Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 840
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 20(7): e1012379, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39037956

RESUMEN

RNA helicases are involved in the innate immune response against pathogens, including bacteria and viruses; however, their mechanism in the human airway epithelial cells is still not fully understood. Here, we demonstrated that DEAH (Asp-Glu-Ala-His) box polypeptide 35 (DHX35), a member of the DExD/H (Asp-Glu-x-Asp/His)-box helicase family, boosts antiviral innate immunity in human airway epithelial cells. DHX35 knockdown attenuated the production of interferon-ß (IFN-ß), IL6, and CXCL10, whereas DHX35 overexpression increased their production. Upon stimulation, DHX35 was constitutively expressed, but it translocated from the nucleus into the cytosol, where it recognized cytosolic poly(I:C) and poly(dA:dT) via its HELICc domain. Mitochondrial antiviral signaling protein (MAVS) acted as an adaptor for DHX35 and interacted with the HELICc domain of DHX35 using amino acids 360-510. Interestingly, DHX35 interacted with retinoic acid-inducible gene 1 (RIG-I), enhanced the binding affinity of RIG-I with poly(I:C) and poly(dA:dT), and formed a signalsome with MAVS to activate interferon regulatory factor 3 (IRF3), NF-κB-p65, and MAPK signaling pathways. These results indicate that DHX35 not only acted as a cytosolic nucleic acid sensor but also synergized with RIG-I to enhance antiviral immunity in human airway epithelial cells. Our results demonstrate a novel molecular mechanism for DHX35 in RIG-I-mediated innate immunity and provide a novel candidate for drug and vaccine design to control viral infections in the human airway.


Asunto(s)
Proteína 58 DEAD Box , ARN Helicasas DEAD-box , Inmunidad Innata , Receptores Inmunológicos , Humanos , Proteína 58 DEAD Box/metabolismo , Proteína 58 DEAD Box/inmunología , ARN Helicasas DEAD-box/metabolismo , ARN Helicasas DEAD-box/inmunología , Receptores Inmunológicos/metabolismo , Poli I-C/inmunología , Poli I-C/farmacología , ARN Helicasas/metabolismo , ARN Helicasas/inmunología , Transducción de Señal/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/inmunología , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/virología , Células HEK293
2.
Nat Immunol ; 15(8): 749-757, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24973821

RESUMEN

Interleukin 15 (IL-15) controls both the homeostasis and the peripheral activation of natural killer (NK) cells. The molecular basis for this duality of action remains unknown. Here we found that the metabolic checkpoint kinase mTOR was activated and boosted bioenergetic metabolism after exposure of NK cells to high concentrations of IL-15, whereas low doses of IL-15 triggered only phosphorylation of the transcription factor STAT5. mTOR stimulated the growth and nutrient uptake of NK cells and positively fed back on the receptor for IL-15. This process was essential for sustaining NK cell proliferation during development and the acquisition of cytolytic potential during inflammation or viral infection. The mTORC1 inhibitor rapamycin inhibited NK cell cytotoxicity both in mice and humans; this probably contributes to the immunosuppressive activity of this drug in different clinical settings.


Asunto(s)
Interleucina-15/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Serina-Treonina Quinasas TOR/inmunología , Animales , Proliferación Celular , Células Cultivadas , Infecciones por Herpesviridae/inmunología , Humanos , Inmunosupresores/farmacología , Inflamación/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Células Asesinas Naturales/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/genética , Complejos Multiproteicos/inmunología , Muromegalovirus/inmunología , Infecciones por Orthomyxoviridae/inmunología , Poli I-C/inmunología , Factor de Transcripción STAT5/metabolismo , Transducción de Señal/inmunología , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/genética
3.
Immunity ; 47(1): 12-14, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28723545

RESUMEN

Sickness in mammals can lead to cognition deficits, although the underlying mechanisms remain elusive. In a recent Nature Medicine article, Garré et al. (2017) report that sickness-induced cortical dendritic spine loss and impaired memory formation is mediated by CX3CR1+ monocyte-derived TNF-α.


Asunto(s)
Espinas Dendríticas/fisiología , Trastornos Mentales/inmunología , Monocitos/fisiología , Neuronas Motoras/fisiología , Red Nerviosa , Plasticidad Neuronal , Virosis/inmunología , Animales , Receptor 1 de Quimiocinas CX3C , Humanos , Memoria , Trastornos Mentales/etiología , Trastornos Mentales/psicología , Ratones , Monocitos/virología , Neuronas Motoras/virología , Poli I-C/inmunología , Receptores de Quimiocina/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Virosis/complicaciones , Virosis/psicología
4.
Proc Natl Acad Sci U S A ; 119(26): e2116738119, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35749366

RESUMEN

Tumor infiltration by T cells profoundly affects cancer progression and responses to immunotherapy. However, the tumor immunosuppressive microenvironment can impair the induction, trafficking, and local activity of antitumor T cells. Here, we investigated whether intratumoral injection of virus-derived peptide epitopes could activate preexisting antiviral T cell responses locally and promote antitumor responses or antigen spreading. We focused on a mouse model of cytomegalovirus (CMV), a highly prevalent human infection that induces vigorous and durable T cell responses. Mice persistently infected with murine CMV (MCMV) were challenged with lung (TC-1), colon (MC-38), or melanoma (B16-F10) tumor cells. Intratumoral injection of MCMV-derived T cell epitopes triggered in situ and systemic expansion of their cognate, MCMV-specific CD4+ or CD8+ T cells. The MCMV CD8+ T cell epitopes injected alone provoked arrest of tumor growth and some durable remissions. Intratumoral injection of MCMV CD4+ T cell epitopes with polyinosinic acid:polycytidylic acid (pI:C) preferentially elicited tumor antigen-specific CD8+ T cells, promoted tumor clearance, and conferred long-term protection against tumor rechallenge. Notably, secondary proliferation of MCMV-specific CD8+ T cells correlated with better tumor control. Importantly, intratumoral injection of MCMV-derived CD8+ T cell-peptide epitopes alone or CD4+ T cell-peptide epitopes with pI:C induced potent adaptive and innate immune activation of the tumor microenvironment. Thus, CMV-derived peptide epitopes, delivered intratumorally, act as cytotoxic and immunotherapeutic agents to promote immediate tumor control and long-term antitumor immunity that could be used as a stand-alone therapy. The tumor antigen-agnostic nature of this approach makes it applicable across a broad range of solid tumors regardless of their origin.


Asunto(s)
Linfocitos T CD8-positivos , Infecciones por Citomegalovirus , Citomegalovirus , Epítopos de Linfocito T , Neoplasias , Animales , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Citomegalovirus/inmunología , Infecciones por Citomegalovirus/inmunología , Epítopos de Linfocito T/administración & dosificación , Epítopos de Linfocito T/inmunología , Inmunoterapia , Ratones , Neoplasias/inmunología , Neoplasias/terapia , Poli I-C/administración & dosificación , Poli I-C/inmunología , Microambiente Tumoral
5.
Proc Natl Acad Sci U S A ; 119(15): e2113310119, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35377817

RESUMEN

Gestational maternal immune activation (MIA) in mice induces persistent brain microglial activation and a range of neuropathologies in the adult offspring. Although long-term phenotypes are well documented, how MIA in utero leads to persistent brain inflammation is not well understood. Here, we found that offspring of mothers treated with polyriboinosinic­polyribocytidylic acid [poly(I:C)] to induce MIA at gestational day 13 exhibit blood­brain barrier (BBB) dysfunction throughout life. Live MRI in utero revealed fetal BBB hyperpermeability 2 d after MIA. Decreased pericyte­endothelium coupling in cerebral blood vessels and increased microglial activation were found in fetal and 1- and 6-mo-old offspring brains. The long-lasting disruptions result from abnormal prenatal BBB formation, driven by increased proliferation of cyclooxygenase-2 (COX2; Ptgs2)-expressing microglia in fetal brain parenchyma and perivascular spaces. Targeted deletion of the Ptgs2 gene in fetal myeloid cells or treatment with the inhibitor celecoxib 24 h after immune activation prevented microglial proliferation and disruption of BBB formation and function, showing that prenatal COX2 activation is a causal pathway of MIA effects. Thus, gestational MIA disrupts fetal BBB formation, inducing persistent BBB dysfunction, which promotes microglial overactivation and behavioral alterations across the offspring life span. Taken together, the data suggest that gestational MIA disruption of BBB formation could be an etiological contributor to neuropsychiatric disorders.


Asunto(s)
Barrera Hematoencefálica , Ciclooxigenasa 2 , Encefalitis , Intercambio Materno-Fetal , Microglía , Efectos Tardíos de la Exposición Prenatal , Animales , Barrera Hematoencefálica/anomalías , Barrera Hematoencefálica/fisiopatología , Celecoxib/farmacología , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , Encefalitis/inmunología , Femenino , Eliminación de Gen , Intercambio Materno-Fetal/inmunología , Ratones , Microglía/enzimología , Poli I-C/inmunología , Embarazo , Efectos Tardíos de la Exposición Prenatal/inmunología
6.
J Biol Chem ; 299(9): 105114, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37524131

RESUMEN

Exosomes, extracellular vesicles (EVs) produced within cells, mediate both the disposal of intracellular waste and communication with distant cells, and they are involved in a variety of disease processes. Although disease modifications of exosome cargos have been well studied, it has been poorly investigated how disease processes, such as endoplasmic reticulum (ER) stress, affect EV production. We previously reported that adiponectin, an adipocyte-secreted salutary factor, increases systemic exosome levels through T-cadherin-mediated enhancement of exosome biogenesis. In the present study, we demonstrated that adiponectin/T-cadherin-dependent EV production was susceptible to ER stress and that low-dose tunicamycin significantly reduced EV production in the presence, but not in the absence, of adiponectin. Moreover, pharmacological or genetic activation of inositol-requiring enzyme 1α, a central regulator of ER stress, downregulated T-cadherin at the mRNA and protein levels as well as attenuated EV production. In addition, adiponectin/T-cadherin-independent EV production was attenuated under ER stress conditions. Repeated administration of tunicamycin to mice decreased circulating small EVs without decreasing tissue T-cadherin expression. Mechanistically, inositol-requiring enzyme 1α activation by silencing of the X-box binding protein 1 transcription factor upregulated the canonical interferon pathway and decreased EV production. The interferon pathway, when it was activated by polyinosinic-polycytidylic acid, also significantly attenuated EV production. Thus, we concluded that ER stress decreases exosome production through adiponectin/T-cadherin-dependent and -independent pathways.


Asunto(s)
Adiponectina , Cadherinas , Estrés del Retículo Endoplásmico , Exosomas , Animales , Ratones , Adiponectina/metabolismo , Cadherinas/biosíntesis , Cadherinas/genética , Cadherinas/metabolismo , Exosomas/efectos de los fármacos , Exosomas/metabolismo , Inositol/metabolismo , Interferones/inmunología , Poli I-C/inmunología , Tunicamicina/farmacología
7.
Clin Exp Immunol ; 217(3): 263-278, 2024 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-38695079

RESUMEN

Neonate responses to pathogen-associated molecular patterns (PAMPS) differ from adults; such understanding is poor in Indian neonates, despite recognized significant infectious risk. Immune profiling analysis was undertaken of 10 secreted mediators contextualized with cellular source induced by six PAMPs in umbilical cord (CB; n = 21) and adult-blood (PBMC; n = 14) from a tertiary care hospital in South India. Differential cytokine expression analysis (minimum log2-fold difference; adj P-value < 0.05) identified bacterial PAMPs induced higher concentrations of IL-1ß, IL-10, TNF-α in adults versus IL-8, GM-CSF, IFN-γ, and IL-2 in CB. CB responded to poly I:C and SARS-CoV-2 lysate with a dominant IL-8 response, whereas in PBMC, CXCL-10 dominated poly I:C, but not SARS-CoV-2, responses, highlighting potential IL-8 importance, in the absence of Type I Interferons, in antiviral CB immunity. Candida albicans was the only PAMP to uniformly induce higher secretion of effectors in CB. The predominant source of IL-8/IL-6/TNF-α/IL-1ß in both CB and PBMC was polyfunctional monocytes and IFN-γ/IL-2/IL-17 from innate lymphocytes. Correlation matrix analyses revealed IL-8 to be the most differentially regulated, correlating positively in CB versus negatively in PBMC with IL-6, GM-CSF, IFN-γ, IL-2, consistent with more negatively regulated cytokine modules in adults, potentially linked to higher anti-inflammatory IL-10. Cord and adult blood from India respond robustly to PAMPs with unique effector combinations. These data provide a strong foundation to monitor, explore, mechanisms that regulate such immunity during the life course, an area of significant global health importance given infection-related infant mortality incidence.


Asunto(s)
COVID-19 , Quimiocina CXCL10 , Sangre Fetal , Interleucina-8 , Leucocitos Mononucleares , Monocitos , SARS-CoV-2 , Humanos , India , Adulto , Sangre Fetal/inmunología , Leucocitos Mononucleares/inmunología , SARS-CoV-2/inmunología , COVID-19/inmunología , Monocitos/inmunología , Interleucina-8/inmunología , Quimiocina CXCL10/inmunología , Femenino , Masculino , Recién Nacido , Poli I-C/inmunología , Interleucina-10 , Candida albicans/inmunología , Citocinas/metabolismo
8.
Semin Immunol ; 49: 101414, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-33011064

RESUMEN

Immunotherapies have become the first line of treatment for many cancer types. Unfortunately, only a small fraction of patients benefits from these therapies. This low rate of success can be attributed to 3 main barriers: 1) low frequency of anti-tumor specific T cells; 2) lack of infiltration of the anti-tumor specific T cells into the tumor parenchyma and 3) accumulation of highly suppressive cells in the tumor mass that inhibit the effector function of the anti-tumor specific T cells. Thus, the identification of immunomodulators that can increase the frequency and/or the infiltration of antitumor specific T cells while reducing the suppressive capacity of the tumor microenvironment is necessary to ensure the effectiveness of T cell immunotherapies. In this review, we discuss the potential of poly-ICLC as a multi-functional immune modulator for treating cancer and its impact on the 3 above mentioned barriers. We describe the unique capacity of poly-ICLC in stimulating 2 separate pattern recognition receptors, TLR3 and cytosolic MDA5 and the consequences of these activations on cytokines and chemokines production. We emphasize the role of poly-ICLC as an adjuvant in the setting of peptide-based cancer vaccines and in situ tumor vaccination by mimicking natural immune responses to infections. Finally, we summarize the impact of poly-ICLC in enhancing T infiltration into the tumor parenchyma and address the implication of this finding in the clinic.


Asunto(s)
Antineoplásicos/farmacología , Carboximetilcelulosa de Sodio/análogos & derivados , Factores Inmunológicos/farmacología , Inmunomodulación , Poli I-C/inmunología , Poli I-C/farmacología , Polilisina/análogos & derivados , Animales , Antineoplásicos/uso terapéutico , Carboximetilcelulosa de Sodio/farmacología , Carboximetilcelulosa de Sodio/uso terapéutico , Citocinas/metabolismo , Humanos , Inmunidad Innata/efectos de los fármacos , Factores Inmunológicos/uso terapéutico , Inmunomodulación/efectos de los fármacos , Helicasa Inducida por Interferón IFIH1/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/etiología , Neoplasias/metabolismo , Neoplasias/patología , Poli I-C/uso terapéutico , Polilisina/inmunología , Polilisina/farmacología , Polilisina/uso terapéutico , Receptores de Reconocimiento de Patrones/metabolismo , Receptor Toll-Like 3/metabolismo
9.
PLoS Genet ; 17(5): e1009578, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34014925

RESUMEN

The Receptor Transporter Protein (RTP) family is present in most, if not all jawed vertebrates. Most of our knowledge of this protein family comes from studies on mammalian RTPs, which are multi-function proteins that regulate cell-surface G-protein coupled receptor levels, influence olfactory system development, regulate immune signaling, and directly inhibit viral infection. However, mammals comprise less than one-tenth of extant vertebrate species, and our knowledge about the expression, function, and evolution of non-mammalian RTPs is limited. Here, we explore the evolutionary history of RTPs in vertebrates. We identify signatures of positive selection in many vertebrate RTP clades and characterize multiple, independent expansions of the RTP family outside of what has been described in mammals. We find a striking expansion of RTPs in the African clawed frog, Xenopus laevis, with 11 RTPs in this species as opposed to 1 to 4 in most other species. RNA sequencing revealed that most X. laevis RTPs are upregulated following immune stimulation. In functional assays, we demonstrate that at least three of these X. laevis RTPs inhibit infection by RNA viruses, suggesting that RTP homologs may serve as antiviral effectors outside of Mammalia.


Asunto(s)
Antivirales , Evolución Molecular , Genómica , Proteínas de Transporte de Membrana/genética , Proteínas de Xenopus/genética , Xenopus laevis/genética , Animales , Antivirales/inmunología , Proteínas de Transporte de Membrana/inmunología , Poli I-C/inmunología , Sintenía , Proteínas de Xenopus/inmunología , Xenopus laevis/inmunología , Xenopus laevis/metabolismo
10.
PLoS Pathog ; 17(10): e1009928, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34695164

RESUMEN

Non-specific protective effects of certain vaccines have been reported, and long-term boosting of innate immunity, termed trained immunity, has been proposed as one of the mechanisms mediating these effects. Several epidemiological studies suggested cross-protection between influenza vaccination and COVID-19. In a large academic Dutch hospital, we found that SARS-CoV-2 infection was less common among employees who had received a previous influenza vaccination: relative risk reductions of 37% and 49% were observed following influenza vaccination during the first and second COVID-19 waves, respectively. The quadrivalent inactivated influenza vaccine induced a trained immunity program that boosted innate immune responses against various viral stimuli and fine-tuned the anti-SARS-CoV-2 response, which may result in better protection against COVID-19. Influenza vaccination led to transcriptional reprogramming of monocytes and reduced systemic inflammation. These epidemiological and immunological data argue for potential benefits of influenza vaccination against COVID-19, and future randomized trials are warranted to test this possibility.


Asunto(s)
COVID-19/inmunología , Protección Cruzada/fisiología , Inmunidad Innata/fisiología , Vacunas contra la Influenza/administración & dosificación , COVID-19/epidemiología , COVID-19/prevención & control , Citocinas/inmunología , Citocinas/metabolismo , Regulación hacia Abajo , Imidazoles/inmunología , Incidencia , Vacunas contra la Influenza/inmunología , Países Bajos/epidemiología , Personal de Hospital , Poli I-C/inmunología , Proteómica , Factores de Riesgo , Análisis de Secuencia de ARN
11.
Nat Immunol ; 12(1): 37-44, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21102435

RESUMEN

The poly(ADP-ribose) polymerases (PARPs) participate in many biological and pathological processes. Here we report that the PARP-13 shorter isoform (ZAPS), rather than the full-length protein (ZAP), was selectively induced by 5'-triphosphate-modified RNA (3pRNA) and functioned as a potent stimulator of interferon responses in human cells mediated by the RNA helicase RIG-I. ZAPS associated with RIG-I to promote the oligomerization and ATPase activity of RIG-I, which led to robust activation of IRF3 and NF-κB transcription factors. Disruption of the gene encoding ZAPS resulted in impaired induction of interferon-α (IFN-α), IFN-ß and other cytokines after viral infection. These results indicate that ZAPS is a key regulator of RIG-I signaling during the innate antiviral immune response, which suggests its possible use as a therapeutic target for viral control.


Asunto(s)
Infecciones por Avulavirus/metabolismo , ARN Helicasas DEAD-box/metabolismo , Virus de la Enfermedad de Newcastle/fisiología , Infecciones por Orthomyxoviridae/metabolismo , Orthomyxoviridae/fisiología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Isoformas de Proteínas/metabolismo , Infecciones por Avulavirus/inmunología , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/inmunología , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Células HEK293 , Humanos , Inmunidad Innata , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Virus de la Enfermedad de Newcastle/patogenicidad , Orthomyxoviridae/patogenicidad , Infecciones por Orthomyxoviridae/inmunología , Poli I-C/inmunología , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/inmunología , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN , Receptores Inmunológicos , Transducción de Señal/genética , Transducción de Señal/inmunología , Replicación Viral/genética
12.
J Immunol ; 206(1): 154-163, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33219146

RESUMEN

Viral RNA in the cytoplasm of mammalian host cells is recognized by retinoic acid-inducible protein-I-like receptors (RLRs), which localize to cytoplasmic stress granules (SGs). Activated RLRs associate with the mitochondrial adaptor protein IPS-1, which activates antiviral host defense mechanisms, including type I IFN induction. It has remained unclear, however, how RLRs in SGs and IPS-1 in the mitochondrial outer membrane associate physically and engage in information transfer. In this study, we show that NUDT21, an RNA-binding protein that regulates alternative transcript polyadenylation, physically associates with IPS-1 and mediates its localization to SGs in response to transfection with polyinosinic-polycytidylic acid [poly(I:C)], a mimic of viral dsRNA. We found that despite its well-established function in the nucleus, a fraction of NUDT21 localizes to mitochondria in resting cells and becomes localized to SGs in response to poly(I:C) transfection. NUDT21 was also found to be required for efficient type I IFN induction in response to viral infection in both human HeLa cells and mouse macrophage cell line RAW264.7 cells. Our results together indicate that NUDT21 links RLRs in SGs to mitochondrial IPS-1 and thereby activates host defense responses to viral infection.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Infecciones por Cardiovirus/metabolismo , Factor de Especificidad de Desdoblamiento y Poliadenilación/metabolismo , Proteína 58 DEAD Box/metabolismo , Virus de la Encefalomiocarditis/fisiología , Mitocondrias/metabolismo , Enfermedad de Newcastle/metabolismo , Virus de la Enfermedad de Newcastle/fisiología , Receptores Inmunológicos/metabolismo , Vesículas Secretoras/metabolismo , Animales , Factor de Especificidad de Desdoblamiento y Poliadenilación/genética , Regulación de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Ratones , Poli I-C/inmunología , Transporte de Proteínas , Células RAW 264.7 , ARN Interferente Pequeño/genética , ARN Viral/inmunología , Estrés Fisiológico
13.
J Immunol ; 207(2): 408-420, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34193600

RESUMEN

Type I IFNs (IFN-I) are important for tumor immune surveillance and contribute to the therapeutic responses for numerous treatment regimens. Nevertheless, certain protumoral activities by IFN-I have been increasingly recognized. Indeed, our recent work showed that systemic poly(I:C)/IFN treatment can undesirably trigger high arginase (ARG1) expression within the tumor-associated monocyte/macrophage compartment. Using a line of CRISPR-generated Arg1-YFP reporter knock-in mice, we have determined that a subset of tumor-associated macrophages represent the major Arg1-expressing cell type following poly(I:C)/IFN stimulation. More detailed analyses from in vitro and in vivo models demonstrate a surprising IFN-to-IL-4 cytokine axis in transitional monocytes, which can subsequently stimulate IL-4 target genes, including Arg1, in macrophages. Intriguingly, IFN stimulation of transitional monocytes yielded concurrent M2 (YFP+)- and M1 (YFP-)-skewed macrophage subsets, correlated with an inhibitory crosstalk between IFN-I and IL-4. Genetic abrogation of IL-4 signaling in mice diminished poly(I:C)/IFN-induced ARG1 in tumors, leading to enhanced activation of CD8+ T cells and an improved therapeutic effect. The present work uncovered a monocyte-orchestrated macrophage phenotype conversion mechanism that may have broad implications.


Asunto(s)
Citocinas/metabolismo , Interferones/metabolismo , Interleucina-4/metabolismo , Macrófagos/metabolismo , Monocitos/metabolismo , Poli I-C/metabolismo , Animales , Arginasa/inmunología , Arginasa/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/inmunología , Citocinas/inmunología , Femenino , Interferones/inmunología , Interleucina-4/inmunología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Neoplasias/inmunología , Neoplasias/metabolismo , Fenotipo , Poli I-C/inmunología , Transducción de Señal/inmunología , Transducción de Señal/fisiología
14.
Nucleic Acids Res ; 49(17): 10034-10045, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34428287

RESUMEN

Quaking (QKI) controls RNA metabolism in many biological processes including innate immunity, where its roles remain incompletely understood. To illuminate these roles, we performed genome scale transcriptome profiling in QKI knockout cells with or without poly(I:C) transfection, a double-stranded RNA analog that mimics viral infection. Analysis of RNA-sequencing data shows that QKI knockout upregulates genes induced by interferons, suggesting that QKI is an immune suppressor. Furthermore, differential splicing analysis shows that QKI primarily controls cassette exons, and among these events, we noted that QKI silences splicing of the extra domain A (EDA) exon in fibronectin (FN1) transcripts. QKI knockout results in elevated production and secretion of FN1-EDA protein, which is a known activator of interferons. Consistent with an upregulation of the interferon response in QKI knockout cells, our results show reduced production of dengue virus-2 and Japanese encephalitis virus in these cells. In conclusion, we demonstrate that QKI downregulates the interferon system and attenuates the antiviral state.


Asunto(s)
Virus del Dengue/crecimiento & desarrollo , Virus de la Encefalitis Japonesa (Especie)/crecimiento & desarrollo , Fibronectinas/genética , Interferón Tipo I/inmunología , Empalme del ARN/genética , Proteínas de Unión al ARN/metabolismo , Células A549 , Línea Celular Tumoral , Virus del Dengue/inmunología , Virus de la Encefalitis Japonesa (Especie)/inmunología , Perfilación de la Expresión Génica , Humanos , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Interferón Tipo I/genética , Poli I-C/inmunología , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/genética , Transcriptoma/genética , Regulación hacia Arriba/genética
15.
Proc Natl Acad Sci U S A ; 117(44): 27502-27508, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33087577

RESUMEN

Cyclic dinucleotides (CDNs) are secondary messengers used by prokaryotic and eukaryotic cells. In mammalian cells, cytosolic CDNs bind STING (stimulator of IFN gene), resulting in the production of type I IFN. Extracellular CDNs can enter the cytosol through several pathways but how CDNs work from outside eukaryotic cells remains poorly understood. Here, we elucidate a mechanism of action on intestinal epithelial cells for extracellular CDNs. We found that CDNs containing adenosine induced a robust CFTR-mediated chloride secretory response together with cAMP-mediated inhibition of Poly I:C-stimulated IFNß expression. Signal transduction was strictly polarized to the serosal side of the epithelium, dependent on the extracellular and sequential hydrolysis of CDNs to adenosine by the ectonucleosidases ENPP1 and CD73, and occurred via activation of A2B adenosine receptors. These studies highlight a pathway by which microbial and host produced extracellular CDNs can regulate the innate immune response of barrier epithelial cells lining mucosal surfaces.


Asunto(s)
Adenosina/metabolismo , Células Epiteliales/metabolismo , Inmunidad Innata , Inmunidad Mucosa , Nucleótidos Cíclicos/metabolismo , 5'-Nucleotidasa/metabolismo , Línea Celular Tumoral , Cloruros/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Células Epiteliales/inmunología , Proteínas Ligadas a GPI/metabolismo , Humanos , Interferón beta/metabolismo , Mucosa Intestinal/citología , Hidrolasas Diéster Fosfóricas/metabolismo , Poli I-C/inmunología , Pirofosfatasas/metabolismo , Receptor de Adenosina A2B/metabolismo , Transducción de Señal/inmunología
16.
Proc Natl Acad Sci U S A ; 117(16): 9022-9031, 2020 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-32284404

RESUMEN

The vast majority of type 1 diabetes (T1D) genetic association signals lie in noncoding regions of the human genome. Many have been predicted to affect the expression and secondary structure of long noncoding RNAs (lncRNAs), but the contribution of these lncRNAs to the pathogenesis of T1D remains to be clarified. Here, we performed a complete functional characterization of a lncRNA that harbors a single nucleotide polymorphism (SNP) associated with T1D, namely, Lnc13 Human pancreatic islets harboring the T1D-associated SNP risk genotype in Lnc13 (rs917997*CC) showed higher STAT1 expression than islets harboring the heterozygous genotype (rs917997*CT). Up-regulation of Lnc13 in pancreatic ß-cells increased activation of the proinflammatory STAT1 pathway, which correlated with increased production of chemokines in an allele-specific manner. In a mirror image, Lnc13 gene disruption in ß-cells partially counteracts polyinosinic-polycytidylic acid (PIC)-induced STAT1 and proinflammatory chemokine expression. Furthermore, we observed that PIC, a viral mimetic, induces Lnc13 translocation from the nucleus to the cytoplasm promoting the interaction of STAT1 mRNA with (poly[rC] binding protein 2) (PCBP2). Interestingly, Lnc13-PCBP2 interaction regulates the stability of the STAT1 mRNA, sustaining inflammation in ß-cells in an allele-specific manner. Our results show that the T1D-associated Lnc13 may contribute to the pathogenesis of T1D by increasing pancreatic ß-cell inflammation. These findings provide information on the molecular mechanisms by which disease-associated SNPs in lncRNAs influence disease pathogenesis and open the door to the development of diagnostic and therapeutic approaches based on lncRNA targeting.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Células Secretoras de Insulina/inmunología , ARN Largo no Codificante/metabolismo , Proteínas de Unión al ARN/metabolismo , Factor de Transcripción STAT1/genética , Regiones no Traducidas 3'/genética , Supervivencia Celular/genética , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/virología , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Células Secretoras de Insulina/patología , Células Secretoras de Insulina/virología , Células Jurkat , Poli I-C/inmunología , Polimorfismo de Nucleótido Simple , Cultivo Primario de Células , Estabilidad del ARN/genética , ARN Mensajero/metabolismo , ARN Viral/inmunología , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Regulación hacia Arriba/inmunología
17.
Scand J Immunol ; 95(3): e13128, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34923667

RESUMEN

Targeting antigens to dendritic cells represent a promising method for enhancing immune responses against specific antigens. However, many studies have focused on systemic delivery (intravenous or intraperitoneally) of targeted antigen, approaches that are not easily transferable to humans. Here we evaluate the efficacy of an influenza vaccine targeting Xcr1+ cDC1 administered by intranasal immunization. Intranasal delivery of antigen fused to the chemokine Xcl1, the ligand of Xcr1, resulted in specific uptake by lung CD103+ cDC1. Interestingly, intranasal immunization with influenza A/PR/8/34 haemagglutinin (HA) fused to Xcl1, formulated with poly(I:C), resulted in enhanced induction of antigen-specific IFNγ+ CD4+ and IFNγ+ CD8+ T cell responses in lung compared non-targeted anti-NIP-HA (αNIP-HA). Induction of antibody responses was, however, similar in Xcl1-HA and αNIP-HA immunized mice, but significantly higher than in mice immunized with monomeric HA. Both Xcl1-HA and αNIP-HA vaccines induced full protection when mice were challenged with a lethal dose of influenza PR8 virus, reflecting the strong induction of HA-specific antibodies. Our results demonstrate that i.n. delivery of Xcl1-HA is a promising vaccine strategy for enhancing T cell responses in addition to inducing strong antibody responses.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Quimiocinas C/metabolismo , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Infecciones por Orthomyxoviridae/inmunología , Poli I-C/inmunología , Animales , Anticuerpos Antivirales/inmunología , Formación de Anticuerpos/inmunología , Antígenos/inmunología , Antígenos CD/inmunología , Línea Celular , Células Dendríticas/inmunología , Perros , Femenino , Células HEK293 , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Cadenas alfa de Integrinas/inmunología , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos BALB C
18.
Immunity ; 38(4): 717-28, 2013 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-23601685

RESUMEN

RIG-I-like receptors (RLRs) sense virus-derived RNA or polyinosinic-polycytidylic acid (poly IC) to exert antiviral immune responses. Here, we examine the mechanisms underlying the adjuvant effects of poly IC. Poly IC was taken up by dendritic cells (DCs), and it induced lysosomal destabilization, which, in turn, activated an RLR-dependent signaling pathway. Upon poly IC stimulation, cathepsin D was released into the cytoplasm from the lysosome to interact with IPS-1, an adaptor molecule for RLRs. This interaction facilitated cathepsin D cleavage of caspase 8 and the activation of the transcription factor NF-κB, resulting in enhanced cytokine production. Further recruitment of the kinase RIP-1 to this complex initiated the necroptosis of a small number of DCs. HMGB1 released by dying cells enhanced IFN-ß production in concert with poly IC. Collectively, these findings suggest that cathepsin D-triggered, IPS-1-dependent necroptosis is a mechanism that propagates the adjuvant efficacy of poly IC.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Catepsina D/metabolismo , Células Dendríticas/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/virología , Proteínas Activadoras de GTPasa/metabolismo , Proteína HMGB1/metabolismo , Inmunidad Innata , Inmunomodulación , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Necrosis/inmunología , Poli I-C/inmunología , Unión Proteica , Transducción de Señal/inmunología
19.
Immunity ; 39(1): 123-35, 2013 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-23871209

RESUMEN

The NLRP3 inflammasome plays a major role in innate immune responses by activating caspase-1, resulting in secretion of interleukin-18 (IL-18) and IL-1ß. Although cytosolic double-stranded RNA (dsRNA) and bacterial RNA are known to activate the NLRP3 inflammasome, the upstream sensor is unknown. We investigated the potential function of DExD/H-box RNA helicase family members (previously shown to sense cytosolic DNA and RNA to induce type 1 interferon responses) in RNA-induced NLRP3 inflammasome activation. Among the helicase family members tested, we found that targeting of DHX33 expression by short hairpin RNA efficiently blocked the activation of caspase-1 and secretion of IL-18 and IL-1ß in human macrophages that were activated by cytosolic poly I:C, reoviral RNA, or bacterial RNA. DHX33 bound dsRNA via the helicase C domain. DHX33 interacted with NLRP3 and formed the inflammasome complex following stimulation with RNA. We therefore identified DHX33 as a cytosolic RNA sensor that activates the NLRP3 inflammasome.


Asunto(s)
Proteínas Portadoras/inmunología , ARN Helicasas DEAD-box/inmunología , Inflamasomas/inmunología , Macrófagos/inmunología , ARN/inmunología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Caspasa 1/inmunología , Caspasa 1/metabolismo , Línea Celular , Citosol/inmunología , Citosol/metabolismo , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Expresión Génica/inmunología , Células HEK293 , Humanos , Immunoblotting , Inflamasomas/genética , Inflamasomas/metabolismo , Interleucina-18/inmunología , Interleucina-18/metabolismo , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Activación de Macrófagos/inmunología , Macrófagos/metabolismo , Microscopía Confocal , Proteína con Dominio Pirina 3 de la Familia NLR , Poli I-C/inmunología , Unión Proteica/inmunología , ARN/genética , ARN/metabolismo , Interferencia de ARN , ARN Bacteriano/inmunología , ARN Bicatenario/genética , ARN Bicatenario/inmunología , ARN Bicatenario/metabolismo , ARN Viral/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
J Immunol ; 205(9): 2511-2518, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32967931

RESUMEN

Microglia are phagocytic cells involved in homeostasis of the brain and are key players in the pathogenesis of multiple sclerosis (MS). A hallmark of MS diagnosis is the presence of IgG Abs, which appear as oligoclonal bands in the cerebrospinal fluid. In this study, we demonstrate that myelin obtained post mortem from 8 out of 11 MS brain donors is bound by IgG Abs. Importantly, we show that IgG immune complexes strongly potentiate activation of primary human microglia by breaking their tolerance for microbial stimuli, such as LPS and Poly I:C, resulting in increased production of key proinflammatory cytokines, such as TNF and IL-1ß. We identified FcγRI and FcγRIIa as the two main responsible IgG receptors for the breaking of immune tolerance of microglia. Combined, these data indicate that IgG immune complexes potentiate inflammation by human microglia, which may play an important role in MS-associated inflammation and the formation of demyelinating lesions.


Asunto(s)
Complejo Antígeno-Anticuerpo/inmunología , Tolerancia Inmunológica/inmunología , Inmunoglobulina G/inmunología , Microglía/inmunología , Adulto , Anciano , Encéfalo/inmunología , Humanos , Inflamación/inmunología , Interleucina-1beta/inmunología , Persona de Mediana Edad , Esclerosis Múltiple/inmunología , Vaina de Mielina/inmunología , Poli I-C/inmunología , Receptores de IgG/inmunología , Factores de Necrosis Tumoral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA