Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 37(8): 1524-1535, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28596374

RESUMEN

OBJECTIVE: Bacterial endotoxin (lipopolysaccharide)-mediated sepsis involves dysregulated systemic inflammation, which injures the lung and other organs, often fatally. Vascular endothelial cells act as both targets and mediators of lipopolysaccharide-induced inflammatory responses. Dysfunction of endothelium results in increases of proinflammatory cytokine production and permeability leakage. BMPER (bone morphogenetic protein-binding endothelial regulator), an extracellular modulator of bone morphogenetic protein signaling, has been identified as a vital component in chronic endothelial inflammatory responses and atherosclerosis. However, it is unclear whether BMPER also regulates inflammatory response in an acute setting such as sepsis. To address this question, we investigated the role of BMPER during lipopolysaccharide-induced acute lung injury. APPROACH AND RESULTS: Mice missing 1 allele of BMPER (BMPER+/- mice used in the place of BMPER-/- mice that die at birth) were used for lipopolysaccharide challenge. Lipopolysaccharide-induced pulmonary inflammation and injury was reduced in BMPER+/- mice as shown by several measures, including survival rate, infiltration of inflammatory cells, edema, and production of proinflammatory cytokines. Mechanistically, we have demonstrated that BMPER is required and sufficient for the activation of nuclear factor of activated T cells c1. This BMPER-induced nuclear factor of activated T cells activation is coordinated by multiple signaling pathways, including bone morphogenetic protein-independent low-density lipoprotein receptor-related protein 1-extracellular signal-regulated kinase activation, calcineurin signaling, and low-density lipoprotein receptor-related protein 1ß-mediated nuclear factor 45 nuclear export in response to BMPER treatment. CONCLUSIONS: We conclude that BMPER plays a pivotal role in pulmonary inflammatory response, which provides new therapeutic options against sepsis shock. The new signaling pathway initiated by BMPER/low-density lipoprotein receptor-related protein 1 axis broadens our understanding about BMPER's role in vascular homeostasis.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Proteínas Portadoras/metabolismo , Células Endoteliales/metabolismo , Endotoxinas , Pulmón/irrigación sanguínea , Neumonía/metabolismo , Receptores de LDL/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Animales , Apoptosis , Permeabilidad Capilar , Proteínas Portadoras/genética , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/patología , Predisposición Genética a la Enfermedad , Haploinsuficiencia , Mediadores de Inflamación/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción NFATC/metabolismo , Proteína del Factor Nuclear 45/metabolismo , Fenotipo , Neumonía/inducido químicamente , Neumonía/genética , Neumonía/patología , Interferencia de ARN , Receptores de LDL/genética , Factores de Tiempo , Transfección , Proteínas Supresoras de Tumor/genética
2.
Curr Osteoporos Rep ; 16(6): 642-647, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30203251

RESUMEN

PURPOSE OF REVIEW: Prostate cancer bone metastasis is the lethal progression of the disease. The disease frequently presents with osteoblastic lesions in bone. The tumor-induced bone can cause complications that significantly hamper the quality of life of patients. A better understanding of how prostate cancer induces aberrant bone formation and how the aberrant bone affects the progression and treatment of the disease may improve the therapies for this disease. RECENT FINDINGS: Prostate cancer-induced bone was shown to enhance tumor growth and confer therapeutic resistance in bone metastasis. Clinically, Radium-223, an alpha emitter that selectively targets bone, was shown to improve overall survival in patients, supporting a role of tumor-induced bone in prostate cancer progression in bone. Recently, it was discovered that PCa-induced aberrant bone formation is due, in part, from tumor-associated endothelial cells that were converted into osteoblasts through endothelial-to-osteoblast (EC-to-OSB) conversion by tumor-secreted BMP4. The unique bone-forming phenotype of prostate cancer bone metastasis plays a role in prostate cancer progression in bone and therapy resistance. Therapies that incorporate targeting the tumor-induced osteoblasts or EC-to-OSB conversion mechanism may reduce tumor-induced bone formation and improve therapy outcomes.


Asunto(s)
Neoplasias Óseas/secundario , Estadificación de Neoplasias , Osteoblastos/patología , Neoplasias de la Próstata/patología , Neoplasias Óseas/diagnóstico , Diferenciación Celular , Progresión de la Enfermedad , Humanos , Masculino , Metástasis de la Neoplasia
3.
J Proteome Res ; 16(8): 2709-2728, 2017 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-28675788

RESUMEN

Osteoblasts communicate both with normal cells in the bone marrow and with tumor cells that metastasized to bone. Here we show that osteoblasts release exosomes, we termed osteosomes, which may be a novel mechanism by which osteoblasts communicate with cells in their environment. We have isolated exosomes from undifferentiated/proliferating (D0 osteosomes) and differentiated/mineralizing (D24 osteosomes) primary mouse calvarial osteoblasts. The D0 and D24 osteosomes were found to be vesicles of 130-140 nm by dynamic light scattering analysis. Proteomics profiling using tandem mass spectrometry (LC-MS/MS) identified 206 proteins in D0 osteosomes and 336 in D24 osteosomes. The proteins in osteosomes are mainly derived from the cytoplasm (∼47%) and plasma membrane (∼31%). About 69% of proteins in osteosomes are also found in Vesiclepedia, and these canonical exosomal proteins include tetraspanins and Rab family proteins. We found that there are differences in both protein content and levels in exosomes isolated from undifferentiated and differentiated osteoblasts. Among the proteins that are unique to osteosomes, 169 proteins are present in both D0 and D24 osteosomes, 37 are unique to D0, and 167 are unique to D24. Among those 169 proteins present in both D0 and D24 osteosomes, 10 proteins are likely present at higher levels in D24 than D0 osteosomes based on emPAI ratios of >5. These results suggest that osteosomes released from different cellular state of osteoblasts may mediate distinct functions. Using live-cell imaging, we measured the uptake of PKH26-labeled osteosomes into C4-2B4 and PC3-mm2 prostate cancer cells. In addition, we showed that cadherin-11, a cell adhesion molecule, plays a role in the uptake of osteosomes into PC3-mm2 cells as osteosome uptake was delayed by neutralizing antibody against cadherin-11. Together, our studies suggest that osteosomes could have a unique role in the bone microenvironment under both physiological and pathological conditions.


Asunto(s)
Calcificación Fisiológica , Proliferación Celular , Exosomas/química , Osteoblastos/patología , Neoplasias de la Próstata/patología , Proteínas/análisis , Animales , Cadherinas/fisiología , Comunicación Celular , Diferenciación Celular , Células Cultivadas , Microambiente Celular/fisiología , Exosomas/patología , Humanos , Masculino , Ratones , Osteoblastos/metabolismo , Neoplasias de la Próstata/metabolismo , Proteómica/métodos
4.
J Cell Sci ; 128(24): 4629-41, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26519476

RESUMEN

Cadherin-11 (Cad11) cell adhesion molecule plays a role in prostate cancer cell migration. Because disassembly of adhesion complexes through endocytosis of adhesion proteins has been shown to play a role in cell migration, we examined whether Cad11 endocytosis plays a role in Cad11-mediated migration. The mechanism by which Cad11 is internalized is unknown. Using a GST pulldown assay, we found that clathrin binds to the Cad11 cytoplasmic domain but not to that of E-cadherin. Using deletion analysis, we identified a unique sequence motif, VFEEE, in the Cad11 membrane proximal region (amino acid residues 11-15) that binds to clathrin. Endocytosis assays using K(+)-depletion buffer showed that Cad11 internalization is clathrin dependent. Proximity ligation assays showed that Cad11 colocalizes with clathrin, and immunofluorescence assays showed that Cad11 localizes in vesicles that stain for the early endosomal marker Rab5. Deletion of the VFEEE sequence from the Cad11 cytoplasmic domain (Cad11-cla-Δ5) leads to inhibition of Cad11 internalization and reduces Cad11-mediated cell migration in C4-2B and PC3-mm2 prostate cancer cells. These observations suggest that clathrin-mediated internalization of Cad11 regulates surface trafficking of Cad11 and that dynamic turnover of Cad11 regulates the migratory function of Cad11 in prostate cancer cells.


Asunto(s)
Cadherinas/metabolismo , Movimiento Celular , Clatrina/metabolismo , Endocitosis , Proteínas de Neoplasias/inmunología , Neoplasias de la Próstata/metabolismo , Cadherinas/genética , Línea Celular Tumoral , Clatrina/genética , Células HEK293 , Humanos , Masculino , Proteínas de Neoplasias/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Unión Proteica
5.
Mol Cell Proteomics ; 14(3): 471-83, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25527621

RESUMEN

A distinct feature of human prostate cancer (PCa) is the development of osteoblastic (bone-forming) bone metastases. Metastatic growth in the bone is supported by factors secreted by PCa cells that activate signaling networks in the tumor microenvironment that augment tumor growth. To better understand these signaling networks and identify potential targets for therapy of bone metastases, we characterized the secretome of a patient-derived xenograft, MDA-PCa-118b (PCa-118b), generated from osteoblastic bone lesion. PCa-118b induces osteoblastic tumors when implanted either in mouse femurs or subcutaneously. To study signaling molecules critical to these unique tumor/microenvironment-mediated events, we performed mass spectrometry on conditioned media of isolated PCa-118b tumor cells, and identified 26 secretory proteins, such as TGF-ß2, GDF15, FGF3, FGF19, CXCL1, galectins, and ß2-microglobulin, which represent both novel and previously published secreted proteins. RT-PCR using human versus mouse-specific primers showed that TGFß2, GDF15, FGF3, FGF19, and CXCL1 were secreted from PCa-118b cells. TGFß2, GDF15, FGF3, and FGF19 function as both autocrine and paracrine factors on tumor cells and stromal cells, that is, endothelial cells and osteoblasts. In contrast, CXCL1 functions as a paracrine factor through the CXCR2 receptor expressed on endothelial cells and osteoblasts. Thus, our study reveals a complex PCa bone metastasis secretome with paracrine and autocrine signaling functions that mediate cross-talk among multiple cell types within the tumor microenvironment.


Asunto(s)
Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , Proteínas de Neoplasias/metabolismo , Neoplasias de la Próstata/metabolismo , Proteómica/métodos , Microambiente Tumoral , Animales , Neoplasias Óseas/patología , Comunicación Celular , Línea Celular Tumoral , Medios de Cultivo Condicionados/metabolismo , Humanos , Masculino , Ratones , Ratones SCID , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Neoplasias de la Próstata/patología , Transducción de Señal , Células del Estroma/fisiología
6.
FASEB J ; 29(3): 1080-91, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25466890

RESUMEN

Loss of E-cadherin and up-regulation of mesenchymal cadherins, a hallmark of the epithelial-mesenchymal transition, contributes to migration and dissemination of cancer cells. Expression of human cadherin-11 (Cad11), also known as osteoblast cadherin, in prostate cancer increases the migration of prostate cancer cells. How Cad11 mediates cell migration is unknown. Using the human Cad11 cytoplasmic domain in pulldown assays, we identified human angiomotin (Amot), known to be involved in cell polarity, migration, and Hippo pathway, as a component of the Cad11 protein complex. Deletion analysis showed that the last C-terminal 10 amino acids in Cad11 cytoplasmic domain are required for Amot binding. Further, Cad11 preferentially interacts with Amot-p80 than Amot-p130 isoform and binds directly to the middle domain of Amot-p80. Cad11-Amot interaction affects Cad11-mediated cell migration, but not homophilic adhesion, as deletion of Amot binding motif of Cad11 (Cad11-ΔAmot) did not abolish Cad11-mediated cell-cell adhesion of mouse L cells, but significantly reduced Cad11-mediated cell migration of human C4-2B4 and PC3-mm2 prostate cancer cells and human HEK293T cells. Together, our studies identified Amot-p80 as a novel component of the Cad11 complex and demonstrated that Amot-p80 is critical for Cad11-mediated cell migration.


Asunto(s)
Cadherinas/metabolismo , Movimiento Celular , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Neoplasias de la Próstata/patología , beta Catenina/metabolismo , Proteína Activadora de GTPasa p120/metabolismo , Secuencia de Aminoácidos , Angiomotinas , Animales , Western Blotting , Cadherinas/genética , Adhesión Celular , Proliferación Celular , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Células HEK293 , Humanos , Técnicas para Inmunoenzimas , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Proteínas de la Membrana/genética , Ratones , Proteínas de Microfilamentos , Datos de Secuencia Molecular , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Isoformas de Proteínas , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , beta Catenina/genética , Proteína Activadora de GTPasa p120/genética
7.
J Immunol ; 192(1): 178-88, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24259505

RESUMEN

The differentiation of CD4(+) T cells into different Th lineages is driven by cytokine milieu in the priming site and the underlying transcriptional circuitry. Even though many positive regulators have been identified, it is not clear how this process is inhibited at transcriptional level. In this study, we report that the E-twenty six (ETS) transcription factor E74-like factor 4 (ELF4) suppresses the differentiation of Th17 cells both in vitro and in vivo. Culture of naive Elf4(-/-) CD4(+) T cells in the presence of IL-6 and TGF-ß (or IL-6, IL-23, and IL-1ß) resulted in increased numbers of IL-17A-positive cells compared with wild-type controls. In contrast, the differentiation to Th1, Th2, or regulatory T cells was largely unaffected by loss of ELF4. The increased expression of genes involved in Th17 differentiation observed in Elf4(-/-) CD4(+) T cells suggested that ELF4 controls their programming into the Th17 lineage rather than only IL-17A gene expression. Despite normal proliferation of naive CD4(+) T cells, loss of ELF4 lowered the requirement of IL-6 and TGF-ß signaling for IL-17A induction in each cell division. ELF4 did not inhibit Th17 differentiation by promoting IL-2 production as proposed for another ETS transcription factor, ETS1. Elf4(-/-) mice showed increased numbers of Th17 cells in the lamina propria at steady state, in lymph nodes after immunization, and, most importantly, in the CNS following experimental autoimmune encephalomyelitis induction, contributing to the increased disease severity. Collectively, our findings suggest that ELF4 restrains Th17 differentiation in dividing CD4(+) T cells by regulating commitment to the Th17 differentiation program.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular/genética , Proteínas de Unión al ADN/genética , Células Th17/citología , Células Th17/metabolismo , Factores de Transcripción/genética , Animales , Linfocitos T CD4-Positivos/inmunología , Línea Celular , Supervivencia Celular/genética , Citocinas/metabolismo , Humanos , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Células Th17/inmunología
8.
EMBO J ; 29(3): 517-31, 2010 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-20019668

RESUMEN

Lissencephaly is a devastating neurological disorder caused by defective neuronal migration. The LIS1 (or PAFAH1B1) gene was identified as the gene mutated in lissencephaly patients, and was found to regulate cytoplasmic dynein function and localization. In particular, LIS1 is essential for anterograde transport of cytoplasmic dynein as a part of the cytoplasmic dynein-LIS1-microtubule complex in a kinesin-1-dependent manner. However, the underlying mechanism by which a cytoplasmic dynein-LIS1-microtubule complex binds kinesin-1 is unknown. Here, we report that mNUDC (mammalian NUDC) interacts with kinesin-1 and is required for the anterograde transport of a cytoplasmic dynein complex by kinesin-1. mNUDC is also required for anterograde transport of a dynactin-containing complex. Inhibition of mNUDC severely suppressed anterograde transport of distinct cytoplasmic dynein and dynactin complexes, whereas motility of kinesin-1 remained intact. Reconstruction experiments clearly demonstrated that mNUDC mediates the interaction of the dynein or dynactin complex with kinesin-1 and supports their transport by kinesin-1. Our findings have uncovered an essential role of mNUDC for anterograde transport of dynein and dynactin by kinesin-1.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Dineínas Citoplasmáticas/metabolismo , Cinesinas/fisiología , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Nucleares/fisiología , Animales , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Complejo Dinactina , Ganglios Espinales/metabolismo , Cinesinas/metabolismo , Ratones , Modelos Biológicos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Unión Proteica/efectos de los fármacos , ARN Interferente Pequeño/farmacología , Porcinos
9.
FASEB J ; 26(10): 4057-67, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22751009

RESUMEN

Protein acetylation has been implicated in playing an important role during mitotic progression. Aurora B kinase is known to play a critical role in mitosis. However, whether Aurora B is regulated by acetylation is not known. Using IP with an anti-acetyl lysine antibody, we identified Aurora B as an acetylated protein in PC3 prostate cancer cells. Knockdown of HDAC3 or inhibiting HDAC3 deacetylase activity led to a significant increase (P<0.01 and P<0.05, respectively) in Aurora B acetylation as compared to siLuc or vehicle-treated controls. Increased Aurora B acetylation is correlated with a 30% reduction in Aurora B kinase activity in vitro and resulted in significant defects in Aurora B-dependent mitotic processes, including kinetochore-microtubule attachment and chromosome congression. Furthermore, Aurora B transiently interacts with HDAC3 at the kinetochore-microtubule interface of congressing chromosomes during prometaphase. This window of interaction corresponded with a transient but significant reduction (P=0.02) in Aurora B acetylation during early mitosis. Together, these results indicate that Aurora B is more active in its deacetylated state and further suggest a new mechanism by which dynamic acetylation/deacetylation acts as a rheostat to fine-tune Aurora B activity during mitotic progression.


Asunto(s)
Mitosis/fisiología , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Acetilación , Aurora Quinasa B , Aurora Quinasas , Línea Celular Tumoral , Histona Desacetilasas/metabolismo , Humanos , Immunoblotting , Cinetocoros/metabolismo , Masculino , Microscopía Fluorescente , Microtúbulos/metabolismo , Mitosis/genética , Procesamiento Proteico-Postraduccional
10.
iScience ; 26(2): 105994, 2023 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-36798441

RESUMEN

Metastatic prostate cancer (PCa) in bone induces bone-forming lesions. We have previously shown that PCa-induced bone originates from endothelial cells (ECs) that have undergone EC-to-osteoblast (OSB) transition. Here, we investigated whether EC-to-OSB transition also occurs during normal bone formation. We developed an EC and OSB dual-color reporter mouse (DRM) model that marks EC-OSB hybrid cells with red and green fluorescent proteins. We observed EC-to-OSB transition (RFP and GFP co-expression) in both endochondral and intramembranous bone formation during embryonic development and in adults. Co-expression was confirmed in cells isolated from DRM. Bone marrow- and lung-derived ECs underwent transition to OSBs and mineralization in osteogenic medium. RNA-sequencing revealed GATA family transcription factors were upregulated in EC-OSB hybrid cells and knockdown of GATA3 inhibited BMP4-induced mineralization. Our findings support that EC-to-OSB transition occurs during normal bone development and suggest a new paradigm regarding the endothelial origin of OSBs.

11.
Prostate ; 72(3): 291-300, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21681775

RESUMEN

BACKGROUND: Katanin p60 is a microtubule-severing protein and is involved in microtubule cytoskeleton organization in both mitotic and non-mitotic processes. Its role in cancer metastasis is unknown. METHODS: Differential protein profiles of bone marrow aspirates were analyzed by chromatography, electrophoresis, and mass spectrometry. Expression of katanin p60 in primary and metastatic prostate cancer was examined by immunohistochemistry. Cellular function of katanin p60 was further examined in prostate cell lines. RESULTS: In a proteomic profiling of bone marrow aspirates from men with prostate cancer, we found that katanin p60 was one of the proteins differentially expressed in bone metastasis samples. Immunohistochemical staining showed that katanin p60 was expressed in the basal cells in normal human prostate glands. In prostatic adenocarcinomas, in which the basal cells were absent, katanin p60 was expressed in the prostate cancer cells. In the specimens from bone metastasis, katanin p60 was detectable in the metastatic cancer cells. Strikingly, some of the metastatic cancer cells also co-expressed basal cell biomarkers including the tumor suppressor p53-homologous protein p63 and the high molecular weight cytokeratins, suggesting that the metastatic prostate cancer cells may have a basal cell-like phenotype. Moreover, overexpression of katanin p60 inhibited prostate cancer cell proliferation but enhanced cell migration activity. CONCLUSIONS: Katanin p60 was aberrantly expressed during prostate cancer progression. Its expression in the metastatic cells in bone was associated with the re-emergence of a basal cell-like phenotype. The elevated katanin p60 expression may contribute to cancer cell metastasis via a stimulatory effect on cell motility.


Asunto(s)
Adenocarcinoma/metabolismo , Adenocarcinoma/secundario , Adenosina Trifosfatasas/metabolismo , Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , Neoplasias de la Próstata/metabolismo , Adenocarcinoma/fisiopatología , Biomarcadores de Tumor/metabolismo , Biopsia , Médula Ósea/metabolismo , Médula Ósea/patología , Médula Ósea/fisiopatología , Neoplasias Óseas/fisiopatología , Movimiento Celular/fisiología , Proliferación Celular , Humanos , Katanina , Masculino , Persona de Mediana Edad , Próstata/metabolismo , Próstata/patología , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/fisiopatología , Estudios Retrospectivos , Regulación hacia Arriba
12.
Cancer Res ; 82(17): 3158-3171, 2022 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-35802768

RESUMEN

Metastatic prostate cancer in the bone induces bone-forming lesions that contribute to progression and therapy resistance. Prostate cancer-induced bone formation originates from endothelial cells (EC) that have undergone endothelial-to-osteoblast (EC-to-OSB) transition in response to tumor-secreted BMP4. Current strategies targeting prostate cancer-induced bone formation are lacking. Here, we show that activation of retinoic acid receptor (RAR) inhibits EC-to-OSB transition and reduces prostate cancer-induced bone formation. Treatment with palovarotene, an RARγ agonist being tested for heterotopic ossification in fibrodysplasia ossificans progressiva, inhibited EC-to-OSB transition and osteoblast mineralization in vitro and decreased tumor-induced bone formation and tumor growth in several osteogenic prostate cancer models, and similar effects were observed with the pan-RAR agonist all-trans-retinoic acid (ATRA). Knockdown of RARα, ß, or γ isoforms in ECs blocked BMP4-induced EC-to-OSB transition and osteoblast mineralization, indicating a role for all three isoforms in prostate cancer-induced bone formation. Furthermore, treatment with palovarotene or ATRA reduced plasma Tenascin C, a factor secreted from EC-OSB cells, which may be used to monitor treatment response. Mechanistically, BMP4-activated pSmad1 formed a complex with RAR in the nucleus of ECs to activate EC-to-OSB transition. RAR activation by palovarotene or ATRA caused pSmad1 degradation by recruiting the E3-ubiquitin ligase Smad ubiquitination regulatory factor1 (Smurf1) to the nuclear pSmad1/RARγ complex, thus blocking EC-to-OSB transition. Collectively, these findings suggest that palovarotene can be repurposed to target prostate cancer-induced bone formation to improve clinical outcomes for patients with bone metastasis. SIGNIFICANCE: This study provides mechanistic insights into how RAR agonists suppress prostate cancer-induced bone formation and offers a rationale for developing RAR agonists for prostate cancer bone metastasis therapy. See related commentary by Bhowmick and Bhowmick, p. 2975.


Asunto(s)
Neoplasias Óseas , Neoplasias de la Próstata , Neoplasias Óseas/metabolismo , Células Endoteliales/patología , Humanos , Masculino , Osteoblastos/metabolismo , Neoplasias de la Próstata/patología , Receptores de Ácido Retinoico/metabolismo , Tretinoina/metabolismo , Tretinoina/farmacología , Ubiquitina-Proteína Ligasas/metabolismo
13.
Oncogene ; 41(6): 757-769, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34845375

RESUMEN

Metastatic prostate cancer (PCa) in bone induces bone-forming lesions that enhance PCa progression. How tumor-induced bone formation enhances PCa progression is not known. We have previously shown that PCa-induced bone originates from endothelial cells (ECs) that have undergone endothelial-to-osteoblast (EC-to-OSB) transition by tumor-secreted bone morphogenetic protein 4 (BMP4). Here, we show that EC-to-OSB transition leads to changes in the tumor microenvironment that increases the metastatic potential of PCa cells. We found that conditioned medium (CM) from EC-OSB hybrid cells increases the migration, invasion, and survival of PC3-mm2 and C4-2B4 PCa cells. Quantitative mass spectrometry (Isobaric Tags for Relative and Absolute Quantitation) identified Tenascin C (TNC) as one of the major proteins secreted from EC-OSB hybrid cells. TNC expression in tumor-induced OSBs was confirmed by immunohistochemistry of MDA PCa-118b xenograft and human bone metastasis specimens. Mechanistically, BMP4 increases TNC expression in EC-OSB cells through the Smad1-Notch/Hey1 pathway. How TNC promotes PCa metastasis was next interrogated by in vitro and in vivo studies. In vitro studies showed that a TNC-neutralizing antibody inhibits EC-OSB-CM-mediated PCa cell migration and survival. TNC knockdown decreased, while the addition of recombinant TNC or TNC overexpression increased migration and anchorage-independent growth of PC3 or C4-2b cells. When injected orthotopically, PC3-mm2-shTNC clones decreased metastasis to bone, while C4-2b-TNC-overexpressing cells increased metastasis to lymph nodes. TNC enhances PCa cell migration through α5ß1 integrin-mediated YAP/TAZ inhibition. These studies elucidate that tumor-induced stromal reprogramming generates TNC that enhances PCa metastasis and suggest that TNC may be a target for PCa therapy.


Asunto(s)
Tenascina
14.
J Pathol ; 221(1): 68-76, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20191612

RESUMEN

Men with castration-resistant prostate cancer (PCa) frequently develop metastasis in bone. The reason for this association is unclear. We have previously shown that cadherin-11 (also known as OB-cadherin), a homophilic cell adhesion molecule that mediates osteoblast adhesion, plays a role in the metastasis of PCa to bone. Here, we report that androgen-deprivation therapy up-regulates cadherin-11 expression in PCa. In human PCa specimens, immunohistochemical staining showed that 22/26 (85%) primary PCa tumours from men with castration-resistant PCa expressed cadherin-11. In contrast, only 7/50 (14%) androgen-dependent PCa tumours expressed cadherin-11. In the MDA-PCa-2b xenograft animal model, cadherin-11 was expressed in the recurrent tumours following castration. In the PCa cell lines, there is an inverse correlation between expression of cadherin-11 and androgen receptor (AR), and cadherin-11 is expressed in very low levels or not expressed in AR-positive cell lines, including LNCaP, C4-2B4 and VCaP cells. We showed that AR likely regulates cadherin-11 expression in PCa through an indirect mechanism. Although re-expression of AR in the AR-negative PC3 cells led to the inhibition of cadherin-11 expression, depletion of androgen from the culture medium or down-regulation of AR by RNA interference in the C4-2B4 cells or VCaP cells only produced a modest increase of cadherin-11 expression. Promoter analysis indicated that cadherin-11 promoter does not contain a typical AR-binding element, and AR elicits a modest inhibition of cadherin-11 promoter activity, suggesting that AR does not regulate cadherin-11 expression directly. Together, these results suggest that androgen deprivation up-regulates cadherin-11 expression in prostate cancer, and this may contribute to the metastasis of PCa to bone. Our study suggests that therapeutic strategies that block cadherin-11 expression or function may be considered when applying androgen-ablation therapy.


Asunto(s)
Andrógenos/deficiencia , Cadherinas/biosíntesis , Proteínas de Neoplasias/biosíntesis , Neoplasias de la Próstata/metabolismo , Animales , Cadherinas/genética , Regulación Enzimológica de la Expresión Génica/fisiología , Humanos , Masculino , Ratones , Ratones Desnudos , Proteínas de Neoplasias/genética , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/cirugía , Orquiectomía , Regiones Promotoras Genéticas , Neoplasias de la Próstata/genética , Receptores Androgénicos/metabolismo , Células Tumorales Cultivadas , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Proc Natl Acad Sci U S A ; 105(11): 4179-84, 2008 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-18326024

RESUMEN

Protein acetylation and deacetylation play key roles in multiple physiological functions. Histone deacetylase 3 (HDAC3) is a highly conserved, ubiquitously expressed protein that forms multiprotein corepressor complexes to repress gene transcription. Recent studies show that HDAC3 may play a role in cell proliferation. Altered HDAC3 level increases G(2)/M cells, but the mechanism remains unknown. Here we show for the first time, to our knowledge, that the HDAC3 complex, including nuclear receptor corepressor (N-CoR), transducin-beta-like protein 1 (TBL1), and TBL1-related protein 1 (TBLR1), is localized on the mitotic spindle. Knockdown of HDAC3 or N-CoR resulted in a collapsed mitotic spindle that was surrounded by chromosomes arranged in a dome-like configuration. Treatment of mitotic cells with Trichostatin A, an HDAC inhibitor, resulted in similar spindle defects independent of transcriptional regulation. In addition, wild-type HDAC3 but not a deacetylase-dead mutant HDAC3 rescued the phenotypes of HDAC3-depleted cells, suggesting that the enzymatic activity of HDAC3 is important for proper spindle function. Whereas the kinetochores and the spindle assembly checkpoint appeared intact in HDAC3-deficient cells, kinetochore-microtubule attachments were impaired because spindle microtubules were unstable in response to cold treatment. These data suggest that the HDAC3 complex is involved in the formation of functional mitotic spindles and proper kinetochore-microtubule attachment. The level or distribution of acetylated alpha-tubulin was not altered in HDAC3-deficient cells. Taken together, our studies raise the interesting possibility that acetylation-deacetylation of mitotic spindle components may be essential for mitotic spindle function.


Asunto(s)
Histona Desacetilasas/metabolismo , Cinetocoros/enzimología , Microtúbulos/enzimología , Huso Acromático/enzimología , Acetilación , Cromosomas/genética , Células HeLa , Humanos , Proteínas Nucleares/metabolismo , Co-Represor 1 de Receptor Nuclear , Unión Proteica , Proteínas Represoras/metabolismo , Tubulina (Proteína)/metabolismo
16.
Oncogene ; 40(27): 4592-4603, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34127814

RESUMEN

A fraction of patients undergoing androgen deprivation therapy (ADT) for advanced prostate cancer (PCa) will develop recurrent castrate-resistant PCa (CRPC) in bone. Strategies to prevent CRPC relapse in bone are lacking. Here we show that the cholesterol-lowering drugs statins decrease castration-induced bone marrow adiposity in the tumor microenvironment and reduce PCa progression in bone. Using primary bone marrow stromal cells (BMSC) and M2-10B4 cells, we showed that ADT increases bone marrow adiposity by enhancing BMSC-to-adipocyte transition in vitro. Knockdown of androgen receptor abrogated BMSC-to-adipocyte transition, suggesting an androgen receptor-dependent event. RNAseq analysis showed that androgens reduce the secretion of adipocyte hormones/cytokines including leptin during BMSC-to-adipocyte transition. Treatment of PCa C4-2b, C4-2B4, and PC3 cells with leptin led to an increase in cell cycle progression and nuclear Stat3. RNAseq analysis also showed that androgens inhibit cholesterol biosynthesis pathway, raising the possibility that inhibiting cholesterol biosynthesis may decrease BMSC-to-adipocyte transition. Indeed, statins decreased BMSC-to-adipocyte transition in vitro and castration-induced bone marrow adiposity in vivo. Statin pre-treatment reduced 22RV1 PCa progression in bone after ADT. Our findings with statin may provide one of the mechanisms to the clinical correlations that statin use in patients undergoing ADT seems to delay progression to "lethal" PCa.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas , Adiposidad , Humanos , Masculino , Neoplasias de la Próstata
17.
iScience ; 24(4): 102388, 2021 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-33981975

RESUMEN

Cell type transition occurs during normal development and under pathological conditions. In prostate cancer bone metastasis, prostate cancer-secreted BMP4 induces endothelial cell-to-osteoblast (EC-to-OSB) transition. Such tumor-induced stromal reprogramming supports prostate cancer progression. We delineate signaling pathways mediating EC-to-OSB transition using EC lines 2H11 and SVR. We found that BMP4-activated pSmad1-Notch-Hey1 pathway inhibits EC migration and tube formation. BMP4-activated GSK3ß-ßcatenin-Slug pathway stimulates Osx expression. In addition, pSmad1-regulated Dlx2 converges with the Smad1 and ß-catenin pathways to stimulate osteocalcin expression. By co-expressing Osx, Dlx2, Slug and Hey1, we were able to achieve EC-to-OSB transition, leading to bone matrix mineralization in the absence of BMP4. In human prostate cancer bone metastasis specimens and MDA-PCa-118b and C4-2b-BMP4 osteogenic xenografts, immunohistochemical analysis showed that ß-catenin and pSmad1 are detected in activated osteoblasts rimming the tumor-induced bone. Our results elucidated the pathways and key molecules coordinating prostate cancer-induced stromal programming and provide potential targets for therapeutic intervention.

18.
J Proteome Res ; 9(9): 4554-64, 2010 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-20812760

RESUMEN

Mitosis is a highly regulated process in which errors can lead to genomic instability, a hallmark of cancer. During this phase of the cell cycle, transcription is silent and RNA translation is inhibited. Thus, mitosis is largely driven by post-translational modification of proteins, including phosphorylation, methylation, ubiquitination, and sumoylation. Here, we show that protein acetylation is prevalent during mitosis. To identify proteins that are acetylated, we synchronized HeLa cells in early prometaphase and immunoprecipitated lysine-acetylated proteins with antiacetyl-lysine antibody. The immunoprecipitated proteins were identified by LC-ESI-MS/MS analysis. These include proteins involved in RNA translation, RNA processing, cell cycle regulation, transcription, chaperone function, DNA damage repair, metabolism, immune response, and cell structure. Immunoprecipitation followed by Western blot analyses confirmed that two RNA processing proteins, eIF4G and RNA helicase A, and several cell cycle proteins, including APC1, anillin, and NudC, were acetylated in mitosis. We further showed that acetylation of APC1 and NudC was enhanced by apicidin treatment, suggesting that their acetylation was regulated by histone deacetylase. Moreover, treating mitotic cells with apicidin or trichostatin A induced spindle abnormalities and cytokinesis failure. These studies suggest that protein acetylation/deacetylation is likely an important regulatory mechanism in mitosis.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Mitosis/fisiología , Procesamiento Postranscripcional del ARN , Acetilación , Proteínas de Ciclo Celular/química , Citocinesis/efectos de los fármacos , ADN/química , ADN/metabolismo , Células HeLa , Humanos , Ácidos Hidroxámicos/farmacología , Immunoblotting , Espectrometría de Masas , Microscopía Fluorescente , Mitosis/efectos de los fármacos , Péptidos Cíclicos/farmacología , Proteómica , Fracciones Subcelulares/química , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo
19.
Curr Biol ; 16(14): 1414-21, 2006 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-16860740

RESUMEN

The equal distribution of chromosomes during mitosis is critical for maintaining the integrity of the genome. Essential to this process are the capture of spindle microtubules by kinetochores and the congression of chromosomes to the metaphase plate . Polo-like kinase 1 (Plk1) is a mitotic kinase that has been implicated in microtubule-kinetochore attachment, tension generation at kinetochores, tension-responsive signal transduction, and chromosome congression . The tension-sensitive substrates of Plk1 at the kinetochore are unknown. Here, we demonstrate that human Nuclear distribution protein C (NudC), a 42 kDa protein initially identified in Aspergillus nidulans and shown to be phosphorylated by Plk1 , plays a significant role in regulating kinetochore function. Plk1-phosphorylated NudC colocalizes with Plk1 at the outer plate of the kinetochore. Depletion of NudC reduced end-on microtubule attachments at kinetochores and resulted in defects in chromosome congression at the metaphase plate. Importantly, NudC-deficient cells exhibited mislocalization of Plk1 and the Kinesin-7 motor CENP-E from prometaphase kinetochores. Ectopic expression of wild-type NudC, but not NudC containing mutations in the Plk1 phosphorylation sites, recovered Plk1 localization at the kinetochore and rescued chromosome congression. Thus, NudC functions as both a substrate and a spatial regulator of Plk1 at the kinetochore to promote chromosome congression.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiología , Cinetocoros/metabolismo , Metafase/genética , Proteínas Nucleares/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas de Ciclo Celular/análisis , Proteínas de Ciclo Celular/genética , Proteínas Cromosómicas no Histona/metabolismo , Posicionamiento de Cromosoma , Humanos , Cinetocoros/ultraestructura , Microtúbulos/metabolismo , Modelos Biológicos , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/análisis , Proteínas Serina-Treonina Quinasas/genética , Transporte de Proteínas , Proteínas Proto-Oncogénicas/análisis , Proteínas Proto-Oncogénicas/genética , Interferencia de ARN , Quinasa Tipo Polo 1
20.
Dev Cell ; 5(1): 127-38, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12852857

RESUMEN

Polo-like kinase 1 (Plk1) plays essential roles at multiple events during cell division, yet little is known about its physiological substrates. In a cDNA phage display screen using Plk1 C-terminal affinity columns, we identified NudC (nuclear distribution gene C) as a Plk1 binding protein. Here, we characterize the interaction between Plk1 and NudC, show that Plk1 phosphorylates NudC at conserved S274 and S326 residues in vitro, and present evidence that NudC is also a substrate for Plk1 in vivo. Downregulation of NudC by RNA interference results in multiple mitotic defects, including multinucleation and cells arrested at the midbody stage, which are rescued by ectopic expression of wild-type NudC, but not by NudC with mutations in the Plk1 phosphorylation sites. These results suggest that Plk1 phosphorylation of NudC may influence cytokinesis.


Asunto(s)
División Celular/fisiología , Proteínas Quinasas/metabolismo , Proteínas Quinasas/fisiología , Proteínas/metabolismo , Animales , Células COS , Proteínas de Ciclo Celular , Chlorocebus aethiops , Regulación de la Expresión Génica , Glutatión Transferasa/metabolismo , Proteínas Fluorescentes Verdes , Células HeLa , Humanos , Proteínas Luminiscentes/metabolismo , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Proteínas Nucleares , Fosforilación , Mutación Puntual , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas , ARN Interferente Pequeño/metabolismo , Quinasa Tipo Polo 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA