Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 185
Filtrar
1.
Mol Biol Rep ; 51(1): 380, 2024 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-38429584

RESUMEN

BACKGROUND: Interferon regulatory factor 6 (IRF6) has a key function in palate fusion during palatogenesis during embryonic development, and mutations in IRF6 cause orofacial clefting disorders. METHODS AND RESULTS: The in silico analysis of IRF6 is done to obtain leads for the domain boundaries and subsequently the sub-cloning of the N-terminal domain of IRF6 into the pGEX-2TK expression vector and successfully optimized the overexpression and purification of recombinant glutathione S-transferase-fused NTD-IRF6 protein under native conditions. After cleavage of the GST tag, NTD-IRF6 was subjected to protein folding studies employing Circular Dichroism and Intrinsic fluorescence spectroscopy at variable pH, temperature, and denaturant. CD studies showed predominantly alpha-helical content and the highest stability of NTD-IRF6 at pH 9.0. A comparison of native and renatured protein depicts loss in the secondary structural content. Intrinsic fluorescence and quenching studies have identified that tryptophan residues are majorly present in the buried areas of the protein and a small fraction was on or near the protein surface. Upon the protein unfolding with a higher concentration of denaturant urea, the peak of fluorescence intensity decreased and red shifted, confirming that tryptophan residues are majorly present in a more polar environment. While regulating IFNß gene expression during viral infection, the N-terminal domain binds to the promoter region of Virus Response Element-Interferon beta (VRE-IFNß). Along with the protein folding analysis, this study also aimed to identify the DNA-binding activity and determine the binding affinities of NTD-IRF6 with the VRE-IFNß promoter region. The protein-DNA interaction is specific as demonstrated by gel retardation assay and the kinetics of molecular interactions as quantified by Biolayer Interferometry showed a strong affinity with an affinity constant (KD) value of 7.96 × 10-10 M. CONCLUSION: NTD-IRF6 consists of a mix of α-helix and ß-sheets that show temperature-dependent cooperative unfolding between 40 °C and 55 °C. Urea-induced unfolding shows moderate tolerance to urea as the mid-transition concentration of urea (Cm) is 3.2 M. The tryptophan residues are majorly buried as depicted by fluorescence quenching studies. NTD-IRF6 has a specific and high affinity toward the promoter region of VRE-IFNß.


Asunto(s)
Factores Reguladores del Interferón , Pliegue de Proteína , Triptófano , Humanos , ADN , Factores Reguladores del Interferón/metabolismo , Factores Reguladores del Interferón/fisiología , Triptófano/metabolismo , Urea
2.
Int J Med Sci ; 18(16): 3794-3799, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34790055

RESUMEN

Psoriasis is a chronic inflammatory disease that involves both the innate and adaptive immune systems. Type I interferons (IFNs), the production of which is partially regulated by toll-like receptors (TLRs), play an important role in the pathogenesis of psoriasis, especially psoriasis caused by skin trauma, known as the Koebner phenomenon. IFN regulatory factors (IRFs) function in both innate and adaptive immune responses, and their effect is associated with the regulation of type I IFNs. In this review, we focus on recent advances in understanding the expression of TLRs, IRFs, and type I IFNs in psoriasis. We also highlight the interplay among TLRs, IRFs, and type I IFNs.


Asunto(s)
Factores Reguladores del Interferón/fisiología , Psoriasis/metabolismo , Animales , Humanos , Inmunidad Innata/fisiología , Interferón Tipo I/metabolismo , Psoriasis/inmunología , Psoriasis/patología , Transducción de Señal/fisiología , Receptores Toll-Like/metabolismo
3.
J Heart Lung Transplant ; 40(10): 1122-1132, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34253454

RESUMEN

BACKGOUND: B cells contribute to chronic transplant rejection by producing donor-specific antibodies and promoting T cell response, but how these processes are regulated at the transcriptional level remains unclear. Herein, we investigate the role of transcription factor interferon regulatory factor 4 (IRF4) in controlling B cell response during chronic transplant rejection. METHODS: We generated the Irf4gfp reporter mice to determine IRF4 expression in B cell lineage. We then used mice with B cell-specific IRF4 deletion to define the role of IRF4 in B cell response after NP-KLH immunization or allogeneic heart transplantation. In particular, graft survival and histology, as well as B and T cell responses, were evaluated after transplantation. RESULTS: IRF4 is dynamically expressed at different stages of B cell development and is absent in germinal center (GC) B cells. However, IRF4 ablation in the B cell lineage primarily eliminates GC B cells in both naïve and NP-KLH immunized mice. In the transplantation setting, IRF4 functions intrinsically in B cells and governs allogeneic B cell responses at multiple levels, including GC B cell generation, plasma cell differentiation, donor-specific antibody production, and support of T cell response. B cell-specific IRF4 deletion combined with transient CTLA4-Ig treatment abrogates acute and chronic cardiac allograft rejection in naïve recipient mice but not in donor skin-sensitized recipients. CONCLUSIONS: B cells require IRF4 to mediate chronic transplant rejection. IRF4 ablation in B cells abrogates allogeneic B cell responses and may also inhibit the ability of B cells to prime allogenic T cells. Targeting IRF4 in B cells represents a potential therapeutic strategy for eliminating chronic transplant rejection.


Asunto(s)
Linfocitos B/fisiología , Rechazo de Injerto/etiología , Rechazo de Injerto/prevención & control , Trasplante de Corazón/efectos adversos , Factores Reguladores del Interferón/fisiología , Trasplante de Piel/efectos adversos , Animales , Modelos Animales de Enfermedad , Centro Germinal/metabolismo , Supervivencia de Injerto , Haptenos , Hemocianinas , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados
4.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33859042

RESUMEN

The transcription factor IRF4 is required for CD8+ T cell activation, proliferation, and differentiation to effector cells and thus is essential for robust CD8+ T cell responses. The function of IRF4 in memory CD8+ T cells yet needs to be explored. To investigate the role of IRF4 for maintaining differentiation state and survival of CD8+ memory T cells, we used a mouse model with tamoxifen-inducible Irf4 knockout to preclude effects due to inefficient memory cell differentiation in absence of IRF4. We infected mice with ovalbumin-recombinant listeria and induced Irf4 knockout after clearance of the pathogen. Loss of IRF4 resulted in phenotypical changes of CD8+ memory T cells but did not cause a reduction of the total memory T cell population. However, upon reencounter of the pathogen, CD8+ memory T cells showed impaired expansion and acquisition of effector functions. When compared to CD8+ effector memory T cells, CD8+ tissue-resident memory T cells (TRM cells) expressed higher IRF4 levels. Mice with constitutive Irf4 knockout had diminished CD8+ TRM-cell populations, and tamoxifen-induced Irf4 deletion caused a reduction of this cell population. In conclusion, our results demonstrate that IRF4 is required for effective reactivation but not for general survival of CD8+ memory T cells. Formation and maintenance of CD8+ TRM cells, in contrast, appear to depend on IRF4.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Memoria Inmunológica/fisiología , Factores Reguladores del Interferón/metabolismo , Animales , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Proliferación Celular , Femenino , Memoria Inmunológica/genética , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/fisiología , Listeria monocytogenes/patogenicidad , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
5.
Front Immunol ; 12: 627072, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33708218

RESUMEN

The accumulation of myeloid-derived suppressor cells (MDSCs) is one of the major obstacles to achieve an appropriate anti-tumor immune response and successful tumor immunotherapy. MDSCs in tumor-bearing hosts are primarily polymorphonuclear (PMN-MDSCs). However, the mechanisms regulating the development of MDSCs remain poorly understood. In this report, we showed that interferon regulatory factor 4 (IRF4) plays a key role in the development of PMN-MDSCs, but not monocytic MDSCs. IRF4 deficiency caused a significant elevation of PMN-MDSCs and enhanced the suppressive activity of PMN-MDSCs, increasing tumor growth and metastasis in mice. Mechanistic studies showed that c-Myc was up-regulated by the IRF4 protein. Over-expression of c-Myc almost abrogated the effects of IRF4 deletion on PMN-MDSCs development. Importantly, the IRF4 expression level was negatively correlated with the PMN-MDSCs frequency and tumor development but positively correlated with c-Myc expression in clinical cancer patients. In summary, this study demonstrated that IRF4 represents a novel regulator of PMN-MDSCs development in cancer, which may have predictive value for tumor progression.


Asunto(s)
Factores Reguladores del Interferón/fisiología , Células Supresoras de Origen Mieloide/fisiología , Neoplasias/inmunología , Proteínas Proto-Oncogénicas c-myc/genética , Transcripción Genética , Animales , Proliferación Celular , Femenino , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas c-myc/fisiología
6.
J Am Soc Nephrol ; 32(5): 1037-1052, 2021 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-33619052

RESUMEN

BACKGROUND: AKI is characterized by abrupt and reversible kidney dysfunction, and incomplete recovery leads to chronic kidney injury. Previous studies by us and others have indicated that macrophage infiltration and polarization play key roles in recovery from AKI. The role in AKI recovery played by IFN regulatory factor 4 (IRF4), a mediator of polarization of macrophages to the M2 phenotype, is unclear. METHODS: We used mice with myeloid or macrophage cell-specific deletion of Irf4 (MΦ Irf4-/- ) to evaluate Irf4's role in renal macrophage polarization and development of fibrosis after severe AKI. RESULTS: Surprisingly, although macrophage Irf4 deletion had a minimal effect on early renal functional recovery from AKI, it resulted in decreased renal fibrosis 4 weeks after severe AKI, in association with less-activated macrophages. Macrophage Irf4 deletion also protected against renal fibrosis in unilateral ureteral obstruction. Bone marrow-derived monocytes (BMDMs) from MΦ Irf4-/- mice had diminished chemotactic responses to macrophage chemoattractants, with decreased activation of AKT and PI3 kinase and increased PTEN expression. PI3K and AKT inhibitors markedly decreased chemotaxis in wild-type BMDMs, and in a cultured macrophage cell line. There was significant inhibition of homing of labeled Irf4-/- BMDMs to postischemic kidneys. Renal macrophage infiltration in response to AKI was markedly decreased in MΦ Irf4-/- mice or in wild-type mice with inhibition of AKT activity. CONCLUSIONS: Deletion of Irf4 from myeloid cells protected against development of tubulointerstitial fibrosis after severe ischemic renal injury in mice, due primarily to inhibition of AKT-mediated monocyte recruitment to the injured kidney and reduced activation and subsequent polarization into a profibrotic M2 phenotype.


Asunto(s)
Lesión Renal Aguda/etiología , Lesión Renal Aguda/patología , Factores Reguladores del Interferón/fisiología , Activación de Macrófagos/fisiología , Células Mieloides/metabolismo , Daño por Reperfusión/complicaciones , Lesión Renal Aguda/metabolismo , Animales , Modelos Animales de Enfermedad , Fibrosis , Masculino , Ratones , Ratones Endogámicos C57BL , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología
7.
PLoS Pathog ; 17(1): e1008299, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33465134

RESUMEN

Host resistance against intracellular pathogens requires a rapid IFN-γ mediated immune response. We reveal that T-bet-dependent production of IFN-γ is essential for the maintenance of inflammatory DCs at the site of infection with a common protozoan parasite, Toxoplasma gondii. A detailed analysis of the cellular sources for T-bet-dependent IFN-γ identified that ILC1s and to a lesser degree NK, but not TH1 cells, were involved in the regulation of inflammatory DCs via IFN-γ. Mechanistically, we established that T-bet dependent innate IFN-γ is critical for the induction of IRF8, an essential transcription factor for cDC1s. Failure to upregulate IRF8 in DCs resulted in acute susceptibility to T. gondii infection. Our data identifies that T-bet dependent production of IFN-γ by ILC1 and NK cells is indispensable for host resistance against intracellular infection via maintaining IRF8+ inflammatory DCs at the site of infection.


Asunto(s)
Células Dendríticas/inmunología , Inmunidad Innata/inmunología , Interferón gamma/metabolismo , Células Asesinas Naturales/inmunología , Linfocitos/inmunología , Proteínas de Dominio T Box/metabolismo , Toxoplasma/inmunología , Toxoplasmosis/inmunología , Animales , Células Dendríticas/metabolismo , Células Dendríticas/microbiología , Femenino , Factores Reguladores del Interferón/fisiología , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/microbiología , Linfocitos/metabolismo , Linfocitos/microbiología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Dominio T Box/genética , Toxoplasma/metabolismo , Toxoplasmosis/metabolismo , Toxoplasmosis/microbiología
8.
Elife ; 92020 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-33373293

RESUMEN

Bone remodeling involves a balance between bone resorption and formation. The mechanisms underlying bone remodeling are not well understood. DEF6 is recently identified as a novel loci associated with bone mineral density. However, it is unclear how Def6 impacts bone remodeling. We identify Def6 as a novel osteoblastic regulator that suppresses osteoblastogenesis and bone formation. Def6 deficiency enhances both bone resorption and osteogenesis. The enhanced bone resorption in Def6-/- mice dominates, leading to osteoporosis. Mechanistically, Def6 inhibits the differentiation of both osteoclasts and osteoblasts via a common mechanism through endogenous type-I IFN-mediated feedback inhibition. RNAseq analysis shows expression of a group of IFN stimulated genes (ISGs) during osteoblastogenesis. Furthermore, we found that Def6 is a key upstream regulator of IFNß and ISG expression in osteoblasts. Collectively, our results identify a novel immunoregulatory function of Def6 in bone remodeling, and shed insights into the interaction between immune system and bone.


Asunto(s)
Interferón gamma/fisiología , Osteoblastos/fisiología , Osteogénesis/fisiología , Animales , Regulación de la Expresión Génica , Factores Reguladores del Interferón/fisiología , Interferón gamma/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
9.
Front Immunol ; 11: 2110, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33042123

RESUMEN

Tumor necrosis factor receptor (TNFR)-associated factors (TRAFs) are vital signaling adaptor proteins for the innate immune response and are involved in many important pathways, such as the NF-κB- and interferon regulatory factor (IRF)-activated signaling pathways. In this study, the TRAF3 ortholog from the shrimp Litopenaeus vannamei (LvTRAF3) was cloned and characterized. LvTRAF3 has a transcript of 3,865 bp, with an open reading frame (ORF) of 1,002 bp and encodes a polypeptide of 333 amino acids, including a conserved TRAF-C domain. The expression of LvTRAF3 in the intestine and hemocyte was up-regulated in response to poly (I:C) challenge and white spot syndrome virus (WSSV) infection. RNAi knockdown of LvTRAF3 in vivo significantly increased WSSV gene transcription, viral loads, and mortality in WSSV-infected shrimp. Next, we found that LvTRAF3 was not able to induce the activation of the NF-κB pathway, which was crucial for synthesis of antimicrobial peptides (AMPs), which mediate antiviral immunity. Specifically, in dual-luciferase reporter assays, LvTRAF3 could not activate several types of promoters with NF-κB binding sites, including those from WSSV genes (wsv069, wsv056, and wsv403), Drosophila AMPs or shrimp AMPs. Accordingly, the mRNA levels of shrimp AMPs did not significantly change when TRAF3 was knocked down during WSSV infection. Instead, we found that LvTRAF3 signaled through the IRF-Vago antiviral cascade. LvTRAF3 functioned upstream of LvIRF to regulate the expression of LvVago4 and LvVago5 during WSSV infection in vivo. Taken together, these data provide experimental evidence of the participation of LvTRAF3 in the host defense to WSSV through the activation of the IRF-Vago pathway but not the NF-κB pathway.


Asunto(s)
Citocinas/fisiología , Factores Reguladores del Interferón/fisiología , Penaeidae/inmunología , Transducción de Señal/fisiología , Factor 3 Asociado a Receptor de TNF/fisiología , Virus del Síndrome de la Mancha Blanca 1/fisiología , Secuencia de Aminoácidos , Animales , Acuicultura , Secuencia de Bases , Línea Celular , Hemocitos/efectos de los fármacos , FN-kappa B/metabolismo , Penaeidae/virología , Filogenia , Interferencia de ARN , ARN Bicatenario/genética , ARN Bicatenario/farmacología , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Factor 3 Asociado a Receptor de TNF/antagonistas & inhibidores , Factor 3 Asociado a Receptor de TNF/biosíntesis , Factor 3 Asociado a Receptor de TNF/genética , Replicación Viral
10.
Life Sci Alliance ; 3(11)2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32958603

RESUMEN

Recent single-cell RNA-sequencing atlases have surveyed and identified major cell types across different mouse tissues. Here, we computationally reconstruct gene regulatory networks from three major mouse cell atlases to capture functional regulators critical for cell identity, while accounting for a variety of technical differences, including sampled tissues, sequencing depth, and author assigned cell type labels. Extracting the regulatory crosstalk from mouse atlases, we identify and distinguish global regulons active in multiple cell types from specialised cell type-specific regulons. We demonstrate that regulon activities accurately distinguish individual cell types, despite differences between individual atlases. We generate an integrated network that further uncovers regulon modules with coordinated activities critical for cell types, and validate modules using available experimental data. Inferring regulatory networks during myeloid differentiation from wild-type and Irf8 KO cells, we uncover functional contribution of Irf8 regulon activity and composition towards monocyte lineage. Our analysis provides an avenue to further extract and integrate the regulatory crosstalk from single-cell expression data.


Asunto(s)
Biología Computacional/métodos , Redes Reguladoras de Genes/genética , Redes Reguladoras de Genes/fisiología , Animales , Fenómenos Fisiológicos Celulares , Bases de Datos Factuales , Bases de Datos Genéticas , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Factores Reguladores del Interferón/fisiología , Ratones , Regulón/genética , Regulón/fisiología , Análisis de Secuencia de ARN/métodos , Factores de Transcripción/genética
11.
Front Immunol ; 11: 606, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32373114

RESUMEN

MyD88 is a conserved intracellular adaptor, which plays an important role in the innate immune system. MyD88 transmits signals for downstream of toll-like and IL-1 receptors to activate NF-κB signaling pathway, which is tightly controlled in the immune response to maintain immune intensity and immune homeostasis at different stages. NF-κB signaling pathway has been extensively studied in mammals, but regulatory molecular mechanism is still unclear in teleost fish. We determined that IRF3 and IRF8 can regulate MyD88-mediated NF-κB signaling pathway in fish. Interestingly, MyD88 is precisely regulated by IRF3 and IRF8 through the same mechanism but in completely opposite ways. IRF3 promotes MyD88-mediated NF-κB signaling pathway, whereas IRF8 inhibits the signaling pathway. MyD88 is regulated via ubiquitin-proteasome degradation, whereas IRF3 or IRF8 inhibited or promoted MyD88 degradation in this pathway. Specifically, in the early stage of lipopolysaccharide (LPS) stimulation or Vibrio infection, up-regulation of IRF3 and down-regulation of IRF8 eventually increased MyD88 expression to activate the NF-κB signaling pathway to trigger immune response. In the late stage of stimulation, down-regulated IRF3 and up-regulated IRF8 synergistically regulate the expression of MyD88 to a normal level, thus maintaining the immune balance of homeostasis and preventing serious damage from persistent over-immunization. This study presents information on Myd88-NF-κB signaling pathway in teleost fish and provides new insights into its regulatory mechanism in fish immune system.


Asunto(s)
Factor 3 Regulador del Interferón/fisiología , Factores Reguladores del Interferón/fisiología , Factor 88 de Diferenciación Mieloide/fisiología , FN-kappa B/fisiología , Perciformes/inmunología , Animales , Células Cultivadas , Células HEK293 , Humanos , Lipopolisacáridos/farmacología , Factor 88 de Diferenciación Mieloide/análisis , Complejo de la Endopetidasa Proteasomal/fisiología , Transducción de Señal/fisiología , Ubiquitinación , Vibriosis/inmunología
12.
Cancer Immunol Immunother ; 69(10): 2101-2112, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32448983

RESUMEN

Pancreatic ductal adenocarcinoma is characterized by a strong immunosuppressive network with a dense infiltration of myeloid cells including myeloid-derived suppressor cells (MDSC). Two distinct populations of MDSC have been defined: polymorphonuclear MDSC (PMN-MDSC) and monocytic MDSC (M-MDSC). Several factors influence the development and function of MDSC including the transcription factor interferon regulatory factor 4 (IRF4). Here, we show that IRF4 deficiency accelerates tumor growth and reduces survival, accompanied with a dense tumor infiltration with PMN-MDSC and reduced numbers of CD8+ T cells. As IRF4 has been described to modulate myeloid cell development and function, particularly of PMN-MDSC, we analyzed its role using MDSC-specific IRF4 knockout mice with the Ly6G or LysM knock-in allele expressing Cre recombinase and Irf4flox. In GM-CSF-driven bone marrow cultures, IRF4 deficiency increased the frequency of MDSC-like cells with a strong T cell suppressive capacity. Myeloid (LysM)-specific depletion of IRF4 led to increased tumor weight and a moderate splenic M-MDSC expansion in tumor-bearing mice. PMN cell (Ly6G)-specific depletion of IRF4, however, did not influence tumor progression or MDSC accumulation in vivo in accordance with our finding that IRF4 is not expressed in PMN-MDSC. This study demonstrates a critical role of IRF4 in the generation of an immunosuppressive tumor microenvironment in pancreatic cancer, which is independent of IRF4 expression in PMN-MDSC.


Asunto(s)
Biomarcadores de Tumor/análisis , Linfocitos T CD8-positivos/inmunología , Factores Reguladores del Interferón/fisiología , Células Supresoras de Origen Mieloide/inmunología , Neoplasias Pancreáticas/inmunología , Microambiente Tumoral/inmunología , Animales , Apoptosis , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Proliferación Celular , Modelos Animales de Enfermedad , Humanos , Terapia de Inmunosupresión , Ratones , Ratones Noqueados , Células Supresoras de Origen Mieloide/metabolismo , Células Supresoras de Origen Mieloide/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Pronóstico , Tasa de Supervivencia , Células Tumorales Cultivadas
13.
Virology ; 541: 160-173, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32056714

RESUMEN

Unique among human viruses, Kaposi's sarcoma-associated herpesvirus (KSHV) encodes several homologs of cellular interferon regulatory factors (vIRFs). Since KSHV expresses multiple factors that can inhibit interferon (IFN) signaling to promote virus production, it is still unclear to what extent vIRFs contribute to these specific processes during KSHV infection. To study the function of vIRFs during viral infection, we engineered 3xFLAG-tagged-vIRF and vIRF-knockout recombinant KSHV clones, which were utilized to test vIRF expression, as well as their requirement for viral replication, virus production, and inhibition of the type I IFN pathway in different models of lytic KSHV infection. Our data show that all vIRFs can be expressed as lytic viral proteins, yet were dispensable for KSHV production and inhibition of type I IFN. Nevertheless, as vIRFs were able to suppress IFN-stimulated antiviral genes, vIRFs may still promote the KSHV lytic cycle in the presence of an ongoing antiviral response.


Asunto(s)
Herpesvirus Humano 8/fisiología , Factores Reguladores del Interferón/fisiología , Interferón Tipo I/antagonistas & inhibidores , Proteínas Virales/fisiología , Replicación Viral , Células Cultivadas , Humanos , Interferón Tipo I/biosíntesis , Interferón beta/genética , Interferón beta/uso terapéutico , Transducción de Señal/fisiología , Activación Viral
14.
Front Immunol ; 11: 620716, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33613551

RESUMEN

Germinal centers play a key role in the adaptive immune system since they are able to produce memory B cells and plasma cells that produce high affinity antibodies for an effective immune protection. The mechanisms underlying cell-fate decisions are not well understood but asymmetric division of antigen, B-cell receptor affinity, interactions between B-cells and T follicular helper cells (triggering CD40 signaling), and regulatory interactions of transcription factors have all been proposed to play a role. In addition, a temporal switch from memory B-cell to plasma cell differentiation during the germinal center reaction has been shown. To investigate if antigen affinity-based Tfh cell help recapitulates the temporal switch we implemented a multiscale model that integrates cellular interactions with a core gene regulatory network comprising BCL6, IRF4, and BLIMP1. Using this model we show that affinity-based CD40 signaling in combination with asymmetric division of B-cells result in switch from memory B-cell to plasma cell generation during the course of the germinal center reaction. We also show that cell fate division is unlikely to be (solely) based on asymmetric division of Ag but that BLIMP1 is a more important factor. Altogether, our model enables to test the influence of molecular modulations of the CD40 signaling pathway on the production of germinal center output cells.


Asunto(s)
Linfocitos B/inmunología , Antígenos CD40/inmunología , Simulación por Computador , Centro Germinal/inmunología , Memoria Inmunológica/inmunología , Linfopoyesis/inmunología , Modelos Inmunológicos , Células Plasmáticas/inmunología , Células T Auxiliares Foliculares/inmunología , División Celular Asimétrica , Linfocitos B/citología , Linaje de la Célula , Redes Reguladoras de Genes , Centro Germinal/citología , Humanos , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/fisiología , Células Plasmáticas/citología , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Factor 1 de Unión al Dominio 1 de Regulación Positiva/fisiología , Proteínas Proto-Oncogénicas c-bcl-6/genética , Proteínas Proto-Oncogénicas c-bcl-6/fisiología , Transducción de Señal , Factores de Tiempo
15.
J Microbiol Biotechnol ; 29(12): 1873-1881, 2019 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-31650769

RESUMEN

The innate immune response serves as a first-line-of-defense mechanism for a host against viral infection. Viruses must therefore subvert this anti-viral response in order to establish an efficient life cycle. In line with this fact, Kaposi's sarcoma-associated herpesvirus (KSHV) encodes numerous genes that function as immunomodulatory proteins to antagonize the host immune system. One such mechanism through which KSHV evades the host immunity is by encoding a viral homolog of cellular interferon (IFN) regulatory factors (IRFs), known as vIRFs. Herein, we summarize recent advances in the study of the immunomodulatory strategies of KSHV vIRFs and their effects on KSHV-associated pathogenesis.


Asunto(s)
Herpesvirus Humano 8/fisiología , Evasión Inmune/fisiología , Factores Reguladores del Interferón/fisiología , Proteínas Virales/fisiología , Apoptosis/fisiología , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/inmunología , Interacciones Huésped-Patógeno/inmunología , Interacciones Huésped-Patógeno/fisiología , Humanos , Evasión Inmune/inmunología , Inmunidad Innata , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/inmunología , Transducción de Señal , Factores de Transcripción , Proteínas Virales/genética , Proteínas Virales/inmunología
17.
Inflammation ; 42(5): 1821-1829, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31243648

RESUMEN

Mounting evidence suggests that aberrant immune responses are involved in the pathogenesis of osteoarthritis (OA). Synovial macrophages are likely involved. In this study, we sought to investigate the role of interferon regulatory factor 5 (IRF5). In vitro M1-polarized macrophages presented significantly higher IRF5 expression than M2-polarized macrophages. Interestingly, IRF5 expression was observed in macrophages from the synovial fluid of OA patients, and the level of IRF expression was positively correlated with disease severity, such that stage 4 OA synovial macrophages presented significantly higher levels of IRF5 than stage 2 and stage 3 OA synovial macrophages. Circulating monocytes from OA patients, on the other hand, expressed little IRF5. However, synovial fluid from OA patients could significantly upregulate IRF5 expression in circulating monocytes. Synovial macrophages also expressed significantly higher IL-12 than circulating monocytes, and circulating monocytes conditioned in OA synovial fluid demonstrated significantly higher IL-12 expression. Direct IRF5 transfection could increase IL-12 expression in circulating monocytes. Interestingly, IRF5-transfected monocytes promoted the expression of Th1-associated genes in naive CD4 T cells via an IL-12-dependent mechanism. Overall, our study demonstrated that IRF5 expression was associated with OA severity and could contribute to the activation of the M1-Th1 axis.


Asunto(s)
Inflamación/etiología , Factores Reguladores del Interferón/fisiología , Macrófagos/patología , Osteoartritis/patología , Células Cultivadas , Femenino , Humanos , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Interleucina-12/metabolismo , Activación de Macrófagos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Células TH1 , Transfección
18.
Blood ; 133(17): 1803-1813, 2019 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-30796024

RESUMEN

Dendritic cells (DCs), which are vital for immune responses, are derived from bone marrow hematopoietic stem cells via common DC progenitors (CDPs). DC lineage fate decisions occurring at stages much earlier than CDPs have recently been recognized, yet the mechanism remains elusive. By single-cell RNA-sequencing, in vivo cell transfer experiments, and an assay for transposase-accessible chromatin sequencing using wild-type, IRF8-GFP chimera knock-in or IRF8-knockout mice, we demonstrate that IRF8 regulates chromatin at the lymphoid-primed multipotent progenitor (LMPP) stage to induce early commitment toward DCs. A low but significant expression of IRF8, a transcription factor essential for DC and monocyte development, was initiated in a subpopulation within LMPPs. These IRF8+ LMPPs were derived from IRF8- LMPPs and predominantly produced DCs, especially classical DC1s, potentially via known progenitors, such as monocyte-DC progenitors, CDPs, and preclassical DCs. IRF8+ LMPPs did not generate significant numbers of monocytes, neutrophils, or lymphocytes. Although IRF8- and IRF8+ LMPPs displayed very similar global gene expression patterns, the chromatin of enhancers near DC lineage genes was more accessible in IRF8+ LMPPs than in IRF8- LMPPs, an epigenetic change dependent on IRF8. The majority of the genes epigenetically primed by IRF8 were still transcriptionally inactive at the LMPP stage, but were highly expressed in the downstream DC lineage populations such as CDPs. Therefore, early expression of the key transcription factor IRF8 changes chromatin states in otherwise multipotent progenitors, biasing their fate decision toward DCs.


Asunto(s)
Linaje de la Célula/genética , Células Dendríticas/citología , Epigénesis Genética , Regulación de la Expresión Génica , Factores Reguladores del Interferón/fisiología , Células Madre Multipotentes/citología , Células Precursoras de Linfocitos B/citología , Animales , Células Cultivadas , Células Dendríticas/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre Multipotentes/metabolismo , Células Precursoras de Linfocitos B/metabolismo
19.
Dev Dyn ; 248(3): 221-232, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30684382

RESUMEN

BACKGROUND: Interferon regulatory factor 6 (IRF6) plays a critical role in embryonic tissue development, including differentiation of epithelial cells. Besides orofacial clefting due to haploinsufficiency of IRF6, recent human genetic studies indicated that mutations in IRF6 are linked to small mandible and digit abnormalities. The function of IRF6 has been well studied in oral epithelium; however, its role in craniofacial skeletal formation remains unknown. In this study, we investigated the role of Irf6 in craniofacial bone development using comparative analyses between wild-type (WT) and Irf6-null littermate mice. RESULTS: Immunostaining revealed the expression of IRF6 in hypertrophic chondrocytes, osteocytes, and bone matrix of craniofacial tissues. Histological analysis of Irf6-null mice showed a remarkable reduction in the number of lacunae, embedded osteocytes in matrices, and a reduction in mineralization during bone formation. These abnormalities may explain the decreased craniofacial bone density detected by micro-CT, loss of incisors, and mandibular bone abnormality of Irf6-null mice. To validate the autonomous role of IRF6 in bone, extracted primary osteoblasts from calvarial bone of WT and Irf6-null pups showed no effect on osteoblastic viability and proliferation. However, a reduction in mineralization was detected in Irf6-null cells. CONCLUSIONS: Altogether, these findings suggest an autonomous role of Irf6 in regulating bone differentiation and mineralization. Developmental Dynamics 248:221-232, 2019. © 2019 Wiley Periodicals, Inc.


Asunto(s)
Desarrollo Óseo/genética , Diferenciación Celular , Labio Leporino/genética , Fisura del Paladar/genética , Factores Reguladores del Interferón/genética , Osteoblastos/citología , Animales , Calcificación Fisiológica/genética , Proliferación Celular , Supervivencia Celular , Anomalías Craneofaciales/genética , Factores Reguladores del Interferón/fisiología , Ratones
20.
Science ; 363(6423)2019 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-30630901

RESUMEN

Cytopenias are an important clinical problem associated with inflammatory disease and infection. We show that specialized phagocytes that internalize red blood cells develop in Toll-like receptor 7 (TLR7)-driven inflammation. TLR7 signaling caused the development of inflammatory hemophagocytes (iHPCs), which resemble splenic red pulp macrophages but are a distinct population derived from Ly6Chi monocytes. iHPCs were responsible for anemia and thrombocytopenia in TLR7-overexpressing mice, which have a macrophage activation syndrome (MAS)-like disease. Interferon regulatory factor 5 (IRF5), associated with MAS, participated in TLR7-driven iHPC differentiation. We also found iHPCs during experimental malarial anemia, in which they required endosomal TLR and MyD88 signaling for differentiation. Our findings uncover a mechanism by which TLR7 and TLR9 specify monocyte fate and identify a specialized population of phagocytes responsible for anemia and thrombocytopenia associated with inflammation and infection.


Asunto(s)
Anemia/fisiopatología , Síndrome de Activación Macrofágica/fisiopatología , Glicoproteínas de Membrana/fisiología , Fagocitos/citología , Transducción de Señal , Receptor Toll-Like 7/fisiología , Receptor Toll-Like 9/fisiología , Animales , Diferenciación Celular , Células Cultivadas , Proteínas de Unión al ADN/fisiología , Inflamación/fisiopatología , Factores Reguladores del Interferón/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/citología , Factor 88 de Diferenciación Mieloide/fisiología , Plasmodium yoelii , Bazo/citología , Trombocitopenia/fisiopatología , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...