RESUMEN
Intestinal microbiome perturbation characterizes Crohn's disease (CD), though specific contributors to pathophysiology remain elusive. In a recent issue of Science, Jain et al. show that Debaryomyces hansenii impairs intestinal healing in mice via effects on type I interferon signaling and chemokine CCL5 expression in macrophages and that it is also prevalent in the inflamed mucosa of CD patients.
Asunto(s)
Enfermedad de Crohn/inmunología , Enfermedad de Crohn/microbiología , Mucosa Intestinal/microbiología , Cicatrización de Heridas/inmunología , Animales , Quimiocina CCL5/inmunología , Microbioma Gastrointestinal/inmunología , Humanos , Interferón Tipo I/inmunología , Mucosa Intestinal/inmunología , Macrófagos/inmunología , Ratones , Micosis/inmunología , Micosis/microbiología , Saccharomycetales/inmunología , Transducción de Señal/inmunologíaRESUMEN
Focal adhesion kinase (FAK) promotes anti-tumor immune evasion. Specifically, the kinase activity of nuclear-targeted FAK in squamous cell carcinoma (SCC) cells drives exhaustion of CD8(+) T cells and recruitment of regulatory T cells (Tregs) in the tumor microenvironment by regulating chemokine/cytokine and ligand-receptor networks, including via transcription of Ccl5, which is crucial. These changes inhibit antigen-primed cytotoxic CD8(+) T cell activity, permitting growth of FAK-expressing tumors. Mechanistically, nuclear FAK is associated with chromatin and exists in complex with transcription factors and their upstream regulators that control Ccl5 expression. Furthermore, FAK's immuno-modulatory nuclear activities may be specific to cancerous squamous epithelial cells, as normal keratinocytes do not have nuclear FAK. Finally, we show that a small-molecule FAK kinase inhibitor, VS-4718, which is currently in clinical development, also drives depletion of Tregs and promotes a CD8(+) T cell-mediated anti-tumor response. Therefore, FAK inhibitors may trigger immune-mediated tumor regression, providing previously unrecognized therapeutic opportunities.
Asunto(s)
Carcinoma de Células Escamosas/inmunología , Quimiocina CCL5/genética , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Neoplasias Cutáneas/inmunología , Linfocitos T Reguladores/inmunología , Escape del Tumor , Aminopiridinas/administración & dosificación , Animales , Carcinoma de Células Escamosas/metabolismo , Quimiocina CCL5/inmunología , Modelos Animales de Enfermedad , Proteína-Tirosina Quinasas de Adhesión Focal/antagonistas & inhibidores , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Humanos , Queratinocitos/metabolismo , Ratones , Ratones Desnudos , Neoplasias Cutáneas/metabolismo , Transcripción GenéticaRESUMEN
The transcription regulator YAP controls organ size by regulating cell growth, proliferation and apoptosis. However, whether YAP has a role in innate antiviral immunity is largely unknown. Here we found that YAP negatively regulated an antiviral immune response. YAP deficiency resulted in enhanced innate immunity, a diminished viral load, and morbidity in vivo. YAP blocked dimerization of the transcription factor IRF3 and impeded translocation of IRF3 to the nucleus after viral infection. Notably, virus-activated kinase IKKÉ phosphorylated YAP at Ser403 and thereby triggered degradation of YAP in lysosomes and, consequently, relief of YAP-mediated inhibition of the cellular antiviral response. These findings not only establish YAP as a modulator of the activation of IRF3 but also identify a previously unknown regulatory mechanism independent of the kinases Hippo and LATS via which YAP is controlled by the innate immune pathway.
Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Fibroblastos/inmunología , Quinasa I-kappa B/metabolismo , Inmunidad Innata/inmunología , Lisosomas/metabolismo , Macrófagos/inmunología , Fosfoproteínas/inmunología , Infecciones por Rhabdoviridae/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Sistemas CRISPR-Cas , Proteínas de Ciclo Celular , Quimiocina CCL5/genética , Quimiocina CCL5/inmunología , Quimiocina CXCL10/genética , Quimiocina CXCL10/inmunología , Técnica del Anticuerpo Fluorescente , Edición Génica , Células HEK293 , Células HeLa , Humanos , Immunoblotting , Inmunoprecipitación , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/inmunología , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/genética , Interferón beta/inmunología , Pulmón/inmunología , Pulmón/patología , Ratones , Microscopía Confocal , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Células RAW 264.7 , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Infecciones por Rhabdoviridae/patología , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/inmunología , Vesiculovirus , Carga Viral , Proteínas Señalizadoras YAPRESUMEN
Although interleukin 1 (IL-1) induces expression of the transcription factor IRF1 (interferon-regulatory factor 1), the roles of IRF1 in immune and inflammatory responses and mechanisms of its activation remain elusive. Here we found that IRF1 was essential for IL-1-induced expression of the chemokines CXCL10 and CCL5, which recruit mononuclear cells into sites of sterile inflammation. Newly synthesized IRF1 acquired Lys63 (K63)-linked polyubiquitination mediated by the apoptosis inhibitor cIAP2 that was enhanced by the bioactive lipid S1P. In response to IL-1, cIAP2 and the sphingosine kinase SphK1 (the enzyme that generates S1P) formed a complex with IRF1, which led to its activation. Thus, IL-1 triggered a hitherto unknown signaling cascade that controlled the induction of IRF1-dependent genes that encode molecules important for sterile inflammation.
Asunto(s)
Quimiocina CCL5/biosíntesis , Quimiocina CXCL10/biosíntesis , Factor 1 Regulador del Interferón/metabolismo , Interleucina-1/metabolismo , Transducción de Señal/inmunología , Animales , Quimiocina CCL5/inmunología , Quimiocina CXCL10/inmunología , Quimiotaxis de Leucocito/inmunología , Ensayo de Inmunoadsorción Enzimática , Humanos , Immunoblotting , Inmunoprecipitación , Inflamación/inmunología , Inflamación/metabolismo , Factor 1 Regulador del Interferón/inmunología , Interleucina-1/inmunología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Lisina , Ratones , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , UbiquitinaciónRESUMEN
Immune thrombocytopenia (ITP) is an autoimmune disease characterized by low platelet counts primarily due to antiplatelet autoantibodies. Anti-D is a donor-derived polyclonal Ab against the rhesus D Ag on erythrocytes used to treat ITP. Unfortunately, adverse inflammatory/hypersensitivity reactions and a Food and Drug Administration-issued black box warning have limited its clinical use. This underscores the imperative to understand the inflammatory pathway associated with anti-erythrocyte Ab-based therapies. TER119 is an erythrocyte-specific Ab with anti-D-like therapeutic activity in murine ITP, while also exhibiting a distinct inflammatory signature involving production of CCL2, CCL5, and CXCL9 but not IFN-γ. Therefore, TER119 has been used to elucidate the potential mechanism underlying the adverse inflammatory activity associated with anti-erythrocyte Ab therapy in murine ITP. Prior work has demonstrated that TER119 administration is associated with a dramatic decrease in body temperature and inflammatory cytokine/chemokine production. The work presented in the current study demonstrates that inhibiting the highly inflammatory platelet-activating factor (PAF) pathway with PAF receptor antagonists prevents TER119-driven changes in body temperature and inhibits the production of the CCL2, CCL5, and CXCL9 inflammatory cytokines in CD-1 mice. Phagocytic cells and a functional TER119 Fc region were found to be necessary for TER119-induced body temperature changes and increases in CXCL9 and CCL2. Taken together, this work reveals the novel requirement of the PAF pathway in causing adverse inflammatory activity associated with anti-erythrocyte Ab therapy in a murine model and provides a strategy of mitigating these potential reactions without altering therapeutic activity.
Asunto(s)
Quimiocina CCL2 , Eritrocitos , Inflamación , Factor de Activación Plaquetaria , Glicoproteínas de Membrana Plaquetaria , Púrpura Trombocitopénica Idiopática , Animales , Ratones , Factor de Activación Plaquetaria/inmunología , Púrpura Trombocitopénica Idiopática/inmunología , Púrpura Trombocitopénica Idiopática/tratamiento farmacológico , Eritrocitos/inmunología , Inflamación/inmunología , Glicoproteínas de Membrana Plaquetaria/antagonistas & inhibidores , Glicoproteínas de Membrana Plaquetaria/inmunología , Quimiocina CCL2/inmunología , Quimiocina CCL5/inmunología , Quimiocina CXCL9/inmunología , Receptores Acoplados a Proteínas G/inmunología , Transducción de Señal/inmunología , Ratones Endogámicos C57BL , Autoanticuerpos/inmunología , Modelos Animales de EnfermedadRESUMEN
Noncoding RNAs have been verified to regulate the infiltration of macrophages to accelerate tumor biological progression, however the regulation of macrophages by circular RNAs in hepatocellular carcinoma (HCC) remains unresolved. Using high-throughput RNA sequencing, we demonstrated that hsa_circ_0003410 was clearly upregulated in HCC. 5-Ethynyl-2'-deoxyuridine and transwell assays showed that hsa_circ_0003410 facilitated the proliferation and migration of HCC cells in vitro. We knocked down the expression of hsa_circ_0003410 in HepG2 cells and performed next-generation sequencing to determine possible target genes of hsa_circ_0003410. Kyoto Encyclopedia of Genes and Genomes analysis revealed that different genes were mainly enriched in immune-related pathways. Mechanistically, we identified CCL5 as the target gene of hsa_circ_0003410. RNA-FISH showed the co-expression of hsa_circ_0003410 and CCL5. Western blot and ELISA also verified that hsa_circ_0003410 could upregulate the expression of CCL5 protein. Flow cytometry and immunofluorescence assays indicated that CCL5 activated and recruited M2 macrophages and increased the ratio of M2/M1 macrophages to promote the progression of HCC. Animal experiments in vitro also confirmed our results. Taken together, our experiments revealed that noncoding RNAs play a critical role in the HCC microenvironment and can be considered as markers for the diagnosis and prognosis of HCC.
Asunto(s)
Carcinoma Hepatocelular/genética , Quimiocina CCL5/genética , Neoplasias Hepáticas/genética , MicroARNs/genética , ARN Circular/genética , Macrófagos Asociados a Tumores/inmunología , Animales , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Quimiocina CCL5/inmunología , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Ratones , Microambiente Tumoral/inmunologíaRESUMEN
Upon detection of viral infections, cells activate the expression of type I interferons (IFNs) and pro-inflammatory cytokines to control viral dissemination. As part of their antiviral response, cells also trigger the translational shutoff response which prevents translation of viral mRNAs and cellular mRNAs in a non-selective manner. Intriguingly, mRNAs encoding for antiviral factors bypass this translational shutoff, suggesting the presence of additional regulatory mechanisms enabling expression of the self-defence genes. Here, we identified the dsRNA binding protein ILF3 as an essential host factor required for efficient translation of the central antiviral cytokine, IFNB1, and a subset of interferon-stimulated genes. By combining polysome profiling and next-generation sequencing, ILF3 was also found to be necessary to establish the dsRNA-induced transcriptional and translational programs. We propose a central role for the host factor ILF3 in enhancing expression of the antiviral defence mRNAs in cellular conditions where cap-dependent translation is compromised.
Asunto(s)
Interferón beta/genética , Proteínas del Factor Nuclear 90/genética , Biosíntesis de Proteínas , ARN Bicatenario/genética , ARN Mensajero/genética , ARN Viral/genética , Células A549 , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/inmunología , Quimiocina CCL5/genética , Quimiocina CCL5/inmunología , Quimiocina CXCL10/genética , Quimiocina CXCL10/inmunología , Citocinas/genética , Citocinas/inmunología , Proteína 58 DEAD Box/genética , Proteína 58 DEAD Box/inmunología , Regulación de la Expresión Génica , Células HeLa , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Interferón beta/inmunología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/inmunología , Proteínas del Factor Nuclear 90/inmunología , Poli I-C/farmacología , Polirribosomas/efectos de los fármacos , Polirribosomas/genética , Polirribosomas/inmunología , ARN Bicatenario/antagonistas & inhibidores , ARN Bicatenario/metabolismo , ARN Mensajero/inmunología , ARN Viral/antagonistas & inhibidores , ARN Viral/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , Receptores Inmunológicos , Transducción de Señal , Ubiquitinas/genética , Ubiquitinas/inmunología , Replicación ViralRESUMEN
Inflammation is central to the pathogenesis of pulmonary vascular remodeling and pulmonary hypertension (PH). Inflammation precedes remodeling in preclinical models, thus supporting the concept that changes in immunity drive remodeling in PH. Platelets are recognized as mediators of inflammation, but whether platelets contribute to hypoxia-driven inflammation has not been studied. We utilized a murine hypoxia model to test the hypothesis that platelets drive hypoxia-induced inflammation. We evaluated male and female 9-wk-old normoxic and hypoxic mice and in selected experiments included hypoxic thrombocytopenic mice. Thrombocytopenic mice were generated with an anti-GP1bα rat IgG antibody. We also performed immunostaining of lung sections from failed donor controls and patients with idiopathic pulmonary arterial hypertension. We found that platelets are increased in the lungs of hypoxic mice and hypoxia induces platelet activation. Platelet depletion prevents hypoxia-driven increases in the proinflammatory chemokines CXCL4 and CCL5 and attenuates hypoxia-induced increase in plasma CSF-2. Pulmonary interstitial macrophages are increased in the lungs of hypoxic mice; this increase is prevented in thrombocytopenic mice. To determine the potential relevance to human disease, lung sections from donors and patients with advanced idiopathic pulmonary arterial hypertension (iPAH) were immunostained for the platelet-specific protein CD41. We observed iPAH lungs had a two-fold increase in CD41, compared with controls. Our data provide evidence that the platelet count is increased in the lungs and activated in mice with hypoxia-induced inflammation and provides rationale for the further study of the potential contribution of platelets to inflammatory mediated vascular remodeling and PH.
Asunto(s)
Plaquetas/inmunología , Hipoxia/inmunología , Pulmón/inmunología , Activación Plaquetaria/inmunología , Neumonía/inmunología , Animales , Plaquetas/patología , Quimiocina CCL5/inmunología , Modelos Animales de Enfermedad , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Hipoxia/patología , Inflamación/inmunología , Inflamación/patología , Pulmón/patología , Masculino , Ratones , Factor Plaquetario 4/inmunología , Neumonía/patología , Trombocitopenia/inducido químicamente , Trombocitopenia/inmunología , Trombocitopenia/patologíaRESUMEN
Fulminant hepatic failure (FHF) is a lethal manifestation of hepatitis A virus (HAV) infection, whose underlying mechanisms are poorly understood. We aimed to evaluate the importance of the modulation of the RANTES-chemokine receptor type 5 (CCR5) signaling axis and its immunomodulatory effects in directing hepatitis A disease pathogenesis using an in silico, in vitro and patient cohort-based approach. In silico interaction studies were performed using computation approaches with suitable software. Differential expression of relevant cytokines and immune cell markers were studied using real-time quantitative reverse transcription PCR (qRT-PCR), enzyme-linked immunosorbent assay, and flow-cytometry-based methods. In the HepG2 cell line, we studied inflammatory responses and susceptibility to HAV infection following RANTES stimulation and antibody blockade of CCR5. The HAV-VP3 region exhibited high interaction in CCR5: HAV complexes. RANTES levels were significantly increased in FHF cases. Reduced monocyte and T-cell activation were observed in FHF cases. RANTES expression inversely correlated with viremia but positively correlated with proinflammatory responses. Hyper Th1-biased immune responses, marked by high interleukin (IL)-12/IL-10 ratio were observed in FHF cases, which were also characterized by upregulated tumor necrosis factor-alpha (TNF-α) expression and reduced interferon-gamma expression. In vitro, RANTES was protective against HAV infection but resulted in upregulated TNF-α expression. Although viral load increased upon the regulation of inflammatory responses by CCR5 blocking, it was still significantly lower compared to control HAV-infected cells. Our study suggests the importance of RANTES-CCR5 signaling and linked-immunomodulation in HAV disease pathogenesis, as well as highlights the utility of CCR5 antagonists as a risk-reduction strategy in FHF patients. Our findings, therefore, have important implications for the management of high-risk HAV infections.
Asunto(s)
Quimiocina CCL5/genética , Quimiocina CCL5/inmunología , Virus de la Hepatitis A/inmunología , Hepatitis A/inmunología , Receptores CCR5/genética , Receptores CCR5/inmunología , Adulto , Quimiocina CCL5/farmacología , Estudios de Cohortes , Simulación por Computador , Femenino , Células Hep G2 , Hepatitis A/virología , Hepatocitos/efectos de los fármacos , Humanos , Inmunomodulación , Fallo Hepático Agudo , Masculino , Persona de Mediana Edad , Pronóstico , Carga ViralRESUMEN
Atopic dermatitis (AD) is a chronic inflammatory skin disease that is characterized by an impaired skin barrier and intense itchiness, which decreases the individual's quality of life. No fully effective therapeutic agents have prevailed for AD due to an insufficient grasp of the complex etiology. Ellagic acid (EA), a natural compound, has anti-inflammatory properties in chronic diseases. The effects of EA on AD have not yet been explored. The present study investigated the effects of EA on TNF-α/IFN-γ-stimulated HaCaT keratinocytes and house dust mite-induced AD-like skin lesions in NC/Nga mice. Treatment with EA suppressed inflammatory responses in keratinocytes by regulating critical inflammatory signaling pathways, such as mitogen-activated protein kinases and signal transducers and activators of transcription. In vivo studies using a DfE-induced AD mouse model showed the effects of EA administration through ameliorated skin lesions via decremented histological inflammatory reactions. These results suggest that EA could be a potential therapeutic alternative for the treatment of AD by inhibiting inflammatory signaling pathways.
Asunto(s)
Antiinflamatorios/farmacología , Dermatitis Atópica/tratamiento farmacológico , Dermatophagoides farinae/química , Ácido Elágico/farmacología , Proteínas Quinasas Activadas por Mitógenos/genética , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT3/genética , Animales , Antígenos Dermatofagoides/administración & dosificación , Quimiocina CCL17/genética , Quimiocina CCL17/inmunología , Quimiocina CCL22/genética , Quimiocina CCL22/inmunología , Quimiocina CCL5/genética , Quimiocina CCL5/inmunología , Mezclas Complejas/administración & dosificación , Citocinas/genética , Citocinas/inmunología , Dermatitis Atópica/inducido químicamente , Dermatitis Atópica/genética , Dermatitis Atópica/inmunología , Dermatophagoides farinae/inmunología , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Células HaCaT , Humanos , Interferón gamma/antagonistas & inhibidores , Interferón gamma/farmacología , Ratones , Proteínas Quinasas Activadas por Mitógenos/inmunología , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT3/inmunología , Transducción de Señal , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/farmacología , Linfopoyetina del Estroma TímicoRESUMEN
Chlamydia trachomatis infection of the human fallopian tubes can lead to damaging inflammation and scarring, ultimately resulting in infertility. To study the human cellular responses to chlamydial infection, researchers have frequently used transformed cell lines that can have limited translational relevance. We developed a primary human fallopian tube epithelial cell model based on a method previously established for culture of primary human bronchial epithelial cells. After protease digestion and physical dissociation of excised fallopian tubes, epithelial cell precursors were expanded in growth factor-containing medium. Expanded cells were cryopreserved to generate a biobank of cells from multiple donors and cultured at an air-liquid interface. Culture conditions stimulated cellular differentiation into polarized mucin-secreting and multiciliated cells, recapitulating the architecture of human fallopian tube epithelium. The polarized and differentiated cells were infected with a clinical isolate of C. trachomatis, and inclusions containing chlamydial developmental forms were visualized by fluorescence and electron microscopy. Apical secretions from infected cells contained increased amounts of proteins associated with chlamydial growth and replication, including transferrin receptor protein 1, the amino acid transporters SLC3A2 and SLC1A5, and the T-cell chemoattractants CXCL10, CXCL11, and RANTES. Flow cytometry revealed that chlamydial infection induced cell surface expression of T-cell homing and activation proteins, including ICAM-1, VCAM-1, HLA class I and II, and interferon gamma receptor. This human fallopian tube epithelial cell culture model is an important tool with translational potential for studying cellular responses to Chlamydia and other sexually transmitted pathogens.
Asunto(s)
Células Epiteliales/inmunología , Regulación de la Expresión Génica/inmunología , Interacciones Microbiota-Huesped/inmunología , Linfocitos T/inmunología , Adulto , Sistema de Transporte de Aminoácidos ASC/genética , Sistema de Transporte de Aminoácidos ASC/inmunología , Antígenos CD/genética , Antígenos CD/inmunología , Biomarcadores/metabolismo , Quimiocina CCL5/genética , Quimiocina CCL5/inmunología , Quimiocina CXCL10/genética , Quimiocina CXCL10/inmunología , Quimiocina CXCL11/genética , Quimiocina CXCL11/inmunología , Infecciones por Chlamydia/genética , Infecciones por Chlamydia/inmunología , Infecciones por Chlamydia/microbiología , Chlamydia trachomatis/crecimiento & desarrollo , Chlamydia trachomatis/inmunología , Células Epiteliales/microbiología , Trompas Uterinas/citología , Trompas Uterinas/cirugía , Femenino , Cadena Pesada de la Proteína-1 Reguladora de Fusión/genética , Cadena Pesada de la Proteína-1 Reguladora de Fusión/inmunología , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Interacciones Microbiota-Huesped/genética , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/inmunología , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/inmunología , Modelos Biológicos , Cultivo Primario de Células , Receptores de Interferón/genética , Receptores de Interferón/inmunología , Receptores de Transferrina/genética , Receptores de Transferrina/inmunología , Salpingectomía , Linfocitos T/microbiología , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/inmunología , Receptor de Interferón gammaRESUMEN
We previously demonstrated that circulating extracellular vesicles (EVs) from patients with valvular heart disease (VHD; vEVs) contain inflammatory components and inhibit endothelium-dependent vasodilation. Neutrophil chemotaxis plays a key role in renal dysfunction, and dexmedetomidine (DEX) can reduce renal dysfunction in cardiac surgery. However, the roles of vEVs in neutrophil chemotaxis and effects of DEX on vEVs are unknown. Here, we investigated the impact of vEVs on neutrophil chemotaxis in kidneys and the influence of DEX on vEVs. Circulating EVs were isolated from healthy subjects and patients with VHD. The effects of EVs on chemokine generation, forkhead box protein O3a (FOXO3a) pathway activation and neutrophil chemotaxis on cultured human umbilical vein endothelial cells (HUVECs) and kidneys in mice and the influence of DEX on EVs were detected. vEVs increased FOXO3a expression, decreased phosphorylation of Akt and FOXO3a, promoted FOXO3a nuclear translocation, and activated the FOXO3a signaling pathway in vitro. DEX pretreatment reduced vEV-induced CXCL4 and CCL5 expression and neutrophil chemotaxis in cultured HUVECs via the FOXO3a signaling pathway. vEVs were also found to suppress Akt phosphorylation and activate FOXO3a signaling to increase plasma levels of CXCL4 and CCL5 and neutrophil accumulation in kidney. The overall mechanism was inhibited in vivo with DEX pretreatment. Our data demonstrated that vEVs induced CXCL4-CCL5 to stimulate neutrophil infiltration in kidney, which can be inhibited by DEX via the FOXO3a signaling. Our findings reveal a unique mechanism involving vEVs in inducing neutrophils chemotaxis and may provide a novel basis for using DEX in reducing renal dysfunction in valvular heart surgery.
Asunto(s)
Quimiotaxis de Leucocito/inmunología , Vesículas Extracelulares/inmunología , Enfermedades de las Válvulas Cardíacas/inmunología , Células Endoteliales de la Vena Umbilical Humana/inmunología , Riñón/inmunología , Neutrófilos/inmunología , Insuficiencia Renal/inmunología , Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Adulto , Animales , Estudios de Casos y Controles , Quimiocina CCL5/efectos de los fármacos , Quimiocina CCL5/inmunología , Quimiocina CCL5/metabolismo , Quimiotaxis de Leucocito/efectos de los fármacos , Dexmedetomidina/farmacología , Vesículas Extracelulares/efectos de los fármacos , Vesículas Extracelulares/metabolismo , Femenino , Proteína Forkhead Box O3/efectos de los fármacos , Proteína Forkhead Box O3/inmunología , Proteína Forkhead Box O3/metabolismo , Enfermedades de las Válvulas Cardíacas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inflamación , Riñón/efectos de los fármacos , Riñón/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Neutrófilos/efectos de los fármacos , Fosforilación , Factor Plaquetario 4/efectos de los fármacos , Factor Plaquetario 4/inmunología , Factor Plaquetario 4/metabolismo , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Insuficiencia Renal/metabolismo , VasodilataciónRESUMEN
Upper respiratory tract is the primary site of SARS-CoV-2 replication. Releasing of pro and anti-inflammatory mediators plays an important role in the immunopathogenesis of Coronavirus Disease 2019 (COVID-19). The aim of this study was to evaluate the early inflammatory response in upper airway by measuring of IFN-γ, TGF-ß1 and RANTES at mRNA level. Forty five SARS-CoV-2 infected patients were enrolled, whose were divided in two groups: asymptomatic and symptomatic. Twenty healthy persons, SARS-CoV-2 negative were included as controls. Higher IFN-γ expression was detected in SARS-CoV-2 infected patients in comparison with controls (p = 0.0393). IFN-γ expression was increased in symptomatic patients (p = 0.0405). TGF-ß1 and RANTES expressions were lower in SARS-CoV-2 infected patients than controls (p < 0.0001; p = 0.0011, respectively). A significant correlation between IFN-γ and TGF-ß1 was observed in SARS-CoV-2 asymptomatic patients (r = +0.61, p = 0.0014). The findings suggest that imbalance between IFN-γ and TGF-ß1 expression could be an impact in clinical expression of SARS-CoV-2 infection.
Asunto(s)
Betacoronavirus/patogenicidad , Quimiocina CCL5/genética , Infecciones por Coronavirus/inmunología , Interferón gamma/genética , Neumonía Viral/inmunología , ARN Mensajero/genética , Factor de Crecimiento Transformador beta1/genética , Adulto , Enfermedades Asintomáticas , Betacoronavirus/inmunología , COVID-19 , Estudios de Casos y Controles , Quimiocina CCL5/inmunología , Infecciones por Coronavirus/diagnóstico , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Femenino , Expresión Génica , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Interferón gamma/inmunología , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Masculino , Persona de Mediana Edad , Nasofaringe/inmunología , Nasofaringe/patología , Nasofaringe/virología , Pandemias , Neumonía Viral/diagnóstico , Neumonía Viral/patología , Neumonía Viral/virología , ARN Mensajero/inmunología , SARS-CoV-2 , Índice de Severidad de la Enfermedad , Factor de Crecimiento Transformador beta1/inmunologíaRESUMEN
An increase in immunosuppressive myeloid-derived suppressor cells (MDSCs) is associated with disease progression and treatment resistance in multiple myeloma (MM). We investigated the mechanisms underlying MDSC induction, and sought to discover a strategy for prevention of MDSC induction in MM. Using a transwell co-culture system, four of nine examined human myeloma-derived cell lines (HMCLs) were potent in inducing monocytic (M)-MDSCs from normal peripheral blood mononuclear cells (PBMCs). As the results, we identified that secretion of C-C motif chemokine ligand 5 (CCL5) and macrophage migration inhibitory factor (MIF) by myeloma cells is a prerequisite for induction of MDSCs in MM. The immunomodulatory drug (IMiD) compounds, such as lenalidomide (LEN) and pomalidomide (POM), were identified as potent inhibitors of MDSC induction through bidirectional molecular effects of cereblon (CRBN)-dependent and -independent downregulation of CCL5 and MIF in myeloma cells; and downregulation of C-C motif chemokine receptor 5, a receptor for CCL5, and induction of interferon regulatory factor 8, a critical transcription factor for monocytic differentiation, in PBMCs. In the present study of the molecular mechanisms underlying MDSC induction, we identified a novel effect of LEN and POM of inhibiting MDSC induction via overlapping regulatory effects in myeloma cells and normal PBMCs.
Asunto(s)
Lenalidomida/farmacología , Mieloma Múltiple/inmunología , Células Supresoras de Origen Mieloide/inmunología , Talidomida/análogos & derivados , Línea Celular Tumoral , Quimiocina CCL5/inmunología , Técnicas de Cocultivo , Humanos , Factores Reguladores del Interferón/inmunología , Oxidorreductasas Intramoleculares/inmunología , Factores Inhibidores de la Migración de Macrófagos/inmunología , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/patología , Células Supresoras de Origen Mieloide/patología , Proteínas de Neoplasias/inmunología , Talidomida/farmacologíaRESUMEN
Dendritic cells (DCs) are activated by pathogens to initiate and shape immune responses. We found that the activation of DCs by Plasmodium falciparum, the main causative agent of human malaria, induces a highly unusual phenotype by which DCs up-regulate costimulatory molecules and secretion of chemokines, but not of cytokines typical of inflammatory responses (IL-1ß, IL-6, IL-10, TNF). Similar results were obtained with DCs obtained from malaria-naïve US donors and malaria-experienced donors from Mali. Contact-dependent cross-talk between the main DC subsets, plasmacytoid and myeloid DCs (mDCs) was necessary for increased chemokine and IFN-α secretion in response to the parasite. Despite the absence of inflammatory cytokine secretion, mDCs incubated with P. falciparum-infected erythrocytes activated antigen-specific naïve CD4+ T cells to proliferate and secrete Th1-like cytokines. This unexpected response of human mDCs to P. falciparum exhibited a transcriptional program distinct from a classical LPS response, pointing to unique P. falciparum-induced activation pathways that may explain the uncharacteristic immune response to malaria.
Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Eritrocitos/parasitología , Interacciones Huésped-Parásitos , Activación de Linfocitos , Plasmodium falciparum/metabolismo , Antígenos CD/genética , Antígenos CD/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/efectos de los fármacos , Quimiocina CCL2/genética , Quimiocina CCL2/inmunología , Quimiocina CCL5/genética , Quimiocina CCL5/inmunología , Quimiocina CXCL10/genética , Quimiocina CXCL10/inmunología , Quimiocina CXCL9/genética , Quimiocina CXCL9/inmunología , Técnicas de Cocultivo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/parasitología , Regulación de la Expresión Génica , Antígenos HLA-DR/genética , Antígenos HLA-DR/inmunología , Humanos , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Lipopolisacáridos/farmacología , Malaria Falciparum/genética , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Malí , Plasmodium falciparum/crecimiento & desarrollo , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunologíaRESUMEN
OBJECTIVES: Yttrium-90 (Y90)-radioembolisation (RE) significantly regresses locally advanced hepatocellular carcinoma and delays disease progression. The current study is designed to deeply interrogate the immunological impact of Y90-RE, which elicits a sustained therapeutic response. DESIGN: Time-of-flight mass cytometry and next-generation sequencing (NGS) were used to analyse the immune landscapes of tumour-infiltrating leucocytes (TILs), tumour tissues and peripheral blood mononuclear cells (PBMCs) at different time points before and after Y90-RE. RESULTS: TILs isolated after Y90-RE exhibited signs of local immune activation: higher expression of granzyme B (GB) and infiltration of CD8+ T cells, CD56+ NK cells and CD8+ CD56+ NKT cells. NGS confirmed the upregulation of genes involved in innate and adaptive immune activation in Y90-RE-treated tumours. Chemotactic pathways involving CCL5 and CXCL16 correlated with the recruitment of activated GB+CD8+ T cells to the Y90-RE-treated tumours. When comparing PBMCs before and after Y90-RE, we observed an increase in tumour necrosis factor-α on both the CD8+ and CD4+ T cells as well as an increase in percentage of antigen-presenting cells after Y90-RE, implying a systemic immune activation. Interestingly, a high percentage of PD-1+/Tim-3+CD8+ T cells coexpressing the homing receptors CCR5 and CXCR6 denoted Y90-RE responders. A prediction model was also built to identify sustained responders to Y90-RE based on the immune profiles from pretreatment PBMCs. CONCLUSION: High-dimensional analysis of tumour and systemic immune landscapes identified local and systemic immune activation that corresponded to the sustained response to Y90-RE. Potential biomarkers associated with a positive clinical response were identified and a prediction model was built to identify sustained responders prior to treatment.
Asunto(s)
Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/radioterapia , Leucocitos Mononucleares/inmunología , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/radioterapia , Radioisótopos de Itrio , Células Presentadoras de Antígenos/inmunología , Biomarcadores de Tumor/inmunología , Quimiocina CCL5/inmunología , Quimiocina CXCL16/inmunología , Progresión de la Enfermedad , Femenino , Citometría de Flujo/métodos , Granzimas/inmunología , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Singapur , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/inmunologíaRESUMEN
Reproductive tract pathology caused by Chlamydia trachomatis infection is an important global cause of human infertility. To better understand the mechanisms associated with Chlamydia-induced genital tract pathogenesis in humans, we used CRISPR genome editing to disrupt Toll-like receptor 3 (TLR3) function in the human oviduct epithelial (hOE) cell line OE-E6/E7 in order to investigate the possible role(s) of TLR3 signaling in the immune response to Chlamydia Disruption of TLR3 function in these cells significantly diminished the Chlamydia-induced synthesis of several inflammation biomarkers, including interferon beta (IFN-ß), interleukin-6 (IL-6), interleukin-6 receptor alpha (IL-6Rα), soluble interleukin-6 receptor beta (sIL-6Rß, or gp130), IL-8, IL-20, IL-26, IL-34, soluble tumor necrosis factor receptor 1 (sTNF-R1), tumor necrosis factor ligand superfamily member 13B (TNFSF13B), matrix metalloproteinase 1 (MMP-1), MMP-2, and MMP-3. In contrast, the Chlamydia-induced synthesis of CCL5, IL-29 (IFN-λ1), and IL-28A (IFN-λ2) was significantly increased in TLR3-deficient hOE cells compared to their wild-type counterparts. Our results indicate a role for TLR3 signaling in limiting the genital tract fibrosis, scarring, and chronic inflammation often associated with human chlamydial disease. Interestingly, we saw that Chlamydia infection induced the production of biomarkers associated with persistence, tumor metastasis, and autoimmunity, such as soluble CD163 (sCD163), chitinase-3-like protein 1, osteopontin, and pentraxin-3, in hOE cells; however, their expression levels were significantly dysregulated in TLR3-deficient hOE cells. Finally, we demonstrate using hOE cells that TLR3 deficiency resulted in an increased amount of chlamydial lipopolysaccharide (LPS) within Chlamydia inclusions, which is suggestive that TLR3 deficiency leads to enhanced chlamydial replication and possibly increased genital tract pathogenesis during human infection.
Asunto(s)
Chlamydia trachomatis/inmunología , Células Epiteliales/microbiología , Regulación de la Expresión Génica/inmunología , Interacciones Huésped-Patógeno/inmunología , Receptor Toll-Like 3/inmunología , Factor Activador de Células B/genética , Factor Activador de Células B/inmunología , Línea Celular Transformada , Quimiocina CCL5/genética , Quimiocina CCL5/inmunología , Chlamydia trachomatis/crecimiento & desarrollo , Chlamydia trachomatis/patogenicidad , Receptor gp130 de Citocinas/genética , Receptor gp130 de Citocinas/inmunología , Células Epiteliales/inmunología , Trompas Uterinas/inmunología , Trompas Uterinas/microbiología , Femenino , Eliminación de Gen , Células HeLa , Interacciones Huésped-Patógeno/genética , Humanos , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Interleucinas/genética , Interleucinas/inmunología , Metaloproteinasa 1 de la Matriz/genética , Metaloproteinasa 1 de la Matriz/inmunología , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/inmunología , Transducción de Señal , Receptor Toll-Like 3/deficiencia , Receptor Toll-Like 3/genéticaRESUMEN
Although Parkinson's disease (PD) is a progressive neurodegenerative disease, the disease does not progress or persist in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model, the most common animal model of PD. Recently, we have described that supplementation of regulated on activation, normal T cell expressed and secreted (RANTES), a chemokine known to drive infiltration of T cells, induces persistent nigrostriatal pathology in MPTP mouse model. However, which particular T cell subsets are recruited to the substantia nigra (SN) by RANTES is not known. Here, by adoptive transfer of different subset of T cells from tomato red transgenic mice to MPTP-intoxicated immunodeficient Rag1-/- mice, we describe that invasion of Th17 cells into the SN is stimulated by exogenous RANTES administration. On the other hand, RANTES supplementation remained unable to influence the infiltration of Th1 and Tregs into the SN of MPTP-insulted Rag1-/- mice. Accordingly, RANTES supplementation increased MPTP-induced TH cell loss in Rag1-/-mice receiving Th17, but neither Th1 nor Tregs. RANTES-mediated aggravation of nigral TH neurons also paralleled with significant DA loss in striatum and locomotor deficits in MPTP-intoxicated Rag1-/- mice receiving Th17 cells. Finally, we demonstrate that levels of IL-17 (a Th17-specific cytokine) and RANTES are higher in serum of PD patients than age-matched controls and that RANTES positively correlated with IL-17 in serum of PD patients. Together, these results highlight the importance of RANTES-Th17 pathway in progressive dopaminergic neuronal loss and associated PD pathology.
Asunto(s)
Quimiocina CCL5/inmunología , Neuronas Dopaminérgicas/patología , Trastornos Parkinsonianos/inmunología , Sustancia Negra/inmunología , Células Th17/inmunología , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Degeneración Nerviosa/inmunología , Degeneración Nerviosa/patología , Enfermedad de Parkinson/inmunología , Enfermedad de Parkinson/patología , Trastornos Parkinsonianos/patología , Sustancia Negra/patologíaRESUMEN
BACKGROUND: Chemotherapy with Docetaxel (Doc) is efficient in a subset of prostate cancer (PCa) cases; however, most patients ultimately develop resistance to Docetaxel. The tumor immune microenvironment and secreted cytokines play a substantial role in development of resistance to chemotherapy. Our previous study has demonstrated that CD4+ T cells in prostate tumor microenvironment contribute to PCa progression; meanwhile, we found increased CD4+ T-cell infiltration in tumor area after Doc treatment; however, their effects on PCa chemosensitivity remain unclear. Here, we aim to explore the role and mechanisms of CD4+ T cells in PCa chemotherapy sensitivity. METHODS: CD4+ T-cell infiltration in Doc-treated paraffin-embedded specimens from transurethral resection of prostate, radical prostatectomy, or bone metastasis was detected by immunohistochemistry. The castration-resistant PCa cell lines-C4-2 and CWR22RV1, and CD4+ T-cell lines-HH and Molt-3 were used in the coculture system. After coculture with the lymphocytes, PCa cell chemosensitivity was detected by cell counting kit-8, terminal deoxynucleotidyl transferase dUTP nick-end labeling assays, and Western blot analysis. Various cell cytokines were determined by cytokine arrays and reverse-transcription polymerase chain reaction. The recombinant human C-C motif chemokine ligand 5 (CCL5) was added to PCa cells for further confirming its effects and anti-CCL5 antibody was used for neutralization. S3I-201, a signal transducer and activator of transcription 3 (STAT3) inhibitor, was added to the coculture system to detect STAT3 role in chemosensitivity. Tumor xenografts in nude mice were used for confirming effects of CD4+ T cells in vivo study. RESULTS: We found more infiltrated CD4+ T cells in human PCa lesions than in the adjacent noncancerous tissues after Doc treatment. In vitro cell line study confirmed that CD4+ T cells increase the PCa Doc resistance. Quantative polymerase chain reaction and cytokine arrays indicated that after coculture with PCa, CD4+ T cells could secrete large amounts of CCL5. Moreover, CCL5 stimulation enhanced PCa resistance to Doc, and anti-CCL5 antibody could partly reverse this process. We found that CD4+ T cells could activate P-STAT3 signaling via secreting CCL5 and adding a STAT3 inhibitor can reverse the chemoresistance. In vivo mouse model with xenografted 22RV1 cells and CD4+ T cells also confirmed the in vitro results. CONCLUSIONS: Together, our results indicate that infiltrating CD4+ T cells could promote PCa chemotherapy resistance via modulation of the CCL5/STAT3 signaling pathway.
Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Quimiocina CCL5/inmunología , Docetaxel/farmacología , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/inmunología , Anciano , Anciano de 80 o más Años , Antineoplásicos/farmacología , Línea Celular Tumoral , Quimiocina CCL5/metabolismo , Resistencia a Antineoplásicos , Humanos , Masculino , Persona de Mediana Edad , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Factor de Transcripción STAT3/inmunología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/inmunología , Microambiente TumoralRESUMEN
We aimed to evaluate the significance of the RANTES-CCR5 axis and resulting immunomodulatory status in Dengue pathogenesis involving a Guwahati, India based population where Dengue cases have increased alarmingly. An increased CC-chemokine receptor type 5 (CCR5) messenger RNA expression and CCR5 positive cell count profile was observed in Dengue cases, the highest being in severe cases. CCR5 ligand RANTES expression was significantly decreased in Dengue cases and inversely correlated with Dengue viremia fold change in severe cases. Monocytes are involved in Dengue virus homing and replication. Its levels and activation profile were higher in Dengue cases. A hyper Th1-biased immunomodulatory profile with upregulated tumor necrosis factor-α levels, and downregulated expression of antiviral cytokine interferon-γ and key regulatory Th2 anti-inflammatory cytokine interleukin 10 was observed in severe Dengue cases compared with mild Dengue cases and controls. The results, therefore, suggest the significance of RANTES-CCR5 axis deregulation and resulting altered immunomodulation in Dengue pathogenesis, and holds prognostic and therapeutic significance.