Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Int Immunol ; 31(8): 499-514, 2019 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-30809639

RESUMEN

Decreased levels of Faecalibacterium prausnitzii (F. prausnitzii), whose supernatant plays an anti-inflammatory effect, are frequently found in inflammatory bowel disease (IBD) patients. However, the anti-inflammatory products in F. prausnitzii supernatant and the mechanism have not been fully investigated. Here we found that F. prausnitzii and F. prausnitzii-derived butyrate were decreased in the intestines of IBD patients. Supplementation with F. prausnitzii supernatant and butyrate could ameliorate colitis in an animal model. Butyrate, but not other substances produced by F. prausnitzii, exerted an anti-inflammatory effect by inhibiting the differentiation of T helper 17 (Th17) cells. The mechanism underlying the anti-inflammatory effects of the butyrate produced by F. prausnitzii involved the enhancement of the acetylation-promoted degradation of c-Myc through histone deacetylase 3 (HDAC3) inhibition. In conclusion, F. prausnitzii produced butyrate to decrease Th17 differentiation and attenuate colitis through inhibiting HDAC3 and c-Myc-related metabolism in T cells. The use of F. prausnitzii may be an effective new approach to decrease the level of Th17 cells in the treatment of inflammatory diseases.


Asunto(s)
Antiinflamatorios/farmacología , Butiratos/farmacología , Diferenciación Celular/efectos de los fármacos , Faecalibacterium prausnitzii/metabolismo , Histona Desacetilasas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Células Th17/efectos de los fármacos , Animales , Antiinflamatorios/química , Antiinflamatorios/metabolismo , Butiratos/química , Butiratos/metabolismo , Colitis/inducido químicamente , Colitis/metabolismo , Colitis/patología , Faecalibacterium prausnitzii/química , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Células Th17/citología , Células Th17/metabolismo , Ácido Trinitrobencenosulfónico/administración & dosificación
2.
Cell Commun Signal ; 17(1): 23, 2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30866966

RESUMEN

BACKGROUND: Cancer cells avidly consume glucose and convert it to lactate, resulting in a low pyruvate level. This phenomenon is known as the Warburg effect, and is important for cell proliferation. Although cMyc has often been described as an oncoprotein that preferentially contributes to the Warburg effect and tumor proliferation, mechanisms of action remain unclear. Histone deacetylase 3 (HDAC3) regulates gene expression by removing acetyl groups from lysine residues, as well as has an oncogenic role in apoptosis and contributes to the proliferation of many cancer cells including cholangiocarcinoma (CCA). HDAC inhibitors display antitumor activity in many cancer cell lines. Cancer cells maintain low levels of pyruvate to prevent inhibition of HDAC but the mechanisms remain elusive. The purpose of our study was to explore the role of cMyc in regulating pyruvate metabolism, as well as to investigate whether the inhibitory effect of pyruvate on HDAC3 could hold promise in the treatment of cancer cells. METHODS: We studied pyruvate levels in CCA cell lines using metabolite analysis, and analyzed the relationship of pyruvate levels and cell proliferation with cell viability analysis. We cultivated CCA cell lines with high or low levels of pyruvate, and then analyzed the protein levels of HDAC3 and apoptotic markers via Western Blotting. We then explored the reasons of low levels of pyruvate by using seahorse analysis and 13C6 metabolites tracing analysis, and then confirmed the results using patient tissue protein samples through Western Blotting. Bioinformatics analysis and transfection assay were used to confirm the upstream target of the low levels of pyruvate status in CCA. The regulation of cMyc by HDAC3 was studied through immunoprecipitation and Western Blotting. RESULTS: We confirmed downregulated pyruvate levels in CCA, and defined that high pyruvate levels correlated with reduced cell proliferation levels. Downregulated pyruvate levels decreased the inhibition to HDAC3 and consequently protected CCA cells from apoptosis. Synergistically upregulated LDHA, PKM2 levels resulted in low levels of pyruvate, as well as poor patient survival. We also found that low levels of pyruvate contributed to proliferation of CCA cells and confirmed that the upstream target is cMyc. Conversely, high activity of HDAC3 stabilized cMyc protein by preferential deacetylating cMyc at K323 site, which further contributed to the low pyruvate levels. Finally, this creates a positive feedback loop that maintained the low levels of pyruvate and promoted CCA proliferation. CONCLUSIONS: Collectively, our findings identify a role for promoting the low pyruvate levels regulated by c-Myc, and its dynamic acetylation in cancer cell proliferation. These targets, as markers for predicting tumor proliferation in patients undergoing clinical treatments, could pave the way towards personalized therapies.


Asunto(s)
Apoptosis , Neoplasias de los Conductos Biliares/patología , Colangiocarcinoma/patología , Histona Desacetilasas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ácido Pirúvico/metabolismo , Animales , Neoplasias de los Conductos Biliares/metabolismo , Carcinogénesis , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Proliferación Celular , Colangiocarcinoma/metabolismo , Humanos , L-Lactato Deshidrogenasa/metabolismo , Proteínas de la Membrana/metabolismo , Ratones Desnudos , Hormonas Tiroideas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas de Unión a Hormona Tiroide
5.
Neoplasia ; 21(5): 429-441, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30933885

RESUMEN

Cholangiocarcinoma (CCA) is a malignant cancer with an unknown etiology and an unfavorable prognosis. Most patients are diagnosed at an advanced stage, thus making it essential to find novel curative targets for CCA. Metabolic reprogramming of the tumor cells includes metabolic abnormalities in glucose (known as the Warburg effect) and other substances such as amino acids and fats. Metabolic reprogramming produces anti-oxidant substances, reduces tumor oxidative stress, and finally promotes the proliferation of tumors. There is increasing evidence to imply that SIRT2, a histone deacetylase, and its downstream target cMYC, play metabolic regulatory roles in tumor cells. However, the role of the SIRT2/cMYC pathway in CCA is unclear. To assess the metabolic reprogramming function of the SIRT2/cMYC pathway in CCA and to determine the downstream targets as well as evaluate the therapeutic effect, the CCA RNA-Seq data were downloaded from the TCGA database. Differentially expressed genes were confirmed and KEGG pathway enrichment analysis was performed. Overall, 48 paired CCA samples were collected and subjected to immunohistochemical detection, and the clinical characteristics of participants were summarized. The CCA cells were suppressed or overexpressed with different downstream targets of SIRT2 and then subjected to apoptosis, immunoblotting, seahorse, and metabolites tracing analysis. In vivo experiments were also performed. We found that the SIRT2/cMYC pathway contributed to the proliferation of CCA cells and confirmed that the downstream target is PHDA1 and the serine synthesis pathway. The up-regulated SIRT2 and cMYC levels resulted in low levels of mitochondrial oxidative phosphorylation and increased conversion of glucose to serine and led to poor patient survival. The highly active SIRT2/cMYC pathway up-regulated the serine synthesis pathway pyruvate and increased antioxidant production, thus consequently protecting the CCA cells from oxidative stress-induced apoptosis. Our data revealed that the SIRT2/cMYC pathway plays a critical role in transforming glucose oxidative metabolism to serine anabolic metabolism, thus providing antioxidants for stress resistance. SIRT2/cMYC-induced metabolic reprogramming may represent a new therapeutic target for treating CCA.


Asunto(s)
Neoplasias de los Conductos Biliares/patología , Reprogramación Celular , Colangiocarcinoma/patología , Regulación Neoplásica de la Expresión Génica , Estrés Oxidativo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Sirtuina 2/metabolismo , Animales , Apoptosis , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/metabolismo , Movimiento Celular , Proliferación Celular , Colangiocarcinoma/genética , Colangiocarcinoma/metabolismo , Femenino , Glucosa/metabolismo , Humanos , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Mitocondrias/metabolismo , Mitocondrias/patología , Fosforilación Oxidativa , Pronóstico , Proteínas Proto-Oncogénicas c-myc/genética , Piruvato Deshidrogenasa (Lipoamida)/genética , Piruvato Deshidrogenasa (Lipoamida)/metabolismo , Serina/metabolismo , Sirtuina 2/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Cancer Med ; 8(5): 2380-2391, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30993888

RESUMEN

Cholangiocarcinoma (CCA) is an extremely invasive malignancy with late diagnosis and unfavorable prognosis. Surgery and chemotherapy are still not effective in improving outcomes in CCA patients. It is crucial to explore a novel therapeutic target for treating CCA. An NAD-dependent deacetylase also known as Sirtuin-3 (SIRT3) has been shown to regulate cellular metabolism in various cancers dynamically. However, the biological function of SIRT3 in CCA remains unclear. In this study, bioinformatics analyses were performed to identify the differentially expressed genes and pathways enriched. CCA samples were collected for immunohistochemical analysis. Three human CCA cell lines (HuCCT1, RBE, and HCCC9810) were used to explore the molecular mechanism of SIRT3 regulation of metabolic reprogramming and malignant behavior in CCA. A CCA xenograft model was then established for further validation in vivo. The data showed that SIRT3 expression was decreased and glycolysis was enhanced in CCA. Similar metabolic reprogramming was also observed in SIRT3 knockout mice. Furthermore, we demonstrated that SIRT3 could play an anti-Warburg effect by inhibiting the hypoxia-inducible factor-1α (HIF1α)/pyruvate dehydrogenase kinase 1 (PDK1)/pyruvate dehydrogenase (PDHA1) pathway in CCA cells. CCA cell proliferation and apoptosis were regulated by SIRT3-mediated metabolic reprogramming. These findings were further confirmed in CCA clinical samples and the xenograft model. Collectively, this study suggests that in the inhibition of CCA progression, SIRT3 acts through an anti-Warburg effect on the downstream pathway HIF1α/PDK1/PDHA1.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Colangiocarcinoma/etiología , Colangiocarcinoma/metabolismo , Glucosa/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Piruvato Deshidrogenasa (Lipoamida)/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/metabolismo , Sirtuina 3/metabolismo , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Colangiocarcinoma/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Glucólisis , Humanos , Inmunohistoquímica , Redes y Vías Metabólicas , Ratones , Ratones Noqueados , Piruvato Deshidrogenasa (Lipoamida)/genética , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/genética , Sirtuina 3/genética , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Oncol Rep ; 42(1): 461, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31002366

RESUMEN

Following the publication of the article, the authors have realized that Fig. 1A contained an error (essentially, the scale bars were drawn incorrectly). The corrected version of Fig. 1 is shown below, also including a modified version of the legend for Fig. 1A. Note that these revisions do not affect the overall conclusions reported in the paper. The authors apologize to the Editor of Oncology Reports and to the readership for any inconvenience caused. [the original article was published in Oncology Reports 39: 1957­1965, 2018; DOI: 10.3892/or.2018.6275].

8.
Endocrine ; 62(3): 576-587, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30117113

RESUMEN

OBJECTIVE: Many studies have confirmed the glucose-lowering effect of berberine in type 2 diabetes patients. Although the mechanism of action of berberine involves the improvement of insulin sensitivity, its hypoglycemic mechanism remains elusive. Here we show a new mechanism by which berberine antagonizes glucagon signaling and find that SIRT3 is involved in the hypoglycemic effect of berberine. METHODS: Gene knockout and overexpression were used to assess the inhibitory effect of berberine on SIRT3. Downstream signaling pathways and the hypoglycemic effect of SIRT3 were evaluated by immunoblotting and metabolic monitoring. RESULTS: We found that berberine led to mitochondrial dysfunction and AMP accumulation by inhibiting deacetylase SIRT3. We confirmed that AMP accumulation activated the AMPK signaling pathway and further promoted glucose uptake. Simultaneously, AMP accumulation reduced cyclic AMP (cAMP) levels and abrogated the phosphorylation of critical protein targets of protein kinase A (PKA). Furthermore, we found that phosphoenolpyruvate carboxykinase 1 (PEPCK1) is a key gluconeogenesis enzyme that can be stabilized by glucagon. Berberine caused significant PEPCK1 ubiquitination and degradation by antagonizing glucagon and was accompanied by high levels of PEPCK1 acetylation. Interestingly, berberine-induced glucagon inhibition is independent of AMPK activation. The in vivo data from sirt3 knockout mice were further confirmed by the in vitro experiments. CONCLUSIONS: Berberine promotes glucose uptake and inhibits gluconeogenesis by inhibiting SIRT3, and regulating mitochondria-related pathways may provide a novel approach to the development of antidiabetic drugs.


Asunto(s)
Berberina/farmacología , Gluconeogénesis/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Sirtuina 3/metabolismo , Animales , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Glucagón/metabolismo , Glucosa/metabolismo , Células HEK293 , Hepatocitos/metabolismo , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Noqueados , Consumo de Oxígeno/efectos de los fármacos , Fosforilación/efectos de los fármacos , Sirtuina 3/genética
9.
Oncol Rep ; 39(4): 1957-1965, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29484415

RESUMEN

Cholangiocarcinoma (CCA) is a highly lethal malignancy with an often late diagnosis and consequent poor prognosis. Chemotherapy is the only therapeutic strategy for most patients. Compared to normal cells, tumor cells preferentially metabolize glucose to lactate, even in aerobic conditions. Such metabolic alterations not only support the growth and invasion of tumor cells, but also promote their chemoresistance. The purpose of our study was to explore the role of metformin in regulating the metabolism of CCA, as well as to investigate whether metformin could act as a chemosensitizer of the HDAC3 inhibitor BG45, and therefore have potential for the treatment of CCA. Through bioinformatic analysis, we found that aberrant metabolism contributed to the proliferation of CCA cells. Seahorse XF96 Extracellular Flux Analyzer analysis and lactate production analysis showed that metformin could act as a suppressor of the Warburg effect in CCA cells. Western blotting showed that metformin decreased the expression of LDHA, which plays a key role in the Warburg effect. However, suppression of the Warburg effect was not sufficient to induce CCA cellular apoptosis. According to our previous research, which showed that an HDAC3 inhibitor (MI192) was involved in CCA apoptosis, we observed that metformin combined with BG45 (a novel specific HDAC3 inhibitor) effectively induced the apoptosis of CCA cells in vitro. Furthermore, in vivo experiments revealed that the combined treatment with metformin and BG45 markedly reduced CCA growth in a CCA xenograft model. Our data revealed that reversing the Warburg effect with metformin sensitizes cells to the antitumor effects of HDAC3 inhibitors. This provides a rationale for using the combination of metformin and BG45 as a new therapeutic strategy in the treatment of CCA.


Asunto(s)
Colangiocarcinoma/tratamiento farmacológico , Inhibidores Enzimáticos/administración & dosificación , Inhibidores de Histona Desacetilasas/administración & dosificación , Histona Desacetilasas/efectos de los fármacos , Metformina/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Colangiocarcinoma/genética , Colangiocarcinoma/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Histona Desacetilasas/genética , Humanos , Lactato Deshidrogenasas/genética , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Inflamm Bowel Dis ; 24(9): 1926-1940, 2018 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-29796620

RESUMEN

BACKGROUND: Inflammatory bowel disease (IBD)-associated dysbiosis is characterized by a loss of Faecalibacterium prausnitzii, whose supernatant exerts an anti-inflammatory effect. However, the anti-inflammatory substances in F. prausnitzii supernatant and the mechanism in ameliorating colitis in IBD have not yet been fully investigated. METHODS: Experimental colitis models were induced and evaluated by clinical examination and histopathology. Levels of cytokines and ratio of T cells were detected by enzyme-linked immunosorbent assay and flow cytometry analysis, respectively. F. prausnitzii supernatant was separated by macroporous resins. After extraction, the substances in supernatant were identified by gas chromatography-mass spectrometer. T-cell differentiation assay was conducted in vitro. Changes in signaling pathways were examined by immunoblot, immunohistochemistry, and immunofluorescent staining. RESULTS: We found that the supernatant of F. prausnitzii could regulate T helper 17 cell (Th17)/regulatory T cell (Treg) differentiation. Then, we identified butyrate produced by F. prausnitzii that played the anti-inflammatory effects by inhibiting interleukin (IL)-6/signal transducer and the activator of transcription 3 (STAT3)/IL-17 pathway and promoting forkhead box protein P3 (Foxp3). Finally, we demonstrated that the target of butyrate was histone deacetylase 1 (HDAC1). CONCLUSIONS: It is butyrate, instead of other substances produced by F. prausnitzii, that maintains Th17/Treg balance and exerts significant anti-inflammatory effects in colorectal colitis rodents, by inhibiting HDAC1 to promote Foxp3 and block the IL-6/STAT3/IL-17 downstream pathway. F. prausnitzii could be an option for further investigation for IBD treatment. Targeting the butyrate-HDAC1-T-cell axis offers an effective novel approach in the treatment of inflammatory disease.


Asunto(s)
Butiratos/metabolismo , Colitis/microbiología , Faecalibacterium prausnitzii/metabolismo , Histona Desacetilasa 1/metabolismo , Linfocitos T Reguladores/microbiología , Células Th17/microbiología , Animales , Diferenciación Celular , Colitis/inducido químicamente , Colon/microbiología , Modelos Animales de Enfermedad , Disbiosis/microbiología , Histona Desacetilasa 1/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Recto/microbiología , Transducción de Señal
11.
Transl Oncol ; 10(6): 917-927, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28992545

RESUMEN

Hepatocellular carcinoma (HCC) is one of the most common neoplasms, and metastasis is the most important feature for HCC-related deaths. Mounting evidence implies the dynamic regulatory role of SIRT2, a histone deacetylase, in cancer cells. Unfortunately, the role of SIRT2 and the antitumor activity of its inhibition are not known in HCC. The present study aims to evaluate the biological function of SIRT2 in HCC and identify the target of SIRT2 as well as evaluate its therapeutic efficacy. We found that SIRT2 was upregulated in HCC tissues compared to adjacent normal tissues, and this was correlated with reduced patient survival. Although CCK8 and colony-formation assays showed that SIRT2 inhibiton marginally promotes proliferation in HCC cell lines, SIRT2 knockdown decreased the invasion of HCC cells. We demonstrated that downregulation of SIRT2 could inhibit its downstream target phosphoenolpyruvate carboxykinase 1 and glutaminase, which is related to mitochondrial metabolism and the E-Cadherin pathway. These results demonstrate, for the first time that downregulation of SIRT2 decreases migration as well as invasion in human HCC cells, indicating that inhibiting SIRT2 may be an effective therapeutic strategy for treating HCC.

12.
Cell Death Dis ; 8(6): e2856, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28569784

RESUMEN

Histone deacetylase 3 (HDAC3) has an oncogenic role in apoptosis and contributes to the proliferation of cancer cells. MI192 is a novel HDAC3-specific inhibitor that displays antitumor activity in many cancer cell lines. However, the role of HDAC3 and the antitumor activity of its inhibitor MI192 are not known in cholangiocarcinoma (CCA). The present study aims to identify the target of MI192 in CCA as well as evaluate its therapeutic efficacy. CCK8 and colony formation assays showed that HDAC3 overexpression promotes proliferation in CCA cell lines. HDAC3 knockdown or treatment with MI192 decreased CCA cell growth and increased caspase-dependent apoptosis, while apoptosis was partially rescued by HDAC3 overexpression. We demonstrated that MI192 can inhibit the deacetylation activity of HDAC3 and its downstream targets in vitro, and MI192 inhibited xenograft tumor growth in vivo. Immunochemistry showed that HDAC3 was upregulated in CCA tissues compared with adjacent normal tissues, and this was correlated with reduced patient survival. Taken together, these results demonstrate for the first time that MI192 targets HDAC3 and induces apoptosis in human CCA cells. MI192 therefore shows the potential as a new drug candidate for CCA therapy.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Colangiocarcinoma/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Anciano , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Benzamidas/farmacología , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/mortalidad , Neoplasias de los Conductos Biliares/patología , Colangiocarcinoma/genética , Colangiocarcinoma/mortalidad , Colangiocarcinoma/patología , Femenino , Histona Desacetilasas/metabolismo , Humanos , Isoquinolinas/farmacología , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA