Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Adv Exp Med Biol ; 889: 105-18, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26658999

RESUMEN

microRNAs are noncoding RNAs that are important for embryonic stem cell development and epithelial to mesenchymal transition (EMT). Tumor cells hijack EMT and stemness to grow and metastasize to distant organs including bone. In the tumor microenvironment, tumor cells interact with the stromal fibroblasts at the primary and metastatic sites and this interaction leads to tumor growth, EMT, and bone metastasis. Tumor-stromal interactions are a dynamic process that involves both cell-cell communications and extracellular vesicles and soluble factors. Growing body of evidence suggests that microRNAs are part of the payload that comprises the extracellular vesicles. microRNAs induce reactive stroma and thus convert normal stroma into tumor-associated stroma to promote aggressive tumorigenicity in vitro and in vivo. Landmark published studies demonstrate that expression of specific microRNAs of DLK1-DIO3 stem cell cluster correlates with patient survival in metastatic prostate cancer. Thus, microRNAs mediate tumor growth, EMT, and metastasis through cell intrinsic mechanisms and extracellular communications and could be novel biomarkers and therapeutic targets in bone metastatic prostate cancer.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Neoplasias de la Próstata/genética , Microambiente Tumoral/genética , Biomarcadores de Tumor/genética , Epigénesis Genética/genética , Humanos , Masculino , Modelos Genéticos , Células Madre Neoplásicas/metabolismo , Transducción de Señal/genética
2.
Semin Cell Dev Biol ; 21(1): 26-32, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19948237

RESUMEN

Cancer development is complex and involves several layers of interactions and pleotropic signaling mechanisms leading to progression. Cancer cells associate with resident stromal fibroblasts, smooth muscle cells, macrophages, endothelium, neurons and migrating cells at metastatic sites and phenotypically and genotypically activate them. These become an integral part of the cancer cell community through activated cell signaling mechanisms. During this process, the cancer cells and cells in the cancer microenvironment "co-evolve" in part due to oxidative stress, and acquire the ability to mimic other cell types (which can be termed osteomimicry, vasculomimicry, neuromimicry and stem cell mimicry), and undergo transition from epithelium to mesenchyme with definitive morphologic and behavioral modifications. In our laboratory, we demonstrated that prostate cancer cells co-evolve in their genotypic and phenotypic characters with stroma and acquire osteomimetic properties allowing them to proliferate and survive in the skeleton as bone metastasis. Several signaling interactions in the bone microenvironment, mediated by reactive oxygen species, soluble and membrane bound factors, such as superoxide, beta2-microglobulin and RANKL have been described. Targeting the signaling pathways in the cancer-associated stromal microenvironment in combination with known conventional therapeutic modalities could have a synergistic effect on cancer treatment. Since cancer cells are constantly interacting and acquiring adaptive and survival changes primarily directed by their microenvironment, it is imperative to delineate these interactions and co-target both cancer and stroma to improve the treatment and overall survival of cancer patients.


Asunto(s)
Neoplasias de la Próstata/patología , Células del Estroma , Neoplasias Óseas/secundario , Progresión de la Enfermedad , Humanos , Masculino , Metástasis de la Neoplasia , Osteoblastos/metabolismo , Osteoblastos/patología , Osteoprotegerina/metabolismo , Estrés Oxidativo , Neoplasias de la Próstata/fisiopatología , Ligando RANK/metabolismo , Células del Estroma/metabolismo , Células del Estroma/patología
3.
Chin J Physiol ; 55(6): 390-7, 2012 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-23286446

RESUMEN

Recent evidence has demonstrated that detection of changes in the levels of urinary vascular endothelial growth factor (VEGF) and tissue a disintegrin and metalloproteinase 9 (ADAM9) is effective in determining prostate cancer progression. To evaluate the combined application of VEGF and ADAM9 as early progression markers of lethal phenotypic cancer, quantification of urinary VEGF and tissue ADAM9 expression was studied in patients with late stage prostate cancer. Tissue biopsies were collected during palliative transurethral resection of prostate (TURP) surgery, and urine samples were collected before hormone therapy and 3, 6 and 12 months post-TURP. We observed a nearly 100% correlation between increasing urinary VEGF levels over time and prostate cancer progression, but no correlation was observed when comparing urinary VEGF concentrations at a single time point and cancer progression. In addition, we also observed correlation of increasing ADAM9 nuclear positive staining and lethal phenotypic transition. Statistical analysis revealed that both the increase in urinary VEGF level and the presence of the tissue ADAM9 nuclear staining were significantly correlated with the risk of patients with relapse prostate cancer (P < 0.05). Thus, we suggest that combination of detection of changes in urinary VEGF and tissue staining of ADAM9 may be accurate for predicting the mortality of patients with prostate cancer during hormone therapy.


Asunto(s)
Recurrencia Local de Neoplasia , Factor A de Crecimiento Endotelial Vascular , Biopsia , Progresión de la Enfermedad , Humanos , Masculino , Neoplasias de la Próstata/metabolismo
4.
Prostate ; 71(3): 232-40, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20672324

RESUMEN

INTRODUCTION: Recent studies demonstrated the importance of ADAM9 in prostate cancer relapse upon therapy. In this study, we determined the role of ADAM9 in the therapeutic resistance to radiation and chemotherapy. MATERIALS AND METHODS: ADAM9 was either transiently or stably knocked down in C4-2 prostate cancer cells. The sensitivity of ADAM9 knockdown cells toward radiation and chemotherapeutic agents were determined. Additionally, the effects of ADAM9 knockdown on prostate cancer cell morphology, biochemical and functional alterations were accessed. RESULTS: Both transient and stable knockdown of ADAM9 resulted in increased apoptosis and increased sensitivity to radiation. ADAM9 knockdown also increased prostate cancer sensitivity to several chemotherapeutic drugs. ADAM9 knockdown resulted in increased E-cadherin and altered integrin expression and underwent phenotypic epithelial transition. These were reflected by the morphological, biochemical, and functional alterations in the ADAM9 knockdown cells. CONCLUSIONS: ADAM9 plays a crucial role in prostate cancer progression and therapeutic resistance in part by altering E-cadherin and integrin expression. ADAM9 is an important target for the consideration of treating prostate cancer patients who developed therapeutic resistance and disease relapse.


Asunto(s)
Proteínas ADAM/fisiología , Epitelio/patología , Proteínas de la Membrana/fisiología , Neoplasias de la Próstata/terapia , Proteínas ADAM/análisis , Proteínas ADAM/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Cadherinas/análisis , Línea Celular Tumoral , Humanos , Integrinas/análisis , Masculino , Proteínas de la Membrana/análisis , Proteínas de la Membrana/antagonistas & inhibidores , Fenotipo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Tolerancia a Radiación , Superóxidos/análisis
5.
Int Immunol ; 22(7): 583-92, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20497960

RESUMEN

MicroRNAs 125a and 125b are predicted to be able to bind to the B lymphocyte-induced maturation protein-1 (BLIMP-1) and IFN regulatory protein-4 (IRF-4) transcription factors, which are essential for plasma cell differentiation. A computational survey of the human and mouse genomes revealed that miR-125a and miR-125b are members of a multigene family located in paralogous clusters. The miR-125a cluster on chromosome 19 in humans includes miR-99b and let-7e, whereas the miR-125b cluster on chromosome 21 includes miR-99a and miR-let-7c. Our analysis of the expression profiles for these six miRs during B lineage differentiation indicated that mature miR-125a, miR-125b, miR-99b and let-7e transcripts are preferentially expressed by the actively dividing centroblasts in germinal centers (GC). However, miR-99b and let-7e are not predicted to bind BLIMP-1 or IRF-4 transcripts, and binding to the untranslated region of BLIMP-1 and IRF-4 messenger RNAs could be confirmed only for miR-125b. When the effect of miR-125b over-expression on terminal B cell differentiation was evaluated in an LPS-responsive B cell line, the induction of BLIMP-1 expression and IgM secretion was inhibited in this model system. Furthermore, miR-125b over-expression inhibited the differentiation of primary B cells and compromised the survival of cultured myeloma cells. These findings suggest that miR-125b promotes B lymphocyte diversification in GC by inhibiting premature utilization of essential transcription factors for plasma cell differentiation.


Asunto(s)
Linfocitos B/citología , Diferenciación Celular , Centro Germinal/citología , MicroARNs/genética , Animales , Linfocitos B/inmunología , Secuencia de Bases , Línea Celular , Análisis por Conglomerados , Secuencia Conservada , Centro Germinal/inmunología , Humanos , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/inmunología , Ratones , Modelos Animales , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Proteínas Represoras/genética , Proteínas Represoras/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética/genética
6.
Mol Cancer Ther ; 7(8): 2367-76, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18723484

RESUMEN

The activation of nuclear factor-kappaB (NF-kappaB) is thought to protect cancer cells against therapy-induced cytotoxicity. RelB, a member of the NF-kappaB family in the alternative pathway, is uniquely expressed at a high level in prostate cancer with high Gleason scores. Here, we show that ionizing radiation (IR) enhances nuclear import of RelB, leading to up-regulation of its target gene, manganese superoxide dismutase (MnSOD), and renders prostate cancer cells resistant to IR. To selectively block RelB nuclear import, we designed a cell-permeable SN52 peptide, a variant of the SN50 peptide that has been shown to block nuclear import of NF-kappaB family members in the classic pathway. Inhibition of IR-induced NF-kappaB activation by SN50 and SN52 was achieved by selectively interrupting the association of p50 and p52 with nuclear import factors importin-alpha1 and importin-beta1. Importantly, SN52 seems to be more efficient for radiosensitization of prostate cancer cells at clinically relevant radiation doses and has less cytotoxicity to normal prostate epithelial cells compared with the toxicity observed with SN50. These results suggest that targeting the alternative pathway is a promising approach to selectively radiosensitize prostate cancers and that SN52 may serve as a prototype biological agent for sensitizing prostate cancers to clinically relevant doses of IR.


Asunto(s)
Núcleo Celular/efectos de los fármacos , FN-kappa B/antagonistas & inhibidores , Péptidos/farmacología , Neoplasias de la Próstata/patología , Fármacos Sensibilizantes a Radiaciones/farmacología , Secuencia de Bases , Línea Celular Tumoral , Núcleo Celular/metabolismo , Células Cultivadas , Cartilla de ADN , Humanos , Masculino , Neoplasias de la Próstata/metabolismo , Tolerancia a Radiación/efectos de los fármacos , Radiación Ionizante
7.
Asian J Urol ; 6(1): 65-81, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30775250

RESUMEN

This article describes cell signaling network of metastatic prostate cancer (PCa) to bone and visceral organs in the context of tumor microenvironment and for the development of novel therapeutics. The article focuses on our recent progress in the understanding of: 1) The plasticity and dynamics of tumor-stroma interaction; 2) The significance of epigenetic reprogramming in conferring cancer growth, invasion and metastasis; 3) New insights on altered junctional communication affecting PCa bone and brain metastases; 4) Novel strategies to overcome therapeutic resistance to hormonal antagonists and chemotherapy; 5) Genetic-based therapy to co-target tumor and bone stroma; 6) PCa-bone-immune cell interaction and TBX2-WNTprotein signaling in bone metastasis; 7) The roles of monoamine oxidase and reactive oxygen species in PCa growth and bone metastasis; and 8) Characterization of imprinting cluster of microRNA, in tumor-stroma interaction. This article provides new approaches and insights of PCa metastases with emphasis on basic science and potential for clinical translation. This article referenced the details of the various approaches and discoveries described herein in peer-reviewed publications. We dedicate this article in our fond memory of Dr. Donald S. Coffey who taught us the spirit of sharing and the importance of focusing basic science discoveries toward translational medicine.

8.
Prostate ; 68(15): 1599-606, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18668526

RESUMEN

BACKGROUND: MicroRNAs (miRNA) are gene regulators and play an important role in response to cellular stress. METHODS: Using multiplexed quantitative real-time PCR we performed global miRNA screening of prostate cancer cells in response to radiation treatment. RESULTS: Several miRNA were significantly altered in response to radiation treatment. Significant changes were observed in miR-521 and miR-34c. To determine the role of miR-521 in radiation response we transiently overexpressed miR-521 using miR-521 mimic. The miR-521 mimic significantly sensitized prostate cancer cells to radiation treatment. Conversely, ectopic inhibition of miR-521 resulted in radiation resistance of prostate cancer cells. To determine the mechanism by which miR-521 modulates radiation sensitivity we measured the expression levels of one of its predicted target protein, Cockayne syndrome protein A (CSA). CSA is a DNA repair protein, and its levels correlated inversely with the levels of miR-521. Radiation treatment downregulated the levels of miR-521 and upregulated CSA protein. Similarly, ectopic inhibition of miR-521 resulted in increased CSA protein levels. Therefore by altering the levels of CSA protein, miR-521 sensitized prostate cancer cells to radiation treatment. CONCLUSION: miR-521 modulates the expression levels of DNA repair protein, CSA and plays an important role, in the radio-sensitivity of prostate cancer cell lines. Thus miR-521 can be a potential target for enhancing the effect of radiation treatment on prostate cancer cells.


Asunto(s)
Línea Celular Tumoral/efectos de la radiación , MicroARNs/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Regulación hacia Abajo , Humanos , Masculino , MicroARNs/farmacología , Tolerancia a Radiación , Superóxido Dismutasa/metabolismo , Factores de Transcripción/metabolismo , Regulación hacia Arriba
9.
Mol Cancer Ther ; 6(7): 2048-56, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17604335

RESUMEN

Nuclear factor-kappaB provides an adaptive response to protect cancer cells against cytotoxicity induced by redox active therapeutics. RelB is uniquely expressed at a high level in prostate cancer with high Gleason scores. Recently, we showed that the level of RelB rapidly increases in androgen-independent prostate cancer cells after exposure to ionizing radiation (IR), leading to a reduction in intrinsic radiosensitivity. Here, we show that interaction of 1alpha,25-dihydroxyvitamin D(3) [1alpha,25-(OH)(2)D(3)] with the vitamin D receptor significantly enhances radiosensitivity of prostate cancer cells at clinically relevant radiation doses. The radiosensitization effect of 1alpha,25-(OH)(2)D(3) is mediated, at least in part, by selectively suppressing IR-mediated RelB activation, leading to a reduced expression of its target gene MnSOD, a primary antioxidant enzyme in mitochondria. These results suggest that suppression of manganese superoxide dismutase is a mechanism by which 1alpha,25-(OH)(2)D(3) exerts its radiosensitization effect and that 1alpha,25-(OH)(2)D(3) may serve as an effective pharmacologic agent for selectively sensitizing prostate cancer cells to IR via suppression of antioxidant responses in mitochondria.


Asunto(s)
Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Tolerancia a Radiación/efectos de los fármacos , Superóxido Dismutasa/antagonistas & inhibidores , Factor de Transcripción ReIB/metabolismo , Vitamina D/análogos & derivados , Antioxidantes/metabolismo , Humanos , Masculino , ARN Interferente Pequeño/metabolismo , Vitamina D/farmacología
10.
Bio Protoc ; 7(4): e2138, 2017 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-34458458

RESUMEN

miRNA are short non-coding RNA which inhibit translation of mRNA. miRNA regulate several cellular processes. Certain miRNA are known to induce oncogenesis. miRNA can be measured by real-time PCR and be imaged using a combination of in situ hybridization (ISH) and quantum dots (QD). The advantage of using quantum dots is that several miRNA can be simultaneously measured using multiplexed QD. Additionally, miRNA can be visualized in different regions of the tissue. Since miRNA are biomarkers of various disease states, miRNA can be visualized and quantitated in tissue sections for diagnostic and prognostic purposes. Here we describe ISH-QD analysis of tissue sections. Tissue sections from xenografts or clinical specimens are used. These are deparaffinized, treated with Proteinase K and hybridized with a biotin-probe to specific to the miRNA. The in situ hybridization is performed by labeling the biotin-probes and followed by labeling with streptavidin tagged quantum dots. Image acquisition of the quantum dots is performed and analyzed for the miRNA expression levels. Combining ISH and QD gives a powerful tool to detect miRNA in different cells of the tissue.

11.
Bio Protoc ; 7(4): e2139, 2017 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-34458459

RESUMEN

Cancer cells and cancer associated stromal cells co-evolve secrete extracellular vesicles to the surrounding regions and regulate several processes involved in cancer metastasis. miRNAs have been known to be mediators of cancer progression and metastasis. miRNAs consist of short noncoding RNA. miRNAs are stable in extracellular fluids such as serum, plasma and urine. miRNAs are secreted by cells in normal and diseased conditions. miRNAs signatures have been identified specific to certain disease conditions. Therefore they are valuable biomarkers for different diseases. In our study we identified certain miRNAs, miR-409-3p and miR-409-5p, which were secreted by activated stromal fibroblast cells and were taken up by cancer cells to induce explosive tumor growth, through activation of epithelial to mesenchymal transition of cancer cells. Here we describe a procedure to determine miRNAs (miR-409-3p and miR-409-5p) in extracellular vesicles, which were secreted by prostate cancer stromal cells expressing miR-409. In this procedure, conditioned media from the stromal fibroblasts was used to extract the vesicular fraction. RNA was purified from the vesicular fraction, and specific miRNA was reverse transcribed and quantitated using real-time PCR assay.

12.
Oncotarget ; 6(42): 44072-83, 2015 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-26624980

RESUMEN

FYN is a SRC family kinase (SFK) that has been shown to be up-regulated in human prostate cancer (PCa) tissues and cell lines. In this study, we observed that FYN is strongly up-regulated in human neuroendocrine PCa (NEPC) tissues and xenografts, as well as cells derived from a NEPC transgenic mouse model. In silico analysis of FYN expression in prostate cancer cell line databases revealed an association with the expression of neuroendocrine (NE) markers such as CHGA, CD44, CD56, and SYP. The loss of FYN abrogated the invasion of PC3 and ARCaPM cells in response to MET receptor ligand HGF. FYN also contributed to the metastatic potential of NEPC cells in two mouse models of visceral metastasis with two different cell lines (PC3 and TRAMPC2-RANKL). The activation of MET appeared to regulate neuroendocrine (NE) features as evidenced by increased expression of NE markers in PC3 cells with HGF. Importantly, the overexpression of FYN protein in DU145 cells was directly correlated with the increase of CHGA. Thus, our data demonstrated that the neuroendocrine differentiation that occurs in PCa cells is, at least in part, regulated by FYN kinase. Understanding the role of FYN in the regulation of NE markers will provide further support for ongoing clinical trials of SFK and MET inhibitors in castration-resistant PCa patients.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Diferenciación Celular , Movimiento Celular , Neoplasias Hepáticas/enzimología , Tumores Neuroendocrinos/enzimología , Neoplasias de la Próstata/enzimología , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Animales , Biomarcadores de Tumor/genética , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular , Cromogranina A/metabolismo , Simulación por Computador , Bases de Datos Genéticas , Relación Dosis-Respuesta a Droga , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/secundario , Masculino , Ratones Endogámicos C57BL , Ratones SCID , Ratones Transgénicos , Invasividad Neoplásica , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/secundario , Fenotipo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-fyn/genética , Proteínas Proto-Oncogénicas c-met/metabolismo , Transducción de Señal , Factores de Tiempo , Transfección , Carga Tumoral , Regulación hacia Arriba
13.
Anticancer Agents Med Chem ; 14(3): 343-52, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23848204

RESUMEN

ß2-microglobulin (ß2-m) has become the focus of intense scrutiny since the discovery of its undesirable roles promoting osteomimicry and cancer progression. ß2-m is a well-known housekeeping protein that forms complexes with the heavy chain of major histocompatibility complex class I molecules, which are heterodimeric cell surface proteins that present antigenic peptides to cytotoxic T cells. On recognition of foreign peptide antigens on cell surfaces, T cells actively bind and lyse antigen-presenting cancer cells. In addition to its roles in tumor immunity, ß2-m has two different functions in cancer cells, either tumor promoting or tumor suppressing, in cancer cell context-dependent manner. Our studies have demonstrated that ß2-m is involved extensively in the functional regulation of growth, survival, apoptosis, and even metastasis of cancer cells. We found that ß2-m is a soluble growth factor and a pleiotropic signaling molecule which interacts with its receptor, hemochromatosis protein, to modulate epithelial-to-mesenchymal transition (EMT) through iron-responsive pathways. Specific antibodies against ß2-m have remarkable tumoricidal activity in cancer, through ß2-m action on iron flux, alterations of intracellular reactive oxygen species, DNA damage and repair enzyme activities, ß-catenin activation and cadherin switching, and tumor responsiveness to hypoxia. These novel functions of ß2-m and ß2-m signaling may be common to several solid tumors including human lung, breast, renal, and prostate cancers. Our experimental results could lead to the development of a novel class of antibody-based pharmaceutical agents for cancer growth control. In this review, we briefly summarize the recent data regarding ß2-m as a promising new cancer therapeutic target and discuss antagonizing this therapeutic target with antibody therapy for the treatment of localized and disseminated cancers.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/terapia , Microglobulina beta-2/metabolismo , Animales , Anticuerpos/uso terapéutico , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/metabolismo , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Transición Epitelial-Mesenquimal , Genes MHC Clase I , Humanos , Células Madre Mesenquimatosas/metabolismo , Terapia Molecular Dirigida , Neoplasias/metabolismo , Neoplasias/patología , Transducción de Señal , Células Tumorales Cultivadas , Microglobulina beta-2/inmunología
14.
Clin Cancer Res ; 20(24): 6559-69, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25324143

RESUMEN

PURPOSE: MicroRNAs in the delta-like 1 homolog-deiodinase, iodothyronine 3 (DLK1-DIO3) cluster have been shown to be critical for embryonic development and epithelial to mesenchymal transition (EMT). DLK1-DIO3 cluster miRNAs are elevated in the serum of patients with metastatic cancer. However, the biologic functions of these miRNAs in the EMT and metastasis of cancer cells are poorly understood. We previously demonstrated the oncogenic and metastatic role of miR-409-3p/5p, a member of this cluster, in prostate cancer. In this study, we defined the role of miR-154* and miR-379, two key members of this cluster, in prostate cancer progression and bone metastasis in both cell line models and clinical specimens. EXPERIMENTAL DESIGN: Genetic manipulation of miR-154* and miR-379 was performed to determine their role in tumor growth, EMT, and bone metastasis in mouse models. We determined the expression of miR-154* in prostate cancer clinical samples and bone metastasis samples using in situ hybridization and quantum dot labeling. RESULTS: Elevated expression of miR-154* and miR-379 was observed in bone metastatic prostate cancer cell lines and tissues, and miR-379 expression correlated with progression-free survival of patients with prostate cancer. Intracardiac inoculation (to mimic systemic dissemination) of miR-154* inhibitor-treated bone metastatic ARCaPM prostate cancer cells in mice led to decreased bone metastasis and increased survival. CONCLUSION: miR-154* and miR-379 play important roles in prostate cancer biology by facilitating tumor growth, EMT, and bone metastasis. This finding has particular translational importance because miRNAs in the DLK1-DIO3 cluster can be attractive biomarkers and possible therapeutic targets to treat bone metastatic prostate cancer.


Asunto(s)
Neoplasias Óseas/secundario , Transición Epitelial-Mesenquimal/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Yoduro Peroxidasa/genética , Proteínas de la Membrana/genética , MicroARNs/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Animales , Proteínas de Unión al Calcio , Línea Celular Tumoral , Modelos Animales de Enfermedad , Expresión Génica , Redes Reguladoras de Genes , Xenoinjertos , Humanos , Masculino , Ratones , Familia de Multigenes , Clasificación del Tumor , Metástasis de la Neoplasia , Neoplasias de la Próstata/metabolismo , Interferencia de ARN
15.
Clin Cancer Res ; 20(17): 4636-46, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24963047

RESUMEN

PURPOSE: miR-409-3p/-5p is a miRNA expressed by embryonic stem cells, and its role in cancer biology and metastasis is unknown. Our pilot studies demonstrated elevated miR-409-3p/-5p expression in human prostate cancer bone metastatic cell lines; therefore, we defined the biologic impact of manipulation of miR-409-3p/-5p on prostate cancer progression and correlated the levels of its expression with clinical human prostate cancer bone metastatic specimens. EXPERIMENTAL DESIGN: miRNA profiling of a prostate cancer bone metastatic epithelial-to-mesenchymal transition (EMT) cell line model was performed. A Gleason score human tissue array was probed for validation of specific miRNAs. In addition, genetic manipulation of miR-409-3p/-5p was performed to determine its role in tumor growth, EMT, and bone metastasis in mouse models. RESULTS: Elevated expression of miR-409-3p/-5p was observed in bone metastatic prostate cancer cell lines and human prostate cancer tissues with higher Gleason scores. Elevated miR-409-3p expression levels correlated with progression-free survival of patients with prostate cancer. Orthotopic delivery of miR-409-3p/-5p in the murine prostate gland induced tumors where the tumors expressed EMT and stemness markers. Intracardiac inoculation (to mimic systemic dissemination) of miR-409-5p inhibitor-treated bone metastatic ARCaPM prostate cancer cells in mice led to decreased bone metastasis and increased survival compared with control vehicle-treated cells. CONCLUSION: miR-409-3p/-5p plays an important role in prostate cancer biology by facilitating tumor growth, EMT, and bone metastasis. This finding bears particular translational importance as miR-409-3p/-5p appears to be an attractive biomarker and/or possibly a therapeutic target to treat bone metastatic prostate cancer.


Asunto(s)
Neoplasias Óseas/genética , Carcinogénesis/genética , Transición Epitelial-Mesenquimal/genética , MicroARNs/biosíntesis , Animales , Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , MicroARNs/genética , Neoplasias de la Próstata
16.
PLoS One ; 8(8): e70987, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23951060

RESUMEN

MicroRNA (miRNA or miR) inhibition of oncogenic related pathways has been shown to be a promising therapeutic approach for cancer. Aberrant lipid and cholesterol metabolism is involved in prostate cancer development and progression to end-stage disease. We recently demonstrated that a key transcription factor for lipogenesis, sterol regulatory element-binding protein-1 (SREBP-1), induced fatty acid and lipid accumulation and androgen receptor (AR) transcriptional activity, and also promoted prostate cancer cell growth and castration resistance. SREBP-1 was overexpressed in human prostate cancer and castration-resistant patient specimens. These experimental and clinical results indicate that SREBP-1 is a potential oncogenic transcription factor in prostate cancer. In this study, we identified two miRNAs, miR-185 and 342, that control lipogenesis and cholesterogenesis in prostate cancer cells by inhibiting SREBP-1 and 2 expression and down-regulating their targeted genes, including fatty acid synthase (FASN) and 3-hydroxy-3-methylglutaryl CoA reductase (HMGCR). Both miR-185 and 342 inhibited tumorigenicity, cell growth, migration and invasion in prostate cancer cell culture and xenograft models coincident with their blockade of lipogenesis and cholesterogenesis. Intrinsic miR-185 and 342 expression was significantly decreased in prostate cancer cells compared to non-cancerous epithelial cells. Restoration of miR-185 and 342 led to caspase-dependent apoptotic death in prostate cancer cells. The newly identified miRNAs, miR-185 and 342, represent a novel targeting mechanism for prostate cancer therapy.


Asunto(s)
Apoptosis/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Neoplasias de la Próstata/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 2 de Unión a Elementos Reguladores de Esteroles/genética , Animales , Caspasas/genética , Caspasas/metabolismo , Línea Celular Tumoral , Células Cultivadas , Células Epiteliales/citología , Células Epiteliales/metabolismo , Acido Graso Sintasa Tipo I/genética , Acido Graso Sintasa Tipo I/metabolismo , Humanos , Hidroximetilglutaril-CoA Reductasas/genética , Hidroximetilglutaril-CoA Reductasas/metabolismo , Lipogénesis/genética , Masculino , Ratones , MicroARNs/metabolismo , Trasplante de Neoplasias , Próstata/metabolismo , Próstata/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Transducción de Señal , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Carga Tumoral
17.
PLoS One ; 8(1): e53795, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23342005

RESUMEN

Cancer cells respond to stress by activating a variety of survival signaling pathways. A disintegrin and metalloproteinase (ADAM) 9 is upregulated during cancer progression and hormone therapy, functioning in part through an increase in reactive oxygen species. Here, we present in vitro and in vivo evidence that therapeutic targeting of ADAM9 gene expression by lentivirus-delivered small hairpin RNA (shRNA) significantly inhibited proliferation of human prostate cancer cell lines and blocked tumor growth in a murine model of prostate cancer bone metastasis. Cell cycle studies confirmed an increase in the G1-phase and decrease in the S-phase population of cancer cells under starvation stress conditions, which correlated with elevated intracellular superoxide levels. Microarray data showed significantly decreased levels of regenerating islet-derived family member 4 (REG4) expression in prostate cancer cells with knockdown of ADAM9 gene expression. This REG4 downregulation also resulted in induction of expression of p21(Cip1/WAF1), which negatively regulates cyclin D1 and blocks the G1/S transition. Our data reveal a novel molecular mechanism of ADAM9 in the regulation of prostate cancer cell proliferation, and suggests a combined modality of ADAM9 shRNA gene therapy and cytotoxic agents for hormone refractory and bone metastatic prostate cancer.


Asunto(s)
Proteínas ADAM/genética , Ciclo Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Lectinas Tipo C/metabolismo , Lentivirus/genética , Proteínas de la Membrana/genética , Neoplasias de la Próstata/patología , ARN Interferente Pequeño/genética , Proteínas ADAM/deficiencia , Andrógenos/metabolismo , Animales , Neoplasias Óseas/fisiopatología , Neoplasias Óseas/secundario , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Terapia Genética , Humanos , Masculino , Proteínas de la Membrana/deficiencia , Ratones , Terapia Molecular Dirigida , Osteólisis/genética , Proteínas Asociadas a Pancreatitis , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/terapia , Puntos de Control de la Fase S del Ciclo Celular/genética
18.
PLoS One ; 8(7): e68366, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23874600

RESUMEN

BACKGROUND: Bone metastasis is the most lethal form of several cancers. The ß2-microglobulin (ß2-M)/hemochromatosis (HFE) complex plays an important role in cancer development and bone metastasis. We demonstrated previously that overexpression of ß2-M in prostate, breast, lung and renal cancer leads to increased bone metastasis in mouse models. Therefore, we hypothesized that ß2-M is a rational target to treat prostate cancer bone metastasis. RESULTS: In this study, we demonstrate the role of ß2-M and its binding partner, HFE, in modulating radiation sensitivity and chemo-sensitivity of prostate cancer. By genetic deletion of ß2-M or HFE or using an anti-ß2-M antibody (Ab), we demonstrate that prostate cancer cells are sensitive to radiation in vitro and in vivo. Inhibition of ß2-M or HFE sensitized prostate cancer cells to radiation by increasing iron and reactive oxygen species and decreasing DNA repair and stress response proteins. Using xenograft mouse model, we demonstrate that anti-ß2-M Ab sensitizes prostate cancer cells to radiation treatment. Additionally, anti-ß2-M Ab was able to prevent tumor growth in an immunocompetent spontaneous prostate cancer mouse model. Since bone metastasis is lethal, we used a bone xenograft model to test the ability of anti-ß2-M Ab and radiation to block tumor growth in the bone. Combination treatment significantly prevented tumor growth in the bone xenograft model by inhibiting ß2-M and inducing iron overload. In addition to radiation sensitive effects, inhibition of ß2-M sensitized prostate cancer cells to chemotherapeutic agents. CONCLUSION: Since prostate cancer bone metastatic patients have high ß2-M in the tumor tissue and in the secreted form, targeting ß2-M with anti-ß2-M Ab is a promising therapeutic agent. Additionally, inhibition of ß2-M sensitizes cancer cells to clinically used therapies such as radiation by inducing iron overload and decreasing DNA repair enzymes.


Asunto(s)
Anticuerpos/farmacología , Sobrecarga de Hierro/inducido químicamente , Proteínas de la Membrana/antagonistas & inhibidores , Neoplasias de la Próstata/terapia , Tolerancia a Radiación/efectos de los fármacos , Fármacos Sensibilizantes a Radiaciones/farmacología , Microglobulina beta-2/antagonistas & inhibidores , Animales , Anticuerpos/uso terapéutico , Terapia Combinada , Proteína de la Hemocromatosis , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Hierro/metabolismo , Sobrecarga de Hierro/metabolismo , Masculino , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/inmunología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Tolerancia a Radiación/genética , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Microglobulina beta-2/inmunología , Microglobulina beta-2/metabolismo
19.
Cancer Res ; 71(7): 2600-10, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21427356

RESUMEN

Bone metastasis is one of the predominant causes of cancer lethality. This study demonstrates for the first time how ß2-microglobulin (ß2-M) supports lethal metastasis in vivo in human prostate, breast, lung, and renal cancer cells. ß2-M mediates this process by activating epithelial to mesenchymal transition (EMT) to promote lethal bone and soft tissue metastases in host mice. ß2-M interacts with its receptor, hemochromatosis (HFE) protein, to modulate iron responsive pathways in cancer cells. Inhibition of either ß2-M or HFE results in reversion of EMT. These results demonstrate the role of ß2-M in cancer metastasis and lethality. Thus, ß2-M and its downstream signaling pathways are promising prognostic markers of cancer metastases and novel therapeutic targets for cancer therapy.


Asunto(s)
Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , Neoplasias/metabolismo , Neoplasias/patología , Microglobulina beta-2/metabolismo , Animales , Neoplasias Óseas/inmunología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Femenino , Técnicas de Silenciamiento del Gen , Proteína de la Hemocromatosis , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Huésped Inmunocomprometido , Inmunohistoquímica , Hierro/metabolismo , Neoplasias Renales/inmunología , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Desnudos , Neoplasias/inmunología , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Trasplante Heterólogo , Microglobulina beta-2/antagonistas & inhibidores , Microglobulina beta-2/biosíntesis , Microglobulina beta-2/inmunología
20.
Oncoscience ; 2(9): 743-4, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26501076
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA