Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Adv Exp Med Biol ; 1286: 107-114, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33725348

RESUMEN

The majority of RNA transcripts are non-coding RNA (ncRNA) transcripts with lengths exceeding 200 nucleotides that are not translated into protein. Unlike microRNAs (miRNAs), long ncRNAs (lncRNAs) are not confined to a single mechanism of action but have a large and diverse role in biological processes as they can function as transcription regulators, decoys, scaffolds, and enhancer RNAs. Currently, many lncRNA molecules are under investigation for their role in tumorigenesis, metastasis, and prognosis in different types of cancer. This review not only summarizes the characteristics and functions of lncRNAs but also discusses the therapeutic implications and applications of lncRNAs with roles associated with head and neck cancer. Our aim is to pinpoint the potential way to perturb specific lncRNAs for future therapeutic use.


Asunto(s)
Neoplasias de Cabeza y Cuello , MicroARNs , ARN Largo no Codificante , Neoplasias de Cabeza y Cuello/genética , Humanos , MicroARNs/genética , ARN Largo no Codificante/genética
2.
Adv Exp Med Biol ; 1255: 165-173, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32949399

RESUMEN

PI3K inhibitors are a common area of research in finding a successful treatment of cancer. The PI3K pathway is important for cell growth, apoptosis, cell metabolism, cell survival, and a multitude of other functions. There are multiple isoforms of PI3K that can be broken down into three categories: class I, II, and III. Each isoform has at least one subunit that helps with the functionality of the isoform. Mutations found in the PI3K isoforms are commonly seen in many different types of cancer and the use of inhibitors is being tested to stop the cell survival of cancer cells. Individual PI3K inhibitors have shown some inhibition of the pathway; however, there is room for improvement. To better treat cancer, PI3K inhibitors are being combined with other pathway inhibitors. These combination therapies have shown better results with cancer treatments. Both the monotherapy and dual therapy treatments are still currently being studied and data collected to better understand cancer and other treatment options.


Asunto(s)
Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Fosfatidilinositol 3-Quinasas/química , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3/uso terapéutico , Humanos , Isoformas de Proteínas/antagonistas & inhibidores
3.
Adv Exp Med Biol ; 1260: 1-12, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32304028

RESUMEN

The control of crosstalk between autophagy and apoptosis in tumor cells can remove a critical barrier to comprehensive and efficacious treatment for cancer. Reactive oxygen species (ROS), by-products of redox homeostasis, are critical for regulating the balance between autophagy and apoptosis in cancer cells upon different drug treatments and gene modifications. The mechanisms and consequences involved in ROS-mediated crosstalk between apoptosis and autophagy are extremely complex in cancer cells. Here, we mainly discuss the closely linked relationship between ROS levels, autophagy, and apoptosis in cancer therapy.


Asunto(s)
Apoptosis , Autofagia , Neoplasias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Humanos , Oxidación-Reducción
4.
Adv Exp Med Biol ; 1134: 259-269, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30919342

RESUMEN

The ATPase family AAA-domain containing protein 3A (ATAD3A), a nuclear-encoded mitochondrial enzyme, is involved in diverse cellular processes, including mitochondrial dynamics, cell death and cholesterol metabolism. Overexpression and/or mutation of the ATAD3A gene have been observed in different types of cancer, associated with cancer development and progression. The dysregulated ATAD3A acts as a broker of a mitochondria-endoplasmic reticulum connection in cancer cells, and inhibition of this enzyme leads to tumor repression and enhanced sensitivity to chemotherapy and radiation. As such, ATAD3A is a promising drug target in cancer treatment.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales/metabolismo , Neoplasias/metabolismo , Animales , Retículo Endoplásmico/metabolismo , Humanos , Mitocondrias/metabolismo
5.
Adv Exp Med Biol ; 1134: 243-258, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30919341

RESUMEN

Phosphofructokinase-1 (PFK-1), a rate-determining enzyme of glycolysis, is an allosteric enzyme that regulates the oxidation of glucose in cellular respiration. Glycolysis phosphofructokinase platelet (PFKP) is the platelet isoform and works as an important mediator of cell metabolism. Considering that PFKP is a crucial player in many steps of cancer initiation and metastasis, we reviewed the specificities and complexities of PFKP and its biological roles in human diseases, especially malignant tumors. The possible use of PFKP as a diagnostic marker or a drug target in the prevention or treatment of cancer is also discussed.


Asunto(s)
Neoplasias/enzimología , Fosfofructoquinasa-1 Tipo C/metabolismo , Transducción de Señal , Glucosa/metabolismo , Glucólisis , Humanos
6.
Mol Carcinog ; 57(11): 1616-1625, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30074276

RESUMEN

Although genetic amplification and overexpression of the fibroblast growth factor 19 (FGF19) gene are found in human breast cancer, mechanisms that contribute to such functional alterations remain elusive. We report here that high expression of FGF19 is associated with the aggressive malignant behavior and poor survival outcome of breast cancer patients. FGF19 is particularly highly expressed in luminal molecular subtype of breast tumors and its expression levels are positively associated with its secretion levels from breast cancer cells. Genetic knockout of FGF19 significantly induces repression of breast tumor progression and metastasis in either an orthotopic mouse model of breast cancer or an experimental metastasis model. The FGF19 specific receptor, FGFR4, can be activated and subsequently upregulate AKT signaling in breast cancer cell upon FGF19, which is critical for oncogenic role of FGF19. Inactivation of FGFR4 by its inhibitor BLU9931 significantly attenuates FGF19-induced tumor-promoting activity, suggesting interruption of FGFR4 function is sufficient to affect FGF19-driven breast cancer. Overall, these insights support the idea that targeting FGFR4 in breast cancer cells overexpressing FGF19 may represent an effective strategy to suppress cancer development, progression, and metastasis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Factores de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Adulto , Anciano , Animales , Biomarcadores , Neoplasias de la Mama/genética , Neoplasias de la Mama/mortalidad , Línea Celular Tumoral , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Factores de Crecimiento de Fibroblastos/genética , Eliminación de Gen , Marcación de Gen , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/secundario , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Exp Clin Cancer Res ; 43(1): 168, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38877579

RESUMEN

PANoptosis represents a novel type of programmed cell death (PCD) with distinctive features that incorporate elements of pyroptosis, apoptosis, and necroptosis. PANoptosis is governed by a newly discovered cytoplasmic multimeric protein complex known as the PANoptosome. Unlike each of these PCD types individually, PANoptosis is still in the early stages of research and warrants further exploration of its specific regulatory mechanisms and primary targets. In this review, we provide a brief overview of the conceptual framework and molecular components of PANoptosis. In addition, we highlight recent advances in the understanding of the molecular mechanisms and therapeutic applications of PANoptosis. By elucidating the complex crosstalk between pyroptosis, apoptosis and necroptosis and summarizing the functional consequences of PANoptosis with a special focus on the tumor immune microenvironment, this review aims to provide a theoretical basis for the potential application of PANoptosis in cancer therapy.


Asunto(s)
Neoplasias , Humanos , Neoplasias/inmunología , Neoplasias/patología , Muerte Celular , Necroptosis , Microambiente Tumoral/inmunología , Animales , Piroptosis , Apoptosis
8.
Exp Hematol Oncol ; 13(1): 10, 2024 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-38287402

RESUMEN

Metabolic reprogramming is an emerging hallmark of cancer cells, enabling them to meet increased nutrient and energy demands while withstanding the challenging microenvironment. Cancer cells can switch their metabolic pathways, allowing them to adapt to different microenvironments and therapeutic interventions. This refers to metabolic heterogeneity, in which different cell populations use different metabolic pathways to sustain their survival and proliferation and impact their response to conventional cancer therapies. Thus, targeting cancer metabolic heterogeneity represents an innovative therapeutic avenue with the potential to overcome treatment resistance and improve therapeutic outcomes. This review discusses the metabolic patterns of different cancer cell populations and developmental stages, summarizes the molecular mechanisms involved in the intricate interactions within cancer metabolism, and highlights the clinical potential of targeting metabolic vulnerabilities as a promising therapeutic regimen. We aim to unravel the complex of metabolic characteristics and develop personalized treatment approaches to address distinct metabolic traits, ultimately enhancing patient outcomes.

9.
J Exp Clin Cancer Res ; 43(1): 203, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39044272

RESUMEN

BACKGROUND: Blacks/African American (BAA) patients diagnosed with head and neck squamous cell carcinoma (HNSCC) have worse survival outcomes than White patients. However, the mechanisms underlying racial disparities in HNSCC have not been thoroughly characterized. METHODS: Data on gene expression, copy number variants (CNVs), gene mutations, and methylation were obtained from 6 head and neck cancer datasets. Comparative bioinformatics analysis of the above genomic features was performed between BAAs and Whites. The expression pattern of GSTM1 was validated by immunohistochemistry using tumor tissue microarray (TMA). Effect of GSTM1 knockdown were assessed by cell proliferation, colony formation, and tumor development in an orthotopic mouse model. The changes in protein kinases were determined using the Proteome Profiler Human Phospho-Kinase Array Kit in HNSCC cells with or without GSTM1 knockdown. RESULTS: We identified ancestry-related differential genomic profiles in HNSCC. Specifically, in BAA HNSCC, FAT1 mutations were associated with its gene expression, SALL3 gene expression correlated with its gene CNVs, and RTP4 gene expression showed an inverse correlation with its methylation. Notably, GSTM1 emerged as a prognostic risk factor for BAA HNSCC, with high gene CNVs and expression levels correlating with poor overall survival in BAA patients. Immunohistochemistry results from newly developed in-house TMA validated the expression pattern of GSTM1 between BAA HNSCC and White HNSCC. In an orthotopic mouse model, GSTM1 knockdown significantly inhibited malignant progression in tumors derived from BAAs. In contrast, loss of GSTM1 did not affect the development of HNSCC originating in Whites. Mechanistically, GSTM1 knockdown suppressed HSP27 phosphorylation and ß-catenin in BAA HNSCC cells, but not in White HNSCC cells. This differential effect at least partially contributes to tumor development in BAA patients. CONCLUSION: This study identifies GSTM1 as a novel molecular determinant of survival in HNSCC patients of African descent. It also provides a molecular basis for future research focused on identifying molecular determinants and developing therapeutic interventions to improve outcomes for BAA patients with HNSCC.


Asunto(s)
Glutatión Transferasa , Neoplasias de Cabeza y Cuello , Animales , Femenino , Humanos , Masculino , Ratones , Biomarcadores de Tumor/genética , Negro o Afroamericano/genética , Línea Celular Tumoral , Variaciones en el Número de Copia de ADN , Glutatión Transferasa/genética , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/mortalidad , Pronóstico , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/mortalidad , Blanco/genética
10.
J Exp Clin Cancer Res ; 42(1): 4, 2023 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-36600271

RESUMEN

BACKGROUND: Immune-related adverse events (irAEs) are a common phenomenon in cancer patients treated with immune checkpoint inhibitors (ICIs). Surprisingly, the toxicity burdens of these irAEs have not been illustrated clearly. In this study, we analyzed irAEs for seven FDA-approved ICIs in cancer treatment to show the pattern of toxicity burden among cancer patients. METHODS: irAEs associated with seven FDA-approved ICIs, including three PD-1 inhibitors (cemiplimab, nivolumab and pembrolizumab), three PD-L1 inhibitors (atezolizumab, avelumab and durvalumab), and one CTLA-4 inhibitor (ipilimumab), were analyzed based on data from 149,303 reported cases (from January 1, 2015 to June 30, 2022) collected from the FDA Adverse Events Reporting System (FAERS) public dashboard. Proportions of serious irAEs and correlations with tumor type, age and sex were assessed via R package and GraphPad software. RESULTS: irAEs related to anti-PD-1 ICIs required less hospital care resources compared with anti-PD-L1 and anti-CTLA-4 ICIs. Patients treated with pembrolizumab had relatively fewer serious cases. Treatment with ICIs led to the highest probability of serious irAEs in patients with lung cancer. 'Respiratory, thoracic and mediastinal disorders' and 'gastrointestinal disorders' were the two most common groups of disorders caused by the seven ICIs studied. 'Cardiac disorders' was the main type of disorders caused by these ICIs in cancer patients aged 65-85, while 'reproductive system and breast disease' was the main type of disorder in cancer patients aged 18-64. 'Respiratory, thoracic, mediastinal diseases' and 'reproductive system and breast diseases' were the main types of disorders associated with treatment with these ICIs in male and female patients, respectively. CONCLUSION: Tissue and organ toxicities of ICIs are age and sex specific. There are risks of respiratory and urinary system toxicity in male patients and reproductive system toxicity in female patients treated with the ICIs studied. Future studies on the toxicity burden of ICIs should incorporate age and sex differences to better understand the relevance of ICI toxicity burden to human immune function to develop appropriate tumor immune and therapeutic intervention strategies.


Asunto(s)
Antineoplásicos Inmunológicos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Inhibidores de Puntos de Control Inmunológico , Neoplasias Pulmonares , Femenino , Humanos , Masculino , Antineoplásicos Inmunológicos/efectos adversos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/epidemiología , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Inmunoterapia/efectos adversos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Estados Unidos/epidemiología , Adolescente , Adulto Joven , Adulto , Persona de Mediana Edad , Anciano , Anciano de 80 o más Años
11.
Cancer Res Commun ; 3(4): 659-671, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37089864

RESUMEN

Radiotherapy plays an essential role in the treatment of head and neck squamous cell carcinoma (HNSCC), yet radioresistance remains a major barrier to therapeutic efficacy. A better understanding of the predominant pathways determining radiotherapy response could help develop mechanism-informed therapies to improve cancer management. Here we report that radioresistant HNSCC cells exhibit increased tumor aggressiveness. Using unbiased proteome profiler antibody arrays, we identify that upregulation of c-Met phosphorylation is one of the critical mechanisms for radioresistance in HNSCC cells. We further uncover that radioresistance-associated HNSCC aggressiveness is effectively exacerbated by c-Met but is suppressed by its genetic knockdown and pharmacologic inactivation. Mechanistically, the resulting upregulation of c-Met promotes elevated expression of plexin domain containing 2 (PLXDC2) through activating ERK1/2-ELK1 signaling, which in turn modulates cancer cell plasticity by epithelial-mesenchymal transition (EMT) induction and enrichment of the cancer stem cell (CSC) subpopulation, leading to resistance of HNSCC cells to radiotherapy. Depletion of PLXDC2 overcomes c-Met-mediated radioresistance through reversing the EMT progress and blunting the self-renewal capacity of CSCs. Therapeutically, the addition of SU11274, a selective and potent c-Met inhibitor, to radiation induces tumor shrinkage and limits tumor metastasis to lymph nodes in an orthotopic mouse model. Collectively, these significant findings not only demonstrate a novel mechanism underpinning radioresistance-associated aggressiveness but also provide a possible therapeutic strategy to target radioresistance in patients with HNSCC. Significance: This work provides novel insights into c-Met-PLXDC2 signaling in radioresistance-associated aggressiveness and suggests a new mechanism-informed therapeutic strategy to overcome failure of radiotherapy in patients with HNSCC.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Animales , Ratones , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas/metabolismo , Neoplasias de Cabeza y Cuello/genética , Células Madre Neoplásicas , Transducción de Señal
12.
Methods Mol Biol ; 2343: 159-164, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34473320

RESUMEN

Natural products, particularly as anticancer agents, continue to provide prototypes for pharmacologically active compounds. Compared with traditional two-dimensional (2D) approaches, 3D cell cultures have shown a clear role in drug discovery and development as they more closely resemble in vivo cell environments and come closer to capturing the in vivo functions of organs and tissues. The growing interest in using more physiological in vitro cancer models has driven the adoption of 3D cell cultures in evaluating anticancer activities of natural products. Here, we establish a protocol to use a novel 3D culture system to evaluate the therapeutic efficacy of epigallocatechin gallate (EGCG), a plant-based natural compound, in head and neck cancer cells. Our findings reveal that the sensitivity of natural products in 3D culture models may differ markedly from that obtained using 2D cultures, suggesting that 3D models will become a more reliable alternative to minimize misleading data.


Asunto(s)
Antineoplásicos , Productos Biológicos , Neoplasias , Antineoplásicos/farmacología , Productos Biológicos/farmacología , Técnicas de Cultivo Tridimensional de Células , Humanos , Neoplasias/tratamiento farmacológico
13.
Methods Mol Biol ; 2343: 3-18, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34473312

RESUMEN

In vitro cancer research models require the utmost accuracy and precision to effectively investigate physiological pathways and mechanisms, as well as test the therapeutic efficacy of anticancer drugs. Although two-dimensional (2D) cell culture models have been the traditional hallmark of cancer research, increasing evidence suggests 2D tumor models cannot accurately recapitulate complex aspects of tumor cells and drug responses. Three-dimensional (3D) cell cultures, however, are more physiologically relevant in oncology as they model the cancer network and microenvironment better, allowing for development and assessment of natural products and other anticancer drugs. The present review outlines unprecedented ways in which multicellular spheroid models, organoid models, hydrogel models, microfluidic devices, microfiber scaffold models, and tissue-engineered scaffold models are used in this research. The future of cancer research lies within 3D cell cultures, and as this approach improves, cancer research will continue to advance.


Asunto(s)
Antineoplásicos , Técnicas de Cultivo Tridimensional de Células , Neoplasias , Animales , Antineoplásicos/farmacología , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Humanos , Neoplasias/tratamiento farmacológico , Esferoides Celulares
14.
Methods Mol Biol ; 2343: 57-70, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34473315

RESUMEN

Phosphoglycerate kinase 1 (PGK1) is the first enzyme in glycolysis to generate a molecule of ATP in the conversion of 1,3-bisphosphoglycerate (1,3-BPG) to 3-phosphoglycerate (3-PG). In addition to the role of glycolysis, PGK-1 acts as a polymerase alpha cofactor protein, with effects on the tricarboxylic acid cycle, DNA replication and repair. Posttranslational modifications such as methylation, phosphorylation, and acetylation have been seen to activate PGK1 in cancer. High levels of intracellular PGK1 are associated with tumorigenesis and progression, and chemoradiotherapy resistance. However, high levels of extracellular PGK1 suppress angiogenesis and subsequently counteract cancer malignancy. Here we have summarized the current knowledge on the mechanisms and effects of PGK1 in various tumor types and evaluated its potential prognostic and therapeutic value in cancer. The data summarized here aims at providing molecular information and new ideas of employing natural products to combat cancer associated with PGK1.


Asunto(s)
Neoplasias , Fosfoglicerato Quinasa , Carcinogénesis , Glucólisis , Humanos , Neoplasias/genética , Fosfoglicerato Quinasa/genética , Fosfoglicerato Quinasa/metabolismo , Fosforilación
15.
J Exp Clin Cancer Res ; 41(1): 43, 2022 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-35093151

RESUMEN

BACKGROUND: Targeting mitochondrial oncoproteins presents a new concept in the development of effective cancer therapeutics. ATAD3A is a nuclear-encoded mitochondrial enzyme contributing to mitochondrial dynamics, cholesterol metabolism, and signal transduction. However, its impact and underlying regulatory mechanisms in cancers remain ill-defined. METHODS: We used head and neck squamous cell carcinoma (HNSCC) as a research platform and achieved gene depletion by lentiviral shRNA and CRISPR/Cas9. Molecular alterations were examined by RNA-sequencing, phospho-kinase profiling, Western blotting, RT-qPCR, immunohistochemistry, and immunoprecipitation. Cancer cell growth was assessed by MTT, colony formation, soft agar, and 3D cultures. The therapeutic efficacy in tumor development was evaluated in orthotopic tongue tumor NSG mice. RESULTS: ATAD3A is highly expressed in HNSCC tissues and cell lines. Loss of ATAD3A expression suppresses HNSCC cell growth and elicits tumor regression in orthotopic tumor-bearing mice, whereas gain of ATAD3A expression produces the opposite effects. From a mechanistic perspective, the tumor suppression induced by the overexpression of the Walker A dead mutant of ATAD3A (K358) produces a potent dominant-negative effect due to defective ATP-binding. Moreover, ATAD3A binds to ERK1/2 in the mitochondria of HNSCC cells in the presence of VDAC1, and this interaction is essential for the activation of mitochondrial ERK1/2 signaling. Most importantly, the ATAD3A-ERK1/2 signaling axis drives HNSCC development in a RAS-independent fashion and, thus, tumor suppression is more effectively achieved when ATAD3A knockout is combined with RAS inhibitor treatment. CONCLUSIONS: These findings highlight the novel function of ATAD3A in regulating mitochondrial ERK1/2 activation that favors HNSCC development. Combined targeting of ATAD3A and RAS signaling may potentiate anticancer activity for HNSCC therapeutics.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Humanos , Ratones , Ratones Endogámicos NOD , Transducción de Señal , Carcinoma de Células Escamosas de Cabeza y Cuello/patología
16.
J Exp Clin Cancer Res ; 40(1): 274, 2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34465361

RESUMEN

BACKGROUND: Metastasis is most often the root cause of cancer-related death. Human short stature homeobox 2 (SHOX2), a homeodomain transcription factor, is a novel inducer of epithelial-to-mesenchymal transition in breast cancer cells, though its exact role and underlying mechanisms in metastasis are not well understood. METHODS: TCGA analysis was performed to identify the clinical relevance of SHOX2 in breast cancer. Gene depletion was achieved by short hairpin RNA and small interfering RNA. Molecular regulations and alterations were assessed by Western blotting, immunoprecipitation, immunohistochemistry, qRT-PCR, chromatin immunoprecipitation coupled with qPCR (ChIP-qPCR), and ChIP/re-ChIP. The impact of SHOX2 signaling on tumor growth and metastasis was evaluated in orthotopic breast tumor mice. RESULTS: The expression level of SHOX2 is strongly associated with poor distant metastasis-free survival in breast cancer patients and inactivation of SHOX2 suppresses breast tumor growth and metastasis in mice. In breast cancer cells, SHOX2 directly activates Wiskott-Aldridge syndrome protein family member 3 (WASF3), a metastasis-promoting gene, at the transcriptional level, leading to a significant increase in metastatic potential. Mechanistically, SHOX2 activates signal transducer and activator of transcription 3 (STAT3) and recruits it to the WASF3 promoter, where STAT3 cooperates with SHOX2 to form a functional immunocomplex to promote WASF3 transcriptional activity in breast cancer cells. WASF3 knockdown abrogates SHOX2-induced metastasis, but not SHOX2-dependent tumorigenesis. CONCLUSIONS: These findings provide a critical link between the SHOX2-STAT3-WASF3 signaling axis and metastasis and suggest that the targeting of this signaling node may represent a valuable alternative strategy for combating breast cancer metastasis.


Asunto(s)
Neoplasias de la Mama/patología , Proteínas de Homeodominio/metabolismo , Metástasis de la Neoplasia , Factor de Transcripción STAT3/metabolismo , Activación Transcripcional , Familia de Proteínas del Síndrome de Wiskott-Aldrich/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Regiones Promotoras Genéticas , Unión Proteica , Transducción de Señal
17.
J Exp Clin Cancer Res ; 40(1): 93, 2021 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-33691750

RESUMEN

BACKGROUND: There is no consensus about the effective dosages of melatonin in cancer management, thus, it is imperative to fully understand the dose-dependent responsiveness of cancer cells to melatonin and the underlying mechanisms. METHODS: Head and neck squamous cell carcinoma (HNSCC) cells with or without melatonin treatment were used as a research platform. Gene depletion was achieved by short hairpin RNA, small interfering RNA, and CRISPR/Cas9. Molecular changes and regulations were assessed by Western blotting, quantitative RT-PCR (qRT-PCR), immunohistochemistry, and chromatin Immunoprecipitation coupled with qPCR (ChIP-qPCR). The therapeutic efficacy of FGF19/FGFR4 inhibition in melatonin-mediated tumor growth and metastasis was evaluated in orthotopic tongue tumor mice. RESULTS: The effect of melatonin on controlling cell motility and metastasis varies in HNSCC cells, which is dose-dependent. Mechanistically, high-dose melatonin facilitates the upregulation of FGF19 expression through activating endoplasmic stress (ER)-associated protein kinase RNA-like endoplasmic reticulum kinase (PERK)-Eukaryotic initiation factor 2 alpha (eIF2α)-activating transcription factor 4 (ATF4) pathway, which in turn promotes FGFR4-Vimentin invasive signaling and attenuates the role of melatonin in repressing metastasis. Intriguingly, following long-term exposure to high-dose melatonin, epithelial HNSCC cells revert the process towards mesenchymal transition and turn more aggressive, which is enabled by FGF19/FGFR4 upregulation and alleviated by genetic depletion of the FGF19 and FGFR4 genes or the treatment of FGFR4 inhibitor H3B-6527. CONCLUSIONS: Our study gains novel mechanistic insights into melatonin-mediated modulation of FGF19/FGFR4 signaling in HNSCC, demonstrating that activating this molecular node confines the role of melatonin in suppressing metastasis and even triggers the switch of its function from anti-metastasis to metastasis promotion. The blockade of FGF19/FGFR4 signaling would have great potential in improving the efficacy of melatonin supplements in cancer treatment.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Melatonina/genética , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Animales , Humanos , Ratones , Metástasis de la Neoplasia , Transducción de Señal , Carcinoma de Células Escamosas de Cabeza y Cuello/patología
18.
J Exp Clin Cancer Res ; 40(1): 393, 2021 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-34906193

RESUMEN

BACKGROUND: Alterations in metabolism are one of the emerging hallmarks of cancer cells and targeting dysregulated cancer metabolism provides a new approach to developing more selective therapeutics. However, insufficient blockade critical metabolic dependencies of cancer allows the development of metabolic bypasses, thus limiting therapeutic benefits. METHODS: A series of head and neck squamous cell carcinoma (HNSCC) cell lines and animal models were used to determine the efficacy of CPI-613 and CB-839 when given alone or in combination. Glutaminase 1 (GLS1) depletion was achieved by lentiviral shRNAs. Cell viability and apoptosis were determined in HNSCC cells cultured in 2D culture dish and SeedEZ™ 3D scaffold. Molecular alterations were examined by Western blotting and immunohistochemistry. Metabolic changes were assessed by glucose uptake, lactate production, glutathione levels, and oxygen consumption rate. RESULTS: We show here that HNSCC cells display strong addiction to glutamine. CPI-613, a novel lipoate analog, redirects cellular activity towards tumor-promoting glutaminolysis, leading to low anticancer efficacy in HNSCC cells. Mechanistically, CPI-613 inhibits the tricarboxylic acid cycle by blocking the enzyme activities of pyruvate dehydrogenase and alpha-ketoglutarate dehydrogenase, which upregulates GLS1 and eventually promotes the compensatory role of glutaminolysis in cancer cell survival. Most importantly, the addition of a GLS1 inhibitor CB-839 to CPI-613 treatment abrogates the metabolic dependency of HNSCC cells on glutamine, achieving a synergistic anticancer effect in glutamine-addicted HNSCC. CONCLUSIONS: These findings uncover the critical role of GLS1-mediated glutaminolysis in CPI-613 treatment and suggest that the CB-839 and CPI-613 combination may potentiate synergistic anticancer activity for HNSCC therapeutic gain.


Asunto(s)
Caprilatos/metabolismo , Glutamina/metabolismo , Neoplasias de Cabeza y Cuello/genética , Sulfuros/metabolismo , Animales , Apoptosis , Línea Celular Tumoral , Supervivencia Celular , Neoplasias de Cabeza y Cuello/patología , Humanos , Ratones
19.
Front Mol Biosci ; 7: 611847, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33521055

RESUMEN

The tumor microenvironment (TME) is a rich and active arena that is strategically evolved overtime by tumors to promote their survival and dissemination. Over the years, attention has been focused to characterize and identify the tumor-supporting roles and subsequent targeting potentials of TME components. Nevertheless, recapitulating the human TME has proved inherently challenging, leaving much to be explored. In this regard, in vivo model systems like zebrafish, with its optical clarity, ease of genetic manipulation, and high engraftment, have proven to be indispensable for TME modeling and investigation. In this review, we discuss the recent ways by which zebrafish models have lent their utility to provide new insights into the various cellular and molecular mechanisms driving TME dynamics and tumor support. Specifically, we report on innate immune cell interactions, cytokine signaling, metastatic plasticity, and other processes within the metastatic cascade. In addition, we reflect on the arrival of adult zebrafish models and the potential of patient-derived xenografts.

20.
Methods Mol Biol ; 2138: 167-173, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32219746

RESUMEN

With the development of new materials and technologies, it is possible to access gene function and drug metabolism using a three-dimensional (3D) cell culture system, which is more suitable for mimicking the in vivo microenvironment of cultured tumor cells ex vivo. SeedEZ is a novel and versatile tool that allows culturing of different types of cells with user convenience and in a desired sequence. This system provides a bridge between traditional 2D culture and animal experiments. Here, we provide two examples demonstrating how to evaluate cancer cell growth by the SeedEZ 3D scaffold and cancer cell invasion by the SeedEZ 3D ring, in order to promote understanding of the necessity of this novel cell culture system.


Asunto(s)
Proliferación Celular/fisiología , Invasividad Neoplásica/patología , Neoplasias/patología , Técnicas de Cultivo de Célula/métodos , Línea Celular Tumoral , Humanos , Andamios del Tejido/química , Microambiente Tumoral/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA