Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Front Oral Health ; 5: 1337582, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38370876

RESUMEN

Introduction: Oral squamous cell carcinoma (OSCC) is often preceded by oral epithelial dysplasia (OED). The role of ribosomal protein S6 (RPS6) and programmed cell death ligand-1 (PD-L1) in the progression of OED to OSCC remains unclear. This study aimed to investigate the expression of phosphorylated RPS6 (p-RPS6) and PD-L1 in OSCC and OED and to examine its relationship with clinicopathological features. Methods: Fifty-two OSCC and 48 OED cases were recruited for immunohistochemical analysis of p-RPS6 and PD-L1 expression. The expression of markers was correlated with clinicopathological features of OSCC and OED. Results: We found p-RPS6 expression in all cases of OSCC and OED, whereas PD-L1 was expressed in 42/48 (87%) OED and in 28/52 (53%) OSCC. The patients with mild OED presented higher expression level of PD-L1 and p-RPS6 significantly, when compared to moderate-differentiated OSCC patients (p < 0.05). Moreover, we found a significant positive correlation between PD-L1 and p-RPS6 expression in OED and OSCC patients (p < 0.01). The PD-L1 expression was significantly related to more than 2 cm tumor size in OSCC patients (p = 0.007). Discussion: Our findings suggest the upregulation of PD-L1 may be related with activation of the mTOR pathway in the early events of tumor progression and the pathogenesis of OSCC.

2.
Anticancer Res ; 42(5): 2689-2699, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35489730

RESUMEN

BACKGROUND/AIM: The prognosis of advanced stage head and neck squamous cell carcinoma (HNSCC) has remained unimproved for the past decades. Therefore, novel diagnostic markers and treatment options are required. Recently, an inhibitor for immune checkpoint program death ligand-1 (PD-L1), was approved by the FDA, and used in HNSCC patients. Histatins (HTNs), one of the common antimicrobial peptides in saliva, have demonstrated wound healing and antifungal capabilities and other functions on the oral epithelium. Dysregulation of HTN1 and HTN3 has also been reported in HNSCC through genomic and proteomic studies. This study aimed to investigate the association between histatins (HTN1 and HTN3) and PD-L1 in advanced HNSCC. PATIENTS AND METHODS: Data of gene expression in HNSCC were collected from TCGA and analyzed using a data-mining platform website (https://ualcan.path.uab.edu/). Tissue microarrays containing 98 samples of HNSCC patients and non-neoplastic controls were immunolabeled against PD-L1, HTN1, and HTN3. The immunohistochemistry results were quantified using ImageJ. RESULTS: The expression of PD-L1 and HTN1 was significantly higher in tumors than normal tissues (p<0.001), but no significant difference was found regarding HTN3. Metastatic HNSCC samples exhibited significantly higher expression of PD-L1 (p<0.018), compared to the non-metastatic group. Association between HTN1 and HTN3 was found using Pearson correlation coefficient (r=0.603, p<0.001). No overall survival difference was evident among our samples. CONCLUSION: PD-L1 and HTN1 are associated with the progression of HNSCC. PD-L1 expression correlated with that of HTN3.


Asunto(s)
Antígeno B7-H1/metabolismo , Neoplasias de Cabeza y Cuello , Histatinas/metabolismo , Neoplasias de Cabeza y Cuello/genética , Humanos , Ligandos , Proteómica , Carcinoma de Células Escamosas de Cabeza y Cuello
3.
Nat Commun ; 10(1): 5546, 2019 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-31804466

RESUMEN

Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide. Tobacco use is the main risk factor for HNSCC, and tobacco-associated HNSCCs have poor prognosis and response to available treatments. Recently approved anti-PD-1 immune checkpoint inhibitors showed limited activity (≤20%) in HNSCC, highlighting the need to identify new therapeutic options. For this, mouse models that accurately mimic the complexity of the HNSCC mutational landscape and tumor immune environment are urgently needed. Here, we report a mouse HNSCC model system that recapitulates the human tobacco-related HNSCC mutanome, in which tumors grow when implanted in the tongue of immunocompetent mice. These HNSCC lesions have similar immune infiltration and response rates to anti-PD-1 (≤20%) immunotherapy as human HNSCCs. Remarkably, we find that >70% of HNSCC lesions respond to intratumoral anti-CTLA-4. This syngeneic HNSCC mouse model provides a platform to accelerate the development of immunotherapeutic options for HNSCC.


Asunto(s)
Carcinoma de Células Escamosas/terapia , Neoplasias de Cabeza y Cuello/terapia , Inmunoterapia/métodos , Ipilimumab/uso terapéutico , Neoplasias de la Boca/terapia , Animales , Antineoplásicos Inmunológicos/uso terapéutico , Carcinoma de Células Escamosas/inducido químicamente , Línea Celular Tumoral , Modelos Animales de Enfermedad , Neoplasias de Cabeza y Cuello/inducido químicamente , Humanos , Ratones , Neoplasias de la Boca/inducido químicamente , Nicotiana/efectos adversos
4.
Cancer Res ; 79(17): 4360-4370, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31292160

RESUMEN

Metformin may reduce the progression of head and neck squamous cell carcinoma (HNSCC); however, whether metformin acts by altering the host metabolism or targets cancer-initiating cells remains poorly understood. This gap in knowledge has prevented the stratification of patient populations who are most likely to benefit from metformin treatment. Here, we explored whether metformin acts directly on HNSCC cells to inhibit aberrant cell growth. To investigate the tumor cell autonomous effects of metformin, we engineered representative HPV- and HPV+ HNSCC cells harboring typical genetic alternations to express the yeast mitochondrial NADH dehydrogenase (NDI1) protein, which is insensitive to metformin. NDI1 expression rescued the inhibitory effects of metformin on mitochondrial complex I, abolished the ability of metformin to activate AMP-activated protein kinase, and inhibited mTOR signaling both in vitro and in vivo, and was sufficient to render metformin ineffective to prevent HNSCC tumor growth. This experimental system provided an opportunity to identify metformin-regulated transcriptional programs linked to cancer cell growth inhibition in the tumor microenvironment. Remarkably, computational analysis of the metformin-induced transcriptome revealed that metformin downregulated gene expression signatures associated with cancer stemness and epithelial-mesenchymal transition, concomitant with increased expression of squamous differentiation genes. These findings support that metformin may act directly on cancer-initiating cells to prevent their progression to HNSCC, which may inform the selection of patients at risk of developing HNSCC in future early-stage clinical trials. SIGNIFICANCE: Metformin's ability to directly target HNSCC-initiating cells instead of exerting cancer preventive activity based solely on its systemic effects may inform the selection of patients in future precision prevention trials.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Metformina/farmacología , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Ratones Desnudos , Piruvatos/farmacología , Proteínas de Saccharomyces cerevisiae/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Serina-Treonina Quinasas TOR/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Dig Dis Sci ; 64(9): 2416-2428, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30863955

RESUMEN

Sepsis is a life-threatening response to systemic infection. In addition to frank gastrointestinal (GI) rupture/puncture, sepsis can also be exacerbated by translocation of pathogen-associated molecular patterns (PAMPs) from the GI tract to the systemic circulation (gut origin of sepsis). In the human gut, Gram-negative bacteria and Candida albicans are abundant, along with their major PAMP components, endotoxin (LPS) and (1 → 3)-ß-D-glucan (BG). Whereas the influence of LPS in bacterial sepsis has been studied extensively, exploration of the role of BG in bacterial sepsis is limited. Post-translocation, PAMPs enter the circulation through lymphatics and the portal vein, and are detoxified and then excreted via the liver and the kidney. Sepsis-induced liver and kidney injury might therefore affect the kinetics and increase circulating PAMPs. In this article, we discuss the current knowledge of the impact of PAMPs from both gut mycobiota and microbiota, including epithelial barrier function and the "gut-liver-kidney axis," on bacterial sepsis severity.


Asunto(s)
Infecciones Bacterianas/metabolismo , Candida/metabolismo , Mucosa Intestinal/metabolismo , Lipopolisacáridos/metabolismo , Sepsis/metabolismo , beta-Glucanos/metabolismo , Animales , Tracto Gastrointestinal/microbiología , Humanos , Mucosa Intestinal/microbiología , Riñón/metabolismo , Lipopolisacáridos/sangre , Hígado/metabolismo , Moléculas de Patrón Molecular Asociado a Patógenos/metabolismo , Proteoglicanos , Sepsis/inmunología , Sepsis/microbiología , beta-Glucanos/sangre
6.
Cancer Res ; 79(7): 1438-1450, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30894372

RESUMEN

Aberrant activation of the PI3K-mTOR signaling pathway occurs in >80% of head and neck squamous cell carcinomas (HNSCC), and overreliance on this signaling circuit may in turn represent a cancer-specific vulnerability that can be exploited therapeutically. mTOR inhibitors (mTORi) promote tumor regression in genetically defined and chemically induced HNSCC animal models, and encouraging results have been recently reported. However, the mTOR-regulated targets contributing to the clinical response have not yet been identified. Here, we focused on EIF4E-BP1 (4E-BP1), a direct target of mTOR that serves as key effector for protein synthesis. A systematic analysis of genomic alterations in the PIK3CA-mTOR pathway in HNSCC revealed that 4E-BP1 is rarely mutated, but at least one 4E-BP1 gene copy is lost in over 35% of the patients with HNSCC, correlating with decreased 4E-BP1 protein expression. 4E-BP1 gene copy number loss correlated with poor disease-free and overall survival. Aligned with a tumor-suppressive role, 4e-bp1/2 knockout mice formed larger and more lesions in models of HNSCC carcinogenesis. mTORi treatment or conditional expression of a mutant 4E-BP1 that cannot be phosphorylated by mTOR was sufficient to disrupt the translation-initiation complex and prevent tumor growth. Furthermore, CRISPR/Cas9-targeted 4E-BP1 HNSCC cells resulted in reduced sensitivity to mTORi in vitro and in vivo. Overall, these findings indicate that in HNSCC, mTOR persistently restrains 4E-BP1 via phosphorylation and that mTORi can restore the tumor-suppressive function of 4E-BP1. Our findings also support 4E-BP1 expression and phosphorylation status as a mechanistic biomarker of mTORi sensitivity in patients with HNSCC. SIGNIFICANCE: These findings suggest that EIF4E-BP1 acts as a tumor suppressor in HNSCC and that 4E-BP1 dephosphorylation mediates the therapeutic response to mTORi, providing a mechanistic biomarker for future precision oncology trials.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Proteínas Supresoras de Tumor/metabolismo , Animales , Benzoxazoles/farmacología , Biomarcadores de Tumor/metabolismo , Sistemas CRISPR-Cas , Línea Celular Tumoral , Proliferación Celular , Neoplasias de Cabeza y Cuello/patología , Humanos , Ratones , Ratones Noqueados , Fosforilación , Pronóstico , Pirimidinas/farmacología , Carcinoma de Células Escamosas de Cabeza y Cuello/patología
7.
Proc Natl Acad Sci U S A ; 115(3): E428-E437, 2018 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-29282319

RESUMEN

Fibrous dysplasia (FD) is a disease caused by postzygotic activating mutations of GNAS (R201C and R201H) that encode the α-subunit of the Gs stimulatory protein. FD is characterized by the development of areas of abnormal fibroosseous tissue in the bones, resulting in skeletal deformities, fractures, and pain. Despite the well-defined genetic alterations underlying FD, whether GNAS activation is sufficient for FD initiation and the molecular and cellular consequences of GNAS mutations remains largely unresolved, and there are no currently available targeted therapeutic options for FD. Here, we have developed a conditional tetracycline (Tet)-inducible animal model expressing the GαsR201C in the skeletal stem cell (SSC) lineage (Tet-GαsR201C/Prrx1-Cre/LSL-rtTA-IRES-GFP mice), which develops typical FD bone lesions in both embryos and adult mice in less than 2 weeks following doxycycline (Dox) administration. Conditional GαsR201C expression promoted PKA activation and proliferation of SSCs along the osteogenic lineage but halted their differentiation to mature osteoblasts. Rather, as is seen clinically, areas of woven bone admixed with fibrous tissue were formed. GαsR201C caused the concomitant expression of receptor activator of nuclear factor kappa-B ligand (Rankl) that led to marked osteoclastogenesis and bone resorption. GαsR201C expression ablation by Dox withdrawal resulted in FD-like lesion regression, supporting the rationale for Gαs-targeted drugs to attempt FD cure. This model, which develops FD-like lesions that can form rapidly and revert on cessation of mutant Gαs expression, provides an opportunity to identify the molecular mechanism underlying FD initiation and progression and accelerate the development of new treatment options.


Asunto(s)
Displasia Fibrosa Ósea/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Células Madre Mesenquimatosas/metabolismo , Animales , Antibacterianos/toxicidad , Desarrollo Óseo/efectos de los fármacos , Huesos/patología , Diferenciación Celular , Doxiciclina/toxicidad , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Regulación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Ratones , Mutación
8.
Clin Cancer Res ; 24(3): 619-633, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29146722

RESUMEN

Purpose: Recently, several comprehensive genomic analyses demonstrated NOTCH1 and NOTCH3 mutations in head and neck squamous cell carcinoma (HNSCC) in approximately 20% of cases. Similar to other types of cancers, these studies also indicate that the NOTCH pathway is closely related to HNSCC progression. However, the role of NOTCH4 in HNSCC is less well understood.Experimental Design: We analyzed NOTCH4 pathway and downstream gene expression in the TCGA data set. To explore the functional role of NOTCH4, we performed in vitro proliferation, cisplatin viability, apoptosis, and cell-cycle assays. We also compared the relationships among NOTCH4, HEY1, and epithelial-mesenchymal transition (EMT)-related genes using the TCGA data set and in vitro assays.Results:HEY1 is specifically upregulated in HNSCC compared with normal tissues in the TCGA data set. NOTCH4 is more significantly related to HEY1 activation in HNSCC in comparison with other NOTCH receptors. NOTCH4 promotes cell proliferation, cisplatin resistance, inhibition of apoptosis, and cell-cycle dysregulation. Furthermore, NOTCH4 and HEY1 upregulation resulted in decreased E-cadherin expression and increased Vimentin, Fibronectin, TWIST1, and SOX2 expression. NOTCH4 and HEY1 expression was associated with an EMT phenotype as well as increased invasion and cell migration.Conclusions: In HNSCC, the NOTCH4-HEY1 pathway is specifically upregulated, induces proliferation and cisplatin resistance, and promotes EMT. Clin Cancer Res; 24(3); 619-33. ©2017 AACR.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Ciclo Celular/metabolismo , Transición Epitelial-Mesenquimal , Receptor Notch4/metabolismo , Transducción de Señal , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Cisplatino/farmacología , Resistencia a Antineoplásicos , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Receptor Notch4/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/genética
9.
Methods ; 128: 3-11, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28780320

RESUMEN

Head and neck squamous cell carcinoma is one of the most common cancers with a 50% 5-year survival rate. Understanding the mechanisms that control development, progression, and spreading of the tumor to distal sites is of paramount importance to develop effective therapies. Here, we describe a minimally invasive procedure, which enables performing intravital microscopy of the mouse tongue in models for oral cancer and provides structural and dynamic information of the tumors at cellular and subcellular level.


Asunto(s)
Carcinoma de Células Escamosas/diagnóstico por imagen , Neoplasias de Cabeza y Cuello/diagnóstico por imagen , Microscopía Intravital/métodos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Línea Celular Tumoral , Femenino , Técnicas de Sustitución del Gen/métodos , Humanos , Ratones , Ratones Desnudos , Ratones SCID
10.
Carcinogenesis ; 37(10): 1014-25, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27538837

RESUMEN

The rising incidence of human papillomavirus (HPV)-associated malignancies, especially for oropharyngeal cancers, has highlighted the urgent need to understand how the interplay between high-risk HPV oncogenes and carcinogenic exposure results in squamous cell carcinoma (SCC) development. Here, we describe an inducible mouse model expressing high risk HPV-16 E6/E7 oncoproteins in adults, bypassing the impact of these viral genes during development. HPV-16 E6/E7 genes were targeted to the basal squamous epithelia in transgenic mice using a doxycycline inducible cytokeratin 5 promoter (cK5-rtTA) system. After doxycycline induction, both E6 and E7 were highly expressed, resulting in rapid epidermal hyperplasia with a remarkable expansion of the proliferative cell compartment to the suprabasal layers. Surprisingly, in spite of the massive growth of epithelial cells and their stem cell progenitors, HPV-E6/E7 expression was not sufficient to trigger mTOR activation, a key oncogenic driver in HPV-associated malignancies, and malignant progression to SCC. However, these mice develop SCC rapidly after a single exposure to a skin carcinogen, DMBA, which was increased by the prolonged exposure to a tumor promoter, 12-O-tetradecanoylphorbol-13-acetate (TPA). Thus, only few oncogenic hits may be sufficient to induce cancer in E6/E7 expressing cells. All HPV-E6/E7 expressing SCC lesions exhibited increased mTOR activation. Remarkably, rapamycin, an mTOR inhibitor, abolished tumor development when administered to HPV-E6/E7 mice prior to DMBA exposure. Our findings revealed that mTOR inhibition protects HPV-E6/E7 expressing tissues form SCC development upon carcinogen exposure, thus supporting the potential clinical use of mTOR inhibitors as a molecular targeted approach for prevention of HPV-associated malignancies.


Asunto(s)
Carcinógenos/toxicidad , Carcinoma de Células Escamosas/genética , Neoplasias Orofaríngeas/genética , Infecciones por Papillomavirus/genética , Serina-Treonina Quinasas TOR/biosíntesis , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Animales , Carcinoma de Células Escamosas/inducido químicamente , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/virología , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/patogenicidad , Humanos , Ratones , Proteínas Oncogénicas Virales/genética , Neoplasias Orofaríngeas/inducido químicamente , Neoplasias Orofaríngeas/tratamiento farmacológico , Neoplasias Orofaríngeas/virología , Proteínas E7 de Papillomavirus/genética , Infecciones por Papillomavirus/patología , Infecciones por Papillomavirus/virología , Ésteres del Forbol/toxicidad , Proteínas Represoras/genética , Sirolimus/administración & dosificación , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/genética
11.
Oncotarget ; 7(10): 10696-709, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26882569

RESUMEN

mTOR inhibition has emerged as a promising strategy for head and neck squamous cell carcinomas (HNSCC) treatment. However, most targeted therapies ultimately develop resistance due to the activation of adaptive survival signaling mechanisms limiting the activity of targeted agents. Thus, co-targeting key adaptive mechanisms may enable more effective cancer cell killing. Here, we performed a synthetic lethality screen using shRNA libraries to identify druggable candidates for combinatorial signal inhibition. We found that the ERK pathway was the most highly represented. Combination of rapamycin with trametinib, a MEK1/2 inhibitor, demonstrated strong synergism in HNSCC-derived cells in vitro and in vivo, including HNSCC cells expressing the HRAS and PIK3CA oncogenes. Interestingly, cleaved caspase-3 was potently induced by the combination therapy in PIK3CA+ cells in vitro and tumor xenografts. Moreover, ectopic expression of PIK3CA mutations into PIK3CA- HNSCC cells sensitized them to the pro-apoptotic activity of the combination therapy. These findings indicate that co-targeting the mTOR/ERK pathways may provide a suitable precision strategy for HNSCC treatment. Moreover, PIK3CA+ HNSCC are particularly prone to undergo apoptosis after mTOR and ERK inhibition, thereby providing a potential biomarker of predictive value for the selection of patients that may benefit from this combination therapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Fosfatidilinositol 3-Quinasa Clase I/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neoplasias de la Boca/tratamiento farmacológico , Piridonas/farmacología , Pirimidinonas/farmacología , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/genética , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Sinergismo Farmacológico , Femenino , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , Terapia Molecular Dirigida , Neoplasias de la Boca/enzimología , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Piridonas/administración & dosificación , Pirimidinonas/administración & dosificación , Interferencia de ARN , Sirolimus/administración & dosificación , Serina-Treonina Quinasas TOR/metabolismo
12.
Nat Commun ; 6: 6725, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25857352

RESUMEN

Histamine-induced vascular leakage is an integral component of many highly prevalent human diseases, including allergies, asthma and anaphylaxis. Yet, how histamine induces the disruption of the endothelial barrier is not well defined. By using genetically modified animal models, pharmacologic inhibitors and a synthetic biology approach, here we show that the small GTPase RhoA mediates histamine-induced vascular leakage. Histamine causes the rapid formation of focal adherens junctions, disrupting the endothelial barrier by acting on H1R Gαq-coupled receptors, which is blunted in endothelial Gαq/11 KO mice. Interfering with RhoA and ROCK function abolishes endothelial permeability, while phospholipase Cß plays a limited role. Moreover, endothelial-specific RhoA gene deletion prevents vascular leakage and passive cutaneous anaphylaxis in vivo, and ROCK inhibitors protect from lethal systemic anaphylaxis. This study supports a key role for the RhoA signalling circuitry in vascular permeability, thereby identifying novel pharmacological targets for many human diseases characterized by aberrant vascular leakage.


Asunto(s)
Anafilaxia/genética , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Histamina/farmacología , Quinasas Asociadas a rho/genética , Proteína de Unión al GTP rhoA/genética , Uniones Adherentes/efectos de los fármacos , Uniones Adherentes/metabolismo , Uniones Adherentes/patología , Amidas/farmacología , Anafilaxia/inducido químicamente , Anafilaxia/metabolismo , Anafilaxia/patología , Animales , Permeabilidad Capilar/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Femenino , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/deficiencia , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Noqueados , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores Histamínicos H1/genética , Receptores Histamínicos H1/metabolismo , Transducción de Señal , Piel/efectos de los fármacos , Piel/metabolismo , Piel/patología , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/deficiencia
13.
FASEB J ; 29(3): 1056-68, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25466898

RESUMEN

Multiple human malignancies rely on C-X-C motif chemokine receptor type 4 (CXCR4) and its ligand, SDF-1/CXCL12 (stroma cell-derived factor 1/C-X-C motif chemokine 12), to metastasize. CXCR4 inhibitors promote the mobilization of bone marrow stem cells, limiting their clinical application for metastasis prevention. We investigated the CXCR4-initiated signaling circuitry to identify new potential therapeutic targets. We used HeLa human cancer cells expressing high levels of CXCR4 endogenously. We found that CXCL12 promotes their migration in Boyden chamber assays and single cell tracking. CXCL12 activated mTOR (mechanistic target of rapamycin) potently in a pertussis-sensitive fashion. Inhibition of mTOR complex 1 (mTORC1) by rapamycin [drug concentration causing 50% inhibition (IC50) = 5 nM] and mTORC1/mTORC2 by Torin2 (IC50 = 6 nM), or by knocking down key mTORC1/2 components, Raptor and Rictor, respectively, decreased directional cell migration toward CXCL12. We developed a CXCR4-mediated spontaneous metastasis model by implanting HeLa cells in the tongue of SCID-NOD mice, in which 80% of the animals develop lymph node metastasis. It is surprising that mTORC1 disruption by Raptor knockdown was sufficient to reduce tumor growth by 60% and spontaneous metastasis by 72%, which were nearly abolished by rapamycin. In contrast, disrupting mTORC2 had no effect in tumor growth or metastasis compared with control short hairpin RNAs. These data suggest that mTORC1 may represent a suitable therapeutic target in human malignancies using CXCR4 for their metastatic spread. .


Asunto(s)
Movimiento Celular , Quimiocina CXCL12/metabolismo , Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , Complejos Multiproteicos/metabolismo , Receptores CXCR4/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Neoplasias del Cuello Uterino/secundario , Animales , Apoptosis , Western Blotting , Proliferación Celular , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas para Inmunoenzimas , Metástasis Linfática , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos NOD , Ratones SCID , Transducción de Señal , Células Tumorales Cultivadas , Neoplasias del Cuello Uterino/metabolismo
14.
Mol Oncol ; 8(1): 105-18, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24216180

RESUMEN

We have recently gained a remarkable understanding of the mutational landscape of head and neck squamous cell carcinoma (HNSCC). However, the nature of the dysregulated signaling networks contributing to HNSCC progression is still poorly defined. Here, we have focused on the role of the family of mitogen activated kinases (MAPKs), extracellular regulated kinase (ERK), c-Jun terminal kinase (JNK) and p38 MAPK in HNSCC. Immunohistochemical analysis of a large collection of human HNSCC tissues revealed that the levels of the phosphorylated active form of ERK1/2 and JNK were elevated in less than 33% and 16% of the cases, respectively. Strikingly, however, high levels of active phospho-p38 were observed in most (79%) of hundreds of tissues analyzed. We explored the biological role of p38 in HNSCC cell lines using three independent approaches: treatment with a specific p38 inhibitor, SB203580; a retro-inhibition strategy consisting in the use of SB203580 combined with the expression of an inhibitor-insensitive mutant form of p38α; and short-hairpin RNAs (shRNAs) targeting p38α. We found that specific blockade of p38 signaling significantly inhibited the proliferation of HNSCC cells both in vitro and in vivo. Indeed, we observed that p38 inhibition in HNSCC cancer cells reduces cancer growth in tumor xenografts and a remarkable decrease in intratumoral blood and lymphatic vessels. We conclude that p38α functions as a positive regulator of HNSCC in the context of the tumor microenvironment, controlling cancer cell growth as well as tumor-induced angiogenesis and lymphangiogenesis.


Asunto(s)
Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/fisiopatología , Neoplasias de Cabeza y Cuello/enzimología , Neoplasias de Cabeza y Cuello/fisiopatología , Linfangiogénesis , Neovascularización Patológica/enzimología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular , Citocinas/análisis , Activación Enzimática , Femenino , Neoplasias de Cabeza y Cuello/patología , Humanos , Ratones , Ratones Desnudos , Neovascularización Patológica/patología , Carcinoma de Células Escamosas de Cabeza y Cuello , Microambiente Tumoral , Proteínas Quinasas p38 Activadas por Mitógenos/análisis , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
15.
J Cell Biol ; 202(6): 951-66, 2013 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-24019537

RESUMEN

Tissue remodeling processes critically depend on the timely removal and remodeling of preexisting collagen scaffolds. Nevertheless, many aspects related to the turnover of this abundant extracellular matrix component in vivo are still incompletely understood. We therefore took advantage of recent advances in optical imaging to develop an assay to visualize collagen turnover in situ and identify cell types and molecules involved in this process. Collagen introduced into the dermis of mice underwent cellular endocytosis in a partially matrix metalloproteinase-dependent manner and was subsequently routed to lysosomes for complete degradation. Collagen uptake was predominantly executed by a quantitatively minor population of M2-like macrophages, whereas more abundant Col1a1-expressing fibroblasts and Cx3cr1-expressing macrophages internalized collagen at lower levels. Genetic ablation of the collagen receptors mannose receptor (Mrc1) and urokinase plasminogen activator receptor-associated protein (Endo180 and Mrc2) impaired this intracellular collagen degradation pathway. This study demonstrates the importance of receptor-mediated cellular uptake to collagen turnover in vivo and identifies a key role of M2-like macrophages in this process.


Asunto(s)
Colágeno Tipo I/fisiología , Colágeno/metabolismo , Macrófagos/metabolismo , Glicoproteínas de Membrana/fisiología , Receptores de Superficie Celular/fisiología , Receptores de Quimiocina/fisiología , Animales , Apoptosis , Western Blotting , Receptor 1 de Quimiocinas CX3C , Proliferación Celular , Células Cultivadas , Cadena alfa 1 del Colágeno Tipo I , Endocitosis/fisiología , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Técnicas para Inmunoenzimas , Lisosomas/metabolismo , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Inmunológicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
16.
J Cell Biol ; 201(7): 969-79, 2013 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-23798727

RESUMEN

Time-lapse fluorescence microscopy is one of the main tools used to image subcellular structures in living cells. Yet for decades it has been applied primarily to in vitro model systems. Thanks to the most recent advancements in intravital microscopy, this approach has finally been extended to live rodents. This represents a major breakthrough that will provide unprecedented new opportunities to study mammalian cell biology in vivo and has already provided new insight in the fields of neurobiology, immunology, and cancer biology.


Asunto(s)
Microscopía Fluorescente/métodos , Imagen de Lapso de Tiempo/métodos , Animales , Biología Celular/tendencias , Células Cultivadas , Técnicas Citológicas , Proteínas Fluorescentes Verdes/análisis , Humanos , Ratones , Microscopía Fluorescente/instrumentación , Imagen de Lapso de Tiempo/instrumentación
17.
Clin Cancer Res ; 19(6): 1375-88, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23340300

RESUMEN

PURPOSE: Head and neck squamous cell carcinoma (HNSCC) is one of the 10 most common cancers with a 50% five-year survival rate, which has remained unchanged for the past three decades. One of the major reasons for the aggressiveness of this cancer is that HNSCCs readily metastasize to cervical lymph nodes that are abundant in the head and neck region. Hence, discovering new molecules controlling the metastatic process as well as understanding their regulation at the molecular level are essential for effective therapeutic strategies. EXPERIMENTAL DESIGN: Rab25 expression level was analyzed in HNSCC tissue microarray. We used a combination of intravital microscopy in live animals and immunofluorescence in an in vitro invasion assay to study the role of Rab25 in tumor cell migration and invasion. RESULTS: In this study, we identified the small GTPase Rab25 as a key regulator of HNSCC metastasis. We observed that Rab25 is downregulated in HNSCC patients. Next, we determined that reexpression of Rab25 in a metastatic cell line is sufficient to block invasion in a three-dimensional collagen matrix and metastasis to cervical lymph nodes in a mouse model for oral cancer. Specifically, Rab25 affects the organization of F-actin at the cell surface, rather than cell proliferation, apoptosis, or tumor angiogenesis. CONCLUSION: These findings suggest that Rab25 plays an important role in tumor migration and metastasis, and that understanding its function may lead to the development of new strategies to prevent metastasis in oral cancer patients.


Asunto(s)
Carcinoma de Células Escamosas/genética , Neoplasias de Cabeza y Cuello/genética , Metástasis Linfática/genética , Neoplasias de la Lengua/genética , Proteínas de Unión al GTP rab/genética , Actinas/metabolismo , Animales , Carcinoma de Células Escamosas/patología , Proliferación Celular , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Células HeLa , Neoplasias de Cabeza y Cuello/patología , Humanos , Metástasis Linfática/patología , Ratones , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Análisis de Matrices Tisulares , Neoplasias de la Lengua/patología , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas de Unión al GTP rab/metabolismo
18.
Bioarchitecture ; 2(5): 143-57, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22992750

RESUMEN

Intravital microscopy is an extremely powerful tool that enables imaging several biological processes in live animals. Recently, the ability to image subcellular structures in several organs combined with the development of sophisticated genetic tools has made possible extending this approach to investigate several aspects of cell biology. Here we provide a general overview of intravital microscopy with the goal of highlighting its potential and challenges. Specifically, this review is geared toward researchers that are new to intravital microscopy and focuses on practical aspects of carrying out imaging in live animals. Here we share the know-how that comes from first-hand experience, including topics such as choosing the right imaging platform and modality, surgery and stabilization techniques, anesthesia and temperature control. Moreover, we highlight some of the approaches that facilitate subcellular imaging in live animals by providing numerous examples of imaging selected organelles and the actin cytoskeleton in multiple organs.


Asunto(s)
Citoesqueleto de Actina , Microscopía/métodos , Orgánulos , Animales
19.
Med Oncol ; 29(2): 690-703, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21380786

RESUMEN

A genetically related pair of human head and neck cancer (HNSCC) cell lines derived from the same patient at different stages of disease was used to investigate the role of extracellular matrix, integrin, and CXCL12-CXCR4 receptor interactions and their signal pathways in MMP-2 and MMP-9 activation and cell invasion. We found that collagen I enhanced MMP-2 and MMP-9 secretion in both primary and metastatic HNSCC cells. Collagen I acted through α(2)ß(1) integrin to activate tyrosine kinases, protein kinase C, ERK1/2, and p38, which in turn activated MMP-2 and MMP-9 production. The signaling function was also involved in the enhancement of cell invasion. Experiments using cocultures between live and fixed cells demonstrated that direct contact between tumor and fibroblast cells was required to activate MMP-2 and MMP-9 secretion in both tumor cells and fibroblasts. The augmentation appears specific for MMP-2. Fibroblasts seem to be responsible for the increased MMP-2 in the coculture. In addition, fibroblast or tumor cell-conditioned media upregulated the secretion of MMP-2 and MMP-9 in HNSCC cells. These findings indicate that autocrine and paracrine factors are involved in the augmented secretion of MMPs in coculture. We also found that CXCL12-enhanced HNSCC cell invasion through paracrine-activated CXCR4, which triggered MMP-dependent cell invasion. Together, our results suggest that cell-matrix and cell-cell interactions including autocrine and paracrine factors play important roles in the invasive behavior of HNSCC via upregulation of MMP-2 and MMP-9.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Movimiento Celular , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Western Blotting , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/secundario , Adhesión Celular , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Colágeno/metabolismo , Matriz Extracelular/patología , Fibroblastos/patología , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/patología , Humanos , Técnicas para Inmunoenzimas , Integrina alfa2beta1/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Invasividad Neoplásica , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Células Tumorales Cultivadas , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
20.
Cancer Res ; 71(22): 7103-12, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21975930

RESUMEN

Despite our improved understanding of cancer, the 5-year survival rate for head and neck squamous cell carcinomas (HNSCC) patients remains relatively unchanged at 50% for the past three decades. HNSCCs often metastasize to locoregional lymph nodes, and lymph node involvement represents one of the most important prognostic factors of poor clinical outcome. Among the multiple dysregulated molecular mechanism in HNSCCs, emerging basic, preclinical, and clinical findings support the importance of the mTOR signaling route in HNSCC progression. Indeed, we observed here that the activation of mTOR is a widespread event in clinical specimens of HNSCCs invading locoregional lymph nodes. We developed an orthotopic model of HNSCC consisting of the implantation of HNSCC cells into the tongues of immunocompromised mice. These orthotopic tumors spontaneously metastasize to the cervical lymph nodes, where the presence of HNSCC cells can be revealed by histologic and immunohistochemical evaluation. Both primary and metastatic experimental HNSCC lesions exhibited elevated mTOR activity. The ability to monitor and quantitate lymph node invasion in this model system enabled us to explore whether the blockade of mTOR could impact HNSCC metastasis. We found that inhibition of mTOR with rapamycin and the rapalog RAD001 diminished lymphangiogenesis in the primary tumors and prevented the dissemination of HNSCC cancer cells to the cervical lymph nodes, thereby prolonging animal survival. These findings may provide a rationale for the future clinical evaluation of mTOR inhibitors, including rapamycin and its analogues, as part of a molecular-targeted metastasis preventive strategy for the treatment of patients with HNSCC.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Linfangiogénesis/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Carcinoma de Células Escamosas/patología , Everolimus , Femenino , Neoplasias de Cabeza y Cuello/patología , Humanos , Metástasis Linfática , Ratones , Ratones SCID , Sirolimus/análogos & derivados , Sirolimus/uso terapéutico , Carcinoma de Células Escamosas de Cabeza y Cuello , Serina-Treonina Quinasas TOR/fisiología , Factores de Transcripción/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...