Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Adv ; 10(25): eadj8650, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38896624

RESUMEN

Pancreatic adenocarcinoma is the fourth leading cause of malignancy-related deaths, with rapid development of drug resistance driven by pancreatic cancer stem cells. However, the mechanisms sustaining stemness and chemotherapy resistance in pancreatic ductal adenocarcinoma (PDAC) remain unclear. Here, we demonstrate that Bicaudal C homolog 1 (BICC1), an RNA binding protein regulating numerous cytoplasmic mRNAs, facilitates chemoresistance and stemness in PDAC. Mechanistically, BICC1 activated tryptophan catabolism in PDAC by up-regulating indoleamine 2,3-dioxygenase-1 (IDO1) expression, a tryptophan-catabolizing enzyme. Increased levels of tryptophan metabolites contribute to NAD+ synthesis and oxidative phosphorylation, leading to a stem cell-like phenotype. Blocking BICC1/IDO1/tryptophan metabolism signaling greatly improves the gemcitabine (GEM) efficacy in several PDAC models with high BICC1 level. These findings indicate that BICC1 is a critical tryptophan metabolism regulator that drives the stemness and chemoresistance of PDAC and thus a potential target for combinatorial therapeutic strategy against chemoresistance.


Asunto(s)
Resistencia a Antineoplásicos , Células Madre Neoplásicas , Neoplasias Pancreáticas , Triptófano , Triptófano/metabolismo , Humanos , Resistencia a Antineoplásicos/genética , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/tratamiento farmacológico , Línea Celular Tumoral , Animales , Ratones , Regulación Neoplásica de la Expresión Génica , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Gemcitabina , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenasa/genética
3.
Oncogene ; 43(11): 776-788, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38243080

RESUMEN

Rapid development of drug resistance after chemotherapy is a major cause of treatment failure in individuals with pancreatic ductal adenocarcinoma (PDAC). In this study, we illustrate that tumor-derived interleukin 35 (IL-35) mediates the accelerated resistance of PDAC to gemcitabine (GEM). We observe that GEM resistance can spread from GEM-resistant PDAC cells to GEM-sensitive cells, and that IL-35 is responsible for the propagation of chemoresistance, which is supported by sequencing and experimental data. Additionally, we discover that GEM-resistant cells have significantly higher levels of IL-35 expression. Mechanistically, aberrantly expressed IL-35 triggers transcriptional activation of SOD2 expression via GP130-STAT1 signaling, scavenging reactive oxygen species (ROS) and leading to GEM resistance. Furthermore, GEM treatment stimulates IL-35 expression through activation of the NF-κB pathway, resulting in acquired chemoresistance. In the mouse model, a neutralizing antibody against IL-35 enhances the tumor suppressive effect of GEM. Collectively, our data suggests that IL-35 is critical in mediating GEM resistance in pancreatic cancer, and therefore could be a valuable therapeutic target in overcoming PDAC chemoresistance.


Asunto(s)
Adenocarcinoma , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Animales , Ratones , Gemcitabina , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/patología , Antimetabolitos Antineoplásicos/farmacología , Antimetabolitos Antineoplásicos/uso terapéutico , Resistencia a Antineoplásicos/genética , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Interleucinas/genética , Línea Celular Tumoral
5.
ACS Appl Mater Interfaces ; 15(33): 39797-39806, 2023 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-37561419

RESUMEN

Multiwavelength organic lasers have attracted considerable interest in recent years due to the cost efficiency, wide luminescence coverage, and simple processability of organics. In this work, by simply spin coating immiscible polymeric gain media in sequence, dual-wavelength (blue-green or blue-red) amplified spontaneous emission (ASE) was achieved in bilayer devices. The blue emission, water/alcohol-soluble conjugated polyelectrolyte, poly[(9,9-bis(3'-((N,N-dimethyl)-N-ethylammonium)-propyl)-2,7-fluorene)-alt-2,7-(9,9-dioctylfluorene)]dibromide (PFN-Br), was used as the bottom layer. The commercially available nonpolar solvent soluble polymer poly(9,9-dioctylfluorene-co-benzothiadiazole) (F8BT) and its blend with poly(3-hexylthiophene) (P3HT) were used as the top active layers offering green and red emission, respectively. This novel compact configuration, without interlayers between the two active layers, offers potential for developing various applications. The carefully selected top and bottom layer polymers not only meet the conditions of immiscibility and different emission wavelength range but also have a common absorption band in UV, which allows simultaneous blue-green or blue-red dual-color ASE behaviors observed in the bilayer devices under the same 390 nm laser excitation. By introducing two-dimension (2D) square distributed feedback (DFB) gratings with different periods (300 nm for blue, 330 nm for green, and 390 nm for red) as cavities, single mode blue-green (Eth = 245 µJ cm-2) and blue-red (Eth = 189 µJ cm-2) lasers were achieved by focusing the excitation laser spot on different 2D DFB gratings area. Furthermore, we found it possible to gain sufficient light confinement for red emission along its diagonal direction (Λ âˆ¼424 nm), whereas the 2D DFB gratings offer feedback for blue emission from the 300 nm period along the rectangle direction. Therefore, both blue and red lasers were eventually achieved in the same PFN-Br/F8BT:P3HT bilayer device on the single 2D DFB gratings with a period of 300 nm in this work.

6.
Signal Transduct Target Ther ; 8(1): 271, 2023 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-37443111

RESUMEN

VEGF inhibitors are one of the most successful antiangiogenic drugs in the treatment of many solid tumors. Nevertheless, pancreatic adenocarcinoma (PAAD) cells can reinstate tumor angiogenesis via activation of VEGF-independent pathways, thereby conferring resistance to VEGF inhibitors. Bioinformatic analysis showed that BICC1 was one of the top genes involved in the specific angiogenesis process of PAAD. The analysis of our own cohort confirmed that BICC1 was overexpressed in human PAAD tissues and was correlated to increased microvessel density and tumor growth, and worse prognosis. In cells and mice with xenograft tumors, BICC1 facilitated angiogenesis in pancreatic cancer in a VEGF-independent manner. Mechanistically, as an RNA binding protein, BICC1 bounds to the 3'UTR of Lipocalin-2 (LCN2) mRNA and post-transcriptionally up-regulated LCN2 expression in PAAD cells. When its level is elevated, LCN2 binds to its receptor 24p3R, which directly phosphorylates JAK2 and activates JAK2/STAT3 signal, leading to increased production of an angiogenic factor CXCL1. Blocking of the BICC1/LCN2 signalling reduced the microvessel density and tumor volume of PAAD cell grafts in mice, and increased the tumor suppressive effect of gemcitabine. In conclusion, BICC1 plays a pivotal role in the process of VEGF-independent angiogenesis in pancreatic cancer, leading to resistance to VEGF inhibitors. BICC1/LCN2 signaling may serve as a promising anti-angiogenic therapeutic target for pancreatic cancer patients.


Asunto(s)
Adenocarcinoma , Neoplasias Pancreáticas , Humanos , Animales , Ratones , Neoplasias Pancreáticas/patología , Factor A de Crecimiento Endotelial Vascular/genética , Adenocarcinoma/genética , Proteínas de Unión al ARN
7.
Cell Death Dis ; 14(5): 335, 2023 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-37217473

RESUMEN

Necroptosis is a caspase-independent form of programmed cell death. Receptor interacting protein kinase 1 (RIPK1) is a key molecule in the initiation of necroptosis and the formation of the necrotic complex. Vasculogenic mimicry (VM) provides a blood supply to tumor cells that is not dependent on endothelial cells. However, the relationship between necroptosis and VM in triple-negative breast cancer (TNBC) is not fully understood. In this study, we found that RIPK1-dependent necroptosis promoted VM formation in TNBC. Knockdown of RIPK1 significantly suppressed the number of necroptotic cells and VM formation. Moreover, RIPK1 activated the p-AKT/eIF4E signaling pathway during necroptosis in TNBC. eIF4E was blocked by knockdown of RIPK1 or AKT inhibitors. Furthermore, we found that eIF4E promoted VM formation by promoting epithelial-mesenchymal transition (EMT) and the expression and activity of MMP2. In addition to its critical role in necroptosis-mediated VM, eIF4E was essential for VM formation. Knockdown of eIF4E significantly suppressed VM formation during necroptosis. Finally, through clinical significance, the results found that eIF4E expression in TNBC was positively correlated with the mesenchymal marker vimentin, the VM marker MMP2, and the necroptosis markers MLKL and AKT. In conclusion, RIPK1-dependent necroptosis promotes VM formation in TNBC. Necroptosis promotes VM formation by activating RIPK1/p-AKT/eIF4E signaling in TNBC. eIF4E promotes EMT and MMP2 expression and activity, leading to VM formation. Our study provides a rationale for necroptosis-mediated VM and also providing a potential therapeutic target for TNBC.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Células Endoteliales/metabolismo , Necroptosis/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo
8.
Cancer Biol Med ; 19(11)2022 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-36476328

RESUMEN

OBJECTIVE: Hypoxia is an important feature of pancreatic ductal adenocarcinoma (PDAC). Previously, we found that hypoxia promotes ENO1 expression and PDAC invasion. However, the underlying molecular mechanism was remains unclear. METHODS: The relationship between ENO1 expression and clinicopathological characteristics was analyzed in 84 patients with PADC. The effects of CoCl2-induced hypoxia and ENO1 downregulation on the apoptosis, invasion, and proliferation of PDAC cells were evaluated in vitro and in vivo. Hypoxia- and ENO1-induced gene expression was analyzed by transcriptomic sequencing. RESULTS: The prognosis of PDAC with high ENO1 expression was poor (P < 0.05). High ENO1 expression was closely associated with histological differentiation and tumor invasion in 84 PDAC cases (P < 0.05). Hypoxia increased ENO1 expression in PDAC and promoted its migration and invasion. Apoptotic cells and the apoptosis marker caspase-3 in the CoCl2-treated ENO1-sh group were significantly elevated (P < 0.05). Transcriptomic sequencing indicated that CoCl2-induced PDAC cells initiated MAPK signaling. Under hypoxic conditions, PDAC cells upregulated ENO1 expression, thereby accelerating ERK phosphorylation and inhibiting apoptosis (P < 0.05). Consistent results were also observed in a PDAC-bearing mouse hindlimb ischemia model. CONCLUSIONS: Hypoxia-induced ENO1 expression promotes ERK phosphorylation and inhibits apoptosis, thus leading to PDAC survival and invasion. These results suggest that ENO1 is a potential therapeutic target for PDAC.


Asunto(s)
Apoptosis , Hipoxia , Animales , Ratones
9.
Cancer Lett ; 548: 215864, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-35981571

RESUMEN

Gemcitabine (GEM) resistance is one of the major causes of treatment failure in pancreatic ductal adenocarcinoma (PDAC) in clinic. Here, through CRISPR/Cas9 activation library screen, we found that MTA3 mediates the GEM resistance of PDAC and thus might be a potential therapeutic target for combination chemotherapy. The CRISPR library screening showed that MTA3 is the most enriched gene in the surviving GEM-treated cells, and bioinformatic and histology analysis implied its high correlation with GEM resistance. MTA3 promoted GEM resistance of PDAC cells in in vitro and in vivo experiments. Mechanistically, as a component of the Mi-2/nucleosome remodeling and deacetylase transcriptional repression complex, MTA3 transcriptionally represses CRIP2, a transcriptional repressor of NF-κB/p65, activating NF-κB signaling and consequently leading to GEM resistance. Furthermore, the treatment of GEM increases MTA3 expression in PDAC cells via activating STAT3 signaling, thereby inducing the acquired chemoresistance of PDAC to GEM. In patients derived xenografts (PDX) mouse model, Colchicine suppresses the expression of MTA3 and increases the sensitivity of tumor cells to GEM. Based on these findings, MTA3 plays a key role in GEM resistance in pancreatic cancer and is a promising therapeutic target for reversing GEM chemotherapy resistance.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Animales , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Proteínas Portadoras/genética , Línea Celular Tumoral , Colchicina , Desoxicitidina/análogos & derivados , Resistencia a Antineoplásicos/genética , Humanos , Proteínas con Dominio LIM/genética , Ratones , FN-kappa B/metabolismo , Proteínas de Neoplasias/metabolismo , Nucleosomas , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina , Neoplasias Pancreáticas
10.
Front Immunol ; 13: 1042835, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36685583

RESUMEN

Introduction: Hypoxia is an important characteristic of solid tumors. However, spatial transcriptomics (ST) of hypoxia-associated heterogeneity is not clear. Methods: This study integrated Spatial Transcriptomics (ST) with immunofluorescence to demonstrate their spatial distribution in human claudin-low breast cancer MDA-MB-231 engraft. ST spots were clustered with differentially expression genes. The data were combined with hypoxia-specific marker and angiogenesis marker-labeled serial sections to indicate the spatial distribution of hypoxia and hypoxia-inducted transcriptional profile. Moreover, marker genes, cluster-specific hypoxia genes, and their co-essential relationship were identified and mapped in every clusters. The clinicopathological association of marker genes of hypoxia-dependent spatial clusters was explored in 1904 breast cancers from METABRIC database. Results: The tumor from center to periphery were enriched into five hypoxia-dependent subgroups with differentially expressed genes, which were matched to necrosis, necrosis periphery, hypoxic tumor, adaptive survival tumor, and invasive tumor, respectively. Different subgroups demonstrated distinct hypoxia condition and spatial heterogeneity in biological behavior and signaling pathways. Cox regression analysis showed that the invasive tumor (cluster 0) and hypoxic tumor (cluster 6) score could be served as independent prognostic factors in claudin-low patients. KM analysis indicated that high invasive tumor (cluster 0) and hypoxic tumor (cluster 6) score was associated with poor prognoses of claudin-low patients. Further analysis showed that hypoxia-induced immune checkpoints, such as CD276 and NRP1, upregulation in invasive tumor to block infiltration and activation of B cells and CD8+ T cells to change tumor immune microenvironment. Discussion: This study reveals hypoxia-dependent spatial heterogeneity in claudin-low breast cancer and highlights its potential value as a predictive biomarker of clinical outcomes and immunotherapy response. The molecules found in this study also provided potential molecular mechanisms and therapeutic targets for subsequent studies.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/genética , Neoplasias de la Mama/terapia , Neoplasias de la Mama/metabolismo , Transcriptoma , Pronóstico , Hipoxia/genética , Claudinas/genética , Claudinas/metabolismo , Inmunoterapia , Microambiente Tumoral/genética , Antígenos B7/genética
11.
Front Oncol ; 12: 1028070, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36703790

RESUMEN

Introduction: Pancreatic ductal adenocarcinoma (PDAC) is characterized by high aggressiveness and a hypoxic tumour microenvironment. Macrophage migration inhibitory factor (MIF) is a hypoxia-related pleiotropic cytokine that plays important roles in cancer. However, its role in PDAC progression has not been fully elucidated. Methods: The clinical significance of MIF and hypoxia inducible factor 1 subunit alpha (HIF1A) in PDAC was analysed using immunohistochemical staining on PDAC tissues and data from KM-Plotter database. Spatial distribution of MIF and HIF1A gene expression was visualized by spatial transcriptomics in PDAC cell xenografts. To monitor the role of MIF in PDAC cell malignancy, immunostaining, lentivirus shRNA, migration assays, flow cytometry, transcriptomics and in vivo tumorigenicity were performed. Results: The spatial distribution of MIF and HIF1A was highly correlated and that high MIF expression was associated with poor prognosis of PDAC patients. MIF knockdown impaired cell invasion, with a decrease in the expression of urokinase-type plasminogen activator receptor (uPAR). Although PLAUR transcript was not reduced, a uPAR endocytic receptor, low-density lipoprotein receptor-related protein 1 (LRP1), was upregulated at both the mRNA and protein levels after MIF knockdown. The LRP1 antagonist RAP restored uPAR expression and invasiveness. MIF attenuated the nuclear translocation of p53, a transcriptional regulator of LRP1. Furthermore, MIF downregulation blunted the growth of PDAC cell xenografts and inhibited cell proliferation under normoxia and hypoxia. Transcriptome analysis also provided evidence for the role of MIF in cancer-associated pathways. Discussion: We demonstrate a novel link between the two pro-invasive agents MIF and uPAR and explain how MIF increases PDAC cell invasion capability. This finding provides a basis for therapeutic intervention of MIF in PDAC progression.

12.
BMC Cancer ; 21(1): 1305, 2021 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-34876062

RESUMEN

BACKGROUND: The expression and activation of eukaryotic translation initiation factor 4E (eIF4E) is associated with cell transformation and tumor initiation, but the functional role and the mechanism whereby it drives immune cell infiltration in breast cancer (BRCA) remain uncertain. METHODS: Oncomine, Timer and UALCAN were used to analyze the expression of eIF4E in various cancers. PrognoScan, Kaplan-Meier plotter, and GEPIA were utilized to analyze the prognostic value of eIF4E in select cancers. In vitro cell experiments were used to verify the role of eIF4E in promoting the progression of BRCA. ImmuCellAI and TIMER database were used to explore the relationship between eIF4E and tumor infiltrating immune cells. The expression of a macrophage marker (CD68+) and an M2-type marker (CD163+) was evaluated using immunohistochemistry in 50 invasive BRCA samples on tissue microarrays. The Human Protein Atlas (HPA) database was used to show the expression of eIF4E and related immune markers. LinkedOmics and NetworkAnalyst were used to build the signaling network. RESULTS: Through multiple dataset mining, we found that the expression of eIF4E in BRCA was higher than that in normal tissues, and patients with increased eIF4E expression had poorer survival and a higher cumulative recurrence rate in BRCA. At the cellular level, BRCA cell migration and invasion were significantly inhibited after eIF4E expression was inhibited by siRNA. Immune infiltration analysis showed that the eIF4E expression level was significantly associated with the tumor purity and immune infiltration levels of different immune cells in BRCA. The results from immunohistochemical (IHC) staining further proved that the expression of CD68+ and CD163+ were significantly increased and correlated with poor prognosis in BRCA patients (P < 0.05). Finally, interaction network and functional enrichment analysis revealed that eIF4E was mainly involved in tumor-related pathways, including the cell adhesion molecule pathway and the JAK-STAT signaling pathway. CONCLUSIONS: Our study has demonstrated that eIF4E expression has prognostic value for BRCA patients. eIF4E may act as an essential regulator of tumor macrophage infiltration and may participate in macrophage M2 polarization.


Asunto(s)
Neoplasias de la Mama/genética , Factor 4E Eucariótico de Iniciación/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Macrófagos/inmunología , Antígenos CD/inmunología , Antígenos de Diferenciación Mielomonocítica/inmunología , Biomarcadores de Tumor/genética , Neoplasias de la Mama/inmunología , Movimiento Celular/genética , Movimiento Celular/inmunología , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Estimación de Kaplan-Meier , Persona de Mediana Edad , Invasividad Neoplásica/genética , Pronóstico , Receptores de Superficie Celular/inmunología
13.
Front Oncol ; 11: 698302, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34804914

RESUMEN

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is a type of malignant tumor with a five-year survival rate of less than 10%. Gemcitabine (GEM) is the most commonly used drug for PDAC chemotherapy. However, a vast majority of patients with PDAC develop resistance after GEM treatment. METHODS: We screened for GEM resistance genes through bioinformatics analysis. We used immunohistochemistry to analyze 3-oxoacid CoA-transferase 1 (OXCT1) expression in PDAC tissues. The survival data were analyzed using the Kaplan-Meier curve. The expression levels of the genes related to OXCT1 and the NF-κB signaling pathway were quantified using real-time quantitative PCR and western blot analyses. We performed flow cytometry to detect the apoptosis rate. Colony formation assay was performed to measure the cell proliferation levels. The cytotoxicity assays of cells were conducted using RTCA. The downstream pathway of OXCT1 was identified via the Gene Set Enrichment Analysis. Tumor growth response to GEM in vivo was also determined in mouse models. RESULTS: Bioinformatics analysis revealed that OXCT1 is the key gene leading to GEM resistance. Patients with high OXCT1 expression exhibited short relapse-free survival under GEM treatment. OXCT1 overexpression in PDAC cell lines exerted inhibitory effect on apoptosis after GEM treatment. However, the down-regulation of OXCT1 showed the opposite effect. Blocking the NF-κB signaling pathway also reduced GEM resistance of PDAC cells. Tumor growth inhibition induced by GEM in vivo reduced after OXCT1 overexpression. Moreover, the effect of OXCT1 on GEM refractoriness in PDAC cell lines was reversed through using an NF-κB inhibitor. CONCLUSION: OXCT1 promoted GEM resistance in PDAC via the NF-κB signaling pathway both in vivo and in vitro. Our results suggest that OXCT1 could be used as a potential therapeutic target for patients with PDAC.

14.
Cancer Biol Med ; 2021 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-34086429

RESUMEN

OBJECTIVE: Hypoxia is a significant feature of solid tumors, including pancreatic ductal adenocarcinoma (PDAC). It is associated with tumor invasion, metastasis, and drug resistance. However, the spatial distribution of hypoxia-related heterogeneity in PDAC remains unclear. METHODS: Spatial transcriptomics (STs), a new technique, was used to investigate the ST features of engrafted human PDAC in the ischemic hind limbs of nude mice. Transcriptomes from ST spots in the hypoxic tumor and the control were clustered using differentially-expressed genes. These data were compared to determine the spatial organization of hypoxia-induced heterogeneity in PDAC. Clinical relevance was validated using the Tumor Cancer Genome Atlas and KM-plotter databases. The CMAP website was used to identify molecules that may serve as therapeutic targets for PDAC. RESULTS: ST showed that the tumor cell subgroups decreased to 7 subgroups in the hypoxia group, compared to 9 subgroups in the control group. Different subgroups showed positional characteristics and different gene signatures. Subgroup 6 located at the invasive front showed a higher proliferative ability under hypoxia. Subgroup 6 had active functions including cell proliferation, invasion, and response to stress. Expressions of hypoxia-related genes, LDHA, TPI1, and ENO1, induced changes. CMAP analysis indicated that ADZ-6482, a PI3K inhibitor, was targeted by the invasive subgroup in hypoxic tumors. CONCLUSIONS: This study is the first to describe hypoxic microenvironment-induced spatial transcriptome changes in PDAC, and to identify potential treatment targets for PDAC. These data will provide the basis for further investigations of the prognoses and treatments of hypoxic tumors.

15.
Cancer Lett ; 515: 86-95, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34052329

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is characterized by remarkable desmoplasia, usually driven by cancer-associated fibroblasts (CAFs), influencing patient prognosis. CAFs are a group of plastic cells responsible for tumor growth and metastasis. Fibroblasts have been reported to directly contribute to angiogenesis by undergoing mesenchymal-endothelial transition (MEndoT) after ischemic injury in the heart, brain, and hindlimbs. However, whether CAFs can undergo similar transdifferentiation in the hostile tumor microenvironment and directly contribute to tumor angiogenesis remains unclear. Herein, we provide evidence that CAFs can adopt an endothelial cell-like phenotype and directly contribute to tumor angiogenesis in vitro and in vivo. Furthermore, this program is regulated by the PERK-eIF2α-ERK1/2 axis. Pharmacological inhibition of PERK with GSK2606414 limited the phenotypic transition of CAFs. In conclusion, our results suggest that CAFs contribute to tumor angiogenesis by undergoing the MEndoT, thus representing therapeutic targets for improving PDAC prognosis.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Transición Epitelial-Mesenquimal/fisiología , Factor 2 Eucariótico de Iniciación/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Neoplasias Pancreáticas/metabolismo , eIF-2 Quinasa/metabolismo , Animales , Fibroblastos Asociados al Cáncer/patología , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Pancreáticas/patología , Pronóstico , Microambiente Tumoral/fisiología
16.
J Cell Mol Med ; 24(13): 7163-7174, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32469152

RESUMEN

The up-regulation of EMT regulator Twist1 has been implicated in vasculogenic mimicry (VM) formation in human triple-negative breast cancer (TNBC). Twist1 targets the Claudin15 promoter in hepatocellular carcinoma cells. Claudin family members are related with TNBC. However, the relationship between Claudin15 and VM formation is not clear. In this study, we first found that Claudin15 expression was frequently down-regulated in human TNBC, and Claudin15 down-regulation was significantly associated with VM and Twist1 nuclear expression. Claudin15 down-regulation correlated with shorter survival compared with high levels. Claudin15 silence significantly enhanced cell motility, invasiveness and VM formation in the non-TNBC MCF-7 cells. Conversely, an up-regulation of Claudin15 remarkably reduced TNBC MDA-MB-231 cell migration, invasion and VM formation. We also showed that down-regulation of Claudin15 was Twist1-dependent, and Twist1 repressed Claudin15 promoter activity. Furthermore, GeneChip analyses of mammary glands of Claudin15-deficient mice indicated that Claudin18 and Jun might be downstream factors of Twist1-Claudin15. Our results suggest that Twist1 induced VM through Claudin15 suppression in TNBC, and Twist1 inhibition of Claudin15 might involve Claudin18 and Jun expression.


Asunto(s)
Claudinas , Regulación Neoplásica de la Expresión Génica , Proteínas Nucleares , Neoplasias de la Mama Triple Negativas , Proteína 1 Relacionada con Twist , Animales , Femenino , Humanos , Antígenos CD/metabolismo , Cadherinas/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Núcleo Celular/metabolismo , Claudinas/deficiencia , Claudinas/genética , Claudinas/metabolismo , Estimación de Kaplan-Meier , Glándulas Mamarias Animales/patología , Invasividad Neoplásica , Proteínas Nucleares/metabolismo , Fenotipo , Transcripción Genética , Neoplasias de la Mama Triple Negativas/irrigación sanguínea , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Proteína 1 Relacionada con Twist/metabolismo , Regulación hacia Arriba/genética , Ratones
17.
Cancer Biol Med ; 16(2): 299-311, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-31516750

RESUMEN

OBJECTIVE: Vasculogenic mimicry (VM) channels that are lined by tumor cells are a functional blood supply in malignant tumors. However, the role of VM-initiating cells remains poorly understood. Cancer stem-like cells (CSCs) are positively correlated with VM. In this study, triple-negative breast cancer (TNBC) enriched with CSCs was used to investigate the relationship between VM and CSCs. METHODS: The expression of several CSC markers was detected by immunohistochemistry in 100 human breast cancer samples. The clinical significance of CSC markers and the relationship between VM, CSCs, breast cancer subtypes, and VM-associated proteins were analyzed. CD133+ and ALDH+ human and mouse TNBC cells were isolated by FACS to examine the ability of VM formation and the spatial relationship between VM and CSCs. RESULTS: CSCs were associated with TNBC subtype and VM in human invasive breast cancer. CSCs in TNBC MDA-MB-231 cells formed more VM channels and expressed more molecules promoting VM than the non-TNBC MCF-7 cells in vitro. MDA-MB-231 cells that encircled VM channels on Matrigel expressed CD133. Moreover, CSCs were located near VM channels in the 3D reconstructed blood supply system in human TNBC grafts. The CD133+ and ALDH+ cells isolated from TA2 mouse breast cancer formed more VM channels in vivo. CONCLUSIONS: CSCs line VM channels directly. Additionally, CSCs provide more VM-related molecules to synergize VM formation. The signaling pathways that control CSC differentiation may also be potential treatment targets for TNBC.

18.
Oncol Lett ; 18(3): 3003-3016, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31452779

RESUMEN

N-myc downstream regulated gene 1 (NDRG1) has been well characterized as a suppressor of metastasis in numerous types of carcinoma. NDRG1 inhibits the metastatic progression of cancer cells via its inhibitory effects on a wide variety of cellular signaling pathways. Vasculogenic mimicry (VM) refers to the unique ability of aggressive tumor cells to mimic the pattern of embryonic vasculogenic networks, and is the main reason for the poor prognosis and failure of antivascular therapy in gastric carcinoma (GC). Tumor cells can mimic the function of endothelial cells to exhibit VM through epithelial-mesenchymal transition (EMT). However, the potential function of NDRG1 in metastatic GC progression in patients has not yet been fully elucidated. To date, data regarding the function of NDRG1 in VM formation in GC have not been reported. The aim of the present study was to elucidate these unknown areas. To this end, 228 samples of human GC were used to identify the protein expression levels of NDRG1, VM-associated proteins and EMT-associated proteins via immunohistochemistry, and their clinical significance was assessed. In addition, the data of 415 patients with GC were collected from The Cancer Genome Atlas database. A functional enrichment analysis concerning NDRG1 was performed using Metascape and the Gene Set Enrichment Analysis (GSEA). In conclusion, the results of the present study indicate that NDRG1 is negatively correlated with poor prognosis through suppression of VM formation in GC. The results of the present study demonstrated that NDRG1 decreases EMT-associated protein expression and that HER2 expression may serve a significant role in this process. The Metascape and GSEA results also indirectly support this conclusion. The present study discusses the status NDRG1 as a prognostic and selective biomarker in GC, as well as current and future NDRG1-targeted therapies.

19.
Cell Death Dis ; 10(3): 200, 2019 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-30814488

RESUMEN

Angiogenesis in solid tumors is divided into two modes: endothelium-dependent vessel (EDV) and vasculogenic mimicry (VM). Sphingosine-1-phosphate receptor 1 (S1PR1) plays a vital role on EDV in a variety of human tumors. However, the relationship between S1PR1 and VM is not clear. The aim of this study is to investigate S1PR1 on the regulation of EDV and mimicry formation in breast cancer. Here we show that S1PR1 phosphorylates the complex of VE-cadherin to regulate the switch of EDV and mimicry formation. Suppression of S1PR1 impairs EDV, but contributes to the generation of VM, invasion, and metastasis in vivo and vitro. By inhibiting RhoA activation, the S1PR1/VE-cadherin signaling is blocked. S1PR1 controls VE-cadherin expression and EDV via RhoA activation. Moreover, the low expression of S1PR1 correlates with VM and poor prognosis in breast cancer patient. The results show that S1PR1 regulated RhoA activation to accelerate VE-cadherin phosphorylation (Y731), leading to increased EDV and reduced VM in breast cancer. S1PR1 may provide a new thinking direction for antiangiogenic therapy for patients with breast cancer.


Asunto(s)
Antígenos CD/metabolismo , Neoplasias de la Mama/irrigación sanguínea , Cadherinas/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/fisiología , Femenino , Células HEK293 , Xenoinjertos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Fosforilación , Transfección , Proteína de Unión al GTP rhoA/metabolismo
20.
Adv Sci (Weinh) ; 6(1): 1801455, 2019 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-30643727

RESUMEN

Electrically pumped organic lasing requires the integration of electrodes contact into the laser cavity in an organic light-emitting diode (OLED) or organic field effect transistor configuration to enable charge injection. Efficient and balanced carrier injection requires in turn alignment of the energy levels of the organic active layers with the Fermi levels of the cathode and anode. This can be achieved through chemical substitution with specific aromatic functional groups, although paying the price for a substantial (and often detrimental) change in the emission and light amplifying properties of the organic gain medium. Here, using host-guest energy transfer mixtures with hosts bearing a systematic and gradual shift in molecular orbitals is proposed, which reduces the amplified spontaneous emission (ASE) threshold of the organic gain medium significantly while leaving the peak emission unaffected. By virtue of the low guest doping required for complete host-to-guest energy transfer, the injection levels in the blends are attributed to the host whereas the gain properties solely depend on the guest. It is demonstrated that the ASE peak and thresholds of blends with different hosts do not differ while the current efficiency of OLEDs devices is deeply influenced by molecular orbital tuning of the hosts.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...