Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 300(6): 107376, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38762176

RESUMEN

Liver fibrosis/cirrhosis is a pathological state caused by excessive extracellular matrix deposition. Sustained activation of hepatic stellate cells (HSC) is the predominant cause of liver fibrosis, but the detailed mechanism is far from clear. In this study, we found that long noncoding RNA Fendrr is exclusively increased in hepatocytes in the murine model of CCl4- and bile duct ligation-induced liver fibrosis, as well as in the biopsies of liver cirrhosis patients. In vivo, ectopic expression of Fendrr aggravated the severity of CCl4-induced liver fibrosis in mice. In contrast, inhibiting Fendrr blockaded the activation of HSC and ameliorated CCl4-induced liver fibrosis. Our mechanistic study showed that Fendrr binds to STAT2 and enhances its enrichment in the nucleus, which then promote the expression of interleukin 6 (IL-6), and, ultimately, activates HSC in a paracrine manner. Accordingly, disrupting the interaction between Fendrr and STAT2 by ectopic expression of a STAT2 mutant attenuated the profibrotic response inspired by Fendrr in the CCl4-induced liver fibrosis. Notably, the increase of Fendrr in patient fibrotic liver is positively correlated with the severity of fibrosis and the expression of IL-6. Meanwhile, hepatic IL-6 positively correlates with the extent of liver fibrosis and HSC activation as well, thus suggesting a causative role of Fendrr in HSC activation and liver fibrosis. In conclusion, these observations identify an important regulatory cross talk between hepatocyte Fendrr and HSC activation in the progression of liver fibrosis, which might represent a potential strategy for therapeutic intervention.


Asunto(s)
Hepatocitos , Interleucina-6 , Cirrosis Hepática , ARN Largo no Codificante , Animales , Humanos , Masculino , Ratones , Tetracloruro de Carbono/toxicidad , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Hepatocitos/metabolismo , Hepatocitos/patología , Interleucina-6/metabolismo , Interleucina-6/genética , Cirrosis Hepática/metabolismo , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Ratones Endogámicos C57BL , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Factor de Transcripción STAT2/metabolismo , Factor de Transcripción STAT2/genética
2.
Free Radic Biol Med ; 220: 236-248, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38704052

RESUMEN

Alcoholic liver disease (ALD) is a common chronic redox disease caused by increased alcohol consumption. Abstinence is a major challenge for people with alcohol dependence, and approved drugs have limited efficacy. Therefore, this study aimed to explore a new treatment strategy for ALD using ferroferric oxide endohedral fullerenol (Fe3O4@C60(OH)n) in combination with static magnetic and electric fields (sBE). The primary hepatocytes of 8-9-week-old female BALB/c mice were used to evaluate the efficacy of the proposed combination treatment. A mouse chronic binge ethanol feeding model was established to determine the alleviatory effect of Fe3O4@C60(OH)n on liver injury under sBE exposure. Furthermore, the ability of Fe3O4@C60(OH)n to eliminate •OH was evaluated. Alcohol-induced hepatocyte and mitochondrial damage were reversed in vitro. Additionally, the combination therapy reduced liver damage, alleviated oxidative stress by improving antioxidant levels, and effectively inhibited liver lipid accumulation in animal experiments. Here, we used a combination of magnetic derivatives of fullerenol and sBE to further improve the ROS clearance rate, thereby alleviating ALD. The developed combination treatment may effectively improve alcohol-induced liver damage and maintain redox balance without apparent toxicity, thereby enhancing therapy aimed at ALD and other redox diseases.


Asunto(s)
Fulerenos , Hepatocitos , Hepatopatías Alcohólicas , Ratones Endogámicos BALB C , Estrés Oxidativo , Especies Reactivas de Oxígeno , Animales , Fulerenos/farmacología , Fulerenos/química , Fulerenos/uso terapéutico , Ratones , Especies Reactivas de Oxígeno/metabolismo , Femenino , Hepatocitos/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Estrés Oxidativo/efectos de los fármacos , Hepatopatías Alcohólicas/metabolismo , Hepatopatías Alcohólicas/patología , Hepatopatías Alcohólicas/tratamiento farmacológico , Hígado/metabolismo , Hígado/patología , Hígado/efectos de los fármacos , Antioxidantes/farmacología , Modelos Animales de Enfermedad , Humanos , Oxidación-Reducción/efectos de los fármacos , Etanol/toxicidad
3.
FASEB J ; 37(8): e23071, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37389924

RESUMEN

The sustained release of profibrotic cytokines, mainly transforming growth factor-ß (TGF-ß), leads to the occurrence of kidney fibrosis and chronic kidney disease (CKD). Connective tissue growth factor (CTGF) appears to be an alternative target to TGF-ß for antifibrotic therapy in CKD. In this study, we found that long noncoding RNA AI662270 was significantly increased in various renal fibrosis models. In vivo, ectopic expression of AI662270 alone was sufficient to activate interstitial fibroblasts and drive kidney fibrosis, whereas inhibition of AI662270 blocked the activation of interstitial fibroblasts and ameliorated kidney fibrosis in various murine models. Mechanistic studies revealed that overexpression of AI662270 significantly increased CTGF product, which was required for the role of AI662270 in driving kidney fibrosis. Furthermore, AI662270 binds to the CTGF promoter and directly interacts with METTL3, the methyltransferase of RNA N6 -methyladenosine (m6 A) modification. Functionally, AI662270-mediated recruitment of METTL3 increased the m6 A methylation of CTGF mRNA and consequently enhanced CTGF mRNA stability. In conclusion, our results support that AI662270 promotes CTGF expression at the posttranscriptional stage by recruiting METTL3 to the CTGF promoter and depositing m6 A modifications on the nascent mRNA, thereby, uncovering a novel regulatory mechanism of CTGF in the pathogenesis of kidney fibrosis.


Asunto(s)
ARN Largo no Codificante , Insuficiencia Renal Crónica , Animales , Ratones , Factor de Crecimiento del Tejido Conjuntivo/genética , Riñón , Metiltransferasas/genética , ARN Largo no Codificante/genética , ARN Mensajero/genética , Factor de Crecimiento Transformador beta/genética
4.
FEBS Lett ; 597(11): 1503-1516, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37171232

RESUMEN

Using a murine model of high-fat diet (HFD)-induced nonalcoholic fatty liver disease (NAFLD), we found that the expression of the epidermal growth factor receptor (EGFR) significantly decreased in hepatocytes. In vitro, free fatty acid influx decreased EGFR in hepatocytes. In HFD-fed mice, ectopic expression of EGFR alleviated intrahepatic lipid accumulation and reduced serum triglyceride and cholesterol, whereas knockdown of EGFR aggravated hepatic steatosis. Notably, EGFR inhibited the induction of lipogenic genes, including Srebf1, Srebf2, Fasn, Acc1 and Ppara, both in vitro and in vivo. Mechanistically, EGFR potentiates TGF-ß/Smad signalling and augments the inhibitory effects of TGF-ß1 on lipogenic genes in hepatocytes. Our findings suggest a hitherto unknown paradigm in the pathogenesis of NAFLD, thereby providing a rational basis for future therapeutic considerations.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Animales , Ratones , Dieta Alta en Grasa/efectos adversos , Receptores ErbB/metabolismo , Hepatocitos/metabolismo , Hígado/metabolismo , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/metabolismo
5.
Drug Deliv ; 29(1): 2984-2994, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36101018

RESUMEN

Colchicine (Col) is used to prevent and treat acute gout flare; however, its therapeutic use is strictly limited owing to severe gastrointestinal side effects after oral administration. Therefore, we developed a dissolvable Col-loaded microneedle (MN) with hyaluronic acid to deliver Col via the transdermal route. We studied the preparation, mechanical properties, skin insertion, skin irritation, drug content, and transdermal release of the Col-loaded MN. The pharmacokinetics of Col after Col-loaded MN application were compared with those of Col solution gavage over 24 h. Knee joint edema evaluation and the hindfoot mechanical threshold test were conducted to determine the pharmacodynamic profile. The dissolvable Col-loaded MN possessed sufficient mechanical strength to penetrate the skin and release the loaded drug. No skin irritation was observed for 3 days after application. We found that 3.36-fold more Col contained in MNs was delivered through the skin compared with that in gel in vitro, and moderate relative bioavailability in vivo. The Col-loaded MN significantly relieved swollen knee joints and mechanical hypernociception in an acute gout model in rats. The dissolvable Col-loaded MN array reduced inflammation and pain via topical administration when acute gout occurred. Reducing the gastrointestinal side effects of Col-loaded MNs is expected to optimize the therapeutic effects of Col and improve patient compliance.


Asunto(s)
Artritis Gotosa , Gota , Administración Cutánea , Animales , Colchicina , Sistemas de Liberación de Medicamentos , Gota/tratamiento farmacológico , Ratas , Brote de los Síntomas
6.
FEBS J ; 289(12): 3535-3549, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35038357

RESUMEN

High rates of metastasis and postsurgical recurrence contribute to the higher mortality of hepatocellular carcinoma (HCC), partly due to cancer stem cell (CSC)-dependent tumorigenesis and metastasis. Sex-determining region Y-box 9 (Sox9) has been previously characterized as a candidate CSC marker of HCC. Here, we observed that the increase of Sox9 significantly promoted HCC cell growth and invasion in cell cultures, whereas knockdown of Sox9 showed the opposite effects, suggesting that Sox9 may regulate the proliferation and invasion of hepatoma cells in an autocrine manner. RNA sequencing, together with functional assays and clinical analyses, identified CXCL5 as a key mediator downstream of Sox9 in HCC cells. Mechanistic studies revealed that Sox9 induced CXCL5 expression by directly binding to a promoter region. Using gain- and loss-of-function approaches, we demonstrated that the intrinsic effective role of Sox9 in hepatoma cell growth and invasion depended on CXCL5, and that blockade of CXCL5/CXCR2 signalling abolished Sox9-triggered HCC cell proliferation and migration. Furthermore, the Sox9/CXCL5 axis activated PI3K-AKT and ERK1/2 signalling which are implicated in regulating HCC cell proliferation and invasion. Finally, the Sox9/CXCL5 axis contributed to the infiltration of neutrophils and macrophages in both tumour and peritumoral tissues from the orthotopic xenograft model. In summary, our data identify the Sox9/CXCL5 axis as an endogenous factor in controlling HCC cell growth and invasion, thereby raising the possibility of pharmacologic intervention with CXCL5/CXCR2 pathway inhibitors in therapy for HCC patients with higher Sox9 expression.


Asunto(s)
Carcinoma Hepatocelular , Quimiocina CXCL5 , Neoplasias Hepáticas , Factor de Transcripción SOX9 , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Quimiocina CXCL5/genética , Quimiocina CXCL5/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/patología , Células Madre Neoplásicas/patología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo
7.
J Pathol ; 256(1): 25-37, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34543458

RESUMEN

Upstream stimuli for myofibroblast activation are of considerable interest for understanding the mechanisms underlying renal fibrosis. Activin B, a member of the TGF-ß family, exists as a homodimer of inhibin subunit beta B (INHBB), but its role in renal fibrosis remains unknown. We found that INHBB expression was significantly increased in various renal fibrosis models and human chronic kidney disease specimens with renal fibrosis. Notably, the increase of INHBB occurred mainly in the tubular epithelial cells (TECs). In vivo, inhibiting INHBB blocked the activation of interstitial fibroblasts and ameliorated the renal fibrosis induced by unilateral ureteral obstruction or ischemia-reperfusion injury, while ectopic expression of INHBB in the TECs was able to activate interstitial fibroblasts and initiate interstitial fibrosis. In vitro, overexpression of INHBB in TECs led to the secretion of activin B, thereby promoting the proliferation and activation of interstitial fibroblasts through activin B/Smad signaling. Furthermore, inhibition of activin B/Smad signaling attenuated the fibrotic response caused by tubular INHBB. Mechanistically, the upregulation of INHBB depended on the transcription factor Sox9 in the injured TECs. Clinical analyses also identified a positive correlation between Sox9 and INHBB expression in human specimens, suggesting the Sox9/INHBB axis as a positive regulator of renal fibrosis. In conclusion, tubule-derived INHBB is implicated in the pathogenesis of renal fibrosis by activating the surrounding fibroblasts in a paracrine manner, thereby exhibiting as a potential therapeutic target. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Fibroblastos/metabolismo , Fibrosis/metabolismo , Subunidades beta de Inhibinas/metabolismo , Animales , Proliferación Celular/fisiología , Fibroblastos/patología , Fibrosis/patología , Humanos , Riñón/metabolismo , Riñón/patología , Ratones Endogámicos C57BL , Miofibroblastos/metabolismo , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología , Regulación hacia Arriba , Obstrucción Ureteral/metabolismo , Obstrucción Ureteral/patología
8.
Biomedicines ; 9(6)2021 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-34071133

RESUMEN

This study aimed to improve the transdermal delivery of lidocaine hydrochloride (LidH) using elastic nano-liposomes (ENLs) and microneedle (MN) array pretreatment. LidH-containing ENLs were prepared using soybean phosphatidylcholine and cholesterol, with Span 80 or Tween 80, using a reverse-phase evaporation method. The ENL particle size, stability, and encapsulation efficiency (EE) were characterized and optimized based on the component ratio, pH, and type of surfactant used. In vitro transdermal diffusion study was performed on MN-pretreated mouse skin using Franz diffusion cells. The anesthetic effects of LidH in various formulations after dermal application were evaluated in vivo in rats by measuring the tail withdrawal latency after photothermic stimulation. Stable LidH-loaded Tween 80 or Span 80 ENLs were obtained with particle sizes of 115.8 and 146.6 nm and EEs of 27% and 20%, respectively. The formulations did not exert any cytotoxicity in HaCaT cells. Tween 80 and Span 80 ENL formulations showed enhanced LidH delivery on pretreated mice skin in vitro and prolonged the anesthetic effect in vivo compared to that by LidH application alone. LidH-loaded ENLs applied to MN-pretreated skin can shorten the onset time and prolong the anesthetic effect safely, which merits their further optimization and practical application.

9.
Mol Immunol ; 130: 104-112, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33309306

RESUMEN

Atopic dermatitis is a severe, chronic relapsing inflammatory disease of the skin with family clustering. It is characterized into acute phase, which is dominated by T helper 2-type immune responses, and chronic phase, which is dominated by T helper 1-type immune responses. Studies have shown that 3,3'-diindolylmethane not only has antitumor effects but also can relieve symptoms of inflammatory diseases by inhibiting the nuclear factor-κB signaling pathway and regulating T cell differentiation. To study the effect of 3,3'-diindolylmethane on atopic dermatitis and the underlying mechanism, a mouse model of acute atopic dermatitis was established using 2,4-dinitrofluorobenzene. After intraperitoneal injection of 3,3'-diindolylmethane, skin erythema and edema in mice were significantly alleviated. Furthermore, 3,3'-diindolylmethane reduced immune activation, probably by inhibiting the secretion of thymic stromal lymphopoietin by keratinocytes. 3,3'-Diindolylmethane also promoted the differentiation of regulatory T cells and inhibited the activation of T helper 2 and T helper 17 cells to reduce atopic dermatitis-related immune responses. However, it showed no significant effect on the differentiation of T helper 1 cells. These results indicate that 3,3'-diindolylmethane has a significant inhibitory effect on T helper 2 cells in the acute phase of atopic dermatitis. Our findings may provide not only more insights into the pathological mechanism of AD, but also a new candidate medicine for it.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Dermatitis Atópica/tratamiento farmacológico , Indoles/uso terapéutico , Linfocitos T/efectos de los fármacos , Enfermedad Aguda , Adulto , Animales , Diferenciación Celular/inmunología , Células Cultivadas , Dermatitis Atópica/inmunología , Modelos Animales de Enfermedad , Femenino , Células HaCaT , Humanos , Indoles/farmacología , Queratinocitos/efectos de los fármacos , Queratinocitos/fisiología , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Piel/efectos de los fármacos , Piel/inmunología , Linfocitos T/fisiología
10.
Cancer Lett ; 499: 243-254, 2021 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-33246092

RESUMEN

The activation of hepatic stellate cells (HSCs) and liver fibrosis in the peri-tumoral tissue contributes to the progression of hepatocellular carcinoma (HCC). However, the mechanisms underlying the crosstalk between hepatoma and peri-tumoral HSCs remain elusive. We found that the Sox9/INHBB axis is upregulated in HCC and is associated with tumor metastasis. Using gain- and loss-of-function approaches, we revealed that the Sox9/INHBB axis promotes the growth and metastasis of an orthotopic HCC tumor by activating the peri-tumoral HSCs. Mechanistically, Sox9 induces INHBB expression by directly binding to its enhancer, thus aiding in the secretion of activin B from hepatoma cells, and in turn, promoting the activation of the surrounding HSCs through activin B/Smad signaling. Furthermore, inhibition of activin B/Smad singaling attenuates the fibrotic response in the peri-tumoral tissue and decreases the incidence of metastasis. Finally, clinical analyses indicated a positive correlation between Sox9 and INHBB expression in HCC specimens and identified the Sox9/INHBB axis as a positive regulator of liver fibrosis. In conclusion, Sox9/INHBB axis-mediated crosstalk between hepatoma cells and HSCs induces a fertile environment favoring HCC metastasis, thereby exhibiting as a potential therapeutic target.


Asunto(s)
Carcinoma Hepatocelular/genética , Células Estrelladas Hepáticas/patología , Subunidades beta de Inhibinas/genética , Cirrosis Hepática/genética , Neoplasias Hepáticas/genética , Factor de Transcripción SOX9/metabolismo , Activinas/metabolismo , Animales , Carcinoma Hepatocelular/secundario , Proliferación Celular/genética , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células Hep G2 , Humanos , Hígado/citología , Hígado/patología , Cirrosis Hepática/patología , Neoplasias Hepáticas/patología , Masculino , Ratones , Comunicación Paracrina/genética , Factor de Transcripción SOX9/genética , Transducción de Señal/genética , Proteínas Smad/metabolismo , Microambiente Tumoral/genética , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Toxicol Sci ; 177(1): 214-225, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32579217

RESUMEN

Sox9 has been previously characterized as a transcription factor responsible for the extracellular matrix production during liver fibrosis. However, the deregulation and functional role of hepatocyte Sox9 in the progression of liver fibrosis remains elusive. Here, we found a significant increase of Sox9 in the hepatocytes isolated from CCl4-induced fibrotic liver and showed that antisense oligoribonucleotides depletion of Sox9 was sufficient to attenuate CCl4-induced liver fibrosis. Notably, the increase of Sox9 in hepatocyte was associated with the upregulation of long noncoding RNA H19 in both in vitro and in vivo systems. Mechanistic studies revealed that Sox9 induced H19 by binding to a conserved promoter region of H19. In vitro, hepatocyte injury triggered the increase of Sox9/H19 axis, whereas silence of H19 greatly alleviated the H2O2-induced hepatocyte apoptosis, suggesting that H19 functions as a downstream effector of Sox9 signaling and is involved in hepatocyte apoptosis. In animal experiments, inhibition of H19 alleviated the activation of hepatic stellate cells and reduced the extent of liver fibrosis, whereas ectopic expression of H19 abolished the inhibitory effects of Sox9 depletion on liver fibrosis, suggesting that the profibrotic effect of hepatocyte Sox9 depends on H19. Finally, we investigated the clinical relevance of Sox9/H19 axis to liver fibrosis and identified the increase of Sox9/H19 axis in liver cirrhosis patients. In conclusion, our findings link Sox9/H19 axis to the intrinsic mechanisms of hepatocyte apoptosis and may represent a hitherto unknown paradigm in hepatocyte injury associated with the progression of liver fibrosis.


Asunto(s)
ARN Largo no Codificante , Animales , Células Estrelladas Hepáticas/patología , Hepatocitos/patología , Humanos , Peróxido de Hidrógeno , Hígado/patología , Cirrosis Hepática/patología , Factor de Transcripción SOX9
12.
Kidney Int ; 96(3): 674-688, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31327474

RESUMEN

Tubular epithelial cell-derived profibrotic factors are known as the driving force in renal fibrosis for their roles in activating the surrounding fibroblast. However, the mechanisms driving their expressions remain undefined. Here, we find that kidney human epidermal growth factor receptor 2 (HER2), a member of the epidermal growth factor receptor family, significantly increased in unilateral ureteral obstruction-induced renal fibrosis, in type 1 and type 2 diabetic nephropathy, and in kidney biopsies from patients with renal fibrosis. Notably, the upregulation of HER2 mainly occurred in proximal tubular epithelial cells (PTECs). In vivo, the ectopic expression of HER2 in these cells was sufficient to activate the interstitial fibroblast and initiate interstitial fibrosis, whereas inhibiting HER2 reduced the accumulation of myofibroblasts and the extent of renal fibrosis in the mouse obstruction model and in streptozotocin (STZ)-induced diabetic mice. We also generated a tubular epithelial cell subline stably expressing HER2 and performed transcriptome RNA sequence analysis. This showed that sustained HER2 expression significantly induced the expression of profibrotic connective tissue growth factor (CTGF). Mechanistically, the induction of CTGF depended on the HER2-mediated activation of Stat3 in the tubular epithelium. In vitro, the incubation of kidney fibroblasts with culture medium from HER2-overexpressed tubular epithelial cells promoted fibroblast proliferation and activation, whereas silencing CTGF impeded the profibrotic effects of the tubular epithelial cell preconditioned media. Thus, our results highlight the significance of HER2 in tubular injury and characterize its role in promoting surrounding fibroblast activation and renal fibrosis in a paracrine manner.


Asunto(s)
Nefropatías Diabéticas/patología , Células Epiteliales/metabolismo , Fibroblastos/metabolismo , Túbulos Renales Proximales/patología , Receptor ErbB-2/metabolismo , Animales , Biopsia , Línea Celular , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Medios de Cultivo Condicionados/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/complicaciones , Nefropatías Diabéticas/etiología , Fibrosis , Técnicas de Silenciamiento del Gen , Humanos , Túbulos Renales Proximales/citología , Comunicación Paracrina/genética , RNA-Seq , Ratas , Receptor ErbB-2/genética , Factor de Transcripción STAT3/metabolismo , Estreptozocina/toxicidad , Regulación hacia Arriba , Obstrucción Ureteral/complicaciones
13.
Vaccine ; 37(30): 4081-4088, 2019 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-31164303

RESUMEN

While vaccination is highly effective for the prevention of many infectious diseases, the number of adjuvants licensed for human use is currently very limited. The aim of this study was to evaluate the safety, efficacy, and to clarify the mechanism of a phosphorothioated interleukin (IL)-10-targeted antisense oligonucleotide (ASO) as an immune adjuvant in intradermal vaccination. The cytotoxicity of IL-10 ASO and its ability to promote T cell proliferation were assessed by Cell Counting Kit-8 (CCK-8) assay. The contents of IL-6, IL-8, TNF-α, IL-1ß, and IL-10 in inoculated local tissue and the antigen-specific antibody titers in mouse serum samples were determined by ELISA. The target cells of IL-10 ASO were observed using immunofluorescent staining. The results showed that the specific antibody titer of ovalbumin (OVA), a model antigen, was increased 100-fold upon addition of IL-10 ASO as an adjuvant compared to that of OVA alone. IL-10 ASO showed an immunopotentiation efficacy similar to that of Freund's incomplete adjuvant, with no detectable cell or tissue toxicity. In vitro and in vivo experiments confirmed that IL-10 ASO enhances immune responses by temporarily suppressing IL-10 expression from local dendritic cells and consequently promoting T cell proliferation. In conclusion, IL-10 ASO significantly enhances immune responses against co-delivered vaccine antigens with high efficacy and low toxicity. It has the potential to be developed into a safe and efficient immune adjuvant.


Asunto(s)
Oligodesoxirribonucleótidos Antisentido/uso terapéutico , Adyuvantes Inmunológicos/uso terapéutico , Animales , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Adyuvante de Freund/uso terapéutico , Interleucina-10/metabolismo , Lípidos/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Células RAW 264.7 , Linfocitos T/metabolismo , Temperatura
14.
Biochim Biophys Acta Mol Basis Dis ; 1864(2): 520-532, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29158184

RESUMEN

TGF-ß signaling plays a principal role in renal fibrosis, but the precise mechanisms and the downstream factors are still largely unknown. Sox9 exhibits diverse roles in regulating the production of extracellular matrix proteins. Here we found that Sox9 was induced by TGF-ß in the kidney fibroblast and acted as an important downstream mediator of TGF-ß signaling in promoting renal fibrosis. TGF-ß/Smad signaling mediated the upregulation of Sox9 in kidney fibroblast by binding to a conserved enhancer. In different mouse models of renal fibrosis, as well as in the kidney biopsy tissue from patients with renal fibrosis, Sox9 expression significantly increased. Immunostaining confirmed the upregulation of Sox9 in the kidney fibroblast during renal fibrosis. Delivery of Sox9 knockdown plasmid to the kidney by ultrasound microbubble-mediated gene transfer suppressed the unilateral ureteral obstruction (UUO) or folic acid-induced mouse renal fibrosis, whereas ectopic expression of Sox9 aggravated renal fibrosis. In addition, we identified Sox9 as a direct target of miR-30. Notably, miR-30 expression was significantly inhibited by TGF-ß1 in the kidney fibroblast and the downregulation of miR-30 was observed in renal fibrosis. Mechanistically, inhibition of miR-30 independently strengthened the effect of TGF-ß/Smad signaling on Sox9 upregulation. Adenovirus-mediated ectopic expression of miR-30 in kidney fibroblast greatly reduced UUO-induced renal fibrosis by targeting Sox9. These findings link Sox9 to intrinsic mechanisms of TGF-ß signaling in renal fibrosis and may have therapeutic potential for tissue fibrosis.


Asunto(s)
Fibroblastos/metabolismo , Riñón/patología , Factor de Transcripción SOX9/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Fibrosis , Ácido Fólico/efectos adversos , Células HEK293 , Humanos , Riñón/metabolismo , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Ratas , Regulación hacia Arriba , Obstrucción Ureteral
15.
Sci Rep ; 7(1): 2957, 2017 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-28592847

RESUMEN

Hepatocyte death, as well as the following inflammatory and fibrogenic signaling cascades, is the key trigger of liver fibrosis. Here, we isolated hepatocytes from CCl4-induced fibrotic liver and found that hepatocyte lincRNA-p21 significantly increased during liver fibrosis. The increase of hepatocyte lincRNA-p21 was associated with the loss of miR-30, which can inhibit TGF-ß signaling by targeting KLF11. We revealed that lincRNA-p21 modulated miR-30 availability by acting as a competing endogenous RNA (ceRNA). The physiological significance of this interaction is highlighted by the feedback loop, in which lincRNA-p21 works as a downstream effector of the TGF-ß signaling to strengthen TGF-ß signaling and mediate its role in promoting liver fibrosis by interacting with miR-30. In vivo results showed that knockdown of hepatocyte lincRNA-p21 greatly reduced CCl4-induced liver fibrosis and inflammation, whereas ectopic expression of miR-30 in hepatocyte exhibited the similar results. Mechanistic studies further revealed that inhibition of miR-30 impaired the effects of lincRNA-p21 on liver fibrosis. Additionally, lincRNA-p21 promoted hepatocyte apoptosis in vitro and in vivo, whereas the proliferation rate of hepatocyte was suppressed by lincRNA-p21. The pleiotropic roles of hepatocyte lincRNA-p21 suggest that it may represent an unknown paradigm in liver fibrosis and serve as a potential target for therapy.


Asunto(s)
Hepatocitos/metabolismo , Cirrosis Hepática/etiología , Cirrosis Hepática/metabolismo , ARN Largo no Codificante/genética , Factor de Crecimiento Transformador beta/metabolismo , Animales , Apoptosis/genética , Biomarcadores , Proliferación Celular , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Hepatocitos/patología , Cirrosis Hepática/patología , Ratones , MicroARNs/genética , Interferencia de ARN , Transducción de Señal , Proteínas Smad/metabolismo
16.
J Cell Physiol ; 232(3): 625-634, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27333771

RESUMEN

Epithelial-mesenchymal transition (EMT) is critical for induction of invasiveness and metastasis in HCC. Growing evidence indicates that upregulation of Snail, the major EMT inducer, significantly correlates with the metastasis and poor prognosis of HCC. Here, we investigate the underlying mechanism of miR-30b in suppressing metastasis of hepatoma cells by targeting Snail. In this study, we found that miR-30b was significantly downregulated and negatively associated with Snail production in HCC cell lines with higher metastatic potentials. Gain- and loss-of-function studies revealed that miR-30b could dramatically inhibit in vitro HCC cell migration and invasion. In vivo orthotopic liver xenograft model further demonstrated that stable over-expression of miR-30b significantly repressed the local invasion and lung metastasis of hepatoma cells. Meanwhile, the restoration of miR-30b expression suppressed the distant colonization of hepatoma cells. Both gain- and loss-of-function studies showed that miR-30b suppressed the EMT of hepatoma cells as indicated by the morphology changes and deregulation of epithelial and mesenchymal markers. Using RNAi, we further investigated the role of Snail in HCC cell EMT and demonstrated that knockdown of Snail significantly inhibited the EMT and cancer cell metastasis. Additionally, miR-30b exhibited inhibitory effects on HCC cell proliferation in vitro and in vivo. In conclusion, our findings highlight the significance of miR-30b downregulation in HCC tumor metastasis and invasiveness, and implicate a new potential therapeutic target for HCC metastasis. J. Cell. Physiol. 232: 625-634, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Transición Epitelial-Mesenquimal/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Neoplasias Pulmonares/secundario , MicroARNs/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Pulmonares/patología , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , Invasividad Neoplásica , Factores de Transcripción de la Familia Snail/metabolismo
17.
PeerJ ; 4: e2451, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27672506

RESUMEN

OBJECTIVE: The skin is an important immunological barrier of the body as well as an optimal route for vaccine administration. Gua Sha, which involves press-stroke treatment of the skin, is an effective folk therapy, widely accepted in East Asia, for various symptoms; however, the mechanisms underlying its therapeutic effects have not been clarified. We investigated the influence of Gua Sha on the immunological features of the skin. METHODS: Gua Sha was performed on BALB/c mice and the effects were evaluated using anatomical, histological, and cytometric methods as well as cytokine determination locally and systemically. The effect on intradermal vaccination was assessed with antigen-specific subtype antibody responses. RESULTS: Blood vessel expansion, erythrocyte extravasation, and increased ratios of immune active cells were observed in the skin tissue following the treatment. Pro-inflammatory cytokines were up-regulated, and immunosuppressive cytokines, down-regulated, in the treated and untreated skin and systemic circulation; no obvious variations were detected in case of anti-inflammatory cytokines. Interestingly, intradermal delivery of a model vaccine following Gua Sha induced about three-fold higher IgG titers with a more Th1-biased antibody subtype profile. CONCLUSION: Gua Sha treatment can up-regulate the innate and adaptive immune functions of the skin and boost the response against intradermal antigens. Thus, Gua Sha may serve as a safe, inexpensive, and independent physical adjuvant for intradermal vaccination.

18.
Biomaterials ; 90: 72-84, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26994345

RESUMEN

Both tumor associated macrophages (TAMs) and tumor infiltrating dendritic cells (TIDCs) are important components in the tumor microenvironment that mediate tumor immunosuppression and promote cancer progression. Targeting these cells and altering their phenotypes may become a new strategy to recover their anti-tumor activities and thereby restore the local immune surveillance against tumor. In this study, we constructed a nucleic acid delivery system for the delivery of let-7b, a synthetic microRNA mimic. Our carrier has an affinity for the mannose receptors on TAMs/TIDCs and is responsive to the low-pH tumor microenvironment. The delivery of let-7b could reactivate TAMs/TIDCs by acting as a TLR-7 agonist and suppressing IL-10 production in vitro. In a breast cancer mouse model, let-7b delivered by this system efficiently reprogrammed the functions of TAMs/TIDCs, reversed the suppressive tumor microenvironment, and inhibited tumor growth. Taken together, this strategy, designed based upon TAMs/TIDCs-targeting delivery and the dual biological functions of let-7b (TLR-7 ligand and IL-10 inhibitor), may provide a new approach for cancer immunotherapy.


Asunto(s)
Neoplasias de la Mama/terapia , Células Dendríticas/patología , Técnicas de Transferencia de Gen , Macrófagos/patología , MicroARNs/uso terapéutico , Animales , Mama/inmunología , Mama/metabolismo , Mama/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Células Cultivadas , Técnicas de Reprogramación Celular/métodos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Terapia Genética , Humanos , Inmunoterapia , Interleucina-10/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones Endogámicos BALB C , MicroARNs/administración & dosificación , MicroARNs/genética , Receptor Toll-Like 7/inmunología , Microambiente Tumoral
19.
Toxicol Sci ; 146(1): 157-69, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25912033

RESUMEN

Transforming growth factor beta (TGF-ß) is crucial for transdifferentiation of hepatic stellate cells (HSCs) and the blunting of TGF-ß signaling in HSCs can effectively prevent liver fibrosis. Krüppel-like factor 11 (KLF11) is an early response transcription factor that potentiates TGF-ß/Smad signaling by suppressing the transcription of inhibitory Smad7. Using a mouse model of carbon tetrachloride (CCl4)-induced liver fibrosis, we observed significant upregulation of KLF11 in the activated HSCs during liver fibrogenesis. Meanwhile, the downregulation of miR-30 was observed in the HSCs isolated from fibrotic liver. Adenovirus-mediated ectopic expression of miR-30 was under the control of smooth muscle α-actin promoter, showing that the increase in miR-30 in HSC greatly reduced CCl4-induced liver fibrosis. Subsequent investigations showed that miR-30 suppressed KLF11 expression in HSC and led to a significant upregulation of Smad7 in vivo. Mechanistic studies further confirmed that KLF11 was the direct target of miR-30, and revealed that miR-30 blunted the profibrogenic TGF-ß signaling in HSC by suppressing KLF11 expression and thus enhanced the negative feedback loop of TGF-ß signaling imposed by Smad7. Finally, we demonstrated that miR-30 facilitated the reversal of activated HSC to a quiescent state as indicated by the inhibition of proliferation and migration, the loss of activation markers, and the gain of quiescent HSC markers. In conclusion, our results define miR-30 as a crucial suppressor of TGF-ß signaling in HSCs activation and provide useful insights into the mechanisms underlying liver fibrosis.


Asunto(s)
Intoxicación por Tetracloruro de Carbono/prevención & control , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Células Estrelladas Hepáticas/patología , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/prevención & control , MicroARNs/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Ratones
20.
J Cell Physiol ; 230(11): 2706-17, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25808945

RESUMEN

Epithelial-to-mesenchymal transition (EMT) has been implicated in embryonic development and various pathological events. However, the involvement of microRNA in the process of EMT remains to be fully defined in hepatocyte. ZEB1 is a well-known transcriptional repressor of E-cadherin and plays a major role in triggering EMT during organ fibrosis and cancer cell metastasis. Computational microRNA target predictions detect a conserved sequence matching to miR-101 in the 3'UTR of ZEB1 mRNA. Our results confirm that miR-101 suppresses ZEB1 expression by targeting the predicted site of ZEB1 3'UTR. Subsequent investigations show that miR-101 is significantly downregulated in the cultured hepatocytes undergoing EMT and in the hepatocytes isolated from fibrotic liver. Along with the loss of miR-101, the ZEB1 expression increases simultaneously in hepatocytes. In addition, miR-101 levels in HCC cell lines are negatively associated with the ZEB1 productions and the metastatic potentials of tumor cells. Mechanistically, we demonstrate that miR-101 significantly inhibits the TGF-ß1-induced EMT in hepatocytes, whereas inhibition of miR-101 promotes the EMT process as indicated by the changes of morphology, cell migration, and the expression profiles of EMT markers. In the fibrotic liver, ectopic expression of miR-101 can significantly downregulate ZEB1 in the hepatocyte and thereby reduces the mesenchymal marker expression. Moreover, miR-101 significantly inhibits the proliferation and migration of HCC cell. Our results demonstrate that miR-101 regulates HCC cell phenotype by upregulating the epithelial marker genes and suppressing the mesenchymal ones.


Asunto(s)
Carcinoma Hepatocelular/genética , Transición Epitelial-Mesenquimal/genética , Proteínas de Homeodominio/genética , Neoplasias Hepáticas/genética , MicroARNs/biosíntesis , Factores de Transcripción/genética , Regiones no Traducidas 3' , Cadherinas/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Hepatocitos/metabolismo , Hepatocitos/patología , Proteínas de Homeodominio/biosíntesis , Humanos , Neoplasias Hepáticas/patología , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Metástasis de la Neoplasia , Factores de Transcripción/biosíntesis , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...