Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(18): e2216587120, 2023 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-37098069

RESUMEN

Innate lymphoid cells (ILCs) play a key role in tissue-mediated immunity and can be controlled by coreceptor signaling. Here, we define a subset of ILCs that are Tbet+NK1.1- and are present within the tumor microenvironment (TME). We show programmed death-1 receptor (PD-1) expression on ILCs within TME is found in Tbet+NK1.1- ILCs. PD-1 significantly controlled the proliferation and function of Tbet+NK1.1- ILCs in multiple murine and human tumors. We found tumor-derived lactate enhanced PD-1 expression on Tbet+NK1.1- ILCs within the TME, which resulted in dampened the mammalian target of rapamycin (mTOR) signaling along with increased fatty acid uptake. In line with these metabolic changes, PD-1-deficient Tbet+NK1.1- ILCs expressed significantly increased IFNγ and granzyme B and K. Furthermore, PD-1-deficient Tbet+NK1.1- ILCs contributed toward diminished tumor growth in an experimental murine model of melanoma. These data demonstrate that PD-1 can regulate antitumor responses of Tbet+NK1.1- ILCs within the TME.


Asunto(s)
Linfocitos , Neoplasias , Ratones , Animales , Humanos , Inmunidad Innata , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo , Microambiente Tumoral , Neoplasias/metabolismo , Apoptosis , Mamíferos/metabolismo
2.
Br J Dermatol ; 190(4): 549-558, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38006317

RESUMEN

BACKGROUND: Combined expression of the autophagy-regulatory protein AMBRA1 (activating molecule in Beclin1-regulated autophagy) and the terminal differentiation marker loricrin in the peritumoral epidermis of stage I melanomas can identify tumour subsets at low risk of -metastasis. OBJECTIVES: To validate the combined expression of peritumoral AMBRA1 and loricrin (AMBLor) as a prognostic biomarker able to identify both stage I and II melanomas at low risk of tumour recurrence. METHODS: Automated immunohistochemistry was used to analyse peritumoral AMBRA1 and loricrin expression in geographically distinct discovery (n = 540) and validation (n = 300) cohorts of nonulcerated American Joint Committee on Cancer (AJCC) stage I and II melanomas. AMBLor status was correlated with clinical outcomes in the discovery and validation cohorts separately and combined. RESULTS: Analysis of AMBLor in the discovery cohort revealed a recurrence-free survival (RFS) rate of 95.5% in the AMBLor low-risk group vs. 81.7% in the AMBLor at-risk group (multivariate log-rank, P < 0.001) and a negative predictive value (NPV) of 96.0%. In the validation cohort, AMBLor analysis revealed a RFS rate of 97.6% in the AMBLor low-risk group vs. 78.3% in the at-risk group (multivariate log-rank, P < 0.001) and a NPV of 97.6%. In a multivariate model considering AMBLor, Breslow thickness, age and sex, analysis of the combined discovery and validation cohorts showed that the estimated effect of AMBLor was statistically significant, with a hazard ratio of 3.469 (95% confidence interval 1.403-8.580, P = 0.007) and an overall NPV of 96.5%. CONCLUSIONS: These data provide further evidence validating AMBLor as a prognostic biomarker to identify nonulcerated AJCC stage I and II melanoma tumours at low risk of disease recurrence.


Asunto(s)
Melanoma , Proteínas de la Membrana , Neoplasias Cutáneas , Humanos , Estados Unidos , Melanoma/patología , Pronóstico , Recurrencia Local de Neoplasia/patología , Epidermis/metabolismo , Biomarcadores , Estadificación de Neoplasias , Proteínas Adaptadoras Transductoras de Señales/metabolismo
3.
Wound Repair Regen ; 32(4): 475-486, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38572659

RESUMEN

Chronic non-healing cutaneous wounds represent a major burden to patients and healthcare providers worldwide, emphasising the continued unmet need for credible and efficacious therapeutic approaches for wound healing. We have recently shown the potential for collagen peptides to promote proliferation and migration during cutaneous wound healing. In the present study, we demonstrate that the application of porcine-derived collagen peptides significantly increases keratinocyte and dermal fibroblast expression of integrin α2ß1 and activation of an extracellular signal-related kinase (ERK)-focal adhesion kinase (FAK) signalling cascade during wound closure in vitro. SiRNA-mediated knockdown of integrin ß1 impaired porcine-derived collagen peptide-induced wound closure and activation of ERK-FAK signalling in keratinocytes but did not impair ERK or FAK signalling in dermal fibroblasts, implying the activation of differing downstream signalling pathways. Studies in ex vivo human 3D skin equivalents subjected to punch biopsy-induced wounding confirmed the ability of porcine-derived collagen peptides to promote wound closure by enhancing re-epithelialisation. Collectively, these data highlight the translational and clinical potential for porcine-derived collagen peptides as a viable therapeutic approach to promote re-epithelialisation of superficial cutaneous wounds.


Asunto(s)
Colágeno , Fibroblastos , Queratinocitos , Repitelización , Transducción de Señal , Cicatrización de Heridas , Animales , Humanos , Porcinos , Colágeno/metabolismo , Colágeno/farmacología , Queratinocitos/metabolismo , Repitelización/efectos de los fármacos , Fibroblastos/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/fisiología , Integrina alfa2beta1/metabolismo , Proliferación Celular , Células Cultivadas , Movimiento Celular , Piel/lesiones , Piel/metabolismo , Péptidos/farmacología
4.
J Cell Biochem ; 116(3): 438-49, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25336069

RESUMEN

Glucose-regulated protein 78 (GRP78) is a stress sensor which interacts with unfolded protein response (UPR) activators in the endoplasmic reticulum (ER). The aim of this study was to test the hypothesis that GRP78 has distinct functional roles in mediating the effects of ER stress in neuroblastoma compared to other neuroectodermal cancer types. GRP78 was knocked down or overexpressed in neuroectodermal tumor cell lines. Protein and transcript expression were measured using Western blotting, confocal microscopy, and real-time polymerase chain reaction; cell stress was assessed by measurement of oxidative stress and accumulation of ubiquitinated proteins and cell response by measurement of apoptosis and cell viability. Neuroblastoma cells were more sensitive to ER stress than melanoma and glioblastoma cells. GRP78 knockdown increased stress sensitivity of melanoma and glioblastoma cells, but not neuroblastoma cells. Over-expression of GRP78 decreased the stress sensitivity of melanoma and glioblastoma cells but, in contrast, increased the stress sensitivity of neuroblastoma cells by activation of caspase-3-independent cell death and substantially increased the expression of UPR activators, particularly inositol-requiring element 1 (IRE1). The results from this study suggest that cell-type specific differences in the relationships between GRP78 and the UPR activators, particularly IRE1, may determine differential sensitivity to ER stress.


Asunto(s)
Proteínas de Choque Térmico/metabolismo , Placa Neural/citología , Placa Neural/metabolismo , Estrés Fisiológico , Biomarcadores/metabolismo , Ácidos Borónicos/farmacología , Bortezomib , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Forma de la Célula/efectos de los fármacos , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Fenretinida/farmacología , Técnicas de Silenciamiento del Gen , Humanos , Pirazinas/farmacología , ARN Interferente Pequeño/metabolismo , Estrés Fisiológico/efectos de los fármacos , Respuesta de Proteína Desplegada/efectos de los fármacos
5.
Exp Dermatol ; 22(11): 767-9, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24118207

RESUMEN

The Bcl-2 family member Mcl-1 is essential for melanoma survival; however, the influence of oncogenic BRAF signalling remains elusive. In this study, Mcl-1 splice variant expression was determined in a panel of melanoma cell lines in relation to BRAF mutational status. Mcl-1L mRNA expression was increased in melanoma cells compared with primary melanocytes with significantly increased mRNA and protein expression observed in BRAF(V600E) mutant melanoma cells. Although no change in Mcl-1S mRNA was observed, Mcl-1S protein expression also increased in BRAF mutant melanoma cells. Additionally, while over-expression of mutant BRAF(V600E) increased both Mcl-1L and Mcl-1S expression, inhibition of hyperactive BRAF signalling resulted in decreased Mcl-1L expression. These studies suggest that the regulation of Mcl-1 expression by BRAF signalling is increased by oncogenic activation of BRAF, revealing a mechanism of apoptotic resistance which may be overcome by the use of more specifically targeted Mcl-1 inhibitors.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Melanoma/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Proteínas Proto-Oncogénicas B-raf/metabolismo , Transducción de Señal , Neoplasias Cutáneas/metabolismo , Apoptosis , Línea Celular Tumoral , Humanos , Melanocitos/metabolismo , Melanoma/genética , Mutación , Neoplasias Cutáneas/genética , Melanoma Cutáneo Maligno
6.
PLoS One ; 17(4): e0265048, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35377887

RESUMEN

OBJECTIVES: Cutaneous melanoma rates are steadily increasing. Up to 20% of patients diagnosed with AJCC Stage I/II melanomas will develop metastatic disease. To date there are no consistently reliable means to accurately identify truly high versus low-risk patient subpopulations. There is hence an urgent need for more accurate prediction of prognosis to determine appropriate clinical management. Validation of a novel prognostic test based on the immunohistochemical expression of two protein biomarkers in the epidermal microenvironment of primary melanomas was undertaken; loss of these biomarkers had previously been shown to be associated with a higher risk of recurrence or metastasis. A parallel qualitative study exploring secondary care health professional and patient views of the test was undertaken and this paper reports the perceived barriers and enablers to its implementation into the melanoma care pathway. METHODS: Qualitative methods were employed drawing upon the Theoretical Domains Framework (TDF) in the exploration and analysis. An inductive-deductive analysis was performed, with all data coded using a thematic then TDF framework. FINDINGS: 20 dermatologists, plastic surgeons, cancer nurse specialists, oncologists and histopathologists participated. Nine TDF domains were relevant to all health professional groups and the 'Skills' and 'Beliefs about Capabilities' domains were relevant only to histopathologists. 'Optimism' and 'Beliefs about consequences' were strong enablers particularly for clinicians. 'Environmental context and resources' (impact on pathology services) and 'Knowledge' (the need for robust evidence about the test reliability) were the main perceived barriers. 19 patients and one carer were interviewed. For the patients eight domains were relevant. ('Knowledge', 'Emotions', 'Beliefs about consequences', 'Social Role and identity', 'Behavioural regulation', 'Memory, attention and decision processes', 'Reinforcement' and 'Skills'). The consequences of the implementation of the test were reassurance about future risk, changes to the follow-up pathway on which there were mixed views, and the need to ensure they maintained self-surveillance (Beliefs about consequences). The test was acceptable to all patient interviewees but the resultant changes to management would need to be supported by mechanisms for fast-track back into the clinic, further information on self-surveillance and clear management plans at the time the result is conveyed (Behavioural regulation). CONCLUSIONS: Health professionals and patients perceived positive consequences-for patients and for health services-of adopting the test. However, its implementation would require exploration of the resource implications for pathology services, psychological support for patients with a high-risk test result and mechanisms to reassure and support patients should the test lead to reduced frequency or duration of follow-up. Exploring implementation at an early stage with health professionals presented challenges related to the provision of specific details of the test and its validation.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Humanos , Melanoma/diagnóstico , Rol Profesional , Pronóstico , Investigación Cualitativa , Reproducibilidad de los Resultados , Neoplasias Cutáneas/diagnóstico , Microambiente Tumoral , Melanoma Cutáneo Maligno
7.
Autophagy ; 18(4): 799-815, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34338148

RESUMEN

Macroautophagy/autophagy is critical for the regulation of pancreatic ß-cell mass and its deregulation has been implicated in the pathogenesis of type 2 diabetes (T2D). We have previously shown that treatment of pancreatic ß-cells with the GLP1R (glucagon like peptide 1 receptor) agonist exendin-4 stimulates autophagic flux in a setting of chronic nutrient excess. The aim of this study was to identify the underlying pathways contributing to enhanced autophagic flux.Pancreatic ß-cells (INS-1E),mouse and human islets were treated with glucolipotoxic stress (0.5 mM palmitate and 25 mM glucose) in the presence of exendin-4. Consistent with our previous work, exendin-4 stimulated autophagic flux. Using chemical inhibitors and siRNA knockdown, we identified RAPGEF4/EPAC2 (Rap guanine nucleotide exchange factor 4) and downstream calcium signaling to be essential for regulation of autophagic flux by exendin-4. This pathway was independent of AMPK and MTOR signaling. Further analysis identified PPP3/calcineurin and its downstream regulator TFEB (transcription factor EB) as key proteins mediating exendin-4 induced autophagy. Importantly, inhibition of this pathway prevented exendin-4-mediated cell survival and overexpression of TFEB mimicked the cell protective effects of exendin-4 in INS-1E and human islets. Moreover, treatment of db/db mice with exendin-4 for 21 days increased the expression of lysosomal markers within the pancreatic islets. Collectively our data identify the RAPGEF4/EPAC2-calcium-PPP3/calcineurin-TFEB axis as a key mediator of autophagic flux, lysosomal function and cell survival in pancreatic ß-cells. Pharmacological modulation of this axis may offer a novel therapeutic target for the treatment of T2D.Abbreviations: AKT1/protein kinase B: AKT serine/threonine kinase 1; AMPK: 5' AMP-activated protein kinase; CAMKK: calcium/calmodulin-dependent protein kinase kinase; cAMP: cyclic adenosine monophosphate; CASP3: caspase 3; CREB: cAMP response element-binding protein; CTSD: cathepsin D; Ex4: exendin-4(1-39); GLP-1: glucagon like peptide 1; GLP1R: glucagon like peptide 1 receptor; GLT: glucolipotoxicity; INS: insulin; MTOR: mechanistic target of rapamycin kinase; NFAT: nuclear factor of activated T-cells; PPP3/calcineurin: protein phosphatase 3; PRKA/PKA: protein kinase cAMP activated; RAPGEF3/EPAC1: Rap guanine nucleotide exchange factor 3; RAPGEF4/EPAC2: Rap guanine nucleotide exchange factor 4; SQSTM1/p62: sequestosome 1; T2D: type 2 diabetes; TFEB: transcription factor EB.


Asunto(s)
Calcineurina , Diabetes Mellitus Tipo 2 , Proteínas Quinasas Activadas por AMP , Animales , Autofagia , Calcineurina/metabolismo , Calcio/metabolismo , Exenatida/farmacología , Receptor del Péptido 1 Similar al Glucagón , Factores de Intercambio de Guanina Nucleótido , Ratones , Serina-Treonina Quinasas TOR/metabolismo
8.
J Biol Chem ; 285(9): 6091-100, 2010 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-20022965

RESUMEN

The neuroectodermal tumors neuroblastoma and melanoma represent biologically aggressive and chemoresistant cancers. The chemotherapeutic agents fenretinide and bortezomib induce apoptosis through endoplasmic reticulum (ER) stress in these tumor types. The aim of this study was to test the hypothesis that the early events of ER stress signaling and response pathways induced by fenretinide and bortezomib are mediated by the eukaryotic initiation factor 2alpha (eIF2alpha)-ATF4 signaling pathway. Treatment of neuroblastoma and melanoma cell lines with fenretinide, bortezomib, or thapsigargin resulted in induction of eIF2alpha signaling, characterized by increased expression of phosphorylated eIF2alpha, ATF4, ATF3, and GADD34. These events correlated with induction of the pro-apoptotic protein Noxa. The cytotoxic response, characterized by up-regulation of Noxa and cell death, was dependent on ATF4, but not the ER-related pro-death signaling pathways involving GADD153 or IRE1. Although PERK-dependent phosphorylation of eIF2alpha enhanced ATF4 protein levels during ER stress, cell death in response to fenretinide, bortezomib, or thapsigargin was not abrogated by inhibition of eIF2alpha phosphorylation through PERK knockdown or overexpression of wild-type eIF2alpha. Furthermore, ATF4 induction in response to ER stress was dependent primarily on transcriptional activation, which occurred in a PERK- and phosphorylated eIF2alpha-independent manner. These results demonstrate that ATF4 mediates ER stress-induced cell death of neuroectodermal tumor cells in response to fenretinide or bortezomib. Understanding the complex regulation of cell death pathways in response to ER stress-inducing drugs has the potential to reveal novel therapeutic targets, thus allowing the development of improved treatment strategies to overcome chemoresistance.


Asunto(s)
Factor de Transcripción Activador 4/fisiología , Muerte Celular , Retículo Endoplásmico/patología , Tumores Neuroectodérmicos/patología , Estrés Fisiológico/efectos de los fármacos , Antineoplásicos/farmacología , Ácidos Borónicos/farmacología , Bortezomib , Línea Celular Tumoral , Fenretinida/farmacología , Humanos , Tumores Neuroectodérmicos/tratamiento farmacológico , Pirazinas/farmacología , Transducción de Señal
9.
Clin Cancer Res ; 15(4): 1192-8, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19228725

RESUMEN

PURPOSE: Single-agent chemotherapy is largely the treatment of choice for systemic therapy of metastatic melanoma, but survival rates are low, and novel adjuvant and systemic therapies are urgently required. Endoplasmic reticulum (ER) stress is a potential therapeutic target, and two relatively new drugs, fenretinide and bortezomib (Velcade), each acting via different cellular mechanisms, induce ER stress leading to apoptosis in melanoma cells. The aim of this study was to test the hypothesis that apoptosis of melanoma cells may be increased by combining clinically achievable concentrations of fenretinide and bortezomib. EXPERIMENTAL DESIGN: Three human melanoma cell lines were used to assess changes in viability and the induction of apoptosis in response to fenretinide, bortezomib, or both drugs together. A s.c. xenograft model was used to test responses in vivo. RESULTS: Fenretinide and bortezomib synergistically decreased viability and increased apoptosis in all three melanoma lines at clinically achievable concentrations. This was also reflected by increased expression of GADD153, a marker of ER stress-induced apoptosis. In vivo, fenretinide in combination with bortezomib gave a marked reduction in xenograft tumor volume and an increase in apoptosis compared with fenretinide or bortezomib alone. The cell cycle stage of tumor cells in vivo were similar to that predicted from the effects of each drug or the combination in vitro. CONCLUSIONS: These results suggest that fenretinide and bortezomib, both of which are available in clinical formulation, warrant clinical evaluation as a combination therapy for metastatic melanoma.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Ácidos Borónicos/administración & dosificación , Retículo Endoplásmico/efectos de los fármacos , Fenretinida/administración & dosificación , Melanoma/tratamiento farmacológico , Pirazinas/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Bortezomib , División Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Retículo Endoplásmico/metabolismo , Femenino , Fase G2/efectos de los fármacos , Humanos , Antígeno Ki-67/análisis , Melanoma/patología , Melanoma/secundario , Ratones
10.
Front Immunol ; 9: 2217, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30319656

RESUMEN

In order to limit the adverse effects of excessive inflammation, anti-inflammatory responses are stimulated at an early stage of an infection, but during sepsis these can lead to deactivation of immune cells including monocytes. In addition, there is emerging evidence that the up-regulation of mitochondrial quality control mechanisms, including mitochondrial biogenesis and mitophagy, is important during the recovery from sepsis and inflammation. We aimed to describe the relationship between the compensatory immune and mitochondrial responses that are triggered following exposure to an inflammatory stimulus in human monocytic cells. Incubation with lipopolysaccharide resulted in a change in the immune phenotype of THP-1 cells consistent with the induction of endotoxin tolerance, similar to that seen in deactivated septic monocytes. After exposure to LPS there was also early evidence of oxidative stress, which resolved in association with the induction of antioxidant defenses and the stimulation of mitochondrial degradation through mitophagy. This was compensated by a parallel up-regulation of mitochondrial biogenesis that resulted in an overall increase in mitochondrial respiratory activity. These observations improve our understanding of the normal homeostatic responses that limit the adverse cellular effects of unregulated inflammation, and which may become ineffective when an infection causes sepsis.


Asunto(s)
Mitocondrias/inmunología , Mitofagia/inmunología , Monocitos/inmunología , Biogénesis de Organelos , Endotoxinas/inmunología , Humanos , Tolerancia Inmunológica , Lipopolisacáridos/inmunología , Mitocondrias/metabolismo , Monocitos/citología , Estrés Oxidativo/inmunología , Células THP-1
11.
Diabetes ; 66(5): 1272-1285, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28232493

RESUMEN

Studies in animal models of type 2 diabetes have shown that glucagon-like peptide 1 (GLP-1) receptor agonists prevent ß-cell loss. Whether GLP-1 mediates ß-cell survival via the key lysosomal-mediated process of autophagy is unknown. In this study, we report that treatment of INS-1E ß-cells and primary islets with glucolipotoxicity (0.5 mmol/L palmitate and 25 mmol/L glucose) increases LC3 II, a marker of autophagy. Further analysis indicates a blockage in autophagic flux associated with lysosomal dysfunction. Accumulation of defective lysosomes leads to lysosomal membrane permeabilization and release of cathepsin D, which contributes to cell death. Our data further demonstrated defects in autophagic flux and lysosomal staining in human samples of type 2 diabetes. Cotreatment with the GLP-1 receptor agonist exendin-4 reversed the lysosomal dysfunction, relieving the impairment in autophagic flux and further stimulated autophagy. Small interfering RNA knockdown showed the restoration of autophagic flux is also essential for the protective effects of exendin-4. Collectively, our data highlight lysosomal dysfunction as a critical mediator of ß-cell loss and shows that exendin-4 improves cell survival via restoration of lysosomal function and autophagic flux. Modulation of autophagy/lysosomal homeostasis may thus define a novel therapeutic strategy for type 2 diabetes, with the GLP-1 signaling pathway as a potential focus.


Asunto(s)
Autofagia/efectos de los fármacos , Diabetes Mellitus Tipo 2/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Glucosa/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Lisosomas/efectos de los fármacos , Palmitatos/farmacología , Adulto , Animales , Apoptosis , Western Blotting , Estudios de Casos y Controles , Catepsina D/efectos de los fármacos , Catepsina D/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Exenatida , Humanos , Inmunohistoquímica , Incretinas/farmacología , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Lisosomas/metabolismo , Ratones , Proteínas Asociadas a Microtúbulos/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/metabolismo , Persona de Mediana Edad , Péptidos/farmacología , ARN Interferente Pequeño , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ponzoñas/farmacología
12.
Cancer Res ; 63(14): 3860-5, 2003 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-12873973

RESUMEN

Glucosylceramide synthase (GCS), the key enzyme in the biosynthesis of glycosphingolipids, has been implicated in many biological phenomena, including multidrug resistance. GCS inhibition, by both antisense and the specific inhibitor (D-threo)-1-phenyl-2-decanoylamino-3-morpholino-1-propanol (PDMP), results in a drastic decrease of apoptosis induced by the p53-independent chemotherapeutic agent N-(4-hydroxyphenyl)retinamide in neuroepithelioma cells. By using the yeast two-hybrid system, we have identified a member of the reticulon (RTN) family (RTN-1C) as the major GCS-protein partner. Interestingly, RTN-1C not only interacts with GCS at Golgi/ER interface but also modulates its catalytic activity in situ. In fact, overexpression of RTN-1C sensitizes CHP-100 cells to fenretinide-induced apoptosis. These findings demonstrate a novel p53-independent pathway of apoptosis regulated by Golgi/endoplasmic reticulum protein interactions, which is relevant for cancer combined therapy.


Asunto(s)
Apoptosis/fisiología , Glucosiltransferasas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Tumores Neuroectodérmicos Periféricos Primitivos/enzimología , Secuencia de Aminoácidos , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Retículo Endoplásmico/enzimología , Retículo Endoplásmico/metabolismo , Fenretinida/farmacología , Glucosiltransferasas/genética , Aparato de Golgi/enzimología , Aparato de Golgi/metabolismo , Humanos , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Tumores Neuroectodérmicos Periféricos Primitivos/genética , Tumores Neuroectodérmicos Periféricos Primitivos/patología , Homología de Secuencia de Aminoácido , Transfección
13.
Cancer Res ; 63(21): 7310-3, 2003 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-14612528

RESUMEN

Unlike 13-cis-retinoic acid, the synthetic retinoid fenretinide [N-(4-hydroxyphenyl)retinamide] induces apoptosis of neuroblastoma cells by mechanisms involving retinoic acid receptors and oxidative stress. After screening a cDNA array for apoptosis-related genes, the Bcl2-related protein Bak was identified as a fenretinide-inducible gene in SH-SY5Y neuroblastoma cells, and this was confirmed by Western blotting and flow cytometry. Although fenretinide acts synergistically in vitro with chemotherapeutic drugs, these drugs did not induce Bak expression. Retinoic acid receptor antagonists did not block the induction of Bak by fenretinide. Conversely, Bak induction was blocked by the antioxidant vitamin C. Overexpression of Bak increased apoptosis in both the presence and absence of fenretinide, whereas expression of antisense Bak inhibited fenretinide-induced apoptosis. Bak expression was also induced in cells overexpressing the stress-induced transcription factor GADD153, but Bak expression was inhibited in cells expressing an antisense GADD153 construct. These results suggest that Bak is a downstream mediator of an oxidative stress pathway leading to apoptosis of SH-SY5Y neuroblastoma cells in response to fenretinide.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Fenretinida/farmacología , Proteínas de la Membrana/fisiología , Neuroblastoma/patología , Apoptosis/fisiología , Western Blotting , Línea Celular Tumoral , Citometría de Flujo , Humanos , Proteínas de la Membrana/biosíntesis , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/metabolismo , Proteína Destructora del Antagonista Homólogo bcl-2
14.
Cancer Res ; 62(18): 5158-67, 2002 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-12234979

RESUMEN

The synthetic retinoid fenretinide induces apoptosis of neuroblastoma cells and in vitro acts synergistically with chemotherapeutic drugs used to treat neuroblastoma. The mechanisms of fenretinide-induced cell death of neuroblastoma cells are complex, involving cellular signaling pathways as yet incompletely defined but, in part, involving the generation of reactive oxygen species (ROS). In an attempt to characterize the mechanism of action of fenretinide, cDNA array filters were screened to identify apoptotic genes regulated in response to treatment of SH-SY5Y cells with fenretinide. Expression of the stress-induced transcription factor, GADD153, was up-regulated at both the protein and mRNA levels in response to fenretinide. Overexpression of GADD153 increased apoptosis in the presence and absence of fenretinide, whereas reduced expression of GADD153 by expression of antisense DNA abrogated the response to fenretinide. Although fenretinide is a partial retinoic acid receptor (RAR)-beta/gamma agonist, RARbeta/gamma antagonists did not block the induction of GADD153 by fenretinide; conversely, the induction of GADD153 was blocked by antioxidants. Enzyme inhibitors were used to identify pathways mediating the ROS-dependent effects of fenretinide: inhibitors of phospholipase A(2) and lypoxygenases (LOX), and specific inhibitors of 12-LOX, but not 5-LOX or 15-LOX, inhibited the induction of ROS, apoptosis, and GADD153 in response to fenretinide. The inhibition of ROS and apoptosis was reversed by the addition of the 12-LOX products, 12 (S)-hydroperoxyeicosatetraenoic acid (12-HpETE) and 12 (S)-hydroxyeicosatetraenoic acid (12-HETE). Fenretinide did not increase free arachidonic acid levels, but increased LOX activity without a detectable increase in 12-LOX protein. These results suggest that fenretinide induces apoptosis via RAR-dependent and -independent pathways in which the RAR-independent pathway is characterized by a fenretinide-dependent increase in 12-LOX activity, leading to the induction of GADD153. The targeting of 12-LOX and/or GADD153 in neuroblastoma cells may thus present a novel pathway for the development of drugs inducing apoptosis of neuroblastoma with improved tumor specificity.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Araquidonato 12-Lipooxigenasa/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/fisiología , Fenretinida/farmacología , Neuroblastoma/patología , Factores de Transcripción/fisiología , Apoptosis/genética , Apoptosis/fisiología , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Proteínas Potenciadoras de Unión a CCAAT/genética , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Especies Reactivas de Oxígeno/metabolismo , Receptores de Ácido Retinoico/antagonistas & inhibidores , Factor de Transcripción CHOP , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/fisiología , Receptor de Ácido Retinoico gamma
15.
Front Oncol ; 6: 236, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27882308

RESUMEN

Prognosis and survival for malignant melanoma is highly dependent on early diagnosis and treatment. While the American Joint Committee on Cancer (AJCC) criterion provides a means of staging melanomas and guiding treatment approaches, it is unable to identify the risk of disease progression of early stage tumors or provide reliable stratification for novel adjuvant therapies. The demand for credible prognostic/companion biomarkers able to identify high-risk melanoma subgroups as well as guide more effective personalized/precision-based therapy is therefore of paramount importance. Autophagy, the principle lysosomal-mediated process for the degradation/recycling of cellular debris, is a hot topic in cancer medicine, and observations of its deregulation in melanoma have brought its potential as a prognostic biomarker to the forefront of current research. Key regulatory proteins, including Atg8/microtubule-associated light chain 3 (LC3) and BECN1 (Beclin 1), have been proposed as potential prognostic biomarkers. However, given the dynamic nature of autophagy, their expression in vitro does not translate to their use as a prognostic biomarker for melanoma in vivo. We have recently identified the expression levels of Sequestosome1/SQSTM1 (p62) and activating molecule in Beclin 1-regulated autophagy protein 1 (AMBRA1) as novel independent prognostic biomarkers for early stage melanomas. While increasing followed by subsequent decreasing levels of p62 expression reflects the paradoxical role of autophagy in melanoma, expression levels additionally define a novel prognostic biomarker for AJCC stage II tumors. Conversely, loss of AMBRA1 in the epidermis overlying primary melanomas defines a novel prognostic biomarker for AJCC stage I tumors. Collectively, the definition of AMBRA1 and p62 as prognostic biomarkers for early stage melanomas provides novel and accurate means through which to identify tumors at risk of disease progression, facilitating earlier patient therapeutic intervention and stratification tools for novel personalized therapeutic approaches to improve clinical outcome.

16.
Cancer Lett ; 228(1-2): 105-10, 2005 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-15907365

RESUMEN

Fenretinide is thought to induce apoptosis via increases in ceramide levels but the mechanisms of ceramide generation and the link between ceramide and subsequent apoptosis in neuroblastoma cells is unclear. In SH-SY5Y neuroblastoma cells, evidence suggests that acid sphingomyelinase activity is essential for the induction of ceramide and apoptosis in response to fenretinide. Downstream of ceramide, apoptosis in response to fenretinide is mediated by increased glucosylceramide synthase activity resulting in increased levels of gangliosides GD3 and GD2 via GD3 synthase. GD3 is a key signalling intermediate leading to apoptosis via the activation of 12-Lipoxygenase, and the parallel induction of GD2 suggests that fenretinide might enhance the response of neuroblastoma to therapy with anti-GD2 antibodies.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/fisiología , Fenretinida/farmacología , Gangliósidos/fisiología , Neuroblastoma/patología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Ceramidas/fisiología , Gangliósidos/inmunología , Humanos , Neuroblastoma/terapia , Transducción de Señal
17.
Mol Cell Oncol ; 2(3): e995016, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-27308477

RESUMEN

Autophagy and endoplasmic reticulum (ER) stress are involved in the development, progression, and chemoresistance of melanoma. We recently reported that oncogenic serine/threonine-protein kinase BRAF induces chronic ER stress, hence increasing baseline autophagy and promoting chemoresistance. The attenuation of ER stress restores basal autophagic activity and resensitizes melanoma cells to apoptosis.

18.
Mol Cancer Ther ; 14(11): 2665-73, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26330548

RESUMEN

Metastatic melanoma remains incurable, emphasizing the acute need for improved research models to investigate the underlying biologic mechanisms mediating tumor invasion and metastasis, and to develop more effective targeted therapies to improve clinical outcome. Available animal models of melanoma do not accurately reflect human disease and current in vitro human skin equivalent models incorporating melanoma cells are not fully representative of the human skin microenvironment. We have developed a robust and reproducible, fully humanized three-dimensional (3D) skin equivalent comprising a stratified, terminally differentiated epidermis and a dermal compartment consisting of fibroblast-generated extracellular matrix. Melanoma cells incorporated into the epidermis were able to invade through the basement membrane and into the dermis, mirroring early tumor invasion in vivo. Comparison of our novel 3D melanoma skin equivalent with melanoma in situ and metastatic melanoma indicates that this model accurately recreates features of disease pathology, making it a physiologically representative model of early radial and vertical growth-phase melanoma invasion.


Asunto(s)
Melanoma/patología , Neoplasias Cutáneas/patología , Piel Artificial , Piel/patología , Células 3T3 , Animales , Biomarcadores de Tumor/metabolismo , Técnicas de Cultivo de Célula/métodos , Línea Celular Tumoral , Células Cultivadas , Dermis/metabolismo , Dermis/patología , Epidermis/metabolismo , Epidermis/patología , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Antígeno MART-1/metabolismo , Melanoma/metabolismo , Ratones , Microscopía Electrónica de Rastreo , Microscopía Fluorescente , Modelos Biológicos , Invasividad Neoplásica , Metástasis de la Neoplasia , Piel/metabolismo , Piel/ultraestructura , Neoplasias Cutáneas/metabolismo , Microambiente Tumoral
19.
J Invest Dermatol ; 135(6): 1629-1637, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25674907

RESUMEN

Although the global incidence of cutaneous melanoma is increasing, survival rates for patients with metastatic disease remain <10%. Novel treatment strategies are therefore urgently required, particularly for patients bearing BRAF/NRAS wild-type tumors. Targeting autophagy is a means to promote cancer cell death in chemotherapy-resistant tumors, and the aim of this study was to test the hypothesis that cannabinoids promote autophagy-dependent apoptosis in melanoma. Treatment with Δ(9)-Tetrahydrocannabinol (THC) resulted in the activation of autophagy, loss of cell viability, and activation of apoptosis, whereas cotreatment with chloroquine or knockdown of Atg7, but not Beclin-1 or Ambra1, prevented THC-induced autophagy and cell death in vitro. Administration of Sativex-like (a laboratory preparation comprising equal amounts of THC and cannabidiol (CBD)) to mice bearing BRAF wild-type melanoma xenografts substantially inhibited melanoma viability, proliferation, and tumor growth paralleled by an increase in autophagy and apoptosis compared with standard single-agent temozolomide. Collectively, our findings suggest that THC activates noncanonical autophagy-mediated apoptosis of melanoma cells, suggesting that cytotoxic autophagy induction with Sativex warrants clinical evaluation for metastatic disease.


Asunto(s)
Autofagia , Cannabinoides/química , Melanoma/patología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/metabolismo , Beclina-1 , Cannabidiol , Cannabinol/química , Muerte Celular , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Dacarbazina/análogos & derivados , Dacarbazina/química , Dronabinol/química , Combinación de Medicamentos , Humanos , Masculino , Melanoma/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Desnudos , Microscopía Confocal , Metástasis de la Neoplasia , Trasplante de Neoplasias , Neoplasias/metabolismo , Extractos Vegetales/química , Proteínas Proto-Oncogénicas B-raf/metabolismo , Neoplasias Cutáneas/metabolismo , Temozolomida , Proteínas ras/metabolismo
20.
Cancer Lett ; 197(1-2): 157-63, 2003 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-12880976

RESUMEN

Unlike 13-cis retinoic acid, the synthetic retinoid fenretinide induces apoptosis of neuroblastoma cells and in vitro acts synergistically with the chemotherapeutic drugs, cisplatin, etoposide and carboplatin. The stress-induced transcription factor GADD153 and the Bcl2-related protein Bak are upregulated in response to fenretinide. Although fenretinide is a partial retinoic acid receptor (RAR)-beta/gamma agonist, RARbeta/gamma antagonists do not block the induction of GADD153 or Bak by fenretinide. Conversely, the induction of GADD153 and Bak is blocked by antioxidants. Neither GADD153 or Bak were induced by chemotherapeutic agents but over expression of GADD153 results in increased sensitivity to fenretinide-induced apoptosis. Therefore, fenretinide induces apoptosis via RAR-dependent and -independent pathways in which the RAR-independent pathway is characterised by the reactive oxygen species-dependent induction of GADD153 and Bak. The targeting of GADD153 and Bak in neuroblastoma cells may be novel pathways for the development of drugs inducing apoptosis of neuroblastoma with improved tumour specificity.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Fenretinida/farmacología , Proteínas de la Membrana/metabolismo , Neuroblastoma/patología , Factores de Transcripción/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/metabolismo , Receptores de Ácido Retinoico/antagonistas & inhibidores , Factor de Transcripción CHOP , Células Tumorales Cultivadas , Proteína Destructora del Antagonista Homólogo bcl-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA