Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Cell Signal ; 121: 111282, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38971568

RESUMEN

Diabetic kidney disease (DKD) is often featured with redox dyshomeostatis. Pyruvate dehydrogenase kinase 4 (PDK4) is the hub for DKD development. However, the mechanism by which PDK4 mediates DKD is poorly understood. The current work aimed to elucidate the relationship between PDK4 and DKD from the perspective of redox manipulation. Oxidative stress was observed in the human proximal tubular cell line (HK-2 cells) treated with a high concentration of glucose and palmitic acid (HGL). The mechanistic study showed that PDK4 could upregulate Kelch-like ECH-associated protein 1 (Keap1) in HGL-treated HK-2 cells through the suppression of autophagy, resulting in the depletion of nuclear factor erythroid 2-related factor 2 (Nrf2), the master regulator of redox homeostasis. At the cellular level, pharmacological inhibition or genetic knockdown of PDK4 could boost Nrf2, followed by the increase of a plethora of antioxidant enzymes and ferroptosis-suppression enzymes. Meanwhile, the inhibition or knockdown of PDK4 remodeled iron metabolism, further mitigating oxidative stress and lipid peroxidation. The same trend was observed in the DKD mice model. The current work highlighted the role of PDK4 in the development of DKD and suggested that PDK4 might be a promising target for the management of DKD.

2.
Int J Biol Macromol ; 275(Pt 1): 133427, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38936586

RESUMEN

Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is an disease characterized by pulmonary edema and widespread inflammation, leading to a notably high mortality rate. The dysregulation of both pro-inflammatory and anti-inflammatory systems, results in cytokine storm (CS), is intricately associated with the development of ALI/ARDS. Tetrastigma hemsleyanum polysaccharide (THP) exerts remarkable anti-inflammatory and immunomodulatory effects against the disease, although its precise role in pathogenesis remains unclear. In the present study, an ALI/ARDS model was established using bacterial lipopolysaccharides. THP administration via aerosol inhalation significantly mitigated lung injury, reduced the number of inflammatory cells, and ameliorated glycerophospholipid metabolism. Furthermore, specific CS-related pathways were investigated by examining the synergy between tumor necrosis factor-α and interferon-γ used to establish CS models. The results indicated that THP effectively decreased inflammatory damage and cell death. The RNA sequencing revealed the involvement of the Janus kinase (JAK) 2-signal transducers and activators of transcription (STAT) signaling pathway in exerting the mentioned effects. Additionally, THP inhibited the activation of the JAK-STAT pathway, thereby alleviating the CS both in vivo and in vitro. Overall, THP exhibited marked therapeutic potential against ALI/ARDS and CS, primarily by targeting the IFN-γ-JAK2/STAT signaling pathway.

3.
Adv Healthc Mater ; : e2400856, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38744431

RESUMEN

Dysregulated skin microbiota and compromised immune responses are the major etiological factors for non-healing diabetic wounds. Current antibacterial strategies fail to orchestrate immune responses and indiscriminately eradicate bacteria at the wound site, exacerbating the imbalance of microbiota. Drawing inspiration from the beneficial impacts that probiotics possess on microbiota, a living microecological hydrogel containing Lactobacillus plantarum and fructooligosaccharide (LP/FOS@Gel) is formulated to remodel dysregulated skin microbiota and reinstate compromised immune responses, cultivating a conducive environment for optimal wound healing. LP/FOS@Gel acts as an "evocator," skillfully integrating the skin microecology, promoting the proliferation of Lactobacillus, Ralstonia, Muribaculum, Bacillus, and Allobaculum, while eradicating colonized pathogenic bacteria. Concurrently, LP/FOS@Gel continuously generates lactic acid to elicit a reparative macrophage response and impede the activation of the nuclear factor kappa-B pathway, effectively alleviating inflammation. As an intelligent microecological system, LP/FOS@Gel reinstates the skin's sovereignty during the healing process and effectively orchestrates the harmonious dialogue between the host immune system and microorganisms, thereby fostering the healing of diabetic infectious wounds. These remarkable attributes render LP/FOS@Gel highly advantageous for pragmatic clinical applications.

4.
Biomed Pharmacother ; 173: 116353, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38432128

RESUMEN

Peiminine, the primary biologically active compound from Fritillaria thunbergii Miq., has demonstrated significant pharmacological activities. Doxorubicin is one of the most potent chemotherapeutic agents for breast cancer (BC). This study was designed to investigate the efficacy and underlying mechanisms of Peiminine combined with Doxorubicin in treating BC. Our results demonstrated that the combination of Peiminine and 1 mg/kg Doxorubicin exhibited more significant suppression of tumor growth compared with the monotherapy in MDA-MB-231 xenograft nude mice model, which is comparable to the effect of 3 mg/kg Doxorubicin in vivo. Notably, the 3 mg/kg Doxorubicin monotherapy resulted in organ toxicity, specifically in the liver and heart, whereas no toxicity was observed in the combination group. In vitro, this combined treatment exhibited a synergistic reduction on the viability of BC cells. Peiminine enhanced the cell cycle arrest and DNA damage induced by Doxorubicin. Furthermore, the combination treatment effectively blocked DNA repair by inhibiting the MAPKs signaling pathways. And ZEB1 knockdown attenuated the combined effect of Peiminine and Doxorubicin on cell viability and DNA damage. In conclusion, our study found that the combination of Peiminine and Doxorubicin showed synergistic inhibitory effects on BC both in vivo and in vitro through enhancing Doxorubicin-induced DNA damage. These findings support that their combination is a novel and promising therapeutic strategy for treating BC.


Asunto(s)
Neoplasias de la Mama , Cevanas , Ratones , Animales , Humanos , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Ratones Desnudos , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Aductos de ADN/farmacología , Aductos de ADN/uso terapéutico , Línea Celular Tumoral , Apoptosis , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
5.
Environ Sci Technol ; 58(9): 4247-4256, 2024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38373403

RESUMEN

Nitrous acid (HONO) is an important source of hydroxyl radicals (OH) in the atmosphere. Precise determination of the absolute ultraviolet (UV) absorption cross section of gaseous HONO lays the basis for the accurate measurement of its concentration by optical methods and the estimation of HONO loss rate through photolysis. In this study, we performed a series of laboratory and field intercomparison experiments for HONO measurement between striping coil-liquid waveguide capillary cell (SC-LWCC) photometry and incoherent broadband cavity-enhanced absorption spectroscopy (IBBCEAS). Specified HONO concentrations prepared by an ultrapure standard HONO source were utilized for laboratory intercomparisons. Results show a consistent ∼22% negative bias in measurements of the IBBCEAS compared with a SC-LWCC photometer. It is confirmed that the discrepancies occurring between these techniques are associated with the overestimation of the absolute UV absorption cross sections through careful analysis of possible uncertainties. We quantified the absorption cross section of gaseous HONO (360-390 nm) utilizing a custom-built IBBCEAS instrument, and the results were found to be 22-34% lower than the previously published absorption cross sections widely used in HONO concentration retrieval and atmospheric chemical transport models (CTMs). This suggests that the HONO concentrations retrieved by optical methods based on absolute absorption cross sections may have been underestimated by over 20%. Plus, the daytime loss rate and unidentified sources of HONO may also have evidently been overestimated in pre-existing studies. In summary, our findings underscore the significance of revisiting the absolute absorption cross section of HONO and the re-evaluation of the previously reported HONO budgets.


Asunto(s)
Contaminantes Atmosféricos , Ácido Nitroso , Ácido Nitroso/análisis , Gases/análisis , Contaminantes Atmosféricos/análisis , Análisis Espectral , Fotólisis
6.
Front Pharmacol ; 14: 1152611, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37251320

RESUMEN

The cardioprotective effects of sodium-glucose cotransporter type 2 (SGLT2) inhibitors have been demonstrated in many studies. However, their benefits for end-stage kidney disease patients, particularly those on peritoneal dialysis, remain unclear. SGLT2 inhibition has shown peritoneal protective effects in some studies, but the mechanisms are still unknown. Herein, we investigated the peritoneal protective mechanisms of Canagliflozin in vitro by simulating hypoxia with CoCl2 in human peritoneal mesothelial cells (HPMCs) and rats by intraperitoneal injection of 4.25% peritoneal dialysate simulating chronic high glucose exposure. CoCl2 hypoxic intervention significantly increased HIF-1α abundance in HPMCs, activated TGF-ß/p-Smad3 signaling, and promoted the production of fibrotic proteins (Fibronectin, COL1A2, and α-SMA). Meanwhile, Canagliflozin significantly improved the hypoxia of HPMCs, decreased HIF-1α abundance, inhibited TGF-ß/p-Smad3 signaling, and decreased the expression of fibrotic proteins. Five-week intraperitoneal injection of 4.25% peritoneal dialysate remarkably increased peritoneal HIF-1α/TGF-ß/p-Smad3 signaling and promoted peritoneal fibrosis and peritoneal thickening. At the same time, Canagliflozin significantly inhibited the HIF-1α/TGF-ß/p-Smad3 signaling, prevented peritoneal fibrosis and peritoneal thickening, and improved peritoneal transportation and ultrafiltration. High glucose peritoneal dialysate increased the expression of peritoneal GLUT1, GLUT3 and SGLT2, all of which were inhibited by Canagliflozin. In conclusion, we showed that Canagliflozin could improve peritoneal fibrosis and function by ameliorating peritoneal hypoxia and inhibiting the HIF-1α/TGF-ß/p-Smad3 signaling pathway, providing theoretical support for the clinical use of SGLT2 inhibitors in patients on peritoneal dialysis.

7.
Comput Biol Med ; 150: 106131, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36195046

RESUMEN

BACKGROUND: Breast cancer is the most aggressive malignant tumor with high morbidity and mortality. Astragalin, a flavonoid widely found in a variety of edible and medicinal plants, is recorded to possess multiple biological and pharmacological activities. However, its effect of anti-breast cancer has been unknown. METHODS: Computational pharmacology was employed to explore the potential mechanism of anti-metastasis and anti-angiogenesis effects of Astragalin on breast cancer. The targets of Astragalin were obtained from TCMSP, Swiss Target Prediction, SEA, BATMAN-TCM, ChemMapper and STITCH databases, and targets of breast cancer were got from OMIM, GeneCards, and DisGeNET databases. Protein-protein interaction network (PPI), Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed to elucidate the interactions of these two groups of targets. Moreover, the anti-metastasis and anti-angiogenesis effects of Astragalin were validated by in vitro and in vivo experiments using wound healing assay, transwell migration and invasion assay, gelatin zymography assay, tube formation assay, and chick embryo chorioallantoic membrane model. RESULTS: Computational pharmacology analysis indicated that the effects of Astragalin against breast cancer were mainly related to the regulation of the cell movement, migration, and angiogenesis, and taking AKT, ZEB1, VEGF, and MMP9 as the promising targets. Further experimental pharmacology indicated that Astragalin exerted anti-metastasis and anti-angiogenesis activities on breast cancer, and verified AKT, ZEB1, VEGF, and MMP9 as the key targets. CONCLUSION: Astragalin suppresses the metastasis and angiogenesis of breast cancer, and AKT, ZEB1, VEGF, and MMP9 are the promising targets for Astragalin against breast cancer. Thus, Astragalin is a potential therapeutic agent for breast cancer.


Asunto(s)
Neoplasias de la Mama , Medicamentos Herbarios Chinos , Embrión de Pollo , Animales , Humanos , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/uso terapéutico , Metaloproteinasa 9 de la Matriz , Proteínas Proto-Oncogénicas c-akt , Factor A de Crecimiento Endotelial Vascular , Simulación del Acoplamiento Molecular
8.
Phytomedicine ; 101: 154143, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35537248

RESUMEN

BACKGROUND: Acute lung injury (ALI) is a complex pulmonary destructive disease with limited therapeutic approaches. Hydnocarpin D (HD) is a flavonolignan isolated from Hydnocarpus wightiana which possesses antioxidant and anti-inflammatory properties. However, whether HD has beneficial effects on ALI as well as its underlying mechanism remains to be elucidated. PURPOSE: This study evaluated the protective effect of HD in ALI and the underlying molecular mechanisms. METHODS: In vivo, the role of HD on lipopolysaccharide (LPS)-induced ALI in mice was tested by determination of neutrophil infiltration, levels of inflammatory cytokines, lung histology and edema, vascular and alveolar barrier disruption. In vitro, murine macrophage RAW 264.7 cells were used to investigate the molecular mechanisms RESULTS: Administration of HD protected mice against LPS-induced ALI, including ameliorating the histological alterations in the lung tissues, and decreasing lung edema, protein content of bronchoalveolar lavage fluid, infiltration of inflammatory cell and secretion of cytokines. Moreover, HD blocked the phosphorylation of TLR-4, NF-κB, and ERK in LPS-induced lung injury. In vitro, HD inhibited LPS-induced oxidative stress and inflammation in RAW 264.7 cells, which largely depend upon the upregulation of antioxidant defensive Nrf2 pathway, thereby suppressing LPS-activated proinflammatory mediator secretion, NLRP3 inflammasome, and MAPK/NF-κB signaling pathway. CONCLUSION: HD attenuates oxidative stress and inflammation against LPS-induced ALI via MAPK/NF-κB and Keap1/Nrf2/HO-1 pathway, and is a promising novel therapeutic candidate for ALI.


Asunto(s)
Lesión Pulmonar Aguda , Flavonolignanos , Animales , Ratones , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/metabolismo , Antioxidantes/metabolismo , Citocinas/metabolismo , Inflamación/tratamiento farmacológico , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Lipopolisacáridos/farmacología , Pulmón , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo
9.
Metabolism ; 131: 155177, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35218794

RESUMEN

INTRODUCTION: Ras guanine nucleotide-releasing protein-4 (RasGRP4) is an activator of Ras protein, which plays significant roles in both the inflammatory response and immune activation. This study determined the role of RasGRP4 in diabetic kidney disease (DKD) progression. METHODS: CRISPR/Cas9 technology was used to establish RasGRP4 knockout (KO) mice. Diabetes was induced by a high-fat diet combined with five consecutive daily intraperitoneal injections of streptozotocin (60 mg/kg) in C57BL/6J wild-type (WT) mice and RasGRP4 KO mice. Hematoxylin and eosin, periodic acid-Schiff, and Masson's trichrome staining were used to observe the histology of pathological injury. Immunohistochemical staining was used to analyze inflammatory cell infiltration. Quantitative PCR and Western blotting were used to detect the expression of inflammatory mediators and the activation of signaling pathways in renal tissues. In vitro cell co-culture experiments were performed to explore the interactions between peripheral blood mononuclear cells (PBMCs) and glomerular endothelial cells (GEnCs). RESULTS: RasGRP4 KO mice developed less severe diabetic kidney injury compared to WT mice, exhibiting lower proteinuria, reduced CD3+ T lymphocyte and F4/80+ macrophage infiltration, less inflammatory mediator expression including interleukin 6, tumor necrosis alpha, intercellular adhesion molecule-1, and vascular cell adhesion molecule-1, and lower expression levels of critical signal transduction molecules in the NLR family pyrin domain-containing 3 inflammasome and mitogen-activated protein kinase (MAPK)/nuclear factor kappa B (NF-κB) signaling pathways in the diabetic kidney. In vitro experiments showed that the adhesion function of PBMCs of RasGRP4 KO mice was reduced compared to that of WT mice. Moreover, the expression of adhesion molecules and critical signal transduction molecules in the NLRP3 inflammasome and MAPK/NF-κB signaling pathways in GEnCs was stimulated by the supernatant of PBMCs, which were derived from RasGRP4 KO mice treated with high glucose and were also significantly reduced compared to those derived from WT mice. CONCLUSION: RasGRP4 promotes the inflammatory injury mediated by PBMCs in diabetes, probably by regulating the interaction between PBMCs and GEnCs and further activating the NLRP3 inflammasome and MAPK/NF-κB signaling pathways.


Asunto(s)
Diabetes Mellitus Tipo 2 , Nefropatías Diabéticas , Animales , Células Endoteliales/metabolismo , Femenino , Nucleótidos de Guanina , Humanos , Inflamasomas/metabolismo , Leucocitos Mononucleares/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Factores de Intercambio de Guanina Nucleótido ras/metabolismo
10.
Zhongguo Zhong Yao Za Zhi ; 46(17): 4488-4496, 2021 Sep.
Artículo en Chino | MEDLINE | ID: mdl-34581054

RESUMEN

This study focused on the ameliorative effects of gypenosides(GPS) on insulin sensitivity and inflammatory factors in rats with type 2 diabetes mellitus(T2 DM) and explored their possible molecular mechanisms. After the successful establishment of T2 DM model, diabetic rats were randomly divided into four groups, including model group, GPS groups(200, 100 mg·kg~(-1)) and metformin group(100 mg·kg~(-1)), with healthy rats serving as the control. After 6-week intragastric administration, fasting blood glucose(FBG) and oral glucose tolerance were examined. The levels of insulin, C-peptide, tumor necrosis factor-α(TNF-α), interleukin-1ß(IL-1ß), interleukin-6(IL-6) and C-reactive protein(CRP) in serum were examined. Then the homeostasis model assessment of insulin resistance(HOMA-IR) and insulin sensitivity index(ISI) were calculated. The protein expression levels of phosphorylated insulin receptor substrate-1(p-IRS-1) and phosphorylated protein kinase B(p-Akt) in skeletal muscle were measured by Western blot, as well as those of phosphorylated inhibitor of nuclear factor-κB(NF-κB) kinase ß(p-IKKß), phosphorylated alpha inhibitor of NF-κB(p-IκBα) and phosphorylated p65 subunit of NF-κB(p-p65) in adipose tissue. The relative expression levels of glucose transporter 4(GLUT4) mRNA in skeletal muscle and NF-κB mRNA in adipose tissue were measured by qRT-PCR, and the morphological changes of pancreatic tissue were observed. Compared with the model group, the GPS groups witnessed significant decrease in FBG, marked amelioration of impaired oral glucose tolerance and significant increase in ISI. Further, the high-dose GPS group saw significantly reduced HOMA-IR, TNF-α, IL-1ß and CRP, significantly increased expression levels of p-IRS-1(Tyr), p-Akt and GLUT4, and markedly inhibited p-IRS-1(Ser), p-IKKß, p-IκBα, p-p65 and NF-κB. The concentration of CRP and the expression levels of p-IRS-1(Ser), p-IKKß, p-IκBα and NF-κB were remarkably reduced in the low-dose GPS group. However, GPS was found less effective in the regulation of serum insulin, C-peptide and IL-6 levels and the alleviation of pancreatic islet injury. The results indicated that GPS can reduce FBG and improve insulin sensitivity in diabetic rats possibly by regulating the NF-κB signaling pathway, inhibiting inflammation, and thereby regulating the expression of key proteins in the insulin signaling pathway.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Animales , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/genética , Gynostemma , Insulina , FN-kappa B/genética , FN-kappa B/metabolismo , Extractos Vegetales , Ratas , Transducción de Señal
11.
Biomed Res Int ; 2021: 6650045, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34124254

RESUMEN

We have previously reported that F1012-2, a sesquiterpene lactone isolated from the Chinese herbal medicine Eupatorium lindleyanum DC., exhibits strong effects against Triple Negative Breast Cancer (TNBC). In this study, we found F1012-2 effectively inhibited cell migration and invasion detected by wound healing and transwell assays. In order to elucidate the potential mechanisms of F1012-2, we further studied its effect on DNA damage in TNBC cell lines. Using single cell gel electrophoresis (comet assay), immunofluorescence, and western blotting assays, we found that F1012-2 treatment induced significant DNA strand breaks and γ-H2AX activation. Moreover, exposure to F1012-2 led to overproduction of reactive oxygen species (ROS). NAC treatment completely eliminated ROS, which may be due to the interaction between NAC and F1012-2. A further study of the molecular mechanisms demonstrated that the MAPK signaling pathway participated in the anti-TNBC effect of F1012-2. Pretreatment with specific inhibitors targeting JNK (SP600125) and ERK (PD98059) could rescue the decrease in cell viability and inhibit expressions of JNK and ERK phosphorylation, but SB203580 had no effects. Finally, in the acute toxicity experiment, there were no obvious symptoms of poisoning in the F1012-2 treatment group. An in vivo study demonstrated that F1012-2 significantly suppressed the tumor growth and induced DNA damage. In conclusion, the activity of F1012-2-induced DNA damage in TNBC was found in vivo and in vitro, which might trigger the MAPK pathway through ROS accumulation. These results indicate that F1012-2 may be an effective anti-TNBC therapeutic agent.


Asunto(s)
Daño del ADN , ADN de Neoplasias/metabolismo , Lactonas/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Sesquiterpenos/farmacología , Neoplasias de la Mama Triple Negativas/metabolismo , Línea Celular Tumoral , ADN de Neoplasias/genética , Femenino , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología
12.
Exp Biol Med (Maywood) ; 246(13): 1541-1553, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33926261

RESUMEN

Hydnocarpin D (HD) is a bioactive flavonolignan compound that possesses promising anti-tumor activity, although the mechanism is not fully understood. Using T cell acute lymphoblastic leukemia (T-ALL) cell lines Jurkat and Molt-4 as model system, we found that HD suppressed T-ALL proliferation in vitro, via induction of cell cycle arrest and subsequent apoptosis. Furthermore, HD increased the LC3-II levels and the formation of autophagolysosome vacuoles, both of which are markers for autophagy. The inhibition of autophagy by either knockdown of ATG5/7 or pre-treatment of 3-MA partially rescued HD-induced apoptosis, thus suggesting that autophagy enhanced the efficacy of HD. Interestingly, this cytotoxic autophagy triggered ferroptosis, as evidenced by the accumulation of lipid ROS and decrease of GSH and GPX4, while inhibition of autophagy impeded ferroptotic cell death. Our study suggests that HD triggers multiple cell death processes and is an interesting compound that should be evaluated in future preclinical studies.


Asunto(s)
Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Ferroptosis/efectos de los fármacos , Flavonolignanos/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Ciclo Celular/efectos de los fármacos , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Humanos , Células Jurkat , Especies Reactivas de Oxígeno/metabolismo
13.
Anticancer Drugs ; 32(8): 842-854, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-33929988

RESUMEN

LFZ-4-46, that is [2-hydroxy-1-phenyl-1,5,6,10b-tetrahydropyrazolo(5,1-a) isoquinolin-3(2H)-yl](phenyl) methanone, a tetrahydroisoquinoline derivative with a pyrazolidine moiety, was synthetically prepared. The anti-cancer mechanism of the compound has not been clarified yet. In this study, the anticancer effects and potential mechanisms of LFZ-4-46 on human breast and prostate cancer cells were explored. (a) 3-(4,5-Dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazoliumbromide assay was first performed to detect the effects of LFZ-4-46 on the viability of human cancer cells. (b) Comet assay was utilized to evaluate DNA damage. (c) Cell cycle, apoptosis and mitochondrial membrane potential were detected by flow cytometry. (d) The expression of relative proteins was detected by western blotting assay. LFZ-4-46 significantly inhibited the viability of cancer cells in a time- and dose-dependent manner and had no obviously inhibitory effect on the viability of mammary epithelial MCF-10A cells. Mechanistic studies demonstrated that LFZ-4-46-induced cell apoptosis and cycle arrest were mediated by DNA damage. It caused DNA damage through activating γ-H2AX and breaking DNA strands. Further studies showed that mitogen-activated protein kinasess pathway was involved in these activated several key molecular events. Finally, LFZ-4-46 showed a potent antitumor effect in vivo. These results suggest that LFZ-4-46 may be a potential lead compound for the treatment of breast and prostate cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Animales , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Masculino , Ratones , Ratones Endogámicos BALB C , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Tetrahidroisoquinolinas , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Toxicol In Vitro ; 73: 105146, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33737050

RESUMEN

Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons in the substantia nigra and striatum. Aging is the most important risk factor of PD. Ferroptosis is an iron-dependent form of cell death associated with PD. However, it is not clear whether ferroptosis accelerates PD by promoting cellular senescence. This study investigated the mechanism of 1-methyl-4-phenylpyridinium (MPP+) -induced PC12 cells injury. We found that MPP+ induced cell senescence with increased ß-galactosidase activity and the expression of p53, p21 and p16 activation in cells. In addition, MPP+ treatment showed smaller mitochondria and increased membrane density, downregulation of ferritin heavy chain 1 expression and upregulation of acyl-CoA synthetase long chain family member 4 expression, and enhanced levels of oxidative stress, which were important characteristics of ferroptosis. Ferrostatin-1 (Fer-1), a ferroptosis inhibitor, was tested to eliminate MPP+-induced cell senescence. Fer-1 downregulated the expression of p53 and upregulated the expression of solute carrier family 7 member 11 (SLC7A11) and glutathione peroxidase-4 (GPX4) in MPP+-induced ferroptosis. Inhibition of p53 eliminated cell senescence by upregulation the expression of of SLC7A11 and GPX4. Thus, these results suggest that MPP+ induces senescence in PC12 cells via the p53/ SLC7A11/ GPX4 signaling pathway in the ferroptosis regulation mechanism.


Asunto(s)
1-Metil-4-fenilpiridinio/farmacología , Senescencia Celular/efectos de los fármacos , Ferroptosis/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Sistema de Transporte de Aminoácidos y+/metabolismo , Animales , Ciclohexilaminas/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Células PC12 , Fenilendiaminas/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , beta-Galactosidasa/metabolismo
15.
J Int Med Res ; 49(2): 300060520981360, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33530801

RESUMEN

OBJECTIVE: To investigate the effects of 1,25(OH)2D3 on renal fibrosis associated with the AMP-activated protein kinase (AMPK)α/mechanistic target of rapamycin (mTOR) signalling pathway in a rat model of unilateral ureteral obstruction (UUO). METHODS: A total of 54 male Sprague Dawley rats were randomly divided into three groups: sham-operation group, UUO group, and UUO plus calcitriol (3 ng/100 g) group. Renal tissue was excised for histological examination by immunohistochemistry and Western blot, and for gene expression analysis using real-time polymerase chain reaction. RESULTS: 1,25(OH)2D3 enhanced AMPKα levels, inhibited mTOR levels and slowed the development of interstitial fibrosis in kidney tissue. Compared with the UUO plus calcitriol group, UUO rats demonstrated more severe renal damage characterized by marked tubular atrophy, interstitial fibrosis and significant induction of fibrogenic transforming growth factor-ß1 and increased extra-cellular matrix proteins (α-smooth muscle actin and collagen type III), and decreased E-cadherin. CONCLUSION: Treatment with 1,25(OH)2D3 altered the AMPKα/mTOR signalling pathway to suppress excessive fibroblast activation observed in UUO rats. This may serve as a novel mechanism to ameliorate renal dysfunction and fibrotic lesions.


Asunto(s)
Enfermedades Renales , Obstrucción Ureteral , Proteínas Quinasas Activadas por AMP/genética , Animales , Fibrosis , Riñón/patología , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/etiología , Enfermedades Renales/patología , Masculino , Ratas , Ratas Sprague-Dawley , Serina-Treonina Quinasas TOR , Factor de Crecimiento Transformador beta1/genética
16.
Int Immunopharmacol ; 90: 107159, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33223467

RESUMEN

PURPOSE: Reported rates of acute kidney injury (AKI) have varied significantly among studies of coronavirus disease 2019 (COVID-19) published to date. The present meta-analysis was conducted to gain clarity regarding AKI incidence and renal replacement therapy (RRT) use in COVID-19 patients. METHODS: The PubMed, Embase, Web of Science, medRxiv, and bioRxiv databases were systematically searched for COVID-19-related case reports published through 25 July 2020. Pooled analyses were conducted using R. RESULTS: The pooled incidence of AKI in 51 studies including 21,531 patients was 12.3% (95% CI 9.5-15.6%), with higher rates of 38.9% in 290 transplant patients (95% CI 27.3-51.9%), 39.0% in 565 ICU patients (95% CI 23.2-57.6%) and 42.0% among 1745 deceased patients (95% CI 30.3-54.7%). RRT usage was reported in 39 studies of 17,664 patients, with an overall pooled use of 5.4% (95% CI 4.0-7.1%), with higher rates of 15.6% in 117 transplant patients (95%CI 9.9-23.8%) and 16.3% in 776 ICU patients (95% CI 11.1-23.3%). CONCLUSION: AKI and RRT use among COVID-19 patients represent a major public health concern, and early and appropriate intervention should be called upon to improve the prognosis of patients suffering from AKI.


Asunto(s)
Lesión Renal Aguda/epidemiología , COVID-19/epidemiología , Terapia de Reemplazo Renal , SARS-CoV-2/fisiología , Lesión Renal Aguda/complicaciones , Lesión Renal Aguda/terapia , COVID-19/complicaciones , COVID-19/terapia , China/epidemiología , Humanos , Incidencia
17.
Int Immunopharmacol ; 88: 106896, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33182045

RESUMEN

BACKGROUND/AIM: IgA nephropathy (IgAN) is the most prevalent primary glomerular disease worldwide and is responsible for 45-50% of primary glomerular diseases in China. We are essentially dependent on the degree of proteinuria to determine prognosis and it has been reported that histopathologic lesions are risk factors for the progression of IgAN. The aim of this study was to investigate the clinicopathologic features and prognosis of IgAN with C1q deposition in adult Chinese patients. METHODS: The patients of primary IgAN diagnosed by renal biopsy from January 2002 to December 2018 at the Second Hospital of Shanxi Medical University were retrospectively analyzed and divided into C1q deposit group and C1q negative group according to glomerular immunofluorescence examination. We evaluated their serologic and histopathologic findings. We collected data of patients during January 1, 2015 to December 31, 2018 and performed the clinical follow-up until the patient's estimated glomerular filtration rate (eGFR) decreased by more than 30%, entering end-stage renal disease (ESRD). Kaplan-Meier survival analysis was used to evaluate the cumulative incidence of renal progression in two groups. Univariate and multivariate Cox proportional hazard regression models were used to analyze the effect of C1q deposition on the prognosis of patients with IgA nephropathy. RESULTS: The baseline data of total 491 cases were available and 172 cases had the follow-up data. The baseline eGFR and plasma albumin (ALB) levels in C1q deposit group were lower than those in the C1q negative group, while the levels of serum creatinine (Scr), total cholesterol (TC), 24 h urine protein, low-density lipoprotein (LDL), ß2 microglobulin, etc. were higher than those in C1q negative group (all P < 0.05). Pathological indexes: glomerular segment sclerosis and adhesion (S), renal tubular atrophy/interstitial fibrosis (T), and cell/fibroblastic crescent (C) scores in C1q deposit group were higher than those in C1q negative group (all P < 0.05). With a median follow-up time of 32.5 (24,42) months, a total of 18 patients (C1q deposit: 11; C1q negative: 7) developed to endpoints. Kaplan-Meier survival curve analysis showed that there was significant decrease in cumulative incidence of renal progression between the two groups (Log-rank test χ2 = 4.78, P = 0.029). Cox regression analysis showed that the risk of renal end-point events in IgAN patients with C1q deposit group was 6.35 times higher than that in C1q negative group (HR = 6.35, 95% CI: 1.21-33.30, P = 0.029). CONCLUSION: The clinical and renal pathological changes of IgAN patients with C1q deposition are more severe than those of C1q negative patients, and has a worse outcome. C1q deposition is an independent risk factor for the progression of renal function and contributes to a poor renal prognosis in adult IgAN patients.


Asunto(s)
Complemento C1q/inmunología , Glomerulonefritis por IGA/inmunología , Adulto , Femenino , Tasa de Filtración Glomerular , Glomerulonefritis por IGA/mortalidad , Glomerulonefritis por IGA/patología , Glomerulonefritis por IGA/fisiopatología , Humanos , Estimación de Kaplan-Meier , Riñón/patología , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Factores de Riesgo , Albúmina Sérica/análisis , Adulto Joven
18.
Oncol Rep ; 44(6): 2455-2464, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33125130

RESUMEN

Zinc finger protein 403 (ZFP403), located on human chromosome 17q12­21, is closely associated with the development of cancer. However, to date, there are a limited number of studies on the biological functions of this gene, particularly in prostate cancer (PCa). The results of the present study demonstrated that compared with normal tissues, the expression of ZFP403 was markedly lower in PCa tissues, as shown by the evaluation of the Gene Expression Profiling Interactive Analysis 2 database. The decreased expression of ZFP403 in PCa clinical tissues and cell lines was confirmed by immunohistochemistry, reverse transcription­quantitative PCR and western blot analysis. Using short harpin (sh)RNA inhibition, stably­silenced ZFP403 cell lines were then constructed by lentiviral transfection (LV­PC3­shRNA­1 and 2; LV­DU145­shRNA­1 and 2). The results revealed that the knockdown of ZFP403 in PCa cells promoted cellular proliferation, colony formation, migration and invasiveness in vitro. Moreover, the levels of tumor growth­ and motility­related proteins were significantly altered after ZFP403­knockdown. A xenograft tumor model using nude mice was established to elucidate the role of ZFP403 in tumorigenesis in vivo. Tumor growth was significantly increased in mice injected with ZFP403­knockdown cells compared with the control mice. Overall, the findings of the present study demonstrate that ZFP403 functions as a tumor suppressor gene in PCa by affecting the proliferation, migration and invasiveness of PCa cells, suggesting its potential use as a clinical diagnostic marker.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Biomarcadores de Tumor/genética , Carcinogénesis/patología , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/genética , Proteínas Adaptadoras Transductoras de Señales/análisis , Anciano , Animales , Apoptosis/genética , Biomarcadores de Tumor/análisis , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación hacia Abajo , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Persona de Mediana Edad , Invasividad Neoplásica/genética , Próstata/patología , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/patología , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Biol Pharm Bull ; 43(3): 480-487, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32115506

RESUMEN

Ferroptosis is a form of necrosis caused by iron-induced accumulation of lipid hydroperoxide, involving several molecular events, and has been implicated in Parkinson's disease. Gastrodin is a component of Gastrodia elata Blume with strong antioxidant activity. We examined whether gastrodin can prevent H2O2-induced cytotoxicity in rat glioma cell line C6. For this purpose, C6 cells were pretreated with gastrodin (1, 5, 25 µM) and then exposed to 100 µM H2O2. Results showed that pretreatment of C6 cells with gastrodin decreased H2O2-induced lactate dehydrogenase (LDH) release and cell death. Moreover, gastrodin decreased intracellular malondialdehyde (MDA) level, whereas increased glutathione peroxidase (GPX) activity and glutathione (GSH) level after H2O2 treatment. In addition, treatment of deferoxamine (DFO), ferrostatin-1, and liproxstatin-1 abolished ferroptosis induced by H2O2 or erastin pretreatment. Treatment with gastrodin attenuated H2O2-induced ferroptosis and decreased lipid reactive oxygen species (ROS) (C11-BODIPY) production in C6 cells. Moreover, gastrodin increased the protein expression of nuclear factor erythroid 2-related factor 2 (Nrf2), GPX4, ferroportin-1 (FPN1), and heme oxygenase-1 (HO-1) in C6 cells treated with H2O2. RSL3, a GPX4 inhibitor, inhibited GPX4 protein level in cells co-treated with gastrodin and 100 µM H2O2. These findings indicate that gastrodin can inhibit H2O2-induced ferroptosis through its antioxidative effect in C6 cells.


Asunto(s)
Antioxidantes/farmacología , Alcoholes Bencílicos/farmacología , Ferroptosis/efectos de los fármacos , Glucósidos/farmacología , Peróxido de Hidrógeno/farmacología , Animales , Muerte Celular/efectos de los fármacos , Línea Celular , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo
20.
Nat Prod Res ; 34(18): 2660-2663, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30638055

RESUMEN

Valtrate is a principle compound isolated from Valeriana jatamansi Jones, a traditional Chinese folk medicine originally used to treat various nervous disorders. Here, we found that valtrate exhibited significant anti-cancer activity in vitro, especially in human breast cancer cells, while displayed relatively low cytotoxicity to normal human breast epithelial cells (MCF 10A). Valtrate induced cell cycle arrest at G2/M stage and apoptosis in MDA-MB-231 and MCF-7 cells, with reduced expression of p-Akt (Ser 473), cyclin B1 and caspase 8, and increased expression of p21, p-cdc2, cleaved-caspase 3, cleaved-caspase 7 and poly (ADP-ribose) polymerase (PARP). In addition, valtrate inhibited cell migration through down-regulation of MMP-9 and MMP-2 expression. These results demonstrate that valtrate possesses anti-breast cancer activities via cell cycle arrest, apoptosis, and inhibition of cell migration, thus supporting valtrate as a potential antitumor agent.


Asunto(s)
Antineoplásicos Fitogénicos/aislamiento & purificación , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Movimiento Celular/efectos de los fármacos , Iridoides/farmacología , Valeriana/química , Antineoplásicos Fitogénicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Caspasa 8/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Humanos , Iridoides/aislamiento & purificación , Células MCF-7
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...