Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 47(3): 538-551.e5, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28930662

RESUMEN

Follicular regulatory T (Tfr) cells differentiate from conventional regulatory T (Treg) cells and suppress excessive germinal center (GC) responses by acting on both GC B cells and T follicular helper (Tfh) cells. Here, we examined the impact of mTOR, a serine/threonine protein kinase that senses and integrates diverse environmental cues, on the differentiation and functional competency of Tfr cells in response to protein immunization or viral infection. By genetically deleting Rptor or Rictor, essential components for mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2), respectively, we found that mTORC1 but not mTORC2 is essential for Tfr differentiation. Mechanistically, mTORC1-mediated phosphorylation of the transcription factor STAT3 induced the expression of the transcription factor TCF-1 by promoting STAT3 binding to the Tcf7 5'-regulatory region. Subsequently, TCF-1 bound to the Bcl6 promoter to induce Bcl6 expression, which launched the Tfr cell differentiation program. Thus, mTORC1 initiates Tfr cell differentiation by activating the TCF-1-Bcl-6 axis during immunization or infection.


Asunto(s)
Inmunomodulación , Complejos Multiproteicos/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Biomarcadores , Diferenciación Celular/inmunología , Análisis por Conglomerados , Perfilación de la Expresión Génica , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Inmunización , Inmunofenotipificación , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Transgénicos , Complejos Multiproteicos/genética , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Linfocitos T Reguladores/citología , Serina-Treonina Quinasas TOR/genética
2.
Cell Mol Life Sci ; 80(9): 242, 2023 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-37552373

RESUMEN

Radiotherapy resistance is a major obstacle to nasopharyngeal carcinoma (NPC) therapy and contributes to tumour recurrence and metastasis. Lipid metabolism is a key regulatory mechanism in cancer biology; however, its role in NPC radiotherapy resistance remains unclear. In this study, we identified hypoxia-inducible lipid droplet-associated protein (HILPDA) as a newly discovered regulator of radioresistance that induces not only lipid droplet (LD) formation but also intracellular lipid remodelling, notably changing mitochondrial cardiolipin (CL) levels. Additionally, we found that the upregulation of CL promotes mitophagy in response to irradiation exposure. Mechanistically, HILPDA inhibits PINK1-mediated CLS1 ubiquitination and degradation. The combination of a mitophagy inhibitor and irradiation significantly increases the radiosensitivity of NPC cells. Human cancer-derived data confirmed that the HILPDA-CLS1 pathway promotes NPC radioresistance. Collectively, these findings suggest that HILPDA plays a critical role in promoting NPC radioresistance and might be targeted to overcome radiotherapeutic resistance in NPC patients in the clinic.


Asunto(s)
Neoplasias Nasofaríngeas , Proteínas de Neoplasias , Humanos , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Lipidómica , Mitofagia , Carcinoma Nasofaríngeo/radioterapia , Carcinoma Nasofaríngeo/genética , Carcinoma Nasofaríngeo/patología , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/radioterapia , Neoplasias Nasofaríngeas/patología , Proteínas de Neoplasias/genética
3.
Horm Metab Res ; 55(6): 420-425, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36599457

RESUMEN

Pancreatic cancer (PC) has the lowest survival rate and the highest mortality rate among all cancers due to lack of effective treatments. The objective of the current study was to identify potential therapeutic targets in PC. Three transcriptome datasets, namely GSE62452, GSE46234, and GSE101448, were analyzed for differentially expressed genes (DEGs) between cancer and normal samples. Several bioinformatics methods, including functional analysis, pathway enrichment, hub genes, and drugs were used to screen therapeutic targets for PC. Fisher's exact test was used to analyze functional enrichments. To screen DEGs, the paired t-test was employed. The statistical significance was considered at p <0.05. Overall, 60 DEGs were detected. Functional enrichment analysis revealed enrichment of the DEGs in "multicellular organismal process", "metabolic process", "cell communication", and "enzyme regulator activity". Pathway analysis demonstrated that the DEGs were primarily related to "Glycolipid metabolism", "ECM-receptor interaction", and "pathways in cancer". Five hub genes were examined using the protein-protein interaction (PPI) network. Among these hub genes, 10 known drugs targeted to the CPA1 gene and CLPS gene were found. Overall, CPA1 and CLPS genes, as well as candidate drugs, may be useful for PC in the future.


Asunto(s)
Perfilación de la Expresión Génica , Neoplasias Pancreáticas , Humanos , Perfilación de la Expresión Génica/métodos , Biomarcadores de Tumor/genética , Detección Precoz del Cáncer/métodos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/diagnóstico , Biología Computacional/métodos , Neoplasias Pancreáticas
4.
Scand J Clin Lab Invest ; 83(2): 119-124, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36847753

RESUMEN

Omentin-1 shows a critical protective role of cardiovascular events in chronic kidney disease. This study aimed to further assess serum omentin-1 level and its relationship with clinical features and accumulating major adverse cardiac/cerebral events (MACCE) risk in end-stage renal disease patients undergoing continuous ambulatory peritoneal dialysis (CAPD-ESRD). Totally, 290 CAPD-ESRD patients and 50 healthy controls (HCs) were recruited, and their serum omentin-1 levels were measured by enzyme-linked immunosorbent assay. All CAPD-ESRD patients were followed up for 36 months to assess accumulating MACCE rate. Omentin-1 level in CAPD-ESRD patients was lower than that in HCs [median (interquartile range): 229.350 (153.575-355.550) vs. 449.800 (354.125-527.450) pg/mL] (p < 0.001). Moreover, omentin-1 level was inversely related to C-reactive protein (CRP) (p = 0.028), total cholesterol (p = 0.023), and low-density lipoprotein cholesterol (p = 0.005), while there was no correlation in omentin-1 level with other clinical features in CAPD-ESRD patients. The accumulating MACCE rate was 4.5%, 13.1%, and 15.5% in the first, second, and third years respectively, and it was lower in CAPD-ESRD patients with high level of omentin-1 than those with low level of omentin-1 (p = 0.004). Furthermore, omentin-1 (hazard ratio (HR)=0.422, p = 0.013) and high-density lipoprotein cholesterol (HR = 0.396, p = 0.010) were independently associated with reduced accumulating MACCE rate; while age (HR = 3.034, p = 0.006), peritoneal dialysis duration (HR = 2.741, p = 0.006), CRP (HR = 2.289, p = 0.026), serum uric acid (HR = 2.538, p = 0.008) were independently related to higher accumulating MACCE rate in CAPD-ESRD patients. In conclusion, serum high omentin-1 level is associated with decreased inflammation, lipid levels, and accumulating MACCE risk in CAPD-ESRD patients.


Asunto(s)
Fallo Renal Crónico , Diálisis Peritoneal Ambulatoria Continua , Humanos , Diálisis Peritoneal Ambulatoria Continua/efectos adversos , Ácido Úrico , Fallo Renal Crónico/complicaciones , Fallo Renal Crónico/terapia , Inflamación/complicaciones , Proteína C-Reactiva/análisis , Colesterol
5.
Nature ; 537(7620): 412-428, 2016 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-27501245

RESUMEN

During chronic viral infection, virus-specific CD8(+) T cells become exhausted, exhibit poor effector function and lose memory potential. However, exhausted CD8(+) T cells can still contain viral replication in chronic infections, although the mechanism of this containment is largely unknown. Here we show that a subset of exhausted CD8(+) T cells expressing the chemokine receptor CXCR5 has a critical role in the control of viral replication in mice that were chronically infected with lymphocytic choriomeningitis virus (LCMV). These CXCR5(+) CD8(+) T cells were able to migrate into B-cell follicles, expressed lower levels of inhibitory receptors and exhibited more potent cytotoxicity than the CXCR5(-) [corrected] subset. Furthermore, we identified the Id2-E2A signalling axis as an important regulator of the generation of this subset. In patients with HIV, we also identified a virus-specific CXCR5(+) CD8(+) T-cell subset, and its number was inversely correlated with viral load. The CXCR5(+) subset showed greater therapeutic potential than the CXCR5(-) [corrected] subset when adoptively transferred to chronically infected mice, and exhibited synergistic reduction of viral load when combined with anti-PD-L1 treatment. This study defines a unique subset of exhausted CD8(+) T cells that has a pivotal role in the control of viral replication during chronic viral infection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Centro Germinal/citología , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Receptores CXCR5/metabolismo , Traslado Adoptivo , Animales , Linfocitos B/inmunología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/trasplante , Diferenciación Celular , Enfermedad Crónica , Femenino , Centro Germinal/inmunología , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Humanos , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Virus de la Coriomeningitis Linfocítica/crecimiento & desarrollo , Masculino , Ratones , Receptores CXCR5/deficiencia , Transducción de Señal , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/trasplante , Carga Viral/inmunología , Replicación Viral/inmunología
6.
Int J Hosp Manag ; 92: 102684, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33052164

RESUMEN

This study investigates how US foodservice conglomerates have embarked on corporate social responsibility (CSR) measures to circumvent dire situations during the COVID-19 pandemic. It explores the evolution of CSR practices from restaurant enterprises to rescue and salvage their stakeholders. By analyzing press releases from ten restaurant chains in three different crisis phases (incubation, acceleration, and climax) through corpus linguistics, we identify a CSR progression mechanism that coevolves with the aftermath of the crisis among their stakeholders. This study improvises the CSR- as-process view to highlight the time-variant dynamic nature of CSR development over the course of major disruption.

7.
Gut ; 67(2): 307-319, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-27849558

RESUMEN

OBJECTIVE: As the modulation of autophagic processes can be therapeutically beneficial to cancer treatment, the identification of novel autophagic enhancers is highly anticipated. However, current autophagy-inducing anticancer agents exert undesired side effects owing to their non-specific biodistribution in off-target tissues. This study aims to develop a multifunctional agent to integrate cancer targeting, imaging and therapy and to investigate its mechanism. DESIGN: A series of mitochondria-targeting near-infrared (NIR) fluorophores were synthesised, screened and identified for their autophagy-enhancing activity. The optical properties and biological effects were tested both in vitro and in vivo. The underlying mechanism was investigated using inhibitors, small interfering RNA (siRNA), RNA sequencing, mass spectrometry and human samples. RESULTS: We have screened and identified a new NIR autophagy-enhancer, IR-58, which exhibits significant tumour-selective killing effects. IR-58 preferentially accumulates in the mitochondria of colorectal cancer (CRC) cells and xenografts, a process that is glycolysis-dependent and organic anion transporter polypeptide-dependent. IR-58 kills tumour cells and induces apoptosis via inducing excessive autophagy, which is mediated through the reactive oxygen species (ROS)-Akt-mammalian target of rapamycin (mTOR) pathway. RNA sequencing, mass spectrometry and siRNA interference studies demonstrate that translocase of inner mitochondrial membrane 44 (TIM44)-superoxide dismutase 2 (SOD2) pathway inhibition is responsible for the excessive ROS, autophagy and apoptosis induced by IR-58. TIM44 expression correlates positively with CRC development and poor prognosis in patients. CONCLUSIONS: A novel NIR small-molecule autophagy-enhancer, IR-58, with mitochondria-targeted imaging and therapy capabilities was developed for CRC treatment. Additionally, TIM44 was identified for the first time as a potential oncogene, which plays an important role in autophagy through the TIM44-SOD2-ROS-mTOR pathway.


Asunto(s)
Autofagia/efectos de los fármacos , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Neoplasias Colorrectales/tratamiento farmacológico , Colorantes Fluorescentes/farmacología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mitocondrias/enzimología , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Femenino , Fluorescencia , Colorantes Fluorescentes/uso terapéutico , Células HCT116 , Células HT29 , Células Hep G2 , Humanos , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Proteínas de Transporte de Membrana Mitocondrial , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Fenómenos Ópticos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/genética , Superóxido Dismutasa/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Pineal Res ; 63(1)2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28247536

RESUMEN

Cellular senescence is an important tumor-suppressive mechanism. However, acquisition of a senescence-associated secretory phenotype (SASP) in senescent cells has deleterious effects on the tissue microenvironment and, paradoxically, promotes tumor progression. In a drug screen, we identified melatonin as a novel SASP suppressor in human cells. Strikingly, melatonin blunts global SASP gene expression upon oncogene-induced senescence (OIS). Moreover, poly(ADP-ribose) polymerase-1 (PARP-1), a sensor of DNA damage, was identified as a new melatonin-dependent regulator of SASP gene induction upon OIS. Here, we report two different but potentially coherent epigenetic strategies for melatonin regulation of SASP. The interaction between the telomeric repeat-containing RNA (TERRA) and PARP-1 stimulates the SASP, which was attenuated by 67.9% (illustrated by the case of IL8) by treatment with melatonin. Through binding to macroH2A1.1, PARP-1 recruits CREB-binding protein (CBP) to mediate acetylation of H2BK120, which positively regulates the expression of target SASP genes, and this process is interrupted by melatonin. Consequently, the findings provide novel insight into melatonin's epigenetic role via modulating PARP-1 in suppression of SASP gene expression in OIS-induced senescent cells. Our studies identify melatonin as a novel anti-SASP molecule, define PARP-1 as a new target by which melatonin regulates SASP, and establish a new epigenetic paradigm for a pharmacological mechanism by which melatonin interrupts PARP-1 interaction with the telomeric long noncoding RNA(lncRNA) or chromatin.


Asunto(s)
Senescencia Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Melatonina/farmacología , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Línea Celular , Células Cultivadas , Senescencia Celular/genética , Fibroblastos/metabolismo , Humanos , Pulmón/citología , Poli(ADP-Ribosa) Polimerasa-1/genética
10.
Clin Sci (Lond) ; 128(8): 493-506, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25431838

RESUMEN

Over-nutrition induces low-grade inflammation that dampens insulin sensitivity, but the underlying molecular mediators are not fully understood. Comparative gene identification-58 (CGI-58) is an intracellular lipolytic activator. In the present study, we show that in mouse visceral fat-derived macrophages or human peripheral blood monocytes, CGI-58 negatively and interleukin (IL)-1ß positively correlate with obesity. Saturated non-esterified fatty acid (NEFA) suppresses CGI-58 expression in macrophages and this suppression activates FOXO1 (forkhead box-containing protein O subfamily-1) through inhibition of FOXO1 phosphorylation. Activated FOXO1 binds to an insulin-responsive element in IL-1ß promoter region to potentiate IL-1ß transcription. Gain- and loss-of-function studies demonstrate that NEFA-induced CGI-58 suppression activates FOXO1 to augment IL-1ß transcription by dampening insulin signalling through induction of SOCS3 (suppressor of cytokine signalling 3) expression. CGI-58 deficiency-induced SOCS3 expression is NLRP3 (nucleotide-binding oligomerization domain-like receptor family, pyrin domain containing 3) inflammasome-dependent. Our data thus identified a vicious cycle (IL-1ß-SOCS3-FOXO1-IL-1ß) that amplifies IL-1ß secretion and is initiated by CGI-58 deficiency-induced activation of the NLRP3 inflammasome in macrophages. We further show that blocking this cycle with a FOXO1 inhibitor, an antioxidant that inhibits FOXO1 or IL-1 receptor antagonist alleviates chronic inflammation and insulin resistance in high-fat diet (HFD)-fed mice. Collectively, our data suggest that obesity-associated factors such as NEFA and lipopolysaccharide (LPS) probably adopt this vicious cycle to promote inflammation and insulin resistance.


Asunto(s)
1-Acilglicerol-3-Fosfato O-Aciltransferasa/deficiencia , Factores de Transcripción Forkhead/metabolismo , Interleucina-1beta/genética , Macrófagos/metabolismo , Transducción de Señal , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Transcripción Genética , 1-Acilglicerol-3-Fosfato O-Aciltransferasa/metabolismo , Animales , Índice de Masa Corporal , Dieta Alta en Grasa , Ácidos Grasos/farmacología , Proteína Forkhead Box O1 , Humanos , Inflamasomas/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Resistencia a la Insulina , Interleucina-1beta/metabolismo , Macrófagos/efectos de los fármacos , Masculino , Ratones , Modelos Biológicos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteína 3 Supresora de la Señalización de Citocinas , Transcripción Genética/efectos de los fármacos , Aumento de Peso
11.
Carcinogenesis ; 35(7): 1661-70, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24743511

RESUMEN

Recent evidence has been suggesting the important roles of endothelial cells (ECs) involved in the pathogenesis of several cancers, including colorectal carcinomas (CRCs), but the underlying mechanism remains elusive. We have demonstrated previously that CRC-derived fibronectin extra domain A (EDA) promotes vasculogenesis, tumorigenesis and metastasis of CRCs. At the current study, we showed that EC-secreted EDA promotes the metastatic capacity CRC cells via inducing an epithelial-mesenchymal transition. In vitro and in vivo experiments showed that EC-secreted EDA, via the interaction with integrin α9ß1 on neighboring CRC cells, leads to the activation of focal adhesion kinase as well as Rac signalings, thus strengthens the polarity of cytoskeleton and promotes the invasion capacity of CRC cells. Furthermore, Erk signaling pathway was revealed to critically mediate the effect of EC-derived EDA on CRC cells. Our findings reveal a novel oncogenic role of ECs in promoting CRC malignancy through secreting EDA.


Asunto(s)
Neoplasias Colorrectales/patología , Endotelio Vascular/patología , Transición Epitelial-Mesenquimal , Fibronectinas/metabolismo , Neoplasias Hepáticas/secundario , Animales , Western Blotting , Movimiento Celular , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Endotelio Vascular/metabolismo , Fibronectinas/genética , Técnica del Anticuerpo Fluorescente , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Técnicas para Inmunoenzimas , Inmunoprecipitación , Integrinas/genética , Integrinas/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
12.
Arterioscler Thromb Vasc Biol ; 33(5): 920-5, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23471229

RESUMEN

OBJECTIVE: Controversies have arisen from recent mouse studies about the essential role of biliary sterol secretion in reverse cholesterol transport (RCT). The objective of this study was to examine the role of biliary cholesterol secretion in modulating macrophage RCT in Niemann-Pick C1-Like 1 (NPC1L1) liver only (L1(LivOnly)) mice, an animal model that is defective in both biliary sterol secretion and intestinal sterol absorption, and determine whether NPC1L1 inhibitor ezetimibe facilitates macrophage RCT by inhibiting hepatic NPC1L1. APPROACH AND RESULTS: L1(LivOnly) mice were generated by crossing NPC1L1 knockout (L1-KO) mice with transgenic mice overexpressing human NPC1L1 specifically in liver. Macrophage-to-feces RCT was assayed in L1-KO and L1(LivOnly) mice injected intraperitoneally with [(3)H]-cholesterol-labeled peritoneal macrophages isolated from C57BL/6 mice. Inhibition of biliary sterol secretion by hepatic overexpression of NPC1L1 substantially reduced transport of [(3)H]-cholesterol from primary peritoneal macrophages to the neutral sterol fraction in bile and feces in L1(LivOnly) mice without affecting tracer excretion in the bile acid fraction. Ezetimibe treatment for 2 weeks completely restored both biliary and fecal excretion of [(3)H]-tracer in the neutral sterol fraction in L1(LivOnly) mice. High-density lipoprotein kinetic studies showed that L1(LivOnly) mice compared with L1-KO mice had a significantly reduced fractional catabolic rate without altered hepatic and intestinal uptake of high-density lipoprotein-cholesterol ether. CONCLUSIONS: In mice lacking intestinal cholesterol absorption, macrophage-to-feces RCT depends on efficient biliary sterol secretion, and ezetimibe promotes macrophage RCT by inhibiting hepatic NPC1L1 function.


Asunto(s)
Anticolesterolemiantes/farmacología , Azetidinas/farmacología , Colesterol/metabolismo , Hígado/metabolismo , Macrófagos/metabolismo , Proteínas de Transporte de Membrana/fisiología , Animales , Bilis/metabolismo , Transporte Biológico/efectos de los fármacos , Ezetimiba , Humanos , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Tritio
13.
Cell Death Discov ; 10(1): 41, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38263401

RESUMEN

Liver metastasis is the major reason for most of colorectal cancer (CRC) related deaths. Accumulating evidence indicates that CRC patients with non-alcoholic fatty liver disease (NAFLD) are at a greater risk of developing liver metastasis. With the growing prevalence of NAFLD, a better understanding of the molecular mechanism in NAFLD-driven CRC liver metastasis is needed. In this study, we demonstrated that NAFLD facilitated CRC liver metastasis as a metabolic disorder and promoted the stemness of metastatic CRC cells for their colonization and outgrowth in hepatic niches. Metabolically, the lipid-rich microenvironment in NAFLD activated de novo palmitate biosynthesis in metastatic CRC cells via upregulating fatty acid synthase (FASN). Moreover, increased intracellular palmitate bioavailability promoted EGFR palmitoylation to enhance its protein stability and plasma membrane localization. Furthermore, we demonstrated that the FDA-approved FASN inhibitor orlistat could reduce NAFLD-activated endogenous palmitate production, thus inhibiting palmitoylation of EGFR to suppress CRC cell stemness and restrict liver metastasis in synergy with conventional chemotherapy. These findings reveal that the NAFLD metabolic microenvironment boosts endogenous palmitate biosynthesis in metastatic CRC cells and promotes cell stemness via EGFR palmitoylation, and FASN inhibitor orlistat could be a candidate adjuvant drug to suppress liver metastasis in CRC patients with NAFLD.

14.
J Lipid Res ; 54(8): 2109-2120, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23733885

RESUMEN

Triglyceride (TG) accumulation in hepatocytes (hepatic steatosis) preludes the development of advanced nonalcoholic fatty liver diseases (NAFLDs) such as steatohepatitis, fibrosis, and cirrhosis. Mutations in human Comparative Gene Identification-58 (CGI-58) cause cytosolic TG-rich lipid droplets to accumulate in almost all cell types including hepatocytes. However, it is unclear if CGI-58 mutation causes hepatic steatosis locally or via altering lipid metabolism in other tissues. To directly address this question, we created liver-specific CGI-58 knockout (LivKO) mice. LivKO mice on standard chow diet displayed microvesicular and macrovesicular panlobular steatosis, and progressed to advanced NAFLD stages over time, including lobular inflammation and centrilobular fibrosis. Compared with CGI-58 floxed control littermates, LivKO mice showed 8-fold and 52-fold increases in hepatic TG content, which was associated with 40% and 58% decreases in hepatic TG hydrolase activity at 16 and 42 weeks, respectively. Hepatic cholesterol also increased significantly in LivKO mice. At 42 weeks, LivKO mice showed increased hepatic oxidative stress, plasma aminotransferases, and hepatic mRNAs for genes involved in fibrosis and inflammation, such as α-smooth muscle actin, collagen type 1 α1, tumor necrosis factor α, and interleukin-1ß. In conclusion, CGI-58 deficiency in the liver directly causes not only hepatic steatosis but also steatohepatitis and fibrosis.


Asunto(s)
1-Acilglicerol-3-Fosfato O-Aciltransferasa/metabolismo , Hígado Graso/metabolismo , Cirrosis Hepática/metabolismo , Hígado/metabolismo , 1-Acilglicerol-3-Fosfato O-Aciltransferasa/deficiencia , 1-Acilglicerol-3-Fosfato O-Aciltransferasa/genética , Animales , Hígado Graso/patología , Femenino , Hígado/patología , Cirrosis Hepática/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
15.
Heliyon ; 9(6): e17550, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37416640

RESUMEN

Background: Tripterygium wilfordii (TW), when formulated in traditional Chinese medicine (TCM), can effectively treat diabetic kidney disease (DKD). However, the pharmacological mechanism associated with its success has not yet been elucidated. The current work adopted network pharmacology and molecular docking for exploring TW-related mechanisms in treating DKD. Methods: In the present work, the Traditional Chinese Medicine Systems Pharmacology (TCMSP) database was employed to obtain the effective components and candidate targets of TW. Additionally, this work utilized the UniProt protein database for screening and standardizing human-derived targets for effective components. The Cytoscape software was utilized to construct an effective component-target network for TW. Targets for DKD were acquired in the GEO, DisGeNET, GeneCards, and OMIM databases. Additionally, a Venn diagram was also plotted to select the possible targets of TW for treating DKD. Gene ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were conducted to explore the TW-related mechanism underlying DKD treatment. This work also built a protein-protein interaction (PPI) network based on the Cytoscape and String platform. Then, molecular docking was conducted in order to assess the affinity of key proteins for related compounds. Results: In total, 29 active components and 134 targets of TW were acquired, including 63 shared targets, which were identified as candidate therapeutic targets. Some key targets and important pathways were included in the effect of TW in treating DKD. Genes with higher degrees, including TNF and AKT1, were identified as hub genes of TW against DKD. Molecular docking showed that TNF and AKT1 bind well to the main components in TW (kaempferol, beta-sitosterol, triptolide, nobiletin, and stigmasterol). Conclusions: TW primarily treats DKD by acting on two targets (AKT1 and TNF) via the five active ingredients kaempferol, beta-sitosterol, triptolide, nobiletin, and stigmasterol.

16.
J Immunother Cancer ; 10(3)2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35236741

RESUMEN

BACKGROUND: The poor immunogenicity of solid tumors limits the efficacy ofanti-programmed cell death protein 1 (anti-PD1)-based immune checkpoint blockade (ICB); thus, less than 30% of patients with cancer exhibit a response. Currently, there is still a lack of effective strategies for improving tumor immunogenicity. METHODS: The antitumor effect of ultrasound-stimulated nanobubbles (USNBs) alone and in combination with an anti-PD1 antibody was evaluated in RM1 (prostate cancer), MC38 (colon cancer) and B16 (melanoma) xenograft mouse models. The phenotypes of antigen-presenting cells and CD8+ T cells were evaluated by flow cytometry. Damage-associated molecular pattern (DAMP) release, antigen release and tumor cell necrosis were assessed via western blot, flow cytometry, transmission electron microscopy and confocal microscopy. RESULTS: USNB promoted the infiltration and antitumor activity of CD8+ T cells. The combination of USNB and anti-PD1 blockade improved systemic antitumor immunity and resulted in an abscopal effect and long-term immune memory protection after complete tumor remission. Mechanistically, tumor-targeting USNB induced tumor cell necrosis through an ultrasound-mediated cavitation effect, which significantly increased DAMP release and tumor antigen presentation, consequently sensitizing tumors to ICB treatment. CONCLUSION: The administration of USNB increased tumor immunogenicity by remodeling the tumor-immune microenvironment, providing a promising strategy for sensitizing poorly immunogenic solid tumors to immunotherapy in the clinic.


Asunto(s)
Inmunoterapia , Melanoma Experimental , Animales , Células Presentadoras de Antígenos/metabolismo , Linfocitos T CD8-positivos , Humanos , Inmunoterapia/métodos , Masculino , Ratones , Microambiente Tumoral
17.
J Cell Biochem ; 112(8): 2106-14, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21465533

RESUMEN

Endostatin is a natural occurring anti-angiogenic peptide and has been shown to inhibit tumor lymphangiogenesis by suppressing the expression of tumor-stimulating growth factors. We have previously shown that fibronectin alternative extra domain A (EDA) facilitates lymphangiogenesis of colorectal tumors. Since it is known that EDA interacts with integrin α9 in the lymphatic endothelial cells (LECs), we hypothesized that endostatin may target EDA-integrin α9 pathway to inhibit colorectal tumor-induced lymphangiogenesis. To test this hypothesis, we examined the effect of endostatin on EDA secreted by SW480 colorectal cancer cells and treated human LECs with different doses of endostatin in the presence of conditional medium from SW480 cells. We found that endostatin significantly reduced EDA secretion by SW480 cells and the expression of integrin α9 in LECs. Immunofluorescence studies showed that EDA and integrin α9 colocalized on the cell membrane of LECs and these colocalizations were dramatically reduced by endostatin. Co-immunoprecipitation studies demonstrated that EDA interacted with integrin α9 in LECs, and showed that endostatin treatment inhibited the formation of EDA-integrin α9 complex in LECs. Furthermore, we found that the arrangement and polarity of LEC cytoskeletons were destroyed by endostatin substantially, leading to a reduced formation of tube-like structures of LECs and a suppressed chemotaxis of LECs toward SW480 cells. Consistently, EDA and integrin α9 expressions as well as lymphangiogenesis were significantly suppressed by endostatin in colorectal cancer xenografts. In conclusion, our results suggest that endostatin reduces colorectal tumor-induced lymphangiogenesis, at least in part, by inhibiting EDA-integrin α9 pathway.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Neoplasias Colorrectales/metabolismo , Endostatinas/farmacología , Células Endoteliales/metabolismo , Fibronectinas/biosíntesis , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Integrinas/biosíntesis , Proteínas de Neoplasias/metabolismo , Neovascularización Patológica/metabolismo , Animales , Línea Celular Tumoral , Membrana Celular/metabolismo , Neoplasias Colorrectales/tratamiento farmacológico , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Complejos Multiproteicos/metabolismo , Trasplante de Neoplasias , Neovascularización Patológica/tratamiento farmacológico , Trasplante Heterólogo
18.
Nat Commun ; 12(1): 6711, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34795238

RESUMEN

Cancer stemness represents a major source of development and progression of colorectal cancer (CRC). c-Met critically contributes to CRC stemness, but how c-Met is activated in CRC remains elusive. We previously identified the lipolytic factor ABHD5 as an important tumour suppressor gene in CRC. Here, we show that loss of ABHD5 promotes c-Met activation to sustain CRC stemness in a non-canonical manner. Mechanistically, we demonstrate that ABHD5 interacts in the cytoplasm with the core subunit of the SET1A methyltransferase complex, DPY30, thereby inhibiting the nuclear translocation of DPY30 and activity of SET1A. In the absence of ABHD5, DPY30 translocates to the nucleus and supports SET1A-mediated methylation of YAP and histone H3, which sequesters YAP in the nucleus and increases chromatin accessibility to synergistically promote YAP-induced transcription of c-Met, thus promoting the stemness of CRC cells. This study reveals a novel role of ABHD5 in regulating histone/non-histone methylation and CRC stemness.


Asunto(s)
1-Acilglicerol-3-Fosfato O-Aciltransferasa/genética , Neoplasias del Colon/genética , Regulación Neoplásica de la Expresión Génica , Células Madre Neoplásicas/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Señalizadoras YAP/genética , 1-Acilglicerol-3-Fosfato O-Aciltransferasa/metabolismo , Animales , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Células HCT116 , Humanos , Masculino , Metilación , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Proteínas Proto-Oncogénicas c-met/metabolismo , Pirazinas/farmacología , Triazinas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Proteínas Señalizadoras YAP/metabolismo
19.
Nat Commun ; 10(1): 1078, 2019 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-30842415

RESUMEN

The efficacy of Fluorouracil (FU) in the treatment of colorectal cancer (CRC) is greatly limited by drug resistance. Autophagy has been implicated in chemoresistance, but the role of selective autophagic degradation in regulating chemoresistance remains unknown. In this study, we revealed a critical role of ABHD5 in charging CRC sensitivity to FU via regulating autophagic uracil yield. We demonstrated that ABHD5 localizes to lysosome and interacts with PDIA5 to prevent PDIA5 from interacting with RNASET2 and inactivating RNASET2. ABHD5 deficiency releases PDIA5 to directly interact with RNASET2 and leave RNASET2 in an inactivate state, which impairs RNASET2-mediated autophagic uracil yield and promotes CRC cells to uptake FU as an exogenous uracil, thus increasing their sensitivity to FU. Our findings for the first time reveal a novel role of ABHD5 in regulating lysosome function, highlighting the significance of ABHD5 as a compelling biomarker predicting the sensitivity of CRCs to FU-based chemotherapy.


Asunto(s)
1-Acilglicerol-3-Fosfato O-Aciltransferasa/metabolismo , Antimetabolitos Antineoplásicos/farmacología , Autofagia , Neoplasias Colorrectales/terapia , Fluorouracilo/farmacología , 1-Acilglicerol-3-Fosfato O-Aciltransferasa/genética , Animales , Antimetabolitos Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/metabolismo , Carcinogénesis/patología , Quimioterapia Adyuvante/métodos , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Conjuntos de Datos como Asunto , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Resistencia a Antineoplásicos , Fluorouracilo/uso terapéutico , Técnicas de Silenciamiento del Gen , Células HCT116 , Humanos , Estimación de Kaplan-Meier , Lisosomas/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Ribonucleasas/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Uracilo/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA