Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Liver Int ; 44(6): 1373-1382, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38441405

RESUMEN

BACKGROUND & AIMS: Short videos, crucial for disseminating health information on metabolic dysfunction-associated steatotic liver disease (MASLD), lack a clear evaluation of quality and reliability. This study aimed to assess the quality and reliability of MASLD-related videos on Chinese platforms. METHODS: Video samples were collected from three platforms (TikTok, Kwai and Bilibili) during the period from November 2019 to July 2023. Two independent reviewers evaluated the integrity of the information contained therein by scoring six key aspects of its content: definition, epidemiology, risk factors, outcomes, diagnosis and treatment. The quality and reliability of the videos were assessed using the Journal of the American Medical Association (JAMA) criteria, the Global Quality Score (GQS) and the modified DISCERN score. RESULTS: A total of 198 videos were included. The video content exhibited an overall unsatisfactory quality, with a primary emphasis on risk factors and treatment, while diagnosis and epidemiology were seldom addressed. Regarding the sources of the videos, the GQS and modified DISCERN scores varied significantly between the platforms (p = .003), although they had generally similar JAMA scores (p = .251). Videos created by medical professionals differed significantly in terms of JAMA scores (p = .046) compared to those created by nonmedical professionals, but there were no statistically significant differences in GQS (p = .923) or modified DISCERN scores (p = .317). CONCLUSIONS: The overall quality and reliability of the videos were poor and varied between platforms and uploaders. Platforms and healthcare professionals should strive to provide more reliable health-related information regarding MASLD.


Asunto(s)
Grabación en Video , Humanos , Reproducibilidad de los Resultados , China/epidemiología , Factores de Riesgo , Enfermedad del Hígado Graso no Alcohólico/epidemiología , Enfermedad del Hígado Graso no Alcohólico/terapia , Hígado Graso/diagnóstico , Hígado Graso/terapia , Información de Salud al Consumidor/normas
2.
Helicobacter ; 29(1): e13048, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38716864

RESUMEN

Current global variations exist in Helicobacter pylori (H. pylori) eradication regimens. Triple therapy (TT), bismuth quadruple therapy (BQT), and high-dose dual therapy (HDDT) currently represent the predominant regimens. These regimens diverge in terms of treatment duration, the utilization of susceptibility testing, acid-inhibiting drug administration, and patient education. We conducted a comprehensive systematic literature review on these H. pylori treatment regimens. Our review aims to provide standardized treatment recommendations for H. pylori, reducing the risk of amalgamating findings from diverse eradication regimens. Recent research suggests that the optimal treatment duration for TT and BQT may be 14 and 10 days, respectively. Selecting the appropriate treatment duration for HDDT should rely on regional research evidence, and 14 days may be the optimal duration. The incorporation of susceptibility testing in TT is of paramount importance. In the case of BQT, the absence of susceptibility testing may be considered as an option, contingent upon cost and availability, and should be determined based on local antibiotic resistance patterns and the efficacy of empirical regimens. The type and dosage of acid-inhibiting drug would affect the efficacy of these regimens. Acid-inhibiting drugs should be selected and applied reasonably according to the population and therapies. Adequate patient education plays a pivotal role in the eradication of H. pylori. In regions with accessible local research evidence, the 10-day empirical BQT regimen may be considered a preferred choice for H. pylori eradication.


Asunto(s)
Antibacterianos , Quimioterapia Combinada , Infecciones por Helicobacter , Helicobacter pylori , Humanos , Antibacterianos/uso terapéutico , Bismuto/uso terapéutico , Infecciones por Helicobacter/tratamiento farmacológico , Infecciones por Helicobacter/microbiología , Helicobacter pylori/efectos de los fármacos , Inhibidores de la Bomba de Protones/uso terapéutico
3.
Gastroenterology ; 162(1): 150-165, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34536451

RESUMEN

BACKGROUND & AIMS: G protein-coupled receptor (GPR) 120 has been implicated in regulating metabolic syndromes with anti-inflammatory function. However, the role of GPR120 in intestinal inflammation is unknown. Here, we investigated whether and how GPR120 regulates CD4+ T cell function to inhibit colitis development. METHODS: Dextran sodium sulfate (DSS)-induced colitis model, Citrobacter rodentium infection model, and CD4+ T cell adoptive transfer model were used to analyze the role of GPR120 in regulating colitis development. The effect of GPR120 on CD4+ T cell functions was analyzed by RNA sequencing, flow cytometry, and Seahorse metabolic assays. Mice were administered GPR120 agonist for investigating the potential of GPR120 agonist in preventing and treating colitis. RESULTS: Deficiency of GPR120 in CD4+ T cells resulted in more severe colitis in mice upon dextran sodium sulfate insult and enteric infection. Transfer of GPR120-deficient CD4+CD45Rbhi T cells induced more severe colitis in Rag-/- mice with lower intestinal interleukin (IL) 10+CD4+ T cells. Treatment with the GPR120 agonist CpdA promoted CD4+ T cell production of IL10 by up-regulating Blimp1 and enhancing glycolysis, which was regulated by mTOR. GPR120 agonist-treated wild-type, but not IL10-deficient and Blimp1-deficient, T helper 1 cells induced less severe colitis. Furthermore, oral administration of GPR120 agonist protected mice from intestinal inflammation in both prevention and treatment schemes. Gpr120 expression was positively correlated with Il10 expression in the human colonic mucosa, including patients with inflammatory bowel diseases. CONCLUSIONS: Our findings show the role of GPR120 in regulating intestinal CD4+ T cell production of IL10 to inhibit colitis development, which identifies GPR120 as a potential therapeutic target for treating inflammatory bowel diseases.


Asunto(s)
Acetatos/farmacología , Antiinflamatorios no Esteroideos/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Colitis/prevención & control , Colon/efectos de los fármacos , Interleucina-10/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Tiramina/análogos & derivados , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/trasplante , Estudios de Casos y Controles , Colitis/inmunología , Colitis/metabolismo , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/metabolismo , Colon/inmunología , Colon/metabolismo , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/metabolismo , Modelos Animales de Enfermedad , Glucólisis/efectos de los fármacos , Interleucina-10/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Tiramina/farmacología
4.
J Immunol ; 207(3): 985-995, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34301843

RESUMEN

An uncontrolled CD4+ T cell response is a critical hallmark of autoimmune diseases. IL-10, which can be produced by both effector and regulatory CD4+ T cells, plays an essential role in the inhibition of autoimmunity. MicroRNAs are key molecules involved in regulating immune responses. However, how miR-10a regulates CD4+ T cell function in the pathogenesis of intestinal immune responses is not fully understood. In this study, we show that the mice with deficient miR-10a in CD4+ T cells were more resistant to intestinal inflammation upon inflammatory insult. miR-10a-deficient CD4+CD45Rbhi T cells were less colitogenic in Rag -/- mice, in which CD4+ T cell production of IL-10 was increased. miR-10a-deficient CD4+ T cells expressed a higher expression of IL-10 in vitro. Blocking the IL-10/IL-10R pathway in vivo aggravated colitis induced by miR-10a-deficient CD4+CD45Rbhi T cells. Mechanically, miR-10a suppressed CD4+ T cell production of IL-10 through targeting Prdm1, which encodes Blimp1. We further show that that CD4+ T cells lacking Blimp1 produced lower levels of IL-10 and induced more severe colitis in Rag -/- mice. These data thus establish the role of miR-10a in the inhibition of IL-10 production in CD4+ T cells to regulate intestinal homeostasis.


Asunto(s)
Colitis , MicroARNs , Animales , Linfocitos T CD4-Positivos , Interleucina-10/genética , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética
5.
J Pathol ; 252(4): 358-370, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32770671

RESUMEN

Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related death worldwide. Lipogenesis has been considered as a critical player in HCC initiation and progression. However, the underlying mechanism is still not fully understood. Here, we identified zinc fingers and homeoboxes 2 (ZHX2), an HCC-associated tumor suppressor, as an important repressor of de novo lipogenesis. Ectopic expression of ZHX2 significantly inhibited de novo lipogenesis in HCC cells and decreased expression of FASN, ACL, ACC1, and SCD1. In accordance with this, ZHX2 was negatively associated with SREBP1c, the master regulator of de novo lipogenesis, in HCC cell lines and human specimens. Results from silencing and overexpression demonstrated that ZHX2 inhibited de novo lipogenesis and consequent HCC progression via repression of SREBP1c. Furthermore, treatment with the SREBP1c inhibitor fatostatin dampened the spontaneous formation of tumors in liver-specific Zhx2 knockout mice. Mechanistically, ZHX2 increased expression of miR-24-3p transcriptionally, which targeted SREBP1c and led to its degradation. In conclusion, our data suggest a novel mechanism through which ZHX2 suppresses HCC progression, which may provide a new strategy for the treatment of HCC. © 2020 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Proteínas de Homeodominio/metabolismo , Lipogénesis/genética , Neoplasias Hepáticas/metabolismo , MicroARNs/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Factores de Transcripción/metabolismo , Adulto , Anciano , Animales , Carcinogénesis/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular/genética , Modelos Animales de Enfermedad , Ácidos Grasos no Esterificados/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/patología , Proteínas de Homeodominio/genética , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Noqueados , MicroARNs/genética , Persona de Mediana Edad , Piridinas/farmacología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/antagonistas & inhibidores , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Tiazoles/farmacología , Factores de Transcripción/genética , Triglicéridos/metabolismo
6.
J Immunol ; 203(1): 282-292, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31076530

RESUMEN

The gut microbiota has been shown critical for mucosal adjuvant activity of cholera toxin (CT), a potent mucosal adjuvant. However, the mechanisms involved remain largely unknown. In this study, we report that depletion of gut bacteria significantly decreased mucosal and systemic Ab responses in mice orally immunized with OVA and CT. Feeding mice short-chain fatty acids (SCFAs) promoted Ab responses elicited by CT, and, more importantly, rescued Ab responses in antibiotic-treated mice. In addition, mice deficient in GPR43, a receptor for SCFAs, showed impaired adjuvant activity of CT. Administering CT did not promote SCFA production in the intestines; thus, SCFAs facilitated but did not directly mediate the adjuvant activity of CT. SCFAs promoted B cell Ab production by promoting dendritic cell production of BAFF and ALDH1a2, which induced B cell expression of IFN regulatory factor 4, Blimp1, and XBP1, the plasma B cell differentiation-related genes. Furthermore, when infected with Citrobacter rodentium, GPR43-/- mice exhibited decreased Ab responses and were more susceptible to infection, whereas the administration of SCFAs promoted intestinal Ab responses in wild-type mice. Our study thereby demonstrated a critical role of gut microbiota and their metabolite SCFAs in promoting mucosal adjuvant activity of CT through GPR43.


Asunto(s)
Adyuvantes Inmunológicos/metabolismo , Linfocitos B/inmunología , Toxina del Cólera/metabolismo , Citrobacter rodentium/fisiología , Células Dendríticas/inmunología , Infecciones por Enterobacteriaceae/inmunología , Ácidos Grasos Volátiles/metabolismo , Microbioma Gastrointestinal/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Familia de Aldehído Deshidrogenasa 1/metabolismo , Animales , Formación de Anticuerpos , Factor Activador de Células B/metabolismo , Diferenciación Celular , Inmunidad Mucosa , Ratones , Ratones Noqueados , Comunicación Paracrina , Receptores Acoplados a Proteínas G/genética , Retinal-Deshidrogenasa/metabolismo
7.
Int J Cancer ; 143(12): 3120-3130, 2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29752719

RESUMEN

Initiation of hepatocellular carcinoma (HCC) by chronic hepatitis B virus (HBV) infection is a complex process that includes both oncogene activation and tumor suppressor inhibition. The HBV X (HBx) protein has an important and complex role in processes leading to HCC. We previously identified the mammalian Zinc fingers and homeoboxes 2 (ZHX2) gene as an HCC-associated tumor suppressor gene. In the present study, we investigated whether the oncogenic properties of HBV and, more specifically, HBx, involved ZHX2 silencing. Our data indicates that ZHX2 expression is significantly decreased in tumor tissues from HBV-positive HCC patients and livers from HBV transgenic mice. In vitro and in vivo studies confirmed that HBV-encoded proteins, particularly HBx, inhibits both the expression and tumor suppression properties of ZHX2. Further analyses identified miR-155, a well-known oncomiR in various cancers, as an important link between HBx and ZHX2 inhibition. Increased miR-155 levels were found in HBV-positive tumors, livers of HBV transgenic mice and HBx-overexpressing hepatoma cell lines. MiR-155 overexpression reduced ZHX2 levels via miR-155 seed sites in the ZHX2 3'UTR, whereas blocking miR-155 levels led to increased ZHX2 levels. Taken together, our data indicate that HCC-promoting properties of HBV may include ZHX2 silencing via a miR-155 dependent pathway and suggests a novel therapy for HBV-related HCC.


Asunto(s)
Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/virología , Proliferación Celular/genética , Virus de la Hepatitis B/fisiología , Hepatitis B Crónica/complicaciones , Proteínas de Homeodominio/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , MicroARNs/genética , Factores de Transcripción/metabolismo , Adulto , Anciano , Animales , Carcinoma Hepatocelular/complicaciones , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo , Femenino , Silenciador del Gen , Proteínas de Homeodominio/genética , Humanos , Neoplasias Hepáticas/complicaciones , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Persona de Mediana Edad , Factores de Riesgo , Transactivadores/metabolismo , Factores de Transcripción/genética , Regulación hacia Arriba , Proteínas Reguladoras y Accesorias Virales
8.
J Innate Immun ; 15(1): 709-723, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37725937

RESUMEN

Piezo1, the mechanosensory ion channel, has attracted increasing attention for its essential roles in various inflammatory responses and immune-related diseases. Although most of the key immune cells in inflammatory bowel disease (IBD) have been reported to be regulated by Piezo1, the specific role of Piezo1 in colitis has yet to be intensively studied. The present study investigated the impact of pharmacological inhibition of Piezo1 on dextran sulfate sodium (DSS)-induced colitis and explored the role of Piezo1 in intestinal immune cells in the context of colitis. We observed upregulated expression of Piezo1 in the colon tissue of mice with DSS-induced colitis. Pharmacological inhibition of Piezo1 by GsMTx4 diminished the severity of colitis. Piezo1 inhibition downregulated the expression of pro-inflammatory mediators Il1b, Il6, and Ptgs2 in colonic tissue and suppressed the production of IL-6 from macrophages and dendritic cells without altering the balance of T helper (Th) cells. In particular, Piezo1 did not affect cell viability but regulated cell proliferation and production of IL-17A in group 3 innate lymphoid cells (ILC3s), which is dependent on the PI3K-Akt-mTOR signaling pathway. Our findings uncover Piezo1 as an effective regulator of gut inflammation. Targeting Piezo1 could be a promising strategy to modulate intestinal immunity in IBD.


Asunto(s)
Colitis , Inmunidad Innata , Canales Iónicos , Linfocitos , Animales , Ratones , Colitis/inducido químicamente , Colitis/metabolismo , Colon/metabolismo , Modelos Animales de Enfermedad , Inflamación/metabolismo , Canales Iónicos/antagonistas & inhibidores , Canales Iónicos/genética , Canales Iónicos/metabolismo , Linfocitos/inmunología , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/metabolismo
9.
Inflamm Bowel Dis ; 28(11): 1766-1775, 2022 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-35486387

RESUMEN

Inflammatory bowel disease (IBD), characterized by chronic inflammation of the gastrointestinal tract, is a global health care problem. Compelling evidence shows sex differences regarding the prevalence, pathophysiology, clinical presentation, and treatment outcome of IBD. Sex hormones, including estrogen, progesterone, and androgen, have been proposed to have a role in the pathogenesis of sexual dimorphism in IBD. Clinical and experimental data support the modulatory effects of sex hormones on various clinical characteristics of the disease, including intestinal barrier dysfunction and mucosal immune activation. Additionally, the potential role of sex hormones in the modulation of gut microbiota is attracting increasing attention. Here, we discuss the sex dimorphic disease profile and address the potential mechanisms involved in the sex-specific pathogenesis of IBD. Improved understanding of these sex differences in the clinic could improve the knowledge of patients with IBD with heterogeneous disease profiles.


Asunto(s)
Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Humanos , Femenino , Masculino , Caracteres Sexuales , Enfermedades Inflamatorias del Intestino/terapia , Hormonas Esteroides Gonadales
10.
Cell Mol Gastroenterol Hepatol ; 14(6): 1177-1198, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35987451

RESUMEN

BACKGROUND & AIMS: Covalently closed circular DNA (cccDNA) of hepatitis B virus (HBV), existing as a stable minichromosome in the hepatocyte, is responsible for persistent HBV infection. Maintenance and sustained replication of cccDNA require its interaction with both viral and host proteins. However, the cccDNA-interacting host factors that limit HBV replication remain elusive. METHODS: Minicircle HBV (MC-HBV), a recombinant cccDNA, was constructed based on chimeric intron and minicircle DNA technology. By mass spectrometry based on pull-down with biotinylated MC-HBV, the cccDNA-hepatocyte interaction profile was mapped. HBV replication was assessed in different cell models that support cccDNA formation. RESULTS: MC-HBV supports persistent HBV replication and mimics the cccDNA minichromosome. The MC-HBV-based screen identified cohesin complex as a cccDNA binding host factor, leading to reduced HBV replication. Mechanistically, with the help of CCCTC-binding factor (CTCF), which has specific binding sites on cccDNA, cohesin loads on cccDNA and reshapes cccDNA confirmation to prevent RNA polymerase II enrichment. Interestingly, HBV X protein transcriptionally reduces structural maintenance of chromosomes complex expression to partially relieve the inhibitory role of the cohesin complex on HBV replication. CONCLUSIONS: Our data not only provide a feasible approach to explore cccDNA-binding factors, but also identify cohesin/CTCF complex as a critical host restriction factor for cccDNA-driven HBV replication. These findings provide a novel insight into cccDNA-host interaction and targeted therapeutic intervention for HBV infection.


Asunto(s)
ADN Circular , Virus de la Hepatitis B , ADN Circular/genética , Proteínas Cromosómicas no Histona/genética , Cromosomas , Cohesinas
11.
Nat Commun ; 11(1): 4457, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32901017

RESUMEN

Innate lymphoid cells (ILCs) and CD4+ T cells produce IL-22, which is critical for intestinal immunity. The microbiota is central to IL-22 production in the intestines; however, the factors that regulate IL-22 production by CD4+ T cells and ILCs are not clear. Here, we show that microbiota-derived short-chain fatty acids (SCFAs) promote IL-22 production by CD4+ T cells and ILCs through G-protein receptor 41 (GPR41) and inhibiting histone deacetylase (HDAC). SCFAs upregulate IL-22 production by promoting aryl hydrocarbon receptor (AhR) and hypoxia-inducible factor 1α (HIF1α) expression, which are differentially regulated by mTOR and Stat3. HIF1α binds directly to the Il22 promoter, and SCFAs increase HIF1α binding to the Il22 promoter through histone modification. SCFA supplementation enhances IL-22 production, which protects intestines from inflammation. SCFAs promote human CD4+ T cell IL-22 production. These findings establish the roles of SCFAs in inducing IL-22 production in CD4+ T cells and ILCs to maintain intestinal homeostasis.


Asunto(s)
Ácidos Grasos Volátiles/inmunología , Microbioma Gastrointestinal/inmunología , Inmunidad Innata , Interleucinas/biosíntesis , Animales , Butiratos/inmunología , Butiratos/metabolismo , Butiratos/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/microbiología , Citrobacter rodentium , Colitis/inmunología , Colitis/microbiología , Colitis/prevención & control , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/microbiología , Infecciones por Enterobacteriaceae/prevención & control , Ácidos Grasos Volátiles/metabolismo , Ácidos Grasos Volátiles/farmacología , Microbioma Gastrointestinal/fisiología , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Técnicas In Vitro , Interleucinas/deficiencia , Interleucinas/genética , Linfocitos/efectos de los fármacos , Linfocitos/inmunología , Linfocitos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Regiones Promotoras Genéticas , Receptores de Hidrocarburo de Aril/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Interleucina-22
12.
EBioMedicine ; 53: 102676, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32114388

RESUMEN

BACKGROUND: Liver cancer stem cells (CSCs) are critical determinants of HCC relapse and therapeutic resistance, but the mechanisms underlying the maintenance of CSCs are poorly understood. We aimed to explore the role of tumor repressor Zinc-fingers and homeoboxes 2 (ZHX2) in liver CSCs. METHODS: CD133+ or EPCAM+ stem-like liver cancer cells were sorted from tumor tissues of HCC patients and HCC cell lines by flow cytometry. In addition, sorafenib-resistant cells, tumor-sphere forming cells and side population (SP) cells were respectively cultured and isolated as hepatic CSCs. The tumor-initiating and chemoresistance properties of ZHX2-overexpressing and ZHX2-knockdown cells were analyzed in vivo and in vitro. Microarray, luciferase reporter assay, chromatin immunoprecipitation (ChIP) and ChIP-on-chip analyses were performed to explore ZHX2 target genes. The expression of ZHX2 and its target gene were determined by quantitative RT-PCR, western blot, immunofluorescence and immunohistochemical staining in hepatoma cells and tumor and adjacent tissues from HCC patients. RESULTS: ZHX2 expression was significantly reduced in liver CSCs from different origins. ZHX2 deficiency led to enhanced liver tumor progression and expansion of CSC populations in vitro and in vivo. Re-expression of ZHX2 restricted capabilities of hepatic CSCs in supporting tumor initiation, self-renewal and sorafenib-resistance. Mechanically, ZHX2 suppressed liver CSCs via inhibiting KDM2A-mediated demethylation of histone H3 lysine 36 (H3K36) at the promoter regions of stemness-associated transcription factors, such as NANOG, SOX4 and OCT4. Moreover, patients with lower expression of ZHX2 and higher expression of KDM2A in tumor tissues showed significantly poorer survival. CONCLUSION: ZHX2 counteracts stem cell traits through transcriptionally repressing KDM2A in HCC. Our data will aid in a better understanding of molecular mechanisms underlying HCC relapse and drug resistance.


Asunto(s)
Carcinoma Hepatocelular/genética , Proteínas F-Box/metabolismo , Código de Histonas , Proteínas de Homeodominio/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Neoplasias Hepáticas/genética , Células Madre Neoplásicas/metabolismo , Factores de Transcripción/metabolismo , Animales , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Proteínas F-Box/genética , Femenino , Células Hep G2 , Proteínas de Homeodominio/genética , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Metilación , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Factores de Transcripción SOXC/genética , Factores de Transcripción SOXC/metabolismo , Factores de Transcripción/genética
13.
World J Gastroenterol ; 25(36): 5469-5482, 2019 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-31576093

RESUMEN

BACKGROUND: Irritable bowel syndrome (IBS) is one of the most common functional gas-troenterological diseases characterized by abnormal visceral sensitivity and low-grade inflammation. The role of Clostridium butyricum (C. butyricum) in reducing intestinal low-grade inflammation via immune pathways has been well defined. However, the detailed mechanisms of the effects of C. butyricum on intestinal mucosal immunity, especially on immune cells of the lamina propria, remain unclear. Dendritic cells (DCs), which are important immune cells, secrete proinflammatory cytokines (IL-1ß, IL-6, and others) and express T cell immuno-globulin and mucin domain-3 (TIM3), promoting proliferation and activation of DCs, and mediating Th1 and Th17 inflammatory responses. AIM: To investigate the role of DCs in the development of IBS in a rat model and to understand the regulation of DCs after C. butyricum intervention. METHODS: An IBS animal model was established using C57BL/6 mice, and C. butyricum was continuously administered via the intragastric route to simulate different intestinal immune states. Intestinal visceral hypersensitivity and histopathology were assessed using the abdominal withdrawal reflex (AWR) test and hematoxylin & eosin (H&E) staining, respectively. The expression of proinflammatory cytokines (IL-1ß and IL-6) and TIM3 was analyzed by Western blot analysis and real-time PCR. Flow cytometry was applied to analyze the quantity, function, and membrane molecule TIM3 of the lamina propria dendritic cells (LPDCs). The regulatory effect of C. butyricum was verified in bone marrow-derived dendritic cells by in vitro experiments. RESULTS: The secretion of proinflammatory cytokines (IL-1ß and IL-6) in mice with IBS was significantly increased compared with that of the control group, which suggested that the intestinal mucosa in mice with IBS was in a low-grade inflammatory state. The expression of CD11C+CD80+ and CD11c+TIM3+ in intestinal LPDCs in mice with IBS increased significantly. Meanwhile, the cytokines (IL-1ß and IL-6) were significantly reduced after the intervention with probiotic C. butyricum. The amount and function of LPDCs and the TIM3 on the surface of the LPDCs were decreased with the alleviation of the intestinal inflammatory response. CONCLUSION: The results suggest that C. butyricum regulates the amount and functional status of LPDCs in the intestinal mucosa of mice with IBS, and therefore modulates the local immune response in the intestine.


Asunto(s)
Clostridium butyricum/inmunología , Células Dendríticas/inmunología , Mucosa Intestinal/inmunología , Síndrome del Colon Irritable/terapia , Probióticos/administración & dosificación , Animales , Modelos Animales de Enfermedad , Humanos , Inmunidad Mucosa , Mucosa Intestinal/citología , Mucosa Intestinal/microbiología , Síndrome del Colon Irritable/inducido químicamente , Síndrome del Colon Irritable/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Resultado del Tratamiento , Ácido Trinitrobencenosulfónico/toxicidad
14.
Mol Metab ; 23: 24-36, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30862474

RESUMEN

OBJECTIVE: Macrophage-mediated inflammation plays a significant role in the development and progression of diabetic nephropathy (DN). However, the underlying mechanisms remain unclear. Studies suggest that T cell immunoglobulin domain and mucin domain-3 (Tim-3) has complicated roles in regulating macrophage activation, but its roles in the progression of DN are still completely unknown. METHODS: We downregulated Tim-3 expression in kidney (intrarenal injection of Tim-3 shRNA expressing lentivirus or global Tim-3 knockout mice) and induced DN by streptozotocin (STZ). We analyzed the degree of renal injury, especially the podocyte injury induced by activated macrophages in vitro and in vivo. Then, we transferred different bone marrow derived macrophages (BMs) into STZ-induced Tim-3 knockdown mice to examine the effects of Tim-3 on macrophages in DN. RESULTS: First, we found that Tim-3 expression on renal macrophages was increased in patients with DN and in two diabetic mouse models, i.e. STZ-induced diabetic mice and db/db mice, and positively correlated with renal dysfunction of DN patients. Tim-3 deficiency ameliorated renal damage in STZ-induced diabetes with concurrent increase in protein levels of Nephrin and WT-1. Similar effects were observed in mice with Tim-3 knockdown diabetic mice. Second, adoptive transfer of Tim-3-expressing macrophages, but not Tim-3 knockout macrophages, accelerated diabetic renal injury in DN mice, suggesting a key role for Tim-3 on macrophages in the development of DN. Furthermore, we found NF-κB activation and TNF-α excretion were upregulated by Tim-3 in diabetic kidneys, and podocyte injury was associated with the Tim-3-mediated activation of the NF-κB/TNF-α signaling pathway in DN macrophages both in vivo and in vitro. CONCLUSIONS: These results suggest that Tim-3 functions as a key regulator in renal inflammatory processes and serves as a potential therapeutic target for renal injury in DN.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Activación de Macrófagos , FN-kappa B/metabolismo , Podocitos/patología , Factor de Necrosis Tumoral alfa/metabolismo , Traslado Adoptivo , Animales , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/inducido químicamente , Nefropatías Diabéticas/patología , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Receptor 2 Celular del Virus de la Hepatitis A/genética , Humanos , Macrófagos/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estreptozocina/farmacología
15.
Inflamm Bowel Dis ; 25(9): 1450-1461, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-30918945

RESUMEN

BACKGROUND: How the gut microbiota regulates intestinal homeostasis is not completely clear. Gut microbiota metabolite short-chain fatty acids (SCFAs) have been reported to regulate T-cell differentiation. However, the mechanisms underlying SCFA regulation of T-cell differentiation and function remain to be investigated. METHODS: CBir1, an immunodominant microbiota antigen, transgenic T cells were treated with butyrate under various T-cell polarization conditions to investigate butyrate regulation of T-cell differentiation and the mechanism involved. Transfer of butyrate-treated CBir T cells into Rag1-/- mice was performed to study the in vivo role of such T cells in inducing colitis. RESULTS: Although butyrate promoted Th1 cell development by promoting IFN-γ and T-bet expression, it inhibited Th17 cell development by suppressing IL-17, Rorα, and Rorγt expression. Interestingly, butyrate upregulated IL-10 production in T cells both under Th1 and Th17 cell conditions. Furthermore, butyrate induced T-cell B-lymphocyte-induced maturation protein 1 (Blimp1) expression, and deficiency of Blimp1 in T cells impaired the butyrate upregulation of IL-10 production, indicating that butyrate promotes T-cell IL-10 production at least partially through Blimp1. Rag1-/- mice transferred with butyrate-treated T cells demonstrated less severe colitis, compared with transfer of untreated T cells, and administration of anti-IL-10R antibody exacerbated colitis development in Rag-/- mice that had received butyrate-treated T cells. Mechanistically, the effects of butyrate on the development of Th1 cells was through inhibition of histone deacetylase but was independent of GPR43. CONCLUSIONS: These data indicate that butyrate controls the capacity of T cells in the induction of colitis by differentially regulating Th1 and Th17 cell differentiation and promoting IL-10 production, providing insights into butyrate as a potential therapeutic for the treatment of inflammatory bowel disease.


Asunto(s)
Butiratos/farmacología , Diferenciación Celular/inmunología , Colitis/etiología , Microbioma Gastrointestinal/fisiología , Mucosa Intestinal/metabolismo , Células TH1/inmunología , Células Th17/inmunología , Animales , Diferenciación Celular/efectos de los fármacos , Colitis/metabolismo , Colitis/patología , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Interleucina-17/inmunología , Interleucina-17/metabolismo , Mucosa Intestinal/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Proteínas de Dominio T Box/metabolismo , Células TH1/efectos de los fármacos , Células TH1/metabolismo , Células TH1/patología , Células Th17/efectos de los fármacos , Células Th17/metabolismo , Células Th17/patología
16.
Antiviral Res ; 153: 114-123, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29580980

RESUMEN

Hepatitis B virus (HBV) covalently closed circular DNA (cccDNA), the stable genomic form as the template for viral transcription, plays a crucial role in viral persistence which remains a major global health problem. While accumulating evidence suggests the involvement of transcription factors and epigenetic machinery in cccDNA transcription, the roles of host transcription factors which contribute to epigenetic modification of cccDNA remain largely unknown. Zinc finger and homeoboxes 2 (ZHX2) is abundantly expressed in adult hepatocytes, where it acts as a transcriptional repressor and tumor suppressor by directly inhibiting the promoter activities of target genes. However, whether ZHX2 influences HBV replication or is involved in cccDNA epigenetic regulation remain unknown. In this study, we investigated the role of ZHX2 in cccDNA transcription. Analysis of immunohistochemistry showed that ZHX2 nuclear expression negatively correlated with serum HBV DNA and HBeAg. Remarkably, ZHX2 significantly decreased HBV antigens expression, pregenomic RNA (pgRNA) and HBV core particle DNA production both in vitro and in mouse livers supporting HBV antigens expression and cccDNA transcription. Dual luciferase and cccDNA ChIP assays confirmed that ZHX2 could bind to cccDNA and transcriptionally inhibit HBV promoter activities. In addition, ZHX2 suppressed the expression of histone regulator genes, such as cccDNA bound p300/CBP, and led to epigenetic repression of cccDNA. These findings highlight the roles of a novel restriction factor, ZHX2, in modulating HBV replication via regulating HBV promoter activities and cccDNA modifications. This study furthers our understanding of HBV transcription from cccDNA and offers new insights on potential HBV therapy.


Asunto(s)
Regulación Viral de la Expresión Génica/efectos de los fármacos , Virus de la Hepatitis B/crecimiento & desarrollo , Virus de la Hepatitis B/inmunología , Proteínas de Homeodominio/metabolismo , Interacciones Huésped-Patógeno , Factores de Transcripción/metabolismo , Transcripción Genética/efectos de los fármacos , Replicación Viral , Animales , Línea Celular , Inmunoprecipitación de Cromatina , Epigénesis Genética/efectos de los fármacos , Genes Reporteros , Antígenos e de la Hepatitis B/sangre , Humanos , Inmunohistoquímica , Hígado/patología , Luciferasas/análisis , Ratones
17.
Antiviral Res ; 145: 20-23, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28709658

RESUMEN

Covalently closed circular DNA (cccDNA) in the hepatocytes nucleus is responsible for persistent infection of Hepatitis B virus (HBV). Current antiviral therapy drugs nucleos(t)ide analogs or interferon fail to eradicate HBV cccDNA. Genome editing technique provides an effective approach for HBV treatment through targeting viral cccDNA. Natronobacterium gregoryi Argonaute (NgAgo)-guide DNA (gDNA) system with powerful genome editing prompts us to explore its application in inhibiting HBV replication. Preliminary function verification indicated that NgAgo/EGFP-gDNA obviously inhibited EGFP expression. To further explore the potential role of NgAgo in restricting HBV replication, 10 of gDNAs targeting the critical region of viral genome were designed, only S-142, P-263 and P-2166 gDNAs led to significant inhibition on HBsAg, HBeAg and pregenomic RNA (pgRNA) level in Huh7 and HepG2 cells transfected with pcDNA-HBV1.1 plasmid. Similar results were also found in HBV infected HLCZ01 cells and Huh7-NTCP cells. However, we failed to detect any DNA editing in S-142, P-263 and P-2166 targeting region through T7E1 assay and Sanger sequencing. Remarkably, we found that NgAgo/P-2166 significantly accelerated the decay of viral pgRNA. Taken together, our results firstly demonstrate the potential of NgAgo/gDNA in inhibiting HBV replication through accelerating pgRNA degradation, but not DNA editing.


Asunto(s)
ADN Circular/genética , Edición Génica/métodos , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/fisiología , Natronobacterium/genética , Estabilidad del ARN , Replicación Viral/genética , Proteínas Argonautas/genética , Línea Celular , Genoma Viral , Células Hep G2 , Humanos , ARN Viral/metabolismo
18.
Oncotarget ; 7(22): 33125-35, 2016 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-27121309

RESUMEN

Over-activation of transforming growth factor-ß (TGF-ß) signaling pathway promotes cell migration and invasion in hepatocellular carcinoma (HCC). The Hepatitis B virus X protein (HBx) is involved in the enhancement of TGF-ß signaling pathway in HCC while the mechanism remains unclear. Protein phosphatase magnesium dependent 1A (PPM1a) functions as a phosphatase essential for terminating the TGF-ß signaling pathway by dephosphorylating p-Smad2/3. In this study, we found that HBx dose-dependently downregulated PPM1a protein level in the presence of TGF-ß, while having no effect on its mRNA level. Further study showed that HBx increased the ubiquitination of PPM1a and accelerated its proteasomal degradation. Restoration of PPM1a almost completely abrogated HBx mediated promotion on HCC migration and invasion. This involvement of PPM1a in HBx-related HCC was further confirmed with immunohistochemical analysis in HCC tissue. Compared with paired pericarcinous tissue, HCC tissue showed decreased PPM1a level. Besides, PPM1a level is negatively correlated with HBx expression. Taken together, our present study suggests that HBx-induced degradation of PPM1a is a novel mechanism for over-activation of TGF-ß pathway in HCC development, which might provide potential candidates for clinical diagnosis and treatment.


Asunto(s)
Carcinoma Hepatocelular/patología , Movimiento Celular/fisiología , Neoplasias Hepáticas/patología , Proteína Fosfatasa 2C/metabolismo , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo , Células Hep G2 , Humanos , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Proteína Fosfatasa 2C/genética , Transactivadores/genética , Transfección , Proteínas Reguladoras y Accesorias Virales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA