Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Immunol ; 212(7): 1161-1171, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38372637

RESUMEN

Monocytes are actively recruited to sites of infection and produce the potent proinflammatory cytokine IL-1ß. We previously showed that IL-1ß release during Toxoplasma gondii infection of primary human monocytes requires the NLRP3 inflammasome and caspase-1 but is independent of gasdermin D and pyroptosis. To investigate mechanisms of IL-1ß release, we generated caspase-1, -4, -5, or -8 knockout (KO) THP-1 monocytic cells. Genetic ablation of caspase-1 or -8, but not caspase-4 or -5, decreased IL-1ß release during T. gondii infection without affecting cell death. In contrast, TNF-α and IL-6 secretion were unperturbed in caspase-8 KO cells during T. gondii infection. Dual pharmacological inhibition of caspase-8 and RIPK1 in primary monocytes also decreased IL-1ß release without affecting cell viability or parasite infection. Caspase-8 was also required for the release of active caspase-1 from T. gondii-infected cells and for IL-1ß release during infection with the related apicomplexan parasite Neospora caninum. Surprisingly, caspase-8 deficiency did not impair synthesis or cleavage of pro-IL-1ß, but resulted in the retention of mature IL-1ß within cells. Generation of gasdermin E KO and ATG7 KO THP-1 cells revealed that the release of IL-1ß was not dependent on gasdermin E or ATG7. Collectively, our data indicate that during T. gondii Infection of human monocytes, caspase-8 functions in a novel gasdermin-independent mechanism controlling IL-1ß release from viable cells. This study expands on the molecular pathways that promote IL-1ß in human immune cells and provides evidence of a role for caspase-8 in the mechanism of IL-1ß release during infection.


Asunto(s)
Caspasa 8 , Interleucina-1beta , Toxoplasma , Toxoplasmosis , Humanos , Caspasa 1/metabolismo , Caspasa 8/metabolismo , Gasderminas , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Monocitos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Toxoplasmosis/metabolismo
2.
Curr Opin Microbiol ; 72: 102264, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36791673

RESUMEN

Intracellular pathogens strike a delicate balance between maintaining their survival within infected cells, while also activating host defense mechanisms. Toxoplasma gondii is a protozoan parasite that initiates a variety of host signaling pathways as it invades host cells and establishes residence in a parasitophorous vacuole. Recent work has highlighted the interplay between T. gondii infection and innate immune pathways that lead to inflammation, several of which converge on caspases. This family of cysteine proteases function at the crossroads of inflammation and cell death and serve as a key target for parasite manipulation. This review focuses on the interaction of T. gondii with caspase-dependent inflammatory and cell death pathways and the role of parasite effector proteins in modulating these processes.


Asunto(s)
Capparis , Toxoplasma , Humanos , Toxoplasma/fisiología , Capparis/metabolismo , Caspasas/metabolismo , Transducción de Señal , Inflamación , Proteínas Protozoarias/metabolismo
3.
Int J Mol Sci ; 23(17)2022 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-36077273

RESUMEN

Despite the decades-old knowledge that males and people with diabetes mellitus (DM) are at increased risk for coronary artery disease (CAD), the reasons for this association are only partially understood. Among the immune cells involved, recent evidence supports a critical role of T cells as drivers and modifiers of CAD. CD4+ T cells are commonly found in atherosclerotic plaques. We aimed to understand the relationship of CAD with sex and DM by single-cell RNA (scRNA-Seq) and antibody sequencing (CITE-Seq) of CD4+ T cells. Peripheral blood mononuclear cells (PBMCs) of 61 men and women who underwent cardiac catheterization were interrogated by scRNA-Seq combined with 49 surface markers (CITE-Seq). CAD severity was quantified using Gensini scores, with scores above 30 considered CAD+ and below 6 considered CAD-. Four pairs of groups were matched for clinical and demographic parameters. To test how sex and DM changed cell proportions and gene expression, we compared matched groups of men and women, as well as diabetic and non-diabetic subjects. We analyzed 41,782 single CD4+ T cell transcriptomes for sex differences in 16 women and 45 men with and without coronary artery disease and with and without DM. We identified 16 clusters in CD4+ T cells. The proportion of cells in CD4+ effector memory cluster 8 (CD4T8, CCR2+ Em) was significantly decreased in CAD+, especially among DM+ participants. This same cluster, CD4T8, was significantly decreased in female participants, along with two other CD4+ T cell clusters. In CD4+ T cells, 31 genes showed significant and coordinated upregulation in both CAD and DM. The DM gene signature was partially additive to the CAD gene signature. We conclude that (1) CAD and DM are clearly reflected in PBMC transcriptomes, and (2) significant differences exist between women and men and (3) between subjects with DM and non-DM.


Asunto(s)
Enfermedad de la Arteria Coronaria , Diabetes Mellitus , Linfocitos T CD4-Positivos , Angiografía Coronaria , Enfermedad de la Arteria Coronaria/genética , Diabetes Mellitus/genética , Femenino , Humanos , Leucocitos Mononucleares , Masculino , Caracteres Sexuales , Análisis de la Célula Individual
4.
J Leukoc Biol ; 112(5): 1053-1063, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35866369

RESUMEN

Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can result in severe immune dysfunction, hospitalization, and death. Many patients also develop long-COVID-19, experiencing symptoms months after infection. Although significant progress has been made in understanding the immune response to acute SARS-CoV-2 infection, gaps remain in our knowledge of how innate immunity influences disease kinetics and severity. We hypothesized that cytometry by time-of-flight analysis of PBMCs from healthy and infected subjects would identify novel cell surface markers and innate immune cell subsets associated with COVID-19 severity. In this pursuit, we identified monocyte and dendritic cell subsets that changed in frequency during acute SARS-CoV-2 infection and correlated with clinical parameters of disease severity. Subsets of nonclassical monocytes decreased in frequency in hospitalized subjects, yet increased in the most severe patients and positively correlated with clinical values associated with worse disease severity. CD9, CD163, PDL1, and PDL2 expression significantly increased in hospitalized subjects, and CD9 and 6-Sulfo LacNac emerged as the markers that best distinguished monocyte subsets amongst all subjects. CD9+ monocytes remained elevated, whereas nonclassical monocytes remained decreased, in the blood of hospitalized subjects at 3-4 months postinfection. Finally, we found that CD9+ monocytes functionally released more IL-8 and MCP-1 after LPS stimulation. This study identifies new monocyte subsets present in the blood of COVID-19 patients that correlate with disease severity, and links CD9+ monocytes to COVID-19 progression.


Asunto(s)
COVID-19 , Humanos , Monocitos , SARS-CoV-2 , Interleucina-8/metabolismo , Lipopolisacáridos/metabolismo , Células Mieloides , Hospitalización , Tetraspanina 29/metabolismo , Síndrome Post Agudo de COVID-19
5.
Sci Adv ; 6(49)2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33277245

RESUMEN

Macrophages are innate immune cells that adhere to the extracellular matrix within tissues. However, how matrix properties regulate their function remains poorly understood. Here, we report that the adhesive microenvironment tunes the macrophage inflammatory response through the transcriptional coactivator YAP. We find that adhesion to soft hydrogels reduces inflammation when compared to adhesion on stiff materials and is associated with reduced YAP expression and nuclear localization. Substrate stiffness and cytoskeletal polymerization, but not adhesive confinement nor contractility, regulate YAP localization. Furthermore, depletion of YAP inhibits macrophage inflammation, whereas overexpression of active YAP increases inflammation. Last, we show in vivo that soft materials reduce expression of inflammatory markers and YAP in surrounding macrophages when compared to stiff materials. Together, our studies identify YAP as a key molecule for controlling inflammation and sensing stiffness in macrophages and may have broad implications in the regulation of macrophages in health and disease.


Asunto(s)
Mecanotransducción Celular , Proteínas Señalizadoras YAP , Matriz Extracelular/metabolismo , Humanos , Inflamación/metabolismo , Macrófagos , Mecanotransducción Celular/fisiología
7.
PLoS Pathog ; 15(8): e1007923, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31449558

RESUMEN

IL-1ß is a potent pro-inflammatory cytokine that promotes immunity and host defense, and its dysregulation is associated with immune pathology. Toxoplasma gondii infection of myeloid cells triggers the production and release of IL-1ß; however, the mechanisms regulating this pathway, particularly in human immune cells, are incompletely understood. We have identified a novel pathway of T. gondii induction of IL-1ß via a Syk-CARD9-NF-κB signaling axis in primary human peripheral blood monocytes. Syk was rapidly phosphorylated during T. gondii infection of primary monocytes, and inhibiting Syk with the pharmacological inhibitors R406 or entospletinib, or genetic ablation of Syk in THP-1 cells, reduced IL-1ß release. Inhibition of Syk in primary cells or deletion of Syk in THP-1 cells decreased parasite-induced IL-1ß transcripts and the production of pro-IL-1ß. Furthermore, inhibition of PKCδ, CARD9/MALT-1 and IKK reduced p65 phosphorylation and pro-IL-1ß production in T. gondii-infected primary monocytes, and genetic knockout of PKCδ or CARD9 in THP-1 cells also reduced pro-IL-1ß protein levels and IL-1ß release during T. gondii infection, indicating that Syk functions upstream of this NF-κB-dependent signaling pathway for IL-1ß transcriptional activation. IL-1ß release from T. gondii-infected primary human monocytes required the NLRP3-caspase-1 inflammasome, but interestingly, was independent of gasdermin D (GSDMD) cleavage and pyroptosis. Moreover, GSDMD knockout THP-1 cells released comparable amounts of IL-1ß to wild-type THP-1 cells after T. gondii infection. Taken together, our data indicate that T. gondii induces a Syk-CARD9/MALT-1-NF-κB signaling pathway and activation of the NLRP3 inflammasome for the release of IL-1ß in a cell death- and GSDMD-independent manner. This research expands our understanding of the molecular basis for human innate immune regulation of inflammation and host defense during parasite infection.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/metabolismo , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Monocitos/metabolismo , FN-kappa B/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Quinasa Syk/metabolismo , Toxoplasmosis/metabolismo , Proteínas Adaptadoras de Señalización CARD/genética , Células Cultivadas , Humanos , Inflamasomas , Péptidos y Proteínas de Señalización Intracelular/genética , Monocitos/inmunología , Monocitos/microbiología , FN-kappa B/genética , Proteínas de Unión a Fosfato/genética , Transducción de Señal , Quinasa Syk/genética , Toxoplasma/fisiología , Toxoplasmosis/inmunología , Toxoplasmosis/microbiología
8.
J Immunol ; 199(8): 2855-2864, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28904126

RESUMEN

IL-1ß is produced by myeloid cells and acts as a critical mediator of host defense during infection and injury. We found that the intracellular protozoan parasite Toxoplasma gondii induced an early IL-1ß response (within 4 h) in primary human peripheral blood monocytes isolated from healthy donors. This process involved upregulation of IL-1ß, IL-1RN (IL-1R antagonist), and NLRP3 transcripts, de novo protein synthesis, and the release of pro- and mature IL-1ß from infected primary monocytes. The released pro-IL-1ß was cleavable to mature bioactive IL-1ß in the extracellular space by the protease caspase-1. Treatment of primary monocytes with the NLRP3 inhibitor MCC950 or with extracellular potassium significantly reduced IL-1ß cleavage and release in response to T. gondii infection, without affecting the release of TNF-α, and indicated a role for the inflammasome sensor NLRP3 and for potassium efflux in T. gondii-induced IL-1ß production. Interestingly, T. gondii infection did not induce an IL-1ß response in primary human macrophages derived from the same blood donors as the monocytes. Consistent with this finding, NLRP3 was downregulated during the differentiation of monocytes to macrophages and was not induced in macrophages during T. gondii infection. To our knowledge, these findings are the first to identify NLRP3 as an inflammasome sensor for T. gondii in primary human peripheral blood cells and to define an upstream regulator of its activation through the release of intracellular potassium.


Asunto(s)
Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Monocitos/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Potasio/metabolismo , Toxoplasma/inmunología , Toxoplasmosis/inmunología , Diferenciación Celular , Células Cultivadas , Furanos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Indenos , Macrófagos/inmunología , Monocitos/parasitología , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Cultivo Primario de Células , Proteolisis/efectos de los fármacos , Sulfonamidas , Sulfonas/farmacología
9.
Mol Cancer Ther ; 16(9): 1942-1953, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28566433

RESUMEN

Elevated activity of mTOR is associated with poor prognosis and higher incidence of relapse in B-cell acute lymphoblastic leukemia (B-ALL). Thus, ongoing clinical trials are testing mTOR inhibitors in combination with chemotherapy in B-ALL. However, the combination of mTOR inhibitors with standard of care chemotherapy drugs has not been studied extensively in high-risk B-ALL subtypes. Therefore, we tested whether mTOR inhibition can augment the efficacy of current chemotherapy agents in Ph+ and Ph-like B-ALL models. Surprisingly, inhibiting mTOR complex 1 (mTORC1) protected B-ALL cells from killing by methotrexate and 6-mercaptopurine, two antimetabolite drugs used in maintenance chemotherapy. The cytoprotective effects correlated with decreased cell-cycle progression and were recapitulated using cell-cycle inhibitors, palbociclib or aphidicolin. Dasatinib, a tyrosine kinase inhibitor currently used in Ph+ patients, inhibits ABL kinase upstream of mTOR. Dasatinib resistance is mainly caused by ABL kinase mutations, but is also observed in a subset of ABL unmutated cases. We identified dasatinib-resistant Ph+ cell lines and patient samples in which dasatinib can effectively reduce ABL kinase activity and mTORC1 signaling without causing cell death. In these cases, dasatinib protected leukemia cells from killing by 6-mercaptopurine. Using xenograft models, we observed that mTOR inhibition or dasatinib increased the numbers of leukemia cells that emerge after cessation of chemotherapy treatment. These results demonstrate that inhibitors targeting mTOR or upstream signaling nodes should be used with caution when combined with chemotherapeutic agents that rely on cell-cycle progression to kill B-ALL cells. Mol Cancer Ther; 16(9); 1942-53. ©2017 AACR.


Asunto(s)
Resistencia a Antineoplásicos , Diana Mecanicista del Complejo 1 de la Rapamicina/antagonistas & inhibidores , Mercaptopurina/farmacología , Metotrexato/farmacología , Cromosoma Filadelfia , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Animales , Antimetabolitos Antineoplásicos/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Daño del ADN , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Modelos Biológicos , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
PLoS One ; 9(5): e96575, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24804781

RESUMEN

Lesch-Nyhan Syndrome (LNS) is a neurodevelopmental disorder caused by mutations in the gene encoding the purine metabolic enzyme hypoxanthine-guanine phosphoribosyltransferase (HPRT). This syndrome is characterized by an array of severe neurological impairments that in part originate from striatal dysfunctions. However, the molecular and cellular mechanisms underlying these dysfunctions remain largely unidentified. In this report, we demonstrate that HPRT-deficiency causes dysregulated expression of key genes essential for striatal patterning, most notably the striatally-enriched transcription factor B-cell leukemia 11b (Bcl11b). The data also reveal that the down-regulated expression of Bcl11b in HPRT-deficient immortalized mouse striatal (STHdh) neural stem cells is accompanied by aberrant expression of some of its transcriptional partners and other striatally-enriched genes, including the gene encoding dopamine- and cAMP-regulated phosphoprotein 32, (DARPP-32). Furthermore, we demonstrate that components of the BDNF/TrkB signaling, a known activator of DARPP-32 striatal expression and effector of Bcl11b transcriptional activation are markedly increased in HPRT-deficient cells and in the striatum of HPRT knockout mouse. Consequently, the HPRT-deficient cells display superior protection against reactive oxygen species (ROS)-mediated cell death upon exposure to hydrogen peroxide. These findings suggest that the purine metabolic defect caused by HPRT-deficiency, while it may provide neuroprotection to striatal neurons, affects key genes and signaling pathways that may underlie the neuropathogenesis of LNS.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/genética , Cuerpo Estriado/patología , Fosfoproteína 32 Regulada por Dopamina y AMPc/genética , Hipoxantina Fosforribosiltransferasa/genética , Síndrome de Lesch-Nyhan/genética , Receptor trkB/genética , Transducción de Señal/genética , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Muerte Celular/genética , Diferenciación Celular/genética , Cuerpo Estriado/metabolismo , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Síndrome de Lesch-Nyhan/metabolismo , Síndrome de Lesch-Nyhan/patología , Ratones , Ratones Noqueados , Neuronas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor trkB/metabolismo
11.
Hum Mol Genet ; 22(22): 4502-15, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23804752

RESUMEN

Lesch-Nyhan syndrome (LNS) is a neurodevelopmental disorder caused by mutations in the gene encoding the purine metabolic enzyme hypoxanthine-guanine phosphoribosyltransferase (HPRT). A series of motor, cognitive and neurobehavioral anomalies characterize this disease phenotype, which is still poorly understood. The clinical manifestations of this syndrome are believed to be the consequences of deficiencies in neurodevelopmental pathways that lead to disordered brain function. We have used microRNA array and gene ontology analysis to evaluate the gene expression of differentiating HPRT-deficient human neuron-like cell lines. We set out to identify dysregulated genes implicated in purine-based cellular functions. Our approach was based on the premise that HPRT deficiency affects preeminently the expression and the function of purine-based molecular complexes, such as guanine nucleotide exchange factors (GEFs) and small GTPases. We found that several microRNAs from the miR-17 family cluster and genes encoding GEF are dysregulated in HPRT deficiency. Most notably, our data show that the expression of the exchange protein activated by cAMP (EPAC) is blunted in HPRT-deficient human neuron-like cell lines and fibroblast cells from LNS patients, and is altered in the cortex, striatum and midbrain of HPRT knockout mouse. We also show a marked impairment in the activation of small GTPase RAP1 in the HPRT-deficient cells, as well as differences in cytoskeleton dynamics that lead to increased motility for HPRT-deficient neuron-like cell lines relative to control. We propose that the alterations in EPAC/RAP1 signaling and cell migration in HPRT deficiency are crucial for neuro-developmental events that may contribute to the neurological dysfunctions in LNS.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/metabolismo , Síndrome de Lesch-Nyhan/genética , MicroARNs/genética , Proteínas de Unión al GTP rap1/metabolismo , Animales , Línea Celular , Movimiento Celular/fisiología , Corteza Cerebral/metabolismo , Cuerpo Estriado/metabolismo , Citoesqueleto/metabolismo , Ontología de Genes , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Hipoxantina Fosforribosiltransferasa/deficiencia , Hipoxantina Fosforribosiltransferasa/genética , Síndrome de Lesch-Nyhan/enzimología , Masculino , Mesencéfalo/metabolismo , Ratones , Ratones Noqueados , MicroARNs/metabolismo , Familia de Multigenes , Análisis de Secuencia por Matrices de Oligonucleótidos , Transducción de Señal , Proteínas de Unión al GTP rap1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA