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












Base de datos
Intervalo de año de publicación
1.
PLoS Pathog ; 19(10): e1011731, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37871034

RESUMEN

Cholesterol derived from the host milieu forms a critical factor for mycobacterial pathogenesis. However, the molecular circuitry co-opted by Mycobacterium tuberculosis (Mtb) to accumulate cholesterol in host cells remains obscure. Here, we report that the coordinated action of WNT-responsive histone modifiers G9a (H3K9 methyltransferase) and SIRT6 (H3K9 deacetylase) orchestrate cholesterol build-up in in vitro and in vivo mouse models of Mtb infection. Mechanistically, G9a, along with SREBP2, drives the expression of cholesterol biosynthesis and uptake genes; while SIRT6 along with G9a represses the genes involved in cholesterol efflux. The accumulated cholesterol in Mtb infected macrophages promotes the expression of antioxidant genes leading to reduced oxidative stress, thereby supporting Mtb survival. In corroboration, loss-of-function of G9a in vitro and pharmacological inhibition in vivo; or utilization of BMDMs derived from Sirt6-/- mice or in vivo infection in haplo-insufficient Sirt6-/+ mice; hampered host cholesterol accumulation and restricted Mtb burden. These findings shed light on the novel roles of G9a and SIRT6 during Mtb infection and highlight the previously unknown contribution of host cholesterol in potentiating anti-oxidative responses for aiding Mtb survival.


Asunto(s)
N-Metiltransferasa de Histona-Lisina , Mycobacterium tuberculosis , Sirtuinas , Animales , Ratones , Colesterol/metabolismo , Histonas/metabolismo , Macrófagos/metabolismo , Mycobacterium tuberculosis/metabolismo , Sirtuinas/genética , Sirtuinas/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo
3.
Mucosal Immunol ; 15(5): 882-895, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35778600

RESUMEN

The chemokine receptor CCR9 equips T cells with the ability to respond to CCL25, a chemokine that is highly expressed in the thymus and the small intestine (SI). Notably, CCR9 is mostly expressed on CD8 but not on CD4 lineage T cells, thus imposing distinct tissue tropism on CD4 and CD8 T cells. The molecular basis and the consequences for such a dichotomy, however, have not been fully examined and explained. Here, we demonstrate that the forced expression of CCR9 interferes with the tissue trafficking and differentiation of CD4 T cells in SI intraepithelial tissues. While CCR9 overexpression did not alter CD4 T cell generation in the thymus, the forced expression of CCR9 was detrimental for the proper tissue distribution of CD4 T cells in the periphery, and strikingly also for their terminal differentiation in the gut epithelium. Specifically, the differentiation of SI epithelial CD4 T cells into immunoregulatory CD4+CD8αα+ T cells was impaired by overexpression of CCR9 and conversely increased by the genetic deletion of CCR9. Collectively, our results reveal a previously unappreciated role for CCR9 in the tissue homeostasis and effector function of CD4 T cells in the gut.


Asunto(s)
Linfocitos Intraepiteliales , Receptores CCR , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Intestinos , Linfocitos Intraepiteliales/metabolismo , Receptores CCR/genética , Receptores CCR/metabolismo
5.
STAR Protoc ; 3(1): 101207, 2022 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-35243383

RESUMEN

Lymphocytes in the mouse small intestine (SI) epithelium are critical to establish effective barrier immunity. Here, we describe a magnetic cell separation protocol that employs anti-epithelial cellular adhesion molecule (EpCAM) antibodies to deplete epithelial cells and to enrich for intraepithelial lymphocytes (IELs) from SI tissues. The resulting IEL preparation was functionally and phenotypically comparable to IELs isolated using conventional density gradient centrifugation protocols. Moreover, the yield and purity of anti-EpCAM-depleted SI IELs were higher than those enriched by conventional isolation. For complete details on the use and execution of this protocol, please refer to Prakhar et al. (2021).


Asunto(s)
Linfocitos Intraepiteliales , Animales , Separación Celular/métodos , Molécula de Adhesión Celular Epitelial , Citometría de Flujo/métodos , Intestino Delgado , Ratones
6.
Autophagy ; 18(2): 391-408, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34074211

RESUMEN

Mycobacterium tuberculosis (Mtb)-driven lipid accumulation is intricately associated with the progression of tuberculosis (TB) disease. Although several studies elucidating the mechanisms for lipid droplet (LD) biosynthesis exist, we provide evidence for the significance of their regulated turnover via macroautophagy/autophagy during Mtb infection. We demonstrate that Mtb utilizes EGFR (epidermal growth factor receptor) signaling to induce the expression of the histone acetylation reader, BRD4 (bromodomain containing 4). The EGFR-BRD4 axis suppresses lipid-specific autophagy, and hence favors cellular lipid accumulation. Specifically, we found that pharmacological inhibition or knockdown of Egfr or Brd4 enhances autophagic flux and concomitantly decreases cellular LDs that is otherwise maintained at a significant level in chloroquine-treated or Atg5 knocked down autophagy-compromised host cells. In line with the enhanced lipophagy, we found that loss of EGFR or BRD4 function restricts mycobacterial burden that is rescued by external replenishment with oleic acid. We also report that the EGFR-BRD4 axis exerts additional effects by modulating pro-angiogenic gene expression and consequently aberrant angiogenesis during mycobacterial infection. This is important in the context of systemic Mtb dissemination as well as for the efficient delivery of anti-mycobacterial therapeutics to the Mtb-rich core of TB granuloma. Finally, utilizing an in vivo mouse model of TB, we show that pharmacological inhibition of EGFR and BRD4 compromises LD buildup via enhanced lipophagy and normalizes angiogenesis, thereby restricting Mtb burden and rescuing mice from severe TB-like pathology. These findings shed light on the novel roles of BRD4 during Mtb infection, and its possible implication in potentiating anti-TB responses.Abbreviations: ATG5: autophagy related 5; BRDs: bromodomain containing; COL18A1: collagen type XVIII alpha 1 chain; EGFR: epidermal growth factor receptor; EP300: E1A binding protein p300; KDR: kinase insert domain receptor; KLF5: Kruppel like factor 5; LDs: lipid droplets; MAP1LC3B: microtubule associated protein 1 light chain 3 beta; Mtb: Mycobacterium tuberculosis; PECAM1: platelet and endothelial cell adhesion molecule 1; SQSTM1/p62: sequestosome 1; TB: tuberculosis; THBS1: thrombospondin 1; VEGF: vascular endothelial growth factor.


Asunto(s)
Mycobacterium tuberculosis , Tuberculosis , Animales , Autofagia/fisiología , Epigénesis Genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Lípidos/farmacología , Ratones , Mycobacterium tuberculosis/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Tuberculosis/microbiología , Factor A de Crecimiento Endotelial Vascular/metabolismo
7.
FEBS J ; 289(6): 1536-1551, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34670010

RESUMEN

Active tuberculosis patients are at high risk of coinfection with opportunistic fungal pathogen Candida albicans. However, the molecular mechanisms that orchestrate pathogenesis of Mycobacterium tuberculosis (Mtb)-C. albicans coinfection remain elusive. In the current study, we utilize a mouse model to demonstrate that Mtb promotes a macrophage environment that is conducive for C. albicans survival. Mtb-dependent protein kinase Cζ-WNT signalling axis induces expression of an E3 ubiquitin ligase, constitutive photomorphogenesis protein 1 (COP1). A secondary infection of C. albicans in such Mtb-infected macrophages causes COP1 to mediate the proteasomal degradation of interferon regulatory factor 9 (IRF9), a cardinal factor that we identified to arbitrate an inflammatory programmed cell death, pyroptosis. In vivo experiments mimicking a pre-existing Mtb infection demonstrate that inhibition of pyroptosis in mice results in increased C. albicans burden and aberrant lung tissue architecture, leading to increased host mortality. Together, our study reveals the crucial role of pyroptosis regulation for manifesting a successful C. albicans-Mtb coinfection.


Asunto(s)
Coinfección , Mycobacterium tuberculosis , Animales , Candida albicans/genética , Humanos , Macrófagos/metabolismo , Ratones , Piroptosis
8.
JCI Insight ; 6(21)2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34747370

RESUMEN

Foxp3+ Tregs are potent immunosuppressive CD4+ T cells that are critical to maintain immune quiescence and prevent autoimmunity. Both the generation and maintenance of Foxp3+ Tregs depend on the cytokine IL-2. Hence, the expression of the IL-2 receptor α-chain (CD25) is not only considered a specific marker, but also a nonredundant requirement for Tregs. Here, we report that Foxp3+ Tregs in the small intestine (SI) epithelium, a critical barrier tissue, are exempt from such an IL-2 requirement, since they had dramatically downregulated CD25 expression, showed minimal STAT5 phosphorylation ex vivo, and were unable to respond to IL-2 in vitro. Nonetheless, SI epithelial Tregs survived and were present at the same frequency as in other lymphoid organs, and they retained potent suppressor function that was associated with high levels of CTLA-4 expression and the production of copious amounts of IL-10. Moreover, adoptive transfer experiments of Foxp3+ Tregs revealed that such IL-2-independent survival and effector functions were imposed by the SI epithelial tissue, suggesting that tissue adaptation is a mechanism that tailors the effector function and survival requirements of Foxp3+ Tregs specific to the tissue environment.


Asunto(s)
Epitelio/metabolismo , Factores de Transcripción Forkhead/metabolismo , Interleucina-2/metabolismo , Intestino Delgado/fisiopatología , Linfocitos T Reguladores/metabolismo , Animales , Homeostasis , Humanos , Ratones
9.
Cell Mol Life Sci ; 78(15): 5789-5805, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34129058

RESUMEN

Integrin CD103 mediates the adhesion and tissue retention of T cells by binding to E-cadherin which is abundant on epithelial cells. Notably, CD103 is highly expressed on CD8 T cells but conspicuously absent on most CD4 T cells. The mechanism controlling such lineage-specific expression of CD103 remains unclear. Using a series of genetically engineered mouse models, here, we demonstrate that the regulatory mechanism of CD103 expression is distinct between CD4 and CD8 T cells, and that the transcription factor Runx3 plays an important but not an essential role in this process. We further found that the availability of integrin ß7 which heterodimerizes with CD103 was necessary but also constrained the surface expression of CD103. Notably, the forced surface expression of CD103 did not significantly alter the thymic development of conventional T cells but severely impaired the generation of MHC-II-restricted TCR transgenic T cells, revealing previously unappreciated aspects of CD103 in the selection and maturation of CD4 T cells. Unlike its effect on CD4 T cell development, however, CD103 overexpression did not significantly affect CD4 T cells in peripheral tissues. Moreover, the frequency and number of CD4 T cells in the small intestine epithelium did not increase even though E-cadherin is highly expressed in this tissue. Collectively, these results suggest that most mature CD4 T cells are refractory to the effects of CD103 expression, and that they presumably utilize CD103-independent pathways to control their tissue retention and residency.


Asunto(s)
Antígenos CD/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Cadenas alfa de Integrinas/metabolismo , Animales , Cadherinas/metabolismo , Femenino , Cadenas beta de Integrinas/metabolismo , Mucosa Intestinal/metabolismo , Linfocitos Infiltrantes de Tumor/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
10.
Eur J Immunol ; 50(7): 986-999, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32144749

RESUMEN

SOCS3 is a cytosolic inhibitor of cytokine signaling that suppresses the activation of cytokine receptor-associated JAK kinases. Mechanistically, SOCS3 is recruited to a site in the cytokine receptors known as the SOCS3-interaction motif, and then binds JAK molecules to inhibit their kinase activity. The SOCS3-interaction motif is found in receptors of the gp130 cytokine family but mostly absent from other cytokine receptors, including γc. Thus, SOCS3 has been considered a selective suppressor of gp130 family cytokines, but not γc cytokines. Considering that γc signaling induces SOCS3 expression in T cells, here we revisited the role of SOCS3 on γc signaling. Using SOCS3 transgenic mice, we found that increased abundance of SOCS3 not only suppressed signaling of the gp130 family cytokine IL-6, but also signaling of the γc family cytokine IL-7. Consequently, SOCS3 transgenic mice were impaired in IL-7-dependent T cell development in the thymus and the homeostasis of mature T cells in peripheral tissues. Moreover, enforced SOCS3 expression interfered with the generation of Foxp3+ regulatory T cells that requires signaling by the γc family cytokine IL-2. Collectively, we report an underappreciated role for SOCS3 in suppressing γc cytokine signaling, effectively expanding its scope of target cytokines in T cell immunity.


Asunto(s)
Citocinas/inmunología , Inmunidad Celular , Transducción de Señal/inmunología , Proteína 3 Supresora de la Señalización de Citocinas/inmunología , Linfocitos T Reguladores/inmunología , Animales , Femenino , Factores de Transcripción Forkhead/inmunología , Masculino , Ratones , Linfocitos T Reguladores/citología
12.
J Infect Dis ; 219(11): 1841-1851, 2019 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-30615126

RESUMEN

The resolution of Shigella flexneri infection-associated hyperinflammation is crucial for host survival. Using in vitro and in vivo models of shigellosis, we found that S. flexneri induces the expression of indoleamine 2,3-dioxygenase 1 (IDO1) through the nucleotide oligomerization domain 2 (NOD2) and epidermal growth factor receptor (EGFR) signaling pathway. Congruently, abrogation of NOD2 or EGFR compromises the ability of S. flexneri to induce IDO1 expression. We observed that the loss of IDO1 function in vivo exacerbates shigellosis by skewing the inflammatory cytokine response, disrupting colon epithelial barrier integrity and consequently limiting the host life-span. Interestingly, administration of recombinant EGF rescued mice from IDO1 inhibition-driven aggravated shigellosis by restoring the cytokine balance and subsequently restricting bacterial growth. This is the first study that underscores the direct implication of the NOD2-EGFR axis in IDO1 production and its crucial homeostatic contributions during shigellosis. Together, these findings reveal EGF as a potential therapeutic intervention for infectious diseases.


Asunto(s)
Citocinas/metabolismo , Disentería Bacilar/inmunología , Receptores ErbB/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Shigella flexneri/inmunología , Transducción de Señal , Animales , Disentería Bacilar/microbiología , Receptores ErbB/genética , Homeostasis , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Ratones , Ratones Endogámicos BALB C , Células RAW 264.7
13.
Front Immunol ; 9: 85, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29449840

RESUMEN

Mycobacteria propelled modulation of host responses is of considerable interest in the face of emerging drug resistance. Although it is known that Abl tyrosine kinases affect entry and persistence of mycobacteria, mechanisms that couple c-Abl to proximal signaling pathways during immunity are poorly understood. Loss-of-function of c-Abl through Imatinib, in a mouse model of tuberculosis or RNA interference, identified bone morphogenesis protein (BMP) signaling as its cellular target. We demonstrate that c-Abl promotes mycobacterial survival through epigenetic modification brought about by KAT5-TWIST1 at Bmp loci. c-Abl-BMP signaling deregulated iNOS, aggravating the inflammatory balance. Interestingly, BMP signaling was observed to have far-reaching effects on host immunity, as it attenuated TLR3 pathway by engaging miR27a. Significantly, these events were largely mediated via WhiB3 and DosR/S/T but not SecA signaling pathway of mycobacteria. Our findings suggest molecular mechanisms of host pathways hijacked by mycobacteria and expand our understanding of c-Abl inhibitors in potentiating innate immune responses.


Asunto(s)
Proteínas Morfogenéticas Óseas/genética , Regulación de la Expresión Génica , MicroARNs/genética , Proteínas Nucleares/genética , Proteínas Proto-Oncogénicas c-abl/genética , Tuberculosis/genética , Tuberculosis/metabolismo , Proteína 1 Relacionada con Twist/genética , Regiones no Traducidas 3' , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Línea Celular , Ensamble y Desensamble de Cromatina , Epigénesis Genética , Femenino , Interacciones Huésped-Patógeno , Masculino , Ratones , Modelos Biológicos , Interferencia de ARN , Especies Reactivas de Oxígeno , Transducción de Señal , Receptor Toll-Like 3/metabolismo , Tuberculosis/inmunología , Tuberculosis/microbiología
14.
J Infect Dis ; 216(10): 1281-1294, 2017 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-28968869

RESUMEN

Background: Human dendritic cell (DC) response to α-(1,3)-glucan polysaccharide of Aspergillus fumigatus and ensuing CD4+ T-cell polarization are poorly characterized. Methods: α-(1,3)-Glucan was isolated from A. fumigatus conidia and mycelia cell wall. For the analysis of polarization, DCs and autologous naive CD4+ T cells were cocultured. Phenotype of immune cells was analyzed by flow cytometry, and cytokines by enzyme-linked immunosorbent assay (ELISA). Blocking antibodies were used to dissect the role of Toll-like receptor 2 (TLR2) and programmed death-ligand 1 (PD-L1) in regulating α-(1,3)-glucan-mediated DC activation and T-cell responses. DCs from TLR2-deficient mice were additionally used to consolidate the findings. Results: α-(1,3)-Glucan induced the maturation of DCs and was dependent in part on TLR2. "α-(1,3)-Glucan-educated" DCs stimulated the activation of naive T cells and polarized a subset of these cells into CD4+CD25+FoxP3+ regulatory T cells (Tregs). Mechanistically, Treg stimulation by α-(1,3)-glucan was dependent on the PD-L1 pathway that negatively regulated interferon-gamma (IFN-γ) secretion. Short α-(1,3)-oligosaccharides lacked the capacity to induce maturation of DCs but significantly blocked α-(1,3)-glucan-induced Treg polarization. Conclusions: PD-L1 dictates the balance between Treg and IFN-γ responses induced by α-(1,3)-glucan. Our data provide a rationale for the exploitation of immunotherapeutic approaches that target PD-1-PD-L1 to enhance protective immune responses to A. fumigatus infections.


Asunto(s)
Aspergillus fumigatus/inmunología , Antígeno B7-H1/genética , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Expresión Génica , Glucanos/inmunología , Activación de Linfocitos/inmunología , Linfocitos T Reguladores/inmunología , Animales , Biomarcadores , Citocinas/metabolismo , Humanos , Interferón gamma/metabolismo , Ratones , Ratones Noqueados , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Linfocitos T Reguladores/metabolismo
15.
J Infect Dis ; 216(4): 477-488, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28931237

RESUMEN

NQO1 and TRXR1 are important host reductases implicated in the regulation of inflammation and apoptosis. Although the transcriptional machinery governing these processes have been extensively investigated, the associated epigenetic regulatory events remain unclear. Here, we report that SET8, a histone H4 lysine 20 monomethylase (H4K20me1), is highly induced during Mycobacterium tuberculosis infection that orchestrates immune evasion strategies through the induction of NQO1 and TRXR1 in vivo. SET8, along with FoxO3a, mediates an active NQO1-PGC1-α complex, which promotes the anti-inflammatory M2 macrophage phenotype, and assists TRXR1-regulated arrest of tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis. Strikingly, the loss-of-function analysis in an in vivo mouse tuberculosis model further corroborated the pivotal role of SET8-responsive NQO1 and TRXR1 in mycobacterial survival. Thus, augmenting host immune responses against Mycobacterium tuberculosis by harnessing the SET8-NQO1/TRXR1 axis with its specific and potent inhibitors could lead to promising host-directed therapeutic adjuvants for tuberculosis treatment.


Asunto(s)
Epigénesis Genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Mycobacterium tuberculosis/crecimiento & desarrollo , Tuberculosis/inmunología , Animales , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Proteína Forkhead Box O3/genética , Proteína Forkhead Box O3/metabolismo , Regulación de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Evasión Inmune , Leucocitos Mononucleares/microbiología , Ratones , NAD(P)H Deshidrogenasa (Quinona)/genética , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Células RAW 264.7 , Reproducibilidad de los Resultados , Transducción de Señal , Tiorredoxina Reductasa 1/genética , Tiorredoxina Reductasa 1/metabolismo , Tuberculosis/microbiología
16.
J Neuroinflammation ; 14(1): 58, 2017 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-28320438

RESUMEN

BACKGROUND: Intravenous immunoglobulin (IVIG) is a polyspecific pooled immunoglobulin G preparation and one of the commonly used therapeutics for autoimmune diseases including those of neurological origin. A recent report in murine model proposed that IVIG expands regulatory T (Treg) cells via induction of interleukin 33 (IL-33). However, translational insight on these observations is lacking. METHODS: Ten newly diagnosed Guillain-Barré syndrome (GBS) patients were treated with IVIG at the rate of 0.4 g/kg for three to five consecutive days. Clinical evaluation for muscular weakness was performed by Medical Research Council (MRC) and modified Rankin scoring (MRS) system. Heparinized blood samples were collected before and 1, 2, and 4-5 weeks post-IVIG therapy. Peripheral blood mononuclear cells were stained for surface CD4 and intracellular Foxp3, IFN-γ, and tumor necrosis factor alpha (TNF-α) and were analyzed by flow cytometry. IL-33 and prostaglandin E2 in the plasma were measured by ELISA. RESULTS: The fold changes in plasma IL-33 at week 1 showed no correlation with the MRC and MRS scores at weeks 1, 2, and ≥4 post-IVIG therapy. Clinical recovery following IVIG therapy appears to be associated with Treg cell response. Contrary to murine study, there was no association between the fold changes in IL-33 at week 1 and Treg cell frequency at weeks 1, 2, and ≥4 post-IVIG therapy. Treg cell-mediated clinical response to IVIG therapy in GBS patients was associated with reciprocal regulation of effector T cells-expressing TNF-α. CONCLUSION: Treg cell expansion by IVIG in patients with autoimmune diseases lack correlation with IL-33. Treg cell frequency, but not plasma IL-33 levels, represents potential immunological biomarker to predict clinical response to IVIG therapy.


Asunto(s)
Síndrome de Guillain-Barré , Inmunoglobulinas Intravenosas/uso terapéutico , Factores Inmunológicos/uso terapéutico , Interleucina-33/sangre , Linfocitos T Reguladores/patología , Anciano , Anciano de 80 o más Años , Dinoprostona/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Estudios de Seguimiento , Síndrome de Guillain-Barré/sangre , Síndrome de Guillain-Barré/tratamiento farmacológico , Síndrome de Guillain-Barré/patología , Humanos , Masculino , Persona de Mediana Edad , Evaluación de Resultado en la Atención de Salud , Valor Predictivo de las Pruebas , Índice de Severidad de la Enfermedad , Estadísticas no Paramétricas
17.
PLoS Pathog ; 12(8): e1005814, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27532872

RESUMEN

Foamy macrophages (FM)s harbor lipid bodies that not only assist mycobacterial persistence within the granulomas but also are sites for intracellular signaling and inflammatory mediators which are essential for mycobacterial pathogenesis. However, molecular mechanisms that regulate intracellular lipid accumulation in FMs during mycobacterial infection are not clear. Here, we report for the first time that jumonji domain containing protein (JMJD)3, a demethylase of the repressive H3K27me3 mark, orchestrates the expression of M. tuberculosis H37Rv-, MDR-JAL2287-, H37Ra- and M. bovis BCG-induced genes essential for FM generation in a TLR2-dependent manner. Further, NOTCH1-responsive RNA-binding protein MUSASHI (MSI), targets a transcriptional repressor of JMJD3, Msx2-interacting nuclear target protein, to positively regulate infection-induced JMJD3 expression, FM generation and M2 phenotype. Investigations in in vivo murine models further substantiated these observations. Together, our study has attributed novel roles for JMJD3 and its regulators during mycobacterial infection that assist FM generation and fine-tune associated host immunity.


Asunto(s)
Histona Demetilasas con Dominio de Jumonji/inmunología , Macrófagos/microbiología , Infecciones por Mycobacterium/inmunología , Mycobacterium tuberculosis/inmunología , Proteínas del Tejido Nervioso/inmunología , Proteínas de Unión al ARN/inmunología , Animales , Inmunoprecipitación de Cromatina , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Regulación Bacteriana de la Expresión Génica/inmunología , Granuloma/inmunología , Granuloma/microbiología , Immunoblotting , Inmunohistoquímica , Inmunoprecipitación , Histona Demetilasas con Dominio de Jumonji/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Infecciones por Mycobacterium/metabolismo , Mycobacterium tuberculosis/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al ARN/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transfección , Tuberculosis/inmunología , Tuberculosis/metabolismo
18.
Sci Rep ; 6: 24193, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-27080341

RESUMEN

CD4(+)CD25(+)FoxP3(+) regulatory T cells (Tregs) are exploited by mycobacteria to subvert the protective host immune responses. The Treg expansion in the periphery requires signaling by professional antigen presenting cells and in particularly dendritic cells (DC). However, precise molecular mechanisms by which mycobacteria instruct Treg expansion via DCs are not established. Here we demonstrate that mycobacteria-responsive sonic hedgehog (SHH) signaling in human DCs leads to programmed death ligand-1 (PD-L1) expression and cyclooxygenase (COX)-2-catalyzed prostaglandin E2 (PGE2) that orchestrate mycobacterial infection-induced expansion of Tregs. While SHH-responsive transcription factor GLI1 directly arbitrated COX-2 transcription, specific microRNAs, miR-324-5p and miR-338-5p, which target PD-L1 were downregulated by SHH signaling. Further, counter-regulatory roles of SHH and NOTCH1 signaling during mycobacterial-infection of human DCs was also evident. Together, our results establish that Mycobacterium directs a fine-balance of host signaling pathways and molecular regulators in human DCs to expand Tregs that favour immune evasion of the pathogen.


Asunto(s)
Antígeno B7-H1/metabolismo , Dinoprostona/metabolismo , Proteínas Hedgehog/metabolismo , Mycobacterium/inmunología , Transducción de Señal , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Ciclooxigenasa 2/metabolismo , Humanos , Activación de Linfocitos/inmunología , Mycobacterium bovis/inmunología , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores Notch/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
19.
J Biol Chem ; 290(44): 26576-86, 2015 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-26391398

RESUMEN

Specific and coordinated regulation of innate immune receptor-driven signaling networks often determines the net outcome of the immune responses. Here, we investigated the cross-regulation of toll-like receptor (TLR)2 and nucleotide-binding oligomerization domain (NOD)2 pathways mediated by Ac2PIM, a tetra-acylated form of mycobacterial cell wall component and muramyl dipeptide (MDP), a peptidoglycan derivative respectively. While Ac2PIM treatment of macrophages compromised their ability to induce NOD2-dependent immunomodulators like cyclooxygenase (COX)-2, suppressor of cytokine signaling (SOCS)-3, and matrix metalloproteinase (MMP)-9, no change in the NOD2-responsive NO, TNF-α, VEGF-A, and IL-12 levels was observed. Further, genome-wide microRNA expression profiling identified Ac2PIM-responsive miR-150 and miR-143 to target NOD2 signaling adaptors, RIP2 and TAK1, respectively. Interestingly, Ac2PIM was found to activate the SRC-FAK-PYK2-CREB cascade via TLR2 to recruit CBP/P300 at the promoters of miR-150 and miR-143 and epigenetically induce their expression. Loss-of-function studies utilizing specific miRNA inhibitors establish that Ac2PIM, via the miRNAs, abrogate NOD2-induced PI3K-PKCδ-MAPK pathway to suppress ß-catenin-mediated expression of COX-2, SOCS-3, and MMP-9. Our investigation has thus underscored the negative regulatory role of Ac2PIM-TLR2 signaling on NOD2 pathway which could broaden our understanding on vaccine potential or adjuvant utilities of Ac2PIM and/or MDP.


Asunto(s)
Quinasas Quinasa Quinasa PAM/metabolismo , Macrófagos/metabolismo , MicroARNs/metabolismo , Proteína Adaptadora de Señalización NOD2/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Acetilmuramil-Alanil-Isoglutamina/farmacología , Animales , Línea Celular , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Ciclooxigenasa 2/farmacología , Epigénesis Genética , Inmunidad Innata , Factores Inmunológicos/farmacología , Quinasas Quinasa Quinasa PAM/genética , Macrófagos/citología , Macrófagos/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Proteínas Quinasas Activadas por Mitógenos , Óxido Nítrico/metabolismo , Proteína Adaptadora de Señalización NOD2/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Polisacáridos Bacterianos/farmacología , Unión Proteica , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Transducción de Señal , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/farmacología , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo
20.
Mol Cell Biol ; 34(23): 4301-14, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25246634

RESUMEN

Macrophages regulate cell fate decisions during microbial challenges by carefully titrating signaling events activated by innate receptors such as dectin-1 or Toll-like receptors (TLRs). Here, we demonstrate that dectin-1 activation robustly dampens TLR-induced proinflammatory signature in macrophages. Dectin-1 induced the stabilization of ß-catenin via spleen tyrosine kinase (Syk)-reactive oxygen species (ROS) signals, contributing to the expression of WNT5A. Subsequently, WNT5A-responsive protein inhibitors of activated STAT (PIAS-1) and suppressor of cytokine signaling 1 (SOCS-1) mediate the downregulation of IRAK-1, IRAK-4, and MyD88, resulting in decreased expression of interleukin 12 (IL-12), IL-1ß, and tumor necrosis factor alpha (TNF-α). In vivo activation of dectin-1 with pathogenic fungi or ligand resulted in an increased bacterial burden of Mycobacteria, Klebsiella, Staphylococcus, or Escherichia, with a concomitant decrease in TLR-triggered proinflammatory cytokines. All together, our study establishes a new role for dectin-1-responsive inhibitory mechanisms employed by virulent fungi to limit the proinflammatory environment of the host.


Asunto(s)
Inflamación/inmunología , Lectinas Tipo C/inmunología , Macrófagos/inmunología , Receptores Toll-Like/inmunología , Vía de Señalización Wnt/inmunología , Animales , Aspergillus flavus/patogenicidad , Aspergillus fumigatus/patogenicidad , Carga Bacteriana/inmunología , Candida albicans/patogenicidad , Regulación hacia Abajo , Activación Enzimática , Escherichia/patogenicidad , Quinasas Asociadas a Receptores de Interleucina-1/biosíntesis , Interleucina-12/biosíntesis , Interleucina-1beta/biosíntesis , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Klebsiella/patogenicidad , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium/patogenicidad , Factor 88 de Diferenciación Mieloide/biosíntesis , Proteínas Inhibidoras de STAT Activados/biosíntesis , Proteínas Tirosina Quinasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Staphylococcus/patogenicidad , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/biosíntesis , Quinasa Syk , Factor de Necrosis Tumoral alfa/biosíntesis , Proteínas Wnt/biosíntesis , Proteína Wnt-5a , beta Catenina/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...