Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 299(1): 102791, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36509142

RESUMEN

Hypoxia-inducible factor 1α (HIF1α) is a transcription factor that regulates angiogenesis under hypoxic conditions. To investigate the posttranscriptional regulatory mechanism of HIF1α, we performed a cell-based screening to reveal potential cis-elements and the regulatory RNA-binding proteins that act as trans-factors. We found that LIN28A promoted HIF1α protein expression independently of the downregulation of microRNA let-7, which is also directly mediated by LIN28A. Transcriptome analysis and evaluation of RNA stability using RNA-seq and SLAM-seq analyses, respectively, revealed that LIN28A upregulates HIF1A expression via mRNA stabilization. To investigate the physical association of LIN28A with HIF1A mRNA, we performed enhanced crosslinking immunoprecipitation in 293FT cells and integrally analyzed the transcriptome. We observed that LIN28A associates with HIF1A mRNA via its cis-element motif "UGAU". The "UGAU" motifs are recognized by the cold shock domain of LIN28A, and the introduction of a loss-of-function mutation to the cold shock domain diminished the upregulatory activities performed by LIN28A. Finally, the microvessel density assay showed that the expression of LIN28A promoted angiogenesis in vivo. In conclusion, our study elucidated the role of LIN28A in enhancing the HIF1α axis at the posttranscription layer.


Asunto(s)
Regulación de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia , Estabilidad del ARN , Proteínas de Unión al ARN , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo , Regulación hacia Arriba
2.
Biochem Biophys Res Commun ; 704: 149636, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38402724

RESUMEN

Osteoclasts are hematopoietic cells attached to the bones containing type I collagen-deposited hydroxyapatite during bone resorption. Two major elements determine the stiffness of bones: regular calcified bone (bone that is resorbable by osteoclasts) and un-calcified osteoid bone (bone that is un-resorbable by osteoclasts). The osteolytic cytokine RANKL promotes osteoclast differentiation; however, the roles of the physical interactions of osteoclasts with calcified and un-calcified bone at the sealing zones and the subsequent cellular signaling remain unclear. In this study, we investigated podosomes, actin-rich adhesion structures (actin-ring) in the sealing zone that participates in sensing hard stiffness with collagen in the physical environment during osteoclast differentiation. RANKL-induced osteoclast differentiation induction was promoted when Raw264.7 cells were cultured on collagen-coated plastic dishes but not on non-coated plastic dishes, which was associated with the increased expression of podosome-related genes and Src. In contrast, when cells were cultured on collagen gel, expression of podosome-related genes and Src were not upregulated. The induction of podosome-related genes and Src requires hard stiffness with RGD-containing substratum and integrin-mediated F-actin polymerization. These results indicate that osteoclasts sense both the RGD sequence and stiffness of calcified collagen through their podosome components regulating osteoclast differentiation via the c-Src pathway.


Asunto(s)
Resorción Ósea , Podosomas , Humanos , Osteoclastos/metabolismo , Podosomas/metabolismo , Actinas/metabolismo , Diferenciación Celular/fisiología , Resorción Ósea/metabolismo , Proteína Tirosina Quinasa CSK/metabolismo , Colágeno/metabolismo , Oligopéptidos/metabolismo
3.
J Biol Chem ; 298(3): 101603, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35101442

RESUMEN

Toll-like receptors (TLRs) are pattern recognition receptors that play a critical role in innate immune diseases. TLR3, which is localized in the endosomal compartments of hematopoietic immune cells, is able to recognize double-stranded RNA (dsRNA) derived from viruses and bacteria and thereby induce innate immune responses. Inflammatory periodontal bone resorption is caused by bacterial infections, which initially is regulated by innate immunity; however, the roles of TLR3 signaling in bone resorption are still not known. We examined the roles of TLR3 signaling in bone resorption using poly(I:C), a synthetic dsRNA analog. In cocultures of mouse bone marrow cells and stromal osteoblasts, poly(I:C) clearly induced osteoclast differentiation. In osteoblasts, poly(I:C) increased PGE2 production and upregulated the mRNA expression of PGE2-related genes, Ptgs2 and Ptges, as well as that of a gene related to osteoclast differentiation, Tnfsf11. In addition, we found that indomethacin (a COX-2 inhibitor) or an antagonist of the PGE2 receptor EP4 attenuated the poly(I:C)-induced PGE2 production and subsequent Tnfsf11 expression. Poly(I:C) also prolonged the survival of the mature osteoclasts associated with the increased mRNA expression of osteoclast marker genes, Nfatc1 and Ctsk. In ex vivo organ cultures of periodontal alveolar bone, poly(I:C) induced bone-resorbing activity in a dose-dependent manner, which was attenuated by the simultaneous administration of either indomethacin or an EP4 antagonist. These data suggest that TLR3 signaling in osteoblasts controls PGE2 production and induces the subsequent differentiation and survival of mature osteoclasts. Endogenous TLR3 in stromal osteoblasts and osteoclasts synergistically induces inflammatory alveolar bone resorption in periodontitis.


Asunto(s)
Resorción Ósea , Dinoprostona , Osteoblastos , Receptor Toll-Like 3 , Animales , Resorción Ósea/genética , Resorción Ósea/metabolismo , Células Cultivadas , Dinoprostona/biosíntesis , Dinoprostona/genética , Dinoprostona/metabolismo , Endosomas/metabolismo , Indometacina/farmacología , Ratones , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Prostaglandinas E/efectos adversos , Prostaglandinas E/metabolismo , Ligando RANK/metabolismo , ARN Mensajero/metabolismo , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/metabolismo
4.
Biochem Biophys Res Commun ; 644: 40-48, 2023 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-36623397

RESUMEN

Prostate cancer highly metastasizes to bone, and such cancer is associated with severe bone resorption and bone formation at the site of metastasis. Prostaglandin E2 (PGE2) promotes bone resorption in inflammatory diseases; however, the roles in prostate cancer-induced bone formation are still unclear. In the present study, we investigated the effects of membrane-bound TGF-α on prostate cancer-induced bone formation through autocrine PGE2 signaling in osteoblasts. In the prostate cancer explant experiment into tibiae, injected prostate cancer cells induced bone formation with the increased expression of osteogenic genes, such as Runx2 and Wnt5a, and prostaglandin synthase Ptgs2. In osteoblasts, PGE2 increased the number of calcified bone nodules with enhanced expression of Runx2 and Wnt5a. We also screened the factors involved in cancer progression, and 11 EGF family members were found to be expressed in the human prostate cancer cell line PC3. PC3 highly expressed amphiregulin, HB-EGF, and especially TGF-α. Treatment with recombinant TGF-α increased the Ptgs2 expression and PGE2 production in osteoblasts, which promoted the formation of calcified bone nodules, suggesting that the interaction between PC3 and osteoblasts promoted PGE2 production. In co-culture of osteoblasts and fixed PC3 cells, the phosphorylation of EGFR and ERK and subsequent Ptgs2 expression and PGE2 production were increased, an effect that was attenuated by treatment with inhibitors of EGFR and ERK. These results indicate that membrane-bound TGF-α enhances ERK signaling while also inducing PGE2-mediated bone formation in osteoblasts, thus suggesting that prostate cancer regulates both PGE2-mediated bone resorption and bone formation at the site of bone metastasis of prostate cancer.


Asunto(s)
Resorción Ósea , Neoplasias de la Próstata , Humanos , Masculino , Resorción Ósea/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Ciclooxigenasa 2/metabolismo , Receptores ErbB/metabolismo , Osteoblastos/metabolismo , Osteogénesis , Prostaglandinas/metabolismo , Neoplasias de la Próstata/patología , Factor de Crecimiento Transformador alfa/metabolismo , Dinoprostona/metabolismo
5.
Int J Mol Sci ; 25(1)2023 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-38203473

RESUMEN

Duchenne muscular dystrophy (DMD) is the most common type of neuromuscular disease caused by mutations in the DMD gene encoding dystrophin protein. To quantitively assess human dystrophin protein in muscle biopsy samples, it is imperative to consistently detect as low as 0.003% of the dystrophin protein relative to the total muscle protein content. The quantitation of dystrophin protein has traditionally been conducted using semiquantitative immunoblotting or immunohistochemistry; however, there is a growing need to establish a more precise quantitative method by employing liquid chromatography-mass spectrometry (LC-MS) to measure dystrophin protein. In this study, a novel quantification method was established using a mouse experiment platform applied to the clinical quantification of human dystrophin protein. The method using a spike-in approach with a triple quadrupole LC-MS quantitated the amount of dystrophin in wild-type and human DMD transgenic mice but not in DMD-null mice. In conclusion, we established a quantitating method of dystrophin using HPLC-LC-MS with a novel spike-in approach. These results indicate that our methodology could be applied to several LC-MS devices to enable the accurate measurement of dystrophin protein in patients with DMD.


Asunto(s)
Distrofina , Distrofia Muscular de Duchenne , Animales , Ratones , Humanos , Distrofina/genética , Cromatografía Líquida con Espectrometría de Masas , Músculo Esquelético , Proteínas Musculares , Ratones Noqueados , Ratones Transgénicos
6.
Am J Physiol Cell Physiol ; 323(4): C1137-C1148, 2022 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-35993515

RESUMEN

Caveolins, encoded by the Cav gene family, are the main components of caveolae. Caveolin-3 (Cav3) is specifically expressed in muscle cells. Mutations in Cav3 are responsible for a group of muscle diseases called caveolinopathies, and Cav3 deficiency is associated with sarcolemmal membrane alterations, disorganization of T-tubules, and disruption of specific cell-signaling pathways. However, Cav3 overexpression increases the number of sarcolemmal caveolae and muscular dystrophy-like regenerating muscle fibers with central nuclei, suggesting that the alteration of Cav3 expression levels or localization influences muscle cell functions. Here, we used mouse C2C12 myoblasts in which Cav3 expression was suppressed with short hairpin RNA and found that Cav3 suppression impaired myotube differentiation without affecting the expression of MyoD and Myog. We also observed an increase of intracellular Ca2+ levels, total calpain activity, and Ca2+-dependent calmodulin kinase II (CaMKII) levels in Cav3-depleted myoblasts. Importantly, those phenotypes due to Cav3 suppression were caused by the ryanodine receptor activation. Furthermore, pharmacological inhibition of CaMKII rescued the impairment of myoblast differentiation due to Cav3 knockdown. Our results suggest that Cav3 regulates intracellular Ca2+ concentrations by modulating ryanodine receptor activity in muscle cells and that CaMKII suppression in muscle could be a novel therapy for caveolinopathies.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Caveolina 3 , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Calpaína/genética , Calpaína/metabolismo , Caveolina 3/genética , Caveolina 3/metabolismo , Caveolinas/metabolismo , Ratones , Músculo Esquelético/metabolismo , ARN Interferente Pequeño/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo
7.
BMC Cancer ; 20(1): 802, 2020 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-32831047

RESUMEN

BACKGROUND: Clinical trials have been conducted to clarify the beneficial effects of VD3 (1α,25-dihydroxy vitamin D3, also known as calcitriol) treatment in prostate cancer. However, the molecular mechanisms underlying these effects are not fully understood. Recent studies on IGFBP-3 have indicated its intracellular functions in cell growth and apoptosis. The aim of this study was to confirm the benefits of low-dose VD3 treatment and clarify the molecular mechanisms underlying these beneficial effects in prostate cancer cells. METHODS: The molecular effects of simultaneous treatment of LNCaP cells and their genetically modified cell lines with low concentration of docetaxel and VD3 were biologically and biochemically analyzed. To further determine the effects of VD3 treatment on IGFBP-3 induction system, cells were temporarily treated with VD3 in combination with a transcriptional inhibitor or protein synthesis inhibitor. Bcl-2 protein and its mRNA behavior were also observed in Igfbp-3 expression-modified LNCaP cells to determine the involvement of IGFBP-3 in the suppression of Bcl-2 by VD3 treatment. RESULTS: Changes in IGFBP-3 expression levels in LNCaP cells indicated that it mediated the inhibition of cell growth induced by VD3 treatment. IGFBP-3 was also found to be a mediator of the enhanced cytotoxicity of prostate cancer cells to VD3 in combination with the anti-cancer drug. We further identified the distinct property of the IGFBP-3 induction system, wherein temporal VD3 stimulation-induced prolonged IGFBP-3 expression and VD3 treatment-induced increase in IGFBP-3 expression were optimized based on the protein concentration rather than the mRNA concentration. Meanwhile, Bcl-2 expression was down-regulated by VD3 treatment in an IGFBP-3-independent manner. CONCLUSION: These findings indicate the molecular mechanisms of IGFBP-3 induction stimulated by VD3 and IGFBP-3 independent Bcl-2 suppression by VD3 treatment in prostate cancer cells. The results could prompt a re-evaluation of VD3 usage in therapy for patients with prostate cancer.


Asunto(s)
Calcitriol/farmacología , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Calcitriol/uso terapéutico , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Próstata/patología , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo
8.
Anal Chem ; 91(16): 10557-10563, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31322341

RESUMEN

Intercellular adhesion strengths between two kinds of murine breast cancer cells with different malignancies were measured quantitatively using a metal cup-attached chip with atomic force microscopy (AFM). The cup-attached chip was used to approach a cell, pick it up, and then approach another cell, and the adhesion strengths were measured according to the contact time of the cells between 0 to 60 s. Separation work was used as a parameter for quantitative comparisons of the strengths. As a result, the work of a highly metastatic cancer cell (FP10SC2) was greater than a low metastatic cancer cell (4T1-LM) throughout all contact times examined. Adhesion was analyzed from a point of a view of binding kinetics of receptors on cells, and two possibilities were found: one was the number of cell adhesive receptors increased, and the other was the work to separate single molecular binding increased with increasing cancer cell malignancy. These results indicated quantitative measurements of intercellular adhesion strengths using AFM yielded information to understand the mechanism of the cancer progression from a new perspective.


Asunto(s)
Neoplasias de la Mama/química , Receptores de Superficie Celular/química , Neoplasias de la Mama/diagnóstico , Adhesión Celular , Línea Celular Tumoral , Humanos , Cinética , Microscopía de Fuerza Atómica
9.
Biol Pharm Bull ; 41(8): 1282-1285, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30068878

RESUMEN

Fungal ß-glucan is a potent immunological stimulator, and that it activates both the innate immune system and adaptive immunity. Curdlan is (1→3)-ß-glucan, a linear form of ß-glucan with a high molecular weight; it modulates the immune response. However, its role in bone tissue is controversial, and the effects of curdlan on bone tissues are unknown. Toll-like receptors (TLRs) play critical roles in innate immunity, and various ligands for TLRs are thought to regulate the host defense mechanisms against pathogens. TLR2 is known to form heterodimers with TLR6, and the TLR2-TLR6 heterodimer (TLR2/6) recognizes diacylated lipopeptides from Gram-positive bacteria. In the present study, we prepared low molecular-weight curdlan, (1→3)-ß-D-glucan, and examined its effects on bone resorption induced by TLR2/6 signaling. In co-cultures of bone marrow cells and osteoblasts, low molecular-weight curdlan suppressed the osteoclast formation induced by TLR2/6 ligand, and attenuated bone resorption in mouse calvarial organ cultures. Curdlan acted on mouse osteoblasts and suppressed the expression of receptor activator of nuclear factor-kappa B (NF-κB) ligand (RANKL), a key molecule for osteoclastogenesis. Curdlan also acted on mouse bone marrow macrophages and suppressed RANKL-dependent osteoclast differentiation from osteoclast precursor cells. The present study indicates that low molecular-weight curdlan attenuated TLR2-induced inflammatory bone resorption. Curdlan, (1→3)-ß-glucan may be a natural agent with beneficial effects on bone health in humans.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Osteoclastos/efectos de los fármacos , beta-Glucanos/farmacología , Animales , Células de la Médula Ósea/citología , Resorción Ósea , Células Cultivadas , Técnicas de Cocultivo , Ratones , Osteoblastos/citología , Osteoblastos/metabolismo , Osteoclastos/citología , Osteoprotegerina/genética , Ligando RANK/genética , ARN Mensajero/metabolismo , Cráneo/efectos de los fármacos , Receptor Toll-Like 2 , beta-Glucanos/química
10.
J Biol Chem ; 291(40): 20891-20899, 2016 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-27539855

RESUMEN

The tyrosine kinase inhibitor TAS-115 that blocks VEGF receptor and hepatocyte growth factor receptor MET signaling exhibits antitumor properties in xenografts of human gastric carcinoma. In this study, we have evaluated the efficacy of TAS-115 in preventing prostate cancer metastasis to the bone and bone destruction using the PC3 cell line. When PC3 cells were injected into proximal tibiae in nude mouse, severe trabecular and cortical bone destruction and subsequent tumor growths were detected. Oral administration of TAS-115 almost completely inhibited both PC3-induced bone loss and PC3 cell proliferation by suppressing osteoclastic bone resorption. In an ex vivo bone organ culture, PC3 cells induced osteoclastic bone resorption when co-cultured with calvarial bone, but TAS-115 effectively suppressed the PC3-induced bone destruction. We found that macrophage colony-stimulating factor-dependent macrophage differentiation and subsequent receptor activator of NF-κB ligand-induced osteoclast formation were largely suppressed by adding TAS-115. The phosphorylation of the macrophage colony-stimulating factor receptor FMS and osteoclast related kinases such as ERK and Akt were also suppressed by the presence of TAS-115. Gene expression profiling showed that FMS expression was only seen in macrophage and in the osteoclast cell lineage. Our study indicates that tyrosine kinase signaling in host pre-osteoclasts/osteoclasts is critical for bone destruction induced by tumor cells and that targeting of MET/VEGF receptor/FMS activity makes it a promising therapeutic candidate for the treatment of prostate cancer patients with bone metastasis.


Asunto(s)
Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , Resorción Ósea/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Osteoclastos/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Quinolinas/farmacología , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Tiourea/análogos & derivados , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Resorción Ósea/tratamiento farmacológico , Resorción Ósea/patología , Diferenciación Celular , Línea Celular Tumoral , Técnicas de Cocultivo , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Masculino , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Osteoclastos/patología , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Tiourea/farmacología
11.
Biol Pharm Bull ; 40(5): 716-721, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28458359

RESUMEN

Lutein is a member of the xanthophyll family of carotenoids, which are known to prevent hypoxia-induced cell damage in the eye by removing free radicals. However, its role in other tissues is controversial, and the effects of lutein on bone tissues are unknown. To identify a possible role of lutein in bone tissues, we examined the effects of lutein on bone formation and bone resorption and on femoral bone mass in mice. Lutein enhanced the formation of mineralized bone nodules in cultures of osteoblasts. On the other hand, lutein clearly suppressed 1α, 25-dihydroxyvitamin D3-induced bone resorption as measured by pit formation in organ culture of mouse calvaria. In co-cultures of bone marrow cells and osteoblasts, lutein suppressed 1α, 25-dihydroxyvitamin D3-induced osteoclast formation. In cultures of bone marrow macrophages, lutein suppressed soluble RANKL, the receptor activator of nuclear factor-kappaB (NF-κB) ligand, induced osteoclast formation. When five-week-old male mice were orally administered lutein for 4 weeks, the femoral bone mass was clearly enhanced in cortical bone, as measured by bone mineral density in dual X-ray absorptiometry and micro computed tomography (µCT) analyses. The present study indicates that lutein enhances bone mass in growing mice by suppressing bone resorption and stimulating bone formation. Lutein may be a natural agent that promotes bone turnover and may be beneficial for bone health in humans.


Asunto(s)
Desarrollo Óseo/efectos de los fármacos , Resorción Ósea/prevención & control , Huesos/anatomía & histología , Luteína/farmacología , Absorciometría de Fotón , Animales , Densidad Ósea/efectos de los fármacos , Huesos/diagnóstico por imagen , Calcificación Fisiológica/efectos de los fármacos , Calcitriol/antagonistas & inhibidores , Calcitriol/farmacología , Células Cultivadas , Fémur/anatomía & histología , Fémur/efectos de los fármacos , Luteína/uso terapéutico , Masculino , Ratones , FN-kappa B/antagonistas & inhibidores , Osteoblastos/efectos de los fármacos , Ligando RANK/antagonistas & inhibidores , Tomografía Computarizada por Rayos X
12.
Biosci Biotechnol Biochem ; 81(2): 302-306, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27776451

RESUMEN

Lutein, a member of the xanthophyll family of carotenoids, suppressed IL-1-induced osteoclast differentiation and bone resorption. The survival of mature osteoclasts was also suppressed by lutein in cultures. When lutein was added to the cultures of osteoblasts, lutein enhanced the formation of mineralized bone nodules by elevating BMP2 expression and inhibiting sclerostin expression. Lutein may be beneficial for bone health.


Asunto(s)
Resorción Ósea/tratamiento farmacológico , Luteína/farmacología , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Animales , Proteína Morfogenética Ósea 2/metabolismo , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Luteína/uso terapéutico , Ratones , Osteoclastos/citología , Osteoclastos/metabolismo , Osteoclastos/patología
13.
J Biol Chem ; 290(50): 29781-93, 2015 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-26475855

RESUMEN

The stromal cells associated with tumors such as melanoma are significant determinants of tumor growth and metastasis. Using membrane-bound prostaglandin E synthase 1 (mPges1(-/-)) mice, we show that prostaglandin E2 (PGE2) production by host tissues is critical for B16 melanoma growth, angiogenesis, and metastasis to both bone and soft tissues. Concomitant studies in vitro showed that PGE2 production by fibroblasts is regulated by direct interaction with B16 cells. Autocrine activity of PGE2 further regulates the production of angiogenic factors by fibroblasts, which are key to the vascularization of both primary and metastatic tumor growth. Similarly, cell-cell interactions between B16 cells and host osteoblasts modulate mPGES-1 activity and PGE2 production by the osteoblasts. PGE2, in turn, acts to stimulate receptor activator of NF-κB ligand expression, leading to osteoclast differentiation and bone erosion. Using eicosanoid receptor antagonists, we show that PGE2 acts on osteoblasts and fibroblasts in the tumor microenvironment through the EP4 receptor. Metastatic tumor growth and vascularization in soft tissues was abrogated by an EP4 receptor antagonist. EP4-null Ptger4(-/-) mice do not support B16 melanoma growth. In vitro, an EP4 receptor antagonist modulated PGE2 effects on fibroblast production of angiogenic factors. Our data show that B16 melanoma cells directly influence host stromal cells to generate PGE2 signals governing neoangiogenesis and metastatic growth in bone via osteoclast erosive activity as well as angiogenesis in soft tissue tumors.


Asunto(s)
División Celular , Dinoprostona/metabolismo , Melanoma Experimental/patología , Metástasis de la Neoplasia , Neovascularización Patológica , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Transducción de Señal , Células del Estroma/patología , Animales , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/metabolismo , Ratones , Ratones Noqueados
14.
Biochem Biophys Res Commun ; 478(1): 154-161, 2016 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-27450806

RESUMEN

The metastasis of tumors to bone is known to be promoted by prostaglandin E2 (PGE2) produced by the tumor host stromal tissue. Although bone metastases frequently occur in prostate cancer patients, the significance of PGE2 in stromal responses to the tumor is not known. In this study, we report that PGE2 and its receptor EP4 play a pivotal role in bone destruction and metastasis in an experimental metastasis model of prostate cancer in nude mice. Using human prostate cancer PC-3 cells that are stably transfected with luciferase, we showed that the development of bone metastasis was accompanied by increased osteoclastic bone resorption in the bone metastasis microenvironment, and could be abrogated by an EP4 receptor antagonist. The growth of PC-3 cells in vitro was not influenced by PGE2 or by the EP4 receptor. However, cell-cell interactions between fixed PC-3 cells and host osteoblasts induced PGE2 production and RANKL expression in the osteoblasts. Addition of an EP4 antagonist suppressed both PGE2 and RANKL expression induced by the PC3-osteoblast interaction, which would have consequent effects on osteoclast activation and osteolysis. These results indicate that the blockage of PGE2-EP4 signaling prevents the bone destruction required for prostate cancer metastases, and that this is, in part due to the abrogation of bone cell responses. The study provides further evidence that an EP4 antagonist is a candidate for the treatment of prostate cancer in the blockade of bone metastasis.


Asunto(s)
Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , Resorción Ósea/metabolismo , Osteoblastos/metabolismo , Osteoblastos/patología , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Animales , Neoplasias Óseas/patología , Resorción Ósea/etiología , Resorción Ósea/patología , Línea Celular Tumoral , Células Cultivadas , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología
15.
Biochem Biophys Res Commun ; 478(1): 279-285, 2016 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-27402268

RESUMEN

Carboranes are a class of carbon-containing polyhedral boron cluster compounds with globular geometry and hydrophobic surface that interact with hormone receptors such as estrogen receptor (ER) and androgen receptor (AR). We have synthesized BA321, a novel carborane compound, which binds to AR. We found here that it also binds to ERs, ERα and ERß. In orchidectomized (ORX) mice, femoral bone mass was markedly reduced due to androgen deficiency and BA321 restored bone loss in the male, whilst the decreased weight of seminal vesicle in ORX mice was not recovered by administration of BA321. In female mice, BA321 acts as a pure estrogen agonist, and restored both the loss of bone mass and uterine atrophy due to estrogen deficiency in ovariectomized (OVX) mice. In bone tissues, the trabecular bone loss occurred in both ORX and OVX mice, and BA321 completely restored the trabecular bone loss in both sexes. Cortical bone loss occurred in ORX mice but not in OVX mice, and BA321 clearly restored cortical bone loss due to androgen deficiency in ORX mice. Therefore, BA321 is a novel selective androgen receptor modulator (SARM) that may offer a new therapy option for osteoporosis in the male.


Asunto(s)
Andrógenos/metabolismo , Boranos/administración & dosificación , Osteoporosis/tratamiento farmacológico , Osteoporosis/metabolismo , Receptores Androgénicos/metabolismo , Receptores de Estrógenos/metabolismo , Animales , Huesos/efectos de los fármacos , Huesos/metabolismo , Boranos/farmacocinética , Relación Dosis-Respuesta a Droga , Femenino , Gónadas/efectos de los fármacos , Gónadas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos , Orquiectomía , Osteoporosis/patología , Ovariectomía , Resultado del Tratamiento
16.
Biol Pharm Bull ; 39(4): 625-30, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26822531

RESUMEN

Parathyroid hormone (PTH) is a potential medicine for osteoporosis, and subcutaneous (s.c.) PTH treatment enhances bone mass; however, continuous infusion of PTH elicits bone resorption and induces bone loss. To clarify this contradictory phenomenon, we examined bone markers and bone mass in rats to assess the optimal duration of PTH(1-34) infusion. Continuous infusion of PTH at 1 µg/kg/h (Css, steady-state concentration ca. 300 pg/mL) for 1-4 h clearly stimulated the expression both of bone formation-related genes (c-fos, Wnt4, EphrinB2) and of bone resorption-related genes (tnfsf11, tnfsf11b, encoding receptor activator of nuclear factor-kappaB ligand (RANKL), osteoprotegerin (OPG)), but s.c. treatment stimulated these genes only 1-h after the injection. Rats were treated with 1-, 2-, or 4-h infusions of PTH daily using a totally implanted catheter system, and the femoral bone mineral density (BMD) was measured at 4 weeks. The 1-h infusion of PTH significantly stimulated serum bone formation markers (procollagen I N-terminal propeptide (PINP) and osteocalcin) on day 14 and femoral BMD at 2 and 4 weeks, but the 4-h infusion of PTH did not enhance BMD. Since the 4-h infusion increased the levels of both the bone formation markers and a bone resorption marker (urinary C-terminal telopeptide of type 1 collagen (CTx)), the increased bone resorption may predominate over bone formation. The intermittent elevation of plasma PTH to 300 pg/mL for 1-h each day is optimal for increasing bone mass in rats. In osteoporosis therapy in human, using the optimal duration for the clinical dose of PTH may selectively stimulate bone formation.


Asunto(s)
Densidad Ósea/efectos de los fármacos , Fémur/efectos de los fármacos , Hormona Paratiroidea/farmacología , Animales , Esquema de Medicación , Fémur/metabolismo , Infusiones Intravenosas , Péptidos y Proteínas de Señalización Intercelular/genética , Hormona Paratiroidea/administración & dosificación , Hormona Paratiroidea/sangre , Hormona Paratiroidea/farmacocinética , ARN Mensajero/metabolismo , Ratas , Factores de Transcripción/genética
17.
Biosci Biotechnol Biochem ; 79(1): 155-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25175163
18.
Biosci Biotechnol Biochem ; 77(1): 201-4, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23291758

RESUMEN

We investigated the effects of equol on adipogenesis by measuring lipid accumulation and analyzing the change in adipocyte-related gene expression in MC3T3-L1 cells. Treatment with 10 µM equol tended to increase adipocyte-related gene expression, whereas 100 µM equol reduced lipid accumulation and suppressed the expression of these genes and proteins. Our results suggest that equol regulated adipogenesis in a bi-phasic fashion.


Asunto(s)
Adipocitos/efectos de los fármacos , Adipogénesis/efectos de los fármacos , Equol/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Fitoestrógenos/farmacología , Células 3T3-L1 , Adipocitos/citología , Adipocitos/metabolismo , Adipogénesis/genética , Animales , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Antígenos CD36/genética , Antígenos CD36/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , PPAR gamma/genética , PPAR gamma/metabolismo , Receptor fas/genética , Receptor fas/metabolismo
19.
Biosci Biotechnol Biochem ; 77(4): 860-2, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23615426

RESUMEN

We examined the effects of ß-cryptoxanthin, a typical carotenoid, on inflammatory periodontitis. ß-Cryptoxanthin suppressed lipopolysaccharide (LPS)-induced osteoclast formation in co-cultures of bone marrow cells and osteoblasts. In a mouse model of periodontitis, it suppressed bone resorption in the mandibular alveolar bone in vitro and restored alveolar bone loss induced by LPS in vivo. ß-Cryptoxanthin might protect against periodontal disease.


Asunto(s)
Resorción Ósea/tratamiento farmacológico , Periodontitis/tratamiento farmacológico , Periodontitis/fisiopatología , Xantófilas/farmacología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Criptoxantinas , Modelos Animales de Enfermedad , Humanos , Inflamación/tratamiento farmacológico , Ratones , Células 3T3 NIH , Osteoblastos/efectos de los fármacos , Osteoblastos/patología , Osteoclastos/efectos de los fármacos , Osteoclastos/patología , Periodontitis/patología , Xantófilas/uso terapéutico
20.
J Clin Biochem Nutr ; 53(1): 41-8, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23874069

RESUMEN

S-equol is a natural metabolite of the soy isoflavone, daidzein, produced by intestinal bacteria. S-equol has been shown to have greater estrogenic activity than other soy isoflavones and prevent bone loss in post-menopausal women. Estrogen regulates both bone remodeling and hemopoiesis in the bone marrow, these processes that communicate closely with each other. In this study, we investigated the effect of S-equol on bone mass and gene expression of bone marrow cells in ovariectomized (OVX) mice. Female ddY strain mice, aged 12 weeks, were either sham operated or OVX. The OVX mice were randomly divided into two groups: (1) OVX control and (2) OVX fed a 0.06% (w/w) S-equol supplemented diet. After 2 weeks, the trabecular bone volume of the femoral distal metaphysis was markedly reduced in OVX mice. However, treatment with equol was observed to ameliorate this. Expression of inflammatory-, osteoclastogenesis- and adipogenesis-related genes was increased in OVX mice compared with sham mice, and equol was observed to suppress their expression. The present study demonstrates that equol might ameliorate bone loss caused by estrogen deficiency through regulating hemopoiesis and production of inflammatory cytokines in bone marrow cells.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA