Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 15: 1368572, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38698852

RESUMEN

Introduction: Interferon-gamma (IFN-γ) is pivotal in orchestrating immune responses during healthy pregnancy. However, its dysregulation, often due to autoimmunity, infections, or chronic inflammatory conditions, is implicated in adverse reproductive outcomes such as pregnancy failure or infertility. Additionally, the underlying immunological mechanisms remain elusive. Methods: Here, we explore the impact of systemic IFN-γ elevation on cytotoxic T cell responses in female reproduction utilizing a systemic lupus-prone mouse model with impaired IFN-γ degradation. Results: Our findings reveal that heightened IFN-γ levels triggered the infiltration of CD8+T cells in the pituitary gland and female reproductive tract (FRT), resulting in prolactin deficiency and subsequent infertility. Furthermore, we demonstrate that chronic IFN-γ elevation increases effector memory CD8+T cells in the murine ovary and uterus. Discussion: These insights broaden our understanding of the role of elevated IFN-γ in female reproductive dysfunction and suggest CD8+T cells as potential immunotherapeutic targets in female reproductive disorders associated with chronic systemic IFN-γ elevation.


Asunto(s)
Linfocitos T CD8-positivos , Interferón gamma , Animales , Femenino , Ratones , Embarazo , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Infertilidad Femenina/inmunología , Interferón gamma/metabolismo , Lupus Eritematoso Sistémico/inmunología , Ratones Endogámicos C57BL , Ovario/inmunología , Hipófisis/inmunología , Hipófisis/metabolismo , Prolactina/metabolismo , Útero/inmunología
2.
STAR Protoc ; 4(4): 102710, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37963022

RESUMEN

Here, we present a validated workflow to isolate sufficient viable single ovary cells from a single mouse without the need to pool from several mice. We provide steps essential for estrous staging, ovary harvesting and dissociation, ovary cell staining, data collection, and analysis. Our approach allows the use of these single-cell suspensions for flow sorting, flow cytometry analysis, or functional in vitro assays. Importantly, our protocol is designed to maximize the isolation of immune cells, including T cell subsets.


Asunto(s)
Ovario , Subgrupos de Linfocitos T , Femenino , Animales , Ratones , Citometría de Flujo/métodos
3.
J Pathol ; 260(3): 276-288, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37185821

RESUMEN

The effect of cytokines on non-traditional immunological targets under conditions of chronic inflammation is an ongoing subject of study. Fatigue is a symptom often associated with autoimmune diseases. Chronic inflammatory response and activated cell-mediated immunity are associated with cardiovascular myopathies which can be driven by muscle weakness and fatigue. Thus, we hypothesize that immune dysfunction-driven changes in myocyte mitochondria may play a critical role in fatigue-related pathogenesis. We show that persistent low-level expression of IFN-γ in designated IFN-γ AU-Rich Element deletion mice (ARE mice) under androgen exposure resulted in mitochondrial and metabolic deficiencies in myocytes from male or castrated ARE mice. Most notably, echocardiography unveiled that low ejection fraction in the left ventricle post-stress correlated with mitochondrial deficiencies, explaining how heart function decreases under stress. We report that inefficiencies and structural changes in mitochondria, with changes to expression of mitochondrial genes, are linked to male-biased fatigue and acute cardiomyopathy under stress. Our work highlights how male androgen hormone backgrounds and active autoimmunity reduce mitochondrial function and the ability to cope with stress and how pharmacological blockade of stress signal protects heart function. These studies provide new insight into the diverse actions of IFN-γ in fatigue, energy metabolism, and autoimmunity. © 2023 The Pathological Society of Great Britain and Ireland. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.


Asunto(s)
Andrógenos , Interferón gamma , Animales , Masculino , Ratones , Andrógenos/metabolismo , Citocinas/metabolismo , Inflamación/metabolismo , Mitocondrias/metabolismo , Células Musculares/metabolismo
4.
Front Immunol ; 13: 886645, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35844500

RESUMEN

Immune regulation of female reproductive function plays a crucial role in fertility, as alterations in the relationship between immune and reproductive processes result in autoimmune subfertility or infertility. The breakdown of immune tolerance leads to ovulation dysfunction, implantation failure, and pregnancy loss. In this regard, immune cells with regulatory activities are essential to restore self-tolerance. Apart from regulatory T cells, double negative T regulatory cells (DNTregs) characterized by TCRαß+/γδ+CD3+CD4-CD8- (and negative for natural killer cell markers) are emerging as effector cells capable of mediating immune tolerance in the female reproductive system. DNTregs are present in the female reproductive tract of humans and murine models. However, their full potential as immune regulators is evolving, and studies so far indicate that DNTregs exhibit features that can also maintain tolerance in the female reproductive microenvironment. This review describes recent progress on the presence, role and mechanisms of DNTregs in the female reproductive system immune regulation and tolerance. In addition, we address how DNTregs can potentially provide a paradigm shift from the known roles of conventional regulatory T cells and immune tolerance by maintaining and restoring balance in the reproductive microenvironment of female fertility.


Asunto(s)
Tolerancia Inmunológica , Linfocitos T Reguladores , Animales , Femenino , Fertilidad , Humanos , Ratones , Embarazo , Reproducción , Autotolerancia
5.
Cancer Res ; 81(23): 5977-5990, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34642183

RESUMEN

The relationship between cancer and autoimmunity is complex. However, the incidence of solid tumors such as melanoma has increased significantly among patients with previous or newly diagnosed systemic autoimmune disease (AID). At the same time, immune checkpoint blockade (ICB) therapy of cancer induces de novo autoinflammation and exacerbates underlying AID, even without evident antitumor responses. Recently, systemic lupus erythematosus (SLE) activity was found to drive myeloid-derived suppressor cell (MDSC) formation in patients, a known barrier to healthy immune surveillance and successful cancer immunotherapy. Cross-talk between MDSCs and macrophages generally drives immune suppressive activity in the tumor microenvironment. However, it remains unclear how peripheral pregenerated MDSC under chronic inflammatory conditions modulates global macrophage immune functions and the impact it could have on existing tumors and underlying lupus nephritis. Here we show that pathogenic expansion of SLE-generated MDSCs by melanoma drives global macrophage polarization and simultaneously impacts the severity of lupus nephritis and tumor progression in SLE-prone mice. Molecular and functional data showed that MDSCs interact with autoimmune macrophages and inhibit cell surface expression of CD40 and the production of IL27. Moreover, low CD40/IL27 signaling in tumors correlated with high tumor-associated macrophage infiltration and ICB therapy resistance both in murine and human melanoma exhibiting active IFNγ signatures. These results suggest that preventing global macrophage reprogramming induced by MDSC-mediated inhibition of CD40/IL27 signaling provides a precision melanoma immunotherapy strategy, supporting an original and advantageous approach to treat solid tumors within established autoimmune landscapes. SIGNIFICANCE: Myeloid-derived suppressor cells induce macrophage reprogramming by suppressing CD40/IL27 signaling to drive melanoma progression, simultaneously affecting underlying autoimmune disease and facilitating resistance to immunotherapy within preexisting autoimmune landscapes.


Asunto(s)
Autoinmunidad , Antígenos CD40/metabolismo , Interleucina-27/metabolismo , Lupus Eritematoso Sistémico/fisiopatología , Macrófagos/patología , Melanoma/patología , Células Supresoras de Origen Mieloide/patología , Animales , Inmunoterapia , Macrófagos/inmunología , Macrófagos/metabolismo , Melanoma/inmunología , Melanoma/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Microambiente Tumoral
6.
J Interferon Cytokine Res ; 39(1): 72-84, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30562133

RESUMEN

Today, improvements in diagnostic and therapeutic options allow patients with autoimmune diseases (ADs) to live longer and have more active lives compared with patients receiving conventional anti-inflammatory therapy just two decades ago. Current therapies for ADs aim to inhibit immune cell activation and effector immune pathways, including those activated by cytokines and cytokine receptors. Understandably, such goals become more complicated in patients with long-term established ADs who develop parallel chronic or comorbid conditions, including life-threatening diseases, such as cancer. Compared with the general population, patients with ADs have an increased risk of developing hematological, lymphoproliferative disorders, and solid tumors. However, the aim of current cancer therapies is to activate the immune system to create autoimmune-like conditions and eliminate tumors. As such, their comorbid presentation creates a paradox on how malignancies must be addressed therapeutically in the context of autoimmunity. Because the physiopathology of malignancies is less understood in the context of autoimmunity than it is in the general population, we undertook this review to highlight the peculiarities and mechanisms governing immune cells in established ADs. Moreover, we examined the role of the autoimmune cytokine milieu in the development of immune-related adverse events during the implementation of conventional or immune-based therapy.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Autoinmunidad/inmunología , Citocinas/inmunología , Neoplasias/inmunología , Enfermedades Autoinmunes/terapia , Humanos , Inmunoterapia , Neoplasias/terapia
7.
Hepatology ; 67(4): 1408-1419, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28921595

RESUMEN

We have reported on a murine model of autoimmune cholangitis, generated by altering the AU-rich element (ARE) by deletion of the interferon gamma (IFN-γ) 3' untranslated region (coined ARE-Del-/- ), that has striking similarities to human primary biliary cholangitis (PBC) with female predominance. Previously, we suggested that the sex bias of autoimmune cholangitis was secondary to intense and sustained type I and II IFN signaling. Based on this thesis, and to define the mechanisms that lead to portal inflammation, we specifically addressed the hypothesis that type I IFNs are the driver of this disease. To accomplish these goals, we crossed ARE-Del-/- mice with IFN type I receptor alpha chain (Ifnar1) knockout mice. We report herein that loss of type I IFN receptor signaling in the double construct of ARE-Del-/- Ifnar1-/- mice dramatically reduces liver pathology and abrogated sex bias. More importantly, female ARE-Del-/- mice have an increased number of germinal center (GC) B cells as well as abnormal follicular formation, sites which have been implicated in loss of tolerance. Deletion of type I IFN signaling in ARE-Del-/- Ifnar1-/- mice corrects these GC abnormalities, including abnormal follicular structure. CONCLUSION: Our data implicate type I IFN signaling as a necessary component of the sex bias of this murine model of autoimmune cholangitis. Importantly these data suggest that drugs that target the type I IFN signaling pathway would have potential benefit in the earlier stages of PBC. (Hepatology 2018;67:1408-1419).


Asunto(s)
Enfermedades Autoinmunes/inmunología , Colangitis/inmunología , Interferón Tipo I/genética , Hígado/patología , Animales , Linfocitos B/inmunología , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Hígado/inmunología , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Factores Sexuales , Transducción de Señal/inmunología
8.
J Biol Chem ; 292(34): 13925-13933, 2017 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-28652404

RESUMEN

Interferon γ (IFNγ) is a pleiotropic protein secreted by immune cells. IFNγ signals through the IFNγ receptor, a protein complex that mediates downstream signaling events. Studies into IFNγ signaling have provided insight into the general concepts of receptor signaling, receptor internalization, regulation of distinct signaling pathways, and transcriptional regulation. Although IFNγ is the central mediator of the adaptive immune response to pathogens, it has been shown to be involved in several non-infectious physiological processes. This review will provide an introduction into IFNγ signaling biology and the functional roles of IFNγ in the autoimmune response.


Asunto(s)
Células Presentadoras de Antígenos/metabolismo , Autoinmunidad , Interferón gamma/metabolismo , Modelos Biológicos , Receptores de Interferón/agonistas , Transducción de Señal , Animales , Células Presentadoras de Antígenos/inmunología , Autofagosomas/inmunología , Autofagosomas/metabolismo , Caveolas/inmunología , Caveolas/metabolismo , Vesículas Cubiertas por Clatrina/inmunología , Vesículas Cubiertas por Clatrina/metabolismo , Dimerización , Endocitosis , Humanos , Interferón gamma/química , Macrófagos/inmunología , Macrófagos/metabolismo , Microdominios de Membrana , Multimerización de Proteína , Receptores de Interferón/química , Receptores de Interferón/metabolismo , Receptor de Interferón gamma
9.
Hepatology ; 64(4): 1189-201, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27178326

RESUMEN

UNLABELLED: In most autoimmune diseases the serologic hallmarks of disease precede clinical pathology by years. Therefore, the use of animal models in defining early disease events becomes critical. We took advantage of a "designer" mouse with dysregulation of interferon gamma (IFNγ) characterized by prolonged and chronic expression of IFNγ through deletion of the IFNγ 3'-untranslated region adenylate uridylate-rich element (ARE). The ARE-Del(-/-) mice develop primary biliary cholangitis (PBC) with a female predominance that mimics human PBC that is characterized by up-regulation of total bile acids, spontaneous production of anti-mitochondrial antibodies, and portal duct inflammation. Transfer of CD4 T cells from ARE-Del(-/-) to B6/Rag1(-/-) mice induced moderate portal inflammation and parenchymal inflammation, and RNA sequencing of liver gene expression revealed that up-regulated genes potentially define early stages of cholangitis. Interestingly, up-regulated genes specifically overlap with the gene expression signature of biliary epithelial cells in PBC, implying that IFNγ may play a pathogenic role in biliary epithelial cells in the initiation stage of PBC. Moreover, differentially expressed genes in female mice have stronger type 1 and type 2 IFN signaling and lymphocyte-mediated immune responses and thus may drive the female bias of the disease. CONCLUSION: Changes in IFNγ expression are critical for the pathogenesis of PBC. (Hepatology 2016;64:1189-1201).


Asunto(s)
Enfermedades Autoinmunes/etiología , Colangitis/inmunología , Interferón gamma/biosíntesis , Animales , Enfermedades Autoinmunes/metabolismo , Colangitis/metabolismo , Femenino , Masculino , Ratones , Factores Sexuales
10.
J Invest Dermatol ; 135(10): 2455-2463, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25950827

RESUMEN

More than 375 genes have been identified that are involved in regulating skin pigmentation and these act during development, survival, differentiation, and/or responses of melanocytes to the environment. Many of these genes have been cloned, and disruptions of their functions are associated with various pigmentary diseases; however, many remain to be identified. We have performed a series of microarray analyses of hyperpigmented compared with less pigmented skin to identify genes responsible for these differences. The rationale and goal for this study was to perform a meta-analysis on these microarray databases to identify genes that may be significantly involved in regulating skin phenotype either directly or indirectly that might not have been identified due to subtle differences by any of these individual studies alone. The meta-analysis demonstrates that 1,271 probes representing 921 genes are differentially expressed at significant levels in the 5 microarray data sets compared, providing new insights into the variety of genes involved in determining skin phenotype. Immunohistochemistry was used to validate two of these markers at the protein level (TRIM63 and QPCT), and we discuss the possible functions of these genes in regulating skin physiology.


Asunto(s)
Proteínas Portadoras/genética , Bases de Datos Genéticas , Regulación de la Expresión Génica , Hiperpigmentación/genética , Análisis por Micromatrices , Proteínas Musculares/genética , Ubiquitina-Proteína Ligasas/genética , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Reproducibilidad de los Resultados , Pigmentación de la Piel/genética , Proteínas de Motivos Tripartitos , Regulación hacia Arriba
11.
Cancer Res ; 75(13): 2708-15, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25832654

RESUMEN

The AMPK-related kinase NUAK2 has been implicated in melanoma growth and survival outcomes, but its therapeutic utility has yet to be confirmed. In this study, we show how its genetic amplification in PTEN-deficient melanomas may rationalize the use of CDK2 inhibitors as a therapeutic strategy. Analysis of array-CGH data revealed that PTEN deficiency is coupled tightly with genomic amplification encompassing the NUAK2 locus, a finding strengthened by immunohistochemical evidence that phospho-Akt overexpression was correlated with NUAK2 expression in clinical specimens of acral melanoma. Functional studies in melanoma cells showed that inactivation of the PI3K pathway upregulated p21 expression and reduced the number of cells in S phase. NUAK2 silencing and inactivation of the PI3K pathway efficiently controlled CDK2 expression, whereas CDK2 inactivation specifically abrogated the growth of NUAK2-amplified and PTEN-deficient melanoma cells. Immunohistochemical analyses confirmed an association of CDK2 expression with NUAK2 amplification and p-Akt expression in melanomas. Finally, pharmacologic inhibition of CDK2 was sufficient to suppress the growth of NUAK2-amplified and PTEN-deficient melanoma cells in vitro and in vivo. Overall, our results show how CDK2 blockade may offer a promising therapy for genetically defined melanomas, where NUAK2 is amplified and PTEN is deleted.


Asunto(s)
Quinasa 2 Dependiente de la Ciclina/metabolismo , Melanoma/genética , Fosfohidrolasa PTEN/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Neoplasias Cutáneas/genética , Anciano , Animales , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/genética , Línea Celular Tumoral , Quinasa 2 Dependiente de la Ciclina/antagonistas & inhibidores , Amplificación de Genes , Humanos , Masculino , Melanoma/tratamiento farmacológico , Melanoma/enzimología , Melanoma/patología , Ratones , Ratones Desnudos , Persona de Mediana Edad , Terapia Molecular Dirigida , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Purinas/farmacología , Roscovitina , Transducción de Señal , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/enzimología , Neoplasias Cutáneas/patología
12.
Chest ; 147(3): 771-777, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25411763

RESUMEN

BACKGROUND: Lymphangioleiomyomatosis (LAM) is characterized by the proliferation in the lung, axial lymphatics (eg, lymphangioleiomyomas), and kidney (eg, angiomyolipomas) of abnormal smooth muscle-like LAM cells, which express melanoma antigens such as Pmel17/gp100 and have dysfunctional tumor suppressor tuberous sclerosis complex (TSC) genes TSC2 or TSC1. Histopathologic diagnosis of LAM in lung specimens is based on identification of the Pmel17 protein with the monoclonal antibody HMB-45. METHODS: We compared the sensitivity of HMB-45 to that of antipeptide antibody αPEP13h, which reacts with a C-terminal peptide of Pmel17. LAM lung nodules were laser-capture microdissected to identify proteins by Western blotting. RESULTS: HMB-45 recognized approximately 25% of LAM cells within the LAM lung nodules, whereas αPEP13h identified > 82% of LAM cells within these structures in approximately 90% of patients. Whereas HMB-45 reacted with epithelioid but not with spindle-shaped LAM cells, αPEP13h identified both spindle-shaped and epithelioid LAM cells, providing greater sensitivity for detection of all types of LAM cells. HMB-45 recognized Pmel17 in premelanosomal organelles; αPEP13h recognized proteins in the cytoplasm as well as in premelanosomal organelles. Both antibodies recognized a Pmel17 variant of approximately 50 kDa. CONCLUSIONS: Based on its sensitivity and specificity, αPEP13h may be useful in the diagnosis of LAM and more sensitive than HMB-45.


Asunto(s)
Anticuerpos Antiidiotipos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/patología , Linfangioleiomiomatosis/diagnóstico , Linfangioleiomiomatosis/patología , Nódulo Pulmonar Solitario/diagnóstico , Nódulo Pulmonar Solitario/patología , Adulto , Anticuerpos Antiidiotipos/inmunología , Biopsia , Broncoscopía , Células Cultivadas , Diagnóstico Diferencial , Femenino , Humanos , Inmunohistoquímica , Pulmón/patología , Neoplasias Pulmonares/inmunología , Linfangioleiomiomatosis/inmunología , Antígenos Específicos del Melanoma/inmunología , Persona de Mediana Edad , Sensibilidad y Especificidad , Nódulo Pulmonar Solitario/inmunología , Antígeno gp100 del Melanoma/inmunología
13.
J Dermatol Sci ; 77(1): 21-7, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25530116

RESUMEN

BACKGROUND: Patients with oculocutaneous albinism (OCA) have severely decreased pigmentation of their skin, hair and eyes. OCA2 and OCA4 result from mutations of the OCA2 and SLC45A2 genes, respectively, both of which disrupt the trafficking of the critical melanogenic enzyme tyrosinase to melanosomes. Both proteins encoded by those loci (termed P and MATP, respectively) have 12 putative transmembrane regions and are thought to function as transporters, although their functions and subcellular localizations remain to be characterized. OBJECTIVE: To generate specific antibodies against unique synthetic peptides encoded by P and MATP that could be used to characterize their functions and subcellular localizations. METHODS: Western blotting and immunohistochemistry were used to assess the specificity of antibodies and to colocalize P and MATP proteins with various subcellular markers. RESULTS: Specific antibodies to the P and MATP proteins were generated that work well for Western blotting and immunohistochemistry. The localizations of P and MATP with various subcellular organelles were characterized using confocal microscopy, which revealed that they colocalize to some extent with LAMP2, but do not significantly colocalize with markers of the ER, Golgi or melanosomes. Interestingly, both P and MATP colocalize significantly with BLOC-1, a sorting component involved in the intracellular trafficking of melanosomal/lysosomal constituents. CONCLUSION: These results provide a basis to understand how disrupted functions of P or MATP result in the misrouting of tyrosinase and cause the hypopigmentation seen in OCA2 and OCA4.


Asunto(s)
Albinismo Oculocutáneo/inmunología , Anticuerpos/química , Hipopigmentación/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Secuencia de Aminoácidos , Antígenos de Neoplasias/metabolismo , Transporte Biológico , Western Blotting , Línea Celular Tumoral , Células Cultivadas , Femenino , Humanos , Inmunohistoquímica , Masculino , Melanocitos/citología , Melanosomas/inmunología , Melanosomas/metabolismo , Datos de Secuencia Molecular , Monofenol Monooxigenasa/inmunología , Monofenol Monooxigenasa/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Péptidos/química , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo
14.
J Pathol ; 236(1): 17-29, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25488118

RESUMEN

Human skin colour, ie pigmentation, differs widely among individuals, as do their responses to various types of ultraviolet radiation (UV) and their risks of skin cancer. In some individuals, UV-induced pigmentation persists for months to years in a phenomenon termed long-lasting pigmentation (LLP). It is unclear whether LLP is an indicator of potential risk for skin cancer. LLP seems to have similar features to other forms of hyperpigmentation, eg solar lentigines or age spots, which are clinical markers of photodamage and risk factors for precancerous lesions. To investigate what UV-induced molecular changes may persist in individuals with LLP, clinical specimens from non-sunburn-inducing repeated UV exposures (UVA, UVB or UVA + UVB) at 4 months post-exposure (short-term LLP) were evaluated by microarray analysis and dataset mining. Validated targets were further evaluated in clinical specimens from six healthy individuals (three LLP+ and three LLP-) followed for more than 9 months (long-term LLP) who initially received a single sunburn-inducing UVA + UVB exposure. The results support a UV-induced hyperpigmentation model in which basal keratinocytes have an impaired ability to remove melanin that leads to a compensatory mechanism by neighbouring keratinocytes with increased proliferative capacity to maintain skin homeostasis. The attenuated expression of SOX7 and other hemidesmosomal components (integrin α6ß4 and plectin) leads to increased melanosome uptake by keratinocytes and points to a spatial regulation within the epidermis. The reduced density of hemidesmosomes provides supporting evidence for plasticity at the epidermal-dermal junction. Altered hemidesmosome plasticity, and the sustained nature of LLP, may be mediated by the role of SOX7 in basal keratinocytes. The long-term sustained subtle changes detected are modest, but sufficient to create dramatic visual differences in skin colour. These results suggest that the hyperpigmentation phenomenon leading to increased interdigitation develops in order to maintain normal skin homeostasis in individuals with LLP.


Asunto(s)
Epidermis/metabolismo , Hemidesmosomas/metabolismo , Queratinocitos/metabolismo , Pigmentación de la Piel/efectos de la radiación , Piel/metabolismo , Rayos Ultravioleta/efectos adversos , Células Cultivadas , Epidermis/efectos de la radiación , Hemidesmosomas/efectos de la radiación , Humanos , Queratinocitos/efectos de la radiación , Piel/efectos de la radiación , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Tiempo
15.
Pigment Cell Melanoma Res ; 27(1): 82-9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24024552

RESUMEN

Through a process known as melanogenesis, melanocyte produces melanin in specialized organelles termed melanosomes, which regulates pigmentation of the skin, eyes, and hair. Gp96 is a constitutively expressed heat shock protein in the endoplasmic reticulum whose expression is further upregulated upon ultraviolet irradiation. However, the roles and mechanisms of this chaperone in pigmentation biology are unknown. In this study, we found that knockdown of gp96 by RNA interference significantly perturbed melanin synthesis and blocked late melanosome maturation. Gp96 knockdown did not impair the expression of tyrosinase, an essential enzyme in melanin synthesis, but compromised its catalytic activity and melanosome translocation. Further, mice with melanocyte-specific deletion of gp96 displayed decreased pigmentation. A mechanistic study revealed that the defect in melanogenesis can be rescued by activation of the canonical Wnt pathway, consistent with the critical roles of gp96 in chaperoning Wnt-coreceptor LRP6. Thus, this work uncovered the essential role of gp96 in regulating melanogenesis.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Melaninas/biosíntesis , Melanosomas/metabolismo , Glicoproteínas de Membrana/biosíntesis , Pigmentación de la Piel/fisiología , Animales , Línea Celular , Melaninas/genética , Melanosomas/genética , Glicoproteínas de Membrana/genética , Ratones , Ratones Transgénicos
16.
J Invest Dermatol ; 132(12): 2791-9, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22895365

RESUMEN

Diacylglycerol (DAG) increases the melanin content of human melanocytes in vitro and increases the pigmentation of guinea pig skin in vivo, but the mechanism(s) underlying those effects remain unknown. In this study, we characterized the role of diacylglycerol kinase (DGK), which phosphorylates DAG to generate phosphatidic acid, in the regulation of pigmentation. Ten isoforms of DGK have been identified, and we show that DGKζ is the most abundant isoform expressed by human melanocytic cells. Melanin content, tyrosinase activity, and tyrosinase protein levels were significantly reduced by a DGK inhibitor, but tyrosinase and microphthalmia-associated transcription factor messenger RNA (mRNA) levels were not changed by that inhibition, and there were no effects on the expression of other melanogenesis-related proteins. Isoform-specific small interfering RNAs showed that knockdown of DGKζ decreased melanin content and tyrosinase expression in melanocytic cells. Overexpression of DGKζ increased tyrosinase protein levels, but did not increase tyrosinase mRNA levels. Glycosidase digestion revealed that inhibition of DGK reduced only the mature form of tyrosinase, and the decrease of tyrosinase resulting from DGK inhibition could be blocked partially by protease inhibitors. These results suggest that DGK regulates melanogenesis via modulation of the posttranslational processing of tyrosinase, which may be related with the protein degradation machinery.


Asunto(s)
Diacilglicerol Quinasa/metabolismo , Regulación Enzimológica de la Expresión Génica/fisiología , Melanocitos/enzimología , Monofenol Monooxigenasa/genética , Adenoviridae/genética , Animales , Línea Celular Tumoral , Diacilglicerol Quinasa/antagonistas & inhibidores , Diacilglicerol Quinasa/genética , Retículo Endoplásmico/enzimología , Inhibidores Enzimáticos/farmacología , Células Epidérmicas , Flavonoides/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hormonas/metabolismo , Hormonas/farmacología , Humanos , Indoles/farmacología , Maleimidas/farmacología , Melaninas/biosíntesis , Melaninas/metabolismo , Melanocitos/citología , Melanoma Experimental , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Procesamiento Proteico-Postraduccional/fisiología , Quinazolinonas/farmacología , Neoplasias Cutáneas , alfa-MSH/metabolismo , alfa-MSH/farmacología
17.
Proc Natl Acad Sci U S A ; 108(16): 6597-602, 2011 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-21460252

RESUMEN

The identification of genes that participate in melanomagenesis should suggest strategies for developing therapeutic modalities. We used a public array comparative genomic hybridization (CGH) database and real-time quantitative PCR (qPCR) analyses to identify the AMP kinase (AMPK)-related kinase NUAK2 as a candidate gene for melanomagenesis, and we analyzed its functions in melanoma cells. Our analyses had identified a locus at 1q32 where genomic gain is strongly associated with tumor thickness, and we used real-time qPCR analyses and regression analyses to identify NUAK2 as a candidate gene at that locus. Associations of relapse-free survival and overall survival of 92 primary melanoma patients with NUAK2 expression measured using immunohistochemistry were investigated using Kaplan-Meier curves, log rank tests, and Cox regression models. Knockdown of NUAK2 induces senescence and reduces S-phase, decreases migration, and down-regulates expression of mammalian target of rapamycin (mTOR). In vivo analysis demonstrated that knockdown of NUAK2 suppresses melanoma tumor growth in mice. Survival analysis showed that the risk of relapse is greater in acral melanoma patients with high levels of NUAK2 expression than in acral melanoma patients with low levels of NUAK2 expression (hazard ratio = 3.88; 95% confidence interval = 1.44-10.50; P = 0.0075). These data demonstrate that NUAK2 expression is significantly associated with the oncogenic features of melanoma cells and with the survival of acral melanoma patients. NUAK2 may provide a drug target to suppress melanoma progression. This study further supports the importance of NUAK2 in cancer development and tumor progression, while AMPK has antioncogenic properties.


Asunto(s)
Movimiento Celular , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Melanoma/enzimología , Melanoma/mortalidad , Proteínas de Neoplasias/biosíntesis , Proteínas Serina-Treonina Quinasas/biosíntesis , Animales , Senescencia Celular/genética , Supervivencia sin Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Sitios Genéticos/genética , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Melanoma/genética , Melanoma/patología , Melanoma/terapia , Ratones , Ratones Desnudos , Proteínas de Neoplasias/genética , Proteínas Serina-Treonina Quinasas/genética , Fase S/genética , Tasa de Supervivencia , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
18.
Pigment Cell Melanoma Res ; 22(6): 740-9, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19725928

RESUMEN

Because melanomas are intrinsically resistant to conventional radiotherapy and chemotherapy, many alternative treatment approaches have been developed such as biochemotherapy and immunotherapy. The most common cause of multidrug resistance (MDR) in human cancers is the expression and function of one or more ATP-binding cassette (ABC) transporters that efflux anticancer drugs from cells. Melanoma cells express a group of ABC transporters (such as ABCA9, ABCB1, ABCB5, ABCB8, ABCC1, ABCC2, and ABCD1) that may be associated with the resistance of melanoma cells to a broad range of anticancer drugs and/or of melanocytes to toxic melanin intermediates and metabolites. In this review, we propose a model (termed the ABC-M model) in which the intrinsic MDR of melanoma cells is at least in part because of the transporter systems that may also play a critical role in reducing the cytotoxicity of the melanogenic pathway in melanocytes. The ABC-M model suggests molecular strategies to reverse MDR function in the context of the melanogenic pathway, which could open therapeutic avenues towards the ultimate goal of circumventing clinical MDR in patients with melanoma.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Resistencia a Múltiples Medicamentos/fisiología , Resistencia a Antineoplásicos/fisiología , Melaninas/biosíntesis , Melanoma/patología , Melanoma/fisiopatología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Antineoplásicos/metabolismo , Antineoplásicos/uso terapéutico , Humanos , Melanocitos/citología , Melanocitos/fisiología , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Melanosomas/metabolismo , Modelos Biológicos , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Metástasis de la Neoplasia , Células Madre Neoplásicas/metabolismo
19.
J Natl Cancer Inst ; 101(18): 1259-71, 2009 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-19704071

RESUMEN

BACKGROUND: Malignant melanomas are intrinsically resistant to many conventional treatments, such as radiation and chemotherapy, for reasons that are poorly understood. Here we propose and test a model that explains drug resistance or sensitivity in terms of melanosome dynamics. METHODS: The growth and sensitivity to cisplatin of MNT-1 cells, which are melanotic and enriched with mature stage III and IV melanosomes, and SK-MEL-28 cells, which have only immature stage I and II melanosomes, were compared using clonogenic assays. Differences in pigmentation, melanosome stages, melanosome number, and cellular structures in different cell lines in response to various treatments were examined by electron microscopy. The relative numbers of melanosomes of different stages were compared after treatment with 1-phenyl-2-thiourea. The relationship between drug transporter function and endogenous melanogenic toxicity was assessed by treating cells with the cyclosporin analog PSC-833 and by assessing vacuole formation and cell growth inhibition. All statistical tests were two-sided. RESULTS: Endogenous melanogenic cytotoxicity, produced by damaged melanosomes, resulted in pronounced cell growth inhibition in MNT-1 cells compared with amelanotic SK-MEL-28 cells. The sensitivity to CDDP of MNT-1 cells was 3.8-fold higher than that of SK-MEL-28 cells (mean IC(50) for SK-MEL-28 and MNT-1 = 2.13 microM and 0.56 microM, respectively; difference = 1.57 microM, 95% confidence interval = 1.45 to 1.69; P = .0017). After treatment with 6.7 microM CDDP for 72 hours, the number of stage II-III melanosomes in surviving MNT-1 cells was 6.8-fold that of untreated cells. Modulation of MNT-1 cells to earlier-stage (II, II-III, III) melanosomes by treatment with the tyrosinase inhibitor 1-phenyl-2-thiourea dramatically increased CDDP resistance. Furthermore, PSC-833 principally suppressed MNT-1 melanotic cell growth via an elevation of autophagosome-like vacuolar structures, possibly by inhibiting melanosome membrane transporters. CONCLUSIONS: Melanosome dynamics (including their biogenesis, density, status, and structural integrity) regulate the drug resistance of melanoma cells. Manipulation of melanosome functions may be an effective way to enhance the therapeutic activity of anticancer drugs against melanoma.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Resistencia a Antineoplásicos , Melanoma/tratamiento farmacológico , Melanoma/patología , Melanosomas/efectos de los fármacos , Melanosomas/ultraestructura , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología , Animales , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Cisplatino/farmacología , Ciclosporinas/farmacología , Doxorrubicina/farmacología , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/patología , Ratones , Microscopía Confocal , Microscopía Electrónica , Verapamilo/farmacología , Vinblastina/farmacología
20.
J Cell Physiol ; 220(3): 640-54, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19452503

RESUMEN

The melanocortin 1 receptor (MC1R), a Gs protein-coupled receptor (GPCR) expressed in melanocytes, is a major determinant of skin pigmentation and phototype. MC1R activation stimulates melanogenesis and increases the ratio of black, strongly photoprotective eumelanins to reddish, poorly photoprotective pheomelanins. Several MC1R alleles are associated with red hair, fair skin, increased sensitivity to ultraviolet radiation (the RHC phenotype) and increased skin cancer risk. Three highly penetrant RHC variants, R151C, R160W, and D294H are loss-of-function MC1R mutants with altered cell surface expression. In this study, we show that forward trafficking was normal for D294H. Conversely, export traffic was impaired for R151C, which accumulated in the endoplasmic reticulum (ER), and for R160W, which was enriched in the cis-Golgi. This is the first report of steady-state retention in a post-ER secretory compartment of a GPCR mutant found in the human population. Residues R151 and R160 are located in the MC1R second intracellular loop (il2). Two other mutations in il2, T157A preventing T157 phosphorylation and R162P disrupting a (160)RARR(163) motif, also caused intracellular retention. Moreover, T157 was phosphorylated in wild-type MC1R and a T157D mutation mimicking constitutive phosphorylation allowed normal traffic, and rescued the retention phenotype of R160W and R162P. Therefore, MC1R export is likely regulated by T157 phosphorylation and the (160)RARR(163) arginine-based motif functions as an ER retrieval signal. These elements are conserved in mammalian MC1Rs and in all five types of human melanocortin receptors. Thus, members of this GPCR subfamily might share common mechanisms for regulation of plasma membrane expression.


Asunto(s)
Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Color del Cabello , Melanocitos/metabolismo , Receptor de Melanocortina Tipo 1/metabolismo , Neoplasias Cutáneas/metabolismo , Pigmentación de la Piel , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Línea Celular , Humanos , Datos de Secuencia Molecular , Mutación , Fosforilación , Conformación Proteica , Transporte de Proteínas , Receptor de Melanocortina Tipo 1/genética , Neoplasias Cutáneas/genética , Relación Estructura-Actividad , Factores de Tiempo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA