Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
EMBO J ; 40(7): e106151, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33616251

RESUMEN

Interleukin (IL)-33 cytokine plays a critical role in allergic diseases and cancer. IL-33 also has a nuclear localization signal. However, the nuclear function of IL-33 and its impact on cancer is unknown. Here, we demonstrate that nuclear IL-33-mediated activation of SMAD signaling pathway in epithelial cells is essential for cancer development in chronic inflammation. Using RNA and ChIP sequencing, we found that nuclear IL-33 repressed the expression of an inhibitory SMAD, Smad6, by interacting with its transcription factor, RUNX2. IL-33 was highly expressed in the skin and pancreatic epithelial cells in chronic inflammation, leading to a markedly repressed Smad6 expression as well as dramatically upregulated p-SMAD2/3 and p-SMAD1/5 in the epithelial cells. Blocking TGF-ß/SMAD signaling attenuated the IL-33-induced cell proliferation in vitro and inhibited IL-33-dependent epidermal hyperplasia and skin cancer development in vivo. IL-33 and SMAD signaling were upregulated in human skin cancer, pancreatitis, and pancreatitis-associated pancreatic cancer. Collectively, our findings reveal that nuclear IL-33/SMAD signaling is a cell-autonomous tumor-promoting axis in chronic inflammation, which can be targeted by small-molecule inhibitors for cancer treatment and prevention.


Asunto(s)
Carcinogénesis/metabolismo , Interleucina-33/metabolismo , Neoplasias Pancreáticas/metabolismo , Transducción de Señal , Neoplasias Cutáneas/metabolismo , Proteína smad6/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Núcleo Celular/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Factor de Crecimiento Transformador beta/metabolismo
2.
Oncologist ; 27(9): e759-e761, 2022 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-35867136

RESUMEN

Skin cancer is the leading malignancy in immunosuppressed patients, including organ transplant recipients (OTRs), which is increasing in incidence as OTRs live longer. We performed a single-center case series of 4 patients with scalp pleomorphic dermal sarcoma and a history of multiple keratinocyte carcinomas. Outcomes included incidence of dermal sarcoma, dermal sarcoma-related mortality, and histopathologic findings. Out of more than 200 patients followed over a 3-year period in Massachusetts General Hospital High Risk Skin Cancer Clinics, all skin cancer-related deaths (2/2) were due to metastatic dermal sarcoma. Three of 4 patients diagnosed with scalp dermal sarcoma were OTRs and had been on at least one immunosuppressive medication for a median of 9 years. For patients who died from dermal sarcoma, the median time between diagnosis and death was 6 months. Our findings suggest pleomorphic dermal sarcoma contributes to skin cancer-related morbidity and mortality in OTRs.


Asunto(s)
Trasplante de Órganos , Sarcoma , Neoplasias Cutáneas , Humanos , Huésped Inmunocomprometido , Trasplante de Órganos/efectos adversos , Sarcoma/complicaciones , Neoplasias Cutáneas/patología , Receptores de Trasplantes
3.
Proc Natl Acad Sci U S A ; 116(7): 2646-2651, 2019 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-30696763

RESUMEN

Chronic inflammation's tumor-promoting potential is well-recognized; however, the mechanism underlying the development of this immune environment is unknown. Studying the transition from acute, tumor-suppressive to chronic, tumor-promoting allergic contact dermatitis (ACD) revealed how tumor-promoting chronic inflammation develops. Epidermis-derived interleukin (IL)-33 up-regulation and its induction of regulatory T cell (Treg) accumulation in the skin preceded the transition from acute to chronic ACD and triggered the tumor-promoting immune environment in chronic ACD. Mice lacking IL-33 were protected from chronic ACD and its skin cancer sequela compared with wild-type controls (P = 0.0002). IL-33's direct signaling onto Tregs was required for the development of the tumor-promoting immune environment in the skin. IL-33-Treg signaling was also required for chronic colitis and its associated colorectal cancer development in a colitis model (P < 0.0001). Significantly increased IL-33 and Tregs marked the perilesional skin and colon in patients with cancer-prone chronic inflammatory diseases. Our findings elucidate the role of the IL-33/Treg axis in creating a tumor-promoting immune environment in chronic inflammatory diseases and suggest therapeutic targets for cancer prevention and treatment in high-risk patients.


Asunto(s)
Colitis/inmunología , Neoplasias Colorrectales/inmunología , Dermatitis Alérgica por Contacto/inmunología , Interleucina-33/inmunología , Linfocitos T Reguladores/inmunología , Animales , Enfermedad Crónica , Colitis/complicaciones , Neoplasias Colorrectales/complicaciones , Dermatitis Alérgica por Contacto/complicaciones , Humanos , Proteína 1 Similar al Receptor de Interleucina-1/genética , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Ratones , Ratones Noqueados , Neoplasias Cutáneas/complicaciones , Neoplasias Cutáneas/inmunología , Regulación hacia Arriba
4.
Oncologist ; 25(5): 366-368, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32073194

RESUMEN

Immune checkpoint blockade (ICB) is highly effective for the treatment of metastatic cancers, but its side effects are incompletely understood. The objective of this article is to highlight hypertrophic lichen planus (HLP) with histological features diagnosed as squamous cell carcinoma (SCC), which is a potential cutaneous reaction to ICB. Two patients (75 and 69 years) presented with lesions diagnosed as SCC on biopsy, which developed after 3-9 months on ICB therapy. Biopsies demonstrated endophytic, atypical, or cystic squamous proliferations consistent with cutaneous SCC. However, the clinical presentation including monomorphic nature of the lesions and lichenoid inflammation in the background were consistent with HLP. Patients initially received topical 5-fluorouracil (5-FU) to reduce the hyperkeratotic lesions followed by topical steroids. The eruptions readily responded to this treatment regimen. Dermatologic immune-related adverse events (irAEs) are the most common irAEs associated with ICB therapy. Our findings indicate that HLP resembling SCC on biopsy is a potential side effect of ICB that can be correctly diagnosed on careful clinical exam and is responsive to ICB cessation and topical steroid with or without 5-FU treatment. KEY POINTS: Immune checkpoint blockade is associated with cutaneous immune-related adverse events including lichen planus. Hypertrophic lichen planus can appear as squamous cell carcinoma histologically and clinical context is key for the proper diagnosis. Hypertrophic lichen planus can be safely treated with topical steroids with or without topical 5-fluorouracil in cases with severe hyperkeratotic lesions. Immune checkpoint blockade may be safely continued if clinical presentation is consistent with hypertrophic lichen planus.


Asunto(s)
Carcinoma de Células Escamosas , Liquen Plano , Neoplasias Cutáneas , Carcinoma de Células Escamosas/tratamiento farmacológico , Humanos , Inhibidores de Puntos de Control Inmunológico , Liquen Plano/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico
6.
Mol Cancer ; 12: 87, 2013 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-23915220

RESUMEN

BACKGROUND: Adrenocortical carcinoma (ACC) is a rare endocrine malignancy with high mutational heterogeneity and a generally poor clinical outcome. Despite implicated roles of deregulated TP53, IGF-2 and Wnt signaling pathways, a clear genetic association or unique mutational link to the disease is still missing. Recent studies suggest a crucial role for epigenetic modifications in the genesis and/or progression of ACC. This study specifically evaluates the potential role of epigenetic silencing of RASSF1A, the most commonly silenced tumor suppressor gene, in adrenocortical malignancy. RESULTS: Using adrenocortical tumor and normal tissue specimens, we show a significant reduction in expression of RASSF1A mRNA and protein in ACC. Methylation-sensitive and -dependent restriction enzyme based PCR assays revealed significant DNA hypermethylation of the RASSF1A promoter, suggesting an epigenetic mechanism for RASSF1A silencing in ACC. Conversely, the RASSF1A promoter methylation profile in benign adrenocortical adenomas (ACAs) was found to be very similar to that found in normal adrenal cortex. Enforced expression of ectopic RASSF1A in the SW-13 ACC cell line reduced the overall malignant behavior of the cells, which included impairment of invasion through the basement membrane, cell motility, and solitary cell survival and growth. On the other hand, expression of RASSF1A/A133S, a loss-of-function mutant form of RASSF1A, failed to elicit similar malignancy-suppressing responses in ACC cells. Moreover, association of RASSF1A with the cytoskeleton in RASSF1A-expressing ACC cells and normal adrenal cortex suggests a role for RASSF1A in modulating microtubule dynamics in the adrenal cortex, and thereby potentially blocking malignant progression. CONCLUSIONS: Downregulation of RASSF1A via promoter hypermethylation may play a role in the malignant progression of adrenocortical carcinoma possibly by abrogating differentiation-promoting RASSF1A- microtubule interactions.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/genética , Carcinoma Corticosuprarrenal/genética , Citoesqueleto/metabolismo , Epigénesis Genética , Silenciador del Gen , Proteínas Supresoras de Tumor/genética , Neoplasias de la Corteza Suprarrenal/metabolismo , Neoplasias de la Corteza Suprarrenal/patología , Carcinoma Corticosuprarrenal/metabolismo , Carcinoma Corticosuprarrenal/patología , Adulto , Línea Celular Tumoral , Islas de CpG , Metilación de ADN , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Microtúbulos/metabolismo , Persona de Mediana Edad , Estadificación de Neoplasias , Fenotipo , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Supresoras de Tumor/metabolismo
7.
J Invest Dermatol ; 142(10): 2668-2676.e4, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35341735

RESUMEN

Fibrosis is a pathological hallmark of systemic sclerosis, a deadly autoimmune disease affecting the connective tissues of multiple organs. However, the immune mechanisms underlying fibrosis and systemic sclerosis remain unclear. To determine the initiating immune pathway in fibrosis, we investigated the role of type 2 alarmin cytokines in the mouse model of skin fibrosis. Wild-type mice that received subcutaneous bleomycin injections developed skin fibrosis accompanied by elevated IL-33 expression in the dermis. Likewise, we found IL-33 upregulation in human skin fibrosis. Mice with germline deletion of IL-33 receptor (ST2 knockout) showed markedly exacerbated skin fibrosis in association with significantly increased T helper 2 cell to regulatory T-cell ratio in the skin. Mice that lacked ST2 specifically on regulatory T cells (Foxp3Cre,ST2flox/flox) showed significantly worse skin fibrosis, increased T helper 2 to regulatory T cell ratio and IL-13 expression in the skin compared with wild-type mice. Our findings show that IL-33 cytokine signaling to regulatory T cells suppresses skin fibrosis and highlight a potential therapeutic axis to alleviate the debilitating manifestations of systemic sclerosis.


Asunto(s)
Interleucina-33 , Esclerodermia Sistémica , Linfocitos T Reguladores , Alarminas/metabolismo , Animales , Bleomicina , Citocinas/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Factores de Transcripción Forkhead/metabolismo , Humanos , Proteína 1 Similar al Receptor de Interleucina-1/genética , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-13/metabolismo , Interleucina-33/metabolismo , Ratones , Piel/patología , Linfocitos T Reguladores/metabolismo
8.
J Invest Dermatol ; 139(1): 62-70, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30030152

RESUMEN

High-risk skin cancer is a rare, but severe, complication associated with discoid lupus erythematosus (DLE). Chronic scar, inflammation, UVR, and immunosuppressive medications are proposed explanations for this heightened skin cancer risk; however, the exact mechanism driving skin carcinogenesis in DLE is unknown. The distinct co-localization of multiple independent skin cancers with areas of active inflammation in two DLE patients followed over 8 years strongly suggested that lupus inflammation promotes skin carcinogenesis in DLE. To investigate this clinical observation, we subjected lupus-prone MRL/lpr and control (MRL/n) mice to a skin carcinogenesis protocol. Skin tumors developed preferentially within the cutaneous lupus inflammation without scarring in MRL/lpr mice (P < 0.01). The inflammation in MRL/lpr skin was characterized by the accumulation of regulatory T cells, mast cells, M2 macrophages, and markedly elevated transforming growth factor-ß1 and IL-6 levels, which have been linked to tumor promotion. Tacrolimus treatment reduced skin inflammation and blocked cancer development in MRL/lpr mice (P = 0.0195). A similar tumor-promoting immune environment was detected in SCCs and the perilesional skin of cancer-prone DLE patients. Therefore, discoid lupus inflammation promotes skin cancer in high-risk DLE patients, and blocking the inflammation may be critical for preventing this life-threatening complication of DLE.


Asunto(s)
Citocinas/metabolismo , Inflamación/patología , Lupus Eritematoso Discoide/patología , Neoplasias Cutáneas/etiología , Piel/patología , Animales , Carcinogénesis , Enfermedad Crónica , Femenino , Humanos , Inflamación/complicaciones , Inflamación/metabolismo , Lupus Eritematoso Discoide/complicaciones , Lupus Eritematoso Discoide/metabolismo , Ratones , Ratones Endogámicos MRL lpr , Persona de Mediana Edad , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología
9.
Oncogene ; 38(35): 6241-6255, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31312026

RESUMEN

Early growth response-1 (EGR1) is a transcription factor correlated with prostate cancer (PC) progression in a variety of contexts. For example, EGR1 levels increase in response to suppressed androgen receptor signaling or loss of the tumor suppressor, PTEN. EGR1 has been shown to regulate genes influencing proliferation, apoptosis, immune cell activation, and matrix degradation, among others. Despite this, the impact of EGR1 on PC metastatic colonization is unclear. We demonstrate using a PC model (DU145/RasB1) of bone and brain metastasis that EGR1 expression regulates angiogenic and osteoclastogenic properties of metastases. We have shown previously that FN14 (TNFRSF12A) and downstream NF-κB signaling is required for metastasis in this model. Here we demonstrate that FN14 ligation also leads to NF-κB-independent, MEK-dependent EGR1 expression. EGR1-depletion in DU145/RasB1 cells reduced both the number and size of metastases but did not affect primary tumor growth. Decreased EGR1 expression led to reduced blood vessel density in brain and bone metastases as well as decreased osteolytic bone lesion area and reduced numbers of osteoclasts at the bone-tumor interface. TWEAK (TNFSF12) induced several EGR1-dependent angiogenic and osteoclastogenic factors (e.g., PDGFA, TGFB1, SPP1, IL6, IL8, and TGFA, among others). Consistent with this, in clinical samples of PC, the level of several genes encoding angiogenic/osteoclastogenic pathway effectors correlated with EGR1 levels. Thus, we show here that EGR1 has a direct effect on prostate cancer metastases. EGR1 regulates angiogenic and osteoclastogenic factors, informing the underlying signaling networks that impact autonomous and microenvironmental mechanisms of cancer metastases.


Asunto(s)
Adenocarcinoma/patología , Proteína 1 de la Respuesta de Crecimiento Precoz/fisiología , Neovascularización Patológica/genética , Osteogénesis/genética , Neoplasias de la Próstata/patología , Adenocarcinoma/irrigación sanguínea , Adenocarcinoma/genética , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/secundario , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Metástasis de la Neoplasia , Neovascularización Patológica/patología , Células PC-3 , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/genética , Células RAW 264.7 , Transducción de Señal/genética , Células Tumorales Cultivadas , Microambiente Tumoral/genética
11.
Anal Chim Acta ; 853: 460-468, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25467491

RESUMEN

The capability of electrospray ionization-conventional ion mobility spectrometry (ESI-IMS) for direct analysis of the samples extracted by solid phase microextraction (SPME) was investigated and evaluated for the first time. To that end, an appropriate new desorption chamber was designed and constructed, resulting in the possibility of direct exposure of the SPME fiber to the electrospray solvent flow. Two different elution methods in dynamic and static modes were exhaustively investigated. The results indicated that the interface could help us to have an accurate and sensitive analysis without disturbing the electrospray process, in static elution method. Venlafaxine as a test compound was extracted from human urine and plasma by a convenient headspace SPME method. The positive ion mobility spectrum of the extracted drug was obtained and the analyte responses were calculated. The coupled method of SPME-ESI-IMS was comprehensively validated in terms of sensitivity, dynamic range, and recovery percentage. Finally, various real samples of human urine and plasma were analyzed, all verifying the feasibility and success of the proposed method for the easy routine analysis.


Asunto(s)
Análisis Químico de la Sangre/métodos , Ciclohexanoles/análisis , Ciclohexanoles/aislamiento & purificación , Microextracción en Fase Sólida/métodos , Espectrometría de Masa por Ionización de Electrospray/métodos , Urinálisis/métodos , Adulto , Animales , Ciclohexanoles/sangre , Ciclohexanoles/orina , Humanos , Concentración de Iones de Hidrógeno , Masculino , Ratas , Reproducibilidad de los Resultados , Sales (Química)/química , Temperatura , Factores de Tiempo , Clorhidrato de Venlafaxina
12.
Mol Cell Biol ; 35(11): 1940-51, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25802280

RESUMEN

Bone metastasis is the hallmark of progressive and castration-resistant prostate cancers. MicroRNA 1 (miR-1) levels are decreased in clinical samples of primary prostate cancer and further reduced in metastases. SRC has been implicated as a critical factor in bone metastasis, and here we show that SRC is a direct target of miR-1. In prostate cancer patient samples, miR-1 levels are inversely correlated with SRC expression and a SRC-dependent gene signature. Ectopic miR-1 expression inhibited extracellular signal-regulated kinase (ERK) signaling and bone metastasis in a xenograft model. In contrast, SRC overexpression was sufficient to reconstitute bone metastasis and ERK signaling in cells expressing high levels of miR-1. Androgen receptor (AR) activity, defined by an AR output signature, is low in a portion of castration-resistant prostate cancer. We show that AR binds to the miR-1-2 regulatory region and regulates miR-1 transcription. Patients with low miR-1 levels displayed correlated low canonical AR gene signatures. Our data support the existence of an AR-miR-1-SRC regulatory network. We propose that loss of miR-1 is one mechanistic link between low canonical AR output and SRC-promoted metastatic phenotypes.


Asunto(s)
Andrógenos/genética , Neoplasias Óseas/genética , Neoplasias Óseas/secundario , MicroARNs/genética , Neoplasias de la Próstata Resistentes a la Castración/genética , Familia-src Quinasas/genética , Animales , Neoplasias Óseas/patología , Línea Celular Tumoral , Quinasas MAP Reguladas por Señal Extracelular/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Masculino , Ratones , Ratones Desnudos , Neoplasias de la Próstata Resistentes a la Castración/patología , Receptores Androgénicos/genética , Transducción de Señal/genética
13.
Cell Rep ; 13(10): 2147-58, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26628377

RESUMEN

Primary prostate cancer almost always has a luminal phenotype. However, little is known about the stem/progenitor properties of transformed cells within tumors. Using the aggressive Pten/Tp53-null mouse model of prostate cancer, we show that two classes of luminal progenitors exist within a tumor. Not only did tumors contain previously described multipotent progenitors, but also a major population of committed luminal progenitors. Luminal cells, sorted directly from tumors or grown as organoids, initiated tumors of adenocarcinoma or multilineage histological phenotypes, which is consistent with luminal and multipotent differentiation potentials, respectively. Moreover, using organoids we show that the ability of luminal-committed progenitors to self-renew is a tumor-specific property, absent in benign luminal cells. Finally, a significant fraction of luminal progenitors survived in vivo castration. In all, these data reveal two luminal tumor populations with different stem/progenitor cell capacities, providing insight into prostate cancer cells that initiate tumors and can influence treatment response.


Asunto(s)
Adenocarcinoma/patología , Células Madre Neoplásicas/patología , Neoplasias de la Próstata/patología , Animales , Linaje de la Célula , Separación Celular , Modelos Animales de Enfermedad , Células Epiteliales/patología , Citometría de Flujo , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Organoides , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA