Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Cancer Res ; 21(12): 1288-1302, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37584671

RESUMEN

Constraints on the p53 tumor suppressor pathway have long been associated with the progression, therapeutic resistance, and poor prognosis of melanoma, the most aggressive form of skin cancer. Likewise, the insulin-like growth factor type 1 receptor (IGF1R) is recognized as an essential coordinator of transformation, proliferation, survival, and migration of melanoma cells. Given that ß-arrestin (ß-arr) system critically governs the anti/pro-tumorigenic p53/IGF1R signaling pathways through their common E3 ubiquitin-protein ligase MDM2, we explore whether unbalancing this system downstream of IGF1R can enhance the response of melanoma cells to chemotherapy. Altering ß-arr expression demonstrated that both ß-arr1-silencing and ß-arr2-overexpression (-ß-arr1/+ß-arr2) facilitated nuclear-to-cytosolic MDM2 translocation accompanied by decreased IGF1R expression, while increasing p53 levels, resulting in reduced cell proliferation/survival. Imbalance towards ß-arr2 (-ß-arr1/+ß-arr2) synergizes with the chemotherapeutic agent, dacarbazine, in promoting melanoma cell toxicity. In both 3D spheroid models and in vivo in zebrafish models, this combination strategy, through dual IGF1R downregulation/p53 activation, limits melanoma cell growth, survival and metastatic spread. In clinical settings, analysis of the TCGA-SKCM patient cohort confirms ß-arr1-/ß-arr2+ imbalance as a metastatic melanoma vulnerability that may enhance therapeutic benefit. Our findings suggest that under steady-state conditions, IGF1R/p53-tumor promotion/suppression status-quo is preserved by ß-arr1/2 homeostasis. Biasing this balance towards ß-arr2 can limit the protumorigenic IGF1R activities while enhancing p53 activity, thus reducing multiple cancer-sustaining mechanisms. Combined with other therapeutics, this strategy improves patient responses and outcomes to therapies relying on p53 or IGF1R pathways. IMPLICATIONS: Altogether, ß-arrestin system bias downstream IGF1R is an important metastatic melanoma vulnerability that may be conductive for therapeutic benefit.


Asunto(s)
Arrestinas , Melanoma , Animales , Humanos , beta-Arrestinas/metabolismo , Arrestinas/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Pez Cebra/metabolismo , beta-Arrestina 1/metabolismo , Isoformas de Proteínas/metabolismo , Melanoma/tratamiento farmacológico , Melanoma/genética , Arrestina beta 2/metabolismo , Línea Celular Tumoral , Receptor IGF Tipo 1/metabolismo
2.
iScience ; 26(8): 107412, 2023 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-37575190

RESUMEN

The human cytomegalovirus (HCMV)-encoded chemokine receptor US28 contributes to various aspects of the viral life cycle and promotes immune evasion by scavenging chemokines from the microenvironment of HCMV-infected cells. In contrast to the plasma membrane localization of most human chemokine receptors, US28 has a predominant intracellular localization. In this study, we used immunofluorescence and electron microscopy to determine the localization of US28 upon exogenous expression, as well as in HCMV-infected cells. We observed that US28 localizes to late endosomal compartments called multivesicular bodies (MVBs), where it is sorted in intraluminal vesicles. Live-cell total internal reflection fluorescence (TIRF) microscopy revealed that US28-containing MVBs can fuse with the plasma membrane, resulting in the secretion of US28 on exosomes. Exosomal US28 binds the chemokines CX3CL1 and CCL5, and US28-containing exosomes inhibited the CX3CL1-CX3CR1 signaling axis. These findings suggest that exosomal release of US28 contributes to chemokine scavenging and immune evasion by HCMV.

4.
Oncogene ; 41(4): 600-611, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34785779

RESUMEN

As the p53 tumor suppressor is rarely mutated in conjunctival melanoma (CM), we investigated its activation as a potential therapeutic strategy. Preventing p53/Mdm2 interaction by Nutlin-3, the prototypical Mdm2 antagonist, or via direct siRNA Mdm2 depletion, increased p53 and inhibited viability in CM cell lines. The sensitivity to Nutlin-3 p53 reactivation with concomitant Mdm2 stabilization was higher than that achieved by siRNA, indicative of effects on alternative Mdm2 targets, identified as the cancer-protective IGF-1R. Nutlin-3 treatment increased the association between IGF-1R and ß-arrestin1, the adaptor protein that brings Mdm2 to the IGF-1R, initiating receptor degradation in a ligand-dependent manner. Controlled expression of ß-arrestin1 augmented inhibitory Nutlin-3 effects on CM survival through enhanced IGF-1R degradation. Yet, the effect of IGF-1R downregulation on cell proliferation is balanced by ß-arrestin1-induced p53 inhibition. As mitomycin (MMC) is a well-established adjuvant treatment for CM, and it triggers p53 activation through genotoxic stress, we evaluated how these alternative p53-targeting strategies alter the cancer-relevant bioactivities of CM. In 2D and 3D in vitro models, Nutlin-3 or MMC alone, or in combination, reduces the overall cell tumor growth ~30%, with double treatment inhibition rate only marginally higher than single-drug regimens. However, histopathological evaluation of the 3D models revealed that Nutlin-3 was the most effective, causing necrotic areas inside spheroids and complete loss of nuclear staining for the proliferative marker Ki67. These findings were further validated in vivo; zebrafish xenografts demonstrate that Nutlin-3 alone has higher efficacy in restraining CM tumor cell growth and preventing metastasis. Combined, these results reveal that ß-arrestin1 directs Mdm2 toward different substrates, thus balancing IGF-1R pro-tumorigenic and p53-tumor suppressive signals. This study defines a potent dual-hit strategy: simultaneous control of a tumor-promoter (IGF-1R) and tumor-suppressor (p53), which ultimately mitigates recurrent and metastatic potential, thus opening up targeted therapy to CM.


Asunto(s)
Neoplasias de la Conjuntiva/genética , Melanoma/genética , Receptor IGF Tipo 1/metabolismo , Proteína p53 Supresora de Tumor/genética , Animales , Neoplasias de la Conjuntiva/patología , Humanos , Masculino , Melanoma/patología , Ratones , Transfección
5.
Neoplasia ; 23(1): 149-157, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33321449

RESUMEN

The discovery that cancer cells discharge vast quantities of extracellular vesicles (EVs), underscored the explosion of the EV field. A large body of evidence now supports their onco-functionality in an array of contexts; stromal crosstalk, immune evasion, metastatic site priming, and drug resistance - justifying therapeutic intervention. The current bottleneck is a lack of clear understanding of why and how EV biogenesis ramps up in cancer cells, and hence where exactly avenues for intervention may reside. We know that EVs also play an array of physiological roles, therefore effective anticancer inhibition requires a target distinct enough from physiology to achieve efficacy. Taking the perspective that EV upregulation may be a consequence of the tumor landscape, we examine classic mutational events and tumor characteristics for EV regulators. All the while, aiming to illuminate topics worth further research in therapeutic development.


Asunto(s)
Vesículas Extracelulares/metabolismo , Neoplasias/metabolismo , Biomarcadores , Comunicación Celular , Susceptibilidad a Enfermedades , Metabolismo Energético , Exosomas/metabolismo , Regulación de la Expresión Génica , Humanos , Neoplasias/etiología , Neoplasias/patología , Oncogenes , Transducción de Señal , Microambiente Tumoral
6.
Cancer Res ; 81(2): 501-514, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33158816

RESUMEN

The ability of a receptor to preferentially activate only a subset of available downstream signal cascades is termed biased signaling. Although comprehensively recognized for the G protein-coupled receptors (GPCR), this process is scarcely explored downstream of receptor tyrosine kinases (RTK), including the cancer-relevant insulin-like growth factor-1 receptor (IGF1R). Successful IGF1R targeting requires receptor downregulation, yet therapy-mediated removal from the cell surface activates cancer-protective ß-arrestin-biased signaling (ß-arr-BS). As these overlapping processes are initiated by the ß-arr/IGF1R interaction and controlled by GPCR-kinases (GRK), we explored GRKs as potential anticancer therapeutic targets to disconnect IGF1R downregulation and ß-arr-BS. Transgenic modulation demonstrated that GRK2 inhibition or GRK6 overexpression enhanced degradation of IGF1R, but both scenarios sustained IGF1-induced ß-arr-BS. Pharmacologic inhibition of GRK2 by the clinically approved antidepressant, serotonin reuptake inhibitor paroxetine (PX), recapitulated the effects of GRK2 silencing with dose- and time-dependent IGF1R downregulation without associated ß-arr-BS. In vivo, PX treatment caused substantial downregulation of IGF1R, suppressing the growth of Ewing's sarcoma xenografts. Functional studies reveal that PX exploits the antagonism between ß-arrestin isoforms; in low ligand conditions, PX favored ß-arrestin1/Mdm2-mediated ubiquitination/degradation of IGF1R, a scenario usually exclusive to ligand abundancy, making PX more effective than antibody-mediated IGF1R downregulation. This study provides the rationale, molecular mechanism, and validation of a clinically feasible concept for "system bias" targeting of the IGF1R to uncouple downregulation from signaling. Demonstrating system bias as an effective anticancer approach, our study reveals a novel strategy for the rational design or repurposing of therapeutics to selectively cross-target the IGF1R or other RTK. SIGNIFICANCE: This work provides insight into the molecular and biological roles of biased signaling downstream RTK and provides a novel "system bias" strategy to increase the efficacy of anti-IGF1R-targeted therapy in cancer.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Óseas/patología , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Quinasas de Receptores Acoplados a Proteína-G/metabolismo , Regulación Neoplásica de la Expresión Génica , Receptor IGF Tipo 1/metabolismo , Sarcoma de Ewing/patología , Animales , Apoptosis , Biomarcadores de Tumor/genética , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Proliferación Celular , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Quinasas de Receptores Acoplados a Proteína-G/genética , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Ratones , Ratones Desnudos , Fosforilación , Receptor IGF Tipo 1/genética , Sarcoma de Ewing/genética , Sarcoma de Ewing/metabolismo , Células Tumorales Cultivadas , Ubiquitinación , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Trends Pharmacol Sci ; 41(9): 627-640, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32711926

RESUMEN

Transmembrane receptors, of which G protein-coupled receptors (GPCRs) constitute the largest group, typically act as cellular antennae that reside at the plasma membrane (PM) to collect and interpret information from the extracellular environment. The discovery of cell-released extracellular vesicles (EVs) has added a new dimension to intercellular communication. These unique nanocarriers reflect cellular topology and can systemically transport functionally competent transmembrane receptors, ligands, and a cargo of signal proteins. Recent developments hint at roles for GPCRs in the EV life cycle and, conversely, at roles for EVs in GPCR signal transduction. We highlight key points of convergence, discuss their relevance to current GPCR and EV paradigms, and speculate on how this intersection could lend itself to future therapeutic avenues.


Asunto(s)
Vesículas Extracelulares , Biología , Comunicación Celular , Receptores Acoplados a Proteínas G , Transducción de Señal
8.
Sci Signal ; 13(633)2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32457114

RESUMEN

The C-terminal tail of insulin-like growth factor 1 receptor (IGF-1R) has long been appreciated to drive much of this receptor's oncogenic power. In this issue of Science Signaling, Rieger et al. have shown that Tyr1250 and Tyr1251 of IGF-1R are autophosphorylated in a cell adhesion-dependent manner, uncovering a previously unknown plasma membrane-Golgi trafficking route for IGF-1R in migratory cells, an integral part of the malignant phenotype.


Asunto(s)
Neoplasias , Receptor IGF Tipo 1 , Movimiento Celular , Neoplasias/genética , Oncogenes , Fosforilación , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Transducción de Señal
9.
Cells ; 8(10)2019 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-31600876

RESUMEN

Ligand-activated plasma membrane receptors follow pathways of endocytosis through the endosomal sorting apparatus. Receptors cluster in clathrin-coated pits that bud inwards and enter the cell as clathrin-coated vesicles. These vesicles travel through the acidic endosome whereby receptors and ligands are sorted to be either recycled or degraded. The traditional paradigm postulated that the endocytosis role lay in signal termination through the removal of the receptor from the cell surface. It is now becoming clear that the internalization process governs more than receptor signal cessation and instead reigns over the entire spatial and temporal wiring of receptor signaling. Governing the localization, the post-translational modifications, and the scaffolding of receptors and downstream signal components established the endosomal platform as the master regulator of receptor function. Confinement of components within or between distinct organelles means that the endosome instructs the cell on how to interpret and translate the signal emanating from any given receptor complex into biological effects. This review explores this emerging paradigm with respect to the cancer-relevant insulin-like growth factor type 1 receptor (IGF-1R) and discusses how this perspective could inform future targeting strategies.


Asunto(s)
Neoplasias/metabolismo , Receptor IGF Tipo 1/metabolismo , Membrana Celular/metabolismo , Endocitosis , Humanos , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Procesamiento Proteico-Postraduccional , Transducción de Señal
10.
Int Rev Cell Mol Biol ; 339: 1-40, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29776602

RESUMEN

Receptor tyrosine kinases (RTKs) such as the insulin-like growth factor type 1 receptor (IGF-1R) control important biological activities as well as being involved in pathological processes. Due to their supportive nature in many human cancers they have long been considered attractive therapeutic targets. However, lessons learnt from early targeting trials highlight that a simple "active versus inactive" state model with classical kinase-only signaling is overly simplistic and does not describe reality. A vast amount of evidence exists disproving this model and hence provides a rational explanation for failure of many targeting agents designed under such a paradigm. In addition, substantial evidence exists that the IGF-1R and other RTKs make direct use of the G protein-coupled receptor (GPCR) components G proteins, GRKs, and ß-arrestins, outside of their traditional receptor family frame. In this chapter we review the evidence that RTKs can undertake a wide range of active conformations, capable of distinct downstream signal cascades and propose an RTK/GPCR functional hybrid model, while discussing the implications of such an update on therapeutic drug development pipelines.


Asunto(s)
Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Quinasas de Receptores Acoplados a Proteína-G/metabolismo , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Activación Transcripcional/genética , beta-Arrestinas/metabolismo
11.
J Invest Dermatol ; 138(4): 882-892, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29104160

RESUMEN

Melanoma is one of the deadliest human cancers with limited therapeutic options. MicroRNAs are a class of short noncoding RNAs regulating gene expression at the post-transcriptional level. To identify important miRNAs in melanoma, we compared the miRNome of primary and metastatic melanomas in The Cancer Genome Atlas dataset and found lower miR-203 abundance in metastatic melanoma. Lower level of miR-203 was associated with poor overall survival in metastatic disease. We found that the methylation levels of several CpGs in the MIR203 promoter negatively correlated with miR-203 expression and that treatment with the demethylating agent 5-aza-2-deoxycytidine induced miR-203 expression, which was associated with demethylation of the promoter CpGs, in melanoma cell lines. In vitro, there was a decreased expression of miR-203 in melanoma cell lines in comparison with primary melanocytes. Ectopic overexpression of miR-203 suppressed cell motility, colony formation, and sphere formation as well as the angiogenesis-inducing capacity of melanoma cells. In vivo, miR-203 inhibited xenograft tumor growth and reduced lymph node and lung metastasis. SLUG was shown as a target of miR-203, and knockdown of SLUG recapitulated the effects of miR-203, whereas its restoration was able to reverse the miR-203-mediated suppression of cell motility. These results establish a role for miR-203 as a tumor suppressor in melanoma which suppresses both early and late steps of metastasis. Hence, restoration of miR-203 has therapeutic potential in melanoma.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Melanoma/genética , MicroARNs/genética , ARN Neoplásico/genética , Neoplasias Cutáneas/genética , Línea Celular Tumoral , Proliferación Celular , Metilación de ADN , Estudio de Asociación del Genoma Completo , Humanos , Melanoma/metabolismo , Melanoma/secundario , MicroARNs/biosíntesis , Regiones Promotoras Genéticas , Transducción de Señal , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología
12.
Oncotarget ; 8(47): 82256-82267, 2017 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-29137261

RESUMEN

Due to its ability to compensate for signals lost following therapeutic MAPK-inhibition, insulin-like growth factor type 1 receptor (IGF-1R) co-targeting is a rational approach for melanoma treatment. However IGF-1R conformational changes associated with its inhibition can preferentially activate MAPK-pathway in a kinase-independent manner, through a process known as biased signaling. We explored the impact of biased IGF-1R signaling, on response to MAPK inhibition in a panel of skin melanoma cell lines with differing MAPK and p53 mutation statuses. Specific siRNA towards IGF-1R down-regulates the receptor and all its signaling in a balanced manner, whilst IGF-1R targeting by small molecule Nutlin-3 parallels receptor degradation with a transient biased pERK1/2 activity, with both strategies synergizing with MEK1/2 inhibition. On the other hand, IGF-1R down-regulation by a targeted antibody (Figitumumab) induces a biased receptor conformation, preserved even when the receptor is exposed to the balanced natural ligand IGF-1. This process sustains MAPK activity and competes with the MEK1/2 inhibition. Our results indicate that IGF-1R down-regulation offers an approach to increase the sensitivity of melanoma cells to MAPK inhibition, and highlights that controlling biased signaling could provide greater specificity and precision required for multi-hit therapy.

13.
Artículo en Inglés | MEDLINE | ID: mdl-25964779

RESUMEN

The insulin-like growth factor type 1 receptor (IGF-1R) plays a key role in the development and maintenance of cancer. Since the first links between growth factor receptors and oncogenes were noted over three decades ago, targeting the IGF-1R has been of great interest. This review follows the progress from inception through intense pharmaceutical development, disappointing clinical trials and recent updates to the signaling paradigm. In light of major developments in signaling understanding and activation complexities, we examine reasons for failure of first line targeting approaches. Recent findings include the fact that the IGF-1R can signal in the absence of the ligand, in the absence of kinase activity, and utilizes components of the GPCR system. With recognition of the unappreciated complexities that this first wave of targeting approaches encountered, we advocate re-recognition of IGF-1R as a valid target for cancer treatment and look to future directions, where both research and pharmaceutical strengths can lend themselves to finally unearthing anti-IGF-1R potential.

14.
Growth Horm IGF Res ; 25(1): 2-12, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25466906

RESUMEN

The prime position of the insulin-like growth factor 1 receptor (IGF-1R), at the head of the principle mitogenic and anti-apoptotic signalling cascades, along with the resilience to transformation of IGF-1R deficient cells fuelled great excitement for its anti-cancer targeting. Yet its potential has not been fulfilled, as clinical trial results fell far short of expectations. Advancements in understanding of other receptors' function have now begun to shed light on this incongruity, with the now apparent parallels highlighting the immaturity of our understanding of IGF-1R biology, with the model used for drug development now recognised as having been too simplistic. Gathering together the many advancements of the field of IGF-1R research over the past decade, alongside those in the GPCR field, advocates for a major paradigm shift in our appreciation of the subtle workings of this receptor. This review will emphasise the updating of the IGF-1R's classification from an RTK, to an RTK/GPCR functional hybrid, which integrates both canonical kinase signalling with many functions characteristic of a GPCR. Recognition of the shortcomings of IGF-1R inhibitor drug development programs and the models used not only allows us to reignite the initial interest in the IGF-1R as an anti-cancer therapeutic target, but also points to the possibility of biased ligand therapeutics, which together may hold a very powerful key to unlocking the true potential of IGF-1R modulation.


Asunto(s)
Neoplasias/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor IGF Tipo 1/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Humanos , Ratones , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Receptor IGF Tipo 1/clasificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...