Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Clin Invest ; 133(22)2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37751299

RESUMEN

The B cell leukemia/lymphoma 2 (BCL-2) inhibitor venetoclax is effective in chronic lymphocytic leukemia (CLL); however, resistance may develop over time. Other lymphoid malignancies such as diffuse large B cell lymphoma (DLBCL) are frequently intrinsically resistant to venetoclax. Although genomic resistance mechanisms such as BCL2 mutations have been described, this probably only explains a subset of resistant cases. Using 2 complementary functional precision medicine techniques - BH3 profiling and high-throughput kinase activity mapping - we found that hyperphosphorylation of BCL-2 family proteins, including antiapoptotic myeloid leukemia 1 (MCL-1) and BCL-2 and proapoptotic BCL-2 agonist of cell death (BAD) and BCL-2 associated X, apoptosis regulator (BAX), underlies functional mechanisms of both intrinsic and acquired resistance to venetoclax in CLL and DLBCL. Additionally, we provide evidence that antiapoptotic BCL-2 family protein phosphorylation altered the apoptotic protein interactome, thereby changing the profile of functional dependence on these prosurvival proteins. Targeting BCL-2 family protein phosphorylation with phosphatase-activating drugs rewired these dependencies, thus restoring sensitivity to venetoclax in a panel of venetoclax-resistant lymphoid cell lines, a resistant mouse model, and in paired patient samples before venetoclax treatment and at the time of progression.


Asunto(s)
Antineoplásicos , Leucemia Linfocítica Crónica de Células B , Linfoma de Células B Grandes Difuso , Ratones , Animales , Humanos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/patología , Resistencia a Antineoplásicos/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Proteína bcl-X/genética , Proteínas Reguladoras de la Apoptosis , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/patología , Línea Celular Tumoral , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo
2.
Theranostics ; 13(2): 621-638, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36632230

RESUMEN

Rationale: Metastasis is a complex process with a molecular underpinning that remains unclear. We hypothesize that cargo proteins conducted by extracellular vesicles (EVs) released from tumors may confer growth and metastasis potential on recipient cells. Here, we report that a cytokine-like secreted protein, FAM3C, contributes to late-stage lung tumor progression. Methods: EV protein profiling was conducted with an unbiased proteomic mass spectrometry analysis on non-small cell lung cancer (NSCLC) and normal lung fibroblast cell lines. Expression of FAM3C was confirmed in a panel of NSCLC cell lines, and correlated to the invasive and metastatic potentials. Functional phenotype of endogenous FAM3C and tumor-derived EVs (TDEs) were further investigated using various biological approaches in RNA and protein levels. Metastasis potential of TDEs secreted by FAM3C-overexpressing carcinoma cells was validated in mouse models. Results: Transcriptomic meta-analysis of pan-cancer datasets confirmed the overexpression of FAM3C - a gene encoding for interleukin-like EMT inducer (ILEI) - in NSCLC tumors, with strong association with poor patient prognosis and cancer metastasis. Aberrant expression of FAM3C in lung carcinoma cells enhances cellular transformation and promotes distant lung tumor colonization. In addition, higher FAM3C concentrations were detected in EVs extracted from plasma samples of NSCLC patients compared to those of healthy subjects. More importantly, we defined a hitherto-unknown mode of microenvironmental crosstalk involving FAM3C in EVs, whereby the delivery and uptake of FAM3C via TDEs enhances oncogenic signaling - in recipient cells that phenocopies the cell-endogenous overexpression of FAM3C. The oncogenicity transduced by FAM3C is executed via a novel interaction with the Ras-related protein RalA, triggering the downstream activation of the Src/Stat3 signaling cascade. Conclusions: Our study describes a novel mechanism for FAM3C-driven carcinogenesis and shed light on EV FAM3C as a driver for metastatic lung tumors that could be exploited for cancer therapeutics.


Asunto(s)
Carcinogénesis , Carcinoma de Pulmón de Células no Pequeñas , Vesículas Extracelulares , Neoplasias Pulmonares , Animales , Humanos , Ratones , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/secundario , Línea Celular Tumoral , Vesículas Extracelulares/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Proteómica
3.
Molecules ; 25(20)2020 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-33092283

RESUMEN

Targeting altered tumour metabolism is an emerging therapeutic strategy for cancer treatment. The metabolic reprogramming that accompanies the development of malignancy creates targetable differences between cancer cells and normal cells, which may be exploited for therapy. There is also emerging evidence regarding the role of stromal components, creating an intricate metabolic network consisting of cancer cells, cancer-associated fibroblasts, endothelial cells, immune cells, and cancer stem cells. This metabolic rewiring and crosstalk with the tumour microenvironment play a key role in cell proliferation, metastasis, and the development of treatment resistance. In this review, we will discuss therapeutic opportunities, which arise from dysregulated metabolism and metabolic crosstalk, highlighting strategies that may aid in the precision targeting of altered tumour metabolism with a focus on combinatorial therapeutic strategies.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Células Madre Neoplásicas/metabolismo , Microambiente Tumoral/efectos de los fármacos , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Redes y Vías Metabólicas/efectos de los fármacos , Neoplasias/metabolismo , Células Madre Neoplásicas/efectos de los fármacos
4.
Clin Cancer Res ; 26(20): 5320-5328, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32816944

RESUMEN

PURPOSE: Induction cisplatin and gemcitabine chemotherapy is a standard treatment for locally advanced nasopharyngeal carcinoma (NPC). Inhibition of VEGF axis has been shown to promote maturation of microvasculature and improve perfusion. We conducted a four-arm study to assess the effect of two doses of either sunitinib or bevacizumab with chemotherapy in NPC. PATIENTS AND METHODS: Patients with treatment-naïve locally advanced NPC were treated with three cycles of 3-weekly cisplatin and gemcitabine preceded by 1 week of anti-VEGF therapy for each cycle, followed by standard concurrent chemoradiation: arm A patients received 7 days of 12.5 mg/day sunitinib; arm B 7 days of 25 mg/day sunitinib; arm C bevacizumab 7.5 mg/kg infusion; arm D bevacizumab 2.5 mg/kg infusion. Patients with metastatic NPC were treated with up to six cycles of similar treatment without concurrent chemoradiation. RESULTS: Complete metabolic response (mCR) by whole body 18FDG PET was highest in arm C (significant difference in four groups Fisher exact test P = 0.001; type 1 error = 0.05), with 42% mCR (95% confidence interval, 18-67) and 3-year relapse-free survival of 88% in patients with locally advanced NPC. Significant increase in pericyte coverage signifying microvascular maturation and increased immune cell infiltration was observed in posttreatment tumor biopsies in Arm C. Myelosuppression was more profound in sunitinib containing arms, and tolerability was established in arm C where hypertension was the most significant toxicity. CONCLUSIONS: Bevacizumab 7.5 mg/kg with cisplatin and gemcitabine was well tolerated. Promising tumor response was observed and supported mechanistically by positive effects on tumor perfusion and immune cell trafficking into the tumor.


Asunto(s)
Cisplatino/administración & dosificación , Desoxicitidina/análogos & derivados , Carcinoma Nasofaríngeo/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Cisplatino/efectos adversos , Desoxicitidina/administración & dosificación , Desoxicitidina/efectos adversos , Supervivencia sin Enfermedad , Femenino , Fluorouracilo/administración & dosificación , Humanos , Masculino , Persona de Mediana Edad , Carcinoma Nasofaríngeo/patología , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/patología , Neovascularización Patológica/patología , Sunitinib/administración & dosificación , Gemcitabina
5.
Nat Commun ; 11(1): 1556, 2020 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-32214092

RESUMEN

c-MET receptors are activated in cancers through genomic events like tyrosine kinase domain mutations, juxtamembrane splicing mutation and amplified copy numbers, which can be inhibited by c-MET small molecule inhibitors. Here, we discover that the most common polymorphism known to affect MET gene (N375S), involving the semaphorin domain, confers exquisite binding affinity for HER2 and enables METN375S to interact with HER2 in a ligand-independent fashion. The resultant METN375S/HER2 dimer transduces potent proliferative, pro-invasive and pro-metastatic cues through the HER2 signaling axis to drive aggressive squamous cell carcinomas of the head and neck (HNSCC) and lung (LUSC), and is associated with poor prognosis. Accordingly, HER2 blockers, but not c-MET inhibitors, are paradoxically effective at restraining in vivo and in vitro models expressing METN375S. These results establish METN375S as a biologically distinct and clinically actionable molecular subset of SCCs that are uniquely amenable to HER2 blocking therapies.


Asunto(s)
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Receptor ErbB-2/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/mortalidad , Línea Celular Tumoral , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/mortalidad , Neoplasias de Cabeza y Cuello/patología , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Ratones , Mutación , Fenotipo , Fosforilación/efectos de los fármacos , Polimorfismo Genético , Pronóstico , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas c-met/química , Receptor ErbB-2/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Antioxid Redox Signal ; 32(5): 285-308, 2020 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-31841375

RESUMEN

Significance: Cancer cells exhibit altered metabolic pathways to keep up with biosynthetic and reduction-oxidation needs during tumor proliferation and metastasis. The common induction of metabolic pathways during cancer progression, regardless of cancer histio- or genotype, makes cancer metabolism an attractive target for therapeutic exploitation. Recent Advances: Emerging data suggest that these altered pathways may even result in resistance to anticancer therapies. Identifying specific metabolic dependencies that are unique to cancer cells has proved challenging in this field, limiting the therapeutic window for many candidate drug approaches. Critical Issues: Cancer cells display significant metabolic flexibility in nutrient-limited environments, hampering the longevity of suppressing cancer metabolism through any singular approach. Combinatorial "synthetic lethal" approaches may have a better chance for success and promising strategies are reviewed here. The dynamism of the immune system adds a level of complexity, as various immune populations in the tumor microenvironment often share metabolic pathways with cancer, with successive alterations during immune activation and quiescence. Decoding the reprogramming of metabolic pathways within cancer cells and stem cells, as well as examining metabolic symbiosis between components of the tumor microenvironment, would be essential to further meaningful drug development within the tumor's metabolic ecosystem. Future Directions: In this article, we examine evidence for the therapeutic potential of targeting metabolic alterations in cancer, and we discuss the drawbacks and successes that have stimulated this field.


Asunto(s)
Antineoplásicos/farmacología , Redes y Vías Metabólicas/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Humanos , Neoplasias/patología
7.
ACS Appl Mater Interfaces ; 11(41): 37513-37523, 2019 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-31547654

RESUMEN

The goal of cancer immunotherapy is the selective killing of malignant cells by the cooperated efforts of immune cells at the primary and secondary sites. Here, we developed folic acid and secondary lymphoid tissue chemokine-loaded mesoporous silica-modified upconversion nanoparticle construct as a targeting, delivery, and imaging system to attract immune cells to folate receptor-expressing tumor cells. The effectiveness of the nanoparticles in targeting dendritic cells and T cells to the tumor compartment was tested in a vasculature-tumor interface model constructed from the co-culture of endothelial cells and ovarian cancer cells, in different interconnected channels in a microfluidic device. In comparison to the unconjugated nanoparticles, the folic acid-conjugated nanoparticles efficiently diffuse across the engineered blood vessel and specifically target the folate receptor-expressing ovarian cancer cells. The developed microfluidic platform was further used to demonstrate increased dendritic cell and T cell migration toward the ovarian cancer cell channel induced by the presence of the chemokine- and folic acid-loaded nanoparticles. The nanoparticle construct did not exhibit any significant cyto- and hemotoxicity. This proof of concept showed the potential of the nanoparticles to target cancer cells as well as to recruit dendritic cells and T cells to tumor sites to augment the weak host immune response.


Asunto(s)
Células Dendríticas/inmunología , Inmunomodulación , Inmunoterapia , Técnicas Analíticas Microfluídicas , Nanopartículas/química , Neovascularización Patológica , Neoplasias Ováricas , Linfocitos T/inmunología , Animales , Línea Celular Tumoral , Técnicas de Cocultivo , Células Dendríticas/patología , Femenino , Humanos , Ratones , Neovascularización Patológica/inmunología , Neovascularización Patológica/patología , Neovascularización Patológica/terapia , Neoplasias Ováricas/irrigación sanguínea , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Neoplasias Ováricas/terapia , Linfocitos T/patología
9.
Hepatology ; 61(3): 965-78, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25363290

RESUMEN

UNLABELLED: CCAAT enhancer binding protein α (C/EBPα) plays an essential role in cellular differentiation, growth, and energy metabolism. Here, we investigate the correlation between C/EBPα and hepatocellular carcinoma (HCC) patient outcomes and how C/EBPα protects cells against energy starvation. Expression of C/EBPα protein was increased in the majority of HCCs examined (191 pairs) compared with adjacent nontumor liver tissues in HCC tissue microarrays. Its upregulation was correlated significantly with poorer overall patient survival in both Kaplan-Meier survival (P=0.017) and multivariate Cox regression (P=0.028) analyses. Stable C/EBPα-silenced cells failed to establish xenograft tumors in nude mice due to extensive necrosis, consistent with increased necrosis in human C/EBPα-deficient HCC nodules. Expression of C/EBPα protected HCC cells in vitro from glucose and glutamine starvation-induced cell death through autophagy-involved lipid catabolism. Firstly, C/EBPα promoted lipid catabolism during starvation, while inhibition of fatty acid beta-oxidation significantly sensitized cell death. Secondly, autophagy was activated in C/EBPα-expressing cells, and the inhibition of autophagy by ATG7 knockdown or chloroquine treatment attenuated lipid catabolism and subsequently sensitized cell death. Finally, we identified TMEM166 as a key player in C/EBPα-mediated autophagy induction and protection against starvation. CONCLUSION: The C/EBPα gene is important in that it links HCC carcinogenesis to autophagy-mediated lipid metabolism and resistance to energy starvation; its expression in HCC predicts poorer patient prognosis.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/fisiología , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Adulto , Anciano , Animales , Autofagia , Carcinoma Hepatocelular/metabolismo , Muerte Celular , Línea Celular Tumoral , Humanos , Metabolismo de los Lípidos , Neoplasias Hepáticas/metabolismo , Masculino , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Pronóstico , Modelos de Riesgos Proporcionales
10.
Blood ; 124(19): 2973-82, 2014 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-25139349

RESUMEN

It is known that cells within the inflammatory background in classical Hodgkin lymphoma (cHL) provide signals essential for the continual survival of the neoplastic Hodgkin and Reed-Sternberg (HRS) cells. However, the mechanisms underlying the recruitment of this inflammatory infiltrate into the involved lymph nodes are less well understood. In this study, we show in vitro that HRS cells secrete lymphotoxin-α (LTα) which acts on endothelial cells to upregulate the expression of adhesion molecules that are important for T cell recruitment. LTα also enhances the expression of hyaluronan which preferentially contributes to the recruitment of CD4(+) CD45RA(+) naïve T cells under in vitro defined flow conditions. Enhanced expression of LTα in HRS cells and tissue stroma; and hyaluronan on endothelial cells are readily detected in involved lymph nodes from cHL patients. Our study also shows that although NF-κB and AP-1 are involved, the cyclooxygenase (COX) pathway is the dominant regulator of LTα production in HRS cells. Using pharmacological inhibitors, our data suggest that activity of COX1, but not of COX2, directly regulates the expression of nuclear c-Fos in HRS cells. Our findings suggest that HRS cell-derived LTα is an important mediator that contributes to T cell recruitment into lesional lymph nodes in cHL.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Comunicación Celular/inmunología , Células Endoteliales/citología , Enfermedad de Hodgkin/metabolismo , Linfotoxina-alfa/metabolismo , Células de Reed-Sternberg/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Moléculas de Adhesión Celular/inmunología , Moléculas de Adhesión Celular/metabolismo , Línea Celular , Ciclooxigenasa 2/inmunología , Ciclooxigenasa 2/metabolismo , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Enfermedad de Hodgkin/inmunología , Enfermedad de Hodgkin/patología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Receptores de Hialuranos/inmunología , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/inmunología , Ácido Hialurónico/metabolismo , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Linfotoxina-alfa/inmunología , Células de Reed-Sternberg/inmunología , Células de Reed-Sternberg/metabolismo
11.
Oncotarget ; 5(15): 5920-33, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25153718

RESUMEN

Triple-negative breast cancer (TNBC) is characterized by unique aggressive behavior and lack of targeted therapies. Among the various molecular subtypes of breast cancer, it was observed that TNBCs express elevated levels of sphingosine kinase 1 (SPHK1) compared to other breast tumor subtypes. High levels of SPHK1 gene expression correlated with poor overall and progression- free survival, as well as poor response to Doxorubicin-based treatment. Inhibition of SPHK1 was found to attenuate ERK1/2 and AKT signaling and reduce growth of TNBC cells in vitro and in a xenograft SCID mouse model. Moreover, SPHK1 inhibition by siRNA knockdown or treatment with SKI-5C sensitizes TNBCs to chemotherapeutic drugs. Our findings suggest that SPHK1 inhibition, which effectively counteracts oncogenic signaling through ERK1/2 and AKT pathways, is a potentially important anti-tumor strategy in TNBC. A combination of SPHK1 inhibitors with chemotherapeutic agents may be effective against this aggressive subtype of breast cancer.


Asunto(s)
Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Animales , Línea Celular Tumoral , Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Femenino , Xenoinjertos , Humanos , Células MCF-7 , Ratones , Ratones SCID , Transfección , Neoplasias de la Mama Triple Negativas/enzimología , Neoplasias de la Mama Triple Negativas/genética
12.
BMC Cell Biol ; 15: 15, 2014 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-24885150

RESUMEN

BACKGROUND: Fetal mesenchymal stem/stromal cells (MSC) represent a developmentally-advantageous cell type with translational potential.To enhance adult MSC migration, studies have focussed on the role of the chemokine receptor CXCR4 and its ligand SDF-1 (CXCL12), but more recent work implicates an intricate system of CXCR4 receptor dimerization, intracellular localization, multiple ligands, splice variants and nuclear accumulation. We investigated the intracellular localization of CXCR4 in fetal bone marrow-derived MSC and role of intracellular trafficking in CXCR4 surface expression and function. RESULTS: We found that up to 4% of human fetal MSC have detectable surface-localized CXCR4. In the majority of cells, CXCR4 is located not at the cell surface, as would be required for 'sensing' migratory cues, but intracellularly. CXCR4 was identified in early endosomes, recycling endosomes, and lysosomes, indicating only a small percentage of CXCR4 travelling to the plasma membrane. Notably CXCR4 was also found in and around the nucleus, as detected with an anti-CXCR4 antibody directed specifically against CXCR4 isoform 2 differing only in N-terminal sequence. After demonstrating that endocytosis of CXCR4 is largely independent of endogenously-produced SDF-1, we next applied the cytoskeletal inhibitors blebbistatin and dynasore to inhibit endocytotic recycling. These increased the number of cells expressing surface CXCR4 by 10 and 5 fold respectively, and enhanced the number of cells migrating to SDF1 in vitro (up to 2.6 fold). These molecules had a transient effect on cell morphology and adhesion, which abated after the removal of the inhibitors, and did not alter functional stem cell properties. CONCLUSIONS: We conclude that constitutive endocytosis is implicated in the regulation of CXCR4 membrane expression, and suggest a novel pharmacological strategy to enhance migration of systemically-transplanted cells.


Asunto(s)
Endocitosis , Feto/citología , Células Madre Mesenquimatosas/citología , Receptores CXCR4/análisis , Receptores CXCR4/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular , Movimiento Celular , Núcleo Celular/metabolismo , Células Cultivadas , Endosomas/metabolismo , Femenino , Humanos , Lisosomas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Transporte de Proteínas
13.
Cancer Discov ; 3(10): 1156-71, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23887393

RESUMEN

UNLABELLED: Although 3-phosphoinositide-dependent protein kinase-1 (PDK1) has been predominately linked to the phosphoinositide 3-kinase (PI3K)-AKT pathway, it may also evoke additional signaling outputs to promote tumorigenesis. Here, we report that PDK1 directly induces phosphorylation of Polo-like kinase 1 (PLK1), which in turn induces MYC phosphorylation and protein accumulation. We show that PDK1-PLK1-MYC signaling is critical for cancer cell growth and survival, and small-molecule inhibition of PDK1/PLK1 provides an effective approach for therapeutic targeting of MYC dependency. Intriguingly, PDK1-PLK1-MYC signaling induces an embryonic stem cell-like gene signature associated with aggressive tumor behaviors and is a robust signaling axis driving cancer stem cell (CSC) self-renewal. Finally, we show that a PLK1 inhibitor synergizes with an mTOR inhibitor to induce synergistic antitumor effects in colorectal cancer by antagonizing compensatory MYC induction. These findings identify a novel pathway in human cancer and CSC activation and provide a therapeutic strategy for targeting MYC-associated tumorigenesis and therapeutic resistance. SIGNIFICANCE: This work identifies PDK1­PLK1-MYC signaling as a new oncogenic pathway driving oncogenic transformation and CSC self-renewal. Targeted inhibition of PDK1/PLK1 is robust in targeting MYC dependency in cancer cells. Thus, our findings provide important insights into cancer and CSC biology and have significant therapeutic implications.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Proteínas de Ciclo Celular/metabolismo , Transformación Celular Neoplásica , Células Madre Neoplásicas/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/genética , Neoplasias Colorrectales/tratamiento farmacológico , Resistencia a Antineoplásicos , Humanos , Terapia Molecular Dirigida , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-myc/genética , Quinasa Tipo Polo 1
14.
Biomaterials ; 34(20): 4860-71, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23562047

RESUMEN

The ability of some malignant cells to evade immunosurveillance has been a major contribution to the inability of the host's immune system to eradicate the neoplastic cells. This has led to the development of various immunological strategies to augment the host immune response as part of cancer treatment. In this study, we developed folic acid (FA)/secondary lymphoid tissue chemokine (CCL21)/upconversion fluorescent nanoparticles (UCNs) conjugates as a targeting and delivery system to attract immune cells to folate receptor (FR) expressing tumor cells. Our data show that FA-conjugated UCNs@mesoporous silica specifically target FR expressing ovarian carcinoma cell line, OVCAR-3, compared to the unconjugated mesoporous silica coated UCNs. Furthermore, the FA-UCNs@mesoporous silica can efficiently cross the endothelial cell monolayer and accumulate in the clusters of OVCAR-3 cells in our endothelial-tumor cell bilayer model. Our migration assay data suggest that the CCL21 loaded into the mesoporous layer is biologically active and can efficiently induce T cells migration in-vitro. No significant cytotoxic effect was observed throughout the study indicating good biocompatibility of the nanoconjugates. As proof-of-concept, we have shown that it is feasible to load biologically active chemokines onto UCNs to modulate T cell migration.


Asunto(s)
Quimiocina CCL21/farmacología , Sistemas de Liberación de Medicamentos , Células Endoteliales/metabolismo , Receptor 1 de Folato/metabolismo , Ácido Fólico/farmacología , Nanopartículas/química , Neoplasias/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Femenino , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Modelos Biológicos , Nanopartículas/ultraestructura , Neoplasias/patología , Porosidad , Dióxido de Silicio/síntesis química , Dióxido de Silicio/química , Espectrometría de Fluorescencia
15.
PLoS One ; 7(8): e43594, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22927998

RESUMEN

Gelsolin is a cytoskeletal protein which participates in actin filament dynamics and promotes cell motility and plasticity. Although initially regarded as a tumor suppressor, gelsolin expression in certain tumors correlates with poor prognosis and therapy-resistance. In vitro, gelsolin has anti-apoptotic and pro-migratory functions and is critical for invasion of some types of tumor cells. We found that gelsolin was highly expressed at tumor borders infiltrating into adjacent liver tissues, as examined by immunohistochemistry. Although gelsolin contributes to lamellipodia formation in migrating cells, the mechanisms by which it induces tumor invasion are unclear. Gelsolin's influence on the invasive activity of colorectal cancer cells was investigated using overexpression and small interfering RNA knockdown. We show that gelsolin is required for invasion of colorectal cancer cells through matrigel. Microarray analysis and quantitative PCR indicate that gelsolin overexpression induces the upregulation of invasion-promoting genes in colorectal cancer cells, including the matrix-degrading urokinase-type plasminogen activator (uPA). Conversely, gelsolin knockdown reduces uPA levels, as well as uPA secretion. The enhanced invasiveness of gelsolin-overexpressing cells was attenuated by treatment with function-blocking antibodies to either uPA or its receptor uPAR, indicating that uPA/uPAR activity is crucial for gelsolin-dependent invasion. In summary, our data reveals novel functions of gelsolin in colorectal tumor cell invasion through its modulation of the uPA/uPAR cascade, with potentially important roles in colorectal tumor dissemination to metastatic sites.


Asunto(s)
Neoplasias Colorrectales/patología , Gelsolina/metabolismo , Transducción de Señal , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Línea Celular Tumoral , Fibrinolisina/metabolismo , Gelsolina/deficiencia , Gelsolina/genética , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Invasividad Neoplásica , ARN Interferente Pequeño/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo
17.
Cancer Cell ; 18(5): 459-71, 2010 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-21075311

RESUMEN

The PP2A serine/threonine protein phosphatase serves as a critical cellular regulator of cell growth, proliferation, and survival. However, how this pathway is altered in human cancer to confer growth advantage is largely unknown. Here, we show that PPP2R2B, encoding the B55ß regulatory subunit of the PP2A complex, is epigenetically inactivated by DNA hypermethylation in colorectal cancer. B55ß-associated PP2A interacts with PDK1 and modulates its activity toward Myc phosphorylation. On loss of PPP2R2B, mTORC1 inhibitor rapamycin triggers a compensatory Myc phosphorylation in PDK1-dependent, but PI3K and AKT-independent manner, resulting in resistance. Reexpression of PPP2R2B, genetic ablation of PDK1 or pharmacologic inhibition of PDK1 abrogates the rapamycin-induced Myc phosphorylation, leading to rapamycin sensitization. Thus, PP2A-B55ß antagonizes PDK1-Myc signaling and modulates rapamycin sensitivity.


Asunto(s)
Antibióticos Antineoplásicos/uso terapéutico , Neoplasias Colorrectales/enzimología , Proteínas del Tejido Nervioso/metabolismo , Proteína Fosfatasa 2/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Sirolimus/uso terapéutico , Animales , Proliferación Celular , Transformación Celular Neoplásica/genética , Senescencia Celular , Fosfatidilinositol 3-Quinasa Clase I , Análisis por Conglomerados , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Metilación de ADN , Resistencia a Antineoplásicos , Epigénesis Genética , Humanos , Ratones , Proteínas del Tejido Nervioso/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositol 3-Quinasas/fisiología , Fosforilación , Proteína Fosfatasa 2/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-akt/fisiología , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Transducción de Señal , Trasplante Heterólogo
18.
Int J Oncol ; 37(4): 909-26, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20811713

RESUMEN

Palladin is a scaffold protein involved in the formation of actin-associated protein complexes. Gene expression array analysis on the poorly metastatic HCT116 colon cancer cell line and a metastatic derivative cell line (E1) with EMT (epithelial-mesenchymal transition) features showed a down-regulation of palladin gene expression in the latter. Knockdown of palladin expression in the HCT116 cells suppressed junctional localization of E-cadherin, reduced intercellular adhesion and collective cell migration, showing that palladin plays an important role in maintaining the integrity of adherens junctions. The acquisition of the EMT features by the E1 cell line was dependent on the Erk pathway. Inhibition of this pathway by U0126 treatment in E1 cells resulted in the re-expression of palladin, relocalization of E-cadherin to the adherens junctions and a reversal of EMT features. The re-establishment of intercellular adhesion was dependent on palladin expression. The down-regulation of palladin was also observed in poorly-differentiated tumor tubules and dissociated tumor cells that have undergone de-differentiation in human primary colon tumors. Our data show that palladin is an integral component of adherens junctions and plays a role in the localization of E-cadherin to the junctions. The loss of palladin may be an integral part of EMT, an early step in the metastatic spread of colon carcinoma.


Asunto(s)
Uniones Adherentes/metabolismo , Adhesión Celular , Movimiento Celular , Neoplasias Colorrectales/metabolismo , Proteínas del Citoesqueleto/metabolismo , Fosfoproteínas/metabolismo , Uniones Adherentes/efectos de los fármacos , Uniones Adherentes/patología , Animales , Antígenos CD , Cadherinas/metabolismo , Adhesión Celular/efectos de los fármacos , Desdiferenciación Celular , Movimiento Celular/efectos de los fármacos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Proteínas del Citoesqueleto/genética , Transición Epitelial-Mesenquimal , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundario , Ratones , Ratones Desnudos , Invasividad Neoplásica , Fenotipo , Fosfoproteínas/genética , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , Neoplasias del Bazo/metabolismo , Neoplasias del Bazo/secundario , Factores de Tiempo , Transfección
19.
J Immunol ; 184(9): 5160-71, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20335533

RESUMEN

Burkholderia pseudomallei is a Gram-negative saprophyte that is the causative agent of melioidosis, a severe infectious disease endemic in Northern Australia and Southeast Asia. This organism has sparked much scientific interest in the West because of its classification as a potential bioterrorism agent by the U.S. Centers for Disease Control and Prevention. However, relatively little is known about its pathogenesis. We demonstrate that B. pseudomallei actively inhibits NF-kappaB and type I IFN pathway activation, thereby downregulating host inflammatory responses. We found the virulence factor TssM to be responsible for this activity. TssM interferes with the ubiquitination of critical signaling intermediates, including TNFR-associated factor-3, TNFR-associated factor-6, and IkappaBalpha. The expression but not secretion of TssM is regulated by the type III secretion system. We demonstrate that TssM is important for B. pseudomallei infection in vivo as inflammation in the tssM mutant-infected mice is more severe and corresponds to a more rapid death compared with wild-type bacteria-infected mice. Abs to TssM can be detected in the sera of melioidosis patients, indicating that TssM is functionally expressed in vivo and thus could contribute to bacterial pathogenesis in human melioidosis.


Asunto(s)
Proteínas Bacterianas/fisiología , Burkholderia pseudomallei/inmunología , Endopeptidasas/fisiología , Inmunidad Innata , Terapia de Inmunosupresión , Factores de Virulencia/fisiología , Enfermedad Aguda , Animales , Línea Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/antagonistas & inhibidores , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/fisiología , Melioidosis/inmunología , Melioidosis/microbiología , Ratones , Ratones Endogámicos BALB C , FN-kappa B/antagonistas & inhibidores , FN-kappa B/fisiología , Transducción de Señal/inmunología , Proteasas Ubiquitina-Específicas
20.
Immunology ; 128(1 Suppl): e881-94, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19740349

RESUMEN

Fve is a fungal protein isolated from the golden needle mushroom Flammulina velutipes and has previously been reported to trigger immunological responses in both mouse and human lymphocytes. In this study, we evaluated the potential application of Fve as an adjuvant for tumour immunotherapy and examined the underlying mechanism(s). When the human papillomavirus (HPV)-16 E7 oncoprotein was used as a model antigen, mice coimmunized with HPV-16 E7 and Fve showed enhanced production of HPV-16 E7-specific antibodies as well as expansion of HPV-16 E7-specific interferon (IFN)-gamma-producing CD4(+) and CD8(+) T cells as compared with mice immunized with HPV-16 E7 alone. Tumour protection assays showed that 60% of mice coimmunized with HPV-16 E7 plus Fve, as compared with 20% of those immunized only with HPV-16 E7, remained tumour-free for up to 167 days after challenge with the tumour cells. Tumour therapeutic assays showed that HPV-16 E7 plus Fve treatment significantly prolonged the survival of tumour-bearing mice as compared with those treated only with HPV-16 E7. In vivo cell depletion and adoptive T-cell transfer assays showed that CD4(+) and CD8(+) T cells and IFN-gamma played critical roles in conferring the antitumour effects. Interestingly, Fve could stimulate the maturation of splenic dendritic cells in vivo and induce antigen-specific CD8(+) T-cell immune responses. In summary, Fve has potent adjuvant properties that enhance T helper type 1 antigen-specific humoral and cellular immune responses which confer strong antitumour effects. The use of Fve as an adjuvant could be an attractive alternative to the current vaccination strategy for cancer immunotherapy.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Flammulina/inmunología , Proteínas Fúngicas/administración & dosificación , Papillomavirus Humano 16 , Inmunoterapia Activa/métodos , Proteínas Oncogénicas Virales/administración & dosificación , Infecciones por Papillomavirus/terapia , Neoplasias del Cuello Uterino/terapia , Adyuvantes Inmunológicos/uso terapéutico , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Células Dendríticas/inmunología , Femenino , Proteínas Fúngicas/inmunología , Proteínas Fúngicas/uso terapéutico , Humanos , Interferón gamma/biosíntesis , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Proteínas Oncogénicas Virales/inmunología , Proteínas Oncogénicas Virales/uso terapéutico , Proteínas E7 de Papillomavirus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...