Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Development ; 150(13)2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37376888

RESUMEN

The reactivation of developmental genes and pathways during adulthood may contribute to pathogenesis of diseases such as prostate cancer. Analysis of the mechanistic links between development and disease could be exploited to identify signalling pathways leading to disease in the prostate. However, the mechanisms underpinning prostate development require further characterisation to interrogate fully the link between development and disease. Previously, our group developed methods to produce prostate organoids using induced pluripotent stem cells (iPSCs). Here, we show that human iPSCs can be differentiated into prostate organoids using neonatal rat seminal vesicle mesenchyme in vitro. The organoids can be used to study prostate development or modified to study prostate cancer. We also elucidated molecular drivers of prostate induction through RNA-sequencing analyses of the rat urogenital sinus and neonatal seminal vesicles. We identified candidate drivers of prostate development evident in the inductive mesenchyme and epithelium involved with prostate specification. Our top candidates included Spx, Trib3, Snai1, Snai2, Nrg2 and Lrp4. This work lays the foundations for further interrogation of the reactivation of developmental genes in adulthood, leading to prostate disease.


Asunto(s)
Células Madre Pluripotentes Inducidas , Neoplasias de la Próstata , Masculino , Humanos , Ratas , Animales , Próstata , Roedores , Sistema Urogenital/fisiología , Diferenciación Celular/genética , Organoides
2.
Ultraschall Med ; 44(1): 36-49, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36228630

RESUMEN

OBJECTIVE: To evaluate the evidence and produce a summary and recommendations for the most common heart and lung point-of-care ultrasound (PoCUS). METHODS: We reviewed 10 clinical domains/questions related to common heart and lung applications of PoCUS. Following review of the evidence, a summary and recommendations were produced, including assigning levels of evidence (LoE) and grading of recommendation, assessment, development, and evaluation (GRADE). 38 international experts, the expert review group (ERG), were invited to review the evidence presented for each question. A level of agreement of over 75 % was required to progress to the next section. The ERG then reviewed and indicated their level of agreement of the summary and recommendation for each question (using a 5-point Likert scale), which was approved in the case of a level of agreement of greater than 75 %. A level of agreement was defined as a summary of "strongly agree" and "agree" on the Likert scale responses. FINDINGS AND RECOMMENDATIONS: One question achieved a strong consensus for an assigned LoE of 3 and a weak GRADE recommendation (question 1), the remaining 9 questions achieved broad agreement with an assigned LoE of 4 and a weak GRADE recommendation (question 2), three achieved an LoE of 3 with a weak GRADE recommendation (questions 3-5), three achieved an LoE of 3 with a strong GRADE recommendation (questions 6-8) and the remaining two were assigned an LoE of 2 with a strong GRADE recommendation (questions 9 and 10). CONCLUSION: These consensus-derived recommendations should aid clinical practice and highlight areas of further research for PoCUS in acute settings.


Asunto(s)
Sistemas de Atención de Punto , Pruebas en el Punto de Atención , Humanos , Pulmón , Ultrasonografía
3.
Ultraschall Med ; 44(1): e1-e24, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36228631

RESUMEN

AIMS: To evaluate the evidence and produce a summary and recommendations for the most common heart and lung applications of point-of-care ultrasound (PoCUS). METHODS: We reviewed 10 clinical domains/questions related to common heart and lung applications of PoCUS. Following review of the evidence, a summary and recommendation were produced, including assignment of levels of evidence (LoE) and grading of the recommendation, assessment, development, and evaluation (GRADE). 38 international experts, the expert review group (ERG), were invited to review the evidence presented for each question. A level of agreement of over 75 % was required to progress to the next section. The ERG then reviewed and indicated their level of agreement regarding the summary and recommendation for each question (using a 5-point Likert scale), which was approved if a level of agreement of greater than 75 % was reached. A level of agreement was defined as a summary of "strongly agree" and "agree" on the Likert scale responses. FINDINGS AND RECOMMENDATIONS: One question achieved a strong consensus for an assigned LoE of 3 and a weak GRADE recommendation (question 1). The remaining 9 questions achieved broad agreement with one assigned an LoE of 4 and weak GRADE recommendation (question 2), three achieving an LoE of 3 with a weak GRADE recommendation (questions 3-5), three achieved an LoE of 3 with a strong GRADE recommendation (questions 6-8), and the remaining two were assigned an LoE of 2 with a strong GRADE recommendation (questions 9 and 10). CONCLUSION: These consensus-derived recommendations should aid clinical practice and highlight areas of further research for PoCUS in acute settings.


Asunto(s)
Sistemas de Atención de Punto , Pruebas en el Punto de Atención , Humanos , Pulmón , Ultrasonografía
4.
Lab Invest ; 101(7): 921-934, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33824421

RESUMEN

Lipid droplet (LD) accumulation in cancer results from aberrant metabolic reprograming due to increased lipid uptake, diminished lipolysis and/or de novo lipid synthesis. Initially implicated in storage and lipid trafficking in adipocytes, LDs are more recently recognized to fuel key functions associated with carcinogenesis and progression of several cancers, including prostate cancer (PCa). However, the mechanisms controlling LD accumulation in cancer are largely unknown. EPHB2, a tyrosine kinase (TKR) ephrin receptor has been proposed to have tumor suppressor functions in PCa, although the mechanisms responsible for these effects are unclear. Given that dysregulation in TRK signaling can result in glutaminolysis we postulated that EPHB2 might have potential effects on lipid metabolism. Knockdown strategies for EPHB2 were performed in prostate cancer cells to analyze the impact on the net lipid balance, proliferation, triacylglycerol-regulating proteins, effect on LD biogenesis, and intracellular localization of LDs. We found that EPHB2 protein expression in a panel of human-derived prostate cancer cell lines was inversely associated with in vivo cell aggressiveness. EPHB2 silencing increased the proliferation of prostate cancer cells and concurrently induced de novo LD accumulation in both cytoplasmic and nuclear compartments as well as a "shift" on LD size distribution in newly formed lipid-rich organelles. Lipid challenge using oleic acid exacerbated the effects on the LD phenotype. Loss of EPHB2 directly regulated key proteins involved in maintaining lipid homeostasis including, increasing lipogenic DGAT1, DGAT2 and PLIN2 and decreasing lipolytic ATGL and PEDF. A DGAT1-specific inhibitor abrogated LD accumulation and proliferative effects induced by EPHB2 loss. In conclusion, we highlight a new anti-tumor function of EPHB2 in lipid metabolism through regulation of DGAT1 and ATGL in prostate cancer. Blockade of DGAT1 in EPHB2-deficient tumors appears to be effective in restoring the lipid balance and reducing tumor growth.


Asunto(s)
Diacilglicerol O-Acetiltransferasa/metabolismo , Lipasa/metabolismo , Gotas Lipídicas/metabolismo , Neoplasias de la Próstata/metabolismo , Receptor EphB2 , Línea Celular Tumoral , Humanos , Metabolismo de los Lípidos/fisiología , Masculino , Receptor EphB2/genética , Receptor EphB2/metabolismo
5.
Prostate ; 80(2): 173-185, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31763714

RESUMEN

BACKGROUND: Carcinoma-associated fibroblasts (CAF) are a heterogeneous group of cells within the tumor microenvironment (TME) that can promote tumorigenesis in the prostate. By understanding the mechanism(s) by which CAF contributes to tumor growth, new therapeutic targets for the management of this disease may be identified. These studies determined whether unique sub-populations of human prostate CAF can be identified and functionally characterized. METHODS: Single-cell RNA-seq of primary human prostate CAF followed by unsupervised clustering was utilized to generate cell clusters based on differentially expressed (DE) gene profiles. Potential communication between CAF and immune cells was analyzed using in vivo tissue recombination by combining CAF or normal prostate fibroblasts (NPF) with non-tumorigenic, initiated prostate epithelial BPH-1 cells. Resultant grafts were assessed for inflammatory cell recruitment. RESULTS: Clustering of 3321 CAF allows for visualization of six subpopulations, demonstrating heterogeneity within CAF. Sub-renal capsule recombination assays show that the presence of CAF significantly increases myeloid cell recruitment to resultant tumors. This is supported by significantly increased expression of chemotactic chemokines CCL2 and CXCL12 in large clusters compared to other subpopulations. Bayesian analysis topologies also support differential communication signals between chemokine-related genes of individual clusters. Migration of THP-1 monocyte cells in vitro is stimulated in the presence of CAF conditioned medium (CM) compared with NPF CM. Further in vitro analyses suggest that CAF-derived chemokine CCL2 may be responsible for CAF-stimulated migration of THP-1 cells, since neutralization of this chemokine abrogates migration capacity. CONCLUSIONS: CAF clustering based on DE gene expression supports the concept that clusters have unique functions within the TME, including a role in immune/inflammatory cell recruitment. These data suggest that CCL2 produced by CAF may be involved in the recruitment of inflammatory cells, but may also directly regulate the growth of the tumor. Further studies aimed at characterizing the subpopulation(s) of CAF which promote immune cell recruitment to the TME and/or stimulate prostate cancer growth and progression will be pursued.


Asunto(s)
Fibroblastos Asociados al Cáncer/patología , Células Mieloides/patología , Neoplasias de la Próstata/patología , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Quimiocina CCL2/genética , Quimiocina CXCL12/genética , Heterogeneidad Genética , Células HEK293 , Humanos , Masculino , Neoplasias de la Próstata/genética , ARN Mensajero/genética , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Células THP-1 , Microambiente Tumoral
6.
J Cell Sci ; 131(13)2018 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-29792311

RESUMEN

Prostate tumors make metabolic adaptations to ensure adequate energy and amplify cell cycle regulators, such as centrosomes, to sustain their proliferative capacity. It is not known whether cancer-associated fibroblasts (CAFs) undergo metabolic re-programming. We postulated that CAFs augment lipid storage and amplify centrosomal or non-centrosomal microtubule-organizing centers (MTOCs) through a pigment epithelium-derived factor (PEDF)-dependent lipid-MTOC signaling axis. Primary human normal prostate fibroblasts (NFs) and CAFs were evaluated for lipid content, triacylglycerol-regulating proteins, MTOC number and distribution. CAFs were found to store more neutral lipids than NFs. Adipose triglyceride lipase (ATGL) and PEDF were strongly expressed in NFs, whereas CAFs had minimal to undetectable levels of PEDF or ATGL protein. At baseline, CAFs demonstrated MTOC amplification when compared to 1-2 perinuclear MTOCs consistently observed in NFs. Treatment with PEDF or blockade of lipogenesis suppressed lipid content and MTOC number. In summary, our data support that CAFs have acquired a tumor-like phenotype by re-programming lipid metabolism and amplifying MTOCs. Normalization of MTOCs by restoring PEDF or by blocking lipogenesis highlights a previously unrecognized plasticity in centrosomes, which is regulated through a new lipid-MTOC axis.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Proteínas del Ojo/metabolismo , Metabolismo de los Lípidos , Centro Organizador de los Microtúbulos/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Neoplasias de la Próstata/metabolismo , Serpinas/metabolismo , Proteínas del Ojo/genética , Fibroblastos/metabolismo , Humanos , Lipasa/genética , Lipasa/metabolismo , Lipogénesis , Masculino , Factores de Crecimiento Nervioso/genética , Próstata/metabolismo , Neoplasias de la Próstata/genética , Serpinas/genética , Triglicéridos/metabolismo
7.
Lab Invest ; 99(12): 1822-1834, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31409893

RESUMEN

Lipid droplets (LDs) utilize microtubules (MTs) to participate in intracellular trafficking of cargo proteins. Cancer cells accumulate LDs and acidify their tumor microenvironment (TME) by increasing the proton pump V-ATPase. However, it is not known whether these two metabolic changes are mechanistically related or influence LD movement. We postulated that LD density and velocity are progressively increased with tumor aggressiveness and are dependent on V-ATPase and the lipolysis regulator pigment epithelium-derived factor (PEDF). LD density was assessed in human prostate cancer (PCa) specimens across Gleason scores (GS) 6-8. LD distribution and velocity were analyzed in low and highly aggressive tumors using live-cell imaging and in cells exposed to low pH and/or treated with V-ATPase inhibitors. The MT network was disrupted and analyzed by α-tubulin staining. LD density positively correlated with advancing GS in human tumors. Acidification promoted peripheral localization and clustering of LDs. Highly aggressive prostate, breast, and pancreatic cell lines had significantly higher maximum LD velocity (LDVmax) than less aggressive and benign cells. LDVmax was MT-dependent and suppressed by blocking V-ATPase directly or indirectly with PEDF. Upon lowering pH, LDs moved to the cell periphery and carried metalloproteinases. These results suggest that acidification of the TME can alter intracellular LD movement and augment velocity in cancer. Restoration of PEDF or blockade of V-ATPase can normalize LD distribution and decrease velocity. This study identifies V-ATPase and PEDF as new modulators of LD trafficking in the cancer microenvironment.


Asunto(s)
Proteínas del Ojo/metabolismo , Gotas Lipídicas/fisiología , Factores de Crecimiento Nervioso/metabolismo , Neoplasias de la Próstata/metabolismo , Serpinas/metabolismo , Microambiente Tumoral , ATPasas de Translocación de Protón Vacuolares/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Células MCF-7 , Masculino , Clasificación del Tumor , Células PC-3 , Próstata/patología , Neoplasias de la Próstata/patología
8.
Prostate ; 79(3): 259-264, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30370673

RESUMEN

BACKGROUND: The tyrosine kinase inhibitors (TKI), imatinib and nilotinib, are used to treat chronic myelogenous leukemia (CML). In three CML patients being monitored for urologic diseases, we observed that switching of TKI therapy affected prostate-specific antigen (PSA) titers. Urologists and other medical professionals need to be aware of the potential side-effects of drugs that patients may be receiving for other indications to modify this important prostate diseases indicator. TKIs may affect PSA titers independent of prostate growth or volume. MATERIALS AND METHODS: We followed PSA levels in urology patients who were also undergoing TKI treatment for CML. We determined the effects of nilotinib and imatinib on proliferation, AR and PSA expression in the LNCaP and 22Rv1 prostate cancer (PCa) cell lines using real-time PCR and Western blotting. RESULTS: Clinically, nilotinib and dasatinib reversibly reduced PSA titers compared to imatinib. At high doses nilotinib and imatinib both demonstrated antiproliferative effects in the PCa cells. At low doses expression of AR and PSA was decreased by both drugs, at mRNA and protein levels. Nilotinib exerted greater effects at lower doses than imatinib. CONCLUSIONS: Nilotinib down-regulates serum PSA in patients being treated for non-urological indications, potentially masking a clinical useful marker, we cannot exclude a similar but smaller effect of imatinib. Nilotinib and imatinib both decreased AR and PSA expression in PCa cell lines with the nilotinib effect evident at lower doses. Urologists must appreciate the effects of drugs provided for other diseases on PSA titers and be aware that sudden changes may not reflect underlying prostatic disease.


Asunto(s)
Mesilato de Imatinib/administración & dosificación , Calicreínas/sangre , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/administración & dosificación , Pirimidinas/administración & dosificación , Anciano , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Mesilato de Imatinib/efectos adversos , Calicreínas/biosíntesis , Calicreínas/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/sangre , Masculino , Antígeno Prostático Específico/biosíntesis , Antígeno Prostático Específico/genética , Hiperplasia Prostática/sangre , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Inhibidores de Proteínas Quinasas/efectos adversos , Pirimidinas/efectos adversos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Androgénicos/biosíntesis , Receptores Androgénicos/genética
9.
Prostate ; 79(9): 980-993, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30999385

RESUMEN

BACKGROUND: Prostatic inflammation and various proinflammatory systemic comorbidities, such as diabetes and obesity are associated with human benign prostatic hyperplasia (BPH). There is a paucity of in vivo models reflecting specific aspects of BPH pathogenesis. Our aim was to investigate the nonobese diabetic (NOD) mouse as a potential model for subsequent intervention studies. MATERIALS AND METHODS: We used the NOD mouse, a model of autoimmune inflammation leading to type 1 diabetes to examine the effects of systemic inflammation and diabetes on the prostate. We assessed changes in prostatic histology, infiltrating leukocytes, and gene expression associated with aging and diabetic status. RESULTS: Both stromal expansion and epithelial hyperplasia were observed in the prostates. Regardless of diabetic status, the degree of prostatic hyperplasia varied. Local inflammation was associated with a more severe prostatic phenotype in both diabetic and nondiabetic mice. Testicular atrophy was noted in diabetic mice, but prostate glands showed persistent focal cell proliferation. In addition, a prostatic intraepithelial neoplasia (PIN)-like phenotype was seen in several diabetic animals with an associated increase in c-Myc and MMP-2 expression. To examine changes in gene and cytokine expression we performed microarray and cytokine array analysis comparing the prostates of diabetic and nondiabetic animals. Microarray analysis revealed several differentially expressed genes including CCL3, CCL12, and TNFS10. Cytokine array analysis revealed increased expression of cytokines and proteases such as LDLR, IL28 A/B, and MMP-2 in diabetic mice. CONCLUSION: Overall, NOD mice provide a model to examine the effects of hyperglycemia and chronic inflammation on the prostate, demonstrating relevance to some of the mechanisms present underlying BPH and potentially the initiation of prostate cancer.


Asunto(s)
Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/inmunología , Hiperglucemia/inmunología , Hiperplasia Prostática/sangre , Hiperplasia Prostática/inmunología , Linfocitos T/inmunología , Animales , Citocinas/inmunología , Diabetes Mellitus Experimental/patología , Células Epiteliales/inmunología , Células Epiteliales/patología , Hiperglucemia/patología , Masculino , Ratones , Ratones Endogámicos NOD , Hiperplasia Prostática/patología , Neoplasia Intraepitelial Prostática/sangre , Neoplasia Intraepitelial Prostática/inmunología , Neoplasia Intraepitelial Prostática/patología , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología , Células del Estroma/inmunología , Células del Estroma/patología , Linfocitos T/patología , Testículo/patología
10.
Emerg Med J ; 36(3): 185-190, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30470688

RESUMEN

INTRODUCTION: It is well-recognised that the detection of rib fractures is unreliable using chest radiograph. The aim of this systematic review was to investigate whether the use of lung ultrasound is superior in accuracy to chest radiography, in the diagnosis of rib fractures following blunt chest wall trauma. METHODS: The search filter was used for international online electronic databases including MEDLINE, EMBASE, Cochrane and ScienceDirect, with no imposed time or language limitations. Grey literature was searched. Two review authors completed study selection, data extraction and data synthesis/analysis process. Quality assessment using the Quality Assessment of Diagnostic Accuracy Studies Tool (QUADAS-2) was completed. RESULTS: 13 studies were included. Overall, study results demonstrated that the use of lung ultrasound in the diagnosis of rib fractures in blunt chest wall trauma patients appears superior compared with chest radiograph. All studies were small, single centre and considered to be at risk of bias on quality assessment. Meta-analysis was not possible due to high levels of heterogeneity, lack of appropriate reference standard and poor study quality. DISCUSSION: The results demonstrate that lung ultrasound may be superior to chest radiography, but the low quality of the studies means that no definitive statement can be made.


Asunto(s)
Diagnóstico por Imagen/normas , Radiografía/normas , Fracturas de las Costillas/diagnóstico , Ultrasonografía/normas , Diagnóstico por Imagen/métodos , Humanos , Pulmón/anomalías , Pulmón/fisiopatología , Radiografía/métodos , Fracturas de las Costillas/diagnóstico por imagen , Factores de Riesgo , Traumatismos Torácicos/complicaciones , Traumatismos Torácicos/diagnóstico por imagen , Ultrasonografía/métodos
11.
Differentiation ; 96: 40-48, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28779656

RESUMEN

Stromal-epithelial interactions play a crucial and poorly understood role in carcinogenesis and tumor progression. Mesenchymal-epithelial interactions have a long history of research in relation to the development of organs. Models designed to study development are often also applicable to studies of benign and malignant disease. Tumor stroma is a complex mixture of cells that includes a fibroblastic component often referred to as cancer-associated fibroblasts (CAF), desmoplasia or "reactive" stroma. Here we discuss the history of, and approaches to, understanding these interactions with particular reference to prostate cancer and to in vivo modeling using human cells and tissues. A series of studies have revealed a complex mixture of signaling molecules acting both within the stromal tissue and between the stromal and epithelial tissues. We are starting to understand the interactions of some of these pathways, however the work is still ongoing. This area of research provide a basis for new medical approaches aimed at stabilizing early stage cancers rendering them chronic rather than acute problems. Such work is especially relevant to slow growing tumors found in older patients, a class that would include many prostate cancers.


Asunto(s)
Carcinogénesis , Carcinoma/patología , Células Epiteliales/patología , Fibroblastos/patología , Neoplasias de la Próstata/patología , Células Epiteliales/metabolismo , Fibroblastos/metabolismo , Humanos , Masculino
12.
Breast Cancer Res ; 19(1): 56, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28506312

RESUMEN

BACKGROUND: The breast tumor microenvironment regulates progression of ductal carcinoma in situ (DCIS) to invasive ductal carcinoma (IDC). However, it is unclear how interactions between breast epithelial and stromal cells can drive this progression and whether there are reliable microenvironmental biomarkers to predict transition of DCIS to IDC. METHODS: We used xenograft mouse models and a 3D pathomimetic model termed mammary architecture and microenvironment engineering (MAME) to study the interplay between human breast myoepithelial cells (MEPs) and cancer-associated fibroblasts (CAFs) on DCIS progression. RESULTS: Our results show that MEPs suppress tumor formation by DCIS cells in vivo even in the presence of CAFs. In the in vitro MAME model, MEPs reduce the size of 3D DCIS structures and their degradation of extracellular matrix. We further show that the tumor-suppressive effects of MEPs on DCIS are linked to inhibition of urokinase plasminogen activator (uPA)/urokinase plasminogen activator receptor (uPAR)-mediated proteolysis by plasminogen activator inhibitor 1 (PAI-1) and that they can lessen the tumor-promoting effects of CAFs by attenuating interleukin 6 (IL-6) signaling pathways. CONCLUSIONS: Our studies using MAME are, to our knowledge, the first to demonstrate a divergent interplay between MEPs and CAFs within the DCIS tumor microenvironment. We show that the tumor-suppressive actions of MEPs are mediated by PAI-1, uPA and its receptor, uPAR, and are sustained even in the presence of the CAFs, which themselves enhance DCIS tumorigenesis via IL-6 signaling. Identifying tumor microenvironmental regulators of DCIS progression will be critical for defining a robust and predictive molecular signature for clinical use.


Asunto(s)
Neoplasias de la Mama/genética , Carcinoma Ductal de Mama/genética , Interleucina-6/genética , Inhibidor 1 de Activador Plasminogénico/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/genética , Animales , Neoplasias de la Mama/patología , Fibroblastos Asociados al Cáncer/patología , Carcinoma Ductal de Mama/patología , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Proteoma/genética , Análisis de Matrices Tisulares , Microambiente Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Prostate ; 77(11): 1213-1220, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28656603

RESUMEN

BACKGROUND: Benign prostatic hyperplasia (BPH) and associated lower urinary tract symptoms (LUTS) are common conditions. Little is known about their etiologies except that studies have suggested a substantial heritable component. Our objective is to provide a comprehensive, genome-wide evaluation of inherited risks and possible mechanisms of etiology in BPH. METHODS: We performed a three-stage, genome-wide association study (GWAS) of men from three independent populations, the REduction by DUtasteride of prostate Cancer Events (REDUCE) trial, the CLUE II cohort, and a Finnish hospital-based population. DNA samples were genotyped using the Illumina HumanOmniExpress BeadChip in REDUCE and CLUE II, and using the Sequenom iPLEX system for the confirmation stage in the Finnish population. A logistic regression model was used to evaluate the association between each SNP and BPH/LUTS. RESULTS: Fourteen SNPs reached P < 5.0 × 10-4 in the meta-analysis of the two GWASs (CLUE II and REDUCE). A total of 773 SNPs were chosen for the confirmation step in the Finish cohort. Only one SNP (rs17144046) located ∼489 kb downstream of GATA3 remained significant after correction for multiple testing (P < 6.5 × 10-5 ). This SNP marginally reached the GWAS significance level after performing a meta-analysis of the three stages (P-meta = 8.89 × 10-7 ). Expression quantitative trait loci (eQTL) analyses showed that the risk allele (G) of rs17144046 was significantly associated with increased expression of GATA3 (P = 0.017). Reported studies indicated a close correlation between GATA3 and BPH pathogenesis and progression. CONCLUSIONS: Rs17144046 located near GATA3 was significantly associated with BPH/LUTS in three independent populations, but did not reach a stringent GWAS significance level. Genetic variants of GATA3 may play a role in the inherited susceptibility and etiology of BPH/LUTS. Further research in this area is needed.


Asunto(s)
Factor de Transcripción GATA3/genética , Predisposición Genética a la Enfermedad/genética , Variación Genética/genética , Estudio de Asociación del Genoma Completo/métodos , Síntomas del Sistema Urinario Inferior/genética , Hiperplasia Prostática/genética , Anciano , Estudios de Cohortes , Método Doble Ciego , Predisposición Genética a la Enfermedad/epidemiología , Humanos , Síntomas del Sistema Urinario Inferior/diagnóstico , Síntomas del Sistema Urinario Inferior/epidemiología , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple/genética , Hiperplasia Prostática/diagnóstico , Hiperplasia Prostática/epidemiología
14.
FASEB J ; 30(10): 3441-3452, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27383183

RESUMEN

The role of tumor heterogeneity in regulating disease progression is poorly understood. We hypothesized that interactions between subpopulations of cancer cells can affect the progression of tumors selecting for a more aggressive phenotype. We developed an in vivo assay based on the immortalized nontumorigenic breast cell line MCF10A and its Ras-transformed derivatives AT1 (mildly tumorigenic) and CA1d (highly tumorigenic). CA1d cells outcompeted MCF10A, forming invasive tumors. AT1 grafts were approximately 1% the size of CA1d tumors when initiated using identical cell numbers. In contrast, CA1d/AT1 mixed tumors were larger than tumors composed of AT1 alone (100-fold) or CA1d (3-fold), suggesting cooperation in tumor growth. One of the mechanisms whereby CA1d and AT1 were found to cooperate was by modulation of TGF-α and TGF-ß signaling. Both of these molecules were sufficient to induce changes in AT1 proliferative potential in vitro. Reisolation of AT1 tumor-derived (AT1-TD) cells from these mixed tumors revealed that AT1-TD cells grew in vivo, forming tumors as large as tumorigenic CA1d cells. Cooperation between subpopulations of cancer epithelium is an understudied mechanism of tumor growth and invasion that may have implications on tumor resistance to current therapies.-Franco, O. E., Tyson, D. R., Konvinse, K. C., Udyavar, A. R., Estrada, L., Quaranta, V., Crawford, S. E., Hayward, S. W. Altered TGF-α/ß signaling drives cooperation between breast cancer cell populations.


Asunto(s)
Neoplasias de la Mama/metabolismo , Movimiento Celular/fisiología , Transformación Celular Neoplásica/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador alfa/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Epitelio/metabolismo , Epitelio/patología , Humanos , Ratones SCID
15.
Differentiation ; 91(4-5): 139-51, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26546040

RESUMEN

The use of lineage tracing in transgenic mouse models has revealed an abundance of subcellular phenotypes responsible for maintaining prostate homeostasis. The ability to use fresh human tissues to examine the hypotheses generated by these mouse experiments has been greatly enhanced by technical advances in tissue processing, flow cytometry and cell culture. We describe in detail the optimization of protocols for each of these areas to facilitate research on solving human prostate diseases through the analysis of human tissue.


Asunto(s)
Diferenciación Celular/genética , Separación Celular/métodos , Células Epiteliales/citología , Próstata/citología , Animales , Citometría de Flujo , Humanos , Masculino , Ratones , Ratones Transgénicos , Próstata/crecimiento & desarrollo
16.
J Biol Chem ; 290(13): 8271-82, 2015 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-25670862

RESUMEN

Prostate cancer remains the second highest contributor to male cancer-related lethality. The transition of a subset of tumors from indolent to invasive disease is associated with a poor clinical outcome. Activation of the epithelial to mesenchymal transition (EMT) genetic program is a major risk factor for cancer progression. We recently reported that secreted extracellular Hsp90 (eHsp90) initiates EMT in prostate cancer cells, coincident with its enhanced expression in mesenchymal models. Our current work substantially extended these findings in defining a pathway linking eHsp90 signaling to EZH2 function, a methyltransferase of the Polycomb repressor complex. EZH2 is also implicated in EMT activation, and its up-regulation represents one of the most frequent epigenetic alterations during prostate cancer progression. We have now highlighted a novel epigenetic function for eHsp90 via its modulation of EZH2 expression and activity. Mechanistically, eHsp90 initiated sustained activation of MEK/ERK, a signal critical for facilitating EZH2 transcriptional up-regulation and recruitment to the E-cadherin promoter. We further demonstrated that an eHsp90-EZH2 pathway orchestrates an expanded repertoire of EMT-related events including Snail and Twist expression, tumor cell motility, and anoikis resistance. To evaluate the role of eHsp90 in vivo, eHsp90 secretion was stably enforced in a prostate cancer cell line resembling indolent disease. Remarkably, eHsp90 was sufficient to induce tumor growth, suppress E-cadherin, and initiate localized invasion, events that are exquisitely dependent upon EZH2 function. In summary, our findings illuminate a hitherto unknown epigenetic function for eHsp90 and support a model wherein tumor eHsp90 functions as a rheostat for EZH2 expression and activity to orchestrate mesenchymal properties and coincident aggressive behavior.


Asunto(s)
Proteínas HSP90 de Choque Térmico/metabolismo , Proteínas del Grupo Polycomb/fisiología , Neoplasias de la Próstata/patología , Animales , Antígenos CD , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular Tumoral , Proteína Potenciadora del Homólogo Zeste 2 , Epigénesis Genética , Transición Epitelial-Mesenquimal , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Proteínas HSP90 de Choque Térmico/fisiología , Humanos , Sistema de Señalización de MAP Quinasas , Masculino , Ratones SCID , Invasividad Neoplásica , Trasplante de Neoplasias , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Carga Tumoral
17.
Prostate ; 76(11): 1004-18, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27197599

RESUMEN

BACKGROUND: Benign prostatic hyperplasia (BPH) is treated with 5α-reductase inhibitors (5ARI). These drugs inhibit the conversion of testosterone to dihydrotestosterone resulting in apoptosis and prostate shrinkage. Most patients initially respond to 5ARIs; however, failure is common especially in inflamed prostates, and often results in surgery. This communication examines a link between activation of NF-κB and increased expression of SRD5A2 as a potential mechanism by which patients fail 5ARI therapy. METHODS: Tissue was collected from "Surgical" patients, treated specifically for lower urinary tract symptoms secondary to advanced BPH; and, cancer free transition zone from "Incidental" patients treated for low grade, localized peripheral zone prostate cancer. Clinical, molecular and histopathological profiles were analyzed. Human prostatic stromal and epithelial cell lines were genetically modified to regulate NF-κB activity, androgen receptor (AR) full length (AR-FL), and AR variant 7 (AR-V7) expression. RESULTS: SRD5A2 is upregulated in advanced BPH. SRD5A2 was significantly associated with prostate volume determined by Transrectal Ultrasound (TRUS), and with more severe lower urinary tract symptoms (LUTS) determined by American Urological Association Symptom Score (AUASS). Synthesis of androgens was seen in cells in which NF-κB was activated. AR-FL and AR-V7 expression increased SRD5A2 expression while forced activation of NF-κB increased all three SRD5A isoforms. Knockdown of SRD5A2 in the epithelial cells resulted in significant reduction in proliferation, AR target gene expression, and response to testosterone (T). In tissue recombinants, canonical NF-κB activation in prostatic epithelium elevated all three SRD5A isoforms and resulted in in vivo growth under castrated conditions. CONCLUSION: Increased BPH severity in patients correlates with SRD5A2 expression. We demonstrate that NF-κB and AR-V7 upregulate SRD5A expression providing a mechanism to explain failure of 5ARI therapy in BPH patients. Prostate 76:1004-1018, 2016. © 2016 Wiley Periodicals, Inc.


Asunto(s)
3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/genética , Inhibidores de 5-alfa-Reductasa/uso terapéutico , Resistencia a Medicamentos , FN-kappa B/fisiología , Hiperplasia Prostática/tratamiento farmacológico , Receptores Androgénicos/fisiología , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/fisiología , Animales , Apoptosis , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Isoenzimas/genética , Isoenzimas/fisiología , Síntomas del Sistema Urinario Inferior/patología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Ratones , Ratones Desnudos , FN-kappa B/antagonistas & inhibidores , Orquiectomía , Próstata/patología , Hiperplasia Prostática/patología , Hiperplasia Prostática/cirugía , Neoplasias de la Próstata Resistentes a la Castración , Testosterona/biosíntesis , Insuficiencia del Tratamiento , Regulación hacia Arriba
18.
Prostate ; 76(5): 491-511, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26709083

RESUMEN

BACKGROUND: Benign prostatic hyperplasia (BPH) is a common, chronic progressive disease. Inflammation is associated with prostatic enlargement and resistance to 5α-reductase inhibitor (5ARI) therapy. Activation of the nuclear factor-kappa B (NF-κB) pathway is linked to both inflammation and ligand-independent prostate cancer progression. METHODS: NF-κB activation and androgen receptor variant (AR-V) expression were quantified in transition zone tissue samples from patients with a wide range of AUASS from incidental BPH in patients treated for low grade, localized peripheral zone prostate cancer to advanced disease requiring surgical intervention. To further investigate these pathways, human prostatic stromal and epithelial cell lines were transduced with constitutively active or kinase dead forms of IKK2 to regulate canonical NF-κB activity. The effects on AR full length (AR-FL) and androgen-independent AR-V expression as well as cellular growth and differentiation were assessed. RESULTS: Canonical NF-κB signaling was found to be upregulated in late versus early stage BPH, and to be strongly associated with non-insulin dependent diabetes mellitus. Elevated expression of AR-variant 7 (AR-V7), but not other AR variants, was found in advanced BPH samples. Expression of AR-V7 significantly correlated with the patient AUASS and TRUS volume. Forced activation of canonical NF-κB in human prostatic epithelial and stromal cells resulted in elevated expression of both AR-FL and AR-V7, with concomitant ligand-independent activation of AR reporters. Activation of NF-κB and over expression of AR-V7 in human prostatic epithelial cells maintained cell viability in the face of 5ARI treatment. CONCLUSION: Activation of NF-κB and AR-V7 in the prostate is associated with increased disease severity. AR-V7 expression is inducible in human prostate cells by forced activation of NF-κB resulting in resistance to 5ARI treatment, suggesting a potential mechanism by which patients may become resistant to 5ARI therapy.


Asunto(s)
FN-kappa B/metabolismo , Próstata/metabolismo , Hiperplasia Prostática/metabolismo , Receptores Androgénicos/metabolismo , Anciano , Línea Celular Tumoral , Supervivencia Celular/genética , Progresión de la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Próstata/patología , Hiperplasia Prostática/genética , Hiperplasia Prostática/patología , Receptores Androgénicos/genética , Transducción de Señal/genética
19.
J Am Soc Nephrol ; 26(3): 597-610, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25071086

RESUMEN

In the kidney, 20-hydroxyeicosatetraenoic acid (20-HETE) is a primary cytochrome P450 4 (Cyp4)-derived eicosanoid that enhances vasoconstriction of renal vessels and induces hypertension, renal tubular cell hypertrophy, and podocyte apoptosis. Hypertension and podocyte injury contribute to diabetic nephropathy and are strong predictors of disease progression. In this study, we defined the mechanisms whereby 20-HETE affects the progression of diabetic nephropathy. We used Cyp4a14KO male mice that exhibit androgen-sensitive hypertension due to increased Cyp4a12-mediated 20-HETE production. We show that, upon induction of diabetes type 1 via streptozotocin injection, Cyp4a14KO male mice developed worse renal disease than streptozotocin-treated wild-type mice, characterized by increased albuminuria, mesangial expansion, glomerular matrix deposition, and thickness of the glomerular basement membranes. Castration blunted androgen-mediated Cyp4a12 synthesis and 20-HETE production, normalized BP, and ameliorated renal damage in diabetic Cyp4a14KO mice. Notably, treatment with a 20-HETE antagonist or agents that normalized BP without affecting Cyp4a12 expression and 20-HETE biosynthesis also ameliorated diabetes-mediated renal damage and albuminuria in Cyp4a14KO male mice. Taken together, these results suggest that hypertension is the major contributor to 20-HETE-driven diabetes-mediated kidney injury.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Nefropatías Diabéticas/etiología , Ácidos Hidroxieicosatetraenoicos/metabolismo , Hipertensión/complicaciones , Animales , Colágeno/metabolismo , Sistema Enzimático del Citocromo P-450/genética , Familia 4 del Citocromo P450 , Nefropatías Diabéticas/patología , Membrana Basal Glomerular/patología , Hidralazina , Hidroclorotiazida , Ácidos Hidroxieicosatetraenoicos/antagonistas & inhibidores , Masculino , Ratones Noqueados , Orquiectomía , Sistema Renina-Angiotensina , Reserpina , Sodio/metabolismo
20.
Am J Pathol ; 184(10): 2598-607, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25088980

RESUMEN

The androgen receptor (AR) in stromal cells contributes significantly to the development and growth of prostate during fetal stages as well as during prostate carcinogenesis and cancer progression. During prostate development, stromal AR induces and promotes epithelial cell growth, as observed from tissue recombinant and mouse knockout studies. During prostate carcinogenesis and progression, the stromal cells begin to lose AR expression as early as at the stage of high-grade prostatic intraepithelial neoplasia. The extent of loss of stromal AR is directly proportional to the degree of differentiation (Gleason grade) and progression of prostate cancer (PCa). Co-culture studies suggested that stromal AR inhibits the growth of malignant epithelial cells, possibly through expression of certain paracrine factors in the presence of androgens. This functional reversal of stromal AR, from growth promotion during fetal prostate development to mediating certain growth-inhibiting effects in cancer, explains to some extent the reason that loss of AR expression in stromal cells may be crucial for development of resistance to androgen ablation therapy for PCa. From a translational perspective, it generates the need to re-examine the current therapeutic options and opens a fundamental new direction for therapeutic interventions, especially in advanced PCa.


Asunto(s)
Andrógenos/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/patología , Receptores Androgénicos/metabolismo , Progresión de la Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Masculino , Próstata/metabolismo , Próstata/patología , Neoplasias de la Próstata/metabolismo , Células del Estroma/metabolismo , Células del Estroma/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA