Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Nature ; 610(7931): 356-365, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36198802

RESUMEN

Hepatocellular carcinoma (HCC), the fourth leading cause of cancer mortality worldwide, develops almost exclusively in patients with chronic liver disease and advanced fibrosis1,2. Here we interrogated functions of hepatic stellate cells (HSCs), the main source of liver fibroblasts3, during hepatocarcinogenesis. Genetic depletion, activation or inhibition of HSCs in mouse models of HCC revealed their overall tumour-promoting role. HSCs were enriched in the preneoplastic environment, where they closely interacted with hepatocytes and modulated hepatocarcinogenesis by regulating hepatocyte proliferation and death. Analyses of mouse and human HSC subpopulations by single-cell RNA sequencing together with genetic ablation of subpopulation-enriched mediators revealed dual functions of HSCs in hepatocarcinogenesis. Hepatocyte growth factor, enriched in quiescent and cytokine-producing HSCs, protected against hepatocyte death and HCC development. By contrast, type I collagen, enriched in activated myofibroblastic HSCs, promoted proliferation and tumour development through increased stiffness and TAZ activation in pretumoural hepatocytes and through activation of discoidin domain receptor 1 in established tumours. An increased HSC imbalance between cytokine-producing HSCs and myofibroblastic HSCs during liver disease progression was associated with increased HCC risk in patients. In summary, the dynamic shift in HSC subpopulations and their mediators during chronic liver disease is associated with a switch from HCC protection to HCC promotion.


Asunto(s)
Carcinogénesis , Carcinoma Hepatocelular , Células Estrelladas Hepáticas , Neoplasias Hepáticas , Animales , Carcinogénesis/patología , Carcinoma Hepatocelular/patología , Proliferación Celular , Colágeno Tipo I/metabolismo , Receptor con Dominio Discoidina 1/metabolismo , Progresión de la Enfermedad , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Factor de Crecimiento de Hepatocito/metabolismo , Hepatocitos , Humanos , Cirrosis Hepática/complicaciones , Neoplasias Hepáticas/patología , Ratones , Miofibroblastos/patología
2.
Acta Pharmacol Sin ; 42(2): 282-289, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32555508

RESUMEN

Identification of the functional impact of mutated and altered genes in cancer is critical for implementing precision oncology and drug repurposing. In recent years, the emergence of multiomics data from large, well-characterized patient cohorts has provided us with an unprecedented opportunity to address this problem. In this study, we investigated survival-associated genes across 26 cancer types and found that these genes tended to be hub genes and had higher K-core values in biological networks. Moreover, the genes associated with adverse outcomes were mainly enriched in pathways related to genetic information processing and cellular processes, while the genes with favorable outcomes were enriched in metabolism and immune regulation pathways. We proposed using the number of survival-related neighbors to assess the impact of mutations. In addition, by integrating other databases including the Human Protein Atlas and the DrugBank database, we predicted novel targets and anticancer drugs using the drug repurposing strategy. Our results illustrated the significance of multidimensional analysis of clinical data in important gene identification and drug development.


Asunto(s)
Antineoplásicos/farmacología , Reposicionamiento de Medicamentos/métodos , Neoplasias/genética , Bases de Datos Factuales , Desarrollo de Medicamentos/métodos , Humanos , Terapia Molecular Dirigida , Mutación , Neoplasias/patología , Sobrevida
3.
Hepatology ; 67(6): 2414-2429, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29091291

RESUMEN

Fibrosis and cancer represent two major complications of chronic liver disease. MicroRNAs have been implicated in the development of fibrosis and cancer, thus constituting potential therapeutic targets. Here, we investigated the role of microRNA-21 (miR-21), a microRNA that has been implicated in the development of fibrosis in multiple organs and has also been suggested to act as an "oncomir." Accordingly, miR-21 was the microRNA that showed the strongest up-regulation in activated hepatic stellate cells (HSCs) in multiple models of fibrogenesis, with an 8-fold to 24-fold induction compared to quiescent HSCs. However, miR-21 antisense inhibition did not suppress the activation of murine or human HSCs in culture or in liver slices. Moreover, genetic deletion of miR-21 in two independently generated knockout mice or miR-21 antisense inhibition did not alter HSC activation or liver fibrosis in models of toxic and biliary liver injury. Despite a strong up-regulation of miR-21 in injury-associated hepatocellular carcinoma and in cholangiocarcinoma, miR-21 deletion or antisense inhibition did not reduce the development of liver tumors. As inhibition of the most up-regulated microRNA did not affect HSC activation, liver fibrosis, or fibrosis-associated liver cancer, we additionally tested the role of microRNAs in HSCs by HSC-specific Dicer deletion. Although Dicer deletion decreased microRNA expression in HSCs and altered the expression of select genes, it only exerted negligible effects on HSC activation and liver fibrosis. CONCLUSION: Genetic and pharmacologic manipulation of miR-21 does not inhibit the development of liver fibrosis and liver cancer. Moreover, suppression of microRNA synthesis does not significantly affect HSC phenotype and activation. (Hepatology 2018;67:2414-2429).


Asunto(s)
ARN Helicasas DEAD-box/fisiología , Células Estrelladas Hepáticas/fisiología , Cirrosis Hepática/etiología , MicroARNs/fisiología , Ribonucleasa III/fisiología , Animales , Femenino , Humanos , Masculino , Ratones Noqueados
4.
J Hepatol ; 65(2): 314-24, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27154061

RESUMEN

BACKGROUND & AIMS: Considerable evidence suggests that adrenergic signaling played an essential role in tumor progression. However, its role in hepatocellular carcinoma (HCC) and the underlying mechanisms remain unknown. METHODS: The effect of adrenaline in hepatocarcinogenesis was observed in a classical diethylnitrosamine-induced HCC mouse model. Effects of ADRB2 signaling inhibition in HCC cell lines were analyzed in proliferation, apoptosis, colony formation assays. Autophagy regulation by ADRB2 was assessed in immunoblotting, immunofluorescence and immunoprecipitation assays. In vivo tumorigenic properties and anticancer effects of sorafenib were examined in nude mice. Expression levels of ADRB2 and hypoxia-inducible factor-1α (HIF1α) in 150 human HCC samples were evaluated by immunohistochemistry. RESULTS: We uncovered that adrenaline promoted DEN-induced hepatocarcinogenesis, which was reversed by the ADRB2 antagonist ICI118,551. ADRB2 signaling also played an essential role in sustaining HCC cell proliferation and survival. Notably, ADRB2 signaling negatively regulated autophagy by disrupting Beclin1/VPS34/Atg14 complex in an Akt-dependent manner, leading to HIF1α stabilization, reprogramming of HCC cells glucose metabolism, and the acquisition of resistance to sorafenib. Conversely, inhibition of ADRB2 signaling by ICI118,551, or knockdown ADRB2 expression, led to enhanced autophagy, HIF1α destabilization, tumor growth suppression, and improved anti-tumor activity of sorafenib. Consistently, ADRB2 expression correlated positively with HIF1α in HCC specimens and was associated with HCC outcomes. CONCLUSIONS: Our results uncover an important role of ADRB2 signaling in regulating HCC progression. Given the efficacy of ADRB2 modulation on HCC inhibition and sorafenib resistance, adrenoceptor antagonist appears to be a putative novel treatment for HCC and chemoresistance. LAY SUMMARY: ADRB2 signaling played an essential role in sustaining hepatocellular carcinoma cell proliferation and survival. ADRB2 signaling negatively regulated autophagy, leading to hypoxia-inducible factor-1α stabilization, reprogramming of hepatocellular carcinoma cells glucose metabolism, and the acquisition of resistance to sorafenib. Adrenoceptor antagonist appears to be a putative novel treatment for hepatocellular carcinoma and chemoresistance.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animales , Autofagia , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Humanos , Ratones , Ratones Desnudos , Niacinamida/análogos & derivados , Compuestos de Fenilurea , Receptores Adrenérgicos beta 2 , Transducción de Señal , Sorafenib
5.
Hepatology ; 61(2): 585-97, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25294684

RESUMEN

UNLABELLED: Hepatocellular carcinoma (HCC) is a prototype of inflammation-associated cancer. Oncoprotein Gankyrin, which mostly increases in HCC, plays a critical role in HCC development and metastasis. However, the exact mechanism of Gankyrin up-regulation in HCC remains unclear. A Gankyrin luciferase reporter was developed to screen a potential regulator for Gankyrin from a list of proinflammatory cytokines, and interleukin (IL)-1ß was found as one of its activators. In clinical premalignant and malignant liver disease samples, enhanced IL-1ß/interleukin-1 receptor-associated kinase 1 (IRAK-1) signaling accompanied by increased Gankyrin was observed. Lower expression of Gankyrin and phospho-IRAK-1 are favorable prognostic markers for HCC. A similar correlation was observed in the diethylnitrosamine (DEN) model of rat hepatocarcinogenesis. The results from Gankyrin reporter activity, real-time polymerase chain reaction, or immunoblotting further confirmed the up-regulation of Gankyrin by IL-1ß/IRAK-1 inflammatory signaling. Moreover, a series of Gankyrin's truncated reporters were constructed, and electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP) were performed to analyze the properties of Gankyrin promoter. Mechanistically, the core promoter of Gankyrin contains the binding site of nuclear factor Y (NF-Y) family members, which can recruit histone acetyltransferase coactivator E1A-binding protein p300 (p300) or CREB-binding protein (CBP) to promote Gankyrin transcription. Conversely, knockdown of NF-Y, p300, or CBP inhibits Gankyrin expression. IL-1ß stimulation causes sequential phosphorylation of IRAK-1, c-Jun N-terminal kinase (JNK), and p300 and enhances recruitment of the p300/CBP/NF-Y complex to Gankyrin promoter. Inhibition of phospho-JNK impairs IL-1ß/IRAK-1 signaling-mediated up-regulation of Gankyrin. CONCLUSION: The finding of IL-1ß/IRAK-1 signaling promoting Gankyrin expression through JNK and NF-Y/p300/CBP complex provides a fresh view on inflammation-enhanced hepatocarcinogenesis.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Interleucina-1beta/metabolismo , Neoplasias Hepáticas Experimentales/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Adulto , Anciano , Animales , Factor de Unión a CCAAT/metabolismo , Proteína de Unión a CREB/metabolismo , Carcinoma Hepatocelular/virología , Estudios de Casos y Controles , Bovinos , Dietilnitrosamina , Proteína p300 Asociada a E1A/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Células Hep G2 , Hepatitis B/complicaciones , Hepatitis B/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Cirrosis Hepática/complicaciones , Cirrosis Hepática/metabolismo , Neoplasias Hepáticas Experimentales/virología , Masculino , Ratones , Persona de Mediana Edad , Regiones Promotoras Genéticas , Complejo de la Endopetidasa Proteasomal/genética , Proteínas Proto-Oncogénicas/genética , Ratas Sprague-Dawley , Adulto Joven
6.
J Biol Chem ; 287(18): 14586-97, 2012 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-22418436

RESUMEN

c-Met, the tyrosine-kinase receptor for hepatocyte growth factor, plays a critical role in the tumorigenesis of hepatocellular carcinoma (HCC). However, the underlying mechanism remains incompletely understood. The mature c-Met protein p190Met(αß) (consists of a α subunit and a ß subunit) is processed from pro-Met. Here we show that pro-Met is processed into p190Met(NC) by sarco/endoplasmic reticulum calcium-ATPase (SERCA) inhibitor thapsigargin. p190Met(NC) compensates for the degradation of p190Met(αß) and protects human HCC cells from apoptosis mediated by endoplasmic reticulum (ER) stress. In comparison with p190Met(αß), p190Met(NC) is not cleaved and is expressed as a single-chain polypeptide. Thapsigargin-initiated p190Met(NC) expression depends on the disturbance of ER calcium homeostasis. Once induced, p190Met(NC) is activated independent of hepatocyte growth factor engagement. p190Met(NC) contributes to sustained high basal activation of c-Met downstream pathways during ER calcium disturbance-mediated ER stress. Both p38 MAPK-promoted glucose-regulated protein 78 (GRP78) expression and sustained high basal activation of PI3K/Akt and MEK/ERK are involved in the cytoprotective function of p190Met(NC). Importantly, the expression of p190Met(NC) is detected in some HCC cases. Taken together, these data provide a potential mechanism to explain how c-Met promotes HCC cells survival in response to ER stress. We propose that context-specific processing of c-Met protein is implicated in HCC progression in stressful microenvironments.


Asunto(s)
Carcinoma Hepatocelular/enzimología , Estrés del Retículo Endoplásmico , Homeostasis , Neoplasias Hepáticas/enzimología , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Transducción de Señal , Calcio/metabolismo , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Supervivencia Celular , Chaperón BiP del Retículo Endoplásmico , Inhibidores Enzimáticos/farmacología , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Neoplasias Hepáticas/genética , Masculino , Proteolisis/efectos de los fármacos , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Tapsigargina/farmacología , Tirosina Quinasa c-Mer
7.
Hepatology ; 55(1): 108-20, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21932402

RESUMEN

UNLABELLED: Hepatitis B virus X (HBx) protein is implicated in hepatitis B virus (HBV)-associated liver carcinogenesis. However, it remains unclear whether HBx-expressing hepatic progenitor cells (HPCs) are attributed to liver tumor formation. In this study, by using HBx transgenic mice and a 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)-induced liver injury model, the relationship between HBx expression and tumorigenicity of HPCs was analyzed. Compared with control mice, an elevated number of EpCAM(+) cells with characteristics of HPCs was observed in HBx mice after 1 month and 4 months of DDC diet feeding. All HBx transgenic mice developed liver tumors characterized by histological features of both hepatocellular carcinoma (HCC) and cholangiocarcinoma after 7 months of DDC feeding. Notably, EpCAM(+) HPCs isolated from premalignant HBx mice exposed to a DDC diet for 4 months formed subcutaneous mixed-lineage tumors (four out of six) in nonobese diabetic/severe-combined immunodeficient (NOD/SCID) mice, and none of the cells from wildtype (WT) induced tumor, indicating that HBx may induce malignant transformation of HPCs that contributes to tumorigenesis. We also found higher titers of circulating interleukin (IL)-6, activities of IL-6/STAT3, and Wnt/ß-catenin signaling pathways in HBx transgenic mice, suggesting HBx may induce intrinsic changes in HPCs by way of the above signaling that enables HPCs with tumorigenicity potential. Finally, clinical evidence showed that high HBx expression in human HBV-related HCC was statistically associated with expansion of EpCAM(+) or OV6(+) tumor cells and aggressive clinicopathologic features. CONCLUSION: HBx induces intrinsic cellular transformation promoting the expansion and tumorigenicity of HPCs in DDC-treated mice, which may be a possible origin for liver cancer induced by chronic hepatitis infection.


Asunto(s)
Carcinoma Hepatocelular/inducido químicamente , Carcinoma Hepatocelular/virología , Neoplasias Hepáticas Experimentales/inducido químicamente , Neoplasias Hepáticas Experimentales/virología , Piridinas/toxicidad , Transactivadores/genética , Animales , Neoplasias de los Conductos Biliares/inducido químicamente , Neoplasias de los Conductos Biliares/fisiopatología , Neoplasias de los Conductos Biliares/virología , Conductos Biliares Intrahepáticos , Carcinoma Hepatocelular/fisiopatología , Transformación Celular Neoplásica/inducido químicamente , Colangiocarcinoma/inducido químicamente , Colangiocarcinoma/fisiopatología , Colangiocarcinoma/virología , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Neoplasias Hepáticas Experimentales/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Persona de Mediana Edad , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Células Madre/efectos de los fármacos , Células Madre/fisiología , Células Madre/virología , Transactivadores/metabolismo , Proteínas Reguladoras y Accesorias Virales
8.
J Hepatol ; 57(3): 613-20, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22612999

RESUMEN

BACKGROUND & AIMS: Accumulating evidence suggests the involvement of tumor-initiating cells (T-ICs) in cancer genesis, but whether liver T-ICs contribute to HCC invasion and metastasis remains unclear. METHODS: OV6(+) T-ICs were isolated from SMMC7721 and HuH7 cell lines by magnetic sorting. Characteristics of T-ICs were assessed by in vitro and mouse xenograft assays. Expression of OV6 was determined by immunostaining in specimens from 218 HCC patients, and Kaplan-Meier survival analysis was used to determine the correlation of OV6 expression with prognosis. RESULTS: OV6(+) T-ICs isolated from HCC cell lines not only possess a higher capacity to form tumor spheroids in vitro, but also had a greater potential to form tumors when implanted in non-obese diabetic/severe combined immunodeficient mice, suggesting their elevated self-renewal capacity and tumorigenicity. Moreover, OV6(+) T-ICs exhibited more invasive and metastatic potentials both in vitro and in vivo. Patients with more OV6(+) tumor cells were associated with aggressive clinicopathologic features and poor prognosis. CXCR4 is expressed at higher levels in OV6(+) cells. Recombinant stromal cell-derived factor-1 (SDF-1) treatment expanded the OV6(+) HCC T-ICs population, by sustaining the stem cell property of OV6(+) cells. The SDF-1 effect was blocked by a specific CXCR4 inhibitor, AMD3100, or transfection of siRNA targeting CXCR4. CONCLUSIONS: OV6(+) HCC cells may represent a subpopulation of T-ICs with augmented invasion and metastasis potential, which contribute to progression and metastasis of HCC. The SDF-1/CXCR4 axis also provides therapeutic targets for elimination of liver T-ICs.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/secundario , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Pulmonares/metabolismo , Células Madre Neoplásicas/metabolismo , Antígeno AC133 , Animales , Antígenos CD/metabolismo , Antígenos de Neoplasias/metabolismo , Bencilaminas , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Movimiento Celular , Quimiocina CXCL12/metabolismo , Ciclamas , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Molécula de Adhesión Celular Epitelial , Femenino , Técnicas de Silenciamiento del Gen , Glicoproteínas/metabolismo , Compuestos Heterocíclicos/farmacología , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/secundario , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Análisis Multivariante , Invasividad Neoplásica , Péptidos/metabolismo , Pronóstico , ARN Interferente Pequeño , Receptores CXCR4/efectos de los fármacos , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Esferoides Celulares
9.
J Hepatol ; 57(4): 803-12, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22727732

RESUMEN

BACKGROUND & AIMS: Due to its anatomic connection, the liver is constantly exposed to gut-derived bacterial products or metabolites. Disruption of gut homeostasis is associated with many human diseases. The aim of this study was to determine the role of gut homeostasis in initiation and progression of hepatocellular carcinoma (HCC). METHODS: Disruption of intestinal homeostasis by penicillin or dextran sulfate sodium (DSS) and its restoration by probiotics were applied in a diethylnitrosamine (DEN) model of rat hepatocarcinogenesis. RESULTS: Patients with liver cirrhosis and HCC had significantly increased serum endotoxin levels. Chronic DEN treatment of rats was associated with an imbalance of subpopulations of the gut microflora including a significant suppression of Lactobacillus species, Bifidobacterium species and Enterococcus species as well as intestinal inflammation. Induction of enteric dysbacteriosis or intestinal inflammation by penicillin or DSS, respectively, significantly promoted tumor formation. Administration of probiotics dramatically mitigated enteric dysbacteriosis, ameliorated intestinal inflammation, and most importantly, decreased liver tumor growth and multiplicity. Interestingly, probiotics not only inhibited the translocation of endotoxin, which bears pathogen-associated molecular patterns (PAMPs) but also the activation of damage-associated molecular patterns (DAMPs) such as high-mobility group box 1 (HMGB1). As a result, the production of pro- and anti-inflammatory cytokines was skewed in favor of a reduced tumorigenic inflammation in the liver. CONCLUSIONS: The data highlights the importance of gut homeostasis in the pathogenesis of HCC. Modulation of the gut microbiota by probiotics may represent a new avenue for therapeutic intervention to treat or prevent HCC development.


Asunto(s)
Carcinoma Hepatocelular/patología , Endotoxinas/metabolismo , Tracto Gastrointestinal/microbiología , Homeostasis , Neoplasias Hepáticas Experimentales/patología , Probióticos/farmacología , Alquilantes/farmacología , Animales , Antibacterianos/farmacología , Bifidobacterium/efectos de los fármacos , Carcinoma Hepatocelular/sangre , Carcinoma Hepatocelular/etiología , Citocinas/biosíntesis , Sulfato de Dextran/farmacología , Dietilnitrosamina/farmacología , Dietilnitrosamina/toxicidad , Progresión de la Enfermedad , Endotoxinas/sangre , Enterococcus/efectos de los fármacos , Gastroenteritis/inducido químicamente , Gastroenteritis/tratamiento farmacológico , Gastroenteritis/metabolismo , Tracto Gastrointestinal/fisiopatología , Proteína HMGB1/metabolismo , Homeostasis/efectos de los fármacos , Humanos , Lactobacillus/efectos de los fármacos , Cirrosis Hepática/sangre , Cirrosis Hepática/complicaciones , Neoplasias Hepáticas Experimentales/inducido químicamente , Neoplasias Hepáticas Experimentales/microbiología , Masculino , Penicilinas/farmacología , Probióticos/uso terapéutico , Ratas , Ratas Sprague-Dawley
10.
Cell Rep ; 39(12): 110987, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35732131

RESUMEN

The liver is one of the most-favored distant metastatic sites for solid tumors, and interactions between cancer cells and components of the hepatic microenvironment are essential for liver metastasis (LM). Although sex is one of the determinants for primary liver cancer, sexual dimorphism in LM (SDLM) and the underlying mechanisms remain unclear. We herein demonstrate a significant male-biased SDLM, which is attributed to host androgen/androgen receptor (Ar) signaling that promotes hepatic seeding of tumor cells and subsequent outgrowth in a neutrophil-dependent manner. Mechanistically, androgen/Ar signaling promotes hepatic accumulation of neutrophils by promoting proliferation and development of neutrophil precursors in the bone marrow, as well as modulating hepatic recruitment of neutrophils and their functions. Antagonizing the androgen/Ar/neutrophil axis significantly mitigates LM in males. Our data thus reveal an important role of androgen in LM and suggest that androgen/Ar modulation represents a promising target for LM therapy in men.


Asunto(s)
Andrógenos , Neoplasias Hepáticas , Neutrófilos , Caracteres Sexuales , Humanos , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/secundario , Masculino , Neutrófilos/citología , Receptores Androgénicos , Microambiente Tumoral
11.
Hepatology ; 52(4): 1322-33, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20803560

RESUMEN

UNLABELLED: Increasing evidence suggests that the presence of endotoxemia is of substantial clinical relevance to patients with cirrhosis, but it is unclear whether and how gut-derived LPS amplifies the tumorigenic response of the liver. We found that the circulating levels of LPS were elevated in animal models of carcinogen-induced hepatocarcinogenesis. Reduction of LPS using antibiotics regimen in rats or genetic ablation of its receptor Toll-like receptor 4 (TLR4) in mice prevented excessive tumor growth and multiplicity. Additional investigation revealed that TLR4 ablation sensitizes the liver to carcinogen-induced toxicity via blocking NF-κB activation and sensitizing the liver to reactive oxygen species (ROS)-induced toxicity, but lessens inflammation-mediated compensatory proliferation. Reconstitution of TLR4-expressing myeloid cells in TLR4-deficient mice restored diethylnitrosamine (DEN)-induced hepatic inflammation and proliferation, indicating a paracrine mechanism of LPS in tumor promotion. Meanwhile, deletion of gut-derived endotoxin suppressed DEN-induced cytokine production and compensatory proliferation, whereas in vivo LPS pre-challenge promotes hepatocyte proliferation. CONCLUSION: Our data indicate that sustained LPS accumulation represents a pathological mediator of inflammation-associated hepatocellular carcinoma (HCC) and manipulation of the gut flora to prevent pathogenic bacterial translocation and endotoxin absorption may favorably influence liver function in patients with cirrhosis who are at risk of developing HCC.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinógenos/farmacología , Carcinoma Hepatocelular/etiología , Lipopolisacáridos/farmacología , Neoplasias Hepáticas/etiología , Receptor Toll-Like 4/fisiología , Animales , Carcinoma Hepatocelular/patología , Proliferación Celular/efectos de los fármacos , Dietilnitrosamina/toxicidad , Intestinos/microbiología , Lipopolisacáridos/sangre , Cirrosis Hepática/complicaciones , Masculino , Ratones , FN-kappa B/antagonistas & inhibidores , Neomicina/farmacología , Polimixina B/farmacología , Ratas , Especies Reactivas de Oxígeno/farmacología
12.
Front Immunol ; 12: 625472, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33763069

RESUMEN

Neutrophils are the largest population of circulating leukocytes and the first responder against invading pathogens or other danger signals. Sophisticated machineries help them play critical roles in immunity and inflammation, including phagocytosis, superoxide production, cytokine and chemokine production, degranulation, and formation of neutrophil extracellular traps (NETs). After maturation and release from the bone marrow, neutrophils migrate to inflamed tissues in response to many stimuli. Increasing evidences indicate that neutrophils are critically involved in the pathogenesis of liver diseases, including liver cancer, thus making them promising target for the treatment of liver diseases. Here, we would like to provide the latest finding about the role of neutrophils in liver diseases and discuss the potentiality of neutrophils as target for liver diseases.


Asunto(s)
Hepatopatías/inmunología , Hígado/inmunología , Activación Neutrófila , Infiltración Neutrófila , Neutrófilos/inmunología , Animales , Enfermedad Crónica , Citocinas/metabolismo , Humanos , Hígado/metabolismo , Hígado/patología , Hepatopatías/metabolismo , Hepatopatías/patología , Neutrófilos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
13.
Clin Transl Med ; 11(4): e379, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33931972

RESUMEN

BACKGROUND AND AIMS: 4-phenylbutyric acid (4-PBA) is a low molecular weight fatty acid that is used in clinical practice to treat inherited urea cycle disorders. In previous reports, it acted as a chemical chaperone inhibiting endoplasmic reticulum (ER) stress and unfolded protein response signaling. A few studies have suggested its function against hepatic fibrosis in mice models. However, its role in hepatocarcinogenesis remained unknown. METHODS: 4-PBA was administered alone or in combination with diethylnitrosamine to investigate its long-term effect on liver tumorigenesis. The role of 4-PBA in oncogene-induced hepatocellular carcinoma (HCC) mice model using sleeping beauty system co-expressed with hMet and ß-catenin point mutation (S45Y) was also observed. RNA-seq and PCR array were used to screen the pathways and genes involved. In vitro and in vivo studies were conducted to explore the effect of 4-PBA on liver and validate the underlying mechanism. RESULTS: 4-PBA alone didn't cause liver tumor in long term. However, it promoted liver tumorigenesis in HCC mice models via initiation of liver cancer stem cells (LCSCs) through Wnt5b-Fzd5 mediating ß-catenin signaling. Peroxisome proliferator-activated receptors (PPAR)-α induced by 4-PBA was responsible for the activation of ß-catenin signaling. Thus, intervention of PPAR-α reversed 4-PBA-induced initiation of LCSCs and HCC development in vivo. Further study revealed that 4-PBA could not only upregulate the expression of PPAR-α transcriptionally but also enhance its stabilization via protecting it from proteolysis. Moreover, high PPAR-α expression predicted poor prognosis in HCC patients. CONCLUSIONS: 4-PBA could upregulate PPAR-α to initiate LCSCs by activating ß-catenin signaling pathway, promoting HCC at early stage. Therefore, more discretion should be taken to monitor the potential tumor-promoting effect of 4-PBA under HCC-inducing environment.


Asunto(s)
Carcinoma Hepatocelular/inducido químicamente , Neoplasias Hepáticas/inducido químicamente , Células Madre Neoplásicas/efectos de los fármacos , PPAR alfa/metabolismo , Fenilbutiratos/farmacología , Animales , Carcinogénesis/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Vía de Señalización Wnt/efectos de los fármacos
14.
Cancer Cell ; 39(6): 866-882.e11, 2021 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-33930309

RESUMEN

Cancer-associated fibroblasts (CAF) are a poorly characterized cell population in the context of liver cancer. Our study investigates CAF functions in intrahepatic cholangiocarcinoma (ICC), a highly desmoplastic liver tumor. Genetic tracing, single-cell RNA sequencing, and ligand-receptor analyses uncovered hepatic stellate cells (HSC) as the main source of CAF and HSC-derived CAF as the dominant population interacting with tumor cells. In mice, CAF promotes ICC progression, as revealed by HSC-selective CAF depletion. In patients, a high panCAF signature is associated with decreased survival and increased recurrence. Single-cell RNA sequencing segregates CAF into inflammatory and growth factor-enriched (iCAF) and myofibroblastic (myCAF) subpopulations, displaying distinct ligand-receptor interactions. myCAF-expressed hyaluronan synthase 2, but not type I collagen, promotes ICC. iCAF-expressed hepatocyte growth factor enhances ICC growth via tumor-expressed MET, thus directly linking CAF to tumor cells. In summary, our data demonstrate promotion of desmoplastic ICC growth by therapeutically targetable CAF subtype-specific mediators, but not by type I collagen.


Asunto(s)
Neoplasias de los Conductos Biliares/patología , Fibroblastos Asociados al Cáncer/patología , Colangiocarcinoma/patología , Anciano , Animales , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/metabolismo , Conductos Biliares Intrahepáticos/patología , Fibroblastos Asociados al Cáncer/metabolismo , Colangiocarcinoma/genética , Colangiocarcinoma/metabolismo , Colágeno Tipo I/metabolismo , Femenino , Células Estrelladas Hepáticas/citología , Células Estrelladas Hepáticas/patología , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Hialuronano Sintasas/genética , Hialuronano Sintasas/metabolismo , Ácido Hialurónico/metabolismo , Masculino , Ratones Transgénicos , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-met/metabolismo , Microambiente Tumoral
15.
Mol Ther ; 17(12): 2049-57, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19773743

RESUMEN

Local recurrence is a therapeutic challenge for radiofrequency ablation (RFA) in treatment of small solid focal malignancies. Here we show that RFA induced heat shock proteins (HSPs) expression and high mobility group box-1 (HMGB1) translocation in xenografted melanoma, which might create a proinflammatory microenvironment that favors tumor antigen presentation and activation of the effector T cells. On this basis, we investigate whether a prime-boost strategy combining a prime with heat-shocked tumor cell lysate-pulsed dendritic cell (HT-DC) followed by an in situ boost with radiofrequency thermal ablation can prevent local tumor recurrence. The combination treatment with HT-DC and RFA showed potent antitumor effects, with >or=90% of tumor recurrence abrogated following RFA treatment. By contrast, prevaccination with unheated tumor lysate-pulsed DC had little effect on tumor relapse. Analysis of the underlying mechanism revealed that splenocytes from mice treated with HT-DC plus RFA contained significantly more tumor-specific, IFN-gamma-secreting T cells compared with control groups. Moreover, adoptive transfer of splenocytes from successfully treated tumor-free mice protected naive animals from tumor recurrence following RFA, and this was mediated mainly by CD8(+) T cells. Therefore, the optimal priming for the DC vaccination before RFA is important for boosting antigen-specific T cell responses and prevention of cancer recurrence.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Ablación por Catéter , Células Dendríticas/inmunología , Recurrencia Local de Neoplasia/prevención & control , Neoplasias Experimentales/terapia , Traslado Adoptivo , Animales , Western Blotting , Linfocitos T CD8-positivos/inmunología , Terapia Combinada , Citotoxicidad Inmunológica , Células Dendríticas/trasplante , Citometría de Flujo , Proteínas de Choque Térmico/metabolismo , Calor , Interferón gamma/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Recurrencia Local de Neoplasia/inmunología , Neoplasias Experimentales/inmunología
16.
Mol Immunol ; 45(11): 3025-35, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18471880

RESUMEN

Recognition of double-stranded RNA (dsRNA) activates interferon-regulatory factor 3 (IRF3)-dependent expression of anti-viral factors. The innate immune system recognizes viral dsRNA through two distinct pathways. First, the Toll-like receptor 3 (TLR3) detects dsRNA phagocytosed in endosomes. In addition, the helicases retinoic acid induced protein I (RIG-I)/melanoma differentiation associated gene 5 (MDA5) binds cytoplasmic dsRNA generated during viral replication. Both RIG-I/MDA5 and TLR3 can bind polyriboinosinic:polyribocytidylic acid (poly(I:C)), the synthetic analog of viral dsRNA, and mediate type I IFN production. Here we show that signal regulatory protein (SIRP) alpha negatively regulates both TLR3- and RIG-1/MDA5-dependent anti-viral pathways. Suppression of SIRPalpha expression by RNA interference results in enhanced activation of IRF3 and MAPK pathways after poly(I:C) treatment, coupled with the up-regulation of IFN-beta and IFN-beta-inducible gene transcriptional activation. The requirement of phosphoinositide 3-kinase (PI3K) activity for the induction of IFN-beta and IFN-beta-inducible genes by dsRNA is supported by the observation that a PI3K inhibitor failed to activate IFN-beta and IFN-beta-inducible gene expression. PI3K, whose activity is essential for activation of IRF3, is recruited to the phosphorylated tyrosine residues of SIRPalpha upon poly(I:C) stimulation, which lead to a reduction in the activity of the downstream kinase AKT. Thus SIRPalpha may accomplish its inhibitory function in type I IFN induction, in part, through its association and sequestration of the signal transducer PI3K.


Asunto(s)
Antígenos de Diferenciación/inmunología , Citoplasma/metabolismo , Interferón beta/inmunología , Receptores Inmunológicos/inmunología , Transducción de Señal , Receptor Toll-Like 3/inmunología , Secuencias de Aminoácidos , Animales , Antígenos de Diferenciación/química , Línea Celular , Citoplasma/efectos de los fármacos , Humanos , Factor 3 Regulador del Interferón/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Poli I-C/farmacología , ARN Bicatenario/farmacología , Receptores Inmunológicos/química , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 3/genética , Activación Transcripcional/efectos de los fármacos
17.
NPJ Precis Oncol ; 2(1): 6, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29872724

RESUMEN

Hepatocellular carcinoma (HCC) has become a leading cause of cancer-related death, making the elucidation of its underlying mechanisms an urgent priority. Inflammation is an adaptive response to infection and tissue injury under strict regulations. When the host regulatory machine runs out of control, nonresolving inflammation occurs. Nonresolving inflammation is a recognized hallmark of cancer that substantially contributes to the development and progression of HCC. The HCC-associated inflammation can be initiated and propagated by extrinsic pathways through activation of pattern-recognition receptors (PRRs) by pathogen-associated molecule patterns (PAMPs) derived from gut microflora or damage-associated molecule patterns (DAMPs) released from dying liver cells. The inflammation can also be orchestrated by the tumor itself through secreting factors that recruit inflammatory cells to the tumor favoring the buildup of a microenvironment. Accumulating datas from human and mouse models showed that inflammation promotes HCC development by promoting proliferative and survival signaling, inducing angiogenesis, evading immune surveillance, supporting cancer stem cells, activating invasion and metastasis as well as inducing genomic instability. Targeting inflammation may represent a promising avenue for the HCC treatment. Some inhibitors targeting inflammatory pathways have been developed and under different stages of clinical trials, and one (sorafenib) have been approved by FDA. However, as most of the data were obtained from animal models, and there is a big difference between human HCC and mouse HCC models, it is challenging on successful translation from bench to bedside.

18.
Nat Commun ; 9(1): 191, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29335551

RESUMEN

The communication between tumor-derived elements and stroma in the metastatic niche has a critical role in facilitating cancer metastasis. Yet, the mechanisms tumor cells use to control metastatic niche formation are not fully understood. Here we report that in the lung metastatic niche, high-metastatic hepatocellular carcinoma (HCC) cells exhibit a greater capacity to convert normal fibroblasts to cancer-associated fibroblasts (CAFs) than low-metastatic HCC cells. We show high-metastatic HCC cells secrete exosomal miR-1247-3p that directly targets B4GALT3, leading to activation of ß1-integrin-NF-κB signaling in fibroblasts. Activated CAFs further promote cancer progression by secreting pro-inflammatory cytokines, including IL-6 and IL-8. Clinical data show high serum exosomal miR-1247-3p levels correlate with lung metastasis in HCC patients. These results demonstrate intercellular crosstalk between tumor cells and fibroblasts is mediated by tumor-derived exosomes that control lung metastasis of HCC, providing potential targets for prevention and treatment of cancer metastasis.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Carcinoma Hepatocelular/metabolismo , Exosomas/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/metabolismo , Neoplasias Pulmonares/metabolismo , MicroARNs/genética , N-Acetil-Lactosamina Sintasa/genética , Animales , Fibroblastos Asociados al Cáncer/patología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/secundario , Comunicación Celular , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Exosomas/química , Humanos , Integrina beta1/genética , Integrina beta1/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundario , Masculino , Ratones , Ratones Desnudos , MicroARNs/metabolismo , N-Acetil-Lactosamina Sintasa/metabolismo , Invasividad Neoplásica , Trasplante de Neoplasias , Células Neoplásicas Circulantes/metabolismo , Células Neoplásicas Circulantes/patología , Transducción de Señal
19.
Nat Rev Gastroenterol Hepatol ; 14(9): 527-539, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28676707

RESUMEN

Hepatocellular carcinoma (HCC) is the third leading cause of worldwide cancer mortality. HCC almost exclusively develops in patients with chronic liver disease, driven by a vicious cycle of liver injury, inflammation and regeneration that typically spans decades. Increasing evidence points towards a key role of the bacterial microbiome in promoting the progression of liver disease and the development of HCC. Here, we will review mechanisms by which the gut microbiota promotes hepatocarcinogenesis, focusing on the leaky gut, bacterial dysbiosis, microbe-associated molecular patterns and bacterial metabolites as key pathways that drive cancer-promoting liver inflammation, fibrosis and genotoxicity. On the basis of accumulating evidence from preclinical studies, we propose the intestinal-microbiota-liver axis as a promising target for the simultaneous prevention of chronic liver disease progression and HCC development in patients with advanced liver disease. We will review in detail therapeutic modalities and discuss clinical settings in which targeting the gut-microbiota-liver axis for the prevention of disease progression and HCC development seems promising.


Asunto(s)
Carcinoma Hepatocelular/microbiología , Microbioma Gastrointestinal , Neoplasias Hepáticas/microbiología , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/prevención & control , Humanos , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/prevención & control
20.
Annu Rev Pathol ; 12: 153-186, 2017 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-27959632

RESUMEN

Liver cancer is the second leading cause of cancer mortality worldwide, causing more than 700,000 deaths annually. Because of the wide landscape of genomic alterations and limited therapeutic success of targeting tumor cells, a recent focus has been on better understanding and possibly targeting the microenvironment in which liver tumors develop. A unique feature of liver cancer is its close association with liver fibrosis. More than 80% of hepatocellular carcinomas (HCCs) develop in fibrotic or cirrhotic livers, suggesting an important role of liver fibrosis in the premalignant environment (PME) of the liver. Cholangiocarcinoma (CCA), in contrast, is characterized by a strong desmoplasia that typically occurs in response to the tumor, suggesting a key role of cancer-associated fibroblasts (CAFs) and fibrosis in its tumor microenvironment (TME). Here, we discuss the functional contributions of myofibroblasts, CAFs, and fibrosis to the development of HCC and CCA in the hepatic PME and TME, focusing on myofibroblast- and extracellular matrix-associated growth factors, fibrosis-associated immunosuppressive pathways, as well as mechanosensitive signaling cascades that are activated by increased tissue stiffness. Better understanding of the role of myofibroblasts in HCC and CCA development and progression may provide the basis to target these cells for tumor prevention or therapy.


Asunto(s)
Fibroblastos Asociados al Cáncer/patología , Cirrosis Hepática/patología , Neoplasias Hepáticas/patología , Miofibroblastos/patología , Microambiente Tumoral , Animales , Fibroblastos Asociados al Cáncer/metabolismo , Humanos , Cirrosis Hepática/metabolismo , Neoplasias Hepáticas/metabolismo , Miofibroblastos/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA