Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Small ; 20(29): e2309842, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38431935

RESUMEN

Triple negative breast cancer (TNBC) cells have a high demand for oxygen and glucose to fuel their growth and spread, shaping the tumor microenvironment (TME) that can lead to a weakened immune system by hypoxia and increased risk of metastasis. To disrupt this vicious circle and improve cancer therapeutic efficacy, a strategy is proposed with the synergy of ferroptosis, immunosuppression reversal and disulfidptosis. An intelligent nanomedicine GOx-IA@HMON@IO is successfully developed to realize this strategy. The Fe release behaviors indicate the glutathione (GSH)-responsive degradation of HMON. The results of titanium sulfate assay, electron spin resonance (ESR) spectra, 5,5'-Dithiobis-(2-nitrobenzoic acid (DTNB) assay and T1-weighted magnetic resonance imaging (MRI) demonstrate the mechanism of the intelligent iron atom (IA)-based cascade reactions for GOx-IA@HMON@IO, generating robust reactive oxygen species (ROS). The results on cells and mice reinforce the synergistic mechanisms of ferroptosis, immunosuppression reversal and disulfidptosis triggered by the GOx-IA@HMON@IO with the following steps: 1) GSH peroxidase 4 (GPX4) depletion by disulfidptosis; 2) IA-based cascade reactions; 3) tumor hypoxia reversal; 4) immunosuppression reversal; 5) GPX4 depletion by immunotherapy. Based on the synergistic mechanisms of ferroptosis, immunosuppression reversal and disulfidptosis, the intelligent nanomedicine GOx-IA@HMON@IO can be used for MRI-guided tumor therapy with excellent biocompatibility and safety.


Asunto(s)
Ferroptosis , Imagen por Resonancia Magnética , Ferroptosis/efectos de los fármacos , Imagen por Resonancia Magnética/métodos , Animales , Humanos , Línea Celular Tumoral , Ratones , Especies Reactivas de Oxígeno/metabolismo , Terapia de Inmunosupresión , Microambiente Tumoral/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Neoplasias de la Mama Triple Negativas/diagnóstico por imagen , Femenino , Glutatión/metabolismo
2.
Cell Commun Signal ; 21(1): 14, 2023 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-36670446

RESUMEN

BACKGROUND: Cellular communication network factor 3 (CCN3) has been implicated in the regulation of osteoblast differentiation. However, it is not known if CCN3 can regulate valvular calcification. While macrophages have been shown to regulate valvular calcification, the molecular and cellular mechanisms of this process remain poorly understood. In the present study, we investigated the role of macrophage-derived CCN3 in the progression of calcific aortic valve disease. METHODS: Myeloid-specific knockout of CCN3 (Mye-CCN3-KO) and control mice were subjected to a single tail intravenous injection of AAV encoding mutant mPCSK9 (rAAV8/D377Y-mPCSK9) to induce hyperlipidemia. AAV-injected mice were then fed a high fat diet for 40 weeks. At the conclusion of high fat diet feeding, tissues were harvested and subjected to histologic and pathologic analyses. In vitro, bone marrow-derived macrophages (BMDM) were obtained from Mye-CCN3-KO and control mice and the expression of bone morphogenic protein signaling related gene were verified via quantitative real-time PCR and Western blotting. The BMDM conditioned medium was cocultured with human valvular intersititial cells which was artificially induced calcification to test the effect of the conditioned medium via Western blotting and Alizarin red staining. RESULTS: Echocardiography revealed that both male and female Mye-CCN3-KO mice displayed compromised aortic valvular function accompanied by exacerbated valve thickness and cardiac dysfunction. Histologically, Alizarin-Red staining revealed a marked increase in aortic valve calcification in Mye-CCN3-KO mice when compared to the controls. In vitro, CCN3 deficiency augmented BMP2 production and secretion from bone marrow-derived macrophages. In addition, human valvular interstitial cells cultured with conditioned media from CCN3-deficient BMDMs resulted in exaggerated pro-calcifying gene expression and the consequent calcification. CONCLUSION: Our data uncovered a novel role of myeloid CCN3 in the regulation of aortic valve calcification. Modulation of BMP2 production and secretion in macrophages might serve as a key mechanism for macrophage-derived CCN3's anti-calcification function in the development of CAVD. Video Abstract.


Asunto(s)
Estenosis de la Válvula Aórtica , Calcinosis , Masculino , Femenino , Humanos , Ratones , Animales , Válvula Aórtica/metabolismo , Válvula Aórtica/patología , Estenosis de la Válvula Aórtica/metabolismo , Estenosis de la Válvula Aórtica/patología , Medios de Cultivo Condicionados , Calcinosis/metabolismo , Calcinosis/patología , Células Cultivadas
3.
Age Ageing ; 52(5)2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-37192506

RESUMEN

BACKGROUND: previous studies have focused on the risk of cardiovascular disease (CVD)-related death in individual cancers, adolescents or all cancers. OBJECTIVE: to evaluate the risk of CVD-related death in older patients with cancer. METHODS: older patients with cancer (over 65 years) of 16 cancers diagnosed between 1975 and 2018 were screened out from the Surveillance, Epidemiology and End Results program. The proportion of deaths, competing risk regression models, standardized mortality ratios (SMRs) and absolute excess risks (AERs) were used to assess the risk of CVD-related death. RESULTS: this study included 1,141,675 older patients (median follow-up: 13.5 years). Of the 16 individual cancers, the risk of CVD death exceeded primary neoplasm death in older patients with cancers of the breast, endometrium, vulva, prostate gland, penis and melanoma of the skin over time (high competing risk group). Compared to the general older population, older patients with cancer had higher SMR and AER of CVD-related death (SMR: 1.58-4.23; AER: 21.16-365.89), heart disease-related death (SMR: 1.14-4.16; AER: 16.29-301.68) and cerebrovascular disease-related death (SMR: 1.11-4.66; AER: 3.02-72.43), with the SMR trend varying with CVD-related death competing risk classifications. The risk of CVD-related death in the high-competing risk group was higher than in the low-competing risk group. CONCLUSIONS: for older patients with cancer, six of 16 individual cancers, including breast, endometrium, vulva, prostate gland, penis and melanoma of the skin was at high risk of CVD-related death. Management for long-term cardiovascular risk in older patients with cancer is needed.


Asunto(s)
Enfermedades Cardiovasculares , Cardiopatías , Melanoma , Masculino , Femenino , Humanos , Adolescente , Anciano , Causas de Muerte , Factores de Riesgo
4.
J Mol Cell Cardiol ; 170: 60-74, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35690006

RESUMEN

AIMS: The main therapeutic strategies for coronary artery disease (CAD) are mainly based on the correction of abnormal cholesterol levels; however, residual risks remain. The newly proven gut microbial metabolite trimethylamine N-oxide (TMAO) linked with CAD has broadened our horizons. In this study, we determined the role of proline/serine-rich coiled-coil protein 1 (PSRC1) in TMAO-driven atherosclerosis. METHODS AND RESULTS: We first analyzed the levels of TMAO and PSRC1 in patients with or without atherosclerosis with a target LDL-C < 1.8 mmol/L. Plasma TMAO levels were increased and negatively associated with decreased PSRC1 in peripheral blood mononuclear cells. Animals and in vitro studies showed that TMAO inhibited macrophage PSRC1 expression due to DNA hypermethylation of CpG islands. ApoE-/- mice fed a choline-supplemented diet exhibited reduced PSRC1 expression accompanied by increased atherosclerotic lesions and plasma TMAO levels. We further deleted PSRC1 in apoE-/- mice and PSRC1 deficiency significantly accelerated choline-induced atherogenesis, characterized by increased macrophage infiltration, foam cell formation and M1 macrophage polarization. Mechanistically, we overexpressed and knocked out PSRC1 in cultured macrophages to explore the mechanisms underlying TMAO-induced cholesterol accumulation and inflammation. PSRC1 deletion impaired reverse cholesterol transport and enhanced cholesterol uptake and inflammation, while PSRC1 overexpression rescued the proatherogenic phenotype observed in TMAO-stimulated macrophages, which was partially attributed to sulfotransferase 2B1b (SULT2B1b) inhibition. CONCLUSIONS: Herein, clinical data provide evidence that TMAO may participate in the development of CAD beyond well-controlled LDL-C levels. Our work also suggests that PSRC1 is a negative regulator mediating the unfavorable effects of TMAO-containing diets. Therefore, PSRC1 overexpression and reduced choline consumption may further alleviate atherosclerosis.


Asunto(s)
Aterosclerosis , Leucocitos Mononucleares , Fosfoproteínas , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Colesterol/sangre , LDL-Colesterol/sangre , Colina , Inflamación , Leucocitos Mononucleares/metabolismo , Metilaminas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Fosfoproteínas/genética , Sulfotransferasas
5.
Cancer ; 128(18): 3330-3339, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35872619

RESUMEN

BACKGROUND: The temporal trend of cardiovascular disease (CVD) mortality in patients with classic Hodgkin lymphoma (cHL) throughout follow-up remains unclear. This study aimed to assess this temporal trend in patients with cHL. METHODS: This multicenter cohort included 15,889 patients with cHL diagnosed between 1983 and 2015, covering all ages. The proportional mortality ratio, cumulative incidence of cause-specific mortality accounting for competing risk, standard mortality ratio, and absolute excess risk were analyzed. RESULTS: Among patients in stage I and stage II cHL, the proportional mortality ratio for CVD exceeded that for cHL, after approximately 60 or 120 months of follow-up, respectively. For almost all the patients with stage I or stage II disease, the cumulative incidence of CVD mortality exceeded that of cHL and other neoplasms over time. In recent decades, the risk of cHL mortality declined sharply, but the risk of CVD mortality among patients with cHL declined quite slowly or even remained unchanged among some populations. Patients with stage I or stage II disease experienced a higher risk of CVD mortality than the general population in almost all follow-up intervals. The absolute excess CVD risk among patients in stage I reached 48.5. CONCLUSIONS: The risk of CVD mortality exceeded that of cHL and other neoplasms and became the leading cause of death over time, among patients with stage I or stage II disease. More effective measures should be taken to reduce the risk of CVD mortality. LAY SUMMARY: Among patients with stage I and stage II classic Hodgkin lymphoma (cHL), the proportional mortality ratio of cardiovascular disease (CVD) exceeded that of cHL after approximately 60 or 120 months of follow-up, respectively. For almost all the patients with stage I or stage II disease, the cumulative incidence of CVD mortality exceeded that of cHL and other neoplasms over time. In the past several decades, the risk of cHL mortality declined sharply, but the risk of CVD mortality among patients with cHL declined quite slowly or even unchanged among some populations. CVD exceeded cHL and has become the leading cause of death over time.


Asunto(s)
Enfermedades Cardiovasculares , Enfermedad de Hodgkin , Estudios de Cohortes , Humanos , Incidencia , Factores de Riesgo , Sobrevivientes
6.
Ecotoxicol Environ Saf ; 244: 114057, 2022 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-36084504

RESUMEN

BACKGROUND: Studies have shown that cadmium (Cd) exposure primarily occurs through diet, and Cd ingestion is a risk factor for atherosclerosis (AS). However, the underlying mechanism remains unclear. As a target organ, the gastrointestinal tract may play a key role in Cd-induced AS. Additionally, as curcumin is insoluble in water but stable in the stomach of acidic pH, it may play regulative roles in the gut. OBJECTIVES: We assess the effect of Cd exposure on gut flora, trimethylamine-N-oxide (TMAO) metabolism and macrophage polarization, further investigate whether curcumin protects against Cd-induced AS by remodeling gut microbiota. METHODS AND RESULTS: The results of 16 S rRNA sequencing show that Cd exposure causes diversity reduction and compositional alteration of the microbial community, resulting in the increasing TMAO synthesis, the imbalance of lipid metabolism, and the M1-type macrophage polarization in the mouse model (ApoE-/-) of AS. As a result, the plaque area is increased with Cd exposure, shown by oil red O staining. TMAO synthesis is positively correlated with the concentration of blood Cd, and the dynamics of specific bacteria in this process were revealed at the phylum to genus levels. Moreover, the effects of intestinal flora and TMAO on Cd-induced AS are further confirmed via microbial transplantation from a mouse model not exposed to Cd, as the transplantation decreases plaque area. Finally, the gavage with curcumin reverses the Cd-induced pathological progression via gut flora restoration. CONCLUSIONS: We first demonstrate that Cd exposure worsens the progression of AS via intestinal flora imbalance and increased TMAO synthesis. Curcumin was verified as a potential novel intervention for preventing Cd-induced AS via remodeling gut microbiota. This study elucidates a new approach for treating AS in regions with significant Cd exposure.


Asunto(s)
Aterosclerosis , Curcumina , Microbioma Gastrointestinal , Animales , Apolipoproteínas E , Aterosclerosis/inducido químicamente , Aterosclerosis/prevención & control , Cadmio/toxicidad , Curcumina/farmacología , Macrófagos , Metilaminas , Ratones , Óxidos/farmacología , Agua/farmacología
7.
J Cell Mol Med ; 25(1): 170-183, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33314583

RESUMEN

Yes-associated protein (YAP), a major effector of the Hippo signalling pathway, is widely implicated in vascular pathophysiology processes. Here, we identify a new role of YAP in the regulation of vascular senescence. The inhibition or deficiency and overexpression of YAP were performed in human umbilical vein endothelial cells (HUVECs) and isolated vascular tissues. Cellular and vascular senescence was assessed by analysis of the senescence-associated ß-galactosidase (SA-ß-gal) and expression of senescence markers P16, P21, P53, TERT and TRF1. We found that YAP was highly expressed in old vascular tissues, inhibition and knockdown of YAP decreased senescence, while overexpression of YAP increased the senescence in both HUVECs and vascular tissues. In addition, autophagic flux blockage and mTOR pathway activation were observed during YAP-induced HUVECs and vascular senescence, which could be relieved by the inhibition and knockdown of YAP. Moreover, YAP-promoted cellular and vascular senescence could be relieved by mTOR inhibition. Collectively, our findings indicate that YAP may serve as a potential therapeutic target for ageing-associated cardiovascular disease.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Autofagia , Vasos Sanguíneos/patología , Senescencia Celular , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción/metabolismo , Animales , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratas Sprague-Dawley , Proteínas Señalizadoras YAP
8.
Clin Sci (Lond) ; 135(21): 2483-2502, 2021 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-34643227

RESUMEN

Vascular calcification is highly prevalent in chronic kidney disease (CKD), and is characterized by transdifferentiation from contractile vascular smooth muscle cells (VSMCs) into an osteogenic phenotype. However, no effective and therapeutic option to prevent vascular calcification is yet available. Dihydromyricetin (DMY), a bioactive flavonoid isolated from Ampelopsis grossedentata, has been found to inhibit VSMCs proliferation and the injury-induced neointimal formation. However, whether DMY has an effect on osteogenic differentiation of VSMCs and vascular calcification is still unclear. In the present study, we sought to investigate the effect of DMY on vascular calcification in CKD and the underlying mechanism. DMY treatment significantly attenuated calcium/phosphate-induced calcification of rat and human VSMCs in a dose-dependent manner, as shown by Alizarin Red S staining and calcium content assay, associated with down-regulation of osteogenic markers including type I collagen (COL I), Runt-related transcription factor 2 (RUNX2), bone morphogenetic protein 2 (BMP2) and osteocalcin (OCN). These results were further confirmed in aortic rings ex vivo. Moreover, DMY ameliorated vascular calcification in rats with CKD. Additionally, we found that AKT signaling was activated during vascular calcification, whereas significantly inhibited by DMY administration. DMY treatment significantly reversed AKT activator-induced vascular calcification. Furthermore, inhibition of AKT signaling efficiently attenuated calcification, which was similar to that after treatment with DMY alone, and DMY had a better inhibitory effect on calcification as compared with AKT inhibitor. The present study demonstrated that DMY has a potent inhibitory role in vascular calcification partially by inhibiting AKT activation, suggesting that DMY may act as a promising therapeutic candidate for patients suffering from vascular calcification.


Asunto(s)
Enfermedades de la Aorta/prevención & control , Flavonoles/farmacología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Insuficiencia Renal Crónica/tratamiento farmacológico , Calcificación Vascular/prevención & control , Animales , Aorta/efectos de los fármacos , Aorta/enzimología , Aorta/patología , Enfermedades de la Aorta/enzimología , Enfermedades de la Aorta/etiología , Enfermedades de la Aorta/patología , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Masculino , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/patología , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Ratas Sprague-Dawley , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/enzimología , Insuficiencia Renal Crónica/patología , Transducción de Señal , Calcificación Vascular/enzimología , Calcificación Vascular/etiología , Calcificación Vascular/patología
9.
Arterioscler Thromb Vasc Biol ; 40(3): 751-765, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31941382

RESUMEN

OBJECTIVES: Vascular calcification is highly prevalent in patients with chronic kidney disease. Increased plasma trimethylamine N-oxide (TMAO), a gut microbiota-dependent product, concentrations are found in patients undergoing hemodialysis. However, a clear mechanistic link between TMAO and vascular calcification is not yet established. In this study, we investigate whether TMAO participates in the progression of vascular calcification using in vitro, ex vivo, and in vivo models. Approach and Results: Alizarin red staining revealed that TMAO promoted calcium/phosphate-induced calcification of rat and human vascular smooth muscle cells in a dose-dependent manner, and this was confirmed by calcium content assay. Similarly, TMAO upregulated the expression of bone-related molecules including Runx2 (Runt-related transcription factor 2) and BMP2 (bone morphogenetic protein-2), suggesting that TMAO promoted osteogenic differentiation of vascular smooth muscle cells. In addition, ex vivo study also showed the positive regulatory effect of TMAO on vascular calcification. Furthermore, we found that TMAO accelerated vascular calcification in rats with chronic kidney disease, as indicated by Mico-computed tomography analysis, alizarin red staining and calcium content assay. By contrast, reducing TMAO levels by antibiotics attenuated vascular calcification in chronic kidney disease rats. Interestingly, TMAO activated NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3) inflammasome and NF-κB (nuclear factor κB) signals during vascular calcification. Inhibition of NLRP3 inflammasome and NF-κB signals attenuated TMAO-induced vascular smooth muscle cell calcification. CONCLUSIONS: This study for the first time demonstrates that TMAO promotes vascular calcification through activation of NLRP3 inflammasome and NF-κB signals, suggesting the potential link between gut microbial metabolism and vascular calcification. Reducing the levels of TMAO could become a potential treatment strategy for vascular calcification in chronic kidney disease.


Asunto(s)
Inflamasomas/efectos de los fármacos , Metilaminas/toxicidad , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Osteogénesis/efectos de los fármacos , Calcificación Vascular/inducido químicamente , Adulto , Anciano , Animales , Antibacterianos/farmacología , Aorta Torácica/efectos de los fármacos , Aorta Torácica/metabolismo , Aorta Torácica/patología , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Inflamasomas/metabolismo , Masculino , Metilaminas/metabolismo , Persona de Mediana Edad , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/metabolismo , Transducción de Señal , Arterias Tibiales/efectos de los fármacos , Arterias Tibiales/metabolismo , Arterias Tibiales/patología , Técnicas de Cultivo de Tejidos , Calcificación Vascular/metabolismo , Calcificación Vascular/patología , Calcificación Vascular/prevención & control
10.
Biochem Genet ; 59(5): 1185-1202, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33715137

RESUMEN

Hypertrophic cardiomyopathy (HCM) is one of the most common genetic heart diseases. Its features include abnormal cardiomyocyte hypertrophy, microvascular dysfunction, and increased accumulation of intercellular matrix. We aim to unravel genes associated with the pathogenesis of HCM and provide a potential target for diagnosis and treatment. Key modules were identified by weighted gene co-expression network analysis (WGCNA). A miRNA-mRNA network was constructed with the predicted miRNA and the most likely hub gene was screened out for gene set enrichment analysis (GSEA). The diagnostic capacity of hub gene was verified by receiver operating characteristic (ROC) curves. Single-cell sequencing (sc-RNA seq) data of normal adult hearts were used to further predict the specific cell types expressing the hub gene. WGCNA assigned genes into different modules and found that the genes contained in the red module had the strongest positive correlation with HCM disease. 2.5% of the genes were common between DEG and hub genes. With the miRNA-mRNA network, osteomodulin (OMD) was identified as the most potential hub gene. GSEA showed that OMD was mainly involved in the synthesis of extracellular matrix and had a certain inhibitory effect on the immune system. The expression of OMD in HCM was validated and ROC curve analysis showed that OMD could distinguish HCM from controls with the area under the curve (AUC) > 0.7. The sc-RNA seq revealed that OMD was mainly expressed in the later stages of cardiac fibroblasts, suggesting that OMD may have an effect on fibroblasts, participating in the pathogenesis of HCM. OMD may serve as a biomarker and therapeutic target for HCM in the future.


Asunto(s)
Biomarcadores/análisis , Cardiomiopatía Hipertrófica/patología , Proteínas de la Matriz Extracelular/genética , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Proteoglicanos/genética , Cardiomiopatía Hipertrófica/genética , Perfilación de la Expresión Génica , Humanos , Curva ROC
11.
J Pathol ; 249(4): 461-471, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31397884

RESUMEN

Vascular calcification is prevalent in patients with chronic kidney disease (CKD) and a major risk factor of cardiovascular disease. Vascular calcification is now recognised as a biological process similar to bone formation involving osteogenic differentiation of vascular smooth muscle cells (VSMCs). Cell division cycle 42 (CDC42), a Rac1 family member GTPase, is essential for cartilage development during endochondral bone formation. However, whether CDC42 affects osteogenic differentiation of VSMCs and vascular calcification remains unknown. In the present study, we observed a significant increase in the expression of CDC42 both in rat VSMCs and in calcified arteries during vascular calcification. Alizarin red staining and calcium content assay revealed that adenovirus-mediated CDC42 overexpression led to an apparent VSMC calcification in the presence of calcifying medium, accompanied with up-regulation of bone-related molecules including RUNX2 and BMP2. By contrast, inhibition of CDC42 by ML141 significantly blocked calcification of VSMCs in vitro and aortic rings ex vivo. Moreover, ML141 markedly attenuated vascular calcification in rats with CKD. Furthermore, pharmacological inhibition of AKT signal was shown to block CDC42-induced VSMC calcification. These findings demonstrate for the first time that CDC42 contributes to vascular calcification through a mechanism involving AKT signalling; this uncovered a new function of CDC42 in regulating vascular calcification. This may provide a potential therapeutic target for the treatment of vascular calcification in the context of CKD. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Osteogénesis , Insuficiencia Renal Crónica/complicaciones , Calcificación Vascular/etiología , Proteína de Unión al GTP cdc42/metabolismo , Anciano , Animales , Estudios de Casos y Controles , Células Cultivadas , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Osteogénesis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirazoles/farmacología , Ratas Sprague-Dawley , Insuficiencia Renal Crónica/sangre , Insuficiencia Renal Crónica/enzimología , Insuficiencia Renal Crónica/patología , Transducción de Señal , Sulfonamidas/farmacología , Técnicas de Cultivo de Tejidos , Calcificación Vascular/sangre , Calcificación Vascular/enzimología , Calcificación Vascular/patología
12.
J Cell Biochem ; 120(10): 17273-17282, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31218741

RESUMEN

Consumption of dietary ellagitannins (ETs) has been proven to benefit multiple chronic health disorders including cancers and cardiovascular diseases. Urolithins, gut microbiota metabolites derived from ETs, are considered as the molecules responsible for these health effects. Previous studies have demonstrated that urolithins exhibit antiproliferative effects on prostate, breast, and colon cancers. However, as for hepatocellular carcinoma (HCC), it remains elusive. Herein, we aim to investigate the function of urolithin B (UB), a member of urolithins family, in HCC. The effects of UB on cell viability, cell cycle and apoptosis were evaluated in HCC cells, and we found UB could inhibit the proliferation of HCC cells, which resulted from cell cycle arrest and apoptosis. Furthermore, UB could increase phosphorylated ß-catenin expression and block its translocation from nuclear to cytoplasm, thus inducing the inactivation of Wnt/ß-catenin signaling. Using a xenograft mice model, UB was found to suppress tumor growth in vivo. In conclusion, our data demonstrated that UB could inhibit the proliferation of HCC cells in vitro and in vivo via inactivating Wnt/ß-catenin signaling, suggesting UB could be a promising candidate in the development of anticancer drugs targeting HCC.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Cumarinas/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Hepáticas/tratamiento farmacológico , Proteínas Wnt/antagonistas & inhibidores , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/antagonistas & inhibidores , Animales , Apoptosis , Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Ciclo Celular , Movimiento Celular , Proliferación Celular , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones , Ratones Desnudos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Lab Invest ; 99(3): 346-357, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30068915

RESUMEN

Trimethylamine N-oxide (TMAO), a gut microbe-derived metabolite of dietary choline and other trimethylamine-containing nutrients, has been linked to increased cardiovascular disease risk. It is unknown whether TMAO plays a role in the development of cardiac hypertrophy. Transverse aortic constriction (TAC) was performed to induce cardiac hypertrophy in Sprague-Dawley (SD) rats. We observed that TMAO levels were significantly elevated in SD rats after 6 weeks of TAC, suggesting the potential role of TMAO in regulating cardiac hypertrophy. In cultured cardiomyocytes, TMAO treatment stimulated cardiac hypertrophy, as indicated by increased cell area of cardiomyocytes and expression of hypertrophic markers including atrial natriuretic peptide (ANP) and beta-myosin heavy chain (ß-MHC). Additionally, TMAO treatment induced cardiac hypertrophy and cardiac fibrosis in SD rats. Reducing TMAO synthesis by antibiotics (Abs) attenuated TAC-induced cardiac hypertrophy and fibrosis. Furthermore, pharmacological inhibition of Smad3 by SIS3 significantly reduced the expression of ANP and ß-MHC, and cardiomyocyte cell size in TMAO-treated group. These data for the first time demonstrate that gut microbe-derived metabolite TMAO induces cardiac hypertrophy and fibrosis involving Smad3 signaling, suggesting that inhibition of gut microbes or generation of TMAO may become a potential target for the prevention and treatment of cardiac hypertrophy.


Asunto(s)
Cardiomegalia/etiología , Microbioma Gastrointestinal/fisiología , Metilaminas/metabolismo , Animales , Cardiomegalia/metabolismo , Cardiomegalia/patología , Células Cultivadas , Modelos Animales de Enfermedad , Fibrosis , Masculino , Metilaminas/antagonistas & inhibidores , Metilaminas/toxicidad , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Proteína smad3/antagonistas & inhibidores , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
14.
Inflamm Res ; 68(9): 739-749, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31218443

RESUMEN

OBJECTIVE AND DESIGN: The purpose of this study was to investigate the roles of SMYD3 and STAT3 in chronic lymphocytic leukemia (CLL) and the possible underlying mechanisms. MATERIALS: Blood samples were collected from 20 patients with CLL and 20 hematologically normal donors. Human cell lines K562, HL-60, MEG-1, and BALL-1 were performed in vitro and BALB/c nude mouse was used in subcutaneous tumor experiments. TREATMENT: WP1066 (30 mg/kg) was also injected intratumorally two days after the first lentivirus treatment and then every four days for a total of four injections and 3 µM WP1066 was carried out for 48 h to downregulate STAT3 phosphorylation. METHODS: We performed studies using the human CLL cell line MEG-1 in vitro and nude mouse subcutaneous tumor experiments in vivo. Differential expression of RNAs was determined using qRT-PCR. The CCK-8 assay and colony formation assay were conducted to evaluate cell proliferation. Flow cytometry was performed to assess cell apoptosis. The relative protein levels were detected using western blotting. Chromatin immunoprecipitation (ChIP) assays, luciferase reporter assays and WP1066, a STAT3 inhibitor, were used to explore the regulatory mechanisms of proteases and transcription factors. A subcutaneous tumor model was constructed to verify the results in vivo. RESULTS: SMYD3 and STAT3 expressions positively correlated with the progression of CLL. Upregulation of SMYD3 significantly promoted the proliferation and inhibited the expression of apoptosis-related genes. The results of the ChIP assays and luciferase reporter assays suggested that STAT3 targeted the promoter region of SMYD3 and, thus, promoted SMYD3 transcription. Downregulation of the phosphorylation of STAT3 by WP1066 notably inhibited the binding of STAT3 to the SMYD3 promoter, and subsequently downregulated SMYD3 transcription. The STAT3 inhibitor inhibited CLL cell growth in vivo, and overexpression of SMYD3 promoted CLL cell growth. Furthermore, overexpression of SMYD3 reversed the inhibitory effects of the STAT3 inhibitor on CLL cell growth. CONCLUSIONS: The STAT3-mediated transcription of SMYD3 plays a role in promoting the progression of chronic lymphocytic leukemia.


Asunto(s)
Regulación Leucémica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/metabolismo , Leucemia Linfocítica Crónica de Células B/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Apoptosis , Células HL-60 , N-Metiltransferasa de Histona-Lisina/sangre , Humanos , Células K562 , Leucemia Linfocítica Crónica de Células B/sangre , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Fosforilación , Unión Proteica , Piridinas/farmacología , Factor de Transcripción STAT3/sangre , Tirfostinos/farmacología
15.
Cell Physiol Biochem ; 45(1): 88-107, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29310113

RESUMEN

Human neuropeptide Y (hNPY) is one of the most widely expressed neurotransmitters in the human central and peripheral nervous systems. It consists of 36 highly conserved amino acid residues, and was first isolated from the porcine hypothalamus in 1982. While it is the most recently discovered member of the pancreatic polypeptide family (which includes neuropeptide Y, gut-derived hormone peptide YY, and pancreatic polypeptide), NPY is the most abundant peptide found in the mammalian brain. In order to exert particular functions, NPY needs to bind to the NPY receptor to activate specific signaling pathways. NPY receptors belong to the class A or rhodopsin-like G-protein coupled receptor (GPCR) family and signal via cell-surface receptors. By binding to GPCRs, NPY plays a crucial role in various biological processes, including cortical excitability, stress response, food intake, circadian rhythms, and cardiovascular function. Abnormal regulation of NPY is involved in the development of a wide range of diseases, including obesity, hypertension, atherosclerosis, epilepsy, metabolic disorders, and many cancers. Thus far, five receptors have been cloned from mammals (Y1, Y2, Y4, Y5, and y6), but only four of these (hY1, hY2, hY4, and hY5) are functional in humans. In this review, we summarize the structural characteristics of human NPY receptors and their role in metabolic diseases.


Asunto(s)
Enfermedades Metabólicas/etiología , Receptores de Neuropéptido Y/metabolismo , Diabetes Mellitus/etiología , Diabetes Mellitus/metabolismo , Diabetes Mellitus/terapia , Dislipidemias/etiología , Dislipidemias/metabolismo , Dislipidemias/terapia , Humanos , Hipertensión/etiología , Hipertensión/metabolismo , Hipertensión/terapia , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/terapia , Obesidad/etiología , Obesidad/metabolismo , Obesidad/terapia , Receptores de Neuropéptido Y/química , Receptores de Neuropéptido Y/genética , Transducción de Señal
16.
J Pharmacol Exp Ther ; 366(3): 458-469, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29945930

RESUMEN

Previous evidence has suggested that puerarin may attenuate cardiac hypertrophy; however, the potential mechanisms have not been determined. Moreover, the use of puerarin is limited by severe adverse events, including intravascular hemolysis. This study used a rat model of abdominal aortic constriction (AAC)-induced cardiac hypertrophy to evaluate the potential mechanisms underlying the attenuating efficacy of puerarin on cardiac hypertrophy, as well as the metabolic mechanisms of puerarin involved. We confirmed that puerarin (50 mg/kg per day) significantly attenuated cardiac hypertrophy, upregulated Nrf2, and decreased Keap1 in the myocardium. Moreover, puerarin significantly promoted Nrf2 nuclear accumulation in parallel with the upregulated downstream proteins, including heme oxygenase 1, glutathione transferase P1, and NAD(P)H:quinone oxidoreductase 1. Similar results were obtained in neonatal rat cardiomyocytes (NRCMs) treated with angiotensin II (Ang II; 1 µM) and puerarin (100 µM), whereas the silencing of Nrf2 abolished the antihypertrophic effects of puerarin. The mRNA and protein levels of UGT1A1 and UGT1A9, enzymes for puerarin metabolism, were significantly increased in the liver and heart tissues of AAC rats and Ang II-treated NRCMs. Interestingly, the silencing of Nrf2 attenuated the puerarin-induced upregulation of UGT1A1 and UGT1A9. The results of chromatin immunoprecipitation-quantitative polymerase chain reaction indicated that the binding of Nrf2 to the promoter region of Ugt1a1 or Ugt1a9 was significantly enhanced in puerarin-treated cardiomyocytes. These results suggest that Nrf2 is the key regulator of antihypertrophic effects and upregulation of the metabolic enzymes UGT1A1 and UGT1A9 of puerarin. The autoregulatory circuits between puerarin and Nrf2-induced UGT1A1/1A9 are beneficial to attenuate adverse effects and maintain the pharmacologic effects of puerarin.


Asunto(s)
Cardiomegalia/metabolismo , Cardiomegalia/prevención & control , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Isoflavonas/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Cardiomegalia/genética , Cardiomegalia/patología , Femenino , Glucuronosiltransferasa/genética , Glucuronosiltransferasa/metabolismo , Masculino , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Estrés Oxidativo/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba/efectos de los fármacos
17.
J Cardiovasc Pharmacol ; 70(3): 184-193, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28678055

RESUMEN

The aim of this study was to explore the effect and mechanism of action of resveratrol (RSV) on cardiac function in diabetic cardiomyopathy (DCM). Hyperglycemia-induced apoptosis contributes to the pathogenic changes in DCM. RSV treatment inhibited high glucose-induced apoptosis of neonatal rat ventricular myocytes. Additionally, high glucose decreased cell viability, prevented serine-threonine kinase (Akt) and FoxO3a phosphorylation, and suppressed cytoplasmic translocation of FoxO3a. However, these effects of apoptosis were reversed by 10 µM of RSV. The PI3K inhibitor LY294002 abolished the RSV protective effect in vitro. RSV (5 or 50 mg·kg·d orally for 8 weeks) prevented the deterioration of cardiac function and structural cardiomyopathy in a streptozotocin-induced rat model of diabetes and reduced apoptosis in diabetic myocardium. Furthermore, it restored streptozotocin-impaired phosphorylation of Akt and FoxO3a (p-Akt and p-FoxO3a) and suppressed nuclear translocation of FoxO3a in vivo. Together, these data indicate that RSV has therapeutic potential against DCM by inhibiting apoptosis via the PI3K/Akt/FoxO3a pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Cardiomiopatías Diabéticas/tratamiento farmacológico , Proteína Forkhead Box O3/antagonistas & inhibidores , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Estilbenos/uso terapéutico , Animales , Apoptosis/fisiología , Cardiomiopatías Diabéticas/metabolismo , Modelos Animales de Enfermedad , Proteína Forkhead Box O3/metabolismo , Masculino , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Resveratrol , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Estilbenos/farmacología
18.
BMC Cardiovasc Disord ; 16(1): 225, 2016 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-27855650

RESUMEN

BACKGROUND: Metabolism remodeling has been recognized as an early event following cardiac pressure overload. However, its temporal association with ventricular hypertrophy has not been confirmed. Moreover, whether trimetazidine could favorably affect this process also needs to be determined. The aim of the study was to explore the temporal changes of myocardial metabolism remodeling following pressure-overload induced ventricular hypertrophy and the potential favorable effect of trimetazidine on myocardial metabolism remodeling. METHODS: A rat model of abdominal aortic constriction (AAC)-induced cardiac pressure overload was induced. These rats were grouped as the AAC (no treatment) or TMZ group according to whether oral trimetazidine (TMZ, 40 mg/kg/d, for 5 days) was administered. Changes in cardiac structures were sequentially evaluated via echocardiography. The myocardial ADP/ATP ratio was determined to reflect the metabolic status, and changes in serum neuropeptide Y systems were evaluated. RESULTS: Myocardial metabolic disorder was acutely induced as evidenced by an increased ADP/ATP ratio within 7 days of AAC before the morphological changes in the myocardium, accompanied by up-regulation of serum oxidative stress markers and expression of fetal genes related to hypertrophy. Moreover, the serum NPY and myocardial NPY-1R, 2R, and 5R levels were increased within the acute phase of AAC-induced cardiac pressure overload. Pretreatment with TMZ could partly attenuate myocardial energy metabolic homeostasis, decrease serum levels of oxidative stress markers, attenuate the induction of hypertrophy-related myocardial fetal genes, inhibit the up-regulation of serum NPY levels, and further increase the myocardial expression of NPY receptors. CONCLUSIONS: Cardiac metabolic remodeling is an early change in the myocardium before the presence of typical morphological ventricular remodeling following cardiac pressure overload, and pretreatment with TMZ may at least partly reverse the acute metabolic disturbance, perhaps via regulation of the NPY system.


Asunto(s)
Aorta Abdominal/cirugía , Presión Arterial , Fármacos Cardiovasculares/farmacología , Metabolismo Energético/efectos de los fármacos , Hipertrofia Ventricular Izquierda/prevención & control , Miocardio/metabolismo , Neuropéptido Y/sangre , Receptores de Neuropéptido Y/metabolismo , Trimetazidina/farmacología , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos , Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Aorta Abdominal/fisiopatología , Constricción , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Hipertrofia Ventricular Izquierda/etiología , Hipertrofia Ventricular Izquierda/metabolismo , Hipertrofia Ventricular Izquierda/fisiopatología , Masculino , Miocardio/patología , Estrés Oxidativo/efectos de los fármacos , Ratas Wistar , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropéptido/metabolismo , Transducción de Señal
19.
Biochem Biophys Res Commun ; 464(3): 908-15, 2015 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-26188094

RESUMEN

This study aimed to explore the effects of puerarin on autophagy in cardiac hypertrophy. Decreased 5'-adenosine monophosphate kinase (AMPK) activity alone with inhibited autophagy could be detected in rats within 3 weeks after aortic banding (AB). Puerarin treatment for 3 weeks in AB rats significantly restored autophagy. Administration of puerarin for 6 weeks effectively restricted cardiomyocyte hypertrophy and apoptosis. In an in vitro study, similar anti-hypertrophy and anti-apoptosis effects of puerarin on isoprenaline-induced H9c2 cells were also observed. After inhibition of autophagy by pretreatment with 3-methyladenine, the protective effects of puerarin were blocked. Further in vivo study demonstrated that puerarin significantly enabled phosphorylation of 5'-AMPK to be activated, subsequently inhibiting expression of the mammalian target of rapamycin (mTOR) target proteins S6 ribosomal protein and 4E-binding protein 1. All these data indicate that puerarin exerts protective effects against cardiomyocyte hypertrophy and apoptosis, partly by restoration of autophagy through AMPK/mTOR-mediated signaling.


Asunto(s)
Autofagia/efectos de los fármacos , Cardiomegalia/prevención & control , Isoflavonas/farmacología , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/metabolismo , Adenina/análogos & derivados , Adenina/farmacología , Animales , Cardiomegalia/etiología , Cardiomegalia/patología , Proteínas Portadoras/metabolismo , Modelos Animales de Enfermedad , Péptidos y Proteínas de Señalización Intracelular , Isoproterenol/farmacología , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Fosfoproteínas/metabolismo , Ratas Sprague-Dawley , Proteínas Quinasas S6 Ribosómicas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Remodelación Ventricular/efectos de los fármacos
20.
Tumour Biol ; 36(11): 8697-702, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26047605

RESUMEN

Breast cancer is one of the most common malignancies and a major cause of cancer-related mortality all over the world. A growing body of reports revealed that microRNAs play essential roles in the progression of cancers. Aberrant expression of miR-503 has been reported in several kinds of cancer. The aim of the current study was to elucidate the role of miR-503 in the pathogenesis of breast cancer. In the present study, our results suggested that miR-503 expression was markedly downregulated in breast cancer tissues and cells. Overexpression of miR-503 in breast cancer cell lines reduced cell proliferation through inducing G0/G1 cell cycle arrest by targeting CCND1. Together, our findings provide new knowledge regarding the role of miR-503 in the progression of breast cancer and indicate the role of miR-503 as a tumor suppressor microRNA (miRNA) in breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Proliferación Celular/genética , Ciclina D1/biosíntesis , MicroARNs/biosíntesis , Neoplasias de la Mama/patología , Ciclina D1/genética , Femenino , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , MicroARNs/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA