on the autonomous production of IFN-γ and the CAMP/autophagy pathway. Together, these data suggest that IL-12+IL-18 cosignaling can trigger the antimicrobial protein cathelicidin and autophagy, resulting in inhibition of intracellular mycobacteria in macrophages and lung epithelial cells.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/metabolismo , Inmunidad Innata/inmunología , Interferón gamma/inmunología , Subunidad p35 de la Interleucina-12/inmunología , Interleucina-18/inmunología , Macrófagos/inmunología , Mycobacterium tuberculosis/crecimiento & desarrollo , Tuberculosis/inmunología , Células A549 , Autofagia/inmunología , Línea Celular Tumoral , Células Epiteliales/inmunología , Humanos , Mycobacterium tuberculosis/inmunología , Receptores de Calcitriol/metabolismo , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología , Factor de Transcripción STAT4/metabolismo , Células THP-1 , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Catelicidinas
4.
J Immunol ; 200(3): 1016-1026, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29255077

RESUMEN

The nucleic acids of Mycobacterium tuberculosis can be detected by intracellular DNA sensors, such as cyclic GMP-AMP synthase and absent in melanoma 2 (AIM2), which results in the release of type I IFN and the proinflammatory cytokine IL-1ß. However, whether cross-talk occurs between AIM2-IL-1ß and cyclic GMP-AMP synthase-type I IFN signaling upon M. tuberculosis infection in vivo is unclear. In this article, we demonstrate that mycobacterial infection of AIM2-/- mice reciprocally induces overreactive IFN-ß and depressive IFN-γ responses, leading to higher infection burdens and more severe pathology. We also describe the underlying mechanism whereby activated apoptosis-associated speck-like protein interacts with a key adaptor, known as stimulator of IFN genes (STING), and inhibits the interaction between STING and downstream TANK-binding kinase 1 in bone marrow-derived macrophages and bone marrow-derived dendritic cells, consequently reducing the induction of type I IFN. Of note, apoptosis-associated speck-like protein expression is inversely correlated with IFN-ß levels in PBMCs from tuberculosis patients. These data demonstrate that the AIM2-IL-1ß signaling pathway negatively regulates the STING-type I IFN signaling pathway by impeding the association between STING and TANK-binding kinase 1, which protects the host from M. tuberculosis infection. This finding has potential clinical significance.


Asunto(s)
Proteínas de Unión al ADN/inmunología , Interferón beta/metabolismo , Interferón gamma/inmunología , Interleucina-1beta/inmunología , Proteínas de la Membrana/metabolismo , Mycobacterium bovis/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Tuberculosis/patología , Animales , Proteínas Adaptadoras de Señalización CARD/metabolismo , Línea Celular , Proteínas de Unión al ADN/genética , Células HEK293 , Humanos , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/inmunología , Células RAW 264.7 , Transducción de Señal/inmunología , Tuberculosis/microbiología
5.
Angew Chem Int Ed Engl ; 59(8): 3226-3234, 2020 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-31756258

RESUMEN

Pathogenesis hallmarks for tuberculosis (TB) are the Mycobacterium tuberculosis (Mtb) escape from phagolysosomal destruction and limited drug delivery into infected cells. Several nanomaterials can be entrapped in lysosomes, but the development of functional nanomaterials to promote phagolysosomal Mtb clearance remains a big challenge. Here, we report on the bactericidal effects of selenium nanoparticles (Se NPs) against Mtb and further introduce a novel nanomaterial-assisted anti-TB strategy manipulating Ison@Man-Se NPs for synergistic drug-induced and phagolysosomal destruction of Mtb. Ison@Man-Se NPs preferentially entered macrophages and accumulated in lysosomes releasing Isoniazid. Surprisingly, Ison@Man-Se/Man-Se NPs further promoted the fusion of Mtb into lysosomes for synergistic lysosomal and Isoniazid destruction of Mtb. Concurrently, Ison@Man-Se/Man-Se NPs also induced autophagy sequestration of Mtb, evolving into lysosome-associated autophagosomal Mtb degradation linked to ROS-mitochondrial and PI3K/Akt/mTOR signaling pathways. This novel nanomaterial-assisted anti-TB strategy manipulating antimicrobial immunity and Mtb clearance may potentially serve in more effective therapeutics against TB and drug-resistant TB.


Asunto(s)
Antibacterianos/uso terapéutico , Sistemas de Liberación de Medicamentos/métodos , Isoniazida/química , Macrófagos/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Nanopartículas/química , Selenio/química , Tuberculosis/tratamiento farmacológico , Humanos , Tuberculosis/patología
6.
J Immunol ; 198(12): 4753-4763, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28526681

RESUMEN

The dominant Vγ2Vδ2 T cell subset recognizes phosphoantigen and exists only in humans and nonhuman primates. Despite the discovery of γδ T cells >30 y ago, a proof-of-concept study has not been done to prove the principle that the Vγ2Vδ2 T cell subset is protective against Mycobacterium tuberculosis and other infections. In this study, we used an adoptive cell-transfer strategy to define the protective role of Vγ2Vδ2 T cells in a primate tuberculosis (TB) model. Vγ2Vδ2 T cells for adoptive transfer displayed central/effector memory and mounted effector functions, including the production of anti-M. tuberculosis cytokines and inhibition of intracellular mycobacteria. They also expressed CXCR3/CCR5/LFA-1 trafficking/tissue-resident phenotypes and consistently trafficked to the airway, where they remained detectable from 6 h through 7 d after adoptive transfer. Interestingly, the test group of macaques receiving transfer of Vγ2Vδ2 T cells at weeks 1 and 3 after high-dose (500 CFU) M. tuberculosis infection exhibited significantly lower levels of M. tuberculosis infection burdens in lung lobes and extrapulmonary organs than did the control groups receiving PBLs or saline. Consistently, adoptive transfer of Vγ2Vδ2 T cells attenuated TB pathology and contained lesions primarily in the infection site of the right caudal lung lobe, with no or reduced TB dissemination to other lobes, spleen, or liver/kidney; in contrast, the controls showed widespread TB dissemination. The proof-of-concept finding supports the view that the dominant Vγ2Vδ2 T cell subset may be included in the rational design of a TB vaccine or host-directed therapy.


Asunto(s)
Traslado Adoptivo , Mycobacterium tuberculosis/inmunología , Fosfoproteínas/uso terapéutico , Receptores de Antígenos de Linfocitos T gamma-delta/uso terapéutico , Subgrupos de Linfocitos T/inmunología , Tuberculosis/inmunología , Tuberculosis/terapia , Animales , Carga Bacteriana , Citocinas/biosíntesis , Citocinas/inmunología , Memoria Inmunológica , Pulmón/inmunología , Pulmón/microbiología , Antígeno-1 Asociado a Función de Linfocito/genética , Antígeno-1 Asociado a Función de Linfocito/inmunología , Macaca fascicularis , Fosfoproteínas/administración & dosificación , Fosfoproteínas/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Receptores CCR5/genética , Receptores CCR5/inmunología , Receptores CXCR3/genética , Receptores CXCR3/inmunología , Tuberculosis/microbiología
7.
Clin Immunol ; 193: 1-9, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29753126

RESUMEN

The PPV23 immunizes healthy elderly and other high-risk populations against pneumococcal disease. Immune mechanisms whereby these populations differently mount antibody(Ab) and cellular responses to PPV23 vaccination remain unknown. Here, healthy elderly, those elderly with prior tuberculosis-cured history (TB-cured), and HIV-infected humans were vaccinated with PPV23, and assessed for opsonophagocytic Ab responses and potential cellular mechanisms. PPV23 vaccination elicited hierarchical responses of opsonophagocytic Ab. PPV23-elicited Ab titers were highest in healthy elderly, significantly lower in TB-cured elderly and lowest in HIV-infected subjects. Mechanistically, high PPV23-elicited Ab titers in healthy elderly were associated with increases in CD19 + CD69+ cells and CD19 + CD138 + plasma cells. Surprisingly, TB-cured elderly failed to show PPV23-induced increases in these cells. While HIV-infected subjects showed a depressed CD19 + CD69+ cellular response, PPV23 vaccination uncovered HIV-related over-reactive increases in CD19 + CD138 + cells. For the first time, we demonstrate that PPV23-elicted opsonophagocytic Ab titers correlate with different cellular responses in healthy, TB-cured and HIV statuses.


Asunto(s)
Infecciones por VIH/inmunología , VIH-1/fisiología , Células Plasmáticas/inmunología , Infecciones Neumocócicas/inmunología , Vacunas Neumococicas/inmunología , Tuberculosis/inmunología , Adulto , Anciano , Formación de Anticuerpos , Antígenos CD/metabolismo , Antígenos CD19/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Femenino , Voluntarios Sanos , Humanos , Inmunidad Celular , Lectinas Tipo C/metabolismo , Masculino , Persona de Mediana Edad , Fagocitosis , Sindecano-1 , Vacunación
8.
J Virol ; 91(14)2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28356537

RESUMEN

Despite a great deal of prior research, the early pathogenic events in natural oral poliovirus infection remain poorly defined. To establish a model for study, we infected 39 macaques by feeding them single high doses of the virulent Mahoney strain of wild type 1 poliovirus. Doses ranging from 107 to 109 50% tissue culture infective doses (TCID50) consistently infected all the animals, and many monkeys receiving 108 or 109 TCID50 developed paralysis. There was no apparent difference in the susceptibilities of the three macaque species (rhesus, cynomolgus, and bonnet) used. Virus excretion in stool and nasopharynges was consistently observed, with occasional viremia, and virus was isolated from tonsils, gut mucosa, and draining lymph nodes. Viral replication proteins were detected in both epithelial and lymphoid cell populations expressing CD155 in the tonsil and intestine, as well as in spinal cord neurons. Necrosis was observed in these three cell types, and viral replication in the tonsil/gut was associated with histopathologic destruction and inflammation. The sustained response of neutralizing antibody correlated temporally with resolution of viremia and termination of virus shedding in oropharynges and feces. For the first time, this model demonstrates that early in the infectious process, poliovirus replication occurs in both epithelial cells (explaining virus shedding in the gastrointestinal tract) and lymphoid/monocytic cells in tonsils and Peyer's patches (explaining viremia), extending previous studies of poliovirus pathogenesis in humans. Because the model recapitulates human poliovirus infection and poliomyelitis, it can be used to study polio pathogenesis and to assess the efficacy of candidate antiviral drugs and new vaccines.IMPORTANCE Early pathogenic events of poliovirus infection remain largely undefined, and there is a lack of animal models mimicking natural oral human infection leading to paralytic poliomyelitis. All 39 macaques fed with single high doses ranging from 107 to 109 TCID50 Mahoney type 1 virus were infected, and many of the monkeys developed paralysis. Virus excretion in stool and nasopharynges was consistently observed, with occasional viremia; tonsil, mesentery lymph nodes, and intestinal mucosa served as major target sites of viral replication. For the first time, this model demonstrates that early in the infectious process, poliovirus replication occurs in both epithelial cells (explaining virus shedding in the gastrointestinal tract) and lymphoid/monocytic cells in tonsils and Peyer's patches (explaining viremia), thereby supplementing historical reconstructions of poliovirus pathogenesis. Because the model recapitulates human poliovirus infection and poliomyelitis, it can be used to study polio pathogenesis, candidate antiviral drugs, and the efficacy of new vaccines.


Asunto(s)
Macaca , Poliomielitis/patología , Poliovirus/crecimiento & desarrollo , Poliovirus/patogenicidad , Estructuras Animales/virología , Animales , Modelos Animales de Enfermedad , Células Epiteliales/virología , Heces/virología , Leucocitos/virología , Nasofaringe/virología , Esparcimiento de Virus
9.
Proc Natl Acad Sci U S A ; 112(29): E3883-92, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-26150504

RESUMEN

Molecular mechanisms for T-cell immune responses modulated by T cell-inhibitory molecules during tuberculosis (TB) infection remain unclear. Here, we show that active human TB infection up-regulates CD244 and CD244 signaling-associated molecules in CD8(+) T cells and that blockade of CD244 signaling enhances production of IFN-γ and TNF-α. CD244 expression/signaling in TB correlates with high levels of a long noncoding RNA (lncRNA)-BC050410 [named as lncRNA-AS-GSTT1(1-72) or lncRNA-CD244] in the CD244(+)CD8(+) T-cell subpopulation. CD244 signaling drives lncRNA-CD244 expression via sustaining a permissive chromatin state in the lncRNA-CD244 locus. By recruiting polycomb protein enhancer of zeste homolog 2 (EZH2) to infg/tnfa promoters, lncRNA-CD244 mediates H3K27 trimethylation at infg/tnfa loci toward repressive chromatin states and inhibits IFN-γ/TNF-α expression in CD8(+) T cells. Such inhibition can be reversed by knock down of lncRNA-CD244. Interestingly, adoptive transfer of lncRNA-CD244-depressed CD8(+) T cells to Mycobacterium tuberculosis (MTB)-infected mice reduced MTB infection and TB pathology compared with lncRNA-CD244-expressed controls. Thus, this work uncovers previously unidentified mechanisms in which T cell-inhibitory signaling and lncRNAs regulate T-cell responses and host defense against TB infection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epigénesis Genética , Inmunidad , ARN Largo no Codificante/genética , Transducción de Señal , Tuberculosis/inmunología , Traslado Adoptivo , Animales , Anticuerpos Monoclonales/farmacología , Antígenos CD , Apoptosis/efectos de los fármacos , Apoptosis/genética , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/efectos de los fármacos , Cromatina/metabolismo , Biología Computacional , Secuencia Conservada , Proteína Potenciadora del Homólogo Zeste 2 , Epigénesis Genética/efectos de los fármacos , Evolución Molecular , Técnicas de Silenciamiento del Gen , Genoma Humano , Células HEK293 , Humanos , Inmunidad/efectos de los fármacos , Inmunidad/genética , Interferón gamma/biosíntesis , Interferón gamma/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Modelos Biológicos , Complejo Represivo Polycomb 2/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Receptores Inmunológicos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria , Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Factores de Transcripción/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética , Regulación hacia Arriba/efectos de los fármacos
10.
J Infect Dis ; 215(3): 420-430, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27789724

RESUMEN

A loss of antigen-specific T-cell responses due to defective cytokine signaling during infections has not been reported. We hypothesize that tuberculosis can destroy signaling effects of selective cytokine(s) and induce exhaustion of antigen-specific T cells. To test this hypothesis, mechanistic studies were performed to examine whether and how tuberculosis blocked interleukin 23 (IL-23) and interleukin 2 (IL-2) signaling effects on a major human γδ T-cell subpopulation, phosphoantigen HMBPP-specific Vγ2Vδ2 T cells. IL-23 and IL-2 significantly expanded HMBPP-stimulated Vγ2Vδ2 T cells from subjects with latent tuberculosis infection, and IL-2 synergized the effect of IL-23. IL-23-induced expansion of Vγ2Vδ2 T cells involved STAT3. Surprisingly, patients with tuberculosis exhibited a selective destruction of IL-23-induced expansion of these cells. The tuberculosis-driven destruction of IL-23 signaling coincided with decreases of expression and phosphorylation of STAT3. Interestingly, impairing of STAT3 was linked to marked increases in the microRNAs (miRNAs) hsa-miR-337-3p and hsa-miR-125b-5p in Vγ2Vδ2 T cells from patients with tuberculosis. Downregulation of hsa-miR-337-3p and hsa-miR-125b-5p by miRNA sponges improved IL-23-mediated expansion of Vγ2Vδ2 T cells and restored the ability of these cells to produce anti-tuberculosis cytokines. These results support our hypothesis that tuberculosis can selectively impair a cytokine effect while sparing another and can induce exhaustion of T cells in response to the respective cytokine.


Asunto(s)
Interleucina-23/antagonistas & inhibidores , Tuberculosis Latente/inmunología , Mycobacterium tuberculosis/inmunología , Subgrupos de Linfocitos T/inmunología , Adulto , Proliferación Celular , Femenino , Humanos , Interleucina-2/antagonistas & inhibidores , Interleucina-2/metabolismo , Interleucina-23/inmunología , Masculino , Organofosfatos/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
11.
Cell Immunol ; 311: 28-35, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27717503

RESUMEN

Despite past extensive studies on B and T lymphocyte attenuator (BTLA)-mediated negative regulation of T cell activation, the role of BTLA in antigen presenting cells (APCs) in patients with active pulmonary tuberculosis (ATB) remains poorly understood. Here, we demonstrate that BTLA expression on CD11c APCs increased in patients with ATB. Particularly, BTLA expression in CD11c APCs was likely associated with the attenuated stimulatory capacity on T cells (especially CD8+ T cell) proliferation. BTLA-expressing CD11c APCs showed lower antigen uptake capacity, lower CD86 expression, higher HLA-DR expression, and enhanced IL-6 secretion, compared to counterpart BTLA negative CD11c APCs in healthy controls (HC). Interestingly, BTLA-expressing CD11c APCs from ATB patients displayed lower expression of HLA-DR and less IL-6 secretion, but higher expression of CD86 than those from HC volunteers. Mixed lymphocyte reaction suggests that BTLA expression is likely associated with positive rather than conventional negative regulation of CD11c APCs stimulatory capacity. This role is impaired in ATB patients manifested by low expression of HLA-DR and low production of IL-6. This previous unappreciated role for BTLA may have implications in the prevention and treatment of patients with ATB.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Linfocitos T/inmunología , Tuberculosis Pulmonar/inmunología , Adolescente , Adulto , Antígeno B7-2/metabolismo , Antígeno CD11c/metabolismo , Proliferación Celular , Células Cultivadas , Femenino , Antígenos HLA-DR/metabolismo , Humanos , Interleucina-6/metabolismo , Activación de Linfocitos , Masculino , Persona de Mediana Edad , Receptores Inmunológicos/metabolismo , Linfocitos T/microbiología , Adulto Joven
12.
Pharmacol Res ; 119: 479-489, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28411855

RESUMEN

As the active anticancer component of Rabdosia Rubescens, oridonin has been proved to show strong anticancer activity in cancer cells, which is also found to be closely related to its specific inhibition effects on the EGFR tyrosine kinase activity. In this study, atomic force microscopy based single molecule force spectroscopy (AFM-SMFS) was used for real-time and in-situ detection of EGF-EGFR interactions in living esophageal cancer KYSE-150 cells to evaluate the anticancer activity of oridonin for the first time. Oridonin was found to induce apoptosis and also reduce EGFR expression in KYSE-150 cells. AFM-SMFS results demonstrated that oridonin could inhibit the binding between EGF and EGFR in KYSE-150 cells by decreasing the unbinding force and binding probability for EGF-EGFR complexes, which was further proved to be closely associated with the intracellular ROS level. More precise mechanism studies based on AFM-SMFS demonstrated that oridonin treatment could decrease the energy barrier width, increase the dissociation off rate constant and decrease the activation energy of EGF-EGFR complexes in ROS dependent way, suggesting oridonin as a strong anticancer agent targeting EGF-EGFR interactions in cancer cells through ROS dependent mechanism. Our results not only suggested oridonin as a strong anticancer agent targeting EGF-EGFR interactions in ROS dependent mechanism, but also highlighted AFM-SMFS as a powerful technique for pharmacodynamic studies by detecting ligand-receptor interactions, which was also expected to be developed into a promising tool for the screening and mechanism studies of drugs.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Diterpenos de Tipo Kaurano/farmacología , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Neoplasias Esofágicas/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , Antineoplásicos Fitogénicos/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Diterpenos de Tipo Kaurano/química , Neoplasias Esofágicas/metabolismo , Esófago/efectos de los fármacos , Esófago/metabolismo , Humanos , Isodon/química , Microscopía de Fuerza Atómica
14.
Biochim Biophys Acta ; 1848(3): 859-68, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25534714

RESUMEN

The migration of mesenchymal stem cells (MSCs) plays a key role in tumor-targeted delivery vehicles and tumor-related stroma formation. However, there so far has been no report on the distribution of cell surface molecules during the VEGF-induced migration of MSCs. Here, we have utilized near-field scanning optical microscopy (NSOM) combined with fluorescent quantum dot (QD)-based nano-technology to capture the functional relationship between CD44 and CD29 adhesion molecules on MSCs and the effect of their spatial rearrangements. Before VEGF-induced migration of MSCs, both CD44 and CD29 formed 200-220 nm nano-domains respectively, with little co-localization between the two types of domains. Surprisingly, the size of the CD44 nano-domain rapidly increased in size to 295 nm and apparently larger aggregates were formed following MSC treatment with VEGF for 10 min, while the area of co-localization increased to 0.327 µm2. Compared with CD44, CD29 was activated obviously later, for the fact that CD29 aggregation didn't appear until 30 min after VEGF treatment. Consistently, its co-localization area increased to 0.917 µm2. The CD44 and CD29 nano-domains further aggregated into larger nano-domains or even formed micro-domains on the membrane of activated MSCs. The aggregation and co-localization of these molecules promoted FAK formation and cytoskeleton rearrangement. All of the above changes induced by VEGF contributed to MSC migration. Taken together, our data of NSOM-based dual color fluorescent imaging demonstrated for the first time that CD44, together with CD29, involved in VEGF-induced migration of MSCs through the interaction between CD44 and its co-receptor of VEGFR-2.


Asunto(s)
Movimiento Celular , Células Madre Mesenquimatosas/citología , Microscopía de Sonda de Barrido/métodos , Puntos Cuánticos , Células Cultivadas , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Fluorescencia , Adhesiones Focales/efectos de los fármacos , Humanos , Receptores de Hialuranos/metabolismo , Integrina beta1/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Microscopía de Fuerza Atómica , Microscopía Confocal , Microscopía Fluorescente , Modelos Biológicos , Unión Proteica/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
15.
Eur J Immunol ; 45(2): 442-51, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25141829

RESUMEN

Whether cytokines can influence the adaptive immune response by antigen-specific γδ T cells during infections or vaccinations remains unknown. We previously demonstrated that, during BCG/Mycobacterium tuberculosis (Mtb) infections, Th17-related cytokines markedly upregulated when phosphoantigen-specific Vγ2Vδ2 T cells expanded. In this study, we examined the involvement of Th17-related cytokines in the recall-like responses of Vγ2Vδ2 T cells following Mtb infection or vaccination against TB. Treatment with IL-17A/IL-17F or IL-22 expanded phosphoantigen 4-hydroxy-3-methyl-but-enyl pyrophosphate (HMBPP)-stimulated Vγ2Vδ2 T cells from BCG-vaccinated macaques but not from naïve animals, and IL-23 induced greater expansion than the other Th17-related cytokines. Consistently, Mtb infection of macaques also enhanced the ability of IL-17/IL-22 or IL-23 to expand HMBPP-stimulated Vγ2Vδ2 T cells. When evaluating IL-23 signaling as a prototype, we found that HMBPP/IL-23-expanded Vγ2Vδ2 T cells from macaques infected with Mtb or vaccinated with BCG or Listeria ΔactA prfA*-ESAT6/Ag85B produced IL-17, IL-22, IL-2, and IFN-γ. Interestingly, HMBPP/IL-23-induced production of IFN-γ in turn facilitated IL-23-induced expansion of HMBPP-activated Vγ2Vδ2 T cells. Furthermore, HMBPP/IL-23-induced proliferation of Vγ2Vδ2 T cells appeared to require APC contact and involve the conventional and novel protein kinase C signaling pathways. These findings suggest that Th17-related cytokines can contribute to recall-like expansion and effector function of Ag-specific γδ T cells after infection or vaccination.


Asunto(s)
Interleucina-17/inmunología , Mycobacterium tuberculosis/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Células Th17/inmunología , Tuberculosis/prevención & control , Animales , Vacunas Bacterianas/administración & dosificación , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Expresión Génica , Interferón gamma/biosíntesis , Interleucina-17/genética , Interleucina-17/farmacología , Interleucina-2/biosíntesis , Interleucina-23/farmacología , Interleucinas/farmacología , Listeria/inmunología , Macaca fascicularis , Mycobacterium bovis/inmunología , Organofosfatos/inmunología , Organofosfatos/farmacología , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Proteínas Recombinantes/farmacología , Células Th17/citología , Células Th17/efectos de los fármacos , Tuberculosis/inmunología , Tuberculosis/microbiología , Vacunación , Interleucina-22
16.
J Virol ; 89(19): 9932-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26202243

RESUMEN

UNLABELLED: Filoviruses, consisting of Ebola virus (EBOV) and Marburg virus (MARV), are among the most lethal infectious threats to mankind. Infections by these viruses can cause severe hemorrhagic fevers in humans and nonhuman primates with high mortality rates. Since there is currently no vaccine or antiviral therapy approved for humans, there is an urgent need to develop prophylactic and therapeutic options for use during filoviral outbreaks and bioterrorist attacks. One of the ideal targets against filoviral infection and diseases is at the entry step, which is mediated by the filoviral glycoprotein (GP). In this report, we screened a chemical library of small molecules and identified numerous inhibitors, which are known G protein-coupled receptor (GPCR) antagonists targeting different GPCRs, including histamine receptors, 5-HT (serotonin) receptors, muscarinic acetylcholine receptor, and adrenergic receptor. These inhibitors can effectively block replication of both infectious EBOV and MARV, indicating a broad antiviral activity of the GPCR antagonists. The time-of-addition experiment and microscopic studies suggest that GPCR antagonists block filoviral entry at a step following the initial attachment but prior to viral/cell membrane fusion. These results strongly suggest that GPCRs play a critical role in filoviral entry and GPCR antagonists can be developed as an effective anti-EBOV/MARV therapy. IMPORTANCE: Infection of Ebola virus and Marburg virus can cause severe illness in humans with a high mortality rate, and currently there is no FDA-approved vaccine or therapeutic treatment available. The 2013-2015 epidemic in West Africa underscores a lack of our understanding in the infection and pathogenesis of these viruses and the urgency of drug discovery and development. In this study, we have identified numerous inhibitors that are known G protein-coupled receptor (GPCR) antagonists targeting different GPCRs. These inhibitors can effectively block replication of both infectious EBOV and MARV, indicating a broad antiviral activity of the GPCR antagonists. Our results strongly suggest that GPCRs play a critical role in filoviral entry and GPCR antagonists can be developed as an effective anti-EBOV/MARV therapy.


Asunto(s)
Ebolavirus/efectos de los fármacos , Ebolavirus/fisiología , Marburgvirus/efectos de los fármacos , Marburgvirus/fisiología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Internalización del Virus/efectos de los fármacos , Animales , Antivirales/farmacología , Benzotropina/farmacología , Línea Celular , Chlorocebus aethiops , Ciproheptadina/farmacología , Ebolavirus/patogenicidad , Células HEK293 , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Heparina/farmacología , Ensayos Analíticos de Alto Rendimiento , Humanos , Macrólidos/farmacología , Enfermedad del Virus de Marburg/tratamiento farmacológico , Marburgvirus/patogenicidad , Receptores Acoplados a Proteínas G/fisiología , Bibliotecas de Moléculas Pequeñas , Células Vero , Zidovudina/farmacología
17.
Microb Pathog ; 93: 1-7, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26792673

RESUMEN

Since BCG, the only vaccine widely used against tuberculosis (TB) in the world, provides varied protective efficacy and may not be effective for inducing long-term cellular immunity, it is in an urgent need to develop more effective vaccines and more potent immune strategies against TB. Prime-boost is proven to be a good strategy by inducing long-term protection. In this study, we tested the protective effect against Mycobacterium tuberculosis (Mtb) challenge of prime-boost strategy by recombinant BCG (rBCG) expressing PPE protein Rv3425 fused with Ag85B and Rv3425. Results showed that the prime-boost strategy could significantly increase the protective efficiency against Mtb infection, characterized by reduction of bacterial load in lung and spleen, attenuation of tuberculosis lesions in lung tissues. Importantly, we found that Rv3425 boost, superior to Ag85B boost, provided better protection against Mtb infection. Further research proved that rBCG prime-Rv3425 boost could obviously increase the expansion of lymphocytes, significantly induce IL-2 production by lymphocytes upon PPD stimulation, and inhibit IL-6 production at an early stage. It implied that rBCG prime-Rv3425 boost opted to induce Th1 immune response and provided a long-term protection against TB. These results implicated that rBCG prime-Rv3425 boost is a potent and promising strategy to prevent acute Mtb infection.


Asunto(s)
Vacuna BCG/administración & dosificación , Mycobacterium tuberculosis/inmunología , Tuberculosis/prevención & control , Animales , Antígenos Bacterianos/administración & dosificación , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Vacuna BCG/genética , Vacuna BCG/inmunología , Proteínas Bacterianas/administración & dosificación , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Femenino , Humanos , Inmunización Secundaria , Ratones , Ratones Endogámicos C57BL , Mycobacterium bovis/inmunología , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/fisiología , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Tuberculosis/inmunología , Tuberculosis/microbiología
19.
J Immunol ; 192(5): 2120-32, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24489088

RESUMEN

The possibility that CD4(+) T cells can act as "innate-like" cells to contain very early Mycobacterium tuberculosis dissemination and function as master helpers to sustain multiple effector functions of CD8(+) T cells and CD3(-) lymphocytes during development of adaptive immunity against primary tuberculosis (TB) has not been demonstrated. We showed that pulmonary M. tuberculosis infection of CD4-depleted macaques surprisingly led to very early extrapulmonary M. tuberculosis dissemination, whereas CD4 deficiency clearly resulted in rapid TB progression. CD4 depletion during M. tuberculosis infection revealed the ability of CD8(+) T cells to compensate and rapidly differentiate to Th17-like/Th1-like and cytotoxic-like effectors, but these effector functions were subsequently unsustainable due to CD4 deficiency. Whereas CD3(-) non-T lymphocytes in the presence of CD4(+) T cells developed predominant Th22-like and NK-like (perforin production) responses to M. tuberculosis infection, CD4 depletion abrogated these Th22-/NK-like effector functions and favored IL-17 production by CD3(-) lymphocytes. CD4-depleted macaques exhibited no or few pulmonary T effector cells constitutively producing IFN-γ, TNF-α, IL-17, IL-22, and perforin at the endpoint of more severe TB, but they presented pulmonary IL-4(+) T effectors. TB granulomas in CD4-depleted macaques contained fewer IL-22(+) and perforin(+) cells despite the presence of IL-17(+) and IL-4(+) cells. These results implicate a previously unknown innate-like ability of CD4(+) T cells to contain extrapulmonary M. tuberculosis dissemination at very early stage. Data also suggest that CD4(+) T cells are required to sustain multiple effector functions of CD8(+) T cells and CD3(-) lymphocytes and to prevent rapid TB progression during M. tuberculosis infection of nonhuman primates.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Citocinas/inmunología , Mycobacterium tuberculosis/inmunología , Células TH1/inmunología , Células Th17/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Linfocitos T CD8-positivos/microbiología , Linfocitos T CD8-positivos/patología , Macaca fascicularis , Células TH1/microbiología , Células TH1/patología , Células Th17/microbiología , Células Th17/patología , Tuberculosis Pulmonar/microbiología , Tuberculosis Pulmonar/patología
20.
Appl Microbiol Biotechnol ; 100(15): 6643-6652, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26883344

RESUMEN

Angiogenesis provides necessary nutrients and oxygen for tumor growth and metastasis; thus, every stage of angiogenesis process is the potential target for cancer therapies. Ursolic acid (UA) is reported to decrease tumor burden through anti-angiogenesis pathway, but its poor water solubility greatly limits its efficiency and clinical application. Here, a simple method for preparing UA-loaded chitosan nanoparticles (CH-UA-NPs) with anti-angiogenesis and anti-tumor activity was demonstrated. In vitro, CH-UA-NPs could significantly inhibit the proliferation, migration, and tube formation of human umbilical vascular endothelial cells (HUVECs). After uptake by HUVECs, CH-UA-NPs were mainly localized in lysosomes and mitochondria, but not nuclei. CH-UA-NPs induced the destruction of lysosome membrane integrity, collapse of mitochondrial membrane potential, and reorganization of cell cytoskeleton. All these changes led to the apoptosis or necrosis in HUVECs. In vivo, CH-UA-NPs could inhibit the angiogenesis in chicken chorioallantoic membrane (CAM) model and H22 xenograft model. Notably, comparing with free UA, such synthesized CH-UA-NPs could save about tenfold of UA doses, implying that this could significantly decrease the side effects induced by high doses of UA in biological organism. Our data showed that CH-UA-NPs and this nanoparticle-based drug delivery system could be as a potential drug candidate for anti-angiogenesis treatment.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Quitosano/química , Membrana Corioalantoides/irrigación sanguínea , Portadores de Fármacos/química , Nanopartículas/química , Neovascularización Patológica/prevención & control , Triterpenos/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Embrión de Pollo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Mitocondrias/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Triterpenos/química , Cicatrización de Heridas/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Ácido Ursólico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA