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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Semin Cancer Biol ; 68: 31-46, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-31711994

RESUMEN

Drug repurposing is a valuable approach in delivering new cancer therapeutics rapidly into the clinic. Existing safety and patient tolerability data for drugs already in clinical use represent an untapped resource in terms of identifying therapeutic agents for off-label protein targets. The multicellular effects of STAT3 mediated by a range of various upstream signaling pathways make it an attractive therapeutic target with utility in a range of diseases including cancer, and has led to the development of a variety of STAT3 inhibitors. Moreover, heightened STAT3 transcriptional activation in tumor cells and within the cells of the tumor microenvironment contribute to disease progression. Consequently, there are many STAT3 inhibitors in preclinical development or under evaluation in clinical trials for their therapeutic efficacy predominantly in inflammatory diseases and cancer. Despite these advances, many challenges remain in ultimately providing STAT3 inhibitors to patients as cancer treatments, highlighting the need not only for a better understanding of the mechanisms associated with STAT3 activation, but also how various pharmaceutical agents suppress STAT3 activity in various cancers. In this review we discuss the importance of STAT3-dependent functions in cancer, review the status of compounds designed as direct-acting STAT3 inhibitors, and describe some of the strategies for repurposing of drugs as STAT3 inhibitors for cancer therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Descubrimiento de Drogas , Reposicionamiento de Medicamentos/métodos , Neoplasias/tratamiento farmacológico , Preparaciones Farmacéuticas/administración & dosificación , Factor de Transcripción STAT3/antagonistas & inhibidores , Animales , Humanos
2.
Int J Mol Sci ; 22(4)2021 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-33669775

RESUMEN

Liver metastasis is the primary contributor to the death of patients with colorectal cancer. Despite the overall success of current treatments including targeted therapy, chemotherapy, and immunotherapy combinations in colorectal cancer patients, the prognosis of patients with liver metastasis remains poor. Recent studies have highlighted the importance of the tumour microenvironment and the crosstalk within that determines the fate of circulating tumour cells in distant organs. Understanding the interactions between liver resident cells and tumour cells colonising the liver opens new therapeutic windows for the successful treatment of metastatic colorectal cancer. Here we discuss critical cellular interactions within the tumour microenvironment in primary tumours and in liver metastases that highlight potential therapeutic targets. We also discuss recent therapeutic advances for the treatment of metastatic colorectal cancer.


Asunto(s)
Neoplasias Colorrectales/patología , Terapia Molecular Dirigida , Microambiente Tumoral , Animales , Humanos , Hígado/patología , Metástasis de la Neoplasia , Pronóstico
3.
Cell Death Dis ; 15(4): 255, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38600086

RESUMEN

Excessive STAT3 signalling via gp130, the shared receptor subunit for IL-6 and IL-11, contributes to disease progression and poor survival outcomes in patients with colorectal cancer. Here, we provide evidence that bazedoxifene inhibits tumour growth via direct interaction with the gp130 receptor to suppress IL-6 and IL-11-mediated STAT3 signalling. Additionally, bazedoxifene combined with chemotherapy synergistically reduced cell proliferation and induced apoptosis in patient-derived colon cancer organoids. We elucidated that the primary mechanism of anti-tumour activity conferred by bazedoxifene treatment occurs via pro-apoptotic responses in tumour cells. Co-treatment with bazedoxifene and the SMAC-mimetics, LCL161 or Birinapant, that target the IAP family of proteins, demonstrated increased apoptosis and reduced proliferation in colorectal cancer cells. Our findings provide evidence that bazedoxifene treatment could be combined with SMAC-mimetics and chemotherapy to enhance tumour cell apoptosis in colorectal cancer, where gp130 receptor signalling promotes tumour growth and progression.


Asunto(s)
Neoplasias del Colon , Indoles , Interleucina-11 , Humanos , Interleucina-11/uso terapéutico , Línea Celular Tumoral , Interleucina-6/metabolismo , Receptor gp130 de Citocinas/metabolismo , Neoplasias del Colon/tratamiento farmacológico , Apoptosis
4.
Life Sci Alliance ; 7(2)2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37957015

RESUMEN

Deregulation of the Hippo pathway is a driver for cancer progression and treatment resistance. In the context of gastric cancer, YAP1 is a biomarker for poor patient prognosis. Although genomic tumor profiling provides information of Hippo pathway activation, the present study demonstrates that inhibition of Yap1 activity has anti-tumor effects in gastric tumors driven by oncogenic mutations and inflammatory cytokines. We show that Yap1 is a key regulator of cell metabolism, proliferation, and immune responses in normal and neoplastic gastric epithelium. We propose that the Hippo pathway is targetable across gastric cancer subtypes and its therapeutic benefits are likely to be mediated by both cancer cell-intrinsic and -extrinsic mechanisms.


Asunto(s)
Neoplasias Gástricas , Humanos , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Microambiente Tumoral , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Vía de Señalización Hippo , Factor de Transcripción STAT3/metabolismo
5.
Dev Cell ; 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38781975

RESUMEN

The transcription factor EHF is highly expressed in the lactating mammary gland, but its role in mammary development and tumorigenesis is not fully understood. Utilizing a mouse model of Ehf deletion, herein, we demonstrate that loss of Ehf impairs mammary lobuloalveolar differentiation at late pregnancy, indicated by significantly reduced levels of milk genes and milk lipids, fewer differentiated alveolar cells, and an accumulation of alveolar progenitor cells. Further, deletion of Ehf increased proliferative capacity and attenuated prolactin-induced alveolar differentiation in mammary organoids. Ehf deletion also increased tumor incidence in the MMTV-PyMT mammary tumor model and increased the proliferative capacity of mammary tumor organoids, while low EHF expression was associated with higher tumor grade and poorer outcome in luminal A and basal human breast cancers. Collectively, these findings establish EHF as a non-redundant regulator of mammary alveolar differentiation and a putative suppressor of mammary tumorigenesis.

6.
Biochem Biophys Res Commun ; 438(3): 533-9, 2013 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-23911789

RESUMEN

The expression of orphan nuclear receptor Liver Receptor Homolog-1 (LRH-1) is elevated in breast cancer and promotes proliferation, migration and invasion in vitro. LRH-1 expression is regulated by oestrogen (E2), with LRH-1 mRNA transcript levels higher in oestrogen receptor α (ERα) positive (ER+) breast cancer cells compared to ER- cells. However, the presence of LRH-1 protein in ER- cells suggests discordance between mRNA transcript levels and protein expression. To understand this, we investigated the impact of mRNA and protein stability in determining LRH-1 protein levels in breast cancer cells. LRH-1 transcript levels were significantly higher in ER+ versus ER- breast cancer cells lines; however LRH-1 protein was expressed at similar levels. We found LRH-1 mRNA and protein was more stable in ER- compared to ER+ cell lines. The tumor-specific LRH-1 variant isoform, LRH-1v4, which is highly responsive to E2, showed increased mRNA stability in ER- versus ER+ cells. In addition, in MCF-7 and T47-D cell lines, LRH-1 total mRNA stability was reduced with E2 treatment, this effect mediated by ERα. Our data demonstrates that in ER- cells, increased mRNA and protein stability contribute to the abundant protein expression levels. Expression and immunolocalisation of LRH-1 in ER- cells as well as ER- tumors suggests a possible role in the development of ER- tumors. The modulation of LRH-1 bioactivity may therefore be beneficial as a treatment option in both ER- and ER+ breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Estradiol/farmacología , Receptor alfa de Estrógeno/genética , Estabilidad Proteica/efectos de los fármacos , Estabilidad del ARN/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Isoformas de Proteínas/genética , Estabilidad del ARN/genética , ARN Mensajero/metabolismo
7.
Breast Cancer Res Treat ; 142(1): 211-23, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24122391

RESUMEN

The interaction between breast tumor epithelial and stromal cells is vital for initial and recurrent tumor growth. While breast cancer-associated stromal cells provide a favorable environment for proliferation and metastasis, the molecular mechanisms contributing to this process are not fully understood. Nuclear receptors (NRs) are intracellular transcription factors that directly regulate gene expression. Little is known about the status of NRs in cancer-associated stroma. Nuclear Receptor Low-Density Taqman Arrays were used to compare the gene expression profiles of all 48 NR family members in a collection of primary cultured cancer-associated fibroblasts (CAFs) obtained from estrogen receptor (ER)α positive breast cancers (n = 9) and normal breast adipose fibroblasts (NAFs) (n = 7). Thirty-three of 48 NRs were expressed in both the groups, while 11 NRs were not detected in either. Three NRs (dosage-sensitive sex reversal, adrenal hypoplasia critical region, on chromosome X, gene 1 (DAX-1); estrogen-related receptor beta (ERR-ß); and RAR-related orphan receptor beta (ROR-ß)) were only detected in NAFs, while one NR (liver receptor homolog-1 (LRH-1)) was unique to CAFs. Of the NRs co-expressed, four were significantly down-regulated in CAFs compared with NAFs (RAR-related orphan receptor-α (ROR-α); Thyroid hormone receptor-ß (TR-ß); vitamin D receptor (VDR); and peroxisome proliferator-activated receptor-γ (PPAR-γ)). Quantitative immunohistochemistry for LRH-1, TR-ß, and PPAR-γ proteins in stromal fibroblasts from an independent panel of breast cancers (ER-positive (n = 15), ER-negative (n = 15), normal (n = 14)) positively correlated with mRNA expression profiles. The differentially expressed NRs identified in tumor stroma are key mediators in aromatase regulation and subsequent estrogen production. Our findings reveal a distinct pattern of NR expression that therefore fits with a sustained and increased local estrogen microenvironment in ER-positive tumors. NRs in CAFs may provide a new avenue for the development of intratumoral-targeted therapies in breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Receptores Citoplasmáticos y Nucleares/metabolismo , Células del Estroma/metabolismo , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/genética , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Clasificación del Tumor , Receptor ErbB-2/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Factores de Riesgo
8.
Cancers (Basel) ; 14(2)2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-35053592

RESUMEN

Interleukin (IL)-6 family cytokines, such as IL-6 and IL-11, are defined by the shared use of the gp130 receptor for the downstream activation of STAT3 signaling and the activation of genes which contribute to the "hallmarks of cancer", including proliferation, survival, invasion and metastasis. Increased expression of these cytokines, or the ligand-specific receptors IL-6R and IL-11RA, in breast tumors positively correlate to disease progression and poorer patient outcome. In this review, we examine evidence from pre-clinical studies that correlate enhanced IL-6 and IL-11 mediated gp130/STAT3 signaling to the progression of breast cancer. Key processes by which the IL-6 family cytokines contribute to the heterogeneous nature of breast cancer, immune evasion and metastatic potential, are discussed. We examine the latest research into the therapeutic targeting of IL-6 family cytokines that inhibit STAT3 transcriptional activity as a potential breast cancer treatment, including current clinical trials. The importance of the IL-6 family of cytokines in cellular processes that promote the development and progression of breast cancer warrants further understanding of the molecular basis for its actions to help guide the development of future therapeutic targets.

9.
Sci Adv ; 8(25): eabl7882, 2022 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-35731867

RESUMEN

Although immunotherapy has revolutionized cancer treatment, many immunogenic tumors remain refractory to treatment. This can be largely attributed to an immunologically "cold" tumor microenvironment characterized by an accumulation of immunosuppressive myeloid cells and exclusion of activated T cells. Here, we demonstrate that genetic ablation or therapeutic inhibition of the myeloid-specific hematopoietic cell kinase (HCK) enables activity of antagonistic anti-programmed cell death protein 1 (anti-PD1), anti-CTLA4, or agonistic anti-CD40 immunotherapies in otherwise refractory tumors and augments response in treatment-susceptible tumors. Mechanistically, HCK ablation reprograms tumor-associated macrophages and dendritic cells toward an inflammatory endotype and enhances CD8+ T cell recruitment and activation when combined with immunotherapy in mice. Meanwhile, therapeutic inhibition of HCK in humanized mice engrafted with patient-derived xenografts counteracts tumor immunosuppression, improves T cell recruitment, and impairs tumor growth. Collectively, our results suggest that therapeutic targeting of HCK activity enhances response to immunotherapy by simultaneously stimulating immune cell activation and inhibiting the immunosuppressive tumor microenvironment.

10.
Hum Reprod ; 26(11): 3102-8, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21914669

RESUMEN

BACKGROUND: Little is known about metabolic processes in the developing ovarian follicle. Using mouse ovarian follicles, we investigated uptake of L-leucine by follicles at varying stages of maturity in the presence of insulin-like growth factor (IGF)-1. METHODS Mouse ovarian follicles were cultured in vitro for 5 days in increasing concentrations of IGF-1, and follicle diameter and atresia measured as endpoints for growth. Uptake of (3)H-leucine was measured in follicles at different stages of development. In optimal IGF-1-mediated growth conditions, competitive inhibition of (3)H-leucine uptake by 2-aminobicyclo-(2,2,1)-heptane-2-carboxylic acid (BCH), a non-metabolizable substrate analogue of L-leucine, was performed to demonstrate specificity of influx, via system L transporters. To test whether uptake rates were dependent on intracellular amino acid availability, follicles from in vitro cultures were pre-treated with L-phenylalanine prior to (3)H-leucine uptake. RESULTS: Follicle development (P< 0.001) and survival (P< 0.001) increased with IGF-1 treatment. As pre-antral follicles progressed to late antral stage, we observed an increase in L-leucine uptake, which was reduced in pre-ovulatory follicles. BCH decreased L-leucine uptake rates in early antral (P< 0.05), antral (P< 0.001) and pre-ovulatory follicles (P< 0.01). L-leucine influx increased in follicles preloaded with phenylalanine (trans-stimulation). In follicles lacking free intracellular amino acids (zero-trans suppression), uptake rate was reduced (P< 0.05). CONCLUSIONS: These results demonstrate, for the first time, evidence of specific system L amino acid transport in intact, mouse ovarian follicles and profile L-leucine uptake during folliculogenesis. A better understanding of ovarian follicle metabolic pathways is necessary for improved in vitro maturation as well as determining the impact of altered metabolism on fertility.


Asunto(s)
Sistema de Transporte de Aminoácidos L/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Folículo Ovárico/embriología , Aminoácidos Cíclicos/farmacología , Animales , Transporte Biológico , Cuerpo Lúteo/metabolismo , Femenino , Fertilidad , Regulación del Desarrollo de la Expresión Génica , Células de la Granulosa/metabolismo , Leucina/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Folículo Ovárico/metabolismo
11.
Biomedicines ; 9(2)2021 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-33673239

RESUMEN

The intestinal epithelium provides a barrier against commensal and pathogenic microorganisms. Barrier dysfunction promotes chronic inflammation, which can drive the pathogenesis of inflammatory bowel disease (IBD) and colorectal cancer (CRC). Although the Signal Transducer and Activator of Transcription-3 (STAT3) is overexpressed in both intestinal epithelial cells and immune cells in IBD patients, the role of the interleukin (IL)-6 family of cytokines through the shared IL-6ST/gp130 receptor and its associated STAT3 signalling in intestinal barrier integrity is unclear. We therefore investigated the role of STAT3 in retaining epithelial barrier integrity using dextran sulfate sodium (DSS)-induced colitis in two genetically modified mouse models, to either reduce STAT1/3 activation in response to IL-6 family cytokines with a truncated gp130∆STAT allele (GP130∆STAT/+), or by inducing short hairpin-mediated knockdown of Stat3 (shStat3). Here, we show that mice with reduced STAT3 activity are highly susceptible to DSS-induced colitis. Mechanistically, the IL-6/gp130/STAT3 signalling cascade orchestrates intestinal barrier function by modulating cytokine secretion and promoting epithelial integrity to maintain a defence against bacteria. Our study also identifies a crucial role of STAT3 in controlling intestinal permeability through tight junction proteins. Thus, therapeutically targeting the IL-6/gp130/STAT3 signalling axis to promote barrier function may serve as a treatment strategy for IBD patients.

12.
Cancer Immunol Res ; 9(7): 735-747, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33906864

RESUMEN

IL11 is a member of the IL6 family of cytokines and signals through its cognate receptor subunits, IL11RA and glycoprotein 130 (GP130), to elicit biological responses via the JAK/STAT signaling pathway. IL11 contributes to cancer progression by promoting the survival and proliferation of cancer cells, but the potential immunomodulatory properties of IL11 signaling during tumor development have thus far remained unexplored. Here, we have characterized a role for IL11 in regulating CD4+ T cell-mediated antitumor responses. Absence of IL11 signaling impaired tumor growth in a sporadic mouse model of colon cancer and syngeneic allograft models of colon cancer. Adoptive bone marrow transfer experiments and in vivo depletion studies demonstrated that the tumor-promoting activity of IL11 was mediated through its suppressive effect on host CD4+ T cells in the tumor microenvironment. Indeed, when compared with Il11ra-proficient CD4+ T cells associated with MC38 tumors, their Il11ra-deficient counterparts displayed elevated expression of mRNA encoding the antitumor mediators IFNγ and TNFα. Likewise, IL11 potently suppressed the production of proinflammatory cytokines (IFNγ, TNFα, IL6, and IL12p70) by CD4+ T cells in vitro, which we corroborated by RNAscope analysis of human colorectal cancers, where IL11RAhigh tumors showed less IFNG and CD4 expression than IL11RAlow tumors. Therefore, our results ascribe a tumor cell-extrinsic immunomodulatory role to IL11 during colon cancer development that could be amenable to an anticytokine-based therapy.See related Spotlight by van der Burg, p. 724.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Neoplasias del Colon/inmunología , Subunidad alfa del Receptor de Interleucina-11/metabolismo , Interleucina-11/metabolismo , Animales , Antígenos CD4/análisis , Antígenos CD4/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Línea Celular Tumoral , Colon/inmunología , Colon/patología , Neoplasias del Colon/patología , Conjuntos de Datos como Asunto , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Interferón gamma/análisis , Interferón gamma/metabolismo , Subunidad alfa del Receptor de Interleucina-11/análisis , Subunidad alfa del Receptor de Interleucina-11/genética , Ratones , Ratones Noqueados , Neoplasias de Tejido Óseo , Receptores de Interleucina-11/metabolismo , Microambiente Tumoral/inmunología
13.
Pharmacol Ther ; 211: 107527, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32173557

RESUMEN

G-protein-coupled receptors (GPCRs) are the largest and most diverse group of cellular membrane receptors identified and characterized. It is estimated that 30 to 50% of marketed drugs target these receptors. The angiotensin II receptor type 1 (AT1R) is a GPCR which signals in response to systemic alterations of the peptide hormone angiotensin II (AngII) in circulation. The enzyme responsible for converting AngI to AngII is the angiotensin-converting enzyme (ACE). Specific inhibitors for the AT1R (more commonly known as AT1R blockers or antagonists) and ACE are well characterized for their effects on the cardiovascular system. Combined with the extensive clinical data available on patient tolerance of AT1R blockers (ARBs) and ACE inhibitors (ACEIs), as well as their non-classical roles in cancer, the notion of repurposing this class of medications as cancer treatment(s) is explored in the current review. Given that AngII-dependent AT1R activity directly regulates angiogenesis, remodeling of vasculature, pro-inflammatory responses, stem cell programming and hematopoiesis, and electrolyte balance; the modulation of these processes with pharmacologically well characterized medications could present a valuable complementary treatment option for cancer patients.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/tratamiento farmacológico , Sistema Renina-Angiotensina/efectos de los fármacos , Antagonistas de Receptores de Angiotensina/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina , Animales , Reposicionamiento de Medicamentos , Humanos , Neoplasias/patología
14.
Cancer Immunol Res ; 8(4): 428-435, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31992566

RESUMEN

Persistent activation of the latent transcription factor STAT3 is observed in gastric tumor epithelial and immune cells and is associated with a poor patient prognosis. Although targeting STAT3-activating upstream kinases offers therapeutically viable targets with limited specificity, direct inhibition of STAT3 remains challenging. Here we provide functional evidence that myeloid-specific hematopoietic cell kinase (HCK) activity can drive STAT3-dependent epithelial tumor growth in mice and is associated with alternative macrophage activation alongside matrix remodeling and tumor cell invasion. Accordingly, genetic reduction of HCK expression in bone marrow-derived cells or systemic pharmacologic inhibition of HCK activity suppresses alternative macrophage polarization and epithelial STAT3 activation, and impairs tumor growth. These data validate HCK as a molecular target for the treatment of human solid tumors harboring excessive STAT3 activity.


Asunto(s)
Proteínas Proto-Oncogénicas c-hck/antagonistas & inhibidores , Pirimidinas/farmacología , Pirroles/farmacología , Factor de Transcripción STAT3/metabolismo , Neoplasias Gástricas/tratamiento farmacológico , Animales , Femenino , Humanos , Activación de Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-hck/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Tasa de Supervivencia
16.
BMC Cancer ; 9: 148, 2009 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-19445691

RESUMEN

BACKGROUND: Until recently, the molecular mechanisms explaining increased incidence of ovarian and breast cancers in carriers of BRCA1 gene mutations had not been clearly understood. Of significance is the finding that BRCA1 negatively regulates aromatase expression in vitro. Our objective was to characterise aromatase gene (CYP19A1) and its promoter expression in breast adipose and ovarian tissue in BRCA1 mutation carriers and unaffected controls. METHODS: We measured aromatase transcripts, total and promoter-specific (PII, PI.3, PI.4) in prophylactic oophorectomy or mastectomy, therapeutic mastectomy, ovarian and breast tissue from unaffected women. RESULTS: We demonstrate that the lack of functional BRCA1 protein correlates to higher aromatase levels in 85% of BRCA1 mutation carriers. This increase is mediated by aberrant transcriptional regulation of aromatase; in breast adipose by increases in promoter II/I.3 and I.4-specific transcripts; and in the ovary with elevation in promoter I.3 and II-specific transcripts. CONCLUSION: Understanding the link between BRCA1 and aromatase is significant in terms of understanding why carcinogenesis is restricted to estrogen-producing tissues in BRCA1 mutation carriers.


Asunto(s)
Aromatasa/genética , Proteína BRCA1/genética , Expresión Génica , Heterocigoto , Mutación , Adipocitos/metabolismo , Adulto , Aromatasa/metabolismo , Australia , Proteína BRCA1/metabolismo , Mama/metabolismo , Femenino , Humanos , Ovario/metabolismo
17.
EMBO Mol Med ; 11(4)2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30885958

RESUMEN

Excessive signaling through gp130, the shared receptor for the interleukin (IL)6 family of cytokines, is a common hallmark in solid malignancies and promotes their progression. Here, we established the in vivo utility of bazedoxifene, a steroid analog clinically approved for the treatment of osteoporosis, to suppress gp130-dependent tumor growth of the gastrointestinal epithelium. Bazedoxifene administration reduced gastric tumor burden in gp130Y757F mice, where tumors arise exclusively through excessive gp130/STAT3 signaling in response to the IL6 family cytokine IL11. Likewise, in mouse models of sporadic colon and intestinal cancers, which arise from oncogenic mutations in the tumor suppressor gene Apc and the associated ß-catenin/canonical WNT pathway, bazedoxifene treatment reduces tumor burden. Consistent with the proposed orthogonal tumor-promoting activity of IL11-dependent gp130/STAT3 signaling, tumors of bazedoxifene-treated Apc-mutant mice retain excessive nuclear accumulation of ß-catenin and aberrant WNT pathway activation. Likewise, bazedoxifene treatment of human colon cancer cells harboring mutant APC did not reduce aberrant canonical WNT signaling, but suppressed IL11-dependent STAT3 signaling. Our findings provide compelling proof of concept to support the repurposing of bazedoxifene for the treatment of gastrointestinal cancers in which IL11 plays a tumor-promoting role.


Asunto(s)
Reposicionamiento de Medicamentos , Neoplasias Gastrointestinales/tratamiento farmacológico , Indoles/uso terapéutico , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Proliferación Celular/efectos de los fármacos , Receptor gp130 de Citocinas/química , Receptor gp130 de Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Neoplasias Gastrointestinales/patología , Humanos , Indoles/metabolismo , Indoles/farmacología , Interleucina-11/química , Interleucina-11/metabolismo , Interleucina-11/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor de Transcripción STAT3/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/metabolismo
18.
Breast Cancer Res Treat ; 112(3): 429-35, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18181018

RESUMEN

Aromatase is a member of the cytochrome P450 superfamily of enzymes which catalyses the rate-limiting step in the biosynthesis of estrogens. A number of clinical studies have highlighted the importance of local estrogen production in adipose tissue. In particular, in the postmenopausal woman, the degree of her estrogenization is mainly determined by the extent of her adiposity and it is this extragonadal source of estrogen that likely contributes to breast cancer development and progression. The mechanisms regulating aromatase expression in adipose tissue however, have not been fully elucidated. In this study, we have characterised the expression of aromatase and its activity in a human preadipocyte cell strain, SGBS. Aromatase is expressed in SGBS cells and its expression and activity are strongly stimulated by forskolin (FSK) and phorbol 12-myristate-13-acetate (PMA) treatment. Consistent with this, FSK and PMA treatment also increased activation of the proximal aromatase promoter, promoter II. These findings mimic those that have previously been shown in isolated primary human preadipocytes. These data suggest that SGBS cells are a valuable model with which to further elucidate the mechanisms regulating aromatase expression, and therefore local estrogen synthesis in human adipose tissue.


Asunto(s)
Adipocitos/enzimología , Aromatasa/biosíntesis , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Aromatasa/metabolismo , Línea Celular , Colforsina/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , ADN Complementario/metabolismo , Progresión de la Enfermedad , Estrógenos/metabolismo , Humanos , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Acetato de Tetradecanoilforbol/farmacología , Factores de Tiempo
19.
J Exp Med ; 215(4): 1205-1225, 2018 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-29472497

RESUMEN

Colorectal cancer is treated with antibodies blocking epidermal growth factor receptor (EGF-R), but therapeutic success is limited. EGF-R is stimulated by soluble ligands, which are derived from transmembrane precursors by ADAM17-mediated proteolytic cleavage. In mouse intestinal cancer models in the absence of ADAM17, tumorigenesis was almost completely inhibited, and the few remaining tumors were of low-grade dysplasia. RNA sequencing analysis demonstrated down-regulation of STAT3 and Wnt pathway components. Because EGF-R on myeloid cells, but not on intestinal epithelial cells, is required for intestinal cancer and because IL-6 is induced via EGF-R stimulation, we analyzed the role of IL-6 signaling. Tumor formation was equally impaired in IL-6-/- mice and sgp130Fc transgenic mice, in which only trans-signaling via soluble IL-6R is abrogated. ADAM17 is needed for EGF-R-mediated induction of IL-6 synthesis, which via IL-6 trans-signaling induces ß-catenin-dependent tumorigenesis. Our data reveal the possibility of a novel strategy for treatment of colorectal cancer that could circumvent intrinsic and acquired resistance to EGF-R blockade.


Asunto(s)
Proteína ADAM17/metabolismo , Receptores ErbB/metabolismo , Interleucina-6/metabolismo , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/patología , Transducción de Señal , Proteína ADAM17/deficiencia , Poliposis Adenomatosa del Colon/metabolismo , Animales , Carcinogénesis/metabolismo , Carcinogénesis/patología , Línea Celular Tumoral , Núcleo Celular/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Intestinales/genética , Intestino Delgado/patología , Antígeno Ki-67/metabolismo , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Estadificación de Neoplasias , Organoides/patología , Pronóstico , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Transcripción STAT3/metabolismo , Carga Tumoral , beta Catenina/metabolismo
20.
Reprod Biol Endocrinol ; 5: 18, 2007 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-17506907

RESUMEN

BACKGROUND: Understanding the pathophysiology of chemokine secretion in endometriosis may offer a novel area of therapeutic intervention. This study aimed to identify chemokines differentially expressed in epithelial glands in eutopic endometrium from normal women and those with endometriosis, and to establish the expression profiles of key chemokines in endometriotic lesions. METHODS: Laser capture microdissection isolated epithelial glands from endometrial eutopic tissue from women with and without endometriosis in the mid-secretory phase of their menstrual cycles. Gene profiling of the excised glands used a human chemokine and receptor cDNA array. Selected chemokines were further examined using real-time PCR and immunohistochemistry. RESULTS: 22 chemokine/receptor genes were upregulated and two downregulated in pooled endometrial epithelium of women with endometriosis compared with controls. CCL16 and CCL21 mRNA was confirmed as elevated in some women with endometriosis compared to controls on individual samples. Immunoreactive CCL16 and CCL21 were predominantly confined to glands in eutopic and ectopic endometrium: leukocytes also stained. Immunoreactive CCL16 was overall higher in glands in ectopic vs. eutopic endometrium from the same woman (P < 0.05). Staining for CCL16 and CCL21 was highly correlated in individual tissues. CONCLUSION: This study provides novel candidate molecules and suggests a potential local role for CCL16 and CCL21 as mediators contributing to the inflammatory events associated with endometriosis.


Asunto(s)
Quimiocinas CC/genética , Quimiocinas CC/metabolismo , Endometriosis/genética , Mediadores de Inflamación/metabolismo , Rayos Láser , Microdisección , Análisis de Secuencia por Matrices de Oligonucleótidos , Adulto , Quimiocina CCL21 , Quimiocinas CC/biosíntesis , Endometriosis/diagnóstico , Endometriosis/inmunología , Endometrio/inmunología , Endometrio/metabolismo , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Mediadores de Inflamación/aislamiento & purificación , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA