Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 43(5): 114249, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38758648

RESUMEN

Signal-regulatory protein alpha (SIRPα) has recently been found to be highly expressed in podocytes and is essential for maintaining podocyte function. However, its immunoregulatory function in podocytes remains elusive. Here, we report that SIRPα controls podocyte antigen presentation in specific T cell activation via inhibiting spleen tyrosine kinase (Syk) phosphorylation. First, podocyte SIRPα under lupus nephritis (LN) conditions is strongly downregulated. Second, podocyte-specific deletion of SIRPα exacerbates renal disease progression in lupus-prone mice, as evidenced by an increase in T cell infiltration. Third, SIRPα deletion or knockdown enhances podocyte antigen presentation, which activates specific T cells, via enhancing Syk phosphorylation. Supporting this, Syk inhibitor GS-9973 prevents podocyte antigen presentation, resulting in a decrease of T cell activation and mitigation of renal disease caused by SIRPα knockdown or deletion. Our findings reveal an immunoregulatory role of SIRPα loss in promoting podocyte antigen presentation to activate specific T cell immune responses in LN.


Asunto(s)
Nefritis Lúpica , Podocitos , Receptores Inmunológicos , Quinasa Syk , Linfocitos T , Podocitos/metabolismo , Podocitos/patología , Podocitos/inmunología , Nefritis Lúpica/patología , Nefritis Lúpica/inmunología , Nefritis Lúpica/metabolismo , Animales , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo , Ratones , Quinasa Syk/metabolismo , Ratones Endogámicos C57BL , Inflamación/patología , Inflamación/metabolismo , Fosforilación , Activación de Linfocitos/inmunología , Humanos , Presentación de Antígeno/inmunología , Femenino
2.
Stem Cell Res Ther ; 13(1): 484, 2022 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-36153633

RESUMEN

BACKGROUND: Anti-inflammatory polarized macrophages are reported to alleviate systemic lupus erythematosus (SLE). Our previous studies have demonstrated that exosomes from adipose-derived stem cells promote the anti-inflammatory polarization of macrophages. However, the possible therapeutic effect of exosomes from stem cells on SLE remains unexplored. METHODS: Exosomes were isolated from the conditioned medium of bone marrow-derived mesenchymal stem cells using ultrafiltration and size-exclusion chromatography and were identified by nanoparticle tracking analysis and immunoblotting of exosomal-specific markers. Macrophages were collected from the MRL/lpr mouse kidney. The phenotype of macrophages was identified by immunoblotting for intracellular markers-inducible nitric oxide synthase (iNOS) and arginase-1 (Arg-1), and flow cytometry for macrophage markers F4/80, CD86, CD206, B7H4, and CD138. Pristane-induced murine lupus nephritis models were employed for in vivo study. RESULTS: When macrophages from the kidney of the MRL/lpr mice were treated with exosomes from bone marrow-derived mesenchymal stem cells (BM-MSCs), the upregulation of CD206, B7H4, CD138, Arg-1, CCL20, and anti-inflammatory cytokines was observed, which suggested that the macrophages were polarized to a specific anti-inflammatory phenotype. These anti-inflammatory macrophages produced low levels of reactive oxygen species (ROS) but had a high efferocytosis activity and promoted regulatory T (Treg) cell recruitment. Moreover, exosome injection stimulated the anti-inflammatory polarization of macrophages and increased the production of IL-17+ Treg cells in a pristane-induced murine lupus nephritis model. We observed that exosomes from BMMSCs depleted of microRNA-16 (miR-16) and microRNA-21 (miR-21) failed to downregulate PDCD4 and PTEN in macrophages, respectively, and attenuated exosome-induced anti-inflammatory polarization. CONCLUSION: Our findings provide evidence that exosomes from BMMSCs promote the anti-inflammatory polarization of macrophages. These macrophages alleviate SLE nephritis in lupus mice by consuming apoptotic debris and inducing the recruitment of Treg cells. We identify that exosomal delivery of miR-16 and miR-21 is a significant contributor to the polarization of macrophages.


Asunto(s)
Exosomas , Lupus Eritematoso Sistémico , Nefritis Lúpica , Células Madre Mesenquimatosas , MicroARNs , Animales , Antiinflamatorios/farmacología , Arginasa , Medios de Cultivo Condicionados/farmacología , Citocinas/metabolismo , Exosomas/metabolismo , Interleucina-17 , Lupus Eritematoso Sistémico/terapia , Nefritis Lúpica/terapia , Macrófagos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , MicroARNs/uso terapéutico , Óxido Nítrico Sintasa de Tipo II , Especies Reactivas de Oxígeno , Linfocitos T Reguladores/metabolismo , Terpenos
3.
J Mol Cell Cardiol ; 162: 10-19, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34474073

RESUMEN

Our previous study has revealed that exosomes from adipose-derived stem cells (ASCs) promote angiogenesis in subcutaneously transplanted gels by delivery of microRNA-31 (miR-31) which targets factor inhibiting hypoxia-inducible factor-1 (FIH1) in recipient cells. Here we hypothesized that ASC exosomes alleviate ischemic diseases through miR-31/FIH1/hypoxia-inducible factor-1α (HIF-1α) signaling pathway. Exosomes from ASCs were characterized with nanoparticle tracking analysis, transmission electron microscopy, and immunoblotting analysis for exosomal markers. Results from immunoblotting and laser imaging of ischemic mouse hindlimb revealed that miR-31 enriched ASC exosomes inhibited FIH1 expression and enhanced the blood perfusion, respectively. These effects were impaired when using miR-31-depleted exosomes. Immunohistochemistry analysis showed that administration of exosomes resulted in a higher arteriole density and larger CD31+ area in ischemic hindlimb than miR-31-delpleted exosomes. Similarly, knockdown of miR-31 in exosomes reduced the effects of the exosomes on increasing ventricular fraction shortening and CD31+ area, and on decreasing infarct size. Exosomes promoted endothelial cell migration and tube formation. These changes were attenuated when miR-31 was depleted in the exosomes or when FIH1 was overexpressed in the endothelial cells. Furthermore, the results from immunocytochemistry, co-immunoprecipitation, and luciferase reporter assay demonstrated that the effects of exosomes on nuclear translocation, binding with co-activator p300, and activation of HIF-1α were decreased when miR-31 was depleted in the exosomes or FIH1 was overexpressed. Our findings provide evidence that exosomes from ASCs promote angiogenesis in both mouse ischemic hindlimb and heart through transport of miR-31 which targets FIH1 and therefore triggers HIF-1α transcriptional activation.


Asunto(s)
Exosomas , MicroARNs , Infarto del Miocardio , Animales , Células Endoteliales/metabolismo , Exosomas/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Infarto del Miocardio/metabolismo , Infarto del Miocardio/terapia , Células Madre/metabolismo
4.
Stem Cell Res Ther ; 11(1): 162, 2020 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-32321589

RESUMEN

BACKGROUND: M2 macrophages and exosomes from adipose-derived stem cells (ASCs) are both reported to promote angiogenesis. However, the possible synergistic effects between exogenous exosomes and endogenous M2 macrophages are poorly understood. METHODS: Exosomes were isolated from conditioned medium of normoxic and hypoxic ASCs using the combined techniques of ultrafiltration and size-exclusion chromatography and were identified with nanoparticle tracking analysis and immunoblotting for exosomal markers. Macrophages were collected from the mouse peritoneal cavity. M1 and M2 macrophages were detected by immunoblotting for the intracellular markers inducible nitric oxide synthase (iNOS) and arginase-1 (Arg-1) and by flow cytometry for the surface markers F4/80, CD86, and CD206. Murine models of Matrigel plug and hindlimb ischemia were employed as in vivo angiogenic assays. RESULTS: When M1 macrophages were treated with exosomes from normoxic ASCs (Nor/Exo), and particularly from hypoxic ASCs (Hyp/Exo), the expression of the M1 marker iNOS decreased, and the M2 marker Arg-1 increased in a time- and dose-dependent manner. Additionally, a decrease in the M1 surface marker CD86 and an increase in the M2 surface marker CD206 were observed, which suggested that M1 macrophages were polarized to an M2-like phenotype. Conditioned medium from these M2-like macrophages presented lower levels of proinflammatory cytokines and higher levels of proangiogenic factors and promoted endothelial cell proliferation, migration, and tube formation. Furthermore, M2 polarization and angiogenesis were induced upon the administration of exosomes in mouse Matrigel plug and hindlimb ischemia (HLI) models. Interestingly, these exosomal effects were attenuated by using a colony stimulating factor 1 receptor (CSF-1R) inhibitor, BLZ945, in vitro and in vivo. Downregulation of microRNA-21 (miR-21) in hypoxic ASCs reduced the exosomal effects on M2 polarization, Akt phosphorylation, and CSF-1 secretion. A similar reduction in exosomal activity was also observed when exosomes were administered along with BLZ945. CONCLUSION: Our findings provide evidence that exosomes from ASCs polarize macrophages toward an M2-like phenotype, which further enhances the exosomal proangiogenic effects. Exosomal delivery of miR-21 and positive feedback of secreted CSF-1 may be involved in macrophage polarization.


Asunto(s)
Exosomas , MicroARNs , Animales , Miembro Posterior , Isquemia/terapia , Macrófagos , Ratones , Células Madre
5.
Am J Physiol Heart Circ Physiol ; 317(4): H765-H776, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31418583

RESUMEN

Induced vascular progenitor cells (iVPCs) were created as an ideal cell type for regenerative medicine and have been reported to positively promote collateral blood flow and improve cardiac function in a rat model of myocardial ischemia. Exosomes have emerged as a novel biomedicine that mimics the function of the donor cells. We investigated the angiogenic activity of exosomes from iPVCs (iVPC-Exo) as a cell-free therapeutic approach for ischemia. Exosomes from iVPCs and rat aortic endothelial cells (RAECs) were isolated using a combination of ultrafiltration and size-exclusion chromatography. Nanoparticle tracking analysis revealed that exosome isolates fell within the exosomal diameter (<150 nm). These exosomes contained known markers Alix and TSG101, and their morphology was validated using transmission electron microscopy. When compared with RAECs, iVPCs significantly increased the secretion of exosomes. Cardiac microvascular endothelial cells and aortic ring explants were pretreated with RAEC-Exo or iVPC-Exo, and basal medium was used as a control. iVPC-Exo exerted an in vitro angiogenic effect on the proliferation, tube formation, and migration of endothelial cells and stimulated microvessel sprouting in an ex vivo aortic ring assay. Additionally, iVPC-Exo increased blood perfusion in a hindlimb ischemia model. Proangiogenic proteins (pentraxin-3 and insulin-like growth factor-binding protein-3) and microRNAs (-143-3p, -291b, and -20b-5p) were found to be enriched in iVPC-Exo, which may mediate iVPC-Exo induced vascular growth. Our findings demonstrate that treatment with iVPC-Exo promotes angiogenesis in vitro, ex vivo, and in vivo. Collectively, these findings indicate a novel cell-free approach for therapeutic angiogenesis.NEW & NOTEWORTHY The results of this work demonstrate exosomes as a novel physiological mechanism by which induced vascular progenitor cells exert their angiogenic effect. Moreover, angiogenic cargo of proteins and microRNAs may define the biological contributors in activating endothelial cells to form a new capillary plexus for ischemic vascular diseases.


Asunto(s)
Células Progenitoras Endoteliales/trasplante , Exosomas/trasplante , Células Madre Pluripotentes Inducidas/trasplante , Isquemia/cirugía , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica , Proteínas Angiogénicas/metabolismo , Animales , Línea Celular , Movimiento Celular , Proliferación Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Progenitoras Endoteliales/metabolismo , Exosomas/metabolismo , Miembro Posterior , Células Madre Pluripotentes Inducidas/metabolismo , Isquemia/metabolismo , Isquemia/fisiopatología , Masculino , MicroARNs/genética , MicroARNs/metabolismo , Músculo Esquelético/metabolismo , Ratas Sprague-Dawley , Recuperación de la Función , Flujo Sanguíneo Regional , Transducción de Señal
6.
Nat Microbiol ; 3(4): 503-513, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29588542

RESUMEN

The precise cell type hosting latent human cytomegalovirus (HCMV) remains elusive. Here, we report that HCMV reprogrammes human haematopoietic progenitor cells (HPCs) into a unique monocyte subset to achieve latency. Unlike conventional monocytes, this monocyte subset possesses higher levels of B7-H4, IL-10 and inducible nitric oxide synthase (iNOS), a longer lifespan and strong immunosuppressive capacity. Cell sorting of peripheral blood from latently infected human donors confirms that only this monocyte subset, representing less than 0.1% of peripheral mononuclear cells, is HCMV genome-positive but immediate-early-negative. Mechanistic studies demonstrate that HCMV promotes the differentiation of HPCs into this monocyte subset by activating cellular signal transducer and activator of transcription 3 (STAT3). In turn, this monocyte subset generates a high level of nitric oxide (NO) to silence HCMV immediate-early transcription and promote viral latency. By contrast, the US28-knockout HCMV mutant, which is incapable of activating STAT3, fails to reprogramme the HPCs and achieve latency. Our findings reveal that via activating the STAT3-iNOS-NO axis, HCMV differentiates human HPCs into a longevous, immunosuppressive monocyte subset for viral latency.


Asunto(s)
Citomegalovirus/inmunología , Citomegalovirus/patogenicidad , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/virología , Interacciones Huésped-Patógeno/inmunología , Tolerancia Inmunológica/genética , Monocitos/virología , Latencia del Virus/inmunología , Diferenciación Celular/fisiología , Reprogramación Celular/genética , Citomegalovirus/genética , Interacciones Huésped-Patógeno/genética , Humanos , Tolerancia Inmunológica/inmunología , Interleucina-10/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Factor de Transcripción STAT3/metabolismo , Inhibidor 1 de la Activación de Células T con Dominio V-Set/metabolismo , Latencia del Virus/genética
7.
Nat Commun ; 8: 14041, 2017 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-28067230

RESUMEN

Tumour cells secrete exosomes that are involved in the remodelling of the tumour-stromal environment and promoting malignancy. The mechanisms governing tumour exosome release, however, remain incompletely understood. Here we show that tumour cell exosomes secretion is controlled by pyruvate kinase type M2 (PKM2), which is upregulated and phosphorylated in tumours. During exosome secretion, phosphorylated PKM2 serves as a protein kinase to phosphorylate synaptosome-associated protein 23 (SNAP-23), which in turn enables the formation of the SNARE complex to allow exosomes release. Direct phosphorylation assay and mass spectrometry confirm that PKM2 phosphorylates SNAP-23 at Ser95. Ectopic expression of non-phosphorylated SNAP-23 mutant (Ser95→Ala95) significantly reduces PKM2-mediated exosomes release whereas expression of selective phosphomimetic SNAP-23 mutants (Ser95→Glu95 but not Ser20→Glu20) rescues the impaired exosomes release induced by PKM2 knockdown. Our findings reveal a non-metabolic function of PKM2, an enzyme associated with tumour cell reliance on aerobic glycolysis, in promoting tumour cell exosome release.


Asunto(s)
Proteínas Portadoras/genética , Exosomas/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de la Membrana/genética , Proteínas Qb-SNARE/genética , Proteínas Qc-SNARE/genética , Hormonas Tiroideas/genética , Células A549 , Animales , Secuencia de Bases , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Proliferación Celular , Células HeLa , Células Hep G2 , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Fosforilación , Proteínas Qb-SNARE/antagonistas & inhibidores , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/antagonistas & inhibidores , Proteínas Qc-SNARE/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Hormonas Tiroideas/metabolismo , Proteínas de Unión a Hormona Tiroide
8.
PLoS Pathog ; 12(11): e1006007, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27824944

RESUMEN

The mechanisms underlying human cytomegalovirus (HCMV) latency remain incompletely understood. Here, we showed that a HCMV-encoded miRNA, miR-UL148D, robustly accumulates during late stages of experimental latent HCMV infection in host cells and promotes HCMV latency by modulating the immediate early response gene 5 (IER5)-cell division cycle 25B (CDC25B) axis in host cells. miR-UL148D inhibited IER5 expression by directly targeting the three-prime untranslated region(3'UTR) of IER5 mRNA and thus rescued CDC25B expression during the establishment of viral latency. Infection with NR-1ΔmiR-UL148D, a derivative of the HCMV clinical strain NR-1 with a miR-UL148D knockout mutation, resulted in sustained induction of IER5 expression but decreased CDC25B expression in host cells. Mechanistically, we further showed that CDC25B plays an important role in suppressing HCMV IE1 and lytic gene transcription by activating cyclin-dependent kinase 1 (CDK-1). Both gain-of-function and lose-of-function assays demonstrated that miR-UL148D promotes HCMV latency by helping maintain CDC25B activity in host cells. These results provide a novel mechanism through which a HCMV miRNA regulates viral latency.


Asunto(s)
Citomegalovirus/fisiología , Proteínas Inmediatas-Precoces/metabolismo , Proteínas Nucleares/metabolismo , Latencia del Virus/fisiología , Fosfatasas cdc25/metabolismo , Western Blotting , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Regulación Viral de la Expresión Génica , Interacciones Huésped-Patógeno/fisiología , Humanos , MicroARNs/genética , ARN Viral/genética , Transfección
9.
Sci Rep ; 6: 23007, 2016 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-26961899

RESUMEN

The efficacy of interferon α (IFNα) therapy for chronic hepatitis B (CHB) patients is about 40% and often associates with adverse side-effects, thus identification of an easy accessible biomarker that can predict the outcome of IFNα treatment for individual CHB patients would be greatly helpful. Recent reports by us and others show that microRNAs encoded by human cytomegalovirus (HCMV) were readily detected in human serum and can interfere with lymphocyte responses required by IFNα therapeutic effect. We thus postulate that differential expression profile of serum HCMV miRNAs in CHB patients may serve as indicator to predict the efficacy of IFNα treatment for CHB patients. Blood was drawn from 56 individual CHB patients prior to IFNα treatment. By quantifying 13 HCMV miRNAs in serum samples, we found that the levels of HCMV-miR-US4-1 and HCMV-miR-UL-148D were significantly higher in IFNα-responsive group than in IFNα-non-responsive group. In a prospective study of 96 new CHB patients, serum level of HCMV-miR-US4-1 alone classified those who were and were not responsive to IFN-α treatment with correct rate of 84.00% and 71.74%, respectively. In conclusion, our results demonstrate that serum HCMV-miR-US4-1 can serve as a novel biomarker for predicting the outcome of IFNα treatment in CHB patients.


Asunto(s)
Biomarcadores/sangre , Citomegalovirus/aislamiento & purificación , Hepatitis B Crónica/sangre , MicroARNs/sangre , Adulto , Citomegalovirus/genética , Femenino , Virus de la Hepatitis B/aislamiento & purificación , Virus de la Hepatitis B/patogenicidad , Hepatitis B Crónica/tratamiento farmacológico , Hepatitis B Crónica/genética , Hepatitis B Crónica/virología , Humanos , Interferón-alfa/administración & dosificación , Masculino , MicroARNs/efectos de los fármacos
10.
Sci Rep ; 6: 23710, 2016 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-27010069

RESUMEN

Signal regulatory protein α (SIRPα) has been shown to operate as a negative regulator in cancer cell survival. The mechanism underneath such function, however, remains poorly defined. In the present study, we demonstrate that overexpression of SIRPα in acute promyelocytic leukemia (APL) cells results in apoptosis possibly via inhibiting the ß-catenin signaling pathway and upregulating Foxo3a. Pharmacological activation of ß-catenin signal pathway attenuates apoptosis caused by SIRPα. Interestingly, we also find that the pro-apoptotic effect of SIRPα plays an important role in arsenic trioxide (ATO)-induced apoptosis in APL cells. ATO treatment induces the SIRPα protein expression in APL cells and abrogation of SIRPα induction by lentivirus-mediated SIRPα shRNA significantly reduces the ATO-induced apoptosis. Mechanistic study further shows that induction of SIRPα protein in APL cells by ATO is mediated through suppression of c-Myc, resulting in reduction of three SIRPα-targeting microRNAs: miR-17, miR-20a and miR-106a. In summary, our results demonstrate that SIRPα inhibits tumor cell survival and significantly contributes to ATO-induced APL cell apoptosis.


Asunto(s)
Antígenos de Diferenciación/fisiología , Apoptosis/efectos de los fármacos , Arsenicales/farmacología , Leucemia Promielocítica Aguda/patología , Óxidos/farmacología , Receptores Inmunológicos/fisiología , Trióxido de Arsénico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , MicroARNs/antagonistas & inhibidores
11.
Biomaterials ; 35(14): 4390-400, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24565517

RESUMEN

Cell-derived microvesicles (MVs) have been recently shown as an efficient carrier to deliver small RNAs into the target cells. In the present study, we characterized the inhibitory effect of TGF-ß1 siRNA delivered by mouse fibroblast L929 cell-derived MVs (L929 MVs) on the growth and metastasis of murine sarcomas 180 cells both in vitro and in vivo. We found that, comparing to the same concentration of free TGF-ß1 siRNA, TGF-ß1 siRNA delivered by L929 MVs much more efficiently decreased the level of TGF-ß1 in the recipient tumor cells. Functionally, MVs containing TGF-ß1 siRNA significantly decreased the viability and migration of sarcomas 180 cells and promoted the apoptosis of tumor cells. Co-immunoprecipitation with Argonaute 2 (AGO2) via anti-AGO2 antibody indicated that the majority of TGF-ß1 siRNA in the MVs were associated with AGO2 complex. A tumor implantation mouse model further showed that intravenous injection of TGF-ß1 siRNA-containing MVs strongly suppressed TGF-ß1 expression and TGF-ß1 signaling downstream in the implanted tumor cells, and thus inhibited the growth and lung metastases of tumor cells. In conclusion, our results collectively demonstrate that the delivery of therapeutic TGF-ß1 siRNA by cell-derived MVs provides an effective strategy to control tumor cell growth and metastasis.


Asunto(s)
Técnicas de Transferencia de Gen , Neoplasias/patología , ARN Interferente Pequeño/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Liposomas Unilamelares/química , Animales , Apoptosis , Proteínas Argonautas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Masculino , Ratones , Ratones Endogámicos BALB C , Neoplasias/metabolismo , Fosforilación , Proteína Smad2/metabolismo
12.
Nat Commun ; 4: 2436, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24026300

RESUMEN

Signal regulatory protein α (SIRPα), an immunoreceptor tyrosine-based inhibitory motif (ITIM)-containing receptor, is an essential negative regulator of leukocyte inflammatory responses. Here we report that SIRPα cytoplasmic signalling ITIMs in neutrophils are cleaved during active inflammation and that the loss of SIRPα ITIMs enhances the polymorphonuclear leukocyte (PMN) inflammatory response. Using human leukocytes and two inflammatory models in mice, we show that the cleavage of SIRPα ITIMs in PMNs but not monocytes occurs at the post-acute stage of inflammation and correlates with increased PMN recruitment to inflammatory loci. Enhanced transmigration of PMNs and PMN-associated tissue damage are confirmed in mutant mice expressing SIRPα but lacking the ITIMs. Moreover, the loss of SIRPα ITIMs in PMNs during colitis is blocked by an anti-interleukin-17 (IL-17) antibody. These results demonstrate a SIRPα-based mechanism that dynamically regulates PMN inflammatory responses by generating a CD47-binding but non-signalling SIRPα 'decoy'.


Asunto(s)
Antígenos de Diferenciación/química , Antígenos de Diferenciación/metabolismo , Citoplasma/metabolismo , Inflamación/metabolismo , Inflamación/patología , Neutrófilos/metabolismo , Proteolisis , Receptores Inmunológicos/química , Receptores Inmunológicos/metabolismo , Secuencias de Aminoácidos , Animales , Western Blotting , Antígeno CD47/metabolismo , Movimiento Celular , Separación Celular , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Eliminación de Gen , Humanos , Ratones , Ratones Endogámicos C57BL , Neutrófilos/enzimología , Neutrófilos/patología , Unión Proteica , Eliminación de Secuencia , Serina Proteasas/metabolismo
13.
J Allergy Clin Immunol ; 132(2): 426-36.e8, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23562609

RESUMEN

BACKGROUND: Signal-regulatory protein α (SIRPα) is an essential signaling molecule that modulates leukocyte inflammatory responses. However, the regulation of selective SIRPα synthesis and its dynamic changes in leukocytes under inflammatory stimulation remain incompletely understood. OBJECTIVE: We sought to identify the microRNAs (miRNAs) that posttranscriptionally regulate SIRPα synthesis and their roles in modulating macrophage inflammatory responses. METHODS: SIRPα was induced in SIRPα-negative promyelocytic cells by retinoic acid or phorbol 12-myristate 13-acetate, and the differential expression of miRNAs was assessed by means of microarray and quantitative RT-PCR assays. The roles of identified miRNAs in controlling SIRPα synthesis in leukocytes and leukocyte inflammatory responses were determined. RESULTS: We identified SIRPα as a common target gene of miR-17, miR-20a, and miR-106a. During SIRPα induction, levels of these 3 miRNAs were all reduced, and their downregulation by retinoic acid or phorbol 12-myristate 13-acetate occurred through suppression of the c-Myc signaling pathway. All miR-17, miR-20a, and miR-106a specifically bound to the same seed sequence within the SIRPα 3' untranslated region and correlated inversely with SIRPα protein levels in various cells. In macrophages upregulation of miR-17, miR-20a, and miR-106a by LPS served as the mechanism underlying LPS-induced SIRPα reduction and macrophage activation. Both in vitro and in vivo assays demonstrate that miR-17, miR-20a, and miR-106a regulate macrophage infiltration, phagocytosis, and proinflammatory cytokine secretion through targeting SIRPα. CONCLUSION: These findings demonstrate for the first time that miR-17, miR-20a, and miR-106a regulate SIRPα synthesis and SIRPα-mediated macrophage inflammatory responses in a redundant fashion, providing a novel pathway in which a panel of miRNAs can modulate immune polarization through regulation of macrophage activation.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Regulación de la Expresión Génica , Macrófagos/inmunología , MicroARNs/genética , MicroARNs/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Antígenos de Diferenciación/genética , Línea Celular , Células HL-60 , Humanos , Inflamación/inmunología , Activación de Macrófagos , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores Inmunológicos/genética , Transducción de Señal , Células U937
14.
PLoS One ; 7(10): e46957, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23077538

RESUMEN

Cell-secreted miRNAs are highly stable and can serve as biomarkers for various diseases and signaling molecules in intercellular communication. The mechanism underlying the stability of circulating miRNAs, however, remains incompletely understood. Here we show that Argonaute 2 (Ago2) complexes and microvesicles (MVs) provide specific and non-specific protection for miRNA in cell-secreted MVs, respectively. First, the resistance of MV-encapsulated miRNAs to RNaseA was both depended on intact vesicular structure of MVs and protease-sensitive. Second, an immunoprecipitation assay using a probe complementary to human miR-16, a miRNA primarily located in the MVs and showed a strong, protease-sensitive resistance to RNaseA, identified Ago2 as a major miR-16-associated protein. Compared with protein-free miR-16, Ago2-associated miR-16 was resistant to RNaseA in a dose- and time-dependent fashion. Third, when the miR-16/Ago2 complex was disrupted by trypaflavine, the resistance of miR-16 to RNaseA was decreased. In contrast, when the association of miR-16 with the Ago2 complexes was increased during cell apoptosis, although the total amount of miR-16 and Ago2 remained unchanged, the resistance of miR-16 to RNaseA in the MVs was enhanced. A similar correlation between the increase of miR-223/Ago2 association and the resistance of miR-223 against RNaseA was observed during all trans retinoic acid (ATRA)-induced cell differentiation of promyelocytic HL60 cells. In conclusion, the association of miRNAs with Ago2 complexes, an event that is linked to cell functional status, plays a critical role in stabilizing the circulating miRNAs in cell-secreted MVs.


Asunto(s)
Proteínas Argonautas/metabolismo , Micropartículas Derivadas de Células/metabolismo , MicroARNs/metabolismo , Proteínas Argonautas/análisis , Secuencia de Bases , Células HeLa , Humanos , MicroARNs/análisis , Ribonucleasas/metabolismo
16.
J Biol Chem ; 287(18): 14851-62, 2012 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-22393047

RESUMEN

Previous studies have indicated that female animals are more resistant to carbon tetrachloride (CCl(4))-induced liver fibrosis than male animals, and that estradiol (E(2)) treatment can inhibit CCl(4)-induced animal hepatic fibrosis. The underlying mechanism governing these phenomena, however, has not been fully elucidated. Here we reported the role of estrogen-induced miRNA-29 (miR-29) expression in CCl(4)-induced mouse liver injury. Hepatic miR-29 levels were differentially regulated in female and male mice during CCl(4) treatment. Specifically, the levels of miR-29a and miR-29b expression were significantly decreased in the livers of male, but not female, mice following 4 weeks of CCl(4) treatment. The down-regulation of miR-29a and miR-29b in male mouse livers correlated with the early development of liver fibrosis, as indicated by increased expressions of fibrotic markers in male mice relative to female mice. In addition, E(2) was maintained at a higher level in female mice than in male mice. In contrast to TGF-ß1 that decreased miR-29a/b expression in murine hepatoma IAR20 cells and normal hepatocytes, E(2) enhanced the expression of miR-29a/b through suppression of the nuclear factor-κB (NF-κB) signal pathway, which negatively regulates miR-29 expression. Furthermore, both E(2) treatment and intravenous injection of the recombinant adenovirus expressing miR-29a/b markedly increased the miR-29a/b level and attenuated the expression of fibrotic markers in mouse livers during CCl(4) treatment, supporting the protective role of E(2)-induced miR-29 in CCl(4)-induced hepatic injury. In conclusion, our results collectively demonstrate that estrogen can inhibit CCl(4)-induced hepatic injury through the induction of hepatic miR-29.


Asunto(s)
Intoxicación por Tetracloruro de Carbono/metabolismo , Tetracloruro de Carbono/farmacología , Estradiol/farmacología , Estrógenos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Hepatocitos/metabolismo , Cirrosis Hepática/metabolismo , MicroARNs/biosíntesis , Animales , Intoxicación por Tetracloruro de Carbono/genética , Intoxicación por Tetracloruro de Carbono/patología , Intoxicación por Tetracloruro de Carbono/prevención & control , Línea Celular Tumoral , Femenino , Hepatocitos/patología , Humanos , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Caracteres Sexuales , Factores de Tiempo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
17.
Cell Res ; 22(3): 504-15, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21862971

RESUMEN

MicroRNAs (miRNAs) are endogenous noncoding RNAs (∼22 nt) that regulate target gene expression at the post-transcriptional level in the cytoplasm. Recent discoveries of the presence of miRNAs and miRNA function-required argonaute family proteins in the cell nucleus have prompted us to hypothesize that miRNAs may also have regulatory functions in the cell nucleus. In this study, we demonstrate that mouse miR-709 is predominantly located in the nucleus of various cell types and that its nuclear localization pattern rapidly changes upon apoptotic stimuli. In the cell nucleus, miR-709 directly binds to a 19-nt miR-709 recognition element on pri-miR-15a/16-1 and prevents its processing into pre-miR-15a/16-1, leading to a suppression of miR-15a/16-1 maturation. Furthermore, nuclear miR-709 participates in the regulation of cell apoptosis through the miR-15a/16-1 pathway. In summary, the present study provides the first evidence that one miRNA can control the biogenesis of other miRNAs by directly targeting their primary transcripts in the nucleus.


Asunto(s)
Núcleo Celular/genética , MicroARNs/biosíntesis , MicroARNs/genética , Procesamiento Postranscripcional del ARN/genética , Animales , Línea Celular , Células HEK293 , Humanos , Ratones , MicroARNs/aislamiento & purificación , Células 3T3 NIH
18.
Cell Res ; 22(1): 107-26, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21931358

RESUMEN

Our previous studies have demonstrated that stable microRNAs (miRNAs) in mammalian serum and plasma are actively secreted from tissues and cells and can serve as a novel class of biomarkers for diseases, and act as signaling molecules in intercellular communication. Here, we report the surprising finding that exogenous plant miRNAs are present in the sera and tissues of various animals and that these exogenous plant miRNAs are primarily acquired orally, through food intake. MIR168a is abundant in rice and is one of the most highly enriched exogenous plant miRNAs in the sera of Chinese subjects. Functional studies in vitro and in vivo demonstrated that MIR168a could bind to the human/mouse low-density lipoprotein receptor adapter protein 1 (LDLRAP1) mRNA, inhibit LDLRAP1 expression in liver, and consequently decrease LDL removal from mouse plasma. These findings demonstrate that exogenous plant miRNAs in food can regulate the expression of target genes in mammals.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Regulación de la Expresión Génica , MicroARNs/metabolismo , Oryza/genética , ARN de Planta/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Adulto , Animales , Pueblo Asiatico , Inmunoprecipitación de Cromatina , Biología Computacional , Dieta , Femenino , Células Hep G2 , Herbivoria , Humanos , Hígado/citología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/sangre , MicroARNs/genética , Persona de Mediana Edad , Plásmidos/genética , Plásmidos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN de Planta/sangre , ARN de Planta/genética , ARN no Traducido , Vesículas Secretoras/metabolismo , Especificidad por Sustrato , Transfección , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...