Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Nat Metab ; 4(5): 559-574, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35606596

RESUMEN

Regulatory T (Treg) cells are critical for maintaining immune homeostasis and preventing autoimmunity. Here, we show that the non-oxidative pentose phosphate pathway (PPP) regulates Treg function to prevent autoimmunity. Deletion of transketolase (TKT), an indispensable enzyme of non-oxidative PPP, in Treg cells causes a fatal autoimmune disease in mice, with impaired Treg suppressive capability despite regular Treg numbers and normal Foxp3 expression levels. Mechanistically, reduced glycolysis and enhanced oxidative stress induced by TKT deficiency triggers excessive fatty acid and amino acid catabolism, resulting in uncontrolled oxidative phosphorylation and impaired mitochondrial fitness. Reduced α-KG levels as a result of reductive TCA cycle activity leads to DNA hypermethylation, thereby limiting functional gene expression and suppressive activity of TKT-deficient Treg cells. We also find that TKT levels are frequently downregulated in Treg cells of people with autoimmune disorders. Our study identifies the non-oxidative PPP as an integrator of metabolic and epigenetic processes that control Treg function.


Asunto(s)
Autoinmunidad , Vía de Pentosa Fosfato , Linfocitos T Reguladores , Transcetolasa , Animales , Autoinmunidad/genética , Autoinmunidad/inmunología , Epigénesis Genética/genética , Epigénesis Genética/inmunología , Glucólisis , Humanos , Ratones , Vía de Pentosa Fosfato/genética , Vía de Pentosa Fosfato/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Transcetolasa/genética , Transcetolasa/inmunología
2.
Onco Targets Ther ; 14: 1673-1687, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33707952

RESUMEN

PURPOSE: Indolamine 2,3-dioxygenase (IDO) is the rate limiting enzyme of tryptophan degradation and is a negative prognostic factor in oral squamous cell carcinoma (OSCC) patients, while the underlying molecular mechanism remains unclear. This research aimed to explore the IDO expression and its biological functions in OSCC. MATERIALS AND METHODS: IDO expression was analyzed by qPCR, Western blots, and immunohistochemistry (IHC) in OSCC cell lines and tissue specimens. Tryptophan and kynurenine content were determined by UPLC-MS/MS in serum samples of OSCC patients and healthy controls. Oncomine databases and Kaplan-Meier survival analyses were used to identify the IDO expression and its correlation with OSCC prognosis. Cell counting, CCK8 assay, flow cytometry, cell cycle, and EdU incorporation assays were used to assess the effect of IDO inhibition on OSCC growth either by shRNA or the IDO-specific inhibitor (epacadostat) in vitro. An OSCC xenograft mouse model was established to verify the predicted function of IDO inhibition in vivo. Mechanistically, an 84-gene apoptosis PCR array and rescue experiment were used to characterize the underlying mechanism involved in IDO-regulated apoptosis in OSCC. RESULTS: IDO expression was upregulated in OSCC cell lines and tissues and was negatively correlated with OSCC progression. Lentivirus-mediated IDO knockdown and epacadostat significantly reduced viability and promoted apoptosis of OSCC cells in vitro and in vivo. The apoptosis PCR array identified BCL2 related protein A1 (BCL2A1) as the most obviously changed gene at the transcriptional level. IDO inhibition downregulated BCL2A1 expression, increased the expression and translocation of cytochrome c, thus promoted apoptosis in OSCC. Overexpression of BCL2A1 reversed the pro-apoptotic effect of IDO inhibition. CONCLUSION: The present results revealed that IDO directly affect the growth of OSCC cells by regulating BCL2A1 expression. IDO and the IDO-BCL2A1-cytochrome c axis may be potential therapeutic targets for OSCC.

3.
Cell Death Discov ; 7(1): 173, 2021 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-34230478

RESUMEN

Sorafenib is the FDA-approved first-line target drug for HCC patients. However, sorafenib only confers 3-5 months of survival benefit with <30% of HCC patients. Thus, it is necessary to develop a sensitizer for hepatocellular carcinoma (HCC) to sorafenib. Here, we report that in representative HCC cell lines (SMMC-7721 and PLC8024) that are insensitive to sorafenib, 3-HAA (50 µM) significantly enhances cell sensitivity to sorafenib to an extent that could not be explained by additive effects. In nude mice carrying HCC xenograft, tumor growth is inhibited by sorafenib (10 mg/kg/day) or 3-HAA (100 mg/kg/day) alone. When used in combination, the treatment effectively prevents the xenograft from growing. In a set of mechanistic experiments, we find enhanced AKT activation and increased proportion of CD44+CD133+ cells in sorafenib-resistant HCC cells and tissues. The proportion of CD44+CD133+ cells is reduced upon 3-HAA treatment in both cultured cells and mouse xenografts, suggesting that 3-HAA could decrease the stemness of HCC. We also detect decreased phosphorylation of AKT, a regulator of the GSK3ß/ß-catenin signaling upon 3-HAA treatment. The AKT activator SC79 activates GSK3 ß/ß-catenin signaling while the Wnt inhibitor XAV-939 abolishes 3-HAA inhibition of HCC growth in vitro and in mice. The current study demonstrates that 3-HAA sensitizes HCC cells to sorafenib by reducing tumor stemness, suggesting it is a promising molecule for HCC therapy.

4.
Front Cell Dev Biol ; 9: 764263, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35004674

RESUMEN

The anticancer properties of erianin have been recently discovered. However, the antitumor effect of erianin in oral squamous cell carcinoma (OSCC) remains unclear. In this study, we demonstrated that erianin can hamper OSCC cells growth both in vitro and in vivo. Erianin induced obvious G2/M arrest as well as apoptosis and gasdermin E (GSDME)-dependent pyroptosis in OSCC cells. Moreover, erianin increased autophagosome formation but decreased autolysosome function. Further study indicated that erianin significantly suppressed the expression of protein-palmitoyl thioesterase 1 (PPT1) and mTOR signaling. PPT1 has been reported to be a critical regulator of cancer progression by its modulation of autophagy and mTOR signaling. According to online databases, higher expression of PPT1 has been observed in OSCC tissues and is associated with poorer patient prognosis. As overexpression of PPT1 significantly reversed erianin-induced growth inhibition in OSCC cells, we identified the importance of PPT1 reduction in erianin-induced growth suppression. With the xenograft model, we confirmed the antitumor effect of erianin in vivo. Erianin efficiently decreased the tumor sizes, together with visibly reduced expression of PPT1 and phosphorylation of mTOR in the xenograft tumor tissues. Therefore, the present study indicated that erianin may be potentially used in OSCC therapy.

5.
Theranostics ; 11(12): 6006-6018, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33897895

RESUMEN

Objectives: Sorafenib is the only FDA-approved first-line target drug for HCC patients. However, sorafenib merely confers 3-5 months of survival benefit with less than 30% of HCC patients sensitive to sorafenib therapy. Thus, it's necessary to develop a sensitizer for hepatocellular carcinoma (HCC) to sorafenib. Methods: The principal component analysis, gene ontology, and KEGG analysis are utilized following RNA-sequencing. The mass spectrometry analysis following immunoprecipitation is performed to discover the phosphatase targets. Most importantly, both the cell line-derived xenograft (CDX) and the patient-derived xenograft (PDX) mouse model are used to determine the effect of 3-HAA on sorafenib-resistant HCC in vivo. Results: In nude mice carrying HCC xenograft, tumor growth is inhibited by sorafenib or 3-HAA alone. When used in combination, the treatment particularly prevents the xenograft from growing. Combined treatment also suppresses the growth of sorafenib-resistant (≥30mg/kg) PDXs. In a set of mechanistic experiments, we find enhanced AKT activation and decreased apoptotic cells in de novo and acquired sorafenib-resistant HCC cells and tissues. 3-HAA decreases AKT phosphorylation and increases the apoptosis of HCC in both cultured cells and mouse xenografts by upregulation of phosphatases PPP1R15A/DUSP6. PPP1R15A/PPP1α directly reduces Akt phosphorylation while DUSP6 decreases Akt activity through inhibiting PDK1. The AKT activator abolishes 3-HAA inhibition of HCC growth in vitro and in mice. Conclusion: This study demonstrates that 3-HAA sensitizes HCC cells to sorafenib by upregulation of phosphatases, suggesting it as a promising molecule for HCC therapy.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Quinurenina/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Monoéster Fosfórico Hidrolasas/metabolismo , Sorafenib/farmacología , Regulación hacia Arriba/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Hematol Oncol ; 14(1): 153, 2021 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-34563230

RESUMEN

The 3-hydroxyanthranilic acid (3-HAA), a derivative of kynurenine, was reported to suppress tumor growth. However, the function of 3-HAA largely remains unclear. Here, we report that 3-hydroxyanthranilic acid (3-HAA) is lower in tumor cells, while adding exogenous 3-HAA induces apoptosis in hepatocellular carcinoma by binding YY1. This 3-HAA binding of YY1 leads to phosphorylation of YY1 at the Thr 398 by PKCζ, concomitantly enhances YY1 chromatin binding activity to increase expression of target genes. These findings demonstrate that 3-HAA is a ligand of YY1, suggesting it is a promising therapeutic candidate for HCC.


Asunto(s)
Ácido 3-Hidroxiantranílico/farmacología , Antineoplásicos/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Quinurenina/análogos & derivados , Neoplasias Hepáticas/tratamiento farmacológico , Factor de Transcripción YY1/metabolismo , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/metabolismo , Células Hep G2 , Humanos , Quinurenina/farmacología , Ligandos , Neoplasias Hepáticas/metabolismo
7.
Artículo en Zh | WPRIM | ID: wpr-1006200

RESUMEN

@#[摘 要] 目的:探讨IL-22/IL-22受体A1(IL-22RA1)通路在口腔鳞状细胞癌(OSCC)恶性进展中的作用及其机制。方法:利用GEO数据库和免疫组化法分析IL-22RA1在OSCC组织及配对癌旁组织中的表达水平,采用组织芯片免疫组化法检测并分析IL-22RA1表达水平与OSCC患者临床病理特征的关系,通过EBI ArrayExpress数据库分析IL-22RA1表达水平与患者预后的关系。采用免疫荧光法检测IL-22和IL-22RA1在OSCC组织中表达水平并分析两者间的相关性。用RNA干扰技术敲减OSCC细胞WSU-HN4和CAL27的IL-22RA1表达,用qPCR法验证敲减效率。采用克隆形成实验、Transwell小室法分别检测IL-22对阴性对照(siNC)组和IL-22RA1敲减(siIL-22RA1)组OSCC细胞克隆形成及迁移能力的影响,WB法检测IL-22对OSCC细胞IL-22RA1表达水平及STAT1、STAT3和ERK1/2磷酸化水平的影响。结果:OSCC组织中IL-22RA1 mRNA的表达水平显著高于癌旁组织(P<0.05)。IL-22RA1表达水平与OSCC患者的肿瘤大小(P<0.05)、淋巴结转移(P<0.01)及预后不良(P<0.05)有关联。OSCC组织中的IL-22和IL-22RA1表达水平无明显关联,IL-22对OSCC细胞IL-22RA1表达无影响(均P>0.05)。IL-22可显著增强OSCC细胞的克隆形成和迁移能力(均P<0.01),并可激活OSCC细胞的STAT1、STAT3及ERK1/2信号分子;敲减OSCC细胞的IL-22RA1后,IL-22则无法发挥上述作用。结论:IL-22/IL-22RA1可通过调控细胞增殖和迁移促进OSCC的生长和转移,其下游机制可能是激活ERK1/2-STAT1/3信号通路。

8.
Theranostics ; 8(5): 1376-1388, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29507627

RESUMEN

Purpose: Elevated glucose uptake is a hallmark of cancer. Fluorodeoxyglucose (FDG) uptake was believed to indicate the aggressiveness of tumors and the standardized uptake value (SUV) is a well-known measurement for FDG uptake in positron emission tomography-computed tomography (PET/CT). However, the SUV is variable due to the heterogeneity of tumors. Methods: 126 patients with colorectal cancer underwent 18F-FDG PET/CT scanning before surgery between Jan 2011 and April 2016. Cancer-associated fibroblast (CAF) densities were calculated with the inForm Advanced image analysis software and were comparatively analyzed between patients with high and low maximum SUV (SUVmax-high and SUVmax-low). Glucose uptake was evaluated in induced and isolated CAFs and CAF-cocultured colon cancer HCT116 cells. Moreover, micro-PET/CT was performed on xenografted tumors and autoradiography was performed in the AOM/DSS induced colon cancer model. Results: CAFs were glycolytic, evidenced by glucose uptake and upregulated HK2 expression. Compared to non-activated fibroblasts (NAFs), CAFs were more dependent on glucose and sensitive to a glycolysis inhibitor. CAFs increased the SUVmax in xenograft tumors and spontaneous colon cancers. Moreover, multivariate analysis revealed that the SUVmax was only associated with tumor size among conventional parameters in colon cancer patients (126 cases, p = 0.009). Besides tumor size, the CAF density was the critical factor associated with SUVmax and outcome, which was 2.27 ± 0.74 and 1.68 ± 0.45 in the SUVmax-high and the SUVmax-low groups, respectively (p = 0.014). Conclusion: CAFs promote tumor progression and increase SUVmax of 18F-FDG, suggesting CAFs lead to the intratumor heterogeneity of the SUV and the SUVmax is a prognostic marker for cancer patients.


Asunto(s)
Fibroblastos Asociados al Cáncer/patología , Fluorodesoxiglucosa F18/farmacocinética , Tomografía Computarizada por Tomografía de Emisión de Positrones , Animales , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Glucólisis , Humanos , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Pronóstico
10.
EBioMedicine ; 17: 45-56, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28258923

RESUMEN

Tumor relapse after radiotherapy is a significant challenge to oncologists, even after recent the advances in technologies. Here, we showed that cancer-associated fibroblasts (CAFs), a major component of cancer stromal cells, promoted irradiated cancer cell recovery and tumor relapse after radiotherapy. We provided evidence that CAFs-produced IGF1/2, CXCL12 and ß-hydroxybutyrate were capable of inducing autophagy in cancer cells post-radiation and promoting cancer cell recovery from radiation-induced damage in vitro and in vivo in mice. These CAF-derived molecules increased the level of reactive oxygen species (ROS) post-radiation, which enhanced PP2A activity, repressing mTOR activation and increasing autophagy in cancer cells. Consistently, the IGF2 neutralizing antibody and the autophagy inhibitor 3-MA reduce the CAF-promoted tumor relapse in mice after radiotherapy. Taken together, our findings demonstrated that CAFs promoted irradiated cancer cell recovery and tumor regrowth post-radiation, suggesting that targeting the autophagy pathway in tumor cells may be a promising therapeutic strategy for radiotherapy sensitization.


Asunto(s)
Autofagia , Fibroblastos/efectos de la radiación , Neoplasias Hepáticas/metabolismo , Neoplasias Pulmonares/metabolismo , Ácido 3-Hidroxibutírico/farmacología , Adenina/análogos & derivados , Adenina/farmacología , Anciano , Anciano de 80 o más Años , Animales , Línea Celular Tumoral , Células Cultivadas , Quimiocina CXCL12/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/farmacología , Factor II del Crecimiento Similar a la Insulina/farmacología , Neoplasias Hepáticas/radioterapia , Neoplasias Pulmonares/radioterapia , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Proteína Fosfatasa 2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA