Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(24): e2305245120, 2023 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-37276392

RESUMEN

The activation and expansion of T cells that recognize cancer cells is an essential aspect to antitumor immunity. Tumors may escape destruction by the immune system through ectopic expression of inhibitory immune ligands typically exemplified by the PD-L1/PD-1 pathway. Here, we reveal another facet of tumor evasion from T cell surveillance. By secretome profiling of necrotic tumor cells, we identified an oncometabolite spermidine as a unique inhibitor of T cell receptor (TCR) signaling. Mechanistically, spermidine causes the downregulation of the plasma membrane cholesterol levels, resulting in the suppression of TCR clustering. Using syngeneic mouse models, we show that spermidine is abundantly detected in the tumor immune microenvironment (TIME) and that administration of the polyamine synthesis inhibitor effectively enhanced CD8+ T cell-dependent antitumor responses. Further, the combination of the polyamine synthesis inhibitor with anti-PD-1 immune checkpoint antibody resulted in a much stronger antitumor immune response. This study reveals an aspect of immunosuppressive TIME, wherein spermidine functions as a metabolic T cell checkpoint that may offer a unique approach for promoting tumor immunotherapy.


Asunto(s)
Antineoplásicos , Neoplasias , Animales , Ratones , Espermidina/farmacología , Espermidina/metabolismo , Linfocitos T CD8-positivos , Neoplasias/metabolismo , Antineoplásicos/farmacología , Inmunoterapia/métodos , Receptores de Antígenos de Linfocitos T/metabolismo , Microambiente Tumoral , Línea Celular Tumoral , Antígeno B7-H1/metabolismo
2.
J Exp Med ; 220(8)2023 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-37163450

RESUMEN

Group 2 innate lymphoid cells (ILC2s) expressing IL-5 and IL-13 are localized at various mucosal tissues and play critical roles in the induction of type 2 inflammation, response to helminth infection, and tissue repair. Here, we reveal a unique ILC2 subset in the mouse intestine that constitutively expresses IL-4 together with GATA3, ST2, KLRG1, IL-17RB, and IL-5. In this subset, IL-4 expression is regulated by mechanisms similar to but distinct from those observed in T cells and is partly affected by IL-25 signaling. Although the absence of the microbiota had marginal effects, feeding mice with a vitamin B1-deficient diet compromised the number of intestinal IL-4+ ILC2s. The decrease in the number of IL-4+ ILC2s caused by the vitamin B1 deficiency was accompanied by a reduction in IL-25-producing tuft cells. Our findings reveal that dietary vitamin B1 plays a critical role in maintaining interaction between tuft cells and IL-4+ ILC2s, a previously uncharacterized immune cell population that may contribute to maintaining intestinal homeostasis.


Asunto(s)
Dieta , Mucosa Intestinal , Tiamina , Animales , Ratones , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Tiamina/metabolismo , Organismos Libres de Patógenos Específicos , Ratones Endogámicos C57BL , Interleucina-4/metabolismo , Microbioma Gastrointestinal , Organoides/citología , Organoides/inmunología , Ácido Trinitrobencenosulfónico
3.
Genes Cells ; 28(3): 202-210, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36550748

RESUMEN

High-mobility group box 1 (HMGB1) is a nucleotide-binding chromatin protein that has also been characterized as a prototypical damage-associate molecular pattern. It triggers inflammatory responses upon release from damaged or dying cells. In fact, HMGB1 has been linked to the induction of many inflammatory diseases through immune cell activation including neutrophil recruitment. In this study, we examined the impact of HMGB1-binding inhibitory oligodeoxynucleotide (ISM ODN) on the development of hepatitis using a murine model of the disease. Our results indicate that ISM ODN effectively suppresses pathological features of hepatitis, including neutrophil accumulation. This study therefore may offer clinical insight into the treatment of hepatitis and possibly other inflammatory diseases.


Asunto(s)
Proteína HMGB1 , Hepatitis , Ratones , Animales , Proteína HMGB1/metabolismo , Oligodesoxirribonucleótidos/farmacología , Modelos Animales de Enfermedad , Infiltración Neutrófila
4.
Sci Rep ; 11(1): 20231, 2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34642357

RESUMEN

Increase of the enteric bacteriophages (phage), components of the enteric virome, has been associated with the development of inflammatory bowel diseases. However, little is known about how a given phage contributes to the regulation of intestinal inflammation. In this study, we isolated a new phage associated with Enterococcus gallinarum, named phiEG37k, the level of which was increased in C57BL/6 mice with colitis development. We found that, irrespective of the state of inflammation, over 95% of the E. gallinarum population in the mice contained phiEG37k prophage within their genome and the phiEG37k titers were proportional to that of E. gallinarum in the gut. To explore whether phiEG37k impacts intestinal homeostasis and/or inflammation, we generated mice colonized either with E. gallinarum with or without the prophage phiEG37k. We found that the mice colonized with the bacteria with phiEG37k produced more Mucin 2 (MUC2) that serves to protect the intestinal epithelium, as compared to those colonized with the phage-free bacteria. Consistently, the former mice were less sensitive to experimental colitis than the latter mice. These results suggest that the newly isolated phage has the potential to protect the host by strengthening mucosal integrity. Our study may have clinical implication in further understanding of how bacteriophages contribute to the gut homeostasis and pathogenesis.


Asunto(s)
Bacteriófagos/clasificación , Colitis/microbiología , Enterococcus/patogenicidad , Mucina 2/metabolismo , Animales , Bacteriófagos/genética , Bacteriófagos/aislamiento & purificación , Colitis/inmunología , Modelos Animales de Enfermedad , Enterococcus/virología , Genoma Viral , Secuenciación de Nucleótidos de Alto Rendimiento , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratones , Ratones Endogámicos C57BL , Filogenia , Secuenciación Completa del Genoma
5.
Mol Ther Nucleic Acids ; 25: 708-715, 2021 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-34589288

RESUMEN

RNA interference-based gene silencing drugs are attracting attention for treating various diseases. Lipid nanoparticles (LNPs) are carriers that efficiently deliver small interfering RNA (siRNA) to the cytoplasm of target cells. Recently, we developed potent and well-tolerated biodegradable LNPs with asymmetric ionizable lipids. Here, we evaluated the effect of LNPs on immune cells in mice. After intravenous administration, LNPs were efficiently incorporated into several tissue-resident macrophages, including liver macrophages, through an apolipoprotein E (ApoE)-independent mechanism. Administration of LNP-encapsulated siRNA against Irf5, encoding the transcription factor critical for inflammatory responses, sharply reduced its expression in macrophages in vivo, and persisted for as long as 7 days. The therapeutic potential of Irf5 siRNA-loaded LNPs in inflammatory diseases was tested in a concanavalin A (Con A)-induced hepatitis model, whose pathogenic mechanisms are dependent on cytokine secretion from macrophages. We found that Con A-induced liver injury was significantly attenuated after LNP injection. Serum aspartate transaminase, alanine aminotransferase, and inflammatory cytokine levels were significantly reduced in mice injected with Irf5 siRNA-loaded LNPs compared to those injected with control siRNA-loaded LNPs. Our results suggest that administering biodegradable LNPs to deliver siRNA is a promising strategy for treating inflammatory disorders.

6.
Int Immunol ; 33(12): 841-846, 2021 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-34357403

RESUMEN

As clinically demonstrated by the success of immunotherapies to improve survival outcomes, tumors are known to gain a survival advantage by circumventing immune surveillance. A defining feature of this is the creation and maintenance of a tumor immune microenvironment (TIME) that directly and indirectly alters the host's immunologic signaling pathways through a variety of mechanisms. Tumor-intrinsic mechanisms that instruct the formation and maintenance of the TIME have been an area of intensive study, such as the identification and characterization of soluble factors actively and passively released by tumor cells that modulate immune cell function. In particular, damage-associated molecular pattern (DAMP) molecules typically released by necrotic tumor cells are recognized by innate immune receptors such as Toll-like receptors (TLRs) and stimulate immune cells within TIME. Given their broad and potent effects on the immune system, a better understanding for how DAMP and TLR interactions sculpt the TIME to favor tumor growth would identify new strategies and approaches for cancer immunotherapy.


Asunto(s)
Neoplasias/inmunología , Receptores Toll-Like/inmunología , Animales , Humanos , Microambiente Tumoral/inmunología
7.
Nat Immunol ; 22(8): 947-957, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34239121

RESUMEN

One of most challenging issues in tumor immunology is a better understanding of the dynamics in the accumulation of myeloid-derived suppressor cells (MDSCs) in the tumor microenvironment (TIME), as this would lead to the development of new cancer therapeutics. Here, we show that translationally controlled tumor protein (TCTP) released by dying tumor cells is an immunomodulator crucial to full-blown MDSC accumulation in the TIME. We provide evidence that extracellular TCTP mediates recruitment of the polymorphonuclear MDSC (PMN-MDSC) population in the TIME via activation of Toll-like receptor-2. As further proof of principle, we show that inhibition of TCTP suppresses PMN-MDSC accumulation and tumor growth. In human cancers, we find an elevation of TCTP and an inverse correlation of TCTP gene dosage with antitumor immune signatures and clinical prognosis. This study reveals the hitherto poorly understood mechanism of the MDSC dynamics in the TIME, offering a new rationale for cancer immunotherapy.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Quimiocina CXCL1/metabolismo , Neoplasias Colorrectales/inmunología , Células Supresoras de Origen Mieloide/inmunología , Receptor Toll-Like 2/inmunología , Microambiente Tumoral/inmunología , Alarminas/genética , Alarminas/metabolismo , Animales , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Femenino , Células HEK293 , Humanos , Inmunoterapia , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Células RAW 264.7 , Proteína Tumoral Controlada Traslacionalmente 1
8.
Nat Commun ; 12(1): 4379, 2021 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-34282144

RESUMEN

The transcription factor IRF5 has been implicated as a therapeutic target for the autoimmune disease systemic lupus erythematosus (SLE). However, IRF5 activation status during the disease course and the effects of IRF5 inhibition after disease onset are unclear. Here, we show that SLE patients in both the active and remission phase have aberrant activation of IRF5 and interferon-stimulated genes. Partial inhibition of IRF5 is superior to full inhibition of type I interferon signaling in suppressing disease in a mouse model of SLE, possibly due to the function of IRF5 in oxidative phosphorylation. We further demonstrate that inhibition of IRF5 via conditional Irf5 deletion and a newly developed small-molecule inhibitor of IRF5 after disease onset suppresses disease progression and is effective for maintenance of remission in mice. These results suggest that IRF5 inhibition might overcome the limitations of current SLE therapies, thus promoting drug discovery research on IRF5 inhibitors.


Asunto(s)
Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Animales , Autoanticuerpos/inmunología , Enfermedades Autoinmunes , Femenino , Regulación de la Expresión Génica , Humanos , Inmunidad Innata , Inmunoglobulina G , Factores Reguladores del Interferón/efectos de los fármacos , Riñón/patología , Lupus Eritematoso Sistémico/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Interferón alfa y beta , Transducción de Señal , Factores de Transcripción , Familia-src Quinasas
9.
Cancer Sci ; 112(7): 2578-2591, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33570784

RESUMEN

The signal-transducing innate receptors represent classes of pattern recognition receptors (PRRs) that play crucial roles in the first line of the host defense against infections by the recognition of pathogen-derived molecules. Because of their poorly discriminative nature compared with antigen receptors of the adaptive immune system, they also recognize endogenous molecules and evoke immune responses without infection, resulting in the regulation of tumor immunity. Therefore, PRRs may be promising targets for effective cancer immunotherapy, either by activating or inhibiting them. Here, we summarize our current knowledge of signal-transducing PRRs in the regulation of tumor immunity.


Asunto(s)
Neoplasias/inmunología , Receptores de Reconocimiento de Patrones/inmunología , Transducción de Señal/inmunología , Animales , Membrana Celular/metabolismo , Citoplasma/metabolismo , Humanos , Inmunidad Innata , Inmunoterapia/tendencias , Neoplasias/terapia , Receptores de Reconocimiento de Patrones/metabolismo , Microambiente Tumoral/inmunología
10.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33443188

RESUMEN

Dysregulation of inflammatory cytokines in keratinocytes promote the pathogenesis of the skin inflammation, such as allergic contact dermatitis (ACD). High-mobility group box 1 protein (HMGB1) has been implicated in the promotion of skin inflammation upon its extracellular release as a damage-associated molecular pattern molecule. However, whether and how HMGB1 in keratinocytes contributes to ACD and other skin disorders remain elusive. In this study, we generated conditional knockout mice in which the Hmgb1 gene is specifically deleted in keratinocytes, and examined its role in ACD models. Interestingly, the mutant mice showed exacerbated skin inflammation, accompanied by increased ear thickening in 2,4-dinitrofluorobenezene-induced ACDs. The mRNA expression of interleukin-24 (IL-24), a cytokine known to critically contribute to ACD pathogenesis, was elevated in skin lesions of the mutant mice. As with constitutively expressed, IL-4-induced Il24 mRNA, expression was also augmented in the Hmgb1-deficient keratinocytes, which would account for the exacerbation of ACD in the mutant mice. Mechanistically, we observed an increased binding of trimethyl histone H3 (lys4) (H3K4me3), a hallmark of transcriptionally active genes, to the promoter region of the Il24 gene in the hmgb1-deficient cells. Thus, the nuclear HMGB1 is a critical "gate keeper" in that the dermal homeostasis is contingent to its function in chromatin remodeling. Our study revealed a facet of nuclear HMGB1, namely its antiinflammatory function in keratinocytes for the skin homeostasis.


Asunto(s)
Ensamble y Desensamble de Cromatina , Dermatitis Alérgica por Contacto/metabolismo , Proteína HMGB1/metabolismo , Histonas/metabolismo , Interleucinas/metabolismo , Queratinocitos/metabolismo , Animales , Dermatitis Alérgica por Contacto/genética , Dermatitis Alérgica por Contacto/prevención & control , Dinitrofluorobenceno/toxicidad , Modelos Animales de Enfermedad , Oído/patología , Eliminación de Gen , Regulación de la Expresión Génica/genética , Proteína HMGB1/deficiencia , Proteína HMGB1/genética , Inflamación/genética , Inflamación/metabolismo , Interleucina-4/farmacología , Interleucinas/genética , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Piel/inmunología , Piel/metabolismo , Piel/patología , Quimera por Trasplante
11.
Proc Natl Acad Sci U S A ; 116(47): 23653-23661, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31694883

RESUMEN

The activation of innate immune receptors by pathogen-associated molecular patterns (PAMPs) is central to host defense against infections. On the other hand, these receptors are also activated by immunogenic damage-associated molecular patterns (DAMPs), typically released from dying cells, and the activation can evoke chronic inflammatory or autoimmune disorders. One of the best known receptors involved in the immune pathogenesis is Toll-like receptor 7 (TLR7), which recognizes RNA with single-stranded structure. However, the causative DAMP RNA(s) in the pathogenesis has yet to be identified. Here, we first developed a chemical compound, termed KN69, that suppresses autoimmunity in several established mouse models. A subsequent search for KN69-binding partners led to the identification of U11 small nuclear RNA (U11snRNA) as a candidate DAMP RNA involved in TLR7-induced autoimmunity. We then showed that U11snRNA robustly activated the TLR7 pathway in vitro and induced arthritis disease in vivo. We also found a correlation between high serum level of U11snRNA and autoimmune diseases in human subjects and established mouse models. Finally, by revealing the structural basis for U11snRNA's ability to activate TLR7, we developed more potent TLR7 agonists and TLR7 antagonists, which may offer new therapeutic approaches for autoimmunity or other immune-driven diseases. Thus, our study has revealed a hitherto unknown immune function of U11snRNA, providing insight into TLR7-mediated autoimmunity and its potential for further therapeutic applications.


Asunto(s)
Glicoproteínas de Membrana/agonistas , ARN Nuclear Pequeño/inmunología , Receptor Toll-Like 7/agonistas , Adulto , Alarminas/química , Animales , Artritis Reumatoide/sangre , Artritis Reumatoide/inmunología , Enfermedades Autoinmunes/sangre , Enfermedades Autoinmunes/inmunología , Secuencia de Bases , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunosupresores/síntesis química , Inmunosupresores/farmacología , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/inmunología , Glicoproteínas de Membrana/deficiencia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Persona de Mediana Edad , ARN/inmunología , ARN/metabolismo , Ribonucleoproteínas Nucleares Pequeñas/química , Ribonucleoproteínas Nucleares Pequeñas/inmunología , Análisis de Secuencia de ARN , Receptor Toll-Like 7/deficiencia , Adulto Joven
12.
Cancer Sci ; 109(9): 2687-2696, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29981256

RESUMEN

Recent years have seen a number of regulatory approvals for immune oncology or immunotherapies based on their ability to enhance antitumor immune responses. Nevertheless, the majority of patients remain refractory to these treatments; hence, new therapies that augment current immunotherapies are required. Innate immune receptors that recognize nucleic acids are potent activators of subsequent T-cell responses and, as a result, can evoke potent antitumor immune responses. Herein, we present a novel compound N-{3-[(1,4'-bipiperidin)-1'-yl]propyl}-6-[4-(4-methylpiperazin-1-yl)phenyl]picolinamide (SINCRO; STING-mediated interferon-inducing and cytotoxic reagent, original) as an anticancer drug that activates the cytosolic DNA-sensing STING (stimulator of interferon genes) signaling pathway leading to the induction of type I interferon (IFN) genes. Indeed, IFN-ß gene induction by SINCRO is abolished in STING-deficient cells. In addition to its IFN-inducing activity, SINCRO shows STING-independent cytotoxic activity against cancer cells. SINCRO does not evoke DNA double-strand break or caspase-3 cleavage. Thus, SINCRO induces cell death in a method different from conventional apoptosis-inducing pathways. Finally, we provide evidence that giving SINCRO significantly attenuates in vivo tumor growth by both type I IFN-dependent and independent mechanisms. Thus, SINCRO is an attractive anticancer compound with dual function in that it evokes type I IFN response to promote antitumor immunity as well as inducing tumor cell death. SINCRO may provide a new platform for the development of drugs for effective cancer therapy.


Asunto(s)
Amidas/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Inmunidad Innata/efectos de los fármacos , Interferón beta/biosíntesis , Proteínas de la Membrana/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Ácidos Picolínicos/farmacología , Piperidinas/farmacología , Células 3T3 , Amidas/química , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de los fármacos , Células HEK293 , Células HeLa , Humanos , Interferón beta/genética , Interferón beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias/patología , Ácidos Picolínicos/química , Piperidinas/química , Transducción de Señal/efectos de los fármacos
13.
Proc Natl Acad Sci U S A ; 115(20): 5253-5258, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29712834

RESUMEN

IFN regulatory factor 3 (IRF3) is a transcription regulator of cellular responses in many cell types that is known to be essential for innate immunity. To confirm IRF3's broad role in immunity and to more fully discern its role in various cellular subsets, we engineered Irf3-floxed mice to allow for the cell type-specific ablation of Irf3 Analysis of these mice confirmed the general requirement of IRF3 for the evocation of type I IFN responses in vitro and in vivo. Furthermore, immune cell ontogeny and frequencies of immune cell types were unaffected when Irf3 was selectively inactivated in either T cells or B cells in the mice. Interestingly, in a model of lipopolysaccharide-induced septic shock, selective Irf3 deficiency in myeloid cells led to reduced levels of type I IFN in the sera and increased survival of these mice, indicating the myeloid-specific, pathogenic role of the Toll-like receptor 4-IRF3 type I IFN axis in this model of sepsis. Thus, Irf3-floxed mice can serve as useful tool for further exploring the cell type-specific functions of this transcription factor.


Asunto(s)
Inmunidad Innata/inmunología , Inflamación/inmunología , Factor 3 Regulador del Interferón/metabolismo , Células Mieloides/inmunología , Linfocitos T/inmunología , Animales , Regulación de la Expresión Génica , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/patología , Lipopolisacáridos/toxicidad , Ratones , Ratones Noqueados , Células Mieloides/metabolismo , Células Mieloides/patología , Transducción de Señal , Linfocitos T/metabolismo , Linfocitos T/patología
14.
Artículo en Inglés | MEDLINE | ID: mdl-28963109

RESUMEN

Interferons (IFNs) are a broad class of cytokines elicited on challenge to the host defense and are essential for mobilizing immune responses to pathogens. Divided into three classes, type I, type II, and type III, all IFNs share in common the ability to evoke antiviral activities initiated by the interaction with their cognate receptors. The nine-member IFN regulatory factor (IRF) family, first discovered in the context of transcriptional regulation of type I IFN genes following viral infection, are pivotal for the regulation of the IFN responses. In this review, we briefly describe cardinal features of the three types of IFNs and then focus on the role of the IRF family members in the regulation of each IFN system.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Factores Reguladores del Interferón/metabolismo , Interferones/metabolismo , Animales , ADN , Humanos , Factores Reguladores del Interferón/genética , Interferones/genética
15.
Cell Res ; 27(12): 1407-1408, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28925385

RESUMEN

Type I interferon (IFN) signaling is critical for intracellular antimicrobial programmes, affecting both innate and adaptive immune responses. The paper recently published in Cell demonstrates a new regulatory mechanism of the type I IFN signaling pathway by histone-lysine N-methyltransferase SETD2.


Asunto(s)
Antivirales , Interferón Tipo I , N-Metiltransferasa de Histona-Lisina , Metilación , Transducción de Señal
16.
Proc Natl Acad Sci U S A ; 114(38): 10178-10183, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28878025

RESUMEN

The commensal microbiota within the gastrointestinal tract is essential in maintaining homeostasis. Indeed, dysregulation in the repertoire of microbiota can result in the development of intestinal immune-inflammatory diseases. Further, this immune regulation by gut microbiota is important systemically, impacting health and disease of organ systems beyond the local environment of the gut. What has not been explored is how distant organs might in turn shape the microbiota via microbe-targeted molecules. Here, we provide evidence that surfactant protein D (SP-D) synthesized in the gallbladder and delivered into intestinal lumen binds selectively to species of gut commensal bacteria. SP-D-deficient mice manifest intestinal dysbiosis and show a susceptibility to dextran sulfate sodium-induced colitis. Further, fecal transfer from SP-D-deficient mice to wild-type, germ-free mice conveyed colitis susceptibility. Interestingly, colitis caused a notable increase in Sftpd gene expression in the gallbladder, but not in the lung, via the activity of glucocorticoids produced in the liver. These findings describe a unique mechanism of interorgan regulation of intestinal immune homeostasis by SP-D with potential clinical implications such as cholecystectomy.


Asunto(s)
Colitis/metabolismo , Vesícula Biliar/metabolismo , Microbioma Gastrointestinal , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Animales , Colitis/microbiología , Factores de Transcripción Forkhead/metabolismo , Glucocorticoides/biosíntesis , Homeostasis , Mucosa Intestinal/inmunología , Hígado/metabolismo , Ratones Endogámicos C57BL , Simbiosis , Linfocitos T Reguladores/metabolismo
17.
Artículo en Inglés | MEDLINE | ID: mdl-28373196

RESUMEN

Although nucleot(s)ide analogues and pegylated interferon alpha 2a (PEG-IFN-α2a) can suppress hepatitis B virus (HBV) replication, it is difficult to achieve complete HBV elimination from hepatocytes. A novel site-specific pegylated recombinant human IFN-ß (TRK-560) was recently developed. In the present study, we evaluated the antiviral effects of TRK-560 on HBV replication in vitro and in vivo. In vitro and in vivo HBV replication models were treated with antivirals including TRK-560, and changes in HBV markers were evaluated. To analyze antiviral mechanisms, cDNA microarray analysis and an enzyme-linked immunoassay (ELISA) were performed. TRK-560 significantly suppressed the production of intracellular HBV replication intermediates and extracellular HBV surface antigen (HBsAg) (P < 0.001 and P < 0.001, respectively), and the antiviral effects of TRK-560 were enhanced in combination with nucleot(s)ide analogues, such as entecavir and tenofovir disoproxil fumarate. The reduction in HBV DNA levels by TRK-560 treatment was significantly higher than that by PEG-IFN-α2a treatment both in vitro and in vivo (P = 0.004 and P = 0.046, respectively), and intracellular HBV covalently closed circular DNA (cccDNA) reduction by TRK-560 treatment was also significantly higher than that by PEG-IFN-α2a treatment in vivo (P = 0.0495). cDNA microarrays and ELISA for CXCL10 production revealed significant differences between TRK-560 and PEG-IFN-α2a in the induction potency of interferon-stimulated genes. TRK-560 shows a stronger antiviral potency via higher induction of interferon-stimulated genes and stronger stimulation of immune cell chemotaxis than PEG-IFN-α2a. As HBsAg loss and HBV cccDNA eradication are important clinical goals, these results suggest a potential role for TRK-560 in the development of more effective treatment for chronic hepatitis B infection.


Asunto(s)
Antivirales/farmacología , Antígenos de Superficie de la Hepatitis B/metabolismo , Virus de la Hepatitis B/efectos de los fármacos , Hepatitis B Crónica/tratamiento farmacológico , Interferón-alfa/farmacología , Polietilenglicoles/farmacología , Animales , Línea Celular Tumoral , Quimiocina CXCL10/biosíntesis , ADN Circular/metabolismo , ADN Viral/metabolismo , Células Hep G2 , Humanos , Ratones , Ratones SCID , Ratones Transgénicos , Proteínas Recombinantes/farmacología , Resultado del Tratamiento , Carga Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos
18.
Cancer Sci ; 108(4): 581-589, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28129467

RESUMEN

Malignant ascites manifests as an end-stage event during the progression of a number of cancers and lacks a generally accepted standard therapy. Interferon-ß (IFN-ß) has been used to treat several cancer indications; however, little is known about the efficacy of IFN-ß on malignant ascites. In the present study, we report on the development of a novel, engineered form of human and murine IFN-ß, each conjugated with a polyethylene glycol molecule (PEG-hIFN-ß and PEG-mIFN-ß, respectively). We provide evidence that these IFN-ß molecules retain anti-viral potency comparable to unmodified IFN-ß in vitro and manifested improved pharmacokinetics in vivo. Interestingly, PEG-mIFN-ß significantly inhibited the accumulation of ascites fluid and vascular permeability of the peritoneal membrane in models of ovarian cancer and gastric cancer cell xenograft mice. We further show that PEG-hIFN-ß directly suppresses VEGF165 -induced hyperpermeability in a monolayer of human vascular endothelial cells and that PEG-mIFN-ß enhanced gene expression for a number of cell adhesion related molecules in mouse vascular endothelial cells. Taken together, these findings unveil a hitherto unrecognized potential of IFN-ß in maintaining vascular integrity, and provide proof-of-mechanism for a novel and long-acting pegylated hIFN-ß for the therapeutic treatment of malignant ascites.


Asunto(s)
Ascitis/tratamiento farmacológico , Interferón beta/farmacología , Neoplasias Peritoneales/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , 5'-Nucleotidasa/metabolismo , Animales , Antivirales/química , Antivirales/farmacocinética , Antivirales/farmacología , Área Bajo la Curva , Ascitis/patología , Línea Celular Tumoral , Permeabilidad de la Membrana Celular/efectos de los fármacos , Células Cultivadas , Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Interferón beta/química , Interferón beta/farmacocinética , Tasa de Depuración Metabólica , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Ratones SCID , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Neoplasias Peritoneales/secundario , Polietilenglicoles/química , Factor A de Crecimiento Endotelial Vascular/farmacología
19.
Proc Natl Acad Sci U S A ; 113(49): 14097-14102, 2016 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-27872290

RESUMEN

Tumor metastasis is the cause of most cancer deaths. Although metastases can form in multiple end organs, the liver is recognized as a highly permissive organ. Nevertheless, there is evidence for immune cell-mediated mechanisms that function to suppress liver metastasis by certain tumors, although the underlying mechanisms for the suppression of metastasis remain elusive. Here, we show that Dectin-2, a C-type lectin receptor (CLR) family of innate receptors, is critical for the suppression of liver metastasis of cancer cells. We provide evidence that Dectin-2 functions in resident macrophages in the liver, known as Kupffer cells, to mediate the uptake and clearance of cancer cells. Interestingly, Kupffer cells are selectively endowed with Dectin-2-dependent phagocytotic activity, with neither bone marrow-derived macrophages nor alveolar macrophages showing this potential. Concordantly, subcutaneous primary tumor growth and lung metastasis are not affected by the absence of Dectin-2. In addition, macrophage C-type lectin, a CLR known to be complex with Dectin-2, also contributes to the suppression of liver metastasis. Collectively, these results highlight the hitherto poorly understood mechanism of Kupffer cell-mediated control of metastasis that is mediated by the CLR innate receptor family, with implications for the development of anticancer therapy targeting CLRs.


Asunto(s)
Macrófagos del Hígado/fisiología , Lectinas Tipo C/metabolismo , Neoplasias Hepáticas Experimentales/secundario , Metástasis de la Neoplasia/inmunología , Fagocitosis , Animales , Línea Celular Tumoral , Humanos , Lectinas Tipo C/genética , Ratones Endogámicos C57BL , Receptores Inmunológicos/metabolismo
20.
Immunity ; 45(2): 319-32, 2016 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-27521268

RESUMEN

Interferon regulatory factor-5 (IRF5), a transcription factor critical for the induction of innate immune responses, contributes to the pathogenesis of the autoimmune disease systemic lupus erythematosus (SLE) in humans and mice. Lyn, a Src family kinase, is also implicated in human SLE, and Lyn-deficient mice develop an SLE-like disease. Here, we found that Lyn physically interacted with IRF5 to inhibit ubiquitination and phosphorylation of IRF5 in the TLR-MyD88 pathway, thereby suppressing the transcriptional activity of IRF5 in a manner independent of Lyn's kinase activity. Conversely, Lyn did not inhibit NF-κB signaling, another major branch downstream of MyD88. Monoallelic deletion of Irf5 alleviated the hyperproduction of cytokines in TLR-stimulated Lyn(-/-) dendritic cells and the development of SLE-like symptoms in Lyn(-/-) mice. Our results reveal a role for Lyn as a specific suppressor of the TLR-MyD88-IRF5 pathway and illustrate the importance of fine-tuning IRF5 activity for the maintenance of immune homeostasis.


Asunto(s)
Autoinmunidad , Células Dendríticas/inmunología , Factores Reguladores del Interferón/metabolismo , Lupus Eritematoso Sistémico/inmunología , Familia-src Quinasas/metabolismo , Animales , Células Cultivadas , Citocinas/metabolismo , Humanos , Tolerancia Inmunológica , Inmunidad Innata , Factores Reguladores del Interferón/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Fosforilación , Unión Proteica , Transducción de Señal , Receptores Toll-Like/metabolismo , Activación Transcripcional , Ubiquitinación , Familia-src Quinasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...