Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mol Med ; 30(1): 79, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38844847

RESUMEN

BACKGROUND: Increased level of serum cholic acid (CA) is often accompanied with decreased CYP2E1 expression in hepatocellular carcinoma (HCC) patients. However, the roles of CA and CYP2E1 in hepatocarcinogenesis have not been elucidated. This study aimed to investigate the roles and the underlying mechanisms of CYP2E1 and CA in HCC cell growth. METHODS: The proteomic analysis of liver tumors from DEN-induced male SD rats with CA administration was used to reveal the changes of protein expression in the CA treated group. The growth of CA-treated HCC cells was examined by colony formation assays. Autophagic flux was assessed with immunofluorescence and confocal microscopy. Western blot analysis was used to examine the expression of CYP2E1, mTOR, AKT, p62, and LC3II/I. A xenograft tumor model in nude mice was used to examine the role of CYP2E1 in CA-induced hepatocellular carcinogenesis. The samples from HCC patients were used to evaluate the clinical value of CYP2E1 expression. RESULTS: CA treatment significantly increased the growth of HCC cells and promoted xenograft tumors accompanied by a decrease of CYP2E1 expression. Further studies revealed that both in vitro and in vivo, upregulated CYP2E1 expression inhibited the growth of HCC cells, blocked autophagic flux, decreased AKT phosphorylation, and increased mTOR phosphorylation. CYP2E1 was involved in CA-activated autophagy through the AKT/mTOR signaling. Finally, decreased CYP2E1 expression was observed in the tumor tissues of HCC patients and its expression level in tumors was negatively correlated with the serum level of total bile acids (TBA) and gamma-glutamyltransferase (GGT). CONCLUSIONS: CYP2E1 downregulation contributes to CA-induced HCC development presumably through autophagy regulation. Thus, CYP2E1 may serve as a potential target for HCC drug development.


Asunto(s)
Autofagia , Carcinoma Hepatocelular , Proliferación Celular , Ácido Cólico , Citocromo P-450 CYP2E1 , Neoplasias Hepáticas , Animales , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/inducido químicamente , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/inducido químicamente , Humanos , Citocromo P-450 CYP2E1/metabolismo , Citocromo P-450 CYP2E1/genética , Masculino , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Ratas , Proliferación Celular/efectos de los fármacos , Ratones , Ratas Sprague-Dawley , Transducción de Señal , Proteómica/métodos , Modelos Animales de Enfermedad , Ratones Desnudos
2.
Int J Mol Sci ; 24(23)2023 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-38069034

RESUMEN

Hepatocellular carcinoma (HCC) is a common malignant tumor with high mortality. Human phenylalanine tRNA synthetase (PheRS) comprises two α catalytic subunits encoded by the FARSA gene and two ß regulatory subunits encoded by the FARSB gene. FARSB is a potential oncogene, but no experimental data show the relationship between FARSB and HCC progression. We found that the high expression of FARSB in liver cancer is closely related to patients' low survival and poor prognosis. In liver cancer cells, the mRNA and protein expression levels of FARSB are increased and promote cell proliferation and migration. Mechanistically, FARSB activates the mTOR complex 1 (mTORC1) signaling pathway by binding to the component Raptor of the mTORC1 complex to play a role in promoting cancer. In addition, we found that FARSB can inhibit erastin-induced ferroptosis by regulating the mTOR signaling pathway, which may be another mechanism by which FARSB promotes HCC progression. In summary, FARSB promotes HCC progression and is associated with the poor prognosis of patients. FARSB is expected to be a biomarker for early screening and treatment of HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proliferación Celular/genética , Línea Celular Tumoral
3.
Physiol Mol Biol Plants ; 29(1): 35-49, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36733834

RESUMEN

Skp1 (S-phase kinase-associated protein 1) is the core gene of SCF ubiquitin ligase, which mediates protein degradation, thereby regulating biological processes such as cell cycle progression, transcriptional regulation, and signal transduction. A variety of plant Skp1 gene family studies have been reported. However, the almond Skp1 gene family has not yet been studied. In this study, we identified 18 members of the Prunus dulcis PdSkp1 family that were unevenly distributed across six chromosomes of the almond genome. Phylogenetic tree analysis revealed that the PdSkp1 members can be divided into three groups: I, II, and III. PdSkp1 members in each subfamily have relatively conserved motif types and exon/intron numbers. There were three pairs of fragment duplication genes and one pair of tandem repeat genes, and their functions were highly evolutionarily conserved. Transcriptome data showed that PdSkp1 is expressed in almond flower tissues, and that its expression shows significant change during cross-pollination. Fluorescence quantitative results showed that eight PdSkp1 genes had different expression levels in five tissues of almond, i.e., branches, leaves, flower buds, flesh, and cores. In addition, we cloned a PsdSSK1 gene based on PdSkp1. The cloned PsdSSK1 showed the same protein sequence as PdSkp1-12. Results of qPCR and western blot analysis showed high expression of PsdSSK1 in almond pollen. In conclusion, we report the first clone of the key gene SSK1 that controls self-incompatibility in almonds. Our research lays a foundation for future functional research on PdSkp1 members, especially for exploring the mechanism of almond self-incompatibility. Supplementary Information: The online version contains supplementary material available at 10.1007/s12298-023-01278-9.

4.
Cancer Cell Int ; 22(1): 115, 2022 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-35277179

RESUMEN

BACKGROUND: Adaptive resistance and side effects of sorafenib treatment result in unsatisfied survival of patients with hepatocellular carcinoma (HCC). Palmitoyl-protein thioesterase 1 (PPT1) plays a critical role in progression of various cancers. However, its role on prognosis and immune infiltrates in HCC remains unclarified. METHODS: By data mining in the Cancer Genome Atlas databases, the role of PPT1 in HCC were initially investigated. Furthermore, HCC cell lines Hep 3B and Hep 1-6 were treated with DC661 or siRNA against PPT1. The biological function of PPT1 was determined by CCK-8 test, colony formation assay, TUNEL staining, immunofluorescence staining, Western blot test, and PI-Annexin V apoptosis assays in vitro. Animal models of subcutaneous injection were applied to investigate the therapeutic role of targeting PPT1. RESULTS: We found that PPT1 levels were significantly upregulated in HCC tissues compared with normal tissues and were significantly associated with a poor prognosis. Multivariate analysis further confirmed that high expression of PPT1 was an independent risk factor for poor overall survival of HCC patients. We initially found that PPT1 was significantly upregulated in sorafenib-resistant cell lines established in this study. Upon sorafenib treatment, HCC cells acquired adaptive resistance by inducing autophagy. We found that DC661, a selective and potent small-molecule PPT1-inhibitor, induced lysosomal membrane permeability, caused lysosomal deacidification, inhibited autophagy and enhanced sorafenib sensitivity in HCC cells. Interestingly, this sensitization effect was also mediated by the induction mitochondrial pathway apoptosis. In addition, the expression level of PPT1 was associated with the immune infiltration in the HCC tumor microenvironment, and PPT1 inhibitor DC661 significantly enhanced the anti-tumor immune response by promoting dendritic cell maturation and further promoting CD8+ T cell activation. Moreover, DC661 combined with sorafenib was also very effective at treating tumor models in immunized mice. CONCLUSIONS: Our findings suggest that targeting PPT1 with DC661 in combination with sorafenib might be a novel and effective alternative therapeutic strategy for HCC.

5.
J Nanobiotechnology ; 19(1): 355, 2021 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-34717654

RESUMEN

BACKGROUND: Hepatocellular carcinoma (HCC) is a common malignant tumor with high rate of metastasis and recurrence. Although immune checkpoint blockade (ICB) has emerged as a promising type of immunotherapy in advanced HCC, treatment with ICB alone achieves an objective remission rate less than 20%. Thus, combination therapy strategies is needed to improve the treatment response rate and therapeutic effect. METHODS:  A light-triggered disassembly of nanoplatform (TB/PTX@RTK) co-loaded an aggregation induced emission (AIE) photosensitizer (TB) and paclitaxel (PTX) was prepared for on-command drug release and synergistic chemo-photodynamic therapy (chemo-PDT). Nano-micelles were characterized for drug loading content, hydrodynamic size, absorption and emission spectra, reactive oxygen species production, and PTX release from micelles. The targeted fluorescence imaging of TB/PTX@RTK micelles and the synergistic anti-tumor efficacy of TB/PTX@RTK micelles-mediated chemo-PDT combined with anti-PD-L1 were assessed both in vitro and in vivo. RESULTS: The TB/PTX@RTK micelles could specifically accumulate at the tumor site through cRGD-mediated active target and facilitate image-guided PDT for tumor ablation. Once irradiated by light, the AIE photosensitizer of TB could produce ROS for PDT, and the thioketal linker could be cleaved by ROS to precise release of PTX in tumor cells. Chemo-PDT could not only synergistically inhibit tumor growth, but also induce immunogenic cell death and elicit anti-tumor immune response. Meanwhile, chemo-PDT significantly upregulated the expression of PD-L1 on tumor cell surface which could efficiently synergize with anti-PD-L1 monoclonal antibodies to induce an abscopal effect, and establish long-term immunological memory to inhibit tumor relapse and metastasis. CONCLUSION: Our results suggest that the combination of TB/PTX@RTK micelle-mediated chemo-PDT with anti-PD-L1 monoclonal antibodies can synergistically enhance systemic anti-tumor effects, and provide a novel insight into the development of new nanomedicine with precise controlled release and multimodal therapy to enhance the therapeutic efficacy of HCC.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Inmunoterapia/métodos , Neoplasias Hepáticas/tratamiento farmacológico , Nanopartículas/química , Fotoquimioterapia/métodos , Medicina de Precisión/métodos , Animales , Línea Celular Tumoral , Liberación de Fármacos , Humanos , Ratones , Micelas , Nanomedicina , Paclitaxel/farmacología , Fármacos Fotosensibilizantes/farmacología , Especies Reactivas de Oxígeno
6.
Arterioscler Thromb Vasc Biol ; 39(3): e91-e105, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30651001

RESUMEN

Objective- Vascular smooth muscle cells (VSMCs) phenotype modulation is critical for the resolution of vascular injury. Genetic and pharmacological inhibition of PI3Kγ (phosphoinositide 3-kinase γ) exerts anti-inflammatory and protective effects in multiple cardiovascular diseases. This study investigated the role of PI3Kγ and its downstream effector molecules in the regulation of VSMC phenotypic modulation and neointimal formation in response to vascular injury. Approach and Results- Increased expression of PI3Kγ was found in injured vessel wall as well in cultured, serum-activated wild-type VSMCs, accompanied by a reduction in the expression of calponin and SM22α, 2 differentiation markers of VSMCs. However, the injury-induced downregulation of calponin and SM22α was profoundly attenuated in PI3Kγ-/- mice. Pharmacological inhibition and short hairpin RNA knockdown of PI3Kγ (PI3Kγ-KD) markedly attenuated YAP (Yes-associated protein) expression and CREB (cyclic AMP-response element binding protein) activation but improved the downregulation of differentiation genes in cultured VSMCs accompanied by reduced cell proliferation and migration. Mechanistically, activated CREB upregulated YAP transcriptional expression through binding to its promoter. Ectopic expression of YAP strikingly repressed the expression of differentiation genes even in PI3Kγ-KD VSMCs. Moreover, established carotid artery ligation and chimeric mice models demonstrate that deletion of PI3Kγ in naïve PI3Kγ-/- mice as well as in chimeric mice lacking PI3Kγ either in bone marrow or vascular wall significantly reduced neointimal formation after injury. Conclusions- PI3Kγ controls phenotypic modulation of VSMCs by regulating transcription factor CREB activation and YAP expression. Modulating PI3Kγ signaling on local vascular wall may represent a new therapeutic approach to treat proliferative vascular disease.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas de Ciclo Celular/fisiología , Fosfatidilinositol 3-Quinasa Clase Ib/fisiología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/enzimología , Neointima/fisiopatología , Transducción de Señal/fisiología , Animales , Arteria Carótida Común , Movimiento Celular , Células Cultivadas , Fosfatidilinositol 3-Quinasa Clase Ib/deficiencia , Fosfatidilinositol 3-Quinasa Clase Ib/genética , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Ligadura , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos del Músculo Liso/patología , Fenotipo , Interferencia de ARN , ARN Interferente Pequeño/genética , Quimera por Radiación , Remodelación Vascular , Proteínas Señalizadoras YAP
7.
World J Surg Oncol ; 18(1): 282, 2020 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-33121478

RESUMEN

BACKGROUND: Hepatic epithelioid hemangioendothelioma (HEHE) is an extremely rare borderline tumor of vascular endothelial origin. Laparoscopic resection of HEHE has never been reported. METHODS: The clinical data of eleven patients with HEHE (4 women and 7 men) who were diagnosed and treated at the Union Hospital (Wuhan, China), and Wuhan Asia General Hospital (Wuhan, China), between March 2012 and July 2020 were analyzed retrospectively. RESULTS: The mean age of HEHE patients was 42.4 ± 13.9 years (range 22-67 years). All patients underwent laparoscopic surgery alone or in combination with radiofrequency ablation. Most tumors showed aggressive growth or metastasis. By immunohistochemistry, tumor cells were positive for CD31, CD34, ERG, PCK, FLi-1, TFE-3, and Ki-67 (labeling index range, 5-15%). In one of the patients, the tumor was accompanied by partial necrosis with a local appearance of epithelioid angiosarcoma. Postoperative adjuvant treatment included chemotherapy, sorafenib, and Huaier granule. As of July 2020, the median follow-up duration was 36 months (range, 9-60 months), with 2 (18.2%) patients experiencing tumor recurrence. CONCLUSIONS: This is the first report of laparoscopic hepatectomy of HEHE. Curative laparoscopic hepatectomy might be an acceptable treatment for appropriate HEHE patients.


Asunto(s)
Hemangioendotelioma Epitelioide , Laparoscopía , Neoplasias Hepáticas , Adulto , Anciano , Asia , China , Femenino , Hemangioendotelioma Epitelioide/cirugía , Humanos , Neoplasias Hepáticas/cirugía , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/cirugía , Pronóstico , Estudios Retrospectivos , Resultado del Tratamiento , Adulto Joven
8.
Phytother Res ; 33(12): 3163-3176, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31468630

RESUMEN

Dysfunction of the blood-brain barrier (BBB) is a prerequisite for the pathogenesis of many cerebral diseases. Oxidative stress and inflammation are well-known factors accounting for BBB injury. Panax notoginseng saponins (PNS), a clinical commonly used drug against cerebrovascular disease, possess efficient antioxidant and anti-inflammatory activity. In the present study, the protective effects of PNS on lipopolysaccharide (LPS)-insulted cerebral microvascular endothelial cells (bEnd.3) were assessed and the underlying mechanisms were investigated. The results showed that PNS mitigated the decrease of Trans-Endothelial Electrical Resistance, increase of paracellular permeability, and loss of tight junction proteins in bEnd.3 BBB model. Meanwhile, PNS suppressed the THP-1 monocytes adhesion on bEnd.3 monolayer. Moreover, PNS prevented the pro-inflammatory cytokines secretion and reactive oxygen species generation in bEnd.3 cells stimulated with LPS. Mechanism investigations suggested that PNS promoted the Akt phosphorylation, activated Nrf2 antioxidant signaling, and inhibited the NF-κB activation. All the effects of PNS could be abolished by PI3K inhibition at different levels. Taken together, these observations suggest that PNS may act as an extrinsic regulator that activates Nrf2 antioxidant defense system depending on PI3K/Akt and inhibits NF-κB inflammatory signaling to attenuate LPS-induced BBB disruption and monocytes adhesion on cerebral endothelial cells in vitro.


Asunto(s)
Antioxidantes/uso terapéutico , Barrera Hematoencefálica/efectos de los fármacos , Lipopolisacáridos/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Panax notoginseng/química , Saponinas/uso terapéutico , Animales , Antioxidantes/farmacología , Humanos , Ratones , Saponinas/farmacología
9.
J Cell Mol Med ; 22(11): 5518-5532, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30134011

RESUMEN

Autophagy promotes invasion of hepatocarcinoma cells through transforming growth factor (TGF)-ß-dependent epithelial-mesenchymal transition (EMT). This study investigated the mechanism by which autophagy induces TGF-ß-triggered EMT and invasion of hepatocarcinoma cells. Autophagy was induced in HepG2 and BEL7402 cells by starvation in Hank's balanced salt solution. Induction of autophagy degraded phosphodiesterase (PDE) 4A and increased intracellular cAMP, PKA activity and PKA phosphorylation, resulting in increased cAMP response element binding (CREB) phosphorylation in hepatocarcinoma cells. Autophagy-induced activation of cAMP/PKA/CREB signalling further enhanced TGF-ß1 expression, downregulated the expression of epithelial markers and upregulated the expression of mesenchymal markers, accelerating invasion of hepatocarcinoma cells. Inhibition of autophagy by Atg3 and Atg7 knockdown or by chloroquine treatment prevented degradation of PDE4A and activation of cAMP/PKA/CREB signalling, suppressing TGF-ß1 expression, EMT and invasion in hepatocarcinoma cells. In addition, inhibition of cAMP/PKA/CREB signalling also blocked autophagy-induced TGF-ß1 expression and prevented EMT and invasion of hepatocarcinoma cells under starvation. Furthermore, exogenous inhibition of PDE4A or activation of cAMP/PKA/CREB signalling rescued TGF-ß1 expression, EMT and invasion in autophagy-deficient hepatocarcinoma cells. These findings suggest that autophagy induces TGF-ß1 expression and EMT in hepatocarcinoma cells via cAMP/PKA/CREB signalling, which is activated by autophagy-dependent PDE4A degradation.


Asunto(s)
Proteína de Unión a CREB/genética , Carcinoma Hepatocelular/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/genética , Factor de Crecimiento Transformador beta1/genética , Autofagia/genética , Proteína 7 Relacionada con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/genética , Carcinoma Hepatocelular/patología , Proliferación Celular/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Células Epiteliales/metabolismo , Células Epiteliales/patología , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica/genética , Células Hep G2 , Humanos , Neoplasias Hepáticas , Invasividad Neoplásica/genética , ARN Interferente Pequeño/genética , Transducción de Señal/genética , Enzimas Ubiquitina-Conjugadoras/genética
10.
J Cell Biochem ; 118(3): 518-529, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27579780

RESUMEN

The phenotypic modulation of VSMCs is a key cellular event driving neointimal formation and vascular remodeling. As a multifaceted cytokine of cell-mediated immunity, IFN-γ has been shown to play a critical role in the pathogenesis of vascular proliferative diseases. Although the important function of IFN-γ on regulating VSMC activation is well established, the molecular mechanisms by which elicits VSMC responses are poorly defined. Recent studies have identified HMGB1 as a principal effector to mediate IFN-γ-dependent biological functions in multiple cell types. Moreover, SIRT1 has emerged as a critical regulator of cellular processes through deacetylating multiple substrates, including HMGB1. Thus, we examined the role of IFN-γ on HMGB1 release, SIRT1 expression, and VSMC phenotypic modulation as well as the underlying molecular mechanisms. We show that IFN-γ dose-dependently induces HMGB1 cytoplasmic accumulation and its active release from VSMCs, resulting in enhanced HMGB1 in the medium. Conversely, IFN-γ treatment led to a dramatic decrease in SIRT1 expression. Additionally, pretreatment with resveratrol, a selective SIRT1 activator, abrogated IFN-γ-induced HMGB1 translocation and its release. Moreover, IFN-γ stimulates VSMC phenotypic modulation to an activated synthetic state characterized by the repression of SMC differentiation markers such as SM22α and calponin and the increase in cell motility. In contrast, blocking HMGB1 release or activity by resveratrol and HMGB1-neutralizing antibody prevents IFN-γ-induced phenotypic modulation of VSMCs. Overall, this study provides the first evidence showing that HMGB1 plays a critical role in regulating VSMC phenotypic modulation, suggesting that HMGB1 may be a potential therapeutic target to prevent vascular occlusive diseases. J. Cell. Biochem. 118: 518-529, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Proteína HMGB1/metabolismo , Interferón gamma/farmacología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Animales , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Masculino , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Ratas , Ratas Sprague-Dawley , Resveratrol , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/biosíntesis , Estilbenos/farmacología
11.
Med Sci Monit ; 23: 5480-5487, 2017 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-29150595

RESUMEN

BACKGROUND Neural stem cells are reported to exist in the hippocampus of adult mammals and are important sources of neurons for repair. The Notch1 signaling pathway is considered as one of the important regulators of neural stem cells, but its role in adult brains is unclear. We aimed to describe the role of Notch1 signaling in the adult rat hippocampus after traumatic brain injury. MATERIAL AND METHODS The model rats were randomly divided into 4 groups as follows: sham, sham-TBI, sham-Ad-TBI, and NICD-Ad-TBI. We used adenovirus-mediated gene transfection to upregulate endogenous NICD in vivo. Firstly, a TBI rat model was constructed with lateral fluid percussion. Then, the hippocampus was collected to detect the expression of Notch1 markers and stem cell markers (DCX) by Western blot analysis, immunohistochemistry, and immunofluorescence. The prognosis after TBI treatment was evaluated by the Morris Water Maze test. RESULTS First, we found the expression of NICD in vivo was significantly increased by adenovirus-mediated gene transfection as assessed by Notch1 immunofluorescence and Western blot analysis. Second, enhancing NICD stimulated the regeneration of neural stem cells in the DG of the adult rat brain following traumatic brain injury, as evaluated by DCX and NeuN double-staining. Furthermore, Notch1 signaling activation can promote behavioral improvement after traumatic brain injury, including spatial learning and memory capacity. CONCLUSIONS Our findings suggest that targeted regulation of Notch1 signaling may have a useful effect on stem cell transformation. Notch1 signaling may have a potential brain-protection effect, which may result from neurogenesis.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Células-Madre Neurales/metabolismo , Receptor Notch1/metabolismo , Animales , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/fisiopatología , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Proteína Doblecortina , Hipocampo/metabolismo , Hipocampo/fisiología , Masculino , Memoria/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Neurogénesis/fisiología , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Recuperación de la Función/fisiología , Transducción de Señal , Lóbulo Temporal/metabolismo
12.
Biochem Biophys Res Commun ; 463(1-2): 130-6, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26002466

RESUMEN

Stromal-derived Factor-1 (SDF-1) derived from vascular smooth muscle cells (VSMCs) contributes to vascular repair and remodeling in various vascular diseases. In this study, the mechanism underlying regulation of SDF-1 expression by interleukin-1α (IL-1α) was investigated in primary rat VSMCs. We found IL-1α promotes SDF-1 expression by up-regulating CCAAT-enhancer-binding protein ß (C/EBPß) in an IκB kinase ß (IKKß) signaling-dependent manner. Moreover, IL-1α-induced expression of C/EBPß and SDF-1 was significantly potentiated by knockdown of transforming growth factor ß-activated kinase 1 (TAK1), an upstream activator of IKKß signaling. In addition, we also demonstrated that TAK1/p38 mitogen-activated protein kinase (p38 MAPK) signaling exerted negative effect on IL-1α-induced expression of C/EBPß and SDF-1 through counteracting ROS-dependent up-regulation of nuclear factor erythroid 2-related factor 2 (NRF2). In conclusion, TAK1 acts as an important regulator of IL-1α-induced SDF-1 expression in VSMCs, and modulating activity of TAK1 may serve as a potential strategy for modulating vascular repair and remodeling.


Asunto(s)
Quimiocina CXCL12/metabolismo , Interleucina-1alfa/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Miocitos del Músculo Liso/metabolismo , Animales , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Células Cultivadas , Quimiocina CXCL12/genética , Activación Enzimática , Quinasa I-kappa B/metabolismo , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/genética , Sistema de Señalización de MAP Quinasas , Masculino , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
World Neurosurg ; 182: e559-e569, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38061540

RESUMEN

BACKGROUND: This study was aimed to investigate the effects of marital status on overall survival (OS) and cancer-specific survival (CSS) in patients with glioblastoma (GBM) and to develop nomograms for predicting prognosis in GBM patients. METHODS: All patients were selected from the Surveillance, Epidemiology, and End Results cancer registry program. We used propensity score matching to balance the baseline characteristics of married and unmarried patients. The effects of marital status on OS and CSS were then assessed using Kaplan-Meier curves and Cox proportional hazard regression, and the magnitude of each factor was visualized in the form of forest maps. The impact of marriage on the survival of GBM patients was further explored by stratifying several demographic factors. Finally, the nomograms were constructed and verified based on Cox proportional risk regression model. RESULTS: A total of 17,517 patients with GBM (11,818 married patients, 67.5%) were enrolled in the study cohort. After propensity score matching, there were 5699 patients in both the married and unmarried groups. Multivariate Cox regression analysis showed that both married and single patients had better OS (married: hazard ratio [HR] 0.824, 95% confidence interval [CI]: 0.788-0.862, P < 0.001; single: HR 0.764, 95% CI: 0.722-0.808, P < 0.001) and CSS (married: HR 0.833, 95% CI: 0.796-0.872, P < 0.001; single: HR 0.761, 95% CI: 0.718-0.806, P < 0.001) than divorced, separated, and widowed patients. CONCLUSIONS: Marital status was an independent prognostic factor in patients with GBM. The nomograms constructed in this study could help medical professionals to provide personalized prognostic assessment and treatment decisions for patients with GBM.


Asunto(s)
Glioblastoma , Humanos , Pronóstico , Programa de VERF , Estado Civil , Matrimonio , Estimación de Kaplan-Meier
14.
Medicine (Baltimore) ; 103(19): e38091, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38728467

RESUMEN

To screen immune-related prognostic biomarkers in low-grade glioma (LGG), and reveal the potential regulatory mechanism. The differential expressed genes (DEGs) between alive and dead patients were initially identified, then the key common genes between DEGs and immune-related genes were obtained. Regarding the key DEGs associated with the overall survival (OS), their clinical value was assessed by Kaplan-Meier, RCS, logistic regression, ROC, and decision curve analysis methods. We also assessed the role of immune infiltration on the association between key DEGs and OS. All the analyses were based on the TGCA-LGG data. Finally, we conducted the molecular docking analysis to explore the targeting binding of key DEGs with the therapeutic agents in LGG. Among 146 DEGs, only interleukin-6 (IL-6) was finally screened as an immune-related biomarker. High expression of IL-6 significantly correlated with poor OS time (all P < .05), showing a linear relationship. The combination of IL-6 with IDH1 mutation had the most favorable prediction performance on survival status and they achieved a good clinical net benefit. Next, we found a significant relationship between IL-6 and immune microenvironment score, and the immune microenvironment played a mediating effect on the association of IL-6 with survival (all P < .05). Detailly, IL-6 was positively related to M1 macrophage infiltration abundance and its biomarkers (all P < .05). Finally, we obtained 4 therapeutic agents in LGG targeting IL-6, and their targeting binding relationships were all verified. IL6, as an immune-related biomarker, was associated with the prognosis in LGG, and it can be a therapeutic target in LGG.


Asunto(s)
Biomarcadores de Tumor , Neoplasias Encefálicas , Glioma , Interleucina-6 , Microambiente Tumoral , Humanos , Interleucina-6/metabolismo , Interleucina-6/genética , Glioma/inmunología , Glioma/genética , Glioma/mortalidad , Microambiente Tumoral/inmunología , Microambiente Tumoral/genética , Pronóstico , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidad , Biomarcadores de Tumor/genética , Femenino , Estimación de Kaplan-Meier , Regulación Neoplásica de la Expresión Génica
15.
J Int Med Res ; 52(1): 3000605231220867, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38190848

RESUMEN

Ectopic adrenocorticotropic hormone syndrome (EAS) is a rare condition caused by pancreatic neuroendocrine tumors (p-NETs). The severe hypercortisolemia that characterizes EAS is associated with a poor prognosis and survival. Mitotane is the only adrenolytic drug approved by the Food and Drug Administration and is often used to treat adrenocortical carcinoma. Combination therapy with mitotane and other adrenal steroidogenesis inhibitors is common for patients with Cushing's syndrome (CS). Here, we describe three patients who developed EAS secondary to the liver metastasis of p-NETs. All three rapidly developed hypercortisolemia but no typical features of CS. They underwent anti-tumor and mitotane therapy, which rapidly reduced their blood cortisol concentrations and ameliorated their symptoms. Their hypercortisolemia was controlled long term using a low dose of mitotane. The principal adverse effects were a slight loss of appetite and occasional dizziness, and there were no severe adverse effects. Importantly, even when the tumor progressed, the patients' circulating cortisol concentrations remained within the normal range. In summary, the present case series suggests that mitotane could be used to treat hypercortisolemia in patients with EAS caused by advanced p-NETs, in the absence of significant adverse effects.


Asunto(s)
Síndrome de Cushing , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Tumores Neuroendocrinos , Neoplasias Pancreáticas , Estados Unidos , Humanos , Mitotano/uso terapéutico , Hidrocortisona , Síndrome de Cushing/tratamiento farmacológico , Neoplasias Pancreáticas/complicaciones , Neoplasias Pancreáticas/tratamiento farmacológico , Hormona Adrenocorticotrópica
16.
Adv Sci (Weinh) ; 11(14): e2307143, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38308097

RESUMEN

Photodynamic therapy (PDT) is a minimally invasive and controllable local cancer treatment for cholangiocarcinoma (CCA). However, the efficacy of PDT is hindered by intratumoral hypoxia and the presence of an antioxidant microenvironment. To address these limitations, combining PDT with gas therapy may be a promising strategy to enhance tumor oxygenation. Moreover, the augmentation of oxidative damage induced by PDT and gas therapy can be achieved by inhibiting NRF2, a core regulatory molecule involved in the antioxidant response. In this study, an integrated nanotherapeutic platform called CMArg@Lip, incorporating PDT and gas therapies using ROS-responsive liposomes encapsulating the photosensitizer Ce6, the NO gas-generating agent L-arginine, and the NRF2 inhibitor ML385, is successfully developed. The utilization of CMArg@Lip effectively deals with challenges posed by tumor hypoxia and antioxidant microenvironment, resulting in elevated levels of oxidative damage and subsequent induction of ferroptosis in CCA. Additionally, these findings suggest that CMArg@Lip exhibits notable immunomodulatory effects, including the promotion of immunogenic cell death and facilitation of dendritic cell maturation. Furthermore, it contributes to the anti-tumor function of cytotoxic T lymphocytes through the downregulation of PD-L1 expression in tumor cells and the activation of the STING signaling pathway in myeloid-derived suppressor cells, thereby reprogramming the immunosuppressive microenvironment via various mechanisms.


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , Fotoquimioterapia , Humanos , Fotoquimioterapia/métodos , Factor 2 Relacionado con NF-E2/metabolismo , Antioxidantes/farmacología , Colangiocarcinoma/tratamiento farmacológico , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Conductos Biliares Intrahepáticos/metabolismo , Microambiente Tumoral
17.
Adv Sci (Weinh) ; : e2309203, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38837691

RESUMEN

Targeted delivery of glutamine metabolism inhibitors holds promise for cholangiocarcinoma therapy, yet effective delivery vehicles remain a challenge. This study reports the development of a biomimetic nanosystem, termed R-CM@MSN@BC, integrating mesoporous organosilicon nanoparticles with reactive oxygen species-responsive diselenide bonds for controlled release of the glutamine metabolism inhibitor bis-2-(5-phenylacetamido-1,3,4-thiadiazol-2-yl) ethyl sulfide (BPTES) and the photosensitizer Ce6. Erythrocyte membrane coating, engineered with Arg-Gly-Asp (RGD) peptides, not only enhanced biocompatibility but also improved tumor targeting and tissue penetration. Upon laser irradiation, R-CM@MSN@BC executed both photodynamic and glutamine-metabolic therapies, inducing necroptosis in tumor cells and triggering significant immunogenic cell death. Time-of-flight mass cytometry analysis revealed that R-CM@MSN@BC can remodel the immunosuppressive tumor microenvironment by polarizing M1-type macrophages, reducing infiltration of M2-type and CX3CR1+ macrophages, and decreasing T cell exhaustion, thereby increasing the effectiveness of anti-programmed cell death ligand 1 immunotherapy. This strategy proposed in this study presents a viable and promising approach for the treatment of cholangiocarcinoma.

18.
Genes Dis ; 10(5): 1883-1893, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37492717

RESUMEN

Cellular metabolism-induced epigenetic regulation is essential for the maintenance of cellular homeostasis. Nicotinamide N-methyltransferase (NNMT) is emerging as a key point of intersection between cellular metabolism and epigenetic regulation and has a central role in various physiological and pathological processes. NNMT catalyzes the methylation of nicotinamide (NAM) using the universal methyl donor S-adenosyl methionine (SAM) to yield S-adeno-syl-L-homocysteine (SAH) and N1-methylnicotinamide (MNAM), directly linking methylation balance with nicotinamide adenosine dinucleotide (NAD+) contents. NNMT acts on either the SAM-methylation balance or both NAD+ metabolism, depending on the tissue involved or pathological settings where metabolic demand is increased. Under physiological conditions, the liver act as an essential metabolic organ with abundant NNMT expression, while NNMT hepatic function is not mediated by its methyltransferase activity due to other major methyltransferases such as glycine N-methyltransferase (GNMT) in the liver. However, hepatic NNMT, as well as its metabolite is improperly regulated and linked to the worse pathological states in liver diseases, including alcoholic liver disease, non-alcoholic fatty liver disease (NAFLD), liver cirrhosis, and hepatocellular carcinoma (HCC), suggesting a potential role in the process of liver diseases. In this review, we summarize how NNMT regulates cell methylation balance and NAD metabolism, and extensively outline the current knowledge concerning the functions of NNMT in hepatic metabolism including glucose, lipid and energy, with a specific focus on the contribution of NNMT to the pathophysiology of liver-related diseases. NNMT is involved in the development and progression of liver diseases. Understanding the complex NNMT regulatory network and its effects on pathogenesis could provide new therapeutic strategies in the context of liver diseases.

19.
J Cancer Res Clin Oncol ; 149(16): 14901-14910, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37604939

RESUMEN

PURPOSE: To explore the efficiency of a contrast-enhanced CT-based radiomics nomogram integrated with radiomics signature and clinically independent predictors to distinguish mass-like thymic hyperplasia (ml-TH) from low-risk thymoma (LRT) preoperatively. METHODS: 135 Patients with histopathology confirmed ml-TH (n = 65) and LRT (n = 70) were randomly divided into training set (n = 94) and validation set (n = 41) at a ratio of 7:3. The least absolute shrinkage and selection operator (LASSO) algorithm was used to obtain the optimal features. Based on the selected features, four machine learning models, support vector machine (SVM), logistic regression (LR), extreme gradient boosting (XGBOOST), and random forest (RF) were constructed. Multivariate logistic regression was used to establish a radiomics nomogram containing clinically independent predictors and radiomics signature. Receiver operating characteristic (ROC), DeLong test, and calibration curves were used to detect the performance of the radiomics nomogram in training set and validation set. RESULTS: In the validation set, the area under the curve (AUC) value of LR (0.857; 95% CI: 0.741, 0.973) was the highest of the four machine learning models. Radiomics nomogram containing radiomics signature and clinically independent predictors (including age, shape, and net enhancement degree) had better calibration and identification in the training set (AUC: 0.959; 95% CI: 0.922, 0.996) and validation set (AUC: 0.895; 95% CI: 0.795, 0.996). CONCLUSION: We constructed a contrast-enhanced CT-based radiomics nomogram containing clinically independent predictors and radiomics signature as a noninvasive preoperative prediction method to distinguish ml-TH from LRT. The radiomics nomogram we constructed has potential for preoperative clinical decision making.


Asunto(s)
Timoma , Hiperplasia del Timo , Neoplasias del Timo , Humanos , Timoma/diagnóstico por imagen , Nomogramas , Neoplasias del Timo/diagnóstico por imagen , Tomografía Computarizada por Rayos X
20.
Cardiovasc Res ; 119(4): 1030-1045, 2023 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-36464772

RESUMEN

AIMS: The loss of vascular wall cells in allotransplanted arteries is the initial event leading to transplant arteriosclerosis (TA) and ensuing loss of allograft function. Pharmacological agents able to prevent TA are currently lacking. We previously showed that selective inhibition of the immunoproteasome prevented the chronic rejection of renal allografts. However, the role and mechanisms of selective inhibition of a single immunoproteasome subunit to prevent immune-mediated vascular allograft rejection and TA is not clear. METHODS AND RESULTS: The effect and potential mechanism of combined or individual inhibition of peptidolytically active immunoproteasome LMP7 (ß5i) and LMP2 (ß1i) subunits on immune rejection-mediated TA was investigated using the epoxyketone inhibitor ONX 0914, and the recently developed LMP7-selective inhibitor KZR-329 and LMP2-selective inhibitor KZR-504 in a rat aorta transplantation model. We find that co-inhibition of LMP7 and LMP2 in allogeneic recipients significantly suppressed T-cell activation and function by expressing inhibitory surface markers and then activating inhibitory signals. Moreover, co-inhibition of LMP7 and LMP2 substantially reduced the number of immunoglobulin G-secreting cells and plasma cells and production of alloantibodies through activating the unfolded protein response and incapacitating the survival niche of plasma cells in the bone marrow. Consequentially, the accumulation of inflammatory cytokines, complement, and antibodies is reduced and the apoptosis of vascular wall cells decreased in aortic allografts via LMP7 and LMP2 co-inhibition with ONX 0914 treatment or combined KZR-329 and KZR-504 treatment. However, neither individual inhibition of LMP7 by KZR-329 nor individual inhibition of LMP2 by KZR-504 showed suppression of immune rejection and TA. CONCLUSIONS: We define a critical role of LMP7 and LMP2 in TA and strongly propose co-inhibition of both immunoproteasome subunits as promising therapeutic approach to suppress TA and allograft rejection.


Asunto(s)
Arteriosclerosis , Riñón , Ratas , Animales , Riñón/metabolismo , Citocinas/metabolismo , Rechazo de Injerto/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA