Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 35.490
Filtrer
1.
Zhongguo Zhong Yao Za Zhi ; 49(14): 3868-3877, 2024 Jul.
Article de Chinois | MEDLINE | ID: mdl-39099360

RÉSUMÉ

The study investigated the effect of Compound Shougong Powder(CSGP) on the biological functions of triple-negative breast cancer(TNBC) cells and whether its mechanism of action was related to the epithelial-mesenchymal transition(EMT) signaling pathway. TNBC cells(MDA-MB-231 and BT-549) were treated with different concentrations of CSGP-containing serum. MTS assay was used to detect the effect of CSGP on the proliferation of TNBC cells. The EdU staining was used to detect the effect of CSGP on the proliferation of TNBC cells. Flow cytometry was used to examine the impact of CSGP on apoptosis of TNBC cells. Wound-healing and Transwell assays were used to evaluate the effects of different concentrations of CSGP on the migration and invasion capabilities of TNBC cells. RNA sequencing technology was utilized to elucidate its mechanism. Subsequently, qRT-PCR was performed to measure the mRNA expression levels of E-cadherin, N-cadherin, Slug, Snail, Vimentin, Twist, Zinc finger E-box-Binding homeobox 1(Zeb1), and Zinc finger E-box-Binding homeobox 2(Zeb2). Western blot was used to assess the protein expression levels of Slug, Vimentin, and E-cadherin. After intervention with CSGP, the proliferation of MDA-MB-231 and BT-549 cells significantly decreased, while the apoptosis rate markedly increased. The expression levels of the epithelial marker protein E-cadherin significantly increased, while the expression levels of the EMT-related transcription factors Slug and Vimentin showed a decrease. In conclusion, CSGP inhibits the EMT, thereby suppressing the malignant progression of TNBC.


Sujet(s)
Apoptose , Prolifération cellulaire , Médicaments issus de plantes chinoises , Transition épithélio-mésenchymateuse , Tumeurs du sein triple-négatives , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Humains , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/génétique , Tumeurs du sein triple-négatives/métabolisme , Tumeurs du sein triple-négatives/anatomopathologie , Médicaments issus de plantes chinoises/pharmacologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Apoptose/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Poudres/composition chimique , Cadhérines/génétique , Cadhérines/métabolisme
2.
Zhongguo Zhong Yao Za Zhi ; 49(14): 3894-3900, 2024 Jul.
Article de Chinois | MEDLINE | ID: mdl-39099363

RÉSUMÉ

This study explored the effect of Tianma Gouteng Decoction on oxidative stress induced by angiotensin Ⅱ(AngⅡ) in vascular smooth muscle cell(VSMC) and its molecular mechanism. Primary rat VSMC were cultured using tissue block method, and VSMC were identified by α-actin immunofluorescence staining. AngⅡ at a concentration of 1×10~(-6) mol·L~(-1) was used as the stimulating factor, and Sprague Dawley(SD) rats were orally administered with Tianma Gouteng Decoction to prepare drug serum. Rat VSMC were divided into normal group, model group, Chinese medicine group, and inhibitor(3-methyladenine, 3-MA) group. Cell counting kit-8(CCK-8) assay was used to detect cell proliferation activity. Bromodeoxyuridine(BrdU) flow cytometry was used to detect cell cycle. Transwell assay was used to detect cell migration ability. Enzyme-linked immunosorbent assay(ELISA) was used to detect the activity of superoxide dismutase(SOD), catalase(CAT), and malondialdehyde(MDA) in VSMC. The intracellular reactive oxygen species(ROS) fluorescence intensity was detected using DCFH-DA fluorescent probe. Western blot was used to detect the expression of PTEN-induced putative kinase 1(PINK1), Parkin, p62, and microtubule-associated protein 1A/1B-light chain 3(LC3-Ⅱ) proteins in VSMC. The results showed that Tianma Gouteng Decoction-containing serum at a concentration of 8% could significantly inhibit VSMC growth after 48 hours of intervention. Compared with the normal group, the model group showed significantly increased cell proliferation activity and migration, significantly decreased levels of SOD and CAT, significantly increased levels of MDA, significantly enhanced ROS fluorescence intensity, significantly decreased expression of PINK1, Parkin, and LC3-Ⅱ proteins, and significantly increased expression of p62 protein. Compared with the model group, the Chinese medicine group showed significantly reduced cell proliferation activity and migration, significantly increased levels of SOD and CAT, significantly decreased levels of MDA, significantly weakened ROS fluorescence intensity, significantly increased expression of PINK1, Parkin, and LC3-Ⅱ proteins, and significantly decreased expression of p62 protein. Compared with the Chinese medicine group, the addition of the mitochondrial autophagy inhibitor 3-MA could block the intervention of Tianma Gouteng Decoction-containing serum on VSMC proliferation, migration, mitochondrial autophagy, and oxidative stress levels, with statistically significant differences. In summary, Tianma Gouteng Decoction has good antioxidant activity and can inhibit cell proliferation and migration. Its mechanism of action may be related to the activation of the mitochondrial autophagy PINK1/Parkin signaling pathway.


Sujet(s)
Angiotensine-II , Prolifération cellulaire , Médicaments issus de plantes chinoises , Muscles lisses vasculaires , Stress oxydatif , Protein kinases , Rat Sprague-Dawley , Ubiquitin-protein ligases , Animaux , Médicaments issus de plantes chinoises/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Rats , Muscles lisses vasculaires/effets des médicaments et des substances chimiques , Muscles lisses vasculaires/cytologie , Muscles lisses vasculaires/métabolisme , Mâle , Prolifération cellulaire/effets des médicaments et des substances chimiques , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Protein kinases/métabolisme , Protein kinases/génétique , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/métabolisme , Espèces réactives de l'oxygène/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Cellules cultivées , Superoxide dismutase/métabolisme
3.
Med Oncol ; 41(9): 217, 2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-39102060

RÉSUMÉ

Multiple myeloma (MM) is a neoplastic condition resulting from the uncontrolled expansion of B-cell-derived plasma cells. The importance of angiogenesis in MM development has also been demonstrated. Extracellular vesicles (EVs) have vital functions in interactions between neighboring cells, such as angiogenesis. The objective of this in vitro study was to examine the transfection and angiogenesis effects of MM-EVs on endothelial cells (ECs) upon treatment with Tetrahydroisoquinoline (THIQ) as a bioactive organic compound derivative from isoquinoline. Following treatment of multiple myeloma cells (U266) with THIQ, MM-EVs were harvested and transmigrated to human umbilical vein endothelial cells (HUVEC) in a co-culture model. EVs transmigration was traced by flow cytometry. Correspondingly, the expression of angiogenic genes and/or proteins in U266 cells and HUVECs was measured by RT-PCR and ELISA methods. Likewise, the proliferation and migration of HUVECs treated with THIQ-treated MM-EVs were visualized and estimated by performing both tube formation and scratch wound healing methods. Surprisingly, the anti-angiogenic effect of THIQ-treated MM-EVs was evident by the decreased expression of CD34, VEGFR2, and IL-6 at the mRNA and/or protein levels after internalization of MM-EVs in HUVEC. Finally, tube formation and scratch wound healing experiments showed inhibition of HUVEC cell proliferation and migration by THIQ-treated MM-EVs compared to control MM-EVs. MM-EVs derived from THIQ-treated myeloma cells (U266) inhibited angiogenesis in HUVECs. This phenomenon is coordinated by the internalized THIQ-treated MM-EVs in HUVECs, and ultimately the reduction of angiogenic factors and inhibition of tube formation and scratch wound healing.


Sujet(s)
Mouvement cellulaire , Vésicules extracellulaires , Cellules endothéliales de la veine ombilicale humaine , Myélome multiple , Néovascularisation pathologique , Tétrahydroisoquinoléines , Humains , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Myélome multiple/anatomopathologie , Myélome multiple/traitement médicamenteux , Myélome multiple/métabolisme , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/effets des médicaments et des substances chimiques , Néovascularisation pathologique/traitement médicamenteux , Néovascularisation pathologique/anatomopathologie , Néovascularisation pathologique/métabolisme , Tétrahydroisoquinoléines/pharmacologie , Mouvement cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Techniques de coculture , Inhibiteurs de l'angiogenèse/pharmacologie ,
4.
Cell Mol Biol (Noisy-le-grand) ; 70(7): 206-211, 2024 Jul 28.
Article de Anglais | MEDLINE | ID: mdl-39097873

RÉSUMÉ

The objective of this study was to investigate the impact of ethyl pyruvate (EP), an HMGB1 inhibitor, on ESCC cells both in vitro and in vivo. The viability of ESCC cells was assessed using the MTT method to evaluate the correlation between EP and cell viability. A scratch test was used to investigate the relationship between EP and cell migration and invasion. The effects of EP on tumor growth and survival in cancerous nude mice were examined using a tumor formation model. Immunohistochemical staining was performed to evaluate the expression levels of HMGB1, TLR4, and MyD88 in tumor tissues. EP, an anti-HMGB1 inhibitor, inhibited ESCC cell proliferation and metastasis in vitro and in vivo. Furthermore, compared with the control treatment, EP improved the activity, diet, and drinking behaviour of nude mice; inhibited tumour growth; and led to lower protein expression levels of HMGB1, TLR4, and MyD88. EP has the potential to regulate the HMGB1/TLR4-MyD88 signaling pathway, thereby inhibiting the proliferation and metastasis of ESCC, suppressing tumor growth, improving quality of life, and serving as an effective drug for ESCC treatment.


Sujet(s)
Carcinome épidermoïde , Prolifération cellulaire , Tumeurs de l'oesophage , Carcinome épidermoïde de l'oesophage , Protéine HMGB1 , Souris nude , Facteur de différenciation myéloïde-88 , Pyruvates , Récepteur de type Toll-4 , Animaux , Pyruvates/pharmacologie , Humains , Protéine HMGB1/métabolisme , Protéine HMGB1/génétique , Récepteur de type Toll-4/métabolisme , Récepteur de type Toll-4/génétique , Facteur de différenciation myéloïde-88/métabolisme , Facteur de différenciation myéloïde-88/génétique , Lignée cellulaire tumorale , Carcinome épidermoïde de l'oesophage/traitement médicamenteux , Carcinome épidermoïde de l'oesophage/anatomopathologie , Carcinome épidermoïde de l'oesophage/métabolisme , Tumeurs de l'oesophage/anatomopathologie , Tumeurs de l'oesophage/traitement médicamenteux , Tumeurs de l'oesophage/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Carcinome épidermoïde/traitement médicamenteux , Carcinome épidermoïde/anatomopathologie , Carcinome épidermoïde/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe , Mouvement cellulaire/effets des médicaments et des substances chimiques , Souris , Transduction du signal/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Survie cellulaire/effets des médicaments et des substances chimiques , Mâle
5.
Int J Mol Sci ; 25(15)2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39125994

RÉSUMÉ

Ocimum gratissimum (O. gratissimum), a medicinal herb with antifungal and antiviral activities, has been found to prevent liver injury and liver fibrosis and induce apoptosis in hepatocellular carcinoma (HCC) cells. In this study, we evaluated the effect of aqueous extracts of O. gratissimum (OGE) on improving the efficacy of chemotherapeutic drugs in HCC cells. Proteomic identification and functional assays were used to uncover the critical molecules responsible for OGE-induced sensitization mechanisms. The antitumor activity of OGE in combination with a chemotherapeutic drug was evaluated in a mouse orthotopic tumor model, and serum biochemical tests were further utilized to validate liver function. OGE sensitized HCC cells to the chemotherapeutic drug cisplatin. Proteomic analysis and Western blotting validation revealed the sensitization effect of OGE, likely achieved through the inhibition of breast cancer type 1 susceptibility protein (BRCA1). Mechanically, OGE treatment resulted in BRCA1 protein instability and increased proteasomal degradation, thereby synergistically increasing cisplatin-induced DNA damage. Moreover, OGE effectively inhibited cell migration and invasion, modulated epithelial-to-mesenchymal transition (EMT), and impaired stemness properties in HCC cells. The combinatorial use of OGE enhanced the efficacy of cisplatin and potentially restored liver function in a mouse orthotopic tumor model. Our findings may provide an alternate approach to improving chemotherapy efficacy in HCC.


Sujet(s)
Protéine BRCA1 , Carcinome hépatocellulaire , Cisplatine , Tumeurs du foie , Ocimum , Extraits de plantes , Cisplatine/pharmacologie , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Animaux , Humains , Ocimum/composition chimique , Extraits de plantes/pharmacologie , Extraits de plantes/composition chimique , Souris , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Protéine BRCA1/métabolisme , Protéine BRCA1/génétique , Lignée cellulaire tumorale , Tests d'activité antitumorale sur modèle de xénogreffe , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Altération de l'ADN/effets des médicaments et des substances chimiques
6.
Zhonghua Xue Ye Xue Za Zhi ; 45(6): 577-585, 2024 Jun 14.
Article de Chinois | MEDLINE | ID: mdl-39134490

RÉSUMÉ

Objective: To investigate the effect and molecular mechanism of hesperadin in inducing ferroptosis in chronic myeloid leukemia cell line K562 cells. Methods: The effects of hesperadin on the viability, proliferation, and migration of K562 cells were detected though CCK8, EDU-594, and Transwell assays, and the apoptotic rate of K562 cells was detected by flow cytometry. In addition, C11-BODIPY and FerroOrange were utilized to detect intracellular lipid peroxidation and Fe(2+) levels. Meanwhile, the expression levels of ferroptosis-associated protein solute carrier family 7 member 11 (SLC7A11) and glutathione peroxidase 4 (GPX4) in cells were detected through Western blot. Lipid peroxidation and Fe(2+) levels were also detected after transfection of cells with SLC7A11 overexpression plasmid. Results: Hesperadin decreased cell viability in a dose-dependent manner with IC(50) of 0.544 µmol/L. Hesperadin concentrations of 0.4 and 0.8 µmol/L were selected for follow-up experiments. EDU-594, Transwell, and flow cytometry showed significantly decreased proliferation and migration rate of K562 cells after 0.4 and 0.8 µmol/L hesperadin treatment for 24 h, and the apoptosis rate was significantly increased compared with the control group (P<0.05). Western blot indicated a downregulated expression of the antiapoptotic protein Bcl-2 and an elevated expression of proapoptotic proteins Bax and Caspase-3. Moreover, hesperadin increased intracellular lipid peroxidation and Fe(2+) levels compared with the control treatment (P<0.05). The combination of ferroptosis inhibitor (Fer-1) and hesperadin could reverse the effect of hesperadin on K562 cells. The mRNA and protein levels of ferroptosis-related genes SLC7A11 and GPX4 were significantly decreased in the 0.8 µmol/L hesperadin-treated group (P<0.05). SLC7A11 overexpression can inhibit hesperadin effect and alleviate ferroptosis. Conclusion: Hesperadin can promote ferroptosis in K562 cells by regulating the SLC7A11/GPX4 axis.


Sujet(s)
Prolifération cellulaire , Ferroptose , Leucémie myéloïde chronique BCR-ABL positive , Humains , Ferroptose/effets des médicaments et des substances chimiques , Cellules K562 , Prolifération cellulaire/effets des médicaments et des substances chimiques , Leucémie myéloïde chronique BCR-ABL positive/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Peroxydation lipidique/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Système y+ de transport d'acides aminés/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques
7.
Int J Nanomedicine ; 19: 7751-7773, 2024.
Article de Anglais | MEDLINE | ID: mdl-39099796

RÉSUMÉ

Endogenous stem cell homing refers to the transport of endogenous mesenchymal stem cells (MSCs) to damaged tissue. The paradigm of using well-designed biomaterials to induce resident stem cells to home in to the injured site while coordinating their behavior and function to promote tissue regeneration is known as endogenous regenerative medicine (ERM). ERM is a promising new avenue in regenerative therapy research, and it involves the mobilizing of endogenous stem cells for homing as the principal means through which to achieve it. Comprehending how mesenchymal stem cells home in and grasp the influencing factors of mesenchymal stem cell homing is essential for the understanding and design of tissue engineering. This review summarizes the process of MSC homing, the factors influencing the homing process, analyses endogenous stem cell homing studies of interest in the field of skin tissue repair, explores the integration of endogenous homing promotion strategies with cellular therapies and details tissue engineering strategies that can be used to modulate endogenous homing of stem cells. In addition to providing more systematic theories and ideas for improved materials for endogenous tissue repair, this review provides new perspectives to explore the complex process of tissue remodeling to enhance the rational design of biomaterial scaffolds and guide tissue regeneration strategies.


Sujet(s)
Matériaux biocompatibles , Cellules souches mésenchymateuses , Ingénierie tissulaire , Cicatrisation de plaie , Humains , Cellules souches mésenchymateuses/cytologie , Cicatrisation de plaie/effets des médicaments et des substances chimiques , Cicatrisation de plaie/physiologie , Matériaux biocompatibles/composition chimique , Matériaux biocompatibles/pharmacologie , Ingénierie tissulaire/méthodes , Animaux , Médecine régénérative/méthodes , Structures d'échafaudage tissulaires/composition chimique , Mouvement cellulaire/effets des médicaments et des substances chimiques , Peau , Transplantation de cellules souches mésenchymateuses/méthodes
8.
PLoS One ; 19(8): e0304010, 2024.
Article de Anglais | MEDLINE | ID: mdl-39150901

RÉSUMÉ

M64HCl, which has drug-like properties, is a water-soluble Focal Adhesion Kinase (FAK) activator that promotes murine mucosal healing after ischemic or NSAID-induced injury. Since M64HCl has a short plasma half-life in vivo (less than two hours), it has been administered as a continuous infusion with osmotic minipumps in previous animal studies. However, the effects of more transient exposure to M64HCl on monolayer wound closure remained unclear. Herein, we compared the effects of shorter M64HCl treatment in vitro to continuous treatment for 24 hours on monolayer wound closure. We then investigated how long FAK activation and downstream ERK1/2 activation persist after two hours of M64HCl treatment in Caco-2 cells. M64HCl concentrations immediately after washing measured by mass spectrometry confirmed that M64HCl had been completely removed from the medium while intracellular concentrations had been reduced by 95%. Three-hour and four-hour M64HCl (100 nM) treatment promoted epithelial sheet migration over 24 hours similar to continuous 24-hour exposure. 100nM M64HCl did not increase cell number. Exposing cells twice with 2-hr exposures of M64HCl during a 24-hour period had a similar effect. Both FAK inhibitor PF-573228 (10 µM) and ERK kinase (MEK) inhibitor PD98059 (20 µM) reduced basal wound closure in the absence of M64HCl, and each completely prevented any stimulation of wound closure by M64HCl. Rho kinase inhibitor Y-27632 (20 µM) stimulated Caco-2 monolayer wound closure but no further increase was seen with M64HCl in the presence of Y-27632. M64HCl (100 nM) treatment for 3 hours stimulated Rho kinase activity. M64HCl decreased F-actin in Caco-2 cells. Furthermore, a two-hour treatment with M64HCl (100 nM) stimulated sustained FAK activation and ERK1/2 activation for up to 16 and hours 24 hours, respectively. These results suggest that transient M64HCl treatment promotes prolonged intestinal epithelial monolayer wound closure by stimulating sustained activation of the FAK/ERK1/2 pathway. Such molecules may be useful to promote gastrointestinal mucosal repair even with a relatively short half-life.


Sujet(s)
Muqueuse intestinale , Cicatrisation de plaie , Humains , Cicatrisation de plaie/effets des médicaments et des substances chimiques , Cellules Caco-2 , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/métabolisme , Focal adhesion protein-tyrosine kinases/métabolisme , Focal adhesion kinase 1/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques , Pyridines/pharmacologie , Animaux , Amides/pharmacologie
9.
Sci Rep ; 14(1): 19003, 2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-39152152

RÉSUMÉ

Gastric cancer (GC) remains a global disease with a high mortality rate, the lack of effective treatments and the high toxicity of side effects are primary causes for its poor prognosis. Hence, urgent efforts are needed to find safe and effective therapeutic strategies. Gypenoside (Gyp) is a widely used natural product that regulates blood glucose to improve disease progression with few toxic side effects. Given the crucial role of abnormal glycometabolism in driving tumor malignancy, it is important to explore the association between Gyp and glycometabolism in GC and understand the mechanism of action by which Gyp influences glycometabolism. In this study, we demonstrated that Gyp suppresses GC proliferation and migration both in vitro and in vivo. We identified that Gyp suppresses the malignant progression of GC by inhibiting glycolysis using network pharmacology and metabolomics. Transcriptome analysis revealed that the Hippo pathway is a key regulator of glycolysis by Gyp in GC. Furthermore, Gyp induced upregulation of LATS1/2 proteins, leading to increased YAP phosphorylation and decreased TAZ protein expression. The YAP agonist XMU-MP-1 rescued the inhibitory effect of Gyp on GC proliferation by reversing glycolysis. These findings confirmed that Gyp inhibits GC proliferation by targeting glycolysis through the Hippo pathway. Our study examined the role of Gyp in the malignant progression of GC, explored its therapeutic prospects, elucidated a mechanism by which Gyp suppresses GC proliferation through interference with the glycolytic process, thus providing a potential novel therapeutic strategy for GC patients.


Sujet(s)
Prolifération cellulaire , Glycolyse , Gynostemma , Voie de signalisation Hippo , Protein-Serine-Threonine Kinases , Tumeurs de l'estomac , Humains , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/traitement médicamenteux , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Glycolyse/effets des médicaments et des substances chimiques , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Lignée cellulaire tumorale , Animaux , Transduction du signal/effets des médicaments et des substances chimiques , Souris , Mouvement cellulaire/effets des médicaments et des substances chimiques , Extraits de plantes/pharmacologie , Souris nude , Tests d'activité antitumorale sur modèle de xénogreffe , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
10.
Sci Rep ; 14(1): 18345, 2024 08 07.
Article de Anglais | MEDLINE | ID: mdl-39112598

RÉSUMÉ

Pressure ulcers (PU) are caused by persistent long-term pressure, which compromises the integrity of the epidermis, dermis, and subcutaneous adipose tissue layer by layer, making it difficult to heal. Platelet products such as platelet lysate (PL) can promote tissue regeneration by secreting numerous growth factors based on clinical studies on skin wound healing. However, the components of PL are difficult to retain in wounds. Gelatin methacrylate (GelMA) is a photopolymerizable hydrogel that has lately emerged as a promising material for tissue engineering and regenerative medicine. The PL liquid was extracted, flow cytometrically detected for CD41a markers, and evenly dispersed in the GelMA hydrogel to produce a surplus growth factor hydrogel system (PL@GM). The microstructure of the hydrogel system was observed under a scanning electron microscope, and its sustained release efficiency and biological safety were tested in vitro. Cell viability and migration of human dermal fibroblasts, and tube formation assays of human umbilical vein endothelial cells were applied to evaluate the ability of PL to promote wound healing and regeneration in vitro. Real-time polymerase chain reaction (PCR) and western blot analyses were performed to elucidate the skin regeneration mechanism of PL. We verified PL's therapeutic effectiveness and histological analysis on the PU model. PL promoted cell viability, migration, wound healing and angiogenesis in vitro. Real-time PCR and western blot indicated PL suppressed inflammation and promoted collagen I synthesis by activating STAT3. PL@GM hydrogel system demonstrated optimal biocompatibility and favorable effects on essential cells for wound healing. PL@GM also significantly stimulated PU healing, skin regeneration, and the formation of subcutaneous collagen and blood vessels. PL@GM could accelerate PU healing by promoting fibroblasts to migrate and secrete collagen and endothelial cells to vascularize. PL@GM promises to be an effective and convenient treatment modality for PU, like chronic wound treatment.


Sujet(s)
, Plaquettes , Gélatine , Méthacrylates , Escarre , Peau , Cicatrisation de plaie , Animaux , Humains , Souris , /effets des médicaments et des substances chimiques , Plaquettes/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Gélatine/composition chimique , Gélatine/pharmacologie , Cellules endothéliales de la veine ombilicale humaine , Hydrogels/composition chimique , Méthacrylates/composition chimique , Méthacrylates/pharmacologie , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Escarre/thérapie , Régénération/effets des médicaments et des substances chimiques , Peau/vascularisation , Peau/effets des médicaments et des substances chimiques , Peau/métabolisme , Peau/anatomopathologie , Facteur de transcription STAT-3/métabolisme , Cicatrisation de plaie/effets des médicaments et des substances chimiques
11.
Sci Rep ; 14(1): 18377, 2024 08 08.
Article de Anglais | MEDLINE | ID: mdl-39112593

RÉSUMÉ

The effects of calcitonin gene-related peptide (CGRP) on atherosclerosis remain unclear. We used apolipoprotein E-deficient (ApoE-/-) mice to generate double-knockout ApoE-/-:CGRP-/- mice lacking alpha CGRP. ApoE-/-:CGRP-/- mice exhibited larger atherosclerotic plaque areas, peritoneal macrophages with enhanced migration functions, and elevated levels of the inflammatory cytokine tumor necrosis factor (TNF)-⍺. Thus, we also explored whether inhibiting TNF-⍺ could improve atherosclerosis in ApoE-/-:CGRP-/- mice by administering etanercept intraperitoneally once a week (5 mg/kg) alongside a high-fat diet for 2 weeks. This treatment led to significant reductions in aortic root lesion size, atherosclerotic plaque area and macrophage migration in ApoE-/-:CGRP-/- mice compared with mice treated with human IgG (5 mg/kg). We further examined whether results observed in ApoE-/-:CGRP-/- mice could similarly be obtained by administering a humanized monoclonal CGRP antibody, galcanezumab, to ApoE-/- mice. ApoE-/- mice were subcutaneously administered galcanezumab at an initial dose of 50 mg/kg, followed by a dose of 30 mg/kg in the second week. Galcanezumab administration did not affect systolic blood pressure, serum lipid levels, or macrophage migration but led to a significant increase in lipid deposition at the aortic root. These findings suggest that alpha CGRP plays a critical role in inhibiting the progression of atherosclerosis.


Sujet(s)
Apolipoprotéines E , Athérosclérose , Peptide relié au gène de la calcitonine , Souris knockout , Plaque d'athérosclérose , Animaux , Athérosclérose/métabolisme , Athérosclérose/génétique , Athérosclérose/anatomopathologie , Peptide relié au gène de la calcitonine/métabolisme , Souris , Apolipoprotéines E/déficit , Apolipoprotéines E/génétique , Plaque d'athérosclérose/anatomopathologie , Plaque d'athérosclérose/métabolisme , Plaque d'athérosclérose/génétique , Alimentation riche en graisse/effets indésirables , Facteur de nécrose tumorale alpha/métabolisme , Mâle , Souris invalidées pour les gènes ApoE , Modèles animaux de maladie humaine , Humains , Anticorps monoclonaux humanisés/pharmacologie , Étanercept/pharmacologie , Souris de lignée C57BL , Mouvement cellulaire/effets des médicaments et des substances chimiques , Aorte/métabolisme , Aorte/anatomopathologie , Aorte/effets des médicaments et des substances chimiques
12.
Sci Rep ; 14(1): 18444, 2024 08 08.
Article de Anglais | MEDLINE | ID: mdl-39117725

RÉSUMÉ

Glioblastoma (GBM) is associated with a median survival rate of less than 15 months, necessitating innovative treatment approaches. This study investigates the safety and efficacy of the low-frequency magnetic field (LFMF) OM-100 instrument in GBM therapy. In vitro experiments utilized normal astrocyte and GBM cell lines, determining that OM-100 at 100 kHz for 72 h selectively targeted GBM cells without harming normal cells. Subsequent analyses revealed OM-100's impact on cell viability, apoptosis, migration, invasion, reactive oxide species levels, and PD-L1 expression. In vivo studies on mice with U87-induced GBM demonstrated OM-100's synergy with anti-PD-1 therapy, leading to significant tumor volume reduction and increased apoptosis. Notably, OM-100 exhibited safety in healthy mice. Overall, OM-100 could enhance anti-PD-1 immunotherapy effectiveness probably by directly inhibiting tumor proliferation and migration as well as promoting PD-L1 expression, offering a promising therapeutic strategy for GBM treatment.


Sujet(s)
Prolifération cellulaire , Glioblastome , Immunothérapie , Récepteur-1 de mort cellulaire programmée , Glioblastome/thérapie , Glioblastome/traitement médicamenteux , Glioblastome/anatomopathologie , Glioblastome/métabolisme , Animaux , Souris , Humains , Lignée cellulaire tumorale , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/métabolisme , Immunothérapie/méthodes , Prolifération cellulaire/effets des médicaments et des substances chimiques , Antigène CD274/métabolisme , Antigène CD274/antagonistes et inhibiteurs , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/thérapie , Apoptose/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Tests d'activité antitumorale sur modèle de xénogreffe , Magnétothérapie/méthodes , Survie cellulaire/effets des médicaments et des substances chimiques
13.
Molecules ; 29(15)2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39125009

RÉSUMÉ

Human pharyngeal squamous cell carcinoma (HPSCC) is the most common malignancy in the head and neck region, characterized by high mortality and a propensity for metastasis. Fucoxanthin, a carotenoid isolated from brown algae, exhibits pharmacological properties associated with the suppression of tumor proliferation and metastasis. Nevertheless, its potential to inhibit HPSCC proliferation and metastasis has not been fully elucidated. This study represents the first exploration of the inhibitory effects of fucoxanthin on two human pharyngeal squamous carcinoma cell lines (FaDu and Detroit 562), as well as the mechanisms underlying those effects. The results showed dose-dependent decreases in the proliferation, migration, and invasion of HPSCC cells after fucoxanthin treatment. Further studies indicated that fucoxanthin caused a significant reduction in the expression levels of proteins in the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mechanistic target of rapamycin (mTOR) pathway, as well as the downstream proteins matrix metalloproteinase (MMP)-2 and MMP-9. Specific activators of PI3K/AKT reversed the effects of fucoxanthin on these proteins, as well as on cell proliferation and metastasis, in FaDu and Detroit 562 cells. Molecular docking assays confirmed that fucoxanthin strongly interacted with PI3K, AKT, mTOR, MMP-2, and MMP-9. Overall, fucoxanthin, a functional food component, is a potential therapeutic agent for HPSCC.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Sérine-thréonine kinases TOR , Xanthophylles , Humains , Sérine-thréonine kinases TOR/métabolisme , Xanthophylles/pharmacologie , Xanthophylles/composition chimique , Protéines proto-oncogènes c-akt/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Phosphatidylinositol 3-kinases/métabolisme , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Tumeurs du pharynx/traitement médicamenteux , Tumeurs du pharynx/anatomopathologie , Tumeurs du pharynx/métabolisme , Carcinome épidermoïde/traitement médicamenteux , Carcinome épidermoïde/métabolisme , Carcinome épidermoïde/anatomopathologie , Métastase tumorale , Simulation de docking moléculaire
14.
Nutrients ; 16(15)2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39125270

RÉSUMÉ

Glioblastoma (GBM) is a severe form of brain tumor that has a high fatality rate. It grows aggressively and most of the time results in resistance to traditional treatments like chemo- and radiotherapy and surgery. Biodiversity, beyond representing a big resource for human well-being, provides several natural compounds that have shown great potential as anticancer drugs. Many of them are being extensively researched and significantly slow GBM progression by reducing the proliferation rate, migration, and inflammation and also by modulating oxidative stress. Here, the use of some natural compounds, such as Allium lusitanicum, Succisa pratensis, and Dianthus superbus, was explored to tackle GBM; they showed their impact on cell number reduction, which was partially given by cell cycle quiescence. Furthermore, a reduced cell migration ability was reported, accomplished by morphological cytoskeleton changes, which even highlighted a mesenchymal-epithelial transition. Furthermore, metabolic studies showed an induced cell oxidative stress modulation and a massive metabolic rearrangement. Therefore, a new therapeutic option was suggested to overcome the limitations of conventional treatments and thereby improve patient outcomes.


Sujet(s)
Tumeurs du cerveau , Glioblastome , Glioblastome/traitement médicamenteux , Glioblastome/anatomopathologie , Humains , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/anatomopathologie , Mouvement cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Stress oxydatif/effets des médicaments et des substances chimiques , Produits biologiques/pharmacologie , Produits biologiques/usage thérapeutique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Extraits de plantes/pharmacologie , Antinéoplasiques d'origine végétale/pharmacologie , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie
15.
Chem Biol Drug Des ; 104(2): e14602, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39134897

RÉSUMÉ

Catalpol, a natural iridoid glycoside, has potential therapeutic benefits, including anti-inflammatory and neuroprotective effects. Investigating catalpol's role in angiogenesis is critical for understanding its potential therapeutic applications, particularly in diseases where modulating angiogenesis is beneficial. This study investigates catalpol's influence on angiogenesis and its mechanisms, combining network pharmacology and in vitro experiments. The target genes corresponding to the catalpol were analyzed by SwissTargetPrediction. Then angiogenesis-related targets were acquired from databases like GeneCards. Subsequently, the Database for Annotation, Visualization and Integrated Discovery was employed for Gene Ontology and pathway analysis, while Cytoscape visualized protein interactions. The effect of catalpol on viability and angiogenesis of HUVECs was further examined using Cell Counting Kit-8 and angiogenesis assays. RT-qPCR and western blot were applied to check the expression of angiogenesis-related proteins. Totally, 312 target genes of catalpol and 823 angiogenesis-related targets were obtained with 56 common targets leading to PPI network analysis, highlighting hub genes (AKT1, EGFR, STAT3, MAPK3, and CASP3). These hub genes were mainly enriched in lipid and atherosclerosis pathway and EGFR-related pathway. The in vitro experimental results showed that catalpol achieved a concentration-dependent increase in HUVECs viability. Catalpol also promoted the migration and angiogenesis of HUVECs and up-regulated the expression of EGFR. EGFR knockdown inhibited the effect of catalpol on HUVECs. Catalpol promotes angiogenesis in HUVECs by upregulating EGFR and angiogenesis-related proteins, indicating its potential therapeutic application in vascular-related diseases.


Sujet(s)
Cellules endothéliales de la veine ombilicale humaine , Glucosides d'iridoïdes , Pharmacologie des réseaux , Humains , Glucosides d'iridoïdes/pharmacologie , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Cartes d'interactions protéiques/effets des médicaments et des substances chimiques , Récepteurs ErbB/métabolisme , Récepteurs ErbB/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques ,
16.
Sci Rep ; 14(1): 18757, 2024 08 13.
Article de Anglais | MEDLINE | ID: mdl-39138279

RÉSUMÉ

Hepatocellular carcinoma (HCC) still presents poor prognosis with low overall survival rates and limited therapeutic options available. Recently, attention has been drawn to peptidomic analysis, an emerging field of proteomics for the exploration of new potential peptide drugs for the treatment of various diseases. However, research on the potential function of HCC peptides is lacking. Here, we analyzed the peptide spectrum in HCC tissues using peptidomic techniques and explored the potentially beneficial peptides involved in HCC. Changes in peptide profiles in HCC were examined using liquid chromatography-mass spectrometry (LC-MS/MS). Analyze the physicochemical properties and function of differently expressed peptides using bioinformatics. The effect of candidate functional peptides on HCC cell growth and migration was evaluated using the CCK-8, colony formation, and transwell assays. Transcriptome sequencing analysis and western blot were employed to delve into the mode of action of potential peptide on HCC. Peptidomic analysis of HCC tissue yielded a total of 8683 peptides, of which 452 exhibited up-regulation and 362 showed down-regulation. The peptides that were differentially expressed, according to bioinformatic analysis, were closely linked to carbon metabolism and the mitochondrial inner membrane. The peptide functional validation identified a novel peptide, PDLC (peptide derived from liver cancer), which was found to dramatically boost HCC cell proliferation through the Ras/Raf/MEK/ERK signaling cascade. Our research defined the peptide's properties and pattern of expression in HCC and identified a novel peptide, PDLC, with a function in encouraging HCC progression, offering an entirely new potential therapeutic target the disease.


Sujet(s)
Carcinome hépatocellulaire , Prolifération cellulaire , Tumeurs du foie , Système de signalisation des MAP kinases , Protéomique , Kinases raf , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Tumeurs du foie/génétique , Humains , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Kinases raf/métabolisme , Protéomique/méthodes , Lignée cellulaire tumorale , Protéines G ras/métabolisme , Protéines G ras/génétique , Peptides/métabolisme , Peptides/pharmacologie , Mouvement cellulaire/effets des médicaments et des substances chimiques , Spectrométrie de masse en tandem , Régulation de l'expression des gènes tumoraux
17.
Mol Cancer ; 23(1): 165, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39138527

RÉSUMÉ

BACKGROUND: Overexpression of receptor tyrosine kinase-like orphan receptor 1 (ROR1) contributes to cancer cell proliferation, survival and migration, playing crucial roles in tumor development. ROR1 has been proposed as a potential therapeutic target for cancer treatment. This study aimed to develop novel humanized ROR1 monoclonal antibodies and investigate their anti-tumor effects. METHODS: ROR1 expression in tumor tissues and cell lines was analyzed by immunohistochemistry and flow cytometry. Antibodies from mouse hybridomas were humanized by the complementarity-determining region (CDR) grafting technique. Surface plasmon resonance spectroscopy, ELISA assay and flow cytometry were employed to characterize humanized antibodies. In vitro cellular assay and in vivo mouse experiment were conducted to comprehensively evaluate anti-tumor activity of these antibodies. RESULTS: ROR1 exhibited dramatically higher expression in lung adenocarcinoma, liver cancer and breast cancer, and targeting ROR1 by short-hairpin RNAs significantly inhibited proliferation and migration of cancer cells. Two humanized ROR1 monoclonal antibodies were successfully developed, named h1B8 and h6D4, with high specificity and affinity to ROR1 protein. Moreover, these two antibodies effectively suppressed tumor growth in the lung cancer xenograft mouse model, c-Myc/Alb-cre liver cancer transgenic mouse model and MMTV-PyMT breast cancer mouse model. CONCLUSIONS: Two humanized monoclonal antibodies targeting ROR1, h1B8 and h6D4, were successfully developed and exhibited remarkable anti-tumor activity in vivo.


Sujet(s)
Anticorps monoclonaux humanisés , Prolifération cellulaire , Récepteurs orphelins de type récepteur à tyrosine kinase , Tests d'activité antitumorale sur modèle de xénogreffe , Récepteurs orphelins de type récepteur à tyrosine kinase/métabolisme , Récepteurs orphelins de type récepteur à tyrosine kinase/antagonistes et inhibiteurs , Récepteurs orphelins de type récepteur à tyrosine kinase/immunologie , Animaux , Humains , Souris , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Anticorps monoclonaux humanisés/pharmacologie , Anticorps monoclonaux humanisés/usage thérapeutique , Femelle , Mouvement cellulaire/effets des médicaments et des substances chimiques , Tumeurs/traitement médicamenteux , Tumeurs/immunologie , Tumeurs/anatomopathologie , Tumeurs/thérapie , Tumeurs/métabolisme , Souris transgéniques , Modèles animaux de maladie humaine , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Tumeurs du poumon/immunologie
18.
Cell Commun Signal ; 22(1): 397, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39138495

RÉSUMÉ

BACKGROUND: Gastric cancer (GC) is a prevalent malignancy with limited therapeutic options for advanced stages. This study aimed to identify novel therapeutic targets for GC by profiling HSP90 client kinases. METHODS: We used mass spectrometry-based activity-based protein profiling (ABPP) with a desthiobiotin-ATP probe, combined with sensitivity analysis of HSP90 inhibitors, to profile kinases in a panel of GC cell lines. We identified kinases regulated by HSP90 in inhibitor-sensitive cells and investigated the impact of MASTL knockdown on GC cell behavior. Global proteomic analysis following MASTL knockdown was performed, and bioinformatics tools were used to analyze the resulting data. RESULTS: Four kinases-MASTL, STK11, CHEK1, and MET-were identified as HSP90-regulated in HSP90 inhibitor-sensitive cells. Among these, microtubule-associated serine/threonine kinase-like (MASTL) was upregulated in GC and associated with poor prognosis. MASTL knockdown decreased migration, invasion, and proliferation of GC cells. Global proteomic profiling following MASTL knockdown revealed NEDD4-1 as a potential downstream mediator of MASTL in GC progression. NEDD4-1 was also upregulated in GC and associated with poor prognosis. Similar to MASTL inhibition, NEDD4-1 knockdown suppressed migration, invasion, and proliferation of GC cells. CONCLUSIONS: Our multi-proteomic analyses suggest that targeting MASTL could be a promising therapy for advanced gastric cancer, potentially through the reduction of tumor-promoting proteins including NEDD4-1. This study enhances our understanding of kinase signaling pathways in GC and provides new insights for potential treatment strategies.


Sujet(s)
Prolifération cellulaire , Protein-Serine-Threonine Kinases , Protéome , Protéomique , Tumeurs de l'estomac , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/traitement médicamenteux , Humains , Lignée cellulaire tumorale , Protéomique/méthodes , Protéome/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Mouvement cellulaire/effets des médicaments et des substances chimiques , Protéines du choc thermique HSP90/métabolisme , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Protéines du choc thermique HSP90/génétique , Ubiquitine protéine ligases NEDD4/métabolisme , Ubiquitine protéine ligases NEDD4/génétique , Régulation de l'expression des gènes tumoraux , Thérapie moléculaire ciblée , Protéines associées aux microtubules
19.
Mediators Inflamm ; 2024: 8828367, 2024.
Article de Anglais | MEDLINE | ID: mdl-39144184

RÉSUMÉ

Background: Bladder cancer (BC) is one of the most common malignancies of the urogenital system. This study assessed the nucleotide-binding oligomerization domain and leucine-rich repeat and pyrin domain-containing protein 3 (NLRP3) in BC as well as the effects of cryptotanshinone on changes in BC malignant behaviors and NLRP3 expression under a lipopolysaccharide (LPS)-induced inflammatory microenvironment. Methods: BC tissue specimens from 62 patients were collected for immunohistochemical detection of NLRP3 protein. BC and normal urothelial cell lines were cultured for the detection of NLRP3 mRNA and protein. Then, BC cells were pretreated with LPS to mimic the inflammatory tumor microenvironment. Next, these cells were incubated with a low or high dose of cryptotanshinone to assess its effects on tumor cell malignant behaviors as well as transfected with NLRP3 cDNA to confirm the role of NLRP3 in BC cells in vitro. Results: High NLRP3 expression was associated with larger tumor diameters (>2 cm), muscle invasion, and metastasis. The levels of NLRP3 mRNA and protein were greater in BC cells than in normal urothelial cells. LPS pretreatment significantly promoted NLRP3 and inflammatory cytokine expression in BC cells, and induced cell viability, migration, and invasion. However, cryptotanshinone was able to reduce the LPS-induced increase of NLRP3 and inflammatory cytokine expression as well as the BC cell malignant progression. NLRP3 overexpression using NLRP3 cDNA further promoted BC cell malignant progression after LPS stimulation and reversed cryptotanshinone-reduced LPS-induced BC cell malignant behaviors. Conclusion: NLRP3 might possess oncogenic activity in BC, and the antitumor activity of cryptotanshinone in BC in vitro might be related to its inhibition of NLRP3 expression.


Sujet(s)
Lipopolysaccharides , Protéine-3 de la famille des NLR contenant un domaine pyrine , Phénanthrènes , Tumeurs de la vessie urinaire , Humains , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Tumeurs de la vessie urinaire/métabolisme , Tumeurs de la vessie urinaire/traitement médicamenteux , Tumeurs de la vessie urinaire/anatomopathologie , Phénanthrènes/pharmacologie , Mâle , Lignée cellulaire tumorale , Femelle , Adulte d'âge moyen , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Sujet âgé , Mouvement cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Inflammation/métabolisme
20.
Steroids ; 210: 109487, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39106908

RÉSUMÉ

Gastric cancer (GC)-diabetes co-morbidity is nowadays growing into a rising concern. However, no separate treatment procedures have been outlined for such patients. Phytochemicals and their derivatives can therefore be used as therapeutics as they have greater effectiveness, reduced toxicity, and a reduced likelihood of developing multi-drug resistance in cancer treatments. The present study intended to assess the therapeutic efficacy of Shatavarin-IV - a major steroidal saponin from the roots of Asparagus racemosus, in human gastric adenocarcinoma cell line under hyperglycemic conditions and explore its mechanism of action in controlling GC progression. For the present study, AGS cells were incubated in high glucose-containing media and the effects of Shatavarin-IV therein have been evaluated. Cell proliferation, confocal microscopic imaging, flow-cytometric analysis for cell cycle and apoptosis, immunoblotting, zymography, reverse zymography, wound-healing, colony formation, and invasion assays were performed. Shatavarin-IV has a prominent effect on AGS cell proliferation; with IC50 of 2.463 µ M under hyperglycemic conditions. Shatavarin-IV induces cell cycle arrest at the G0/G1 phase, thereby preventing hyperglycemia-induced excessive cell proliferation that later on leads to apoptotic cell death at 36 h of incubation. Shatavarin-IV further inhibits the migratory and invasive potential of AGS cells by altering the expression patterns of different EMT markers. It also inhibits MMP-9 while promoting TIMP-1 activity and expression; thereby regulating ECM turnover. This is the first report demonstrating the therapeutic efficacy of Shatavarin-IV against AGS cells grown in hyperglycemic conditions, implicating new insights into the treatment paradigm of patients with GC-diabetes co-morbidity.


Sujet(s)
Asparagus , Prolifération cellulaire , Transition épithélio-mésenchymateuse , Hyperglycémie , Saponines , Humains , Saponines/pharmacologie , Saponines/composition chimique , Saponines/isolement et purification , Asparagus/composition chimique , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Hyperglycémie/traitement médicamenteux , Lignée cellulaire tumorale , Cycle cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/traitement médicamenteux , Mouvement cellulaire/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE