Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Oncogene ; 42(5): 351-363, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36463370

RESUMEN

Interferon-gamma (IFNG) has long been regarded as the flag-bearer for the anti-cancer immunosurveillance mechanisms. However, relatively recent studies have suggested a dual role of IFNG, albeit there is no direct experimental evidence for its potential pro-tumor functions. Here we provide in vivo evidence that treatment of mouse melanoma cell lines with Ifng enhances their tumorigenicity and metastasis in lung colonization allograft assays performed in immunocompetent syngeneic host mice, but not in immunocompromised host mice. We also show that this enhancement is dependent on downstream signaling via Stat1 but not Stat3, suggesting an oncogenic function of Stat1 in melanoma. The experimental results suggest that melanoma cell-specific Ifng signaling modulates the tumor microenvironment and its pro-tumorigenic effects are partially dependent on the γδ T cells, as Ifng-enhanced tumorigenesis was inhibited in the TCR-δ knockout mice. Overall, these results show that Ifng signaling may have tumor-promoting effects in melanoma by modulating the immune cell composition of the tumor microenvironment.


Asunto(s)
Interferón gamma , Melanoma , Animales , Ratones , Interferón gamma/metabolismo , Melanoma/patología , Transducción de Señal , Línea Celular , Carcinogénesis , Ratones Noqueados , Línea Celular Tumoral , Microambiente Tumoral
2.
EMBO J ; 41(19): e110046, 2022 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-36039850

RESUMEN

The role of store-operated Ca2+ entry (SOCE) in melanoma metastasis is highly controversial. To address this, we here examined UV-dependent metastasis, revealing a critical role for SOCE suppression in melanoma progression. UV-induced cholesterol biosynthesis was critical for UV-induced SOCE suppression and subsequent metastasis, although SOCE suppression alone was both necessary and sufficient for metastasis to occur. Further, SOCE suppression was responsible for UV-dependent differences in gene expression associated with both increased invasion and reduced glucose metabolism. Functional analyses further established that increased glucose uptake leads to a metabolic shift towards biosynthetic pathways critical for melanoma metastasis. Finally, examination of fresh surgically isolated human melanoma explants revealed cholesterol biosynthesis-dependent reduced SOCE. Invasiveness could be reversed with either cholesterol biosynthesis inhibitors or pharmacological SOCE potentiation. Collectively, we provide evidence that, contrary to current thinking, Ca2+ signals can block invasive behavior, and suppression of these signals promotes invasion and metastasis.


Asunto(s)
Señalización del Calcio , Melanoma , Calcio/metabolismo , Canales de Calcio/metabolismo , Colesterol , Glucosa , Humanos , Melanoma/genética , Melanoma/metabolismo , Proteína ORAI1/metabolismo , Molécula de Interacción Estromal 1/metabolismo
3.
Adv Exp Med Biol ; 1360: 109-116, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35505166

RESUMEN

Gadd45a, Gadd45b, and Gadd45g have been implicated in cell cycle arrest, DNA repair, apoptosis, innate immunity, genomic stability, and more recently in senescence. Evidence has accumulated that Gadd45a deficiency results in escape of mouse embryo fibroblasts from senescence, whereas Gadd45b deficiency promotes premature senescence and skin aging. Moreover, recently Gadd45b deficiency was found to promote senescence and attenuate liver fibrosis, whereas Gadd45a was observed to exert a protective effect against hepatic fibrosis. These findings indicate that the Gadd45 stress response proteins play important roles in modulating cellular responses to senescence. Thus, exploring how Gadd45 proteins modulate cellular senescence has the potential to provide new and innovative tools to treat cancer as well as liver disease.


Asunto(s)
Apoptosis , Envejecimiento de la Piel , Animales , Antígenos de Diferenciación , Apoptosis/genética , Puntos de Control del Ciclo Celular , Senescencia Celular/genética , Reparación del ADN , Ratones
4.
Pigment Cell Melanoma Res ; 35(3): 342-355, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35266648

RESUMEN

Melanogenesis (melanin pigment production) in melanocytes is canonically stimulated by the alpha melanocyte stimulating hormone (αMSH), which activates the cyclic-AMP-mediated expression of the melanocyte inducing transcription factor (MITF) and its downstream melanogenic genes, including the principal rate-limiting melanogenic enzyme tyrosinase (TYR). Here, we report that interferon-gamma (IFNG; type II interferon), but not interferon-alpha (a type I interferon), induces a noncanonical melanogenic pathway in mouse and human melanocytic cells. Inhibition of IFNG pathway by the JAK1/2 inhibitor ruxolitinib or knocking out Stat1 gene abrogated the IFNG-induced melanogenesis. Interestingly, IFNG-induced melanogenesis was independent of MITF. IFNG markedly increased the TYR protein expression but did not affect the mRNA expression, suggesting a post-translational regulatory mechanism. In contrast, IFNG had no effect on the expression of other melanogenesis-related proteins, for example, tyrosinase-related protein 1 (TYRP1) and dopachrome tautomerase (DCT). Glycosidase digestion assays revealed that IFNG treatment increased the mature glycosylated form of TYR, but not its de novo synthesis. Moreover, cycloheximide chase assay showed that degradation of TYR was decreased in IFNG-treated cells. These results suggest that the IFNG-STAT1 pathway regulates melanogenesis via regulation of the post-translational processing and protein stability of TYR.


Asunto(s)
Interferón gamma , Monofenol Monooxigenasa , Animales , Interferón gamma/metabolismo , Interferón gamma/farmacología , Melaninas/metabolismo , Melanocitos/metabolismo , Glicoproteínas de Membrana , Ratones , Monofenol Monooxigenasa/metabolismo , Oxidorreductasas , Unión Proteica
6.
Nat Cell Biol ; 22(5): 591-602, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32284542

RESUMEN

Triple-negative breast cancer (TNBC) is characterized by a high degree of immune infiltrate in the tumour microenvironment, which may influence the fate of TNBC cells. We reveal that loss of the tumour suppressive transcription factor Elf5 in TNBC cells activates intrinsic interferon-γ (IFN-γ) signalling, promoting tumour progression and metastasis. Mechanistically, we find that loss of the Elf5-regulated ubiquitin ligase FBXW7 ensures stabilization of its putative protein substrate IFN-γ receptor 1 (IFNGR1) at the protein level in TNBC. Elf5low tumours show enhanced IFN-γ signalling accompanied by an increase of immunosuppressive neutrophils within the tumour microenvironment and increased programmed death ligand 1 expression. Inactivation of either programmed death ligand 1 or IFNGR1 elicited a robust anti-tumour and/or anti-metastatic effect. A positive correlation between ELF5 and FBXW7 expression and a negative correlation between ELF5, FBXW7 and IFNGR1 expression in the tumours of patients with TNBC strongly suggest that this signalling axis could be exploited for patient stratification and immunotherapeutic treatment strategies for Elf5low patients with TNBC.


Asunto(s)
Proliferación Celular/fisiología , Proteínas de Unión al ADN/metabolismo , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Interferón gamma/metabolismo , Metástasis de la Neoplasia/patología , Receptores de Interferón/metabolismo , Factores de Transcripción/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Transducción de Señal/fisiología , Microambiente Tumoral/fisiología , Receptor de Interferón gamma
7.
EMBO Rep ; 21(5): e48904, 2020 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-32212315

RESUMEN

While the zinc finger transcription factors EGR1, EGR2, and EGR3 are recognized as critical for T-cell function, the role of EGR4 remains unstudied. Here, we show that EGR4 is rapidly upregulated upon TCR engagement, serving as a critical "brake" on T-cell activation. Hence, TCR engagement of EGR4-/- T cells leads to enhanced Ca2+ responses, driving sustained NFAT activation and hyperproliferation. This causes profound increases in IFNγ production under resting and diverse polarizing conditions that could be reversed by pharmacological attenuation of Ca2+ entry. Finally, an in vivo melanoma lung colonization assay reveals enhanced anti-tumor immunity in EGR4-/- mice, attributable to Th1 bias, Treg loss, and increased CTL generation in the tumor microenvironment. Overall, these observations reveal for the first time that EGR4 is a key regulator of T-cell differentiation and function.


Asunto(s)
Señalización del Calcio , Factores de Transcripción de la Respuesta de Crecimiento Precoz , Neoplasias , Animales , Diferenciación Celular , Activación de Linfocitos , Ratones , Microambiente Tumoral , Dedos de Zinc
8.
Nat Commun ; 11(1): 333, 2020 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-31949145

RESUMEN

Cutaneous malignant melanoma is an aggressive cancer of melanocytes with a strong propensity to metastasize. We posit that melanoma cells acquire metastatic capability by adopting an embryonic-like phenotype, and that a lineage approach would uncover metastatic melanoma biology. Using a genetically engineered mouse model to generate a rich melanoblast transcriptome dataset, we identify melanoblast-specific genes whose expression contribute to metastatic competence and derive a 43-gene signature that predicts patient survival. We identify a melanoblast gene, KDELR3, whose loss impairs experimental metastasis. In contrast, KDELR1 deficiency enhances metastasis, providing the first example of different disease etiologies within the KDELR-family of retrograde transporters. We show that KDELR3 regulates the metastasis suppressor, KAI1, and report an interaction with the E3 ubiquitin-protein ligase gp78, a regulator of KAI1 degradation. Our work demonstrates that the melanoblast transcriptome can be mined to uncover targetable pathways for melanoma therapy.


Asunto(s)
Perfilación de la Expresión Génica , Melanoma/genética , Melanoma/metabolismo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Transcriptoma , Animales , Línea Celular Tumoral , Retículo Endoplásmico , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Proteína Kangai-1/genética , Proteína Kangai-1/metabolismo , Pulmón/patología , Melanocitos/metabolismo , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia/genética , Neoplasias Primarias Secundarias/patología , Fenotipo , Receptores de Péptidos/genética , Receptores de Péptidos/metabolismo , Neoplasias Cutáneas/patología , Ubiquitina-Proteína Ligasas/metabolismo , Melanoma Cutáneo Maligno
10.
Adv Cancer Res ; 144: 1-54, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31349897

RESUMEN

Cutaneous malignant melanoma is one of the few major cancers that continue to exhibit a positive rate of increase in the developed world. A wealth of epidemiological data has undisputedly implicated ultraviolet radiation (UVR) from sunlight and artificial sources as the major risk factor for melanomagenesis. However, the molecular mechanisms of this cause-and-effect relationship remain murky and understudied. Recent efforts on multiple fronts have brought unprecedented expansion of our knowledge base on this subject and it is now clear that melanoma is caused by a complex interaction between genetic predisposition and environmental exposure, primarily to UVR. Here we provide an overview of the effects of the macroenvironment (UVR) on the skin microenvironment and melanocyte-specific intrinsic (mostly genetic) landscape, which conspire to produce one of the deadliest malignancies.


Asunto(s)
Transformación Celular Neoplásica/genética , Interacción Gen-Ambiente , Melanoma/genética , Neoplasias Inducidas por Radiación/genética , Neoplasias Cutáneas/genética , Rayos Ultravioleta/efectos adversos , Animales , Transformación Celular Neoplásica/patología , Modelos Animales de Enfermedad , Humanos , Melanoma/etiología , Melanoma/patología , Proteínas de Neoplasias/genética , Neoplasias Inducidas por Radiación/etiología , Neoplasias Inducidas por Radiación/patología , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/patología , Melanoma Cutáneo Maligno
11.
Cancer Res ; 79(11): 2909-2922, 2019 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-30737234

RESUMEN

Solar ultraviolet radiation (UVR) suppresses skin immunity, which facilitates initiation of skin lesions and establishment of tumors by promoting immune evasion. It is unclear whether immune checkpoints are involved in the modulation of skin immunity by UVR. Here, we report that UVR exposure significantly increased expression of immune checkpoint molecule PD-L1 in melanoma cells. The damage-associated molecular patterns molecule HMGB1 was secreted by melanocytes and keratinocytes upon UVR, which subsequently activated the receptor for advanced glycation endproducts (RAGE) receptor to promote NF-κB- and IRF3-dependent transcription of PD-L1 in melanocytes. UVR exposure significantly reduced the susceptibility of melanoma cells to CD8+ T-cell-dependent cytotoxicity, which was mitigated by inhibiting the HMGB1/TBK1/IRF3/NF-κB cascade or by blocking the PD-1/PD-L1 checkpoint. Taken together, our findings demonstrate that UVR-induced upregulation of PD-L1 contributes to immune suppression in the skin microenvironment, which may promote immune evasion of oncogenic cells and drive melanoma initiation and progression. SIGNIFICANCE: These findings identify PD-L1 as a critical component of UV-induced immune suppression in the skin, which facilitates immunoevasion of oncogenic melanocytes and development of melanoma.See related commentary by Sahu, p. 2805.


Asunto(s)
Proteína HMGB1 , Rayos Ultravioleta , Antígeno B7-H1/genética , FN-kappa B , Receptor para Productos Finales de Glicación Avanzada , Regulación hacia Arriba
12.
J Interferon Cytokine Res ; 39(1): 30-38, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30388040

RESUMEN

Interferon-gamma (IFNG) has long been implicated as a central orchestrator of antitumor immune responses in the elimination stage of the immunoediting paradigm. However, mounting evidence suggests that IFNG may also have important and significant protumor roles to play in the equilibrium and escape phases through its regulatory effects on immunoevasive functions that promote tumorigenesis. These seemingly contradictory effects of IFNG undoubtedly play profound roles in not only the activation of inflammatory response to cancer but also in the determination of its outcome. In the face of the recent explosion of anticancer immunotherapeutic strategies in the clinic, it is critical that a complete understanding is achieved of the underpinnings of the mechanisms that determine the two faces of IFNG signaling in cancer. Here, the current state of this dichotomy is reviewed.


Asunto(s)
Interferón gamma/inmunología , Neoplasias/inmunología , Animales , Humanos , Transducción de Señal/inmunología
13.
Int J Mol Sci ; 19(5)2018 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-29762513

RESUMEN

Melanocytes are pigment producing cells in the skin that give rise to cutaneous malignant melanoma, which is a highly aggressive and the deadliest form of skin cancer. Studying melanocytes in vivo is often difficult due to their small proportion in the skin and the lack of specific cell surface markers. Several genetically-engineered mouse models (GEMMs) have been created to specifically label the melanocyte compartment. These models give both spatial and temporal control over the expression of a cellular 'beacon' that has an added benefit of inducible expression that can be activated on demand. Two powerful models that are discussed in this review include the melanocyte-specific, tetracycline-inducible green fluorescent protein expression system (iDct-GFP), and the fluorescent ubiquitination-based cell cycle indicator (FUCCI) model that allows for the monitoring of the cell-cycle. These two systems are powerful tools in studying melanocyte and melanoma biology. We discuss their current uses and how they could be employed to help answer unresolved questions in the fields of melanocyte and melanoma biology.


Asunto(s)
Ciclo Celular , Proteínas Fluorescentes Verdes/genética , Melanocitos/citología , Ratones Transgénicos/genética , Animales , Linaje de la Célula , Proteínas Fluorescentes Verdes/metabolismo , Melanocitos/metabolismo , Melanoma/patología , Ratones , Ratones Transgénicos/metabolismo , Microscopía Fluorescente/métodos
14.
EMBO J ; 37(5)2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29449324
15.
Cancer Res ; 78(2): 436-450, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29150430

RESUMEN

CTLA4 is a cell surface receptor on T cells that functions as an immune checkpoint molecule to enforce tolerance to cognate antigens. Anti-CTLA4 immunotherapy is highly effective at reactivating T-cell responses against melanoma, which is postulated to be due to targeting CTLA4 on T cells. Here, we report that CTLA4 is also highly expressed by most human melanoma cell lines, as well as in normal human melanocytes. Interferon-γ (IFNG) signaling activated the expression of the human CTLA4 gene in a melanocyte and melanoma cell-specific manner. Mechanistically, IFNG activated CTLA4 expression through JAK1/2-dependent phosphorylation of STAT1, which bound a specific gamma-activated sequence site on the CTLA4 promoter, thereby licensing CBP/p300-mediated histone acetylation and local chromatin opening. In melanoma cell lines, elevated baseline expression relied upon constitutive activation of the MAPK pathway. Notably, RNA-seq analyses of melanoma specimens obtained from patients who had received anti-CTLA4 immunotherapy (ipilimumab) showed upregulation of an IFNG-response gene expression signature, including CTLA4 itself, which correlated significantly with durable response. Taken together, our results raise the possibility that CTLA4 targeting on melanoma cells may contribute to the clinical immunobiology of anti-CTLA4 responses.Significance: These findings show that human melanoma cells express high levels of the immune checkpoint molecule CTLA4, with important possible implications for understanding how anti-CTLA4 immunotherapy mediates its therapeutic effects. Cancer Res; 78(2); 436-50. ©2017 AACR.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Antígeno CTLA-4/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Interferón gamma/farmacología , Melanocitos/metabolismo , Melanoma/metabolismo , Antineoplásicos/farmacología , Antivirales/farmacología , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/genética , Antígeno CTLA-4/genética , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Perfilación de la Expresión Génica , Humanos , Inmunoterapia , Ipilimumab/farmacología , Melanocitos/efectos de los fármacos , Melanocitos/patología , Melanoma/tratamiento farmacológico , Melanoma/patología , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Regiones Promotoras Genéticas , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Transducción de Señal
16.
Br J Cancer ; 117(11): 1583-1591, 2017 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-29065107

RESUMEN

Macrophages are conventionally classified into M1 and M2 subtypes according to their differentiation status and functional role in the immune system. However, accumulating evidence suggests that this binary classification system is insufficient to account for the remarkable plasticity of macrophages that gives rise to an immense diversity of subtypes. This diverse spectrum of macrophage subtypes play critical roles in various homeostatic and immune functions, but remain far from being fully characterised. In addition to their roles in normal physiological conditions, macrophages also play crucial roles in disease conditions such as cancer. In this review, we discuss the roles tumour-associated macrophages (TAMs) play in regulating different steps of tumour progression and metastasis, and the opportunities to target them in the quest for cancer prevention and treatment.


Asunto(s)
Macrófagos/fisiología , Metástasis de la Neoplasia , Neoplasias/inmunología , Animales , Progresión de la Enfermedad , Humanos , Tolerancia Inmunológica , Invasividad Neoplásica , Células Madre Neoplásicas/fisiología , Neovascularización Patológica/etiología , Microambiente Tumoral
17.
Cancer ; 123(S11): 2089-2103, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28543694

RESUMEN

Melanoma is a complex disease that exhibits highly heterogeneous etiological, histopathological, and genetic features, as well as therapeutic responses. Genetically engineered mouse (GEM) models provide powerful tools to unravel the molecular mechanisms critical for melanoma development and drug resistance. Here, we expound briefly the basis of the mouse modeling design, the available technology for genetic engineering, and the aspects influencing the use of GEMs to model melanoma. Furthermore, we describe in detail the currently available GEM models of melanoma. Cancer 2017;123:2089-103. © 2017 American Cancer Society.


Asunto(s)
Modelos Animales de Enfermedad , Melanoma/genética , Ratones , Neoplasias Cutáneas/genética , Animales , Ingeniería Genética , Ratones Noqueados , Ratones Transgénicos , Transcriptoma
18.
Sci China Life Sci ; 59(8): 764-9, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27417567

RESUMEN

Calcium is a key regulator of many physiological processes that are perturbed in cancer, such as migration, proliferation and apoptosis. The proteins STIM and Orai mediate store-operated calcium entry (SOCE), the main pathway for calcium entry in non-excitable cells. Changes in the expression and function of STIM and Orai have been found in a range of cancer types and thus implicated in disease progression. Here we discuss the role of STIM, Orai and the SOCE pathway in the progression of melanoma and explore how the heterogeneous nature of melanoma may explain the lack of consensus in the field regarding the role of SOCE in the progression of this disease.


Asunto(s)
Calcio/metabolismo , Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Melanoma/metabolismo , Animales , Señalización del Calcio , Humanos , Modelos Biológicos , Proteína ORAI1/metabolismo , Molécula de Interacción Estromal 1/metabolismo
19.
Blood ; 127(25): 3270-80, 2016 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-27143255

RESUMEN

Alloreactive T cells play a critical role in eliminating hematopoietic malignant cells but are also the mediators of graft-versus-host disease (GVHD), a major complication that subverts the success of allogeneic hematopoietic stem cell transplantation (HSCT). However, induction of alloreactive T cells does not necessarily lead to GVHD. Here we report the development of a cellular programming approach to render alloreactive T cells incapable of causing severe GVHD in both major histocompatibility complex (MHC)-mismatched and MHC-identical but minor histocompatibility antigen-mismatched mouse models. We established a novel platform that produced δ-like ligand 4-positive dendritic cells (Dll4(hi)DCs) from murine bone marrow using Flt3 ligand and Toll-like receptor agonists. Upon allogeneic Dll4(hi)DC stimulation, CD4(+) naïve T cells underwent effector differentiation and produced high levels of interferon γ (IFN-γ) and interleukin-17 in vitro, depending on Dll4 activation of Notch signaling. Following transfer, allogeneic Dll4(hi)DC-induced T cells were unable to mediate severe GVHD but preserved antileukemic activity, significantly improving the survival of leukemic mice undergoing allogeneic HSCT. This effect of Dll4(hi)DC-induced T cells was associated with their impaired expansion in GVHD target tissues. IFN-γ was important for Dll4(hi)DC programming to reduce GVHD toxicities of alloreactive T cells. Absence of T-cell IFN-γ led to improved survival and expansion of Dll4(hi)DC-induced CD4(+) T cells in transplant recipients and caused lethal GVHD. Our findings demonstrate that Dll4(hi)DC programming can overcome GVHD toxicity of donor T cells and produce leukemia-reactive T cells for effective immunotherapy.


Asunto(s)
Técnicas de Reprogramación Celular/métodos , Reprogramación Celular , Células Dendríticas/metabolismo , Enfermedad Injerto contra Huésped/prevención & control , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Linfocitos T/inmunología , Animales , Células Dendríticas/fisiología , Enfermedad Injerto contra Huésped/inmunología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Leucemia/inmunología , Leucemia/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Donantes de Tejidos , Trasplante Homólogo
20.
Cancer Cell ; 27(1): 41-56, 2015 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-25584893

RESUMEN

Braf(V600E) induces benign, growth-arrested melanocytic nevus development, but also drives melanoma formation. Cdkn2a loss in Braf(V600E) melanocytes in mice results in rare progression to melanoma, but only after stable growth arrest as nevi. Immediate progression to melanoma is prevented by upregulation of miR-99/100, which downregulates mTOR and IGF1R signaling. mTORC1 activation through Stk11 (Lkb1) loss abrogates growth arrest of Braf(V600E) melanocytic nevi, but is insufficient for complete progression to melanoma. Cdkn2a loss is associated with mTORC2 and Akt activation in human and murine melanocytic neoplasms. Simultaneous Cdkn2a and Lkb1 inactivation in Braf(V600E) melanocytes results in activation of both mTORC1 and mTORC2/Akt, inducing rapid melanoma formation in mice. In this model, activation of both mTORC1/2 is required for Braf-induced melanomagenesis.


Asunto(s)
Melanoma Experimental/patología , Complejos Multiproteicos/metabolismo , Nevo/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas B-raf/metabolismo , Neoplasias Cutáneas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Quinasas Activadas por AMP , Animales , Línea Celular Tumoral , Proliferación Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Melanocitos/metabolismo , Melanoma Experimental/metabolismo , Ratones , MicroARNs/metabolismo , Datos de Secuencia Molecular , Mutación , Nevo/patología , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal , Neoplasias Cutáneas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...