Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.620
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Cell ; 177(2): 384-398.e11, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30853218

RESUMEN

The signaling organelles of the innate immune system consist of oligomeric protein complexes known as supramolecular organizing centers (SMOCs). Examples of SMOCs include myddosomes and inflammasomes, which respectively induce transcription-dependent and -independent inflammatory responses. The common use of oligomeric structures as signaling platforms suggests multifunctionality, but each SMOC has a singular biochemically defined function. Here, we report that the myddosome is a multifunctional organizing center. In addition to promoting inflammatory transcription factor activation, the myddosome drives the rapid induction of glycolysis. We identify the kinase TBK1 as a myddosome component that promotes glycolysis, but not nuclear factor κB (NF-κB) activation. Synthetic immunology approaches further diversified SMOC activities, as we created interferon- or necroptosis-inducing myddosomes, inflammasomes that induce interferon responses instead of pyroptosis, and a SMOC-like nanomachine that induces interferon expression in response to a chemical ligand. These discoveries demonstrate the flexibility of immune signaling organelles, which permits the design of user-defined innate immune responses.


Asunto(s)
Inmunidad Innata/inmunología , Inmunidad Innata/fisiología , Transducción de Señal/inmunología , Animales , Glucólisis/inmunología , Inflamasomas , Ratones , Ratones Endogámicos C57BL , Enzimas Multifuncionales/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Orgánulos/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores Toll-Like
2.
Cell ; 171(4): 783-794.e13, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-28942917

RESUMEN

Intestinal intraepithelial lymphocytes (IELs) are located at the critical interface between the intestinal lumen, which is chronically exposed to food and microbes, and the core of the body. Using high-resolution microscopy techniques and intersectional genetic tools, we investigated the nature of IEL responses to luminal microbes. We observed that TCRγδ IELs exhibit unique microbiota-dependent location and movement patterns in the epithelial compartment. This behavioral pattern quickly changes upon exposure to different enteric pathogens, resulting in increased interepithelial cell (EC) scanning, expression of antimicrobial genes, and glycolysis. Both dynamic and metabolic changes to γδ IEL depend on pathogen sensing by ECs. Direct modulation of glycolysis is sufficient to change γδ IEL behavior and susceptibility to early pathogen invasion. Our results uncover a coordinated EC-IEL response to enteric infections that modulates lymphocyte energy utilization and dynamics and supports maintenance of the intestinal epithelial barrier. VIDEO ABSTRACT.


Asunto(s)
Intestinos/citología , Intestinos/inmunología , Infecciones por Salmonella/inmunología , Linfocitos T/inmunología , Animales , Células Epiteliales/metabolismo , Vigilancia Inmunológica , Mucosa Intestinal/inmunología , Ratones , Infecciones por Salmonella/microbiología , Salmonella typhimurium/fisiología
3.
Mol Cell ; 84(13): 2436-2454.e10, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38925114

RESUMEN

Signal transduction proteins containing a pLxIS motif induce interferon (IFN) responses central to antiviral immunity. Apart from their established roles in activating the IFN regulator factor (IRF) transcription factors, the existence of additional pathways and functions associated with the pLxIS motif is unknown. Using a synthetic biology-based platform, we identified two orphan pLxIS-containing proteins that stimulate IFN responses independent of all known pattern-recognition receptor pathways. We further uncovered a diversity of pLxIS signaling mechanisms, where the pLxIS motif represents one component of a multi-motif signaling entity, which has variable functions in activating IRF3, the TRAF6 ubiquitin ligase, IκB kinases, mitogen-activated protein kinases, and metabolic activities. The most diverse pLxIS signaling mechanisms were associated with the highest antiviral activities in human cells. The flexibility of domains that regulate IFN signaling may explain their prevalence in nature.


Asunto(s)
Factor 3 Regulador del Interferón , Interferones , Transducción de Señal , Factor 6 Asociado a Receptor de TNF , Humanos , Interferones/metabolismo , Células HEK293 , Factor 3 Regulador del Interferón/metabolismo , Factor 3 Regulador del Interferón/genética , Factor 6 Asociado a Receptor de TNF/metabolismo , Factor 6 Asociado a Receptor de TNF/genética , Quinasa I-kappa B/metabolismo , Quinasa I-kappa B/genética , Dominios Proteicos , Animales , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Secuencias de Aminoácidos , Proteínas Quinasas Activadas por Mitógenos/metabolismo
4.
Immunity ; 53(2): 277-289, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32814026

RESUMEN

The steep rise in food allergy (FA) has evoked environmental factors involved in disease pathogenesis, including the gut microbiota, diet, and their metabolites. Early introduction of solid foods synchronizes with the "weaning reaction," a time during which the microbiota imprints durable oral tolerance. Recent work has shown that children with FA manifest an early onset dysbiosis with the loss of Clostridiales species, which promotes the differentiation of ROR-γt+ regulatory T cells to suppress FA. This process can be reversed in pre-clinical mouse models by targeted bacteriotherapy. Here, we review the dominant tolerance mechanisms enforced by the microbiota to suppress FA and discuss therapeutic intervention strategies that act to recapitulate the early life window of opportunity in stemming the FA epidemic.


Asunto(s)
Dieta , Disbiosis/microbiología , Hipersensibilidad a los Alimentos/inmunología , Microbioma Gastrointestinal/fisiología , Animales , Clostridiales/aislamiento & purificación , Desensibilización Inmunológica/métodos , Humanos , Tolerancia Inmunológica/inmunología , Inmunoglobulina E/inmunología , Ratones , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología
5.
Immunity ; 51(2): 367-380.e4, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31350179

RESUMEN

Epithelial barrier defects are implicated in the pathogenesis of inflammatory bowel disease (IBD); however, the role of microbiome dysbiosis and the cytokine networks orchestrating chronic intestinal inflammation in response to barrier impairment remain poorly understood. Here, we showed that altered Schaedler flora (ASF), a benign minimal microbiota, was sufficient to trigger colitis in a mouse model of intestinal barrier impairment. Colitis development required myeloid-cell-specific adaptor protein MyD88 signaling and was orchestrated by the cytokines IL-12, IL-23, and IFN-γ. Colon inflammation was driven by IL-12 during the early stages of the disease, but as the mice aged, the pathology shifted toward an IL-23-dependent inflammatory response driving disease chronicity. These findings reveal that IL-12 and IL-23 act in a temporally distinct, biphasic manner to induce microbiota-driven chronic intestinal inflammation. Similar mechanisms might contribute to the pathogenesis of IBD particularly in patients with underlying intestinal barrier defects.


Asunto(s)
Colitis/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Interleucina-12/metabolismo , Interleucina-23/metabolismo , Mucosa Intestinal/patología , Microbiota/inmunología , Animales , Enfermedad Crónica , Modelos Animales de Enfermedad , Humanos , Inflamación , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-12/genética , Interleucina-23/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/metabolismo , Transducción de Señal , Quimera por Trasplante
6.
Immunity ; 49(4): 695-708.e4, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30291027

RESUMEN

B cells can present antigens to CD4+ T cells, but it is thought that dendritic cells (DCs) are the primary initiators of naive CD4+ T cell responses. Nanoparticles, including virus-like particles (VLPs), are attractive candidates as carriers for vaccines and drug delivery. Using RNA phage Qß-derived VLP (Qß-VLP) as a model antigen, we found that antigen-specific B cells were the dominant antigen-presenting cells that initiated naive CD4+ T cell activation. B cells were sufficient to induce T follicular helper cell development in the absence of DCs. Qß-specific B cells promoted CD4+ T cell proliferation and differentiation via cognate interactions and through Toll-like receptor signaling-mediated cytokine production. Antigen-specific B cells were also involved in initiating CD4+ T cell responses during immunization with inactivated influenza virus. These findings have implications for the rational design of nanoparticles as vaccine candidates, particularly for therapeutic vaccines that aim to break immune tolerance.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Inmunización/métodos , Vacunas contra la Influenza/inmunología , Animales , Presentación de Antígeno/inmunología , Antígenos Virales/química , Antígenos Virales/inmunología , Diferenciación Celular/inmunología , Citocinas/inmunología , Citocinas/metabolismo , Subtipo H1N1 del Virus de la Influenza A/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Nanopartículas/química , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Receptores Toll-Like/inmunología , Vacunas de Productos Inactivados/inmunología
7.
Trends Immunol ; 44(3): 153-155, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36740514

RESUMEN

Activation of Toll-like receptor 7 (TLR7) can induce lupus in mice, whereas activation of TLR9 can prevent it, even though both receptors interact with myeloid differentiation primary response gene 88 (MyD88) for downstream signaling. How TLR9 triggers anti-inflammatory responses in autoimmunity is unclear. Leibler et al. recently reported that TLR9 initiates anti-inflammatory signaling and inhibits lupus pathogenesis in a MyD88-independent but ligand-dependent manner.


Asunto(s)
Factor 88 de Diferenciación Mieloide , Receptor Toll-Like 9 , Ratones , Animales , Receptor Toll-Like 9/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Ratones Noqueados , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo , Antiinflamatorios
8.
Immunity ; 46(4): 635-648, 2017 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-28410990

RESUMEN

Mice carrying a hypomorphic point mutation in the Ptpn6 gene (Ptpn6spin mice) develop an inflammatory skin disease that resembles neutrophilic dermatosis in humans. Here, we demonstrated that interleukin-1α (IL-1α) signaling through IL-1R and MyD88 in both stromal and immune cells drive inflammation in Ptpn6spin mice. We further identified SYK as a critical kinase that phosphorylates MyD88, promoted MyD88-dependent signaling and mediates dermatosis in Ptpn6spin mice. Our studies further demonstrated that SHP1 encoded by Ptpn6 binds and suppresses SYK activation to inhibit MyD88 phosphorylation. Downstream of SHP1 and SYK-dependent counterregulation of MyD88 tyrosine phosphorylation, we have demonstrated that the scaffolding function of receptor interacting protein kinase 1 (RIPK1) and tumor growth factor-ß activated kinase 1 (TAK1)-mediating signaling were required to spur inflammatory disease. Overall, these studies identify SHP1 and SYK crosstalk as a critical regulator of MyD88 post-translational modifications and IL-1-driven inflammation.


Asunto(s)
Inflamación/inmunología , Interleucina-1alfa/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Enfermedades de la Piel/inmunología , Quinasa Syk/inmunología , Animales , Citometría de Flujo , Células HEK293 , Humanos , Immunoblotting , Inflamación/genética , Inflamación/metabolismo , Interleucina-1alfa/genética , Interleucina-1alfa/metabolismo , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/inmunología , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones Noqueados , Modelos Inmunológicos , Mutación , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/inmunología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Receptores de Interleucina-1/inmunología , Receptores de Interleucina-1/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Enfermedades de la Piel/genética , Enfermedades de la Piel/metabolismo , Quinasa Syk/genética , Quinasa Syk/metabolismo
9.
Development ; 149(8)2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-34528666

RESUMEN

B cell participation in early embryo/fetal development and the underlying molecular pathways have not been explored. To understand whether maternal B cell absence or impaired signaling interferes with placental and fetal growth, we paired CD19-deficient (CD19-/-) mice, females with B cell-specific MyD88 (BMyD88-/-) or IL10 (BIL10-/-) deficiency as well as wild-type and MyD88-/- controls on C57Bl/6 background with BALB/c males. Pregnancies were followed by ultrasound and Doppler measurements. Implantation number was reduced in BMyD88-/- and MyD88-/- mice. Loss of MyD88 or B cell-specific deletion of MyD88 or IL10 resulted in decreased implantation areas at gestational day (gd) 5, gd8 and gd10, accompanied by reduced placental thickness, diameter and areas at gd10. Uterine artery resistance was enhanced in BIL10-/- dams at gd10. Challenge with 0.4 mg lipopolysaccharide/kg bodyweight at gd16 revealed that BMyD88-/-, BIL10-/- and CD19-/- mothers delivered preterm, whereas controls maintained their pregnancy. B cell-specific MyD88 and IL10 expression is essential for appropriate in utero development. IL10+B cells are involved in uterine blood flow regulation during pregnancy. Finally, B cell-specific CD19, MyD88 and IL10 expression influences susceptibility towards preterm birth.


Asunto(s)
Linfocitos B/metabolismo , Desarrollo Fetal , Feto/embriología , Transducción de Señal , Arteria Uterina/metabolismo , Útero , Resistencia Vascular , Animales , Antígenos CD19/genética , Antígenos CD19/metabolismo , Femenino , Interleucina-10/deficiencia , Interleucina-10/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/metabolismo , Embarazo , Útero/irrigación sanguínea , Útero/metabolismo
10.
J Virol ; 98(6): e0043424, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38690875

RESUMEN

The globally reemerging respiratory pathogen enterovirus D68 (EV-D68) is implicated in outbreaks of severe respiratory illness and associated with acute flaccid myelitis. However, there remains a lack of effective treatments for EV-D68 infection. In this work, we found that the host Toll-like receptor 7 (TLR7) proteins, which function as powerful innate immune sensors, were selectively elevated in expression in response to EV-D68 infection. Subsequently, we investigated the impact of Vesatolimod (GS-9620), a Toll-like receptor 7 agonist, on EV-D68 replication. Our findings revealed that EV-D68 infection resulted in increased mRNA levels of TLR7. Treatment with Vesatolimod significantly inhibited EV-D68 replication [half maximal effective concentration (EC50) = 0.1427 µM] without inducing significant cytotoxicity at virucidal concentrations. Although Vesatolimod exhibited limited impact on EV-D68 attachment, it suppressed RNA replication and viral protein synthesis after virus entry. Vesatolimod broadly inhibited the replication of circulating isolated strains of EV-D68. Furthermore, our findings demonstrated that treatment with Vesatolimod conferred resistance to both respiratory and neural cells against EV-D68 infection. Overall, these results present a promising strategy for drug development by pharmacologically activating TLR7 to initiate an antiviral state in EV-D68-infected cells selectively.IMPORTANCEConventional strategies for antiviral drug development primarily focus on directly targeting viral proteases or key components, as well as host proteins involved in viral replication. In this study, based on our intriguing discovery that enterovirus D68 (EV-D68) infection specifically upregulates the expression of immune sensor Toll-like receptor 7 (TLR7) protein, which is either absent or expressed at low levels in respiratory cells, we propose a potential antiviral approach utilizing TLR7 agonists to activate EV-D68-infected cells into an anti-viral defense state. Notably, our findings demonstrate that pharmacological activation of TLR7 effectively suppresses EV-D68 replication in respiratory tract cells through a TLR7/MyD88-dependent mechanism. This study not only presents a promising drug candidate and target against EV-D68 dissemination but also highlights the potential to exploit unique alterations in cellular innate immune responses induced by viral infections, selectively inducing a defensive state in infected cells while safeguarding uninfected normal cells from potential adverse effects associated with therapeutic interventions.


Asunto(s)
Antivirales , Enterovirus Humano D , Receptor Toll-Like 7 , Replicación Viral , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 7/metabolismo , Humanos , Replicación Viral/efectos de los fármacos , Enterovirus Humano D/efectos de los fármacos , Antivirales/farmacología , Indoles/farmacología , Infecciones por Enterovirus/virología , Inmunidad Innata/efectos de los fármacos , Línea Celular , Internalización del Virus/efectos de los fármacos , Pteridinas
11.
Traffic ; 23(12): 558-567, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36224049

RESUMEN

Intracellular compartmentalization of ligands, receptors and signaling molecules has been recognized as an important regulator of inflammation. The toll-like receptor (TLR) 2 pathway utilizes the trafficking molecule adaptor protein 3 (AP-3) to activate interleukin (IL)-6 signaling from within phagosomal compartments. To better understand the vesicular pathways that may contribute to intracellular signaling and cooperate with AP-3, we performed a vesicular siRNA screen. We identified Rab8 and Rab11 GTPases as important in IL-6 induction upon stimulation with the TLR2 ligand Pam3 CSK4 or the pathogen, Borrelia burgdorferi (Bb), the causative agent of Lyme disease. These Rabs were recruited to late and lysosomal stage phagosomes and co-transported with TLR2 signaling adaptors and effectors, such as MyD88, TRAM and TAK1, in an AP-3-dependent manner. Our data support a model where AP-3 mediates the recruitment of recycling and secretory vesicles and the assembly of signaling complexes at the phagosome.


Asunto(s)
Borrelia burgdorferi , Enfermedad de Lyme , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Borrelia burgdorferi/metabolismo , Ligandos , Enfermedad de Lyme/genética , Enfermedad de Lyme/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Fagosomas/metabolismo , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Proteínas de Unión al GTP rab , Animales , Ratones
12.
J Biol Chem ; 299(8): 104803, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37172723

RESUMEN

Interleukin-1ß is one of the most potent inducers of beta cell inflammation in the lead-up to type 1 diabetes. We have previously reported that IL1ß-stimulated pancreatic islets from mice with genetic ablation of stress-induced pseudokinase TRB3(TRB3KO) show attenuated activation kinetics for the MAP3K MLK3 and JNK stress kinases. However, JNK signaling constitutes only a portion of the cytokine-induced inflammatory response. Here we report that TRB3KO islets also show a decrease in amplitude and duration of IL1ß-induced phosphorylation of TAK1 and IKK, kinases that drive the potent NF-κB proinflammatory signaling pathway. We observed that TRB3KO islets display decreased cytokine-induced beta cell death, preceded by a decrease in select downstream NF-κB targets, including iNOS/NOS2 (inducible nitric oxide synthase), a mediator of beta cell dysfunction and death. Thus, loss of TRB3 attenuates both pathways required for a cytokine-inducible, proapoptotic response in beta cells. In order to better understand the molecular basis of TRB3-enhanced, post-receptor IL1ß signaling, we interrogated the TRB3 interactome using coimmunoprecipitation followed by mass spectrometry to identify immunomodulatory protein Flightless homolog 1 (Fli1) as a novel, TRB3-interacting protein. We show that TRB3 binds and disrupts Fli1-dependent sequestration of MyD88, thereby increasing availability of this most proximal adaptor required for IL1ß receptor-dependent signaling. Fli1 sequesters MyD88 in a multiprotein complex resulting in a brake on the assembly of downstream signaling complexes. By interacting with Fli1, we propose that TRB3 lifts the brake on IL1ß signaling to augment the proinflammatory response in beta cells.


Asunto(s)
Proteínas de Ciclo Celular , Interleucina-1beta , Transducción de Señal , Animales , Ratones , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Citocinas/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Transducción de Señal/genética , Inhibidores Enzimáticos/farmacología , Apoptosis/efectos de los fármacos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/fisiología , Activación Transcripcional/genética
13.
Angiogenesis ; 2024 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-39316206

RESUMEN

Inflammasome activation is implicated in diseases of aberrant angiogenesis such as age-related macular degeneration (AMD), though its precise role in choroidal neovascularization (CNV), a characteristic pathology of advanced AMD, is ill-defined. Reports on inhibition of inflammasome constituents on CNV are variable and the precise role of inflammasome in mediating pathological angiogenesis is unclear. Historically, subretinal injection of inflammasome agonists alone has been used to investigate retinal pigmented epithelium (RPE) degeneration, while the laser photocoagulation model has been used to study pathological angiogenesis in a model of CNV. Here, we report that the simultaneous introduction of any of several disease-relevant inflammasome agonists (Alu or B2 RNA, Alu cDNA, or oligomerized amyloid ß (1-40)) exacerbates laser-induced CNV. These activities were diminished or abrogated by genetic or pharmacological targeting of inflammasome signaling constituents including P2rx7, Nlrp3, caspase-1, caspase-11, and Myd88, as well as in myeloid-specific caspase-1 knockout mice. Alu RNA treatment induced inflammasome activation in macrophages within the CNV lesion, and increased accumulation of macrophages in an inflammasome-dependent manner. Finally, IL-1ß neutralization prevented inflammasome agonist-induced chemotaxis, macrophage trafficking, and angiogenesis. Collectively, these observations support a model wherein inflammasome stimulation promotes and exacerbates CNV and may be a therapeutic target for diseases of angiogenesis such as neovascular AMD.

14.
Clin Immunol ; 266: 110327, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39053866

RESUMEN

This study retrospectively investigated the impact of interleukin-1 receptor-associated kinase-3 (IRAK-3/IRAK-M) silencing by methylation on the likelihood of multiple sclerosis (MS) activity. This cross-sectional study included 90 patients with MS: 45 with active disease (Group 1), 45 in remission (Group 2), and 45 healthy controls. The study included quantitation of IRAK-3 methylation index (MI%), IRAK-3 mRNA, and myeloid differentiation factor88 (MyD88) and assessment of NF-κB activity. IRAK-3 MI% was significantly higher in group 1 compared to group 2, accompanied by lower IRAK-3 mRNA expression, elevated circulating MyD88, and increased NF-κB activity. IRAK-3 MI% correlated negatively with its transcript and positively with MyD88 and NF-κB activity. A logistic regression model was created to predict active demyelination. The C-index was 0.924, which indicates a very strong prediction model. Within the limitations of current work, IRAK-3 methylation level seems to be a promising candidate biomarker for identifying MS patients at risk of relapse.


Asunto(s)
Quinasas Asociadas a Receptores de Interleucina-1 , Esclerosis Múltiple , Factor 88 de Diferenciación Mieloide , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/genética , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Femenino , Masculino , Adulto , Esclerosis Múltiple/genética , Esclerosis Múltiple/sangre , Esclerosis Múltiple/inmunología , Factor 88 de Diferenciación Mieloide/genética , Persona de Mediana Edad , Estudios Transversales , FN-kappa B/metabolismo , FN-kappa B/genética , Recurrencia , Estudios Retrospectivos , Metilación de ADN , Biomarcadores/sangre , ARN Mensajero/genética , ARN Mensajero/metabolismo , Adulto Joven
15.
Br J Haematol ; 2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39295138

RESUMEN

Covalent Bruton's tyrosine kinase-inhibitors (cBTK-i) are highly active in MYD88-mutated (MYD88Mut) Waldenstrom's macroglobulinaemia and suppress nuclear factor kappa-light-chain-enhancer of activated B cells and extracellular signal-regulated kinases-1/2 (ERK1/2)-related signalling. BTKCys481 mutations are associated with cBTK-i acquired resistance and are accompanied by reactivation of ERK1/2 that promotes inflammatory cytokine secretion and paracrine-mediated resistance of BTK wild-type (BTKWT) tumour cells. Pirtobrutinib is a non-covalent BTK-inhibitor that binds at non-BTKCys481 sites. We show that pirtobrutinib blocked p-ERK1/2, ERK1/2-driven inflammatory cytokines, and overcame paracrine-mediated resistance in MYD88Mut lymphoma cells expressing mutated BTKCys481. Our results provide important mechanistic insights for the activity of pirtobrutinib in MYD88Mut lymphomas carrying BTKCys481 mutations.

16.
Biochem Biophys Res Commun ; 739: 150569, 2024 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-39186869

RESUMEN

Lactation mastitis is a debilitating inflammatory mammary disease in postpartum animals. Myeloid differentiation primary response protein MyD88 is the key downstream adapter for innate pattern recognition receptor toll-like receptor 4 (TLR4), which plays an important role in inflammation. However, the specific role of MyD88 in mammary epithelial cells in the progression of mastitis has not been investigated. In this study, lipopolysaccharide (LPS)-induced mouse mastitis model was used and cytokines such as Tnf-α, Il-1ß, Il-6, Cxcl1, Cxcl2 and Ccl2 were significantly increased in inflammatory mammary gland as shown by real time-qPCR. However, the mice with MyD88-deficienet in mammary epithelial cells (cKO) showed a reduction in the expression of Tnf-α, Il-1ß, Il-6, Cxcl1 and Cxcl2 in mammary gland compared with control mice, when subjected to LPS induced mastitis. Immunohistochemical staining of cleaved caspase-3 showed that the cell apoptosis induced by inflammation were decreased in MyD88 cKO mice. Furthermore, there were significantly fewer infiltrating inflammatory cells in alveolar lumen of MyD88 cKO mice, including Ly6G-positive neutrophils and F4/80-positive macrophages. RNA-seq in LPS treated mammary glands showed that MyD88 cKO mice had significantly downregulated inflammation-related genes and upregulated genes related to anti-inflammation processes and lipid metabolism compared with control mice. Thus, these results demonstrate that MyD88 in mammary epithelial cells is essential for mastitis progression. And this study not only has important implications for understanding the innate immune response in mammary epithelial cells, but also potentially helps the development of new therapeutic drugs for treating mastitis.

17.
Biochem Biophys Res Commun ; 705: 149756, 2024 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-38460440

RESUMEN

Exacerbated expression of TLR4 protein (foremost pattern recognition receptor) during obesity could trigger NF-κB/iNOS signaling through linker protein (MyD88), predisposed to an indispensable inflammatory response. The induction of this detrimental cascade leads to myocardial and vascular abnormalities. Molecular docking was studied for protein-ligand interaction between these potential targets and resveratrol. The pre-treatment of resveratrol (20 mg/kg/p.o/per day for ten weeks) was given to investigate the therapeutic effect against HFD-induced obesity and associated vascular endothelial dysfunction (VED) and myocardial infarction (MI) in Wistar rats. In addition to accessing the levels of serum biomarkers for VED and MI, oxidative stress, inflammatory cytokines, and histopathology of these tissues were investigated. Lipopolysaccharide (for receptor activation) and protein expression analysis were introduced to explore the mechanistic involvement of TLR4/MyD88/NF-κB/iNOS signaling. Assessment of in-silico analysis showed significant interaction between protein and ligand. The involvement of this proposed signaling (TLR4/MyD88/NF-κB/iNOS) was further endorsed by the impact of lipopolysaccharide and protein expression analysis in obese and treated rats. Moreover, resveratrol pre-treated rats showed significantly lowered cardio and vascular damage measured by the distinct down expression of the TLR4/MyD88/NF-κB/iNOS pathway by resveratrol treatment endorses its ameliorative effect against VED and MI.


Asunto(s)
Infarto del Miocardio , Estilbenos , Ratas , Animales , FN-kappa B/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Receptor Toll-Like 4/metabolismo , Resveratrol/farmacología , Estilbenos/farmacología , Estilbenos/uso terapéutico , Lipopolisacáridos/farmacología , Ligandos , Simulación del Acoplamiento Molecular , Ratas Wistar , Infarto del Miocardio/tratamiento farmacológico , Dieta
18.
Cell Tissue Res ; 395(3): 251-260, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38291253

RESUMEN

Osteoarthritis (OA) is one of the most common diseases of the skeleton. Long non-coding RNAs (lncRNAs) are emerging as key players in OA pathogenesis. This work sets out to determine the function of lncRNA MALAT1 in OA and the mechanisms by which it does so. Mesenchymal stem cells isolated from the human synovial membrane are called hSMSCs. The hSMSCs' surface markers were studied using flow cytometry. To determine whether or not hSMSC might differentiate, researchers used a number of different culture settings and labeling techniques. The expression levels of associated genes and proteins were determined using quantitative real-time polymerase chain reaction (RT-qPCR), western blotting (WB), and immunostaining. A dual luciferase reporter experiment and RNA immunoprecipitation (RIP) test demonstrated the direct association between miR-212-5p and MALAT1 or MyD88. MALAT1 was downregulated during the chondrogenic differentiation of hSMSCs, and underexpression of MALAT1 promotes chondrogenesis in hSMSCs. Using dual luciferase reporter and RIP assays facilitated the identification of MALAT1 as a competitive endogenous RNA (ceRNA) that sequesters miR-212-5p. Additionally, the expression of MYD88 was regulated by MALAT1 through direct binding with miR-212-5p. Significantly, the effects of MALAT1 on the chondrogenic differentiation of hSMSCs were counteracted by miR-212-5p/MYD88. Furthermore, our in vivo investigation revealed that the inhibition of MALAT1 mitigated osteoarthritis progression in rat models. In conclusion, the promotion of chondrogenic differentiation in hSMSCs and the protective effect on cartilage tissue in OA can be achieved by suppressing MALAT1, which regulates the miR-212-5p/MyD88 axis.


Asunto(s)
Células Madre Mesenquimatosas , MicroARNs , Osteoartritis de la Rodilla , ARN Largo no Codificante , Animales , Humanos , Ratas , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Condrogénesis , Luciferasas/metabolismo , Células Madre Mesenquimatosas/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
19.
Am J Physiol Regul Integr Comp Physiol ; 327(4): R410-R422, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39133777

RESUMEN

Inflammation and fibrosis play important roles in diabetic kidney disease (DKD). Previous studies have shown that glucagon-like peptide-1 receptor (GLP-1R) agonists had renal protective effects. However, the mechanisms are not clear. The present study explored the effect of liraglutide (LR), a GLP-1R agonist, on the downregulation of glomerular inflammation and fibrosis in DKD by regulating the Toll-like receptor (TLR)4/myeloid differentiation marker 88 (MyD88)/nuclear factor κB (NF-κB) signaling pathway in mesangial cells (MCs). In vitro, rat MCs were cultured in high glucose (HG). We found that liraglutide treatment significantly reduced the HG-mediated activation of the TLR4/MYD88/NF-κB signaling pathway, extracellular matrix (ECM)-related proteins, and inflammatory factors. A combination of TLR4 inhibitor (TAK242) and liraglutide did not synergistically inhibit inflammatory factors and ECM proteins. Furthermore, in the presence of TLR4 siRNA, liraglutide significantly blunted HG-induced expression of fibronectin protein and inflammatory factors. Importantly, TLR4 selective agonist LPS or TLR4 overexpression eliminated the improvement effects of liraglutide on the HG-induced response. In vivo, administration of liraglutide for 8 wk significantly improved the glomerular damage in streptozotocin-induced diabetic mice and reduced the expression of TLR4/MYD88/NF-κB signaling proteins, ECM protein, and inflammatory factors in renal cortex. TLR4-/- diabetic mice showed significant amelioration in urine protein excretion rate, glomerular pathological damage, inflammation, and fibrosis. Liraglutide attenuated glomerular hypertrophy, renal fibrosis, and inflammatory response in TLR4-/- diabetic mice. Taken together, our findings suggest that TLR4/MYD88/NF-κB signaling is involved in the regulation of inflammatory response and ECM protein proliferation in DKD. Liraglutide alleviates inflammation and fibrosis by downregulating the TLR4/MYD88/NF-κB signaling pathway in MCs.NEW & NOTEWORTHY Liraglutide, a glucagon-like peptide-1 receptor agonist (GLP-1RA), has renoprotective effect in diabetic kidney disease (DKD). In DKD, TLR4/MYD88/NF-κB signaling is involved in the regulation of inflammatory responses and extracellular matrix (ECM) protein proliferation. Liraglutide attenuates renal inflammation and overexpression of ECM proteins by inhibiting TLR4/MYD88/NF-κB signaling pathway. Therefore, we have identified a new mechanism that contributes to the renal protection of GLP-1RA, thus helping to design innovative treatment strategies for diabetic patients with various complications.


Asunto(s)
Diabetes Mellitus Experimental , Nefropatías Diabéticas , Fibrosis , Liraglutida , Factor 88 de Diferenciación Mieloide , FN-kappa B , Transducción de Señal , Receptor Toll-Like 4 , Animales , Liraglutida/farmacología , Liraglutida/uso terapéutico , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 4/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/metabolismo , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Masculino , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/complicaciones , Ratones , Ratones Endogámicos C57BL , Ratas , Regulación hacia Abajo/efectos de los fármacos , Ratas Sprague-Dawley , Células Mesangiales/efectos de los fármacos , Células Mesangiales/metabolismo , Células Mesangiales/patología , Ratones Noqueados , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico
20.
Toxicol Appl Pharmacol ; 487: 116958, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38735591

RESUMEN

Acute lung injury (ALI) remains a significant clinical challenge due to the absence of effective treatment alternatives. This study presents a new method that employs a screening platform focusing on MyD88 affinity, anti-inflammatory properties, and toxicity. This platform was used to evaluate a 300-compound library known for its anti-inflammatory potential. Among the screened compounds, Bicyclol emerged as a standout, exhibiting MyD88 binding and a significant reduction in LPS-stimulated pro-inflammatory factors production in mouse primary peritoneal macrophages. By targeting MyD88, Bicyclol disrupts the MyD88/TLR4 complex and MyD88 polymer formation, thereby mitigating the MAPKs and NF-κB signaling pathways. In vivo experiments further confirmed Bicyclol's efficacy, demonstrating alleviated ALI symptoms, decreased inflammatory cytokines level, and reduced inflammatory cells presence in lung tissues. These findings were associated with a decrease in mortality in LPS-challenged mice. Overall, Bicyclol represents a promising treatment option for ALI by specifically targeting MyD88 and limiting inflammatory responses.


Asunto(s)
Lesión Pulmonar Aguda , Compuestos de Bifenilo , Lipopolisacáridos , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide , Animales , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/prevención & control , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Lipopolisacáridos/toxicidad , Factor 88 de Diferenciación Mieloide/metabolismo , Ratones , Masculino , Compuestos de Bifenilo/farmacología , Antiinflamatorios/farmacología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/metabolismo , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Citocinas/metabolismo , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA