Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
J Neurosci ; 44(21)2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38565292

RESUMEN

Glucagon-like peptide-1 (GLP-1) and its analogs are widely used for diabetes treatment. The paraventricular nucleus (PVN) is crucial for regulating cardiovascular activity. This study aims to determine the roles of GLP-1 and its receptors (GLP-1R) in the PVN in regulating sympathetic outflow and blood pressure. Experiments were carried out in male normotensive rats and spontaneously hypertensive rats (SHR). Renal sympathetic nerve activity (RSNA) and mean arterial pressure (MAP) were recorded. GLP-1 and GLP-1R expressions were present in the PVN. PVN microinjection of GLP-1R agonist recombinant human GLP-1 (rhGLP-1) or EX-4 increased RSNA and MAP, which were prevented by GLP-1R antagonist exendin 9-39 (EX9-39) or GLP-1R antagonist 1, superoxide scavenger tempol, antioxidant N-acetylcysteine, NADPH oxidase (NOX) inhibitor apocynin, adenylyl cyclase (AC) inhibitor SQ22536 or protein kinase A (PKA) inhibitor H89. PVN microinjection of rhGLP-1 increased superoxide production, NADPH oxidase activity, cAMP level, AC, and PKA activity, which were prevented by SQ22536 or H89. GLP-1 and GLP-1R were upregulated in the PVN of SHR. PVN microinjection of GLP-1 agonist increased RSNA and MAP in both WKY and SHR, but GLP-1 antagonists caused greater effects in reducing RSNA and MAP in SHR than in WKY. The increased superoxide production and NADPH oxidase activity in the PVN of SHR were augmented by GLP-1R agonists but attenuated by GLP-1R antagonists. These results indicate that activation of GLP-1R in the PVN increased sympathetic outflow and blood pressure via cAMP-PKA-mediated NADPH oxidase activation and subsequent superoxide production. GLP-1 and GLP-1R upregulation in the PVN partially contributes to sympathetic overactivity and hypertension.


Asunto(s)
Péptido 1 Similar al Glucagón , Receptor del Péptido 1 Similar al Glucagón , Hipertensión , Núcleo Hipotalámico Paraventricular , Ratas Endogámicas SHR , Sistema Nervioso Simpático , Animales , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Masculino , Hipertensión/fisiopatología , Hipertensión/metabolismo , Ratas , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/fisiología , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/antagonistas & inhibidores , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Ratas Endogámicas WKY , Ratas Sprague-Dawley
2.
Artículo en Inglés | MEDLINE | ID: mdl-38581321

RESUMEN

Objective: The biomimetic coating on titanium surface affects the adhesion, proliferation, and differentiation of bone cells on the surface of implants. Naringin-7-O-Neohesperidoside (NRG) positively affects the proliferation and differentiation of bone cells, while inhibiting the formation of osteoclasts, thereby affecting the osteogenic effect. This study aimed to construct biomimetic coatings on pure titanium surfaces using layer by layer (LBL) self-assembly of NRGat different concentrations. The effects of the assembled NRG biomimetic coatings on the proliferation and differentiation of mouse preosteoblast cells (MC3T3-E1) in vitro were investigated. The influence of NRG concentration and culture time on MC3T3-E1 cells was also explored. Methods: LBL is a technology that allows for the creation of thin membranes made of polyelectrolytes through electrostatic attraction between polyanions and polycations, which effectively incorporates charged polyelectrolytes onto solid surfaces while preserving their biological activity. Alkaline phosphatase (ALP) plays a crucial role in biomineralization, and its activity is considered as a marker for osteoblast differentiation. Real-time quantitative PCR accurately and quantitatively measures gene expression levels, which reflect the transcriptional activity of genes and thus reflect the proliferation and differentiation of osteoblasts. The research different concentrations of NRG biomimetic coatings (1×10-4 mol/L, 1×10-5 mol/L, 1×10-6 mol/L, and 1×10-7 mol/L) were constructed on titanium surfaces using the LBL self-assembly technique. The control groups included the blank group and the group without drugs. The effects of the coatings on the proliferation of MC3T3-E1 cells were evaluated by ALP activity assay. The differentiation of MC3T3-E1 cells was evaluated by ALP activity assay. Real-time quantitative PCR was performed to detect the gene expressions of OC mRNA, Runx2 mRNA, and Col1a1 mRNA in MC3T3-E1 cells grown on the titanium samples of different experimental groups. Results: The proliferation indices of all NRG concentration groups were higher than those of the groups without drug and blank groups. The highest ALP value was detected at a concentration of 10-4 mol/L. All NRG concentrations upregulated the expression of Col1al mRNA compared to the group without the drug, and the concentrations of 10-5 mol/L and 10-6 mol/L showed statistically significant differences (P < .01). NRG at a concentration of 10-6 mol/L significantly upregulated the expression of Runx2 mRNA (P < .05), while all NRG concentration groups upregulated the expression of OC mRNA. NRG at a concentration of 10-6 mol/L demonstrated a 4 times increase in Runx2 mRNA expression, indicating a significant impact on osteogenic differentiation. Conclusions: NRG biomimetic coatings on titanium surfaces were successfully constructed using the LBL technique. NRG at different concentrations had stronger effects on the proliferation and differentiation of MC3T3-E1 cells compared to the groups without drug and blank groups, with the concentration of 10-6 mol/L demonstrating the best effect. These findings suggest that NRG-loaded biomimetic coatings may enhance the osseointegration of titanium implants, offering promising prospects for dental and orthopedic applications.

3.
Gastroenterology ; 163(2): 481-494, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35489428

RESUMEN

BACKGROUND & AIMS: YES-associated protein (YAP) aberrant activation is implicated in intrahepatic cholangiocarcinoma (iCCA). Transcriptional enhanced associate domain (TEAD)-mediated transcriptional regulation is the primary signaling event downstream of YAP. The role of Wnt/ß-Catenin signaling in cholangiocarcinogenesis remains undetermined. Here, we investigated the possible molecular interplay between YAP and ß-Catenin cascades in iCCA. METHODS: Activated AKT (Myr-Akt) was coexpressed with YAP (YapS127A) or Tead2VP16 via hydrodynamic tail vein injection into mouse livers. Tumor growth was monitored, and liver tissues were collected and analyzed using histopathologic and molecular analysis. YAP, ß-Catenin, and TEAD interaction in iCCAs was investigated through coimmunoprecipitation. Conditional Ctnnb1 knockout mice were used to determine ß-Catenin function in murine iCCA models. RNA sequencing was performed to analyze the genes regulated by YAP and/or ß-Catenin. Immunostaining of total and nonphosphorylated/activated ß-Catenin staining was performed in mouse and human iCCAs. RESULTS: We discovered that TEAD factors are required for YAP-dependent iCCA development. However, transcriptional activation of TEADs did not fully recapitulate YAP's activities in promoting cholangiocarcinogenesis. Notably, ß-Catenin physically interacted with YAP in human and mouse iCCA. Ctnnb1 ablation strongly suppressed human iCCA cell growth and Yap-dependent cholangiocarcinogenesis. Furthermore, RNA-sequencing analysis revealed that YAP/ transcriptional coactivator with PDZ-binding motif (TAZ) regulate a set of genes significantly overlapping with those controlled by ß-Catenin. Importantly, activated/nonphosphorylated ß-Catenin was detected in more than 80% of human iCCAs. CONCLUSION: YAP induces cholangiocarcinogenesis via TEAD-dependent transcriptional activation and interaction with ß-Catenin. ß-Catenin binds to YAP in iCCA and is required for YAP full transcriptional activity, revealing the functional crosstalk between YAP and ß-Catenin pathways in cholangiocarcinogenesis.


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , Proteínas Señalizadoras YAP , beta Catenina , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/patología , Conductos Biliares Intrahepáticos/patología , Carcinogénesis , Colangiocarcinoma/genética , Colangiocarcinoma/patología , Humanos , Ratones , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Señalizadoras YAP/genética , Proteínas Señalizadoras YAP/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
4.
J Hepatol ; 76(1): 123-134, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34464659

RESUMEN

BACKGROUND & AIMS: Mounting evidence implicates the Hippo downstream effectors Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) in hepatocellular carcinoma (HCC). We investigated the functional contribution of YAP and/or TAZ to c-MYC-induced liver tumor development. METHODS: The requirement for YAP and/or TAZ in c-Myc-driven hepatocarcinogenesis was analyzed using conditional Yap, Taz, and Yap;Taz knockout (KO) mice. An hepatocyte-specific inducible TTR-CreERT2 KO system was applied to evaluate the role of YAP and TAZ during tumor progression. Expression patterns of YAP, TAZ, c-MYC, and BCL2L12 were analyzed in human HCC samples. RESULTS: We found that the Hippo cascade is inactivated in c-Myc-induced mouse HCC. Intriguingly, TAZ mRNA levels and activation status correlated with c-MYC activity in human and mouse HCC, but YAP mRNA levels did not. We demonstrated that TAZ is a direct transcriptional target of c-MYC. In c-Myc induced murine HCCs, ablation of Taz, but not Yap, completely prevented tumor development. Mechanistically, TAZ was required to avoid c-Myc-induced hepatocyte apoptosis during tumor initiation. The anti-apoptotic BCL2L12 gene was identified as a novel target regulated specifically by YAP/TAZ, whose silencing strongly suppressed c-Myc-driven murine hepatocarcinogenesis. In c-Myc murine HCC lesions, conditional knockout of Taz, but not Yap, led to tumor regression, supporting the requirement of TAZ for c-Myc-driven HCC progression. CONCLUSIONS: TAZ is a pivotal player at the crossroad between the c-MYC and Hippo pathways in HCC. Targeting TAZ might be beneficial for the treatment of patients with HCC and c-MYC activation. LAY SUMMARY: The identification of novel treatment targets and approaches for patients with hepatocellular carcinoma is crucial to improve survival outcomes. We identified TAZ as a transcriptional target of c-MYC which plays a critical role in c-MYC-dependent hepatocarcinogenesis. TAZ could potentially be targeted for the treatment of patients with c-MYC-driven hepatocellular carcinoma.


Asunto(s)
Carcinoma Hepatocelular/genética , Proteínas de Unión al ADN/genética , Factores de Transcripción/genética , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ/efectos adversos , Proteínas Señalizadoras YAP/efectos adversos , Animales , Carcinoma Hepatocelular/fisiopatología , Proteínas de Unión al ADN/efectos adversos , Proteínas de Unión al ADN/análisis , Modelos Animales de Enfermedad , Redes Reguladoras de Genes/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/fisiopatología , Ratones , Ratones Noqueados , Estadísticas no Paramétricas , Factores de Transcripción/efectos adversos , Factores de Transcripción/análisis , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ/genética , Proteínas Señalizadoras YAP/genética
5.
Am J Pathol ; 191(9): 1651-1667, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34129844

RESUMEN

Intrahepatic cholangiocarcinoma (iCCA) is a lethal malignant neoplasm with limited therapeutic options. Previous studies have found that Notch1 overexpression alone suffices to induce iCCA in the mouse, albeit after long latency. The current study found that activation of the Yes-associated protein (Yap) proto-oncogene occurs during Notch1-driven iCCA progression. After co-expressing activated Notch1 intracellular domain (Nicd) and Yap (YapS127A) in the mouse liver, rapid iCCA formation and progression occurred in Nicd/Yap mice. Mechanistically, an increased expression of amino acid transporters and activation of the mammalian target of rapamycin complex 1 (mTORC1) signaling pathway was detected in Nicd/Yap mouse liver tumors. Significantly, the genetic deletion of Raptor, the major mTORC1 component, completely suppressed iCCA development in Nicd/Yap mice. Elevated expression of Notch1, YAP, amino acid transporters, and members of the mTORC1 pathway was also detected ubiquitously in a collection of human iCCA specimens. Their levels were associated with a poor patient outcome. This study demonstrates that Notch and YAP concomitant activation is frequent in human cholangiocarcinogenesis. Notch and YAP synergize to promote iCCA formation by activating the mTORC1 pathway.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias de los Conductos Biliares/metabolismo , Colangiocarcinoma/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Receptor Notch1/metabolismo , Factores de Transcripción/metabolismo , Adulto , Anciano , Animales , Neoplasias de los Conductos Biliares/patología , Colangiocarcinoma/patología , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Proto-Oncogenes Mas , Proteínas Señalizadoras YAP
6.
Clin Oral Investig ; 26(3): 2491-2502, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35091817

RESUMEN

OBJECTIVES: In order to verify the hypothesis that fibrin glue (FG) is able to seal the area of bone grafting and facilitate bone regeneration. MATERIALS AND METHODS: Twenty-one Sprague-Dawley rats with critical-sized calvarial bone defects were randomly assigned to three groups: (A) co-administrated deproteinized bovine bone (DBB) and autologous bone grafts with FG [fibrin ( +)], (B) co-administrated DBB and autologous bone grafts without FG [fibrin ( -)], and (C) no graft as control. Four weeks and 8 weeks later, micro-CT analysis and histomorphometric analysis were carried out to evaluate following parameters: bone volume fraction (BV/TV), trabecular number (Tb.N), trabecular thickness (Tb.Th) and trabecular separation (Tb.Sp), percentage of new bone area (Pe.NB), average thickness of bone defect (Th.BD), average thickness of basal bone (Th.BB), and percentage of new bone in center of the skull defect (Pe.NBc). RESULTS: BV/TV, Tb.Th, and Tb.N in fibrin ( -) group were significantly higher than that of fibrin ( +) group (p = 0.008, 0.000, 0.007, respectively) and control group (p = 0.004, 0.001, and 0.007, respectively) at 8 weeks. Pe.NB in fibrin ( -) group (33.67 ± 11.72%) was significantly higher than that of fibrin ( +) group (12.33 ± 3.21%) (p = 0.038) and control group (9.66 ± 8.50%) (p = 0.045) at 8 weeks. Pe.NBc in fibrin ( -) group (12.05 ± 3.91%) was significantly higher than that of fibrin ( +) group (4.79 ± 1.21%) (p = 0.005) and control group (0.00 ± 0.00%) (p = 0.000) at 4 weeks. CONCLUSIONS: Administration of both DBB and autograft stimulates calvarial bone defect regeneration, while combination of FG does not additionally accelerate new bone formation. CLINICAL RELEVANCE: The use of fibrin to cement traditional bone graft materials in oral clinical practice requires caution.


Asunto(s)
Regeneración Ósea , Adhesivo de Tejido de Fibrina , Animales , Huesos , Bovinos , Adhesivo de Tejido de Fibrina/farmacología , Ratas , Ratas Sprague-Dawley , Cicatrización de Heridas
7.
Int J Mol Sci ; 23(20)2022 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-36293450

RESUMEN

Asprosin is a newly discovered adipokine that is involved in regulating metabolism. Sympathetic overactivity contributes to the pathogenesis of several cardiovascular diseases. The paraventricular nucleus (PVN) of the hypothalamus plays a crucial role in the regulation of sympathetic outflow and blood pressure. This study was designed to determine the roles and underlying mechanisms of asprosin in the PVN in regulating sympathetic outflow and blood pressure. Experiments were carried out in male adult SD rats under anesthesia. Renal sympathetic nerve activity (RSNA), mean arterial pressure (MAP), and heart rate (HR) were recorded, and PVN microinjections were performed bilaterally. Asprosin mRNA and protein expressions were high in the PVN. The high asprosin expression in the PVN was involved in both the parvocellular and magnocellular regions according to immunohistochemical analysis. Microinjection of asprosin into the PVN produced dose-related increases in RSNA, MAP, and HR, which were abolished by superoxide scavenger tempol, antioxidant N-acetylcysteine (NAC), and NADPH oxidase inhibitor apocynin. The asprosin promoted superoxide production and increased NADPH oxidase activity in the PVN. Furthermore, it increased the cAMP level, adenylyl cyclase (AC) activity, and protein kinase A (PKA) activity in the PVN. The roles of asprosin in increasing RSNA, MAP, and HR were prevented by pretreatment with AC inhibitor SQ22536 or PKA inhibitor H89 in the PVN. Microinjection of cAMP analog db-cAMP into the PVN played similar roles with asprosin in increasing the RSNA, MAP, and HR, but failed to further augment the effects of asprosin. Pretreatment with PVN microinjection of SQ22536 or H89 abolished the roles of asprosin in increasing superoxide production and NADPH oxidase activity in the PVN. These results indicated that asprosin in the PVN increased the sympathetic outflow, blood pressure, and heart rate via cAMP-PKA signaling-mediated NADPH oxidase activation and the subsequent superoxide production.


Asunto(s)
Núcleo Hipotalámico Paraventricular , Superóxidos , Masculino , Ratas , Animales , Núcleo Hipotalámico Paraventricular/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Superóxidos/metabolismo , Adenilil Ciclasas/metabolismo , Antioxidantes/farmacología , Acetilcisteína/farmacología , Ratas Sprague-Dawley , Sistema Nervioso Simpático , Presión Sanguínea , NADPH Oxidasas/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Adipoquinas/metabolismo , ARN Mensajero/metabolismo
8.
Gut ; 70(9): 1746-1757, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33144318

RESUMEN

OBJECTIVE: Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer with limited treatment options. Cabozantinib, an orally bioavailable multikinase inhibitor is now approved by Food and Drug Administration (FDA) for HCC patients. We evaluated the therapeutic efficacy of cabozantinib, either alone or in combination, in vitro and in vivo. DESIGN: Human HCC cell lines and HCC mouse models were used to assess the therapeutic efficacy and targeted molecular pathways of cabozantinib, either alone or in combination with the pan-mTOR inhibitor MLN0128 or the checkpoint inhibitor anti-PD-L1 antibody. RESULTS: Cabozantinib treatment led to stable disease in c-Met/ß-catenin and Akt/c-Met mouse HCC while possessing limited efficacy on Akt/Ras and c-Myc liver tumours. Importantly, cabozantinib effectively inhibited c-MET and ERK activity, leading to decreased PKM2 and increased p21 expression in HCC cells and in c-Met/ß-catenin and Akt/c-Met HCC. However, cabozantinib was ineffective in inhibiting the Akt/mTOR cascade. Intriguingly, a strong inhibition of angiogenesis by cabozantinib occurred regardless of the oncogenic drivers. However, cabozantinib had limited impact on other tumour microenvironment parameters, including tumour infiltrating T cells, and did not induce programmed death-ligand 1 (PD-L1) expression. Combining cabozantinib with MLN0128 led to tumour regression in c-Met/ß-catenin mice. In contrast, combined treatment with cabozantinib and the checkpoint inhibitor anti-PD-L1 antibody did not provide any additional therapeutic benefit in the four mouse HCC models tested. CONCLUSION: c-MET/ERK/p21/PKM2 cascade and VEGFR2-induced angiogenesis are the primary targets of cabozantinib in HCC treatment. Combination therapies with cabozantinib and mTOR inhibitors may be effective against human HCC.


Asunto(s)
Anilidas/uso terapéutico , Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Piridinas/uso terapéutico , Anilidas/administración & dosificación , Animales , Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica , Benzoxazoles/administración & dosificación , Benzoxazoles/uso terapéutico , Línea Celular Tumoral , Femenino , Humanos , Inhibidores de Puntos de Control Inmunológico/administración & dosificación , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias Hepáticas Experimentales/tratamiento farmacológico , Piridinas/administración & dosificación , Pirimidinas/administración & dosificación , Pirimidinas/uso terapéutico , Microambiente Tumoral/efectos de los fármacos
9.
J Hepatol ; 75(4): 888-899, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34052254

RESUMEN

BACKGROUND & AIMS: Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase that is upregulated in many tumor types and is a promising target for cancer therapy. Herein, we elucidated the functional role of FAK in intrahepatic cholangiocarcinoma (iCCA) development and progression. METHODS: Expression levels and activation status of FAK were determined in human iCCA samples. The functional contribution of FAK to Akt/YAP murine iCCA initiation and progression was investigated using conditional Fak knockout mice and constitutive Cre or inducible Cre mice, respectively. The oncogenic potential of FAK was further examined via overexpression of FAK in mice. In vitro cell line studies and in vivo drug treatment were applied to address the therapeutic potential of targeting FAK for iCCA treatment. RESULTS: FAK was ubiquitously upregulated and activated in iCCA lesions. Ablation of FAK strongly delayed Akt/YAP-driven mouse iCCA initiation. FAK overexpression synergized with activated AKT to promote iCCA development and accelerated Akt/Jag1-driven cholangiocarcinogenesis. Mechanistically, FAK was required for YAP(Y357) phosphorylation, supporting the role of FAK as a central YAP regulator in iCCA. Significantly, ablation of FAK after Akt/YAP-dependent iCCA formation strongly suppressed tumor progression in mice. Furthermore, a remarkable iCCA growth reduction was achieved when a FAK inhibitor and palbociclib, a CDK4/6 inhibitor, were administered simultaneously in human iCCA cell lines and Akt/YAP mice. CONCLUSIONS: FAK activation contributes to the initiation and progression of iCCA by inducing the YAP proto-oncogene. Targeting FAK, either alone or in combination with anti-CDK4/6 inhibitors, may be an effective strategy for iCCA treatment. LAY SUMMARY: We found that the protein FAK (focal adhesion kinase) is upregulated and activated in human and mouse intrahepatic cholangiocarcinoma samples. FAK promotes intrahepatic cholangiocarcinoma development, whereas deletion of FAK strongly suppresses its initiation and progression. Combined FAK and CDK4/6 inhibitor treatment had a strong anti-cancer effect in in vitro and in vivo models. This combination therapy might represent a valuable and novel treatment against human intrahepatic cholangiocarcinoma.


Asunto(s)
Proteína-Tirosina Quinasas de Adhesión Focal/efectos adversos , Proteínas Señalizadoras YAP/efectos de los fármacos , Animales , California , Colangiocarcinoma/etiología , Estudios de Cohortes , Modelos Animales de Enfermedad , Proteína-Tirosina Quinasas de Adhesión Focal/administración & dosificación , Ratones , Transducción de Señal/efectos de los fármacos , Proteínas Señalizadoras YAP/administración & dosificación
10.
J Hepatol ; 75(1): 120-131, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33577921

RESUMEN

BACKGROUND & AIMS: Gain of function (GOF) mutations in the CTNNB1 gene are one of the most frequent genetic events in hepatocellular carcinoma (HCC). T-box transcription factor 3 (TBX3) is a liver-specific target of the Wnt/ß-catenin pathway and thought to be an oncogene mediating activated ß-catenin-driven HCC formation. METHODS: We evaluated the expression pattern of TBX3 in human HCC specimens. Tbx3 was conditionally knocked out in murine HCC models by hydrodynamic tail vein injection of Cre together with c-Met and ΔN90-ß-catenin (c-Met/ß-catenin) in Tbx3flox/flox mice. TBX3 was overexpressed in human HCC cell lines to investigate the functions of TBX3 in vitro. RESULTS: A bimodal expression pattern of TBX3 in human HCC samples was detected: high expression of TBX3 in GOF CTNNB1 HCC and downregulation of TBX3 in non-CTNNB1 mutant tumors. High expression of TBX3 was associated with increased differentiation and decreased expression signatures of tumor growth. Using Tbx3flox/flox mice, we found that ablation of Tbx3 significantly accelerates c-Met/ß-catenin-driven HCC formation. Moreover, Tbx3(-) HCC demonstrated increased YAP/TAZ activity. The accelerated tumor growth induced by loss of TBX3 in c-Met/ß-catenin mouse HCC was successfully prevented by overexpression of LATS2, which inhibited YAP/TAZ activity. In human HCC cell lines, overexpression of TBX3 inhibited HCC cell growth as well as YAP/TAZ activation. A negative correlation between TBX3 and YAP/TAZ target genes was observed in human HCC samples. Mechanistically, phospholipase D1 (PLD1), a known positive regulator of YAP/TAZ, was identified as a novel transcriptional target repressed by TBX3. CONCLUSION: Our study suggests that TBX3 is induced by GOF CTNNB1 mutants and suppresses HCC growth by inactivating PLD1, thus leading to the inhibition of YAP/TAZ oncogenes. LAY SUMMARY: TBX3 is a liver-specific target of the Wnt/ß-catenin pathway and thought to be an oncogene in promoting liver cancer development. Herein, we demonstrate that TBX3 is in fact a tumor suppressor gene that restricts liver tumor growth. Strategies which increase TBX3 expression and/or activities may be effective for HCC treatment.


Asunto(s)
Carcinogénesis/genética , Carcinoma Hepatocelular , Neoplasias Hepáticas , beta Catenina , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Descubrimiento de Drogas , Mutación con Ganancia de Función , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor/fisiología , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Ratones , Ratones Noqueados , Fosfolipasa D/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Vía de Señalización Wnt , beta Catenina/genética , beta Catenina/metabolismo
11.
Am J Pathol ; 190(4): 817-829, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32035060

RESUMEN

Liver regeneration is a fundamental biological process required for sustaining body homeostasis and restoring liver function after injury. Emerging evidence demonstrates that cytokines, growth factors, and multiple signaling pathways contribute to liver regeneration. Mammalian target of rapamycin complex 2 (mTORC2) regulates cell metabolism, proliferation and survival. The major substrates for mTORC2 are the AGC family members of kinases, including AKT, SGK, and PKC-α. We investigated the functional roles of mTORC2 during liver regeneration. Partial hepatectomy (PHx) was performed in liver-specific Rictor (the pivotal unit of mTORC2 complex) knockout (RictorLKO) and wild-type (Rictorfl/fl) mice. Rictor-deficient mice were found to be more intolerant to PHx and displayed higher mortality after PHx. Mechanistically, loss of Rictor resulted in decreased Akt phosphorylation, leading to a delay in hepatocyte proliferation and lipid droplets formation along liver regeneration. Overall, these results indicate an essential role of the mTORC2 signaling pathway during liver regeneration.


Asunto(s)
Proliferación Celular , Hepatectomía , Regeneración Hepática , Hígado/citología , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Proteína Asociada al mTOR Insensible a la Rapamicina/fisiología , Animales , Puntos de Control del Ciclo Celular , Femenino , Lípidos/análisis , Hígado/metabolismo , Hígado/cirugía , Masculino , Diana Mecanicista del Complejo 2 de la Rapamicina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Transducción de Señal
12.
Gut ; 69(1): 177-186, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-30954949

RESUMEN

OBJECTIVE: Increased de novo fatty acid (FA) synthesis and cholesterol biosynthesis have been independently described in many tumour types, including hepatocellular carcinoma (HCC). DESIGN: We investigated the functional contribution of fatty acid synthase (Fasn)-mediated de novo FA synthesis in a murine HCC model induced by loss of Pten and overexpression of c-Met (sgPten/c-Met) using liver-specific Fasn knockout mice. Expression arrays and lipidomic analysis were performed to characterise the global gene expression and lipid profiles, respectively, of sgPten/c-Met HCC from wild-type and Fasn knockout mice. Human HCC cell lines were used for in vitro studies. RESULTS: Ablation of Fasn significantly delayed sgPten/c-Met-driven hepatocarcinogenesis in mice. However, eventually, HCC emerged in Fasn knockout mice. Comparative genomic and lipidomic analyses revealed the upregulation of genes involved in cholesterol biosynthesis, as well as decreased triglyceride levels and increased cholesterol esters, in HCC from these mice. Mechanistically, loss of Fasn promoted nuclear localisation and activation of sterol regulatory element binding protein 2 (Srebp2), which triggered cholesterogenesis. Blocking cholesterol synthesis via the dominant negative form of Srebp2 (dnSrebp2) completely prevented sgPten/c-Met-driven hepatocarcinogenesis in Fasn knockout mice. Similarly, silencing of FASN resulted in increased SREBP2 activation and hydroxy-3-methyl-glutaryl-CoA (HMG-CoA) reductase (HMGCR) expression in human HCC cell lines. Concomitant inhibition of FASN-mediated FA synthesis and HMGCR-driven cholesterol production was highly detrimental for HCC cell growth in culture. CONCLUSION: Our study uncovers a novel functional crosstalk between aberrant lipogenesis and cholesterol biosynthesis pathways in hepatocarcinogenesis, whose concomitant inhibition might represent a therapeutic option for HCC.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Colesterol/biosíntesis , Acido Graso Sintasa Tipo I/metabolismo , Ácidos Grasos/biosíntesis , Neoplasias Hepáticas/metabolismo , Animales , Vías Biosintéticas/efectos de los fármacos , Vías Biosintéticas/genética , Carcinogénesis/genética , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Acido Graso Sintasa Tipo I/genética , Femenino , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Genómica , Humanos , Hidroximetilglutaril-CoA Reductasas/genética , Hidroximetilglutaril-CoA Reductasas/metabolismo , Lipidómica , Neoplasias Hepáticas/genética , Masculino , Ratones , Ratones Noqueados , Fosfohidrolasa PTEN/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteína 2 de Unión a Elementos Reguladores de Esteroles/genética , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Transcriptoma
13.
Hepatology ; 70(6): 2003-2017, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-30737831

RESUMEN

Inactivating mutations of axis inhibition protein 1 (AXIN1), a negative regulator of the Wnt/ß-Catenin cascade, are among the common genetic events in human hepatocellular carcinoma (HCC), affecting approximately 10% of cases. In the present manuscript, we sought to define the genetic crosstalk between Axin1 mutants and Wnt/ß-catenin as well as Notch signaling cascades along hepatocarcinogenesis. We discovered that c-MET activation and AXIN1 mutations occur concomitantly in ~3%-5% of human HCC samples. Subsequently, we generated a murine HCC model by means of CRISPR/Cas9-based gene deletion of Axin1 (sgAxin1) in combination with transposon-based expression of c-Met in the mouse liver (c-Met/sgAxin1). Global gene expression analysis of mouse normal liver, HCCs induced by c-Met/sgAxin1, and HCCs induced by c-Met/∆N90-ß-Catenin revealed activation of the Wnt/ß-Catenin and Notch signaling in c-Met/sgAxin1 HCCs. However, only a few of the canonical Wnt/ß-Catenin target genes were induced in c-Met/sgAxin1 HCC when compared with corresponding lesions from c-Met/∆N90-ß-Catenin mice. To study whether endogenous ß-Catenin is required for c-Met/sgAxin1-driven HCC development, we expressed c-Met/sgAxin1 in liver-specific Ctnnb1 null mice, which completely prevented HCC development. Consistently, in AXIN1 mutant or null human HCC cell lines, silencing of ß-Catenin strongly inhibited cell proliferation. In striking contrast, blocking the Notch cascade through expression of either the dominant negative form of the recombinant signal-binding protein for immunoglobulin kappa J region (RBP-J) or the ablation of Notch2 did not significantly affect c-Met/sgAxin1-driven hepatocarcinogenesis. Conclusion: We demonstrated here that loss of Axin1 cooperates with c-Met to induce HCC in mice, in a ß-Catenin signaling-dependent but Notch cascade-independent way.


Asunto(s)
Proteína Axina/fisiología , Carcinoma Hepatocelular/etiología , Neoplasias Hepáticas Experimentales/etiología , Receptores Notch/fisiología , beta Catenina/fisiología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-met/fisiología , Vía de Señalización Wnt/fisiología
14.
Pediatr Blood Cancer ; 67(5): e28221, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32124532

RESUMEN

BACKGROUND: Hepatoblastoma (HB) is the most common pediatric liver malignancy, occurring mainly during the first 4 years of life. Recent studies unraveled the frequent, coordinated activation of Wnt/ß-catenin and YAP/Hippo (where YAP is yes-associated protein) pathways in human HB samples. Furthermore, it was found that concomitant overexpression of activated forms of ß-catenin and YAP in the mouse liver triggers HB formation in YAP/ß-catenin mice. Cyclin-dependent kinases 9 (CDK9) is an elongating kinase, which has been shown to mediate YAP-driven tumorigenesis. The role of CDK9 in HB molecular pathogenesis has not been investigated to date. METHODS: CDK9 expression was determined in human HB lesions, HB cell lines, and YAP/ß-catenin mouse livers. CDK9 was silenced in human HB cell lines and the effects on growth rate and YAP targets were analyzed. Hydrodynamic transfection of YAPS127A and ∆N90-ß-catenin together with either shCdk9 or control shLuc (where Luc is luciferase) plasmids was employed to assess the requirement of Cdk9 for HB development in vivo. RESULTS: Nuclear immunoreactivity for CDK9 protein was more pronounced in human HB samples and YAP/ß-catenin mouse HB tumor tissues than in corresponding surrounding nontumorous liver tissues. CDK9 protein was also expressed in human HB cell lines. Silencing of CDK9 in human HB cell lines did not lead to consistent effects on HB cell growth or YAP target gene expression. Surprisingly, silencing of Cdk9 led to accelerated liver tumorigenesis in YAP/ß-catenin mice. CONCLUSION: CDK9 is not a major downstream mediator of YAP oncogenic function in HB development and progression.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Carcinogénesis , Carcinoma Hepatocelular , Proteínas de Ciclo Celular , Quinasa 9 Dependiente de la Ciclina , Neoplasias Hepáticas Experimentales , Factores de Transcripción , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Quinasa 9 Dependiente de la Ciclina/genética , Quinasa 9 Dependiente de la Ciclina/metabolismo , Células Hep G2 , Humanos , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas Experimentales/metabolismo , Ratones , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Señalizadoras YAP
15.
J Hepatol ; 71(4): 742-752, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31195063

RESUMEN

BACKGROUND & AIMS: The ubiquitin ligase F-box and WD repeat domain-containing 7 (FBXW7) is recognized as a tumor suppressor in many cancer types due to its ability to promote the degradation of numerous oncogenic target proteins. Herein, we aimed to elucidate its role in intrahepatic cholangiocarcinoma (iCCA). METHODS: Herein, we first confirmed that FBXW7 gene expression was reduced in human iCCA specimens. To identify the molecular mechanisms by which FBXW7 dysfunction promotes cholangiocarcinogenesis, we generated a mouse model by hydrodynamic tail vein injection of Fbxw7ΔF, a dominant negative form of Fbxw7, either alone or in association with an activated/myristylated form of AKT (myr-AKT). We then confirmed the role of c-MYC in human iCCA cell lines and its relationship to FBXW7 expression in human iCCA specimens. RESULTS: FBXW7 mRNA expression is almost ubiquitously downregulated in human iCCA specimens. While forced overexpression of Fbxw7ΔF alone did not induce any appreciable abnormality in the mouse liver, co-expression with AKT triggered cholangiocarcinogenesis and mice had to be euthanized by 15 weeks post-injection. At the molecular level, a strong induction of Fbxw7 canonical targets, including Yap, Notch2, and c-Myc oncoproteins, was detected. However, only c-MYC was consistently confirmed as a FBXW7 target in human CCA cell lines. Most importantly, selected ablation of c-Myc completely impaired iCCA formation in AKT/Fbxw7ΔF mice, whereas deletion of either Yap or Notch2 only delayed tumorigenesis in the same model. In human iCCA specimens, an inverse correlation between the expression levels of FBXW7 and c-MYC transcriptional activity was observed. CONCLUSIONS: Downregulation of FBXW7 is ubiquitous in human iCCA and cooperates with AKT to induce cholangiocarcinogenesis in mice via c-Myc-dependent mechanisms. Targeting c-MYC might represent an innovative therapy against iCCA exhibiting low FBXW7 expression. LAY SUMMARY: There is mounting evidence that FBXW7 functions as a tumor suppressor in many cancer types, including intrahepatic cholangiocarcinoma, through its ability to promote the degradation of numerous oncoproteins. Herein, we have shown that the low expression of FBXW7 is ubiquitous in human cholangiocarcinoma specimens. This low expression is correlated with increased c-MYC activity, leading to tumorigenesis. Our findings suggest that targeting c-MYC might be an effective treatment for intrahepatic cholangiocarcinoma.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinogénesis/metabolismo , Proteínas de Ciclo Celular/metabolismo , Colangiocarcinoma , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Neoplasias Hepáticas , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptor Notch2/metabolismo , Animales , Colangiocarcinoma/metabolismo , Colangiocarcinoma/patología , Modelos Animales de Enfermedad , Genes Supresores de Tumor , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones , Terapia Molecular Dirigida , Transducción de Señal , Células Tumorales Cultivadas , Proteínas Señalizadoras YAP
16.
Am J Pathol ; 188(4): 995-1006, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29378174

RESUMEN

Primary liver cancer consists mainly of hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC). A subset of human HCCs expresses a ICC-like gene signature and is classified as ICC-like HCC. The Hippo pathway is a critical regulator of normal and malignant liver development. However, the precise function(s) of the Hippo cascade along liver carcinogenesis remain to be fully delineated. The role of the Hippo pathway in a murine mixed HCC/ICC model induced by activated forms of AKT and Ras oncogenes (AKT/Ras) was investigated. The authors demonstrated the inactivation of Hippo in AKT/Ras liver tumors leading to nuclear localization of Yap and TAZ. Coexpression of AKT/Ras with Lats2, which activates Hippo, or the dominant negative form of TEAD2 (dnTEAD2), which blocks Yap/TAZ activity, resulted in delayed hepatocarcinogenesis and elimination of ICC-like lesions in the liver. Mechanistically, Notch2 expression was found to be down-regulated by the Hippo pathway in liver tumors. Overexpression of Lats2 or dnTEAD2 in human HCC cell lines inhibited their growth and led to the decreased expression of ICC-like markers, as well as Notch2 expression. Altogether, this study supports the key role of the Hippo cascade in regulating the differentiation status of liver tumors.


Asunto(s)
Linaje de la Célula , Neoplasias Hepáticas/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Animales , Conductos Biliares/patología , Carcinogénesis/metabolismo , Carcinogénesis/patología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular , Colangiocarcinoma/metabolismo , Colangiocarcinoma/patología , Femenino , Vía de Señalización Hippo , Humanos , Masculino , Ratones , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Notch/metabolismo , Transcripción Genética , Proteínas ras/metabolismo
17.
BMC Cancer ; 19(1): 343, 2019 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-30975125

RESUMEN

BACKGROUND: Hepatocellular carcinoma (HCC) is a leading cause of cancer related deaths worldwide. The PI3K cascade is one of the major signaling pathways underlying HCC development and progression. Activating mutations of PI3K catalytic subunit alpha (PIK3CA) and/or loss of Pten often occur in human HCCs. Serum and glucocorticoid kinase 3 (SGK3) belongs to the SGK family of AGK kinases and functions in parallel to AKT downstream of PI3K. Previous studies have shown that SGK3 may be the major kinase responsible for the oncogenic potential of PIK3CA helical domain mutants, such as PIK3CA(E545K), but not kinase domain mutants, such as PIK3CA(H1047R). METHODS: We investigated the functional contribution of SGK3 in mediating activated PIK3CA mutant or loss of Pten induced HCC development using Sgk3 knockout mice. RESULTS: We found that ablation of Sgk3 does not affect PIK3CA(H1047R) or PIK3CA(E545K) induced lipogenesis in the liver. Using PIK3CA(H1047R)/c-Met, PIK3CA(E545K)/c-Met, and sgPten/c-Met murine HCC models, we also demonstrated that deletion of Sgk3 moderately delays PIK3CA(E545K)/c-Met driven HCC, while not affecting PIK3CA(H1047R)/c-Met or sgPten/c-Met HCC formation in mice. Similarly, in human HCC cell lines, silencing of SGK3 reduced PIK3CA(E545K) -but not PIK3CA(H1047R)- induced accelerated tumor cell proliferation. CONCLUSION: Altogether, our data suggest that SGK3 plays a role in transducing helical domain mutant PIK3CA signaling during liver tumor development.


Asunto(s)
Carcinoma Hepatocelular/patología , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Neoplasias Hepáticas/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Anciano , Animales , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Proliferación Celular , Fosfatidilinositol 3-Quinasa Clase I/genética , Modelos Animales de Enfermedad , Femenino , Humanos , Hígado/patología , Neoplasias Hepáticas/genética , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Dominios Proteicos/genética , Proteínas Serina-Treonina Quinasas/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal
18.
Chin J Cancer Res ; 30(1): 112-121, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29545725

RESUMEN

OBJECTIVE: Triple-negative breast cancer (TNBC) is highly invasive and metastatic, which is in urgent need of transformative therapeutics. Tubeimu (TBM), the rhizome of Bolbostemma paniculatum (Maxim.) Franquet, is one of the Chinese medicinal herbs used for breast diseases since the ancient times. The present study evaluated the efficacy, especially the anti-metastatic effects of the dichloromethane extract of Tubeimu (ETBM) on TNBC orthotopic mouse models and cell lines. METHODS: We applied real-time imaging on florescent orthotopic TNBC mice model and tested cell migration and invasion abilities with MDA-MB-231 cell line. Digital gene expression sequencing was performed and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis applied to explore the pathways influenced by ETBM. Moreover, quantitative real-time polymerase chain reactions (qRT-PCR) and Western blot were delivered to confirm the gene expression changes. RESULTS: ETBM exhibited noticeable control on tumor metastasis and growth of TNBC tumors with no obvious toxicity. In compliance with this, it also showed inhibition of cell migration and invasion in vitro. Its impact on the changed biological behavior in TNBC may be a result of decreased expression of integrin ß1 (ITGß1), integrin ß8 (ITGß8) and Rho GTPase activating protein 5 (ARHGAP5), which disabled the focal adhesion pathway and caused change in cell morphology. CONCLUSIONS: This study reveals that ETBM has anti-metastatic effects on MDA-MB-231-GFP tumor and may lead to a new therapeutic agent for the integrative treatment of highly invasive TNBC.

20.
Int J Mol Sci ; 17(3): 334, 2016 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-26950123

RESUMEN

The repair of infected bone defects is still challenging in the fields of orthopedics, oral implantology and maxillofacial surgery. In these cases, the self-healing capacity of bone tissue can be significantly compromised by the large size of bone defects and the potential/active bacterial activity. Infected bone defects are conventionally treated by a systemic/local administration of antibiotics to control infection and a subsequent implantation of bone grafts, such as autografts and allografts. However, these treatment options are time-consuming and usually yield less optimal efficacy. To approach these problems, novel biomaterials with both antibacterial and osteoinductive properties have been developed. The antibacterial property can be conferred by antibiotics and other novel antibacterial biomaterials, such as silver nanoparticles. Bone morphogenetic proteins are used to functionalize the biomaterials with a potent osteoinductive property. By manipulating the carrying modes and release kinetics, these biomaterials are optimized to maximize their antibacterial and osteoinductive functions with minimized cytotoxicity. The findings, in the past decade, have shown a very promising application potential of the novel biomaterials with the dual functions in treating infected bone defects. In this review, we will summarize the current knowledge of novel biomaterials with both antibacterial and osteoinductive properties.


Asunto(s)
Antibacterianos/uso terapéutico , Materiales Biocompatibles/uso terapéutico , Proteínas Morfogenéticas Óseas/uso terapéutico , Regeneración Ósea/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Animales , Antibacterianos/administración & dosificación , Antibacterianos/farmacología , Infecciones Bacterianas/complicaciones , Infecciones Bacterianas/tratamiento farmacológico , Materiales Biocompatibles/administración & dosificación , Materiales Biocompatibles/farmacología , Proteínas Morfogenéticas Óseas/administración & dosificación , Proteínas Morfogenéticas Óseas/farmacología , Huesos/efectos de los fármacos , Huesos/lesiones , Huesos/microbiología , Huesos/fisiopatología , Sistemas de Liberación de Medicamentos/métodos , Humanos , Cicatrización de Heridas/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda