Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
J Proteome Res ; 23(1): 277-288, 2024 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-38085828

RESUMEN

Given the pressing clinical problem of making a decision in diagnosis for subjects with pulmonary nodules, we aimed to discover novel plasma protein biomarkers for lung adenocarcinoma (LUAD) and benign pulmonary nodules (BPNs) and then develop an integrative multianalytical model to guide the clinical management of LUAD and BPN patients. Through label-free quantitative plasma proteomic analysis (data are available via ProteomeXchange with identifier PXD046731), 12 differentially expressed proteins (DEPs) in LUAD and BPN were screened. The diagnostic abilities of DEPs were validated in two independent validation cohorts. The results showed that the levels of three candidate proteins (PRDX2, PON1, and APOC3) were lower in the plasma of LUAD than in BPN. The three candidate proteins were combined with three promising computed tomography indicators (spiculation, vascular notch sign, and lobulation) and three traditional markers (CEA, CA125, and CYFRA21-1) to construct an integrative multianalytical model, which was effective in distinguishing LUAD from BPN, with an AUC of 0.904, a sensitivity of 81.44%, and a specificity of 90.14%. Moreover, the model possessed impressive diagnostic performance between early LUADs and BPNs, with the AUC, sensitivity, specificity, and accuracy of 0.868, 65.63%, 90.14%, and 82.52%, respectively. This model may be a useful auxiliary diagnostic tool for LUAD and BPN by achieving a better balance of sensitivity and specificity.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , Nódulos Pulmonares Múltiples , Humanos , Neoplasias Pulmonares/patología , Proteómica , Adenocarcinoma del Pulmón/diagnóstico , Nódulos Pulmonares Múltiples/diagnóstico , Nódulos Pulmonares Múltiples/patología , Biomarcadores , Proteínas Sanguíneas , Biomarcadores de Tumor , Arildialquilfosfatasa
2.
Br J Cancer ; 130(11): 1770-1782, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38600327

RESUMEN

BACKGROUND: Tumour-associated macrophages (TAMs) are an important component of the tumour microenvironment (TME). However, the crosstalk between oesophageal squamous cell carcinoma (ESCC) cells and TAMs remains largely unexplored. METHODS: Clinical samples and the TCGA database were used to evaluate the relevance of SPP1 and TAM infiltration in ESCC. Mouse models were constructed to investigate the roles of macrophages educated by SPP1 in ESCC. Macrophage phenotypes were determined using qRT‒PCR and immunohistochemical staining. RNA sequencing was performed to elucidate the mechanism. RESULTS: Increasing expression of SPP1 correlated with M2-like TAM accumulation in ESCC, and they both predicted poor prognosis in the ESCC cohort. Knockdown of SPP1 significantly inhibited the infiltration of M2 TAMs in xenograft tumours. In vivo mouse model experiments showed that SPP1-mediated education of macrophages plays an essential role in the progression of ESCC. Mechanistically, SPP1 recruited macrophages and promoted M2 polarisation via CD44/PI3K/AKT signalling activation and then induced VEGFA and IL6 secretion to sustain ESCC progression. Finally, blockade of SPP1 with RNA aptamer significantly inhibited tumour growth and M2 TAM infiltration in xenograft mouse models. CONCLUSIONS: This study highlights SPP1-mediated crosstalk between ESCC cells and TAMs in ESCC. SPP1 could serve as a potential target in ESCC therapy.


Asunto(s)
Progresión de la Enfermedad , Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Osteopontina , Microambiente Tumoral , Macrófagos Asociados a Tumores , Humanos , Animales , Carcinoma de Células Escamosas de Esófago/patología , Carcinoma de Células Escamosas de Esófago/inmunología , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/metabolismo , Ratones , Neoplasias Esofágicas/patología , Neoplasias Esofágicas/inmunología , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/metabolismo , Microambiente Tumoral/inmunología , Osteopontina/genética , Osteopontina/metabolismo , Línea Celular Tumoral , Macrófagos/metabolismo , Macrófagos/inmunología , Femenino , Ensayos Antitumor por Modelo de Xenoinjerto , Masculino , Pronóstico , Transducción de Señal , Fosfatidilinositol 3-Quinasas/metabolismo , Ratones Desnudos , Receptores de Hialuranos/metabolismo , Receptores de Hialuranos/genética
3.
BMC Cancer ; 23(1): 52, 2023 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-36647029

RESUMEN

BACKGROUND: TGF-ß-induced factor homeobox 2 (TGIF2) is a transcription regulator that is phosphorylated by EGFR/ERK signaling. However, the functions of phosphorylated (p)-TGIF2 in cancer are largely unknown. Here, we investigated the roles of p-TGIF2 in promoting epithelial-mesenchymal transition (EMT) and metastasis in lung adenocarcinoma (LUAD). METHODS: In vitro and in vivo experiments were conducted to investigate the role of TGIF2 in LUAD EMT and metastasis. Dual-luciferase reporter and ChIP assays were employed to observe the direct transcriptional regulation of E-cadherin by TGIF2 and HDAC1. Co-immunoprecipitation was performed to identify the interaction between TGIF2 and HDAC1. RESULTS: Downregulating the expression of TGIF2 inhibited LUAD cell migration, EMT and metastasis in vitro and in vivo. Phosphorylation of TGIF2 by EGFR/ERK signaling was required for TGIF2-promoted LUAD EMT and metastasis since phosphorylation-deficient TGIF2 mutant lost these functions. Phosphorylation of TGIF2 was necessary to recruit HDAC1 to the E-cadherin promoter sequence and subsequently suppress E-cadherin transcription. Meanwhile, inhibition of HDAC1 repressed the TGIF2 phosphorylation-induced migration and EMT of LUAD cells. In xenograft mouse models, both inhibition of ERK and HDAC1 could significantly inhibited TGIF2-enhanced metastasis. Furthermore, TGIF2-positive staining was significantly correlated with E-cadherin-negative staining in human lung cancer specimens. CONCLUSIONS: Our study reveals the novel function of p-TGIF2 in promoting EMT and metastasis in LUAD; p-TGIF2 could be a potential therapeutic target to inhibit LUAD metastasis.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , Humanos , Animales , Ratones , Transición Epitelial-Mesenquimal/genética , Fosforilación , Adenocarcinoma del Pulmón/genética , Neoplasias Pulmonares/patología , Cadherinas/genética , Cadherinas/metabolismo , Receptores ErbB/metabolismo , Línea Celular Tumoral , Movimiento Celular , Regulación Neoplásica de la Expresión Génica , Proteínas Represoras/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo
4.
BMC Cancer ; 22(1): 932, 2022 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-36038839

RESUMEN

BACKGROUND: Esophageal squamous cell carcinoma (ESCC) has poor prognosis mainly due to lacking of effective diagnostic biomarkers. Aberrant expression of secreted phosphoprotein 1 (SPP1) protein has been observed in several cancers. The purpose of this study is to assess the feasibility of serum autoantibody to SPP1 in detection of ESCC. METHODS: The SPP1 protein levels in 108 ESCC tissues and 72 adjacent normal tissues were analyzed by immunohistochemistry. Discovery group containing 62 serum samples from ESCC patients and 62 serum samples from normal controls (NC) were used to detect the levels of anti-SPP1 autoantibody by enzyme-linked immunosorbent assay (ELISA). Validation group containing another 100 ESCC and 100 NC serum samples were tested to confirm the levels of autoantibody to SPP1. Western blotting was performed to further confirm the results of ELISA. RESULTS: SPP1 protein was significantly overexpressed in ESCC tissues compared to adjacent normal tissues. ELISA results showed that serum autoantibody to SPP1 was significantly increased in ESCC compared to NC in both discovery and validation groups. Autoantibody to SPP1 could discriminate patients with ESCC from NC with the area under curve (AUC) values of 0.653 and 0.739 in discovery and validation group, respectively. The results of ELISA and the occurrence of immunoreactivity to SPP1 in ESCC sera were confirmed by western blotting. CONCLUSION: Our study indicated the potential significance of anti-SPP1 autoantibody as a novel biomarker for detection of ESCC.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Autoanticuerpos , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/patología , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/diagnóstico , Humanos , Osteopontina , Pronóstico
5.
Bioorg Chem ; 109: 104717, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33647744

RESUMEN

Efforts toward finding potent CDK4 inhibitor for cancer therapy, a series of fluorine substituted pyrrolo[2,3-d]pyrimidine derivatives were designed, synthesized, and evaluated. Among them, the optimal lead compound 18i was discovered with potent activity against CDK4 at the nanomolar level (IC50 = 2.5 nM) and exquisite selectivity which demonstrated only modest activity against 3 out of the 394 protein kinases. 18i exhibited a much greater in vitro antiproliferative activity against several human cancer cell lines than that of the approved drug ribociclib. Further mechanism studies revealed that 18i effectively stimulated cancer cell cycle arrest in G1 phase and induced tumor cell apoptosis. In the comparison of in vivo therapeutic effects in xenograft mouse models of breast cancer, oral administration of 18i showed a significantly better degree of inhibitory effect to ribociclib without obvious toxicity. All of the results indicated that 18i could be a promising CDK4 inhibitor for treating malignancies.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/uso terapéutico , Supervivencia Celular/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Diseño de Fármacos , Animales , Antineoplásicos/química , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Desnudos , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Filogenia , Distribución Aleatoria , Transducción de Señal
6.
Br J Cancer ; 123(5): 730-741, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32528119

RESUMEN

BACKGROUND: The anticancer potential of ibuprofen has created a broad interest to explore the clinical benefits of ibuprofen in cancer therapy. However, the current understanding of the molecular mechanisms involved in the anticancer potential of ibuprofen remains limited. METHODS: Cancer stemness assays to validate ibuprofen function in vitro and in vivo. Histone modification assays to check the effect of ibuprofen on histone acetylation/methylation, as well as the activity of HDAC and KDM6A/B. Inhibitors' in vivo assays to evaluate therapeutic effects of various inhibitors' combination manners. RESULTS: In our in vitro studies, we report that ibuprofen diminishes cancer cell stemness properties that include reducing the ALDH + subpopulation, side population and sphere formation in three cancer types. In our in vivo studies, we report that ibuprofen decreases tumour growth, metastasis and prolongs survival. In addition, our results showed that ibuprofen inhibits inflammation-related stemness gene expression (especially ICAM3) identified by a high-throughput siRNA platform. In regard to the underlying molecular mechanism of action, we report that ibuprofen reduces HDACs and histone demethylase (KDM6A/B) expression that mediates histone acetylation and methylation, and suppresses gene expression via a COX2-dependent way. In regard to therapeutic strategies, we report that ibuprofen combined HDAC/HDM inhibitors prevents cancer progression in vivo. CONCLUSIONS: The aforementioned findings suggest a molecular model that explains how ibuprofen diminishes cancer cell stemness properties. These may provide novel targets for therapeutic strategies involving ibuprofen in the prevention of cancer progression.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Histonas/metabolismo , Ibuprofeno/farmacología , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Células A549 , Acetilación/efectos de los fármacos , Animales , Línea Celular Tumoral , Resistencia a Antineoplásicos , Femenino , Células Hep G2 , Histona Desacetilasas/metabolismo , Humanos , Molécula 3 de Adhesión Intercelular/metabolismo , Metilación/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Metástasis de la Neoplasia , Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Distribución Aleatoria
7.
Biochim Biophys Acta Mol Basis Dis ; 1864(8): 2566-2578, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29729315

RESUMEN

ICAM3 was reported to promote metastasis in tumors. However, the underlying mechanism remains elusive. Here, we disclosed that the expression of ICAM3 was closely correlated with the TNM stage of human breast and lung cancer, as well as the dominant overexpression in high aggressive tumor cell lines (231 and A549 cells). Moreover, the knockdown of ICAM3 inhibited tumor metastasis whereas the ectopic expression of ICAM3 promoted tumor metastasis both in vitro and in vivo. In addition, exploration of the underlying mechanism demonstrated that ICAM3 not only binds to LFA-1 with its extracellular domain and structure protein ERM but also to lamellipodia with its intracellular domain which causes a tension that pulls cells apart (metastasis). Furthermore, ICAM3 extracellular or intracellular mutants alternatively abolished ICAM3 mediated tumor metastasis in vitro and in vivo. As a therapy strategy, LFA-1 antibody or Lifitegrast restrained tumor metastasis via targeting ICAM3-LFA-1 interaction. In summary, the aforementioned findings suggest a model of ICAM3 in mediating tumor metastasis. This may provide a promising target or strategy for the prevention of tumor metastasis.


Asunto(s)
Antígenos CD/metabolismo , Neoplasias de la Mama/metabolismo , Moléculas de Adhesión Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Neoplasias Pulmonares/metabolismo , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Proteínas de Neoplasias/metabolismo , Factores de Transcripción/metabolismo , Células A549 , Animales , Antígenos CD/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Moléculas de Adhesión Celular/genética , Proteínas de Unión al ADN/genética , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Antígeno-1 Asociado a Función de Linfocito/genética , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Factores de Transcripción/genética
8.
J Biol Inorg Chem ; 23(6): 939-947, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30014256

RESUMEN

Arsenic trioxide (As2O3) induces cell apoptosis and reduces the invasive and metastatic activities in various cancer types. However, the role of As2O3 in ovarian cancer angiogenesis remains unclear. In this study, we investigated the role of As2O3 in ovarian cancer angiogenesis and found that a low concentration of As2O3 causes no effects on epithelial ovarian cancer cell viability or apoptosis. Moreover, we found that As2O3-treated epithelial ovarian cancer cells demonstrate a reduced tube formation of endothelial cells in Matrigel. In addition, As2O3-treated epithelial ovarian cancer cells show a decreased VEGFA, VEGFR2 and CD31 mRNA expression. As per the underlying mechanisms involved in As2O3 treatment, we found that As2O3 inhibits VEGFA and VEGFR2 expression that thereby inhibits the VEGFA-VEGFR2-PI3K/ERK signaling pathway. This leads to a suppression in both VEGFA synthesis and angiogenesis-related gene expression. A decreased VEGFA synthesis and secretion also inhibits the VEGFA-VEGFR2-PI3K/ERK signaling pathway in human umbilical vein endothelial cells (HUVECs). In summary, our results may provide strategies for the use of As2O3 in the prevention of tumor angiogenesis.


Asunto(s)
Apoptosis , Trióxido de Arsénico/farmacología , Carcinoma Epitelial de Ovario/irrigación sanguínea , Neovascularización Patológica/prevención & control , Neoplasias Ováricas/irrigación sanguínea , Trióxido de Arsénico/administración & dosificación , Carcinoma Epitelial de Ovario/metabolismo , Carcinoma Epitelial de Ovario/patología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , ARN Mensajero/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
9.
Cell Oncol (Dordr) ; 46(5): 1269-1283, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37067748

RESUMEN

PURPOSE: Previous studies have shown that TBX21 (T-Box Transcription Factor 21) plays a vital role in coordinating multiple aspects of the immune response especially type 1 immune response as well as tumor progression. However, the function of TBX21 in colorectal cancer (CRC) remains unclear. METHODS: IHC to investigate TBX21 expression in CRC tissues. Cell proliferation and apoptosis assays to validate TBX21 function in vitro and in vivo. RNA-seq assay to explore target genes of TBX21. Human phospho-kinase array assay to explore down-stream signaling of TBX21. RESULTS: We disclosed that the expression of TBX21 was marked decreased in CRC versus normal tissue, and negatively correlated with CRC TNM stages. Surprisingly, we found that the CRC and normal cell lines show no TBX21 expression levels. Ectopic expression of TBX21 inhibited cell proliferation and promoted cell apoptosis in vitro. Moreover, RNA-sequence data first time showed that ARHGAP29 acts as the target gene of TBX21 to mediate down-stream signaling activation. Human phospho-kinase array data first time displayed that ectopic expression of TBX21 reduced kinase RSK and GSK3ß activation. In contrast, knocked down the expression of TBX21 or ARHGAP29 alternatively abolished TBX21 mediated cell proliferation suppression, cell apoptosis enhancement and RSK/GSK3ß activation. In addition, xenograft model studies demonstrated that TBX21 inhibits colorectal tumor progression via ARHGAP29/ RSK/ GSK3ß signaling in vivo. CONCLUSIONS: In summary, the aforementioned findings suggest a model of TBX21 in suppressing CRC progression. This may provide a promising target for CRC therapy.


Asunto(s)
Neoplasias Colorrectales , Humanos , Apoptosis/genética , Secuencia de Bases , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Neoplasias Colorrectales/metabolismo , Regulación Neoplásica de la Expresión Génica , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Transducción de Señal , Proteínas Quinasas S6 Ribosómicas 90-kDa
10.
Anticancer Res ; 43(11): 4923-4935, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37909971

RESUMEN

BACKGROUND: MicroRNAs (miRNAs) are emerging as potential blood-based biomarkers involved in various types of carcinogenesis, including lung adenocarcinoma (LUAD). MATERIALS AND METHODS: In the present study, microarray was used to screen 2,549 miRNAs in serum samples from seven patients with LUAD and seven from healthy controls. Quantitative real-time polymerase chain reaction was used to validate the expression of miRNA in serum samples from 30 patients with LUAD and 30 heathy individuals. The area under the receiver operating characteristic curve was determined to evaluate the diagnostic capability of miR-625-3p. Cell counting kit-8 assay and Transwell assays were used to explore cell proliferation, migration and invasion. Bioinformatics prediction was applied in the search for the target genes of miR-625-3p. Quantitative real-time polymerase chain reaction, western blot and dual luciferase assay were used to validate target genes of miR-625-3p. A xenograft tumor model was established to evaluate cell proliferation in vivo. RESULTS: miR-625-3p was the miRNA most highly expressed in serum samples from patients with LUAD according to microarray analysis, this finding was verified in sera from an independent cohort, as well as in tissues based on The Cancer Genome Atlas database. Serum miR-625-3p provided a high diagnostic accuracy for LUAD (area under the curve=0.790, 95% confidence interval=0.6640-0.9152). Functionally, miR-625-3p promoted LUAD cell proliferation, migration and invasion both in vivo and in vitro. Mechanistically, we found miR-625-3p promoted cell proliferation and metastasis of LUAD by directly targeting KLF transcription factor 9 (Kruppel-like factor 9, KLF9). CONCLUSION: Our study identified that miR-625-3p plays an oncogenic role in LUAD, targeting KLF9. miR-625-3p might be a potential novel diagnostic biomarker and target for LUAD therapy.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , MicroARNs , Animales , Humanos , MicroARNs/genética , Adenocarcinoma del Pulmón/genética , Biomarcadores , Proliferación Celular/genética , Modelos Animales de Enfermedad , Neoplasias Pulmonares/genética , Factores de Transcripción de Tipo Kruppel/genética
11.
Front Bioinform ; 2: 836981, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36304284

RESUMEN

Background: Hepatocellular carcinoma (HCC) is a common malignant cancer. Metastasis plays a critical role in tumor progression, and vascular invasion is considered one of the most crucial factors for HCC metastasis. However, comprehensive analysis focusing on competitive endogenous RNA (ceRNA) and immune infiltration in the vascular invasion of HCC is lacking. Methods: The gene expression profiles of 321 samples, including 210 primary HCC cases and 111 HCC cases with vascular invasion, were downloaded from The Cancer Genome Atlas-Liver Hepatocellular Carcinoma project, and used in identifying significant differentially expressed lncRNAs (DElncRNAs), miRNAs (DEmiRNAs), and mRNAs (DEmRNAs). The RNAs associated with vascular invasion were used in constructing a ceRNA network. A multigene-based risk signature was constructed using the least absolute shrinkage and selection operator algorithm. We detected the fractions of 28 immune cell types in HCC through single-sample gene set enrichment analysis (ssGSEA). Finally, the relationship between the ceRNA network and immune cells was determined through correlation analysis and used in clarifying the potential mechanism involved in vascular invasion. Results: Overall, 413 DElncRNAs, 27 DEmiRNAs, and 397 DEmRNAs were recognized in HCC. A specific ceRNA network based on the interaction among 3 lncRNA-miRNA pairs and 24 miRNA-mRNA pairs were established. A ceRNA-based prognostic signature was constructed and used in dividing samples into high- and low-risk subgroups. The signature showed significant efficacy; its 3- and 5-year areas under the receiver operating characteristic curves were 0.712 and 0.653, respectively. ceRNA and ssGSEA integration analysis demonstrated that PART1 (p = 0, R = -0.33) and CDK5R2 (p = 0.01, R = -0.15) were negatively correlated to natural killer cells. Conclusion: This study demonstrated that vascular invasion in HCC might be related to PART1, and its role in regulating CDK5R2 and NK cells. A nomogram was developed to predict the prognosis of patients with HCC and demonstrated the value of the ceRNA network and tumor-infiltrating immune cells value in improving personalized management.

12.
Theranostics ; 11(6): 2564-2580, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33456560

RESUMEN

Rationale: Tumor microenvironment interacts with tumor cells to regulate their stemness properties through various cytokines and cytokine receptors. Previous studies revealed the possible role of interleukin 20 receptor subunit alpha (IL20RA) signaling in the progression of several types of tumors. However, its regulatory effects on the stemness and the microenvironment of breast cancer need to be studied. Methods: Immunohistochemical staining and western blot analysis were used to evaluate the association between IL20RA and SOX2 in breast tumors and noncancerous tissues. Enzyme-linked immunosorbent assay and TCGA dataset analysis were performed to determine the function of IL20RA signaling in breast cancer progression. Gain- and loss-of-function methods were performed to examine the effects of IL20RA on the stemness of breast cancer cells. The stemness features were analyzed by detecting the expression of core stemness genes, side population (SP), sphere formation ability, and aldehyde dehydrogenase (ALDH) activity. Flow cytometric analysis was applied to detect the changes of tumor-infiltration lymphocytes in tumor tissues in mice. Based on the relevant molecular mechanisms elucidated in this study, a novel IL20RA-targeted liposomal nanoparticle encapsulating the signal transducer and activator of transcription 3 (STAT3) inhibitor stattic (NP-Stattic-IL20RA) was synthesized. These NPs were combined with anti-programmed death ligand 1 (PD-L1) antibody and chemotherapy to inhibit the development of breast tumors in mice. Results: IL20RA is highly expressed in human breast cancers and is positively associated with the SOX2 expression. IL20RA increases the SP and ALDHbr proportions of breast cancer cells, enhances the sphere formation ability, and promotes the expression of core stemness genes, such as Sox2 and Oct4, as well as increases chemoresistance of breast cancer cells. IL20RA promotes the tumor-initiating ability and lung metastasis of breast cancer cells in vivo. In addition, IL20RA activates the Janus kinase 1 (JAK1)-STAT3-SOX2 signaling pathway, leading to increased expression of PD-L1 and reduced recruitment of anti-cancer lymphocytes, including CD8+ T cells and natural killer cells. Meanwhile, IL20RA signaling enhances the proportion of myeloid-derived suppressor cells. Combined with anti-PD-L1 antibody and NPs-Stattic-IL20RA, the chemotherapeutic efficacy was increased in breast cancer mouse models in vivo. Conclusion: Collectively, our results reveal that the IL20RA pathway is a novel signaling pathway involved in promoting the stemness features of breast cancer along with the formation of a tumor-favorable immune microenvironment. Targeting the IL20RAhi population with STAT3 signaling inhibition combined with anti-PD-L1 antibody can increase the therapeutic efficacy of chemotherapeutic agents for breast cancer. This study thus introduces a promising novel strategy for breast cancer therapy.


Asunto(s)
Neoplasias de la Mama/metabolismo , Células Madre Neoplásicas/metabolismo , Receptores de Interleucina/metabolismo , Transducción de Señal/fisiología , Microambiente Tumoral/fisiología , Aldehído Deshidrogenasa/metabolismo , Animales , Antígeno B7-H1/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Factor de Transcripción STAT3/metabolismo
14.
Theranostics ; 11(5): 2297-2317, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33500726

RESUMEN

Rationale: Considerable evidence suggests that breast cancer metastasis and recurrence occur due to emergence of cancer stem cells (CSCs). In our previous study, we designed a high-throughput siRNA screening platform that identifies inflammation genes involved in the regulation of cancer cell stemness. We reported that CCL16 protein decreases OCT4 expression and reduces the ALDH+ subpopulation. However, the mechanism by which CCL16 maintains stem cell-like properties remains unclear. Methods: Tissue microarrays were used to evaluate CCL16 expression. Cancer stemness assays were performed in CCL16 knockdown and overexpressing cells in vitro and in a xenograft model in vivo. Human phosphokinase array, immunofluorescence and chromatin immunoprecipitation assays were performed to explore the underlying mechanism. Results: We report that CCL16 was overexpressed in breast tumors and significantly correlated with clinical progression. We found that silencing CCL16 in MDA-MB-231 and BT549 cells diminished CSC properties including ALDH+ subpopulation, side population, chemo-resistance, and sphere formation. Furthermore, mice bearing CCL16-silenced MDA-MB-231 xenografts had lower tumorigenic frequency and developed smaller tumors. Exploration of the underlying mechanism found that CCL16 selects CCR2 to activate p-AKT/GSK3ß signaling and facilitate ß-catenin nuclear translocation. Further, CCL16 binds to the OCT4 promoter and promotes OCT4 expression. In addition, shRNAs targeting CCR2 and XAV939 targeting ß-catenin abolished CCL16-mediated cancer stemness. Upstream, IL10 mediates STAT3 activation, which binds to the CCL16 promoter and enhances its expression. The STAT3-targeted inhibitor Stattic suppressed CCL16 expression in vitro and restrained tumor progression in vivo. Conclusions: We identified a potential CSC regulator and suggest a novel mechanism for how CCL16 governs cancer cell stemness. We propose that CCL16 could be an effective target for breast cancer therapy.


Asunto(s)
Neoplasias de la Mama/patología , Quimiocinas CC/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células Madre Neoplásicas/patología , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Receptores CCR2/metabolismo , beta Catenina/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proliferación Celular , Quimiocinas CC/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Glucógeno Sintasa Quinasa 3 beta/genética , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Receptores CCR2/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/genética
15.
Stem Cell Res Ther ; 11(1): 370, 2020 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-32854760

RESUMEN

BACKGROUND: The widely recognized anti-cancer potential of aspirin has created a broad interest to explore the clinical benefits of aspirin in cancer therapy. However, the current understanding of the molecular mechanisms involved in the anti-cancer potential of aspirin remains limited. METHODS: Cancer stemness assays which contained ALDH, side population, chemo-resistance, sphere formation, and tumorigenesis were performed to validate aspirin function in vitro and in vivo. Histone modification assay was performed to check the effect of aspirin on histone methylation as well as the activity of HDAC and KDM6A/B. Inhibitor in vivo assay was performed to evaluate therapeutic effects of various inhibitor combination manners. RESULTS: In regards to in vitro studies, aspirin diminishes cancer cell stemness properties which include reducing the ALDH+ subpopulation, side population, chemo-resistance, and sphere formation in three cancer types. In regards to in vivo studies, aspirin decreases tumor growth and metastasis and prolongs survival. In addition, our results showed that aspirin inhibits inflammation-related stemness gene expression (especially ICAM3) identified by a high-throughput siRNA platform. In regards to the underlying molecular mechanism of action, aspirin reduces histone demethylase (KDM6A/B) expression that mediates histone methylation and suppresses gene expression via a COX-independent manner. In regards to therapeutic strategies, aspirin combined HDM inhibitors, ICAM3 downstream signaling Src/PI3K inhibitors, or ICAM3 inhibitor Lifitigrast prevents cancer progression in vivo. CONCLUSIONS: The aforementioned findings suggest a molecular model that explains how aspirin diminishes cancer cell stemness properties. These findings may provide novel targets for therapeutic strategies involving aspirin in the prevention of cancer progression.


Asunto(s)
Aspirina , Neoplasias , Aspirina/farmacología , Línea Celular Tumoral , Expresión Génica , Histonas/genética , Histonas/metabolismo , Humanos , Inflamación , Metilación , Células Madre Neoplásicas/metabolismo , Fosfatidilinositol 3-Quinasas
16.
Mol Cancer Ther ; 19(3): 868-881, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31871268

RESUMEN

Considerable evidence suggests that as breast cancer progresses, genetic and epigenetic mechanisms contribute to the emergence of self-renewing cells (CSC), which may also arise as a consequence of metastasis. Although the molecular pathways that trigger stemness and metastasis are known, key molecular and mechanistic gaps in our understanding of these processes remain unclear. Here, we first screened the inflammation-associated stemness gene phosphodiesterase 3A (PDE3A) using a medium-throughput siRNA library, which was overexpressed in breast tumors and significantly correlated with clinical progression. PDE3A induced the inflammatory nuclear factor NFκB signaling pathway by suppressing cAMP/PKA, which promotes the expression of the stem cell marker OCT4. In addition, PDE3A also promoted the translocation of CCDC88A from the cytoplasm to nuclei, thereby boosting the invasion-metastasis cascade in breast cancer. Most importantly, the PDE3A-selective inhibitor cilostazol dramatically suppressed breast tumor growth and reduced metastasis to the lungs in xenograft breast cancer models, with minimum toxicity. Taken together, we show that PDE3A could predispose patients with breast cancer to metastases by acting as a mediator of cancer stemness. PDE3A is a potential therapeutic target for advanced breast cancer.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Cilostazol/farmacología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/química , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células Madre Neoplásicas/efectos de los fármacos , Animales , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Neoplasias de la Mama/secundario , Proliferación Celular , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas de Microfilamentos/metabolismo , Persona de Mediana Edad , Células Madre Neoplásicas/enzimología , Células Madre Neoplásicas/metabolismo , Inhibidores de Fosfodiesterasa 3/farmacología , Pronóstico , Transporte de Proteínas , Transducción de Señal , Células Tumorales Cultivadas , Proteínas de Transporte Vesicular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
17.
ACS Appl Mater Interfaces ; 11(15): 13935-13944, 2019 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-30915833

RESUMEN

Photoactivated therapy, including photodynamic therapy (PDT) and photothermal therapy (PTT), is a spatiotemporally precise, controllable, and noninvasive method for tumor therapy and has therefore attracted increasing attention in recent years. However, it is still a challenge to obtain highly efficient therapeutic photoactive agents (PAAs) and deliver them into tumor, especially the core of solid tumors. Here, we have developed a newly engineered monocyte (MNC)-based PAA system that realizes precise and highly efficient tumor diagnosis and therapy. First, a near-infrared emissive PAA molecule with both strong singlet oxygen (1O2) production and high photothermal conversion efficiency was precisely designed for realizing simultaneous PDT and PTT of tumor and was further fabricated to form PAA nanoparticles (NPs). After loading the PAA NPs into MNCs, the MNCs were then decorated with cyclic Arg-Gly-Asp (cRGD) groups through a metabolic labeling method to further improve their ability of targeting and homing into the deep regions of tumors. Using this strategy, we have achieved highly efficient solid tumor ablation results both in vitro and in vivo, indicating that our strategy has a promising prospect for solid tumor therapy.


Asunto(s)
Rayos Láser , Monocitos/química , Nanopartículas/química , Neoplasias/terapia , Fármacos Fotosensibilizantes/química , Animales , Línea Celular Tumoral , Femenino , Humanos , Cetonas/química , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Monocitos/citología , Monocitos/metabolismo , Neoplasias/diagnóstico , Neoplasias/patología , Oligopéptidos/química , Péptidos Cíclicos/química , Fotoquimioterapia , Fármacos Fotosensibilizantes/uso terapéutico , Fototerapia , Pirroles/química , Teoría Cuántica , Oxígeno Singlete/metabolismo , Trasplante Heterólogo
18.
Cancer Med ; 8(2): 593-605, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30632703

RESUMEN

BACKGROUND: Current predictive model is not developed by inflammation-related genes to evaluate clinical outcome of breast cancer patients. METHODS: With mRNA expression profiling, we identified 3 mRNAs with significant expression between 15 normal samples and 669 breast cancer patients. Using 7 cell lines and 150 paraffin-embedded specimens, we verified the expression pattern by bio-experiments. Then, we constructed a three-mRNA model by Cox regression method and approved its predictive accuracy in both training set (n = 1095) and 4 testing sets (n = 703). RESULTS: We developed a three-mRNA (TBX21, TGIF2, and CYCS) model to stratify patients into high- and low-risk subgroup with significantly different prognosis. In training set, 5-year OS rate was 84.5% (78.8%-90.5%) vs 73.1% (65.9%-81.2%) for the low- and high-risk group (HR = 1.573 (1.090-2.271); P = 0.016). The predictive value was similar in four independent testing sets (HR>1.600; P < 0.05). This model could assess survival independently with better predictive power compared with single clinicopathological risk factors and any of the three mRNAs. Patients with both low-risk values and any poor prognostic factors had more favorable survival from nonmetastatic status (HR = 1.740 (1.028-2.945), P = 0.039). We established two nomograms for clinical application that integrated this model and another three significant risk factors to forecast survival rates precisely in patients with or without metastasis. CONCLUSIONS: This model is a dependable tool to predict the disease recurrence precisely and could improve the predictive accuracy of survival probability for breast cancer patients with or without metastasis.


Asunto(s)
Neoplasias de la Mama/genética , Citocromos c/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas Represoras/genética , Proteínas de Dominio T Box/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Humanos , Inflamación/genética , Masculino , Persona de Mediana Edad , Pronóstico , ARN Mensajero/genética
19.
Artículo en Inglés | MEDLINE | ID: mdl-31871777

RESUMEN

TGF-ß-induced factor homeobox 2 (TGIF2) is a transcription regulator that plays essential roles in the regulation of development and cell fate decisions. Aberrant expression of TGIF family proteins has been observed in several cancers, including ovarian, esophageal, and colorectal cancers. Here, we report that TGIF2 mediates the EGFR-RAS-ERK signaling pathway to enhance the stemness of lung adenocarcinoma (LUAD) cells and, therefore, promote the progression and metastasis of LUAD. We found that high TGIF2 expression was closely correlated with tumor growth, lymph node metastasis, and survival of patients with LUAD. Mice bearing TGIF2-silenced H1299 xenografts developed smaller tumors and fewer lung metastases. Importantly, silencing TGIF2 decreased the cancer stem cell (CSC)-like properties in A549 and H1299 cells. Furthermore, we identified that TGIF2 binding to the OCT4 promoter promotes its expression. In both LUAD cells and in vivo LUAD mouse models, we revealed that EGFR-RAS-ERK signaling phosphorylated TGIF2 and increased its stability, which was important for TGIF2-promoted LUAD stemness since phosphorylation-deficient TGIF2 mutants lost these functions. Thus, our study revealed that an important factor, TGIF2, bridges EGFR signaling to the CSC characteristics of LUAD cells, which can be utilized as an effective target for LUAD therapy.

20.
Cancer Med ; 8(5): 2325-2337, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30957973

RESUMEN

Toll-like receptor 4 (TLR4) plays an essential role in cancer progress. Here, we find that the expression of TLR4 in relapsed human hepatocellular carcinoma (HCC) clinical samples is higher than that in the non-relapsed ones, which leads us to explore the role of TLR4 in cancer stemness. We reported that TLR4-AKT signaling pathway was activated by lipopolysaccharides (LPS) in HCC cell lines to enhance the cancer stemness capacity, which was reflected by the increased percentage of CD133+ CD49f+ population and side population, enhanced sphere formation, and the upregulation of stemness marker gene-SOX2. Downregulation of SOX2 attenuated the enhanced HCC stemness induced by LPS, indicating SOX2 as a downstream mediator of LPS-TLR4 signaling. The role of LPS-TLR4 signaling in inducing HCC stemness was further confirmed by tumor xenograft experiment in vivo. Taken together, our findings provide a novel therapeutic target to prevent the recurrence of HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Células Madre Neoplásicas/metabolismo , Receptor Toll-Like 4/genética , Animales , Apoptosis , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Autorrenovación de las Células/genética , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Células Madre Neoplásicas/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Recurrencia , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal , Receptor Toll-Like 4/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA