Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.961
Filtrar
1.
Shanghai Kou Qiang Yi Xue ; 33(1): 13-21, 2024 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-38583019

RESUMO

PURPOSE: To clarify the effect of genistein(GEN) on osteogenic differentiation and explore the effect of GEN loaded by platelet-rich fibrin (PRF) on the repair process of bone defects in obese mice. METHODS: In in vitro experiments, the effect of GEN(0, 0.1, 1, 10, 50 µmol/L) on the proliferation of mouse embryonic osteoblast precursor cells (MC3T3-E1) was determined by CCK 8. Alkaline phosphatase(ALP) staining and quantitative detection of ALP activity were performed to determine the changes of ALP activity in cells; RNA and protein expression levels of ALP, osteopontin (OPN) and osteocalcin (OCN) were detected by quantitative real-time PCR(qRT-PCR) and Western blot. Alizarin red staining was used to define the effect of GEN on mineralization of MC3T3-E1. To verify the feasibility of the PRF drug loading, the ultrastructure of PRF was subsequently observed under SEM. In in vivo experiments, obese C57 mouse models were established by high-fat diet feeding. On this basis, skull defect models with a diameter of 2.8 mm were established, and the prepared GEN/PRF complexes were placed into the bone defect area. The effects of GEN on skull defect repair in obese mice were evaluated by Micro-CT scanning and hematoxylin-eosin(H-E) staining. Statistical analysis was performed with GraphPad Prism 5.0 software package. RESULTS: CCK 8 results showed that 0.1, 1 µmol/L GEN promoted cell proliferation within 7 days(P<0.05); 10 µmol/L GEN had no significant effect on the process of cell proliferation. From the second day, 50 µmol/L GEN significantly inhibited cell growth and showed cytotoxicity(P<0.05). These two concentrations had similar effects in promoting cellular osteogenic differentiation. SEM results showed that PRF presented a 3-dimensional network structure, providing space for loading drug molecules. In in vivo experiments, the body weight of mice in the high-fat diet (HFD) group was 27.7% greater than that in the normal diet group(P<0.05) and had abnormal glucose tolerance (P<0.05). Micro-CT showed that compared with the normal diet group, the number of bone trabeculae in the femur of obese mice was decreased(P<0.05), the distance between bone trabeculae was widened(P<0.05), and the bone density was decreased (P<0.05). In addition, GEN (0.1, 1.0 µmol/L) loaded by PRF increased bone volume fraction in the skull of obese mice (P<0.05). H-E results showed that GEN/PRF promoted the healing of the bone defects. CONCLUSIONS: GEN promotes osteogenic differentiation of MC3T3-E1, and it can effectively accelerate the healing of cranial bone defects after loading with PRF in obese mice.


Assuntos
Osteogênese , Fibrina Rica em Plaquetas , Animais , Camundongos , Osteogênese/genética , Genisteína/farmacologia , Camundongos Obesos , Sincalida/farmacologia , Diferenciação Celular/genética , Osteoblastos
2.
J Orthop Surg Res ; 19(1): 198, 2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38528538

RESUMO

PURPOSE: This study aimed to evaluate the protective effects of gentiopicroside against lipopolysaccharide-induced chondrocyte inflammation. METHODS: SW 1353 chondrosarcoma cells were stimulated with LPS (5 µg/ml) for 24 h and treated with different concentrations of gentiopicroside (GPS) for 24 h. The toxic effects of GPS on chondrocytes were determined using a CCK-8 assay and EdU staining. Western blotting, qPCR, and immunofluorescence analysis were used to examine the protective effect of GPS against the inflammatory response in chondrocytes induced by lipopolysaccharide (LPS). One-way ANOVA was used to compare the differences between the groups (significance level of 0.05). RESULTS: The CCK-8 results showed that 10, 20 and 40 µM GPS had no significant toxic effects on chondrocytes; GPS effectively reduced the production of IL-1ß and PGE2, reversed LPS-induced extracellular matrix degradation in cartilage by inhibiting the Stat3/Runx2 signaling pathway, and suppressed the hypertrophic transformation of SW 1353 chondrosarcoma cells. CONCLUSION: Our study demonstrated that GPS significantly inhibited the LPS-induced inflammatory response and hypertrophic cellular degeneration in SW 1353 chondrosarcoma cells and is a valuable traditional Chinese medicine for the treatment of knee osteoarthritis.


Assuntos
Condrossarcoma , Glucosídeos Iridoides , Osteoartrite , Humanos , Condrócitos/metabolismo , Lipopolissacarídeos/toxicidade , Osteoartrite/metabolismo , Sincalida/metabolismo , Sincalida/farmacologia , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Hipertrofia , Condrossarcoma/tratamento farmacológico , Interleucina-1beta/metabolismo , NF-kappa B/metabolismo
3.
Crit Rev Eukaryot Gene Expr ; 34(4): 45-54, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38505872

RESUMO

HDAC1 functions as an oncogene in multi-type cancers. This study aimed to investigate the roles of histone deacetylase 1 (HDAC1) in cervical cancer (CC). mRNA expression was determined using reverse transcription quantitative polymerase chain reaction. The protein-protein complexes was analyzed using co-immunoprecipitation assay. The binding sites between NRF2 and NEU1 were confirmed by chromatin immunoprecipitation assay. Cell viability was detected by CCK-8. Cell proliferation was measured using CCK-8 and colony formation assays. Cell migrative and invasive ability were determined using transwell assay. We found that HDAC1 was upregulated in CC patients and cells. Trichostatin A (TSA) treatment decreased the number of colonies and migrated and invaded cells. Moreover, HDAC1 interacted with NRF2 to downregulate NEU1 expression. NEU1 knockdown attenuated the effects of TSA and enhanced the aggressiveness of CC cells. In conclusion, HDAC1 functions as an oncogene in CC. Targeting HDAC1 may be an alternative strategy for CC.


Assuntos
Neoplasias do Colo do Útero , Feminino , Humanos , Regulação para Baixo , Neoplasias do Colo do Útero/genética , Histona Desacetilase 1/genética , Histona Desacetilase 1/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Sincalida/genética , Sincalida/metabolismo , Neuraminidase/genética , Neuraminidase/metabolismo
4.
Dig Dis Sci ; 69(4): 1156-1168, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38448762

RESUMO

BACKGROUND/AIMS: We examined the involvement of cholecystokinin (CCK) in the exacerbation of indomethacin (IND)-induced gastric antral ulcers by gastroparesis caused by atropine or dopamine in mice. METHODS: Male mice were fed for 2 h (re-feeding) following a 22-h fast. Indomethacin (IND; 10 mg/kg, s.c.) was administered after re-feeding; gastric lesions were examined 24 h after IND treatment. In another experiment, mice were fed for 2 h after a 22-h fast, after which the stomachs were removed 1.5 h after the end of the feeding period. Antral lesions, the amount of gastric contents, and the gastric luminal bile acids concentration were measured with or without the administration of the pro- and antimotility drugs CCK-octapeptide (CCK-8), atropine, dopamine, SR57227 (5-HT3 receptor agonist), apomorphine, lorglumide (CCK1 receptor antagonist), ondansetron, and haloperidol alone and in combination. RESULTS: IND produced severe lesions only in the gastric antrum in re-fed mice. CCK-8, atropine, dopamine, SR57227 and apomorphine administered just after re-feeding increased bile reflux and worsened IND-induced antral lesions. These effects were significantly prevented by pretreatment with lorglumide. Although atropine and dopamine also increased the amount of gastric content, lorglumide had no effect on the delayed gastric emptying provoked by atropine and dopamine. Both ondansetron and haloperidol significantly inhibited the increase of bile reflux and the exacerbation of antral lesions induced by atropine and dopamine, respectively, but did not affect the effects of CCK-8. CONCLUSIONS: These results suggest that CCK-CCK1 receptor signal increases bile reflux during gastroparesis induced by atropine and dopamine, exacerbating IND-induced antral ulcers.


Assuntos
Refluxo Biliar , Gastroparesia , Úlcera Gástrica , Camundongos , Masculino , Animais , Indometacina , Úlcera , Receptor de Colecistocinina A , Sincalida/efeitos adversos , Apomorfina/efeitos adversos , Dopamina , Haloperidol/efeitos adversos , Ondansetron , Úlcera Gástrica/induzido quimicamente , Colecistocinina/efeitos adversos , Receptores da Colecistocinina , Atropina/efeitos adversos
5.
Neurochem Int ; 175: 105683, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38341034

RESUMO

BACKGROUND: Oxidative stress and neuroinflammation are proven to play critical roles in the pathogenesis of Parkinson's disease (PD). As reported, patients with PD have lower level of STAT4 compared with healthy subjects. However, the biological functions and mechanisms of STAT4 in PD pathogenesis remain uncertain. This study aimed to investigate the roles and related mechanisms of STAT4 in PD development. METHODS: The intraperitoneal injection of MPTP (20 mg/kg) dissolved in physiological saline was performed to mimic PD-like conditions in vivo. MPP + solution was prepared for cell model of PD. Cell viability was measured by CCK-8. Griess reaction was conducted to measure NO concentrations. The mRNA and protein levels were evaluated by RT-qPCR and western blotting. ROS generation was assessed by DCFH-DA. The levels of inflammatory cytokines were measured by ELISA. Cell apoptosis was examined by flow cytometry and western blotting. Moreover, the SH-SY5Y cells were treated with conditioned medium from LPS-stimulated microglia and subjected to CCK-8 assays and ELISA. Mechanistically, CHIP assays and luciferase reporter assays were performed to verify the binding relationship between KISS1 and STAT4. For in vivo analysis, the histological changes of midbrain tissues of mice were determined by hematoxylin and eosin staining. The expression of tyrosine hydroxylase (TH) was detected by immunohistochemistry staining. Iba-1 positive microglial cells in the striatum were assessed by immunofluorescence staining. RESULTS: For in vitro analysis, STAT4 level was downregulated after MPP+ treatment, and STAT4 upregulation inhibited the oxidative damage, inflammation and apoptosis in SH-SY5Y cells. STAT4 bound at +215-228 region of KISS1, and KISS1 upregulation counteracted the protection of STAT4 upregulation against cell damage. Moreover, STAT4 upregulation inhibited cell viability loss and inflammation induced by conditioned medium from LPS-treated microglia, whereas KISS1 upregulation had the opposite effect. For in vivo analysis, the protective effects of STAT4 upregulation against inflammatory response, oxidative stress, dopaminergic neuronal loss and microglia activation were attenuated by KISS1 upregulation. Moreover, the inactivation of MAPK pathway caused by STAT4 upregulation was reversed by KISS1 upregulation, and MAPK inhibition attenuated the MPP+-induced inflammation, oxidative stress and apoptosis in SH-SY5Y cells. CONCLUSION: STAT4 inhibits KISS1 to attenuate the oxidative damage, inflammation and neuronal apoptosis in PD by inactivating the MAPK pathway.


Assuntos
Neuroblastoma , Doença de Parkinson , Animais , Humanos , Camundongos , Apoptose , Linhagem Celular Tumoral , Meios de Cultivo Condicionados/farmacologia , Inflamação/induzido quimicamente , Inflamação/metabolismo , Kisspeptinas , Lipopolissacarídeos/farmacologia , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Doença de Parkinson/metabolismo , Sincalida/efeitos adversos , Sincalida/metabolismo , Fator de Transcrição STAT4/metabolismo
6.
Beijing Da Xue Xue Bao Yi Xue Ban ; 56(1): 17-24, 2024 Feb 18.
Artigo em Chinês | MEDLINE | ID: mdl-38318891

RESUMO

OBJECTIVE: To explore the effects of different polymers on in vitro biomimetic mineralization of small intestinal submucosa (SIS) scaffolds, and to evaluate the physicochemical properties and biocompatibility of the SIS scaffolds. METHODS: The SIS scaffolds prepared by freeze-drying method were immersed in simulated body fluid (SBF), mineralized liquid containing polyacrylic acid (PAA) and mine-ralized liquid containing PAA and polyaspartic acid (PASP). After two weeks in the mineralized solution, the liquid was changed every other day. SBF@SIS, PAA@SIS, PAA/PASP@SIS scaffolds were obtained. The SIS scaffolds were used as control group to evaluate their physicochemical properties and biocompatibility. We observed the bulk morphology of the scaffolds in each group, analyzed the microscopic morphology by environment scanning electron microscopy and determined the porosity and pore size. We also analyzed the surface elements by energy dispersive X-ray spectroscopy (EDX), analyzed the structure of functional groups by Flourier transformed infrared spectroscopy (FTIR), detected the water absorption rate by using specific gravity method, and evaluated the compression strength by universal mechanical testing machine. The pro-cell proliferation effect of each group of scaffolds were evaluated by CCK-8 cell proliferation method. RESULTS: Under scanning electron microscopy, the scaffolds of each group showed a three-dimensional porous structure with suitable pore size and porosity, and crystal was observed in all the mineralized scaffolds of each group, in which the crystal deposition of PAA/PASP@SIS scaffolds was more regular. At the same time, the collagen fibers could be seen to thicken. EDX analysis showed that the characteristic peaks of Ca and P were found in the three groups of mineralized scaffolds, and the highest peaks were found in the PAA/PASP@SIS scaffolds. FTIR analysis proved that all the three groups of mineralized scaffolds were able to combine hydroxyapatite with SIS. All the scaffolds had good hydrophilicity. The compressive strength of the mineralized scaffold in the three groups was higher than that in the control group, and the best compressive strength was found in PAA/PASP@SIS scaffold. The scaffolds of all the groups could effectively adsorb proteins, and PAA/PASP@SIS group had the best adsorption capacity. In the CCK-8 cell proliferation experiment, the PAA/PASP@SIS scaffold showed the best ability to promote cell proliferation with the largest number of living cells observed. CONCLUSION: Compared with other mineralized scaffolds, PAA/PASP@SIS scaffolds prepared by mineralized solution containing both PAA and PASP have better physicochemical properties and biocompatibility and have potential applications in bone tissue engineering.


Assuntos
Polímeros , Tecidos Suporte , Tecidos Suporte/química , Polímeros/química , Biomimética , Sincalida , Engenharia Tecidual/métodos , Intestino Delgado , Porosidade
7.
J Exp Clin Cancer Res ; 43(1): 62, 2024 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-38419081

RESUMO

BACKGROUND: In recent years, the development of adjunctive therapeutic hyperthermia for cancer therapy has received considerable attention. However, the mechanisms underlying hyperthermia resistance are still poorly understood. In this study, we investigated the roles of cold­inducible RNA binding protein (Cirbp) in regulating hyperthermia resistance and underlying mechanisms in nasopharyngeal carcinoma (NPC). METHODS: CCK-8 assay, colony formation assay, tumor sphere formation assay, qRT-PCR, Western blot were employed to examine the effects of hyperthermia (HT), HT + oridonin(Ori) or HT + radiotherapy (RT) on the proliferation and stemness of NPC cells. RNA sequencing was applied to gain differentially expressed genes upon hyperthermia. Gain-of-function and loss-of-function experiments were used to evaluate the effects of RNAi-mediated Cirbp silencing or Cirbp overexpression on the sensitivity or resistance of NPC cells and cancer stem-like cells to hyperthermia by CCK-8 assay, colony formation assay, tumorsphere formation assay and apoptosis assay, and in subcutaneous xenograft animal model. miRNA transient transfection and luciferase reporter assay were used to demonstrate that Cirbp is a direct target of miR-377-3p. The phosphorylation levels of key members in ATM-Chk2 and ATR-Chk1 pathways were detected by Western blot. RESULTS: Our results firstly revealed that hyperthermia significantly attenuated the stemness of NPC cells, while combination treatment of hyperthermia and oridonin dramatically increased the killing effect on NPC cells and cancer stem cell (CSC)­like population. Moreover, hyperthermia substantially improved the sensitivity of radiation­resistant NPC cells and CSC­like cells to radiotherapy. Hyperthermia noticeably suppressed Cirbp expression in NPC cells and xenograft tumor tissues. Furthermore, Cirbp inhibition remarkably boosted anti­tumor­killing activity of hyperthermia against NPC cells and CSC­like cells, whereas ectopic expression of Cirbp compromised tumor­killing effect of hyperthermia on these cells, indicating that Cirbp overexpression induces hyperthermia resistance. ThermomiR-377-3p improved the sensitivity of NPC cells and CSC­like cells to hyperthermia in vitro by directly suppressing Cirbp expression. More importantly, our results displayed the significantly boosted sensitization of tumor xenografts to hyperthermia by Cirbp silencing in vivo, but ectopic expression of Cirbp almost completely counteracted hyperthermia-mediated tumor cell-killing effect against tumor xenografts in vivo. Mechanistically, Cirbp silencing-induced inhibition of DNA damage repair by inactivating ATM-Chk2 and ATR-Chk1 pathways, decrease in stemness and increase in cell death contributed to hyperthermic sensitization; conversely, Cirbp overexpression-induced promotion of DNA damage repair, increase in stemness and decrease in cell apoptosis contributed to hyperthermia resistance. CONCLUSION: Taken together, these findings reveal a previously unrecognized role for Cirbp in positively regulating hyperthermia resistance and suggest that thermomiR-377-3p and its target gene Cirbp represent promising targets for therapeutic hyperthermia.


Assuntos
Diterpenos do Tipo Caurano , Hipertermia Induzida , MicroRNAs , Neoplasias Nasofaríngeas , Animais , Humanos , Neoplasias Nasofaríngeas/patologia , Sincalida/metabolismo , Carcinoma Nasofaríngeo/genética , Carcinoma Nasofaríngeo/terapia , Carcinoma Nasofaríngeo/patologia , MicroRNAs/genética , Células-Tronco Neoplásicas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica
8.
Int Immunopharmacol ; 129: 111629, 2024 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-38346377

RESUMO

BACKGROUND: As a common chronic musculoskeletal condition, osteoarthritis (OA) presently lacks particular treatment strategies. The aim of this study was to examine how AT-III therapies affected macrophage repolarity in order to slow down the advancement of OA. METHODS: RAW264.7 macrophages were polarized to M1 subtypes then administered with different concentrations of AT-III. Immunofluorescence, qRT-PCR and flow cytometry were used to assess the polarization of the macrophages. The mechanism of AT-III repolarize macrophages was evaluated by western blot. Furthermore, the effects of macrophage conditioned media (CM) on the migration, proliferation, and chondrogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) were investigated using CCK-8 assays, the scratch test, and alcian blue staining. The effects of macrophage CM on chondrocyte proliferation and degeneration were investigated using CCK-8 and qRT-PCR. In vivo micro-CT and histological observations were performed on rats with anterior cruciate ligament transection and partial medial meniscectomy, either with or without AT-III treatment. RESULTS: AT-III repolarized M1 macrophages to M2 phenotype. Mechanistically, AT-III reduced the expression of Toll-like receptor(TLR) 4 induced by lipopolysaccharide in RAW264.7 and lowered nuclear factor-κB (NF-κB) signaling molecules p-p65 and p-IκBα. The TLR4 agonist RS09 reversed the effects of AT-III on macrophage repolarization. AT-III-induced macrophages CM stimulated BMSCs migration, proliferation and chondrogenic differentiation. AT-III-treated macrophage CM promoted chondrocyte proliferation while inhibiting chondrocyte degeneration. In vivo, AT-III treatment alleviated the degree of synovitis, inhibited subchondral bone remodeling and reduced cartilage destruction in the rat OA model. CONCLUSIONS: AT-III attenuates OA by repolarizing macrophages through inactivating TLR4/NF-κB signaling. These data suggest that AT-III may be an effective therapeutic candidate for OA treatment.


Assuntos
NF-kappa B , Osteoartrite , Ratos , Animais , NF-kappa B/metabolismo , Receptor 4 Toll-Like/metabolismo , Sincalida/metabolismo , Sincalida/farmacologia , Sincalida/uso terapêutico , Macrófagos , Osteoartrite/tratamento farmacológico , Osteoartrite/metabolismo
9.
Brasília; CONITEC; fev. 2024.
Não convencional em Português | BRISA/RedTESA, Inca | ID: biblio-1551260

RESUMO

INTRODUÇÃO: Os tumores neuroendócrinos (TNE) são neoplasias, com origem mais comum no trato gastrointestinal, que podem cursar com liberação de hormônios associado a sintomas, levando a síndrome carcinoide, com incidência anual estimada em 0,25/1.000.000 na Europa (ano de 2008). As manifestações clínicas mais comuns incluem diarreia secretória e rubor súbito, mas a diarreia é considerada mais debilitante com potencial risco de morte. Quando o tratamento curativo com ressecção completa não é viável pela presença de doença metastática, o tratamento é direcionado para o controle dos sintomas da síndrome carcinoide e os análogos da somatostatina (octreotida ou lanreotida) são considerados terapia de primeira linha na SC. PERGUNTAS DE PESQUISA: O acetato de octreotida de liberação prolongada (octreotida LAR) e o acetato de lanreotida de liberação prolongada (lanreotida LP) são eficazes, seguros e custo-efetivos para o tratamento dos sintomas relacionados à SC associados ao TNE gastroenteropancreático funcional em pacientes adultos? EVIDÊNCIAS CLÍNICAS: Foram selecionados três ensaios clínico


Assuntos
Humanos , Sincalida/análogos & derivados , Octreotida/uso terapêutico , Neoplasias das Glândulas Endócrinas/etiologia , Trato Gastrointestinal/patologia , Enteropatias/patologia , Síndrome do Carcinoide Maligno/tratamento farmacológico , Sistema Único de Saúde , Brasil , Eficácia , Análise Custo-Benefício/economia
10.
Aging (Albany NY) ; 16(2): 1767-1780, 2024 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-38244583

RESUMO

Approximately 10% of gastric cancers are associated with Epstein-Barr virus (EBV). Tremella fuciformis polysaccharides (TFPs) are characterized by antioxidative and anti-inflammatory effects in different diseases. However, whether TFP improves EBV-associated gastric cancer (EBVaGC) has never been explored. The effects of TFP on EBV-infected GC cell viability were determined using a CCK-8 assay and flow cytometry. Western blotting and RT-qPCR were performed to explore the expression of ferroptosis-related proteins. The CCK-8 assay showed that TFP decreased EBV-infected GC cell viability in a dose- and time-dependent manner. Flow cytometry assays indicated that TFP significantly induced EBV-infected GC cell death. TFP also reduced the migratory capacity of EBV-infected GC cells. Furthermore, treatment with TFP significantly increased the mRNA levels of PTGS2 and Chac1 in EBV-infected GC cells. Western blot assays indicated that TFP suppressed the expression of NRF2, HO-1, GPX4 and xCT in EBV-infected GC cells. More importantly, overexpression of NRF2 could obviously rescue TFP-induced downregulation of GPX4 and xCT in EBV-infected GC cells. In summary, we showed novel data that TFP induced ferroptosis in EBV-infected GC cells by inhibiting NRF2/HO-1 signaling. The current findings may shed light on the potential clinical application of TFP in the treatment of EBVaGC.


Assuntos
Basidiomycota , Infecções por Vírus Epstein-Barr , Ferroptose , Neoplasias Gástricas , Humanos , Herpesvirus Humano 4/genética , Neoplasias Gástricas/genética , Infecções por Vírus Epstein-Barr/complicações , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Sincalida/metabolismo
11.
Gene ; 902: 148156, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38211899

RESUMO

BACKGROUND: Cuproptosis is a newly discovered cell death mechanism that relies on mitochondrial respiration, for which oxidative phosphorylation (OXPHOS) is an essential part. However, the detailed mechanisms of cuproptosis associated with OXPHOS in esophageal squamous cell carcinoma (ESCC) and how this correlation affects prognosis still remains unclear. METHODS: scRNA-seq data of ESCC were downloaded from SRA (Sequence Read Archive) database. "AUCell" algorithm was used to grouping epithelial cells according to cuproptosis and OXPHOS score. Cell-cell communication, Pseudo-time Trajectory and transcription factor enrichment analysis were repectively conducted by "CellChat", "monocle3" package and "pySCENIC" algorithm. Univariate and LASSO cox regression analysis were used to construct the prognostic cuproptosis-OXPHOS signature. Finally, CCK-8 assay and DCFH-DA staining assay were respectively validated the sensitive and ROS production of elesclomol. RESULTS: scRNA-seq data were analyzed to identify 10 core cell types. According to the median scores for cuproptosis and OXPHOS, malignant epithelial cells were divided into double high, double low, and mixed groups. The double high group distributed at the end of the pseudo-time trajectory and harbored HMGA1(+) as specific transcriptional regulons. Knockdown of HMGA1 partly reversed the inhibition of cell viability visualized by CCK-8 assay, while reactive oxygen species (ROS) production by elesclomol was enhanced after HMGA1 silencing. Furthermore, the immunosuppressive signal was significantly increased in the double high group detected by 'CellChat' in single-cell data and 'ssGSEA' in bulk data followed by 'CIBERSORTx' algorithm. Finally, a new cuproptosis-OXPHOS prognostic signature (CNN2, ATP6V0E1, PSMD6, CCDC25, IGFBP2, MT1E, and RPS4Y1) was constructed for the prediction of the prognosis, and a high-risk group corresponding to a more sensitive tendency to erlotinib, dasatinib, and bosutinib treatment was identified. CONCLUSIONS: Our study revealed the relationship between OXPHOS and tendency of cuproptosis in ESCC, and malignant cells with this characteristic exerted immunosuppressive signals and indicated poor prognosis. Furthermore, we constructed the regulatory network in high cuproptosis-OXPHOS ESCC and identified HMGA1 as a potential regulator molecule of cuproptosis mediated by elesclomol.


Assuntos
Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Hidrazinas , Humanos , Carcinoma de Células Escamosas do Esôfago/genética , Fosforilação Oxidativa , Proteína HMGA1a , Neoplasias Esofágicas/genética , Espécies Reativas de Oxigênio , Sincalida , Biologia Computacional , Apoptose , Cobre , Microambiente Tumoral/genética
12.
Pathol Res Pract ; 254: 155095, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38237399

RESUMO

BACKGROUND: To explore the role of Kelch repeat and BTB (POZ) domain containing 2 (KBTBD2) in Gastric cancer(GC) via studying the level of KBTBD2 and its impact on GC cells and mice model. METHODS: Expression of KBTBD2 in GC was analyzed by analysis of TCGA data, Western blotting and Real-time quantitative polymerasechain reaction (RT-qPCR). The role of KBTBD2 on GC cells proliferation, viability, invasion, migration and apoptosis in vitro were assessed by using western blotting,RT-qPCR,CCK-8, EDU, Colony Formation Assay, Wound healing assay, Transwell, JC-1 mitochondrial membrane potential and flow cytometry assay, respectively. And levels of Bcl-2, BAX, PARP, E-cadherin, Vimentin, N-cadherin, EGFR, SOS1, NROS, BRAF,ERK1/2 and GAPDH were tested by western blotting. Relation of KBTBD2 and epidermal growth factor receptor (EGFR) was predicted by KEGG analysis. KBTBD2 gene GSEA enrichment was analyzed by using R language. Moreover, CCK-8, western blotting, and wound healing assays were used to verify the correlation of KBTBD2 and EGFR pathway. Finally, tumor growth in mice was also investigated. Cells proliferation, migration and apoptosis were detected by Ki67 staining, Tunnel staining and mouse lung metastasis model. RESULTS: KBTBD2 was highly expressed in GC, and was related to poor prognosis. Moreover, silencing KBTBD2 suppressed GC cell proliferation, migration and invasion, while also inhibited the EMT, but promoted apoptosis. At the same time, KBTBD2 overexpression showed opposite results. In addition, KBTBD2 regulated the EGFR pathway. Further, silencing KBTBD2 inhibited tumor growth, cell proliferation and migration but promoted apoptosis in vivo, and KBTBD2 overexpression showed opposite results. CONCLUSIONS: KBTBD2 was highly expressed in GC. KBTBD2 promotes the progress of GC by activating EGFR signal pathway. KBTBD2 may thus be a novel target for treating GC.


Assuntos
Neoplasias Gástricas , Animais , Camundongos , Neoplasias Gástricas/patologia , Sincalida/genética , Sincalida/metabolismo , Linhagem Celular Tumoral , Transdução de Sinais , Receptores ErbB/genética , Proliferação de Células/genética , Modelos Animais de Doenças , Movimento Celular/genética , Regulação Neoplásica da Expressão Gênica
13.
Arab J Gastroenterol ; 25(1): 51-57, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38220478

RESUMO

BACKGROUND AND STUDY AIMS: Glypican 2 (GPC2) is a member of the glypican gene family and is expressed in multiple kinds of cancer. However, the function and mechanism of GPC2 in colorectal cancer remains unclear. In this study, we aimed to identify the role of GPC2 on tumor cell proliferation and survival in colorectal cancer. PATIENTS AND METHODS: Ten pairs of colon cancer and matched normal colon tissues were collected in this research. GEPIA was used to analysis the GPC2 gene expression profile in TGCA data base. RT-qPCR and western blot assay were performed to determine the mRNA and protein expressions. CCK-8, Flow cytometry and colon formation assay were applied to evaluate cell viability. IHC staining was performed to evaluate the protein expression in tissues. The function of GPC2 in vivo was verified by an animal model of colon cancer. RESULTS: Through the bioinformatics analysis and qRT-PCR validation, we found that GPC2 was upregulated in the colon cancer tissues and cells. GPC2 knockdown suppressed cell proliferation in vitro and in vivo was confirmed by the results of CCK-8, colony formation assays, and tumor xenograft models. Moreover, by the analysis of flow cytometry assay and gain-or-loss function experiments, we discovered that CEP164 was highly associated with the expression state of GPC2, and mediated G2/M-phase arrest in GPC2-downregulated tumor cells. CONCLUSION: GPC2 might be a novel oncogenic gene in colorectal cancer, suggesting that it could be a considerable marker for the diagnosis and prognosis of colorectal cancer.


Assuntos
Neoplasias do Colo , Neoplasias Colorretais , Animais , Humanos , Glipicanas/genética , Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Prognóstico , Sincalida/genética , Sincalida/metabolismo , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Regulação Neoplásica da Expressão Gênica
14.
Appl Biochem Biotechnol ; 196(2): 717-728, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37178251

RESUMO

This study aimed to explore the molecular mechanism of LCN2 regulating aerobic glycolysis on abnormal proliferation of HCC cells. Based on the prediction of GEPIA database, the expression levels of LCN2 in hepatocellular carcinoma tissues were detected by RT-qPCR analysis, western blot, and immunohistochemical staining, respectively. In addition, CCK-8 kit, clone formation, and EdU staining were used to analyze the effect of LCN2 on the proliferation of hepatocellular carcinoma cells. Glucose uptake and lactate production were detected using kits. In addition, western blot was used to detect the expressions of aerobic glycolysis-related proteins. Finally, western blot was used to detect the expressions of phosphorylation of JAK2 and STAT3. We found LCN2 was upregualted in hepatocellular carcinoma tissues. CCK-8 kit, clone formation, and EdU staining results showed that LCN2 could promote the proliferation in hepatocellular carcinoma cells (Huh7 and HCCLM3 cells). Western blot results and kits confirmed that LCN2 significantly promotes aerobic glycolysis in hepatocellular carcinoma cells. Western blot results showed that LCN2 could significantly upregulate the phosphorylation of JAK2 and STAT3. Our results indicated that LCN2 activated the JAK2/STAT3 signaling pathway, promoted aerobic glycolysis, and accelerated malignant proliferation of hepatocellular carcinoma cells.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Glicólise , Janus Quinase 2/metabolismo , Lipocalina-2/metabolismo , Neoplasias Hepáticas/patologia , Transdução de Sinais , Sincalida/metabolismo , Sincalida/farmacologia , Fator de Transcrição STAT3/metabolismo
15.
Int J Toxicol ; 43(2): 134-145, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-37859596

RESUMO

Antioxidant 1 copper chaperone (Atox1) may contribute to preventing DDP cochlear damage by regulating copper transport family and cell cycle proteins. A rat model of cochlear damage was developed by placing gelatin sponges treated with DDP in the cochlea. HEI-OC1 cells were treated with 133 µM DDP as a cell model. DDP-induced ototoxicity in rats was confirmed by immunofluorescence (IF) imaging. The damage of DDP to HEI-OC1 cells was assessed by using CCK-8, TUNEL, and flow cytometry. The relationship between Atox1, a member of the copper transport protein family, and the damage to in vivo/vitro models was explored by qRT-PCR, western blot, CCK-8, TUNEL, and flow cytometry. DDP had toxic and other side effects causing cochlear damage and promoted HEI-OC1 cell apoptosis and cell cycle arrest. The over-expression of Atox1 (oe-Atox1) was accomplished by transfecting lentiviral vectors into in vitro/vivo models. We found that oe-Atox1 increased the levels of Atox1, copper transporter 1 (CTR1), and SOD3 in HEI-OC1 cells and decreased the expression levels of ATPase copper transporting α (ATP7A) and ATPase copper transporting ß (ATP7B). In addition, the transfection of oe-Atox1 decreased cell apoptosis rate and the number of G2/M stage cells. Similarly, the expression of myosin VI and phalloidin of cochlea cells in vivo decreased. Atox1 ameliorated DDP-induced damage to HEI-OC1 cells or rats' cochlea by regulating the levels of members of the copper transport family.


Assuntos
Cisplatino , Cobre , Ratos , Animais , Cisplatino/toxicidade , Cobre/toxicidade , Sincalida/farmacologia , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Chaperonas Moleculares/farmacologia , Cóclea , Ciclo Celular
16.
Angle Orthod ; 94(2): 200-206, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38052230

RESUMO

OBJECTIVES: To develop a photochromic bracket adhesive (PCA) with modification using photochromic material and evaluate the biocompatibility, bond strength, photochromic property, and adhesive removal efficiency. MATERIALS AND METHODS: The resin-modified glass ionomer powder was mixed with the photochromic material and then blended with the liquid agent to form PCA. Biocompatibility was evaluated by CCK-8 kit, and shear bond strength (SBS) was measured. Stereoscopic microscopy and quantitative color analysis were used to assess the photochromic property. Bracket bonding and debonding procedures were performed on a head simulator with the assistance of an ultraviolet radiator. The effectiveness of adhesive removal during bonding and debonding procedures was assessed using a stereomicroscope. Removal time was recorded, and the enamel damage index after debonding was analyzed. RESULTS: CCK-8 assay and SBS test indicated that 5wt.% mixing ratios of the photochromic material did not compromise the biocompatibility and SBS of the adhesive (PCA5). PCA5 showed photochromic properties and could help the operator remove adhesive more thoroughly without increasing enamel damage. CONCLUSIONS: Photochromic adhesive (PCA5) can be good for orthodontic adhesive removal and therefore has good clinical translation potential.


Assuntos
Colagem Dentária , Braquetes Ortodônticos , Cimentos Dentários , Cimentos de Resina/química , Sincalida , Propriedades de Superfície , Colagem Dentária/métodos , Teste de Materiais , Resistência ao Cisalhamento , Análise do Estresse Dentário
17.
Fish Shellfish Immunol ; 144: 109245, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38000652

RESUMO

Irisin, a secreted myokine generated by fibronectin type III domain-containing protein 5, has recently shown the potential to alleviate inflammation. Cholecystokinin-octapeptide (CCK-8) is closely associated with the inflammatory factor TNF-α, a central cytokine in inflammatory reactions. However, the interactions between irisin and CCK-8 in regulating TNF-α production and the underlying mechanism have not yet been elucidated. In the present study, irisin treatment inhibited the basal and the CCK-8-induced TNF-α production in vivo. Additionally, neutralizing circulating irisin using an irisin antiserum significantly augmented the CCK-8-induced stimulation of TNF-α levels. Moreover, the incubation of head kidney cells with irisin or CCK-8 has opposite effects on TNF-α secretion. Notably, irisin treatment inhibited basal and CCK-8-stimulated TNF-α release and gene transcription in head kidney cells. Mechanistically, the inhibitory actions of irisin on basal and CCK-8-induced TNF-α production could be negated by co-administered with the selective integrin αVß5 inhibitor cilengitide. In addition, the inhibitory effect of irisin on basal and CCK-8-triggered TNF-α production could be abolished by the inhibition of the nuclear factor-kappa B (NF-κB) signaling pathway. Furthermore, irisin impeded CCK-8-induced phosphorylation and degradation of IκBα, simultaneously inhibiting NF-κB phosphorylation, preventing its translocation into the nucleus, and suppressing its DNA-binding activity induced by CCK-8. Collectively, these results suggest that the inhibitory effect of irisin on TNF-α production caused by CCK-8 is mediated via the integrin αVß5-NF-κB signaling pathways in tilapia.


Assuntos
Ciclídeos , NF-kappa B , Animais , NF-kappa B/metabolismo , Sincalida/efeitos adversos , Fator de Necrose Tumoral alfa/farmacologia , Fibronectinas/genética , Ciclídeos/metabolismo , Transdução de Sinais , Inflamação/induzido quimicamente
18.
Cell Oncol (Dordr) ; 47(1): 343-359, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37672204

RESUMO

BACKGROUND: Chemotherapeutic agents such as cisplatin are commonly used in patients with clinically unresectable or recurrent esophageal cancer (ESCA). However, patients often develop resistance to cisplatin, which in turn leads to a poor prognosis. Studies have shown that FAM111B may be involved in the development of tumors as an oncogene or tumor suppressor gene. However, the pathological role and corresponding mechanism of FAM111B in ESCA are still unclear. METHODS: The GEPIA web tool, ENCORI Pan-Cancer Analysis Platform and UALCAN-TCGA database were used to study the expression of FAM111B in ESCA. CCK-8, angiogenesis, Transwell and xenograft assays were applied to explore the biological function of FAM111B in ESCA. Western blot, RT-qPCR, and RNA-seq analyses were applied to study the FAM111B/GSDMA axis in the progression of ESCA cells. CCK-8 and xenograft assays were used to study the role of the FAM111B/GSDMA axis in determining the sensitivity of ESCA to cisplatin. RESULTS: Our results demonstrated that FAM111B is highly expressed in ESCA tissues compared to normal tissues. We showed that FAM111B promotes the progression of ESCC cells by binding to GSDMA and that the trypsin protease domain is essential for the activity of FAM111B. Furthermore, we showed that the FAM111B/GSDMA axis regulates cisplatin sensitivity in ESCA. CONCLUSIONS: Overall, we identified a novel FAM111B/GSDMA axis regulating ESCA tumorigenesis and chemosensitivity, at least in ESCC cells.


Assuntos
Proteínas de Ciclo Celular , Cisplatino , Neoplasias Esofágicas , Gasderminas , Humanos , Carcinogênese , Proteínas de Ciclo Celular/metabolismo , Transformação Celular Neoplásica , Cisplatino/farmacologia , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Gasderminas/metabolismo , Sincalida , Resistencia a Medicamentos Antineoplásicos
19.
Proteomics Clin Appl ; 18(1): e2300061, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37672800

RESUMO

PURPOSE: This research aimed to find potential HER2 mutations that would have an impact on breast cancer and investigate the underlying mechanism. EXPERIMENTAL DESIGN: This study first investigated 238 pairs of breast cancer and para-cancerous tissue samples from patients on the targeted next-generation sequencing (tNGS) platform. CCK-8 and clone formation assay were used to investigate whether the mutation exerts proliferative effects on breast cancer cells. In addition, mass spectrometry-based comparative proteomic and phosphoproteomic analyses of the mutation types and wild types of MCF-7 cell lines were carried out. RESULTS: Among the identified mutations, a new mutation HER2 L796P promoted the proliferation of breast cancer cells and had resistance to lapatinib using CCK-8 cell proliferation assay and clone formation assay. The bioinformatic analysis showed that RAS family proteins and ERK phosphorylated proteins significantly increased in the L796P mutant cells. The Gene Ontology (GO) analysis revealed that L796P mutation affected the function of breast cancer at the level of upstream genes in the MAPK and PI3K-AKT-TOR pathways. CONCLUSIONS AND CLINICAL RELEVANCE: This study demonstrated that a rare mutation HER2 L796P could be a potential therapeutic target for the clinical management of breast cancer.


Assuntos
Neoplasias da Mama , Feminino , Humanos , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Mutação/genética , Mutação de Sentido Incorreto/genética , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteômica , Quinazolinas/farmacologia , Quinazolinas/uso terapêutico , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Sincalida/genética , Sincalida/uso terapêutico
20.
J Periodontal Res ; 59(1): 84-93, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37814383

RESUMO

BACKGROUND AND OBJECTIVES: The utilization of natural products to enhance the function of periodontal ligament cells (PDLCs) has emerged as a popular area of research. Recent investigations have demonstrated that sappanchalcone (SC) possesses pharmacological properties such as anti-inflammatory and osteoprotective effects. This study aims to explore the impact of SC on the in vivo and in vitro osteogenic differentiation ability of PDLCs. MATERIALS: Cell proliferation was quantified using the CCK-8 assay, while gene expression levels were assessed through qRT-PCR analysis. Osteoblast differentiation capacity was evaluated by employing Alizarin red staining (ARS), alkaline phosphatase (ALP) staining and western blot (WB) analysis. A rat model of periodontitis was established utilizing the tether-wire method. Micro-CT imaging and hematoxylin and eosin (HE) staining were employed to evaluate alveolar bone resorption. Masson's trichrome staining was utilized to observe fiber alignment, whereas immunohistochemistry (IHC) techniques were applied for detecting osteogenic and inflammatory factors. RESULTS: The results from the CCK-8 assay indicate no observed cytotoxicity for concentrations of 1, 5, or 10 nM for SC treatment (p < .05), while qRT-PCR analysis demonstrates a significant decrease in inflammatory factors such as MMP-1 and IL-6 with treatment by SC (p < .05). Additionally, western blotting reveals an increase in protein expression levels of Runx2 and OPN within PDLCs treated with SC compared to control groups (p < .05), which is further supported by ARS and ALP staining indicating an increase in mineralized nodules formation along with elevated ALP content within these cells following treatment with this compound (p < .05). Finally, both HE staining as well as micro-CT imaging suggest potential benefits associated with using this compound including slowing alveolar bone resorption while simultaneously promoting junctional epithelium proliferation. CONCLUSIONS: Our in vitro and in vivo findings suggest that SC can effectively enhance the inflammatory response of PDLCs and promote their osteogenic differentiation ability under inflammatory conditions, indicating its potential as a promising therapeutic agent for improving periodontal inflammation and bone formation.


Assuntos
Reabsorção Óssea , Chalconas , Osteogênese , Ratos , Animais , Sincalida/farmacologia , Diferenciação Celular , Ligamento Periodontal , Células Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...