Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Cell Mol Med ; 27(18): 2792-2803, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37610061

RESUMEN

Bisphenol A (BPA) is a ubiquitous environmental xenobiotic impacting millions of people worldwide. BPA has long been proposed to promote ovarian carcinogenesis, but the detrimental mechanistic target remains unclear. Cancer stem cells (CSCs) are considered as the trigger of tumour initiation and progression. Here, we show for the first time that nanomolar (environmentally relevant) concentration of BPA can markedly increase the formation and expansion of ovarian CSCs concomitant. This effect is observed in both oestrogen receptor (ER)-positive and ER-defective ovarian cancer cells, suggesting that is independent of the classical ERs. Rather, the signal is mediated through alternative ER G-protein-coupled receptor 30 (GPR30), but not oestrogen-related receptor α and γ. Moreover, we report a novel role of BPA in the regulation of Exportin-5 that led to dysregulation of microRNA biogenesis through miR-21. The use of GPR30 siRNA or antagonist to inhibit GPR30 expression or activity, respectively, resulted in significant inhibition of ovarian CSCs. Similarly, the CSCs phenotype can be reversed by expression of Exportin-5 siRNA. These results identify for the first time non-classical ER and microRNA dysregulation as novel mediators of low, physiological levels of BPA function in CSCs that may underlie its significant tumour-promoting properties in ovarian cancer.


Asunto(s)
MicroARNs , Neoplasias Ováricas , Humanos , Femenino , Neoplasias Ováricas/genética , MicroARNs/genética , Carioferinas
2.
Exp Cell Res ; 416(1): 113150, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35405118

RESUMEN

Among all gynecological malignancies, ovarian cancer (OC) accounts for the highest mortality rate due to high therapeutic resistance, prolonged latency and a lack of effective treatments. This calls for preclinical models that could recapitulate the histological, molecular and pathophysiological features of distinct OC subtypes. Various mouse models including tumor xenografts, genetically modified models, and novel 3D tumor models including organoids and organotypic co-culture models have been developed, and they serve as valuable assets to fulfill this demand. These models, particularly those patient-derived, can address the heterogeneity of OC and simulate OC progression in patients, hence bringing important insights for personalized treatments. In this review, we will discuss the merits and challenges of these models, and summarize their current preclinical applications in patient stratification and therapeutic research. Though limitations are inevitable, further optimization will render these models more clinically translatable in OC research.


Asunto(s)
Neoplasias Ováricas , Animales , Carcinoma Epitelial de Ovario/patología , Modelos Animales de Enfermedad , Femenino , Xenoinjertos , Humanos , Ratones , Organoides , Neoplasias Ováricas/patología
3.
Mod Pathol ; 33(1): 138-152, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31383965

RESUMEN

Female genital melanomas are rare. At diagnosis, most affected patients have advanced disease. Surgery remains the primary treatment, and adjuvant therapy is largely ineffective. Recently, immune checkpoints and the mitogen-activated protein kinase pathway have been explored as treatment targets. However, evaluation of these biomarkers in genital melanomas is limited. We evaluated the clinicopathological features of 20 vulvar, 32 vaginal, and three cervical melanomas and assessed programmed cell death ligand 1 (PD-L1) expression, CD8 tumor-infiltrating lymphocyte density, mismatch repair proteins, VE1 immunohistochemistry, and KIT and BRAF mutations. The median age of the patients was 66 years, and median tumor sizes were 25, 30, and 20 mm for vulvar, vaginal, and cervical tumors, respectively. Mean mitotic figures were 18, 19, and 30 per mm2. Thirty-seven patients (67%) had operable tumors. After a median follow-up of 15 months, only nine patients (16%) were alive. Eight of the nine survivors did not have lymph node metastasis. Using 5% as the threshold, PD-L1 expression was observed in 55%, 50%, and 33% of vulvar, vaginal, and cervical tumors, respectively, when the Roche SP263 antibody was used and 20%, 53%, and 0%, respectively, when the Dako 28-8 antibody was used. The median CD8 tumor-infiltrating lymphocyte density was significantly higher in vulvar/vaginal than cervical melanomas and correlated with PD-L1 expression. No cases exhibited loss of mismatch repair proteins. Five cases harbored KIT mutations, three of which were hotspots. BRAF V600E mutation was not detected. Univariable analysis showed that tumor size greater than or equal to 33 mm, mitotic figures of greater than or equal to 10 per mm2, lymph node metastasis, and low CD8+ tumor-infiltrating lymphocyte density were adverse prognostic factors. Thus, patients with genital melanomas have a poor prognosis, and evaluation of multiple biomarkers is necessary to identify patients who may benefit from immunotherapy or targeted therapy.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias de los Genitales Femeninos/patología , Melanoma/patología , Microambiente Tumoral/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Neoplasias de los Genitales Femeninos/genética , Neoplasias de los Genitales Femeninos/inmunología , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma/genética , Melanoma/inmunología , Persona de Mediana Edad
4.
Mol Ther ; 26(1): 70-83, 2018 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-29241971

RESUMEN

Metastasis is the cause of most (>90%) cancer deaths and currently lacks effective treatments. Approaches to understanding the biological process, unraveling the most effective molecular target(s), and implementing nanotechnology to increase the therapeutic index are expected to facilitate cancer therapy against metastasis. Here, we demonstrate the potential advantages of bringing these three approaches together through the rational design of a small interfering RNA (siRNA) that targets p70S6K in cancer stem cells (CSCs) in combination with dendrimer nanotechnology-based siRNA delivery. Our results demonstrated that the generation 6 (G6) poly(amidoamine) dendrimer can be used as a nanovector to effectively deliver p70S6K siRNA by forming uniform dendriplex nanoparticles that protect the siRNA from degradation. These nanoparticles were able to significantly knock down p70S6K in ovarian CSCs, leading to a marked reduction in CSC proliferation and expansion without obvious toxicity toward normal ovarian surface epithelial cells. Furthermore, treatment with the p70S6K siRNA/G6 dendriplexes substantially decreased mesothelial interaction, migration and invasion of CSCs in vitro, as well as tumor growth and metastasis in vivo. Collectively, these results suggest that p70S6K constitutes a promising therapeutic target, and the use of siRNA in combination with nanotechnology-based delivery may constitute a new approach for molecularly targeted cancer therapy to treat metastasis.


Asunto(s)
Dendrímeros , Técnicas de Transferencia de Gen , Células Madre Neoplásicas/metabolismo , Neoplasias Ováricas/genética , ARN Interferente Pequeño/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Animales , Adhesión Celular , Movimiento Celular/genética , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Neoplasias Ováricas/patología , Interferencia de ARN , Estabilidad del ARN , ARN Interferente Pequeño/administración & dosificación , Recurrencia , Nanomedicina Teranóstica
5.
Anal Chem ; 90(22): 13173-13177, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30354065

RESUMEN

In this study, we develop a method to detect multiple DNAs of foodborne pathogens by encapsulating emulsion droplets for loop-mediated isothermal amplification (LAMP). In contrast to the traditional bulk-phase LAMP, which involves a labor-intensive mixing process, with our method, different primers are automatically mixed with DNA samples and LAMP buffers after picoinjection. By directly observing and analyzing the fluorescence intensity of the resultant droplets, one can detect DNA from different pathogens, with a detection limit 500 times lower than that obtained by bulk-phase LAMP. We further demonstrate the ability to quantify bacteria concentration by detecting bacterial DNA in practical samples, showing great potential in monitoring water resources and their contamination by pathogenic bacteria.


Asunto(s)
Bacterias/aislamiento & purificación , ADN Bacteriano/análisis , Contaminación de Alimentos/análisis , Técnicas Analíticas Microfluídicas/métodos , Bacterias/genética , Enfermedades Transmitidas por los Alimentos/prevención & control , Dispositivos Laboratorio en un Chip , Límite de Detección , Técnicas de Amplificación de Ácido Nucleico/métodos , Sensibilidad y Especificidad , Aguas Residuales/análisis
6.
Nat Prod Rep ; 32(2): 256-72, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25347695

RESUMEN

Cancer is one of the leading causes of death worldwide. Ginseng, a key ingredient in traditional Chinese medicine, shows great promise as a new treatment option. As listed by the U.S. National Institutes of Health as a complementary and alternative medicine, its anti-cancer functions are being increasingly recognized. This review covers the mechanisms of action of ginsenosides and their metabolites, which can modulate signaling pathways associated with inflammation, oxidative stress, angiogenesis, metastasis, and stem/progenitor-like properties of cancer cells. The emerging use of structurally modified ginsenosides and recent clinical studies on the use of ginseng either alone or in combination with other herbs or Western medicines which are exploited as novel therapeutic strategies will also be explored.


Asunto(s)
Neoplasias/tratamiento farmacológico , Panax/química , Ginsenósidos/química , Ginsenósidos/aislamiento & purificación , Ginsenósidos/farmacología , Estructura Molecular , National Institutes of Health (U.S.) , Estados Unidos
7.
Mol Ther ; 21(1): 78-90, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23164934

RESUMEN

Ovarian cancer has a clear predilection to metastasize to the peritoneum, which represents one of the most important prognostic factors of poor clinical outcome. Gonadotropin-releasing hormone (GnRH) receptor is significantly overexpressed during the malignant progression of human ovarian cancer. Here, using lentiviral-based small interfering RNA (siRNA) technology to downregulate GnRH receptor in metastatic ovarian cancer cells, we show that GnRH receptor is an important mediator of ovarian cancer peritoneal metastasis. GnRH receptor downregulation dramatically attenuated their adhesion to the peritoneal mesothelium. By inhibiting the expression of GnRH receptor, we showed decreased expression of α2ß1 and α5ß1 integrin and adhesion to specific extracellular matrix (ECM) proteins. This was also associated with a reduction of P-cadherin. Furthermore, adhesion of ovarian cancer cells to different ECMs and the mesothelium were abrogated in response to ß1 integrin and P-cadherin reduction, confirming that the effects were ß1 integrin- and P-cadherin-specific. Using a mouse model of human ovarian cancer metastasis, we found that the inhibition of GnRH receptor, ß1 integrin, and P-cadherin significantly attenuated tumor growth, ascites formation, and the number of metastatic implants. These results define a new role for GnRH receptor in early metastasis and offer the possibility of novel therapeutic targets.


Asunto(s)
Adhesión Celular , Epitelio/patología , Metástasis de la Neoplasia/prevención & control , Neoplasias Ováricas/patología , Receptores LHRH/genética , Animales , Secuencia de Bases , Western Blotting , Línea Celular Tumoral , Cartilla de ADN , Femenino , Humanos , Integrina beta1/fisiología , Lentivirus/genética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
8.
Sci Transl Med ; 16(763): eadl3598, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39231239

RESUMEN

With the revolutionary progress of immune checkpoint inhibitors (ICIs) in non-small cell lung cancer, identifying patients with cancer who would benefit from ICIs has become critical and urgent. Here, we report protein tyrosine phosphatase receptor type T (PTPRT) loss as a precise and convenient predictive marker independent of PD-L1 expression for anti-PD-1/PD-L1 axis therapy. Anti-PD-1/PD-L1 axis treatment markedly increased progression-free survival in patients with PTPRT-deficient tumors. PTPRT-deficient tumors displayed cumulative DNA damage, increased cytosolic DNA release, and higher tumor mutation burden. Moreover, the tyrosine residue 240 of STING was identified as a direct substrate of PTPRT. PTPRT loss elevated phosphorylation of STING at Y240 and thus inhibited its proteasome-mediated degradation. PTPRT-deficient tumors released more IFN-ß, CCL5, and CXCL10 by activation of STING pathway and increased immune cell infiltration, especially of CD8 T cells and natural killer cells, ultimately enhancing the efficacy of anti-PD-1 therapy in multiple subcutaneous and orthotopic tumor mouse models. The response of PTPRT-deficient tumors to anti-PD-1 therapy depends on the tumor-intrinsic STING pathway. In summary, our findings reveal the mechanism of how PTPRT-deficient tumors become sensitive to anti-PD-1 therapy and highlight the biological function of PTPRT in innate immunity. Considering the prevalence of PTPRT mutations and negative expression, this study has great value for patient stratification and clinical decision-making.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Proteínas de la Membrana , Receptor de Muerte Celular Programada 1 , Transducción de Señal , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Humanos , Animales , Proteínas de la Membrana/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/genética , Transducción de Señal/efectos de los fármacos , Receptor de Muerte Celular Programada 1/metabolismo , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Ratones , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Línea Celular Tumoral , Fosforilación , Femenino
9.
Clin Cancer Res ; 30(17): 3904-3918, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-38848043

RESUMEN

PURPOSE: Uterine leiomyosarcoma (LMS) is an aggressive sarcoma and a subset of which exhibits DNA repair defects. Polo-like kinase 4 (PLK4) precisely modulates mitosis, and its inhibition causes chromosome missegregation and increased DNA damage. We hypothesize that PLK4 inhibition is an effective LMS treatment. EXPERIMENTAL DESIGN: Genomic profiling of clinical uterine LMS samples was performed, and homologous recombination (HR) deficiency scores were calculated. A PLK4 inhibitor (CFI-400945) with and without an ataxia telangiectasia mutated (ATM) inhibitor (AZD0156) was tested in vitro on gynecologic sarcoma cell lines SK-UT-1, SKN, and SK-LMS-1. Findings were validated in vivo using the SK-UT-1 xenograft model in the Balb/c nude mouse model. The effects of CFI-400945 were also evaluated in a BRCA2-knockout SK-UT-1 cell line. The mechanisms of DNA repair were analyzed using a DNA damage reporter assay. RESULTS: Uterine LMS had a high HR deficiency score, overexpressed PLK4 mRNA, and displayed mutations in genes responsible for DNA repair. CFI-400945 demonstrated effective antitumor activity in vitro and in vivo. The addition of AZD0156 resulted in drug synergism, largely due to a preference for nonhomologous end-joining DNA repair. Compared with wild-type cells, BRCA2 knockouts were more sensitive to PLK4 inhibition when both HR and nonhomologous end-joining repairs were impaired. CONCLUSIONS: Uterine LMS with DNA repair defects is sensitive to PLK4 inhibition because of the effects of chromosome missegregation and increased DNA damage. Loss-of-function BRCA2 alterations or pharmacologic inhibition of ATM enhanced the efficacy of the PLK4 inhibitor. Genomic profiling of an advanced-stage or recurrent uterine LMS may guide therapy.


Asunto(s)
Daño del ADN , Reparación del ADN , Leiomiosarcoma , Proteínas Serina-Treonina Quinasas , Neoplasias Uterinas , Ensayos Antitumor por Modelo de Xenoinjerto , Femenino , Humanos , Animales , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Leiomiosarcoma/genética , Leiomiosarcoma/tratamiento farmacológico , Leiomiosarcoma/patología , Neoplasias Uterinas/genética , Neoplasias Uterinas/tratamiento farmacológico , Neoplasias Uterinas/patología , Ratones , Línea Celular Tumoral , Reparación del ADN/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Proteínas de la Ataxia Telangiectasia Mutada/genética , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Morfolinas/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Indoles/farmacología , Indoles/uso terapéutico , Piridinas , Quinolinas
10.
Cancer Lett ; 535: 215593, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35176419

RESUMEN

p70 S6 kinase (p70S6K) is best known for its regulatory roles in protein synthesis and cell growth by phosphorylating its primary substrate, ribosomal protein S6, upon mitogen stimulation. The enhanced expression/activation of p70S6K has been correlated with poor prognosis in some cancer types, suggesting that it may serve as a biomarker for disease monitoring. p70S6K is a critical downstream effector of the oncogenic PI3K/Akt/mTOR pathway and its activation is tightly regulated by an ordered cascade of Ser/Thr phosphorylation events. Nonetheless, it should be noted that other upstream mechanisms regulating p70S6K at both the post-translational and post-transcriptional levels also exist. Activated p70S6K could promote various aspects of cancer progression such as epithelial-mesenchymal transition, cancer stemness and drug resistance. Importantly, novel evidence showing that p70S6K may also regulate different cellular components in the tumor microenvironment will be discussed. Therapeutic targeting of p70S6K alone or in combination with traditional chemotherapies or other microenvironmental-based drugs such as immunotherapy may represent promising approaches against cancers with aberrant p70S6K signaling. Currently, the only clinically available p70S6K inhibitors are rapamycin analogs (rapalogs) which target mTOR. However, there are emerging p70S6K-selective drugs which are going through active preclinical or clinical trial phases. Moreover, various screening strategies have been used for the discovery of novel p70S6K inhibitors, hence bringing new insights for p70S6K-targeted therapy.


Asunto(s)
Neoplasias , Proteínas Quinasas S6 Ribosómicas 70-kDa , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Oncogenes , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Quinasas S6 Ribosómicas/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Microambiente Tumoral
11.
Cells ; 11(23)2022 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-36497127

RESUMEN

Hepatocellular carcinoma (HCC) commonly possesses chronical elevation of IRE1α-ASK1 signaling. Orphan nuclear receptor Nur77, a promising therapeutic target in various cancer types, is frequently silenced in HCC. In this study, we show that cryptomeridiol (Bkh126), a naturally occurring sesquiterpenoid derivative isolated from traditional Chinese medicine Magnolia officinalis, has therapeutic efficacy in HCC by aggravating the pre-activated UPR and activating the silenced Nur77. Mechanistically, Nur77 is induced to sense IRE1α-ASK1-JNK signaling and translocate to the mitochondria, which leads to the loss of mitochondrial membrane potential (Δψm). The Bkh126-induced aggravation of ER stress and mitochondrial dysfunction result in increased cytotoxic product of reactive oxygen species (ROS). The in vivo anti-HCC activity of Bkh126 is superior to that of sorafenib, currently used to treat advanced HCC. Our study shows that Bkh126 induces Nur77 to connect ER stress to mitochondria-mediated cell killing. The identification of Nur77 as a molecular target of Bhk126 provides a basis for improving the leads for the further development of anti-HCC drugs.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Receptores Nucleares Huérfanos , Humanos , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/patología , Estrés del Retículo Endoplásmico , Endorribonucleasas , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Receptores Nucleares Huérfanos/metabolismo , Proteínas Serina-Treonina Quinasas
12.
Cell Death Dis ; 13(6): 537, 2022 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-35676254

RESUMEN

Overcoming drug resistance is an inevitable challenge to the success of cancer treatment. Recently, in ovarian cancer, a highly chemoresistant tumor, we demonstrated an important role of shear stress in stem-like phenotype and chemoresistance using a three-dimensional microfluidic device, which most closely mimics tumor behavior. Here, we examined a new mechanosensitive microRNA-miR-199a-3p. Unlike most key microRNA biogenesis in static conditions, we found that Dicer, Drosha, and Exportin 5 were not involved in regulating miR-199a-3p under ascitic fluid shear stress (0.02 dynes/cm2). We further showed that hepatocyte growth factor (HGF), but not other ascitic cytokines/growth factors such as epidermal growth factor and tumor necrosis factor α or hypoxia, could transcriptionally downregulate miR-199a-3p through its primary transcript miR-199a-1 and not miR-199a-2. Shear stress in the presence of HGF resulted in a concerted effect via a specific c-Met/PI3K/Akt signaling axis through a positive feedback loop, thereby driving cancer stemness and drug resistance. We also showed that miR-199a-3p expression was inversely correlated with enhanced drug resistance properties in chemoresistant ovarian cancer lines. Patients with low miR-199a-3p expression were more resistant to platinum with a significantly poor prognosis. miR-199a-3p mimic significantly suppressed ovarian tumor metastasis and its co-targeting in combination with cisplatin or paclitaxel further decreased the peritoneal dissemination of ovarian cancer in mice. These findings unravel how biophysical and biochemical cues regulate miR-199a-3p and is important in chemoresistance. miR-199a-3p mimics may serve as a novel targeted therapy for effective chemosensitization.


Asunto(s)
MicroARNs , Neoplasias Ováricas , Animales , Líquido Ascítico/metabolismo , Líquido Ascítico/patología , Línea Celular Tumoral , Proliferación Celular/genética , Resistencia a Antineoplásicos/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Ratones , MicroARNs/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Transducción de Señal/genética
13.
Adv Sci (Weinh) ; 9(16): e2103230, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35403834

RESUMEN

Tumor heterogeneity plays a key role in cancer relapse and metastasis, however, the distinct cellular behaviors and kinetics of interactions among different cancer cell subclones and the tumor microenvironment are poorly understood. By profiling an isogenic model that resembles spontaneous human ovarian cancer metastasis with an highly metastatic (HM) and non-metastatic (NM) tumor cell pair, one finds an upregulation of Wnt/ß-catenin signaling uniquely in HM. Using humanized immunocompetent mice, one shows for the first time that activated ß-catenin acts nonautonomously to modulate the immune microenvironment by enhancing infiltrating tumor-associated macrophages (TAM) at the metastatic site. Single-cell time-lapse microscopy further reveals that upon contact with macrophages, a significant subset of HM, but not NM, becomes polyploid, a phenotype pivotal for tumor aggressiveness and therapy resistance. Moreover, HM, but not NM, polarizes macrophages to a TAM phenotype. Mechanistically, ß-catenin upregulates cancer cell surface metadherin, which communicates through CEACAM1 expressed on macrophages to produce CCL3. Tumor xenografts in humanized mice and clinical patient samples both corroborate the relevance of enhanced metastasis, TAM activation, and polyploidy in vivo. The results thus suggest that targeting the ß-catenin-metadherin/CEACAM1-CCL3 positive feedback cascade holds great therapeutic potential to disrupt polyploidization of the cancer subclones that drive metastasis.


Asunto(s)
Vía de Señalización Wnt , beta Catenina , Animales , Antígenos CD , Antígeno Carcinoembrionario/metabolismo , Moléculas de Adhesión Celular , Línea Celular Tumoral , Quimiocina CCL3/metabolismo , Humanos , Macrófagos/metabolismo , Ratones , Recurrencia Local de Neoplasia/metabolismo , Factores de Transcripción/metabolismo , Microambiente Tumoral , Vía de Señalización Wnt/fisiología , beta Catenina/genética , beta Catenina/metabolismo
14.
Endocr Rev ; 28(4): 440-61, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17463396

RESUMEN

Ovarian epithelial cancer (OEC) accounts for 90% of all ovarian cancers and is the leading cause of death from gynecological cancers in North America and Europe. Despite its clinical significance, the factors that regulate the development and progression of ovarian cancer are among the least understood of all major human malignancies. The two gonadotropins, FSH and LH, are key regulators of ovarian cell functions, and the potential role of gonadotropins in the pathogenesis of ovarian cancer is suggested. Ovarian carcinomas have been found to express specific receptors for gonadotropins. The presence of gonadotropins in ovarian tumor fluid suggests the importance of these factors in the transformation and progression of ovarian cancers as well as being prognostic indicators. Functionally, there is evidence showing a direct action of gonadotropins on ovarian tumor cell growth. This review summarizes the key findings and recent advances in our understanding of these peptide hormones in ovarian cancer development and progression and their role in potential future cancer therapy. We will first discuss the supporting evidence and controversies in the "gonadotropin theory" and the use of animal models for exploring the involvement of gonadotropins in the etiology of ovarian cancer. The role of gonadotropins in regulating the proliferation, survival, and metastasis of OEC is next summarized. Relevant data from ovarian surface epithelium, which is widely believed to be the precursor of OEC, are also described. Finally, we will discuss the clinical applications of gonadotropins in ovarian cancer and the recent progress in drug development.


Asunto(s)
Gonadotropinas/fisiología , Neoplasias Glandulares y Epiteliales/etiología , Neoplasias Glandulares y Epiteliales/fisiopatología , Neoplasias Ováricas/etiología , Neoplasias Ováricas/fisiopatología , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/patología , Ratas , Receptores de Gonadotropina/fisiología
15.
Angiogenesis ; 14(4): 515-22, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21964931

RESUMEN

Hypoxia-inducible factor (HIF-1) is the key transcription regulator for multiple angiogenic factors and is an appealing target. Ginsenoside-Rg1, a nontoxic saponin isolated from the rhizome of Panax ginseng, exhibits potent proangiogenic activity and has the potential to be developed as a new angiotherapeutic agent. However, the mechanisms by which Rg1 promotes angiogenesis are not fully understood. Here, we show that Rg1 is an effective stimulator of HIF-1α under normal cellular oxygen conditions in human umbilical vein endothelial cells. HIF-1α steady-state mRNA was not affected by Rg1. Rather, HIF-1α protein synthesis was stimulated by Rg1. This effect was associated with constitutive activation of phosphatidylinositol 3-kinase (PI3K)/Akt and its effector p70 S6 kinase (p70(S6K)), but not extracellular-signal regulated kinase 1/2. We further revealed that HIF-1α induction triggered the expression of target genes, including vascular endothelial growth factor (VEGF). The use of small molecule inhibitors LY294002 or rapamycin to inhibit PI3K/Akt and p70(S6K) activities, respectively, resulted in diminished HIF-1α activation and subsequent VEGF expression. RNA interference-mediated knockdown of HIF-1α suppressed Rg1-induced VEGF synthesis and angiogenic tube formation, confirming that the effect was HIF-1α specific. Similarly, the angiogenic phenotype could be reversed by inhibition of PI3K/Akt and p70(S6K). These results define a hypoxia-independent activation of HIF-1α, uncovering a novel mechanism for Rg1 that could play a major role in angiogenesis and vascular remodeling.


Asunto(s)
Células Endoteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Ginsenósidos/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neovascularización Fisiológica/fisiología , Panax/química , Western Blotting , Células Cultivadas , Cromonas/farmacología , Cartilla de ADN/genética , Ginsenósidos/aislamiento & purificación , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasa/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rizoma/química , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Venas Umbilicales/citología , Factor A de Crecimiento Endotelial Vascular/metabolismo
16.
Theranostics ; 11(13): 6526-6541, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33995674

RESUMEN

The treatment for metastatic castration-resistant prostate cancer patients remains a great challenge in the clinic and continuously demands discoveries of new targets and therapies. Here, we assess the function and therapeutic value of SIRT6 in metastatic castration-resistant prostate cancer. Methods: The expression of SIRT6 was examined in prostate cancer tissue microarray by immunohistochemistry staining. The functions of SIRT6 and underlying mechanisms were elucidated by in vitro and in vivo experiments. We also developed an efficient method to silence SIRT6 by aptamer-modified exosomes carrying small interfering RNA and tested the therapeutic effect in the xenograft mice models. Results: SIRT6 expression is positively correlated with prostate cancer progression. Loss of SIRT6 significantly suppressed proliferation and metastasis of prostate cancer cell lines both in vitro and in vivo. SIRT6-driven prostate cancer displays activation of multiple cancer-related signaling pathways, especially the Notch pathway. Silencing SIRT6 by siRNA delivered through engineered exosomes inhibited tumor growth and metastasis. Conclusions: SIRT6 is identified as a driver and therapeutic target for metastatic prostate cancer in our findings, and inhibition of SIRT6 by engineered exosomes can serve as a promising therapeutic tool for clinical application.


Asunto(s)
Adenocarcinoma/terapia , Exosomas , Terapia Molecular Dirigida , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias de la Próstata/terapia , Sirtuinas/antagonistas & inhibidores , Adenocarcinoma/patología , Animales , Aptámeros de Nucleótidos , Carcinogénesis , Línea Celular Tumoral , ADN Complementario/genética , Progresión de la Enfermedad , Electroporación , Vectores Genéticos/farmacología , Vectores Genéticos/uso terapéutico , Células HEK293 , Humanos , Lentivirus/genética , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Metástasis de la Neoplasia , Neoplasias de la Próstata/patología , ARN Interferente Pequeño/genética , Receptores Notch/fisiología , Transducción de Señal , Ensayo de Tumor de Célula Madre , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Theranostics ; 11(7): 3376-3391, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33537093

RESUMEN

Background: Colorectal cancer (CRC) and the associated metastatic lesions are reported to be hypoxic. Hypoxia is a common feature in the tumor microenvironment and a potent stimulant of CRC. We have identified a regulatory role of Nur77 on Akt activation to enhance ß-catenin signaling essential for CRC progression under hypoxic conditions. Methods: The functional role of Nur77 in hypoxia-induced EMT was examined by scattering assays to monitor the morphologies of CRC cell lines under 1% O2. Sphere formation assays were performed to investigate whether Nur77 induced cancer stem cell-like properties in hypoxic CRC cells. The expression of various epithelial-to-mesenchymal transition (EMT) and stemness markers was analyzed by qPCR and Western blotting. Finally, Nur77 function and signaling in vivo was ascertained in subcutaneous tumor xenograft or liver metastasis model in nude mice using CRC cells stably transfected with appropriate constructs. Results: Herein, we show, for the first time, that Nur77 is a novel regulator of microRNA biogenesis that may underlie its significant tumor-promoting activities in CRC cells under hypoxia. Mechanistically, Nur77 interacted with the tumor suppressor protein p63, leading to the inhibition of p63-dependent transcription of Dicer, an important miRNA processor and subsequent decrease in the biogenesis of let-7i-5p which targeted the 3'UTR of p110α mRNA and regulated its stability. Knockdown of Nur77 or overexpression of let-7i-5p inhibited the tumor metastasis in vivo. Conclusion: Our data uncovered a novel mechanistic link connecting Nur77, Akt, and invasive properties of CRC in the hypoxic microenvironment.


Asunto(s)
Adenocarcinoma/genética , Fosfatidilinositol 3-Quinasa Clase I/genética , Neoplasias Colorrectales/genética , ARN Helicasas DEAD-box/genética , Hipoxia/genética , Neoplasias Hepáticas/genética , MicroARNs/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Proteínas Proto-Oncogénicas c-akt/genética , Ribonucleasa III/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/mortalidad , Adenocarcinoma/secundario , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , ARN Helicasas DEAD-box/metabolismo , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Hipoxia/metabolismo , Hipoxia/mortalidad , Hipoxia/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/secundario , Ratones , Ratones Desnudos , MicroARNs/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/antagonistas & inhibidores , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ribonucleasa III/metabolismo , Transducción de Señal , Análisis de Supervivencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Carga Tumoral , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Cancers (Basel) ; 12(8)2020 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-32785160

RESUMEN

Ovarian cancer is the most lethal gynecological malignancy worldwide. Unlike most other tumor types that metastasize via the vasculature, ovarian cancer metastasizes predominantly via the transcoelomic route within the peritoneal cavity. As cancer metastasis accounts for the majority of deaths, there is an urge to better understand its determinants. In the peritoneal cavity, tumor-mesothelial adhesion is an important step for cancer dissemination. Selectins are glycan-binding molecules that facilitate early steps of this adhesion cascade by mediating heterotypic cell-cell interaction under hydrodynamic flow. Here, we review the function and regulation of selectins in peritoneal carcinomatosis of ovarian cancer, and highlight how dysregulation of selectin ligand biogenesis affects disease outcome. Further, we will introduce the latest tools in studying selectin-glycan interaction. Finally, an overview of potential therapeutic intervention points that may lead to the development of efficacious therapies for ovarian cancer is provided.

19.
J Cell Biol ; 161(6): 1191-203, 2003 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-12810698

RESUMEN

Loss of E-cadherin expression or function in tumors leads to a more invasive phenotype. In this study, we investigated whether the invasion suppressor activity of E-cadherin is mediated directly by tighter physical cell adhesion, indirectly by sequestering beta-catenin and thus antagonizing beta-catenin/T cell factor (TCF) signaling, or by other signaling pathways. To distinguish mechanisms, we expressed wild-type E-cadherin and various E-cadherin mutants in invasive E-cadherin-negative human breast (MDA-MB-231) and prostate (TSU-Pr1) epithelial carcinoma cell lines using a tetracycline-inducible system. Our data confirm that E-cadherin inhibits human mammary and prostate tumor cell invasion. We find that adhesion is neither necessary nor sufficient for suppressing cancer invasion. Rather, the invasion suppressor signal is mediated through the beta-catenin-binding domain of the E-cadherin cytoplasmic tail but not through the p120ctn-binding domain. beta-catenin depletion also results in invasion suppression. However, alteration in the beta-catenin/TCF transcriptional regulation of target genes is not required for the invasion suppressor activity of E-cadherin, suggesting the involvement of other beta-catenin-binding proteins.


Asunto(s)
Cadherinas/metabolismo , Adhesión Celular/fisiología , Membrana Celular/metabolismo , Invasividad Neoplásica/fisiopatología , Neoplasias/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Cadherinas/genética , Carcinoma/genética , Carcinoma/metabolismo , Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Factor de Unión 1 al Potenciador Linfoide , Masculino , Neoplasias/fisiopatología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Unión Proteica/fisiología , Estructura Terciaria de Proteína/fisiología , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas , beta Catenina
20.
Mol Endocrinol ; 22(9): 2085-98, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18550773

RESUMEN

Tumorigenesis is a multistep process involving dysregulated cell growth and metastasis. Considerable evidence implicates a mitogenic action of estrogen in early ovarian carcinogenesis. In contrast, its influence in the metastatic cascade of ovarian tumor cells remains obscure. In the present study, we showed that 17beta-estradiol (E2) increased the metastatic potential of human epithelial ovarian cancer cell lines. E2 treatment led to clear morphological changes characteristic of epithelial-mesenchymal transition (EMT) and an enhanced cell migratory propensity. These morphological and functional alterations were associated with changes in the abundance of EMT-related genes. Upon E2 stimulation, expression and promoter activity of the epithelial marker E-cadherin were strikingly suppressed, whereas EMT-associated transcription factors, Snail and Slug, were significantly up-regulated. This up-regulation was attributed to the increase in gene transcription activated by E2. Depletion of endogenous Snail or Slug using small interfering RNA (siRNA) attenuated E2-mediated decrease in E-cadherin. In addition, E2-induced cell migration was also neutralized by the siRNAs, suggesting that both transcription factors are indispensable for the prometastatic actions of E2. More importantly, by using selective estrogen receptor (ER) agonists, forced expression, and siRNA approaches, we identified that E2 triggered the metastatic behaviors exclusively through an ERalpha-dependent pathway. We also showed that ERbeta had an opposing action on ERalpha because the presence of ERbeta completely inhibited the EMT and down-regulation of E-cadherin induced by ERalpha. Collectively, this study provides a compelling argument that estrogen can potentiate tumor progression by EMT induction and highlights the crucial role of ERalpha in ovarian tumorigenesis.


Asunto(s)
Cadherinas/genética , Cadherinas/metabolismo , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Secuencia de Bases , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Receptor beta de Estrógeno/metabolismo , Femenino , Humanos , Neoplasias Ováricas/fisiopatología , Neoplasias Ováricas/secundario , Regiones Promotoras Genéticas/efectos de los fármacos , ARN Interferente Pequeño/genética , Factores de Transcripción de la Familia Snail , Factores de Transcripción/antagonistas & inhibidores , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA