Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
In Vitro Cell Dev Biol Anim ; 59(4): 300-311, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37002492

RESUMEN

The inflammatory cytokine IL-17A is known to have the capacity to promote osteoclastogenesis, thereby enhancing bone loss. Moreover, IL-17A can promote the expression of RANKL in osteoblasts, contributing to its pro-osteoclastogenic effect. IL-17A is an autophagy regulator, which is also responsible for its regulation on RANKL expression. However, the specific role of autophagy in IL-17A-regulated RANKL expression and the underlying mechanism of IL-17A-regulated osteoblast autophagy remain unclear. IL-17A is known to inhibit autophagy by preventing BCL2 degradation. This study aimed to explore the significance of BCL2-dependent autophagy in IL-17A-regulated RANKL expression. Our results showed that IL-17A at 50 ng/mL could inhibit autophagic activity and promote RANKL protein expression in MC3T3-E1 osteoblast line. Moreover, the corresponding concentration of IL-17A could enhance BCL2 protein expression and the protein interaction between BCL2 and Beclin1 in MC3T3-E1 cells. However, the protein expression of RANKL and BCL2 promoted by 50 ng/mL of IL-17A was blocked by autophagy activation with Beclin1 pharmacological upregulation. Furthermore, RANKL protein expression promoted by 50 ng/mL of IL-17A was also reversed by autophagy activation with BCL2 knockdown. Importantly, the supernatant from osteoblasts treated with 50 ng/mL of IL-17A made osteoclast precursors (OCPs) form larger osteoclasts, which was reversed by BCL2 knockdown in osteoblasts. In conclusion, high levels of IL-17A prevent the degradation of RANKL by inhibiting BCL2-Beclin1-autophagy activation signal transduction in osteoblasts, thereby indirectly promoting osteoclastogenesis.


Asunto(s)
Interleucina-17 , Ligando RANK , Animales , Beclina-1/genética , Ligando RANK/farmacología , Ligando RANK/metabolismo , Interleucina-17/farmacología , Interleucina-17/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Transducción de Señal , Autofagia/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo
2.
Exp Biol Med (Maywood) ; 247(19): 1764-1775, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35957534

RESUMEN

Morroniside is known to improve osteoporosis by promoting osteoblastogenesis. The activation of PI3K/Akt/mTOR signaling is a significant mechanism in morroniside-promoted osteoblastogenesis. It is well known that protective autophagy is an important factor in osteoblastogenesis. However, the activation of mTOR signaling can inhibit autophagy. This study aimed to investigate the relationship between mTOR signaling and autophagy in morroniside-regulated osteoblastogenesis. In this study, we investigated the effect of morroniside on the autophagic activity (LC3 conversion rate, LC3-puncta formation, and autophagosome number) of differentiated osteoblast precursors (MC3T3-E1 cells). Then, we identified the roles of mTOR knockdown in morroniside-regulated alterations of autophagy and osteogenic parameters in MC3T3-E1 cells. Next, mTOR knockdown and overexpression were used to observe the roles of mTOR in morroniside-regulated alterations of autophagic molecules (Atg7, Atg13, and Beclin1). Subsequently, the additional value of the above autophagic molecules on morroniside-regulated osteogenic parameters in MC3T3-E1 cells was analyzed based on lentiviral transduction. Finally, combined with morroniside and TAT-Beclin1, the roles of Beclin1 upregulation in the in vivo effects of morroniside was investigated. Our experimental data showed that morroniside promoted both the mTOR activity and autophagy in MC3T3-E1 cells. Morroniside-upregulated autophagic activity and Atg13 or Beclin1 protein level in MC3T3-E1 cells were enhanced by mTOR knockdown. Furthermore, Morroniside-upregulated Atg13 and Beclin1 expression was reversed by mTOR overexpression. Importantly, autophagy upregulation with overexpression of the autophagic gene, Atg13 or BECN1 (gene form of Beclin1), significantly promoted osteoblastogenesis regulated by morroniside. The promotional effect of morroniside on bone microarchitecture, bone mass, and bone parameters (including trabecular bone area and OCN expression in trabecular bone) in ovariectomized (OVX) mice was enhanced by TAT-Beclin1 administration. In conclusion, the autophagy-enhancing drugs related to Beclin1 or Atg13 may be an effective adjuvant therapy in the treatment of osteoporosis with morroniside.


Asunto(s)
Proteínas Reguladoras de la Apoptosis , Beclina-1 , Osteoporosis , Fosfatidilinositol 3-Quinasas , Animales , Ratones , Autofagia , Beclina-1/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo
3.
Int J Biol Sci ; 18(4): 1508-1520, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35280670

RESUMEN

Sustained activation of multiple receptor tyrosine kinases (RTKs) simultaneously is vital for tumorigenesis and progression of osteosarcoma (OS). Gαi proteins recruitment to various RTKs mediates downstream oncogenic signaling activation. The expression, functions and underlying mechanisms of Gαi3 in human OS were examined. Expression of Gαi3 is robustly elevated in human OS tissues and is correlated with a poor overall survival. In patient-derived primary OS cells and immortalized lines (MG63 and U2OS), Gαi3 depletion, by shRNA and CRISPR/Cas9 strategies, robustly suppressed cell viability, proliferation and migration, while provoking G1-S arrest and apoptosis activation. Conversely, Gαi3 overexpressing ectopically can accelerate proliferation and migration of OS cells. In OS cells, Gαi3 immunoprecipitated with VEGFR2, FGFR, PGDFR and EGFR, mediating downstream cascade transduction. Akt-mTOR activation in primary OS cells was potently inhibited by Gαi3 shRNA, knockout or dominant negative mutation, but augmented after Gαi3 overexpression. In vivo studies showed that Gαi3 shRNA AAV (adeno-associated viruses) intratumoral injection largely inhibited the growth of subcutaneous xenografts of primary OS cells. Moreover, the growth of the Gαi3-knockout primary OS xenografts was much slower than that of OS xenografts with empty vector. In Gαi3-depleted OS xenografts tissues, Gαi3 downregulation and Akt-mTOR inactivation were detected. Taken together, overexpressed Gαi3 mediates RTK-Akt signaling to drive OS progression.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Apoptosis/genética , Neoplasias Óseas/genética , Línea Celular Tumoral , Proliferación Celular/genética , Humanos , Osteosarcoma/genética , Osteosarcoma/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño , Proteínas Tirosina Quinasas Receptoras , Serina-Treonina Quinasas TOR/metabolismo
4.
Cell Death Discov ; 7(1): 393, 2021 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-34907167

RESUMEN

POLRMT (RNA polymerase mitochondrial) is essential for transcription of mitochondrial genome encoding components of oxidative phosphorylation process. The current study tested POLRMT expression and its potential function in osteosarcoma (OS). The Cancer Genome Atlas (TCGA) cohorts and Gene Expression Profiling Interactive Analysis (GEPIA) database both show that POLRMT transcripts are elevated in OS tissues. In addition, POLRMT mRNA and protein levels were upregulated in local OS tissues as well as in established and primary human OS cells. In different OS cells, shRNA-induced stable knockdown of POLRMT decreased cell viability, proliferation, migration, and invasion, whiling inducing apoptosis activation. CRISPR/Cas9-induced POLRMT knockout induced potent anti-OS cell activity as well. Conversely, in primary OS cells ectopic POLRMT overexpression accelerated cell proliferation and migration. In vivo, intratumoral injection of adeno-associated virus-packed POLRMT shRNA potently inhibited U2OS xenograft growth in nude mice. Importantly, levels of mitochondrial DNA, mitochondrial transcripts and expression of respiratory chain complex subunits were significantly decreased in U2OS xenografts with POLRMT shRNA virus injection. Together, POLRMT is overexpressed in human OS, promoting cell growth in vitro and in vivo. POLRMT could be a novel therapeutic target for OS.

5.
In Vitro Cell Dev Biol Anim ; 57(9): 878-885, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34780049

RESUMEN

As the active form of vitamin D3, 1α,25-(OH)2-D3 promotes receptor activator for nuclear factor-κB ligand (RANKL)-induced autophagy in osteoclast precursors (OCPs). However, the relationship between 1α,25-(OH)2-D3 and RANKL signaling is still unknown. This study aimed to explore whether 1α,25-(OH)2-D3 regulates OCP autophagy and osteoclastogenesis through RANKL signaling. Our results showed that 1α,25-(OH)2-D3 directly decreased OCP autophagy while significantly enhancing the ability of RANKL to promote OCP autophagy. Moreover, 1α,25-(OH)2-D3 not only promoted the expression of key signaling proteins in OCPs induced by RANKL but also enhanced the coimmunoprecipitation levels of RANK and TRAF6. Notably, 1α,25-(OH)2-D3 significantly enhanced the autophagic activity and osteoclast differentiation of RANK-positive OCPs but did not affect the autophagic activity or osteoclast differentiation of RANK-negative OCPs. More importantly, 1α,25-(OH)2-D3 had no effect on autophagy or osteoclastogenesis in TRAF6-silenced OCPs. Overall, 1α,25-(OH)2-D3 could upregulate RANKL-RANK-TRAF6 signaling in OCPs, thereby promoting OCP autophagy and osteoclastogenesis.


Asunto(s)
Autofagia/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Vitamina D/análogos & derivados , Animales , Autofagia/fisiología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Ratones Endogámicos C57BL , Osteoclastos/citología , Osteoclastos/metabolismo , Osteogénesis/fisiología , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Transducción de Señal/efectos de los fármacos , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo , Vitamina D/farmacología
6.
Front Oncol ; 11: 659181, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34336655

RESUMEN

Osteosarcoma (OS) is a common primary bone malignancy. We here investigated the potential activity of PF-06409577, a novel, potent, and direct activator of AMP-activated protein kinase (AMPK), against human OS cells. In established (U2OS, MG-63, and SaOs-2 lines) and primary human OS cells, PF-06409577 inhibited cell viability and proliferation, while inducing cell apoptosis and cell cycle arrest. PF-06409577 induced AMPK activation, mTORC1 inhibition, autophagy induction, and downregulation of multiple receptor tyrosine kinase inOS cells. AMPK inactivation by AMPKα1 shRNA, CRISPR/Cas9 knockout, or dominant negative mutation (T172A) was able to abolish PF-06409577-induced activity in OS cells. In vivo, PF-06409577 oral administration at well-tolerated doses potently inhibited growth of U2OS cells and primary human OS cells in severe combined immunodeficient mice. AMPK activation, mTORC1 inhibition, autophagy induction, as well as RTK degradation and apoptosis activation were detected in PF-06409577-treated xenografts. In conclusion, activation of AMPK by PF-06409577 inhibits OS cell growth.

7.
Theranostics ; 11(10): 4599-4615, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33754016

RESUMEN

HBO1 (KAT7 or MYST2) is a histone acetyltransferase that acetylates H3 and H4 histones. Methods: HBO1 expression was tested in human OS tissues and cells. Genetic strategies, including shRNA, CRISPR/Cas9 and overexpression constructs, were applied to exogenously alter HBO1 expression in OS cells. The HBO1 inhibitor WM-3835 was utilized to block HBO1 activation. Results:HBO1 mRNA and protein expression is significantly elevated in OS tissues and cells. In established (MG63/U2OS lines) and primary human OS cells, shRNA-mediated HBO1 silencing and CRISPR/Cas9-induced HBO1 knockout were able to potently inhibit cell viability, growth, proliferation, as well as cell migration and invasion. Significant increase of apoptosis was detected in HBO1-silenced/knockout OS cells. Conversely, ectopic HBO1 overexpression promoted OS cell proliferation and migration. We identified ZNF384 (zinc finger protein 384) as a potential transcription factor of HBO1. Increased binding between ZNF384 and HBO1 promoter was detected in OS cell and tissues, whereas ZNF384 silencing via shRNA downregulated HBO1 and produced significant anti-OS cell activity. In vivo, intratumoral injection of HBO1 shRNA lentivirus silenced HBO1 and inhibited OS xenograft growth in mice. Furthermore, growth of HBO1-knockout OS xenografts was significantly slower than the control xenografts. WM-3835, a novel and high-specific small molecule HBO1 inhibitor, was able to potently suppressed OS cell proliferation and migration, and led to apoptosis activation. Furthermore, intraperitoneal injection of a single dose of WM-3835 potently inhibited OS xenograft growth in SCID mice. Conclusion: HBO1 overexpression promotes OS cell growth in vitro and in vivo.


Asunto(s)
Apoptosis/genética , Neoplasias Óseas/genética , Proliferación Celular/genética , Histona Acetiltransferasas/genética , Osteosarcoma/genética , Animales , Apoptosis/efectos de los fármacos , Sistemas CRISPR-Cas , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Inhibidores Enzimáticos/farmacología , Femenino , Técnicas de Inactivación de Genes , Histona Acetiltransferasas/antagonistas & inhibidores , Humanos , Masculino , Ratones , Ratones SCID , Trasplante de Neoplasias , Oncogenes , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , ARN Interferente Pequeño , Transactivadores/metabolismo
8.
Aging (Albany NY) ; 12(6): 5399-5410, 2020 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-32203055

RESUMEN

Sphingosine kinase 1 (SphK1) is a potential therapeutic target for human osteosarcoma (OS). SphK1-targeting microRNAs (miRNAs) could have important therapeutic value for OS. We discovered that micorRNA-3677 (miR-3677) is a SphK1-targeting miRNA, inhibiting OS cell progression. The results of RNA-Pull down assay confirmed direct binding between biotinylated-miR-3677 and SphK1 mRNA in primary human OS cells. In established and primary human OS cells forced overexpression of miR-3677, by a lentiviral construct, decreased SphK1 3'-UTR (untranslated region) activity and downregulated SphK1 expression. Both were however enhanced with miR-3677 inhibition in OS cells. Function studies demonstrated that OS cell growth, proliferation and migration were inhibited with miR-3677 overexpression, but augmented with miR-3677 inhibition. MiR-3677 overexpression-induced anti-OS cell activity was reversed with re-expression of the 3'-UTR-depleted SphK1. Additionally, in SphK1 knockout OS cells (by CRISPR/Cas9 strategy), altering miR-3677 expression failed to further alter cell functions. Finally, we show that miR-3677 expression was significantly downregulated in primary human OS tissues, correlating with SphK1 mRNA upregulation. We conclude that targeting SphK1 by miR-3677 inhibits human OS cell progression.


Asunto(s)
MicroARNs/metabolismo , Osteosarcoma/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Regiones no Traducidas 3' , Apoptosis/genética , Ciclo Celular , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Humanos , ARN Mensajero/metabolismo , Regulación hacia Arriba
9.
Cell Prolif ; 53(4): e12789, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32157750

RESUMEN

OBJECTIVES: Oestrogen is known to inhibit osteoclastogenesis, and numerous studies have identified it as an autophagic activator. To date, the role of oestrogen in the autophagy of osteoclast precursors (OCPs) during osteoclastogenesis remains unclear. This study aimed to determine the effect of autophagy regulated by the biologically active form of oestrogen (17ß-estradiol) on osteoclastogenesis. MATERIALS AND METHODS: After treatment with 17ß-estradiol in OCPs (from bone marrow-derived macrophages, BMMs) and ovariectomy (OVX) mice, we measured the effect of 17ß-estradiol on the autophagy of OCPs in vitro and in vivo. In addition, we studied the role of autophagy in the OCP proliferation, osteoclast differentiation and bone loss regulated by 17ß-estradiol using autophagic inhibitor or knock-down of autophagic genes. RESULTS: The results showed that direct administration of 17ß-estradiol enhanced the autophagic response of OCPs. Interestingly, 17ß-estradiol inhibited the stimulatory effect of receptor activator of nuclear factor-κB ligand (RANKL) on the autophagy and osteoclastogenesis of OCPs. Moreover, 17ß-estradiol inhibited the downstream signalling of RANKL. Autophagic suppression by pharmacological inhibitors or gene silencing enhanced the inhibitory effect of 17ß-estradiol on osteoclastogenesis. In vivo assays showed that the autophagic inhibitor 3-MA not only inhibited the autophagic activity of the OCPs in the trabecular bone of OVX mice but also enhanced the ability of 17ß-estradiol to ameliorate bone loss. CONCLUSIONS: In conclusion, our study showed that oestrogen directly enhanced the autophagy of OCPs, which inhibited its anti-osteoclastogenic effect. Drugs based on autophagic inhibition may enhance the efficacy of oestrogen on osteoporosis.


Asunto(s)
Autofagia/efectos de los fármacos , Estradiol/farmacología , Estrógenos/farmacología , Osteogénesis/efectos de los fármacos , Animales , Células Cultivadas , Estradiol/uso terapéutico , Estrógenos/uso terapéutico , Femenino , Ratones , Ratones Endogámicos C57BL , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteoporosis/tratamiento farmacológico , Osteoporosis/metabolismo , Ligando RANK/metabolismo
10.
Biochimie ; 167: 217-227, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31654668

RESUMEN

RANKL induces osteoclastogenesis via JNK1 signal that exerts an anti-apoptotic effect during osteoclastogenesis. However, the classic downstream c-Jun/AP-1 pathway is not included in anti-apoptosis of JNK1. Thus, the detail mechanism underlying JNK1-resisted apoptosis remains unknown during RANKL-induced osteoclastogenesis. RANKL-induced autophagy results in the degradation of the osteoclastogenesis-inhibitor TRAF3, and TRAF3 is thought as a regulator of apoptosis. Given the key effect of JNK1 in mediating autophagy, our study aims to investigate the significance of TRAF3 in bridging JNK1-mediated autophagy and apoptotic resistance during osteoclastogenesis. In this study, by using Bone Marrow-derived macrophages (BMMs) as osteoclast precursors (OCPs), we found that RANKL-induced TRAF3 degradation was significantly suppressed by JNK inhibitor (SP600125), which was restored by overexpression of Beclin1 (key autophagic protein). Nevertheless, TRAF3 silencing partially reversed the reduced osteoclastogenesis under SP600125 intervention. Besides, OCP apoptosis was positively regulated by TRAF3 overexpression, regardless of the application of autophagy inhibitor or SP600125. Remarkably, the enhanced apoptosis caused by the pharmacological inhibition of Beclin1 was reversed by TRAF3 silencing. Together, these results suggest that JNK1-mediated autophagy could promote RANKL-induced osteoclastogenesis via enhancing TRAF3 degradation. Importantly, JNK1 could prevent OCP apoptosis through autophagy-TRAF3 signaling, which provides more potential targets for clinical treatment of pathological bone loss.


Asunto(s)
Macrófagos/metabolismo , Osteoclastos/metabolismo , Osteogénesis , Factor 3 Asociado a Receptor de TNF/fisiología , Animales , Apoptosis , Autofagia , Beclina-1/metabolismo , Células Cultivadas , Macrófagos/citología , Ratones Endogámicos C57BL , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Osteoclastos/citología , Ligando RANK/metabolismo , Transducción de Señal
11.
Oncol Lett ; 17(1): 360-364, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30655775

RESUMEN

Clinical and radiographic outcomes of multilevel cervical myelopathy (MCM) patients treated with a hybrid technique combining cervical anterior corpectomy and fusion (ACF) with cervical artificial disc replacement (C-ADR) were evaluated. A total of 23 patients including 14 females and 9 males (mean age, 48.3 years) were treated with the hybrid technique and they were followed up for an average time of 35 months (range from 24 to 40 months). Arm- and neck-pain visual analogue scale (VAS) scores, neck disability index (NDI) scores and C2-7 range of motion (ROM) preoperation and 6 weeks, 3 and 24 months postoperation were assessed and compared. Fusion success and system failure based on anteroposterior (AP), lateral and flexion/extension X-rays were examined by an independent reviewer. Postoperative VAS, NDI and ROM were decreased significantly at all the time-points examined, as compared to preoperative scores (P<0.05). The results revealed that this hybrid technique is both technically feasible and effective for the treatment of MCM. However, future comparative studies will be required to determine the potential benefits and pitfalls of this hybrid technique.

12.
Oncotarget ; 8(16): 26424-26433, 2017 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-28460435

RESUMEN

Forced-activation of AMP-activated protein kinase (AMPK) can possibly inhibit osteoblastoma cells. Here, we aim to provoke AMPK activation via microRNA silencing its phosphatase Ppm1e (protein phosphatase Mg2+/Mn2+-dependent 1e). We showed that microRNA-135b-5p ("miR-135b-5p"), the anti-Ppm1e microRNA, was significantly downregulated in human osteoblastoma tissues. It was correlated with Ppm1e upregulation and AMPKα1 de-phosphorylation. Forced-expression of miR-135b-5p in human osteoblastoma cells (MG-63 and U2OS lines) silenced Ppm1e, and induced a profound AMPKα1 phosphorylation (at Thr-172). Osteoblastoma cell proliferation was inhibited after miR-135b-5p expression. Intriguingly, Ppm1e shRNA knockdown similarly induced AMPKα1 phosphorylation, causing osteoblastoma cell proliferation. Reversely, AMPKα1 shRNA knockdown or dominant negative mutation almost abolished miR-135b-5p's actions in osteoblastoma cells. Further in vivo studies demonstrated that U2OS tumor growth in mice was dramatically inhibited after expressing miR-135b-5p or Ppm1e shRNA. Together, our results suggest that miR-135b-induced Ppm1e silence induces AMPK activation to inhibit osteoblastoma cell proliferation.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Silenciador del Gen , MicroARNs/genética , Osteoblastoma/genética , Osteoblastoma/metabolismo , Proteína Fosfatasa 2C/genética , Animales , Neoplasias Óseas/patología , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Activación Enzimática , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Mutación , Osteoblastoma/patología , Fosforilación , ARN Interferente Pequeño/genética , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Oncotarget ; 7(48): 79417-79427, 2016 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-27765904

RESUMEN

Malignant osteosarcoma (OS) is still a deadly disease for many affected patients. The search for the novel anti-OS agent is extremely urgent and important. Our previous study has proposed that salinomycin is a novel anti-OS agent. Here we characterized DNA-dependent protein kinase catalytic subunit (DNA-PKcs) as a primary salinomycin resistance factor in OS cells. DNA-PKcs inhibitors (NU7026, NU7441 and LY294002) or DNA-PKcs shRNA knockdown dramatically potentiated salinomycin-induced death and apoptosis of OS cells (U2OS and MG-63 lines). Further, forced-expression of microRNA-101 ("miR-101") downregulated DNA-PKcs and augmented salinomycin's cytotoxicity against OS cells. Reversely, over-expression of DNA-PKcs in OS cells inhibited salinomycin's lethality. For the mechanism study, we show that DNA-PKcs is required for salinomycin-induced pro-survival autophagy activation. DNA-PKcs inhibition (by NU7441), shRNA knockdown or miR-101 expression inhibited salinomycin-induced Beclin-1 expression and autophagy induction. Meanwhile, knockdown of Beclin-1 by shRNA significantly sensitized salinomycin-induced OS cell lethality. In vivo, salinomycin administration suppressed U2OS xenograft tumor growth in severe combined immuno-deficient (SCID) mice, and its anti-tumor activity was dramatically potentiated with co-administration of the DNA-PKcs inhibitor NU7026. Together, these results suggest that DNA-PKcs could be a primary resistance factor of salinomycin in OS cells. DNA-PKcs inhibition or silence may thus significantly increase salinomycin's sensitivity in OS cells.


Asunto(s)
Neoplasias Óseas/metabolismo , Cromonas/administración & dosificación , Proteína Quinasa Activada por ADN/genética , Resistencia a Antineoplásicos , Morfolinas/administración & dosificación , Proteínas Nucleares/genética , Osteosarcoma/metabolismo , Piranos/administración & dosificación , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Neoplasias Óseas/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cromonas/farmacología , Proteína Quinasa Activada por ADN/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Ratones , MicroARNs/genética , Morfolinas/farmacología , Trasplante de Neoplasias , Proteínas Nucleares/metabolismo , Osteosarcoma/genética , Piranos/farmacología
14.
Oncotarget ; 7(31): 49527-49538, 2016 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-27385099

RESUMEN

In the present study, we investigated the activity of XL388, a novel mammalian target of rapamycin (mTOR) complex 1/2 (mTORC1/2) dual inhibitor, in preclinical osteosarcoma (OS) models. XL388 was cytotoxic, cytostatic and pro-apoptotic to multiple established OS cell lines and primary human OS cells. XL388 blocked mTORC1/2 activation and downregulated cyclin D1/B1 expressions in OS cells, leaving AKT Thr-308 phosphorylation un-affected. Intriguingly, AKT1 T308A mutation potentiated XL388-induced cytotoxicity in OS cells. XL388 activated cytoprotective autophagy in OS cells. Autophagy inhibition, either pharmacologically or genetically, augmented XL388-induced anti-OS activity. Further, XL388 oral administration inhibited U2OS xenografts growth in severe combined immuno-deficient (SCID) mice. Such activity was enhanced with co-administration of the autophagy inhibitor 3-methyladenine (3-MA). Similarly, Beclin-1-silenced U2OS xenografts were remarkably more sensitive to XL388. Thus, concurrent blockage of mTORC1/2 with XL388 may have therapeutic value for OS.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Óseas/tratamiento farmacológico , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Osteosarcoma/tratamiento farmacológico , Sulfonas/farmacología , Adenina/análogos & derivados , Adenina/farmacología , Adolescente , Animales , Apoptosis/efectos de los fármacos , Autofagia , Beclina-1/metabolismo , Caspasas/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Silenciador del Gen , Humanos , Masculino , Ratones , Ratones SCID , Trasplante de Neoplasias , Osteosarcoma/metabolismo , Fosforilación , Resultado del Tratamiento , Células Tumorales Cultivadas , Adulto Joven
15.
Tumour Biol ; 37(9): 11743-11752, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27020593

RESUMEN

Mammalian target of rapamycin (mTOR) complex 1 (mTORC1) and mTORC2 are frequently dysregulated in human colon cancers. In the present study, we evaluated the potential anti-colon cancer cell activity by a novel mTORC1/2 dual inhibitor WYE-354. We showed that WYE-354 was anti-survival and anti-proliferative when adding to primary (patient-derived) and established (HCT-116, HT-29, Caco-2, LoVo, and DLD-1 lines) colon cancer cells. In addition, WYE-354 treatment activated caspase-dependent apoptosis in the colon cancer cells. Mechanistically, WYE-354 blocked mTORC1 and mTORC2 activation. Meanwhile, it also induced autophagy activation in the colon cancer cells. Autophagy inhibitors (bafilomycin A1 and 3-methyladenine), or shRNA-mediated knockdown of autophagy elements (Beclin-1 and ATG-5), remarkably sensitized WYE-354-mediated anti-colon cancer cell activity in vitro. Further studies showed that WYE-354 administration inhibited HT-29 xenograft growth in severe combined immunodeficient (SCID) mice. Importantly, its activity in vivo was further potentiated with co-administration of the autophagy inhibitor 3-MA. Phosphorylations of Akt (Ser-473) and S6 were also decreased in WYE-354-treated HT-29 xenografts. Together, these pre-clinical results demonstrate the potent anti-colon cancer cell activity by WYE-354, and its activity may be further augmented with autophagy inhibition.


Asunto(s)
Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Complejos Multiproteicos/antagonistas & inhibidores , Purinas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Adenina/análogos & derivados , Adenina/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Neoplasias del Colon/patología , Femenino , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones
16.
Cancer Biol Ther ; 16(4): 602-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25869769

RESUMEN

Recent studies have identified that constitutively active phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling is an important feature of osteosarcoma, where it promotes cell proliferation, survival, and chemo-resistance. Here, we studied the therapeutic potential of NVP-BEZ235, a novel dual PI3K/mTOR dual inhibitor, on osteosarcoma cells in vivo and in vitro. NVP-BEZ235 was cytotoxic and cytostatic to a panel of osteosarcoma lines (MG-63, U2OS and SaOs-2), where it induce apoptosis and cell-cycle arrest. At the molecular level, NVP-BEZ235 inhibited PI3K-AKT-mTORC1 activation and downregulated cyclin D1/cyclin B1 expressions, while increasing MEK/Erk phosphorylation in osteosarcoma cells. MEK/Erk inhibitors PD98059 and MEK-162 increased NVP-BEZ235 activity on osteosarcoma cells. In vivo, oral NVP-BEZ235 inhibited U2OS xenograft in SCID mice, and its anti-tumor efficiency was further enhanced by MEK-162 co-administration. Taken together, our findings indicate that dual inhibition of PI3K and mTOR with NVP-BEZ235, either alone or in combination with MEK/Erk inhibitors, may be an efficient treatment for osteosarcoma.


Asunto(s)
Imidazoles/farmacología , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Quinolinas/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Apoptosis/efectos de los fármacos , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones SCID , Complejos Multiproteicos/metabolismo , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Zhonghua Yi Xue Za Zhi ; 94(17): 1326-9, 2014 May 06.
Artículo en Chino | MEDLINE | ID: mdl-25142854

RESUMEN

OBJECTIVE: To evaluate the mid-term clinical outcomes of unilateral pedicle screw fixation with lumbar interbody fusion for lumber degenerative diseases. METHODS: From October 2008 to December 2010, unilateral pedicle screw fixation with lumbar interbody fusion was performed for a consecutive cohort of 42 patients. There were 18 males and 24 females with an average age of 52 (38-69) years. The operation level at L3-4 (n = 2), L4-5 (n = 19), L5-S1 (n = 21) and multilevel (n = 0). Their clinical outcomes were assessed by Oswestry (ODI) scores and Japanese Orthopedics Association (JOA) questionnaires before and after operation. Operative duration, intraoperative blood loss, incision status and complications were recorded. Radiological examination was performed to assess the height of intervertebral space, the postoperative intervertebrai fusion conditions and the degeneration of adjacent segments. RESULTS: The mean operative duration was 90 minutes and mean blood loss 150 ml. All incisions healed primarily. The mean follow-up period was 40 (36-58) months. The ODI scores decreased significantly from 59.1 ± 9.6 preoperatively to 8.5 ± 2.7 postoperatively (P < 0.05). The JOA scores improved markedly from 8.3 ± 2.7 preoperatively to 23.3 ± 2.1 postoperatively (P < 0.05). And the proportion of optimal outcomes was 88.1%. The ventral and dorsal heights of intervertebal disc were significantly higher than those before operation. The fusion rate was 95.2% and the incidence of adjacent segment degeneration 9.5%. There was no occurrence of such complications as secondary scoliosis, screw loosening, internal fixation failure and cage slippage. CONCLUSION: Unilateral pedicle screw fixation with lumbar interbody fusion is efficacious for lumber degenerative diseases with little surgical trauma.


Asunto(s)
Degeneración del Disco Intervertebral/cirugía , Vértebras Lumbares , Adulto , Anciano , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Tornillos Pediculares , Estudios Retrospectivos , Fusión Vertebral/métodos , Resultado del Tratamiento
18.
Biochem Biophys Res Commun ; 451(1): 112-8, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25068992

RESUMEN

The search for novel and more efficient chemo-agents against malignant osteoblastoma is important. In this study, we examined the potential anti-osteoblastoma function of bufotalin, and studied the underlying mechanisms. Our results showed that bufotalin induced osteoblastoma cell death and apoptosis in dose- and time-dependent manners. Further, bufotalin induced endoplasmic reticulum (ER) stress activation in osteoblastoma cells, the latter was detected by the induction of C/EBP homologous protein (CHOP), phosphorylation of inositol-requiring enzyme 1 (IRE1) and PKR-like endoplasmic reticulum kinase (PERK), as well as caspase-12 activation. Conversely, the ER stress inhibitor salubrinal, the caspase-12 inhibitor z-ATAD-fmk as well as CHOP depletion by shRNA significantly inhibited bufotalin-induced osteoblastoma cell death and apoptosis. Finally, by using a mice xenograft model, we demonstrated that bufotalin inhibited U2OS osteoblastoma cell growth in vivo. In summary, our results suggest that ER stress contributes to bufotalin-induced apoptosis in osteoblastoma cells. Bufotalin might be investigated as a novel anti-osteoblastoma agent.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias Óseas/tratamiento farmacológico , Bufanólidos/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Osteoblastoma/tratamiento farmacológico , Animales , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Caspasa 12/metabolismo , Cinamatos/farmacología , Relación Dosis-Respuesta a Droga , Silenciador del Gen , Humanos , Masculino , Ratones , Ratones SCID , Osteoblastoma/metabolismo , Osteoblastoma/patología , Tiourea/análogos & derivados , Tiourea/farmacología , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
19.
J Int Med Res ; 41(5): 1768-72, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24029021

RESUMEN

We report a case of primary empty sella syndrome (ESS) resulting in osteoporotic fractures and persistent non-fusion of the hand epiphyses, and discuss the potential pathogenesis of this disease. A 41-year-old man presented with pain in the right hand and back after a fall. X-radiographs revealed persistent epiphyses and severe osteoporosis. Serum phosphorus and prolactin levels were above normal levels, and free triiodothyronine, free thyroxine and testosterone levels were below normal limits. Magnetic resonance imaging of the head revealed empty sella. A lumbar bone mineral density examination indicated severe osteoporosis. ESS caused a systemic hormone disorder in this patient, resulting in osteoporotic fractures and persistent non-fusion of the hand epiphyses. Possible causes of this anomaly are chronic or congenital abnormities of the pituitary gland.


Asunto(s)
Síndrome de Silla Turca Vacía/patología , Epífisis/patología , Fracturas Osteoporóticas/patología , Hipófisis/patología , Adulto , Síndrome de Silla Turca Vacía/sangre , Síndrome de Silla Turca Vacía/complicaciones , Síndrome de Silla Turca Vacía/diagnóstico por imagen , Epífisis/diagnóstico por imagen , Humanos , Masculino , Fracturas Osteoporóticas/sangre , Fracturas Osteoporóticas/diagnóstico por imagen , Fracturas Osteoporóticas/etiología , Hipófisis/diagnóstico por imagen , Hipófisis/metabolismo , Radiografía , Testosterona/sangre , Testosterona/deficiencia , Tiroxina/sangre , Tiroxina/deficiencia , Triyodotironina/sangre , Triyodotironina/deficiencia
20.
Commun Comput Phys ; 11(1): 179-214, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21949541

RESUMEN

We consider the design of an effective and reliable adaptive finite element method (AFEM) for the nonlinear Poisson-Boltzmann equation (PBE). We first examine the two-term regularization technique for the continuous problem recently proposed by Chen, Holst, and Xu based on the removal of the singular electrostatic potential inside biomolecules; this technique made possible the development of the first complete solution and approximation theory for the Poisson-Boltzmann equation, the first provably convergent discretization, and also allowed for the development of a provably convergent AFEM. However, in practical implementation, this two-term regularization exhibits numerical instability. Therefore, we examine a variation of this regularization technique which can be shown to be less susceptible to such instability. We establish a priori estimates and other basic results for the continuous regularized problem, as well as for Galerkin finite element approximations. We show that the new approach produces regularized continuous and discrete problems with the same mathematical advantages of the original regularization. We then design an AFEM scheme for the new regularized problem, and show that the resulting AFEM scheme is accurate and reliable, by proving a contraction result for the error. This result, which is one of the first results of this type for nonlinear elliptic problems, is based on using continuous and discrete a priori L(∞) estimates to establish quasi-orthogonality. To provide a high-quality geometric model as input to the AFEM algorithm, we also describe a class of feature-preserving adaptive mesh generation algorithms designed specifically for constructing meshes of biomolecular structures, based on the intrinsic local structure tensor of the molecular surface. All of the algorithms described in the article are implemented in the Finite Element Toolkit (FETK), developed and maintained at UCSD. The stability advantages of the new regularization scheme are demonstrated with FETK through comparisons with the original regularization approach for a model problem. The convergence and accuracy of the overall AFEM algorithm is also illustrated by numerical approximation of electrostatic solvation energy for an insulin protein.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...