Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.981
Filter
1.
Braz J Med Biol Res ; 57: e13537, 2024.
Article in English | MEDLINE | ID: mdl-39258669

ABSTRACT

The clinical application of 5-fluorouracil (5-Fu), a potent chemotherapeutic agent, is often hindered by its well-documented cardiotoxic effects. Nevertheless, natural polyphenolic compounds like resveratrol (RES), known for their dual anti-tumor and cardioprotective properties, are potential adjunct therapeutic agents. In this investigation, we examined the combined utilization of RES and 5-Fu for the inhibition of gastric cancer using both in vitro and in vivo models, as well as their combined impact on cardiac cytotoxicity. Our study revealed that the co-administration of RES and 5-Fu effectively suppressed MFC cell viability, migration, and invasion, while also reducing tumor weight and volume. Mechanistically, the combined treatment prompted p53-mediated apoptosis and autophagy, leading to a considerable anti-tumor effect. Notably, RES mitigated the heightened oxidative stress induced by 5-Fu in cardiomyocytes, suppressed p53 and Bax expression, and elevated Bcl-2 levels. This favorable influence enhanced primary cardiomyocyte viability, decreased apoptosis and autophagy, and mitigated 5-Fu-induced cardiotoxicity. In summary, our findings suggested that RES holds promise as an adjunct therapy to enhance the efficacy of gastric cancer treatment in combination with 5-Fu, while simultaneously mitigating cardiotoxicity.


Subject(s)
Apoptosis , Cell Survival , Fluorouracil , Resveratrol , Stomach Neoplasms , Resveratrol/pharmacology , Resveratrol/therapeutic use , Fluorouracil/pharmacology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Cell Line, Tumor , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Stilbenes/pharmacology , Stilbenes/therapeutic use , Humans , Oxidative Stress/drug effects , Antimetabolites, Antineoplastic/pharmacology , Autophagy/drug effects , Male , Myocytes, Cardiac/drug effects , Mice , Cell Movement/drug effects
2.
Cell Mol Biol Lett ; 29(1): 114, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39198723

ABSTRACT

BACKGROUND: Stroke is a type of acute brain damage that can lead to a series of serious public health challenges. Demonstrating the molecular mechanism of stroke-related neural cell degeneration could help identify a more efficient treatment for stroke patients. Further elucidation of factors that regulate microglia and nuclear factor (erythroid-derived 2)-like 1 (Nrf1) may lead to a promising strategy for treating neuroinflammation after ischaemic stroke. In this study, we investigated the possible role of pterostilbene (PTS) in Nrf1 regulation in cell and animal models of ischaemia stroke. METHODS: We administered PTS, ITSA1 (an HDAC activator) and RGFP966 (a selective HDAC3 inhibitor) in a mouse model of middle cerebral artery occlusion-reperfusion (MCAO/R) and a model of microglial oxygen‒glucose deprivation/reperfusion (OGD/R). The brain infarct size, neuroinflammation and microglial availability were also determined. Dual-luciferase reporter, Nrf1 protein stability and co-immunoprecipitation assays were conducted to analyse histone deacetylase 3 (HDAC3)/Nrf1-regulated Nrf1 in an OGD/R-induced microglial injury model. RESULTS: We found that PTS decreased HDAC3 expression and activity, increased Nrf1 acetylation in the cell nucleus and inhibited the interaction of Nrf1 with p65 and p65 accumulation, which reduced infarct volume and neuroinflammation (iNOS/Arg1, TNF-α and IL-1ß levels) after ischaemic stroke. Furthermore, the CSF1R inhibitor PLX5622 induced elimination of microglia and attenuated the therapeutic effect of PTS following MCAO/R. In the OGD/R model, PTS relieved OGD/R-induced microglial injury and TNF-α and IL-1ß release, which were dependent on Nrf1 acetylation through the upregulation of HDAC3/Nrf1 signalling in microglia. However, the K105R or/and K139R mutants of Nrf1 counteracted the impact of PTS in the OGD/R-induced microglial injury model, which indicates that PTS treatment might be a promising strategy for ischaemia stroke therapy. CONCLUSION: The HDAC3/Nrf1 pathway regulates the stability and function of Nrf1 in microglial activation and neuroinflammation, which may depend on the acetylation of the lysine 105 and 139 residues in Nrf1. This mechanism was first identified as a potential regulatory mechanism of PTS-based neuroprotection in our research, which may provide new insight into further translational applications of natural products such as PTS.


Subject(s)
Histone Deacetylases , Ischemic Stroke , Mice, Inbred C57BL , Microglia , Neuroinflammatory Diseases , Stilbenes , Animals , Histone Deacetylases/metabolism , Microglia/metabolism , Microglia/drug effects , Mice , Ischemic Stroke/drug therapy , Ischemic Stroke/metabolism , Male , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Stilbenes/pharmacology , Stilbenes/therapeutic use , Disease Models, Animal , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/complications , Signal Transduction/drug effects , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Brain Ischemia/drug therapy , Brain Ischemia/metabolism
3.
Mediators Inflamm ; 2024: 5273198, 2024.
Article in English | MEDLINE | ID: mdl-39108992

ABSTRACT

Tendinopathy is one of the most frequent musculoskeletal disorders characterized by sustained tissue inflammation and oxidative stress, accompanied by extracellular matrix remodeling. Patients suffering from this pathology frequently experience pain, swelling, stiffness, and muscle weakness. Current pharmacological interventions are based on nonsteroidal anti-inflammatory drugs; however, the effectiveness of these strategies remains ambiguous. Accumulating evidence supports that oral supplementation of natural compounds can provide preventive, and possibly curative, effects. Vitamin C (Vit C), collagen peptides (Coll), resveratrol (Res), and astaxanthin (Asx) were reported to be endowed with potential beneficial effects based on their anti-inflammatory and antioxidant activities. Here, we analyzed the efficacy of a novel combination of these compounds (Mix) in counteracting proinflammatory (IL-1ß) and prooxidant (H2O2) stimuli in human tenocytes. We demonstrated that Mix significantly impairs IL-6-induced IL-1ß secretion, NF-κB nuclear translocation, and MMP-2 production; notably, a synergistic effect of Mix over the single compounds could be observed. Moreover, Mix was able to significantly counteract H2O2-triggered ROS production. Together, these results point out that Mix, a novel combination of Vit C, Coll, Resv, and Asx, significantly impairs proinflammatory and prooxidant stimuli in tenocytes, mechanisms that contribute to the onset of tendinopathies.


Subject(s)
Anti-Inflammatory Agents , Antioxidants , Ascorbic Acid , Collagen , Resveratrol , Tendinopathy , Tenocytes , Xanthophylls , Humans , Ascorbic Acid/pharmacology , Ascorbic Acid/therapeutic use , Resveratrol/pharmacology , Antioxidants/pharmacology , Xanthophylls/pharmacology , Xanthophylls/therapeutic use , Tendinopathy/drug therapy , Tendinopathy/metabolism , Collagen/metabolism , Anti-Inflammatory Agents/pharmacology , Tenocytes/metabolism , Tenocytes/drug effects , Interleukin-1beta/metabolism , Peptides/chemistry , Peptides/pharmacology , Hydrogen Peroxide/metabolism , Stilbenes/pharmacology , Stilbenes/therapeutic use , Reactive Oxygen Species/metabolism , NF-kappa B/metabolism , Cells, Cultured , Oxidative Stress/drug effects
4.
Acta Cir Bras ; 39: e395329, 2024.
Article in English | MEDLINE | ID: mdl-39109783

ABSTRACT

PURPOSE: To evaluate the neuroprotective effect of resveratrol, urapidil, and a combined administration of these drugs against middle cerebral artery occlusion (MCAO) induced ischemia/reperfusion (IR) injury model in rats. METHODS: Thirty-five rats were divided into five groups of seven animals each. Animals in IR, IR resveratrol (IRr), IR urapidil (IRu), and IR + combination of resveratrol and urapidil (IRc) were exposed to MCAO induced cerebral ischemia reperfusion injury model. Rats in IRr and IRu groups received 30-mg/kg resveratrol and 5-mg/kg urapidil respectively. Animals in IRc received a combined treatment of both drugs. At the end of the study, brain tissues were used for oxidative stress (malondialdehyde, glutathione, and superoxide dismutase), pro-apoptotic caspase-3, anti-apoptotic Bcl-2, and pro-inflammatory tumor necrosis factor-α cytokine level measurements. RESULTS: The MCAO model successfully replicated IR injury with significant histopathological changes, elevated tissue oxidative stress, and upregulated apoptotic and inflammatory protein expression in IR group compared to control group (p < 0.001). All parameters were significantly alleviated in IRr group compared to IR group (all p < 0.05). In IRu group, all parameters except for caspase-3 and Bcl-2 were also significantly different than IR group (all p < 0.05). The IRc group showed the biggest difference compared to IR group in all parameters (all p < 0.001). The IRc had higher superoxide dismutase and Bcl-2 levels, and lower caspase-3 levels compared to both IRr and IRu groups (all p < 0.05). Also, the IRc group had lower MDA and TNF-α levels compared to IRu group (all p < 0.05). CONCLUSIONS: The results indicate that combined treatment of resveratrol and urapidil may be a novel strategy to downregulate neurodegeneration in cerebral IR injury.


Subject(s)
Disease Models, Animal , Neuroprotective Agents , Oxidative Stress , Reperfusion Injury , Resveratrol , Stilbenes , Animals , Resveratrol/pharmacology , Resveratrol/therapeutic use , Reperfusion Injury/drug therapy , Reperfusion Injury/prevention & control , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Male , Oxidative Stress/drug effects , Stilbenes/therapeutic use , Stilbenes/pharmacology , Drug Therapy, Combination , Rats, Wistar , Infarction, Middle Cerebral Artery/drug therapy , Treatment Outcome , Rats , Tumor Necrosis Factor-alpha/analysis , Superoxide Dismutase/analysis , Superoxide Dismutase/metabolism , Malondialdehyde/analysis , Malondialdehyde/metabolism , Reproducibility of Results , Apoptosis/drug effects , Random Allocation , Brain Ischemia/drug therapy , Antioxidants/therapeutic use , Antioxidants/pharmacology , Caspase 3/metabolism , Caspase 3/analysis
5.
Thromb Res ; 241: 109111, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39098189

ABSTRACT

Thrombosis is the primary cause of death in patients with cancer. Resveratrol inhibits platelet activation, a crucial pathophysiological basis of thrombosis, in healthy individuals. However, its effects and mechanisms of action in patients with colon cancer remain unknown. Here, we investigated the effect of resveratrol on platelet adhesion and aggregation in patients with colon cancer. Through numerous in vitro and in vivo analyses, including flow cytometry, western blotting, ELISA, and immunofluorescence and colon cancer rat models, we demonstrated that resveratrol reduced thrombosis in patients with colon cancer by inhibiting the phosphorylation of the MAPK and activating the cyclic-GMP/vasodilator-stimulated phosphoprotein pathway. These findings demonstrate the potential of resveratrol in reducing thrombosis in patients with colon cancer and could be used to develop novel therapeutic strategies for this condition.


Subject(s)
Cell Adhesion Molecules , Colonic Neoplasms , Cyclic GMP , Phosphoproteins , Platelet Activation , Resveratrol , Thrombosis , Resveratrol/pharmacology , Resveratrol/therapeutic use , Thrombosis/drug therapy , Colonic Neoplasms/drug therapy , Colonic Neoplasms/complications , Humans , Animals , Platelet Activation/drug effects , Male , Rats , Cyclic GMP/metabolism , Cell Adhesion Molecules/metabolism , Phosphoproteins/metabolism , Female , Stilbenes/pharmacology , Stilbenes/therapeutic use , Rats, Sprague-Dawley , Middle Aged , Signal Transduction/drug effects , Blood Platelets/drug effects , Blood Platelets/metabolism , Platelet Aggregation/drug effects , Microfilament Proteins
6.
Zhonghua Nan Ke Xue ; 30(2): 145-150, 2024 Feb.
Article in Chinese | MEDLINE | ID: mdl-39177348

ABSTRACT

OBJECTIVE: To investigate the effects of resveratrol (RSV) on ovarian morphology, plasma anti-Müllerian hormone (AMH) and insulin-like growth factor 1 levels (IGF-1), and oxidative stress parameters in rats with polycystic ovary syndrome (PCOS). METHODS: Forty-six rats were randomly divided into a normal control (n = 12), a PCOS model control (n = 12), a rosiglitazone (RSG, n = 11), and an RSV group (n = 11). The PCOS model was established in the latter three groups by rejection of epidehydroandrosterone. The rats in the normal control and PCOS model control groups were treated by gavage of normal saline and those in the RSG and RSV groups by intragastric administration of RSG at 10 mg/(kg·d) and RSV at 3.0 mg/(kg·d), respectively. After 4 weeks of treatment, the ovarian histology was observed under the light microscope, the levels of plasma AMH and IGF-1 measured by ELISA, and the activities of glutathione peroxidase (GSH-Px), superoxide dismutase (SOD) and catalase (CAT) in the ovarian tissue detected using the Ellman, Sun and AEBI methods, respectively. RESULTS: After a 4-week treatment, statistically significant differences were observed in the above indicators between the normal control and PCOS model control groups (P<0.05). The rats treated with RSG and RSV also showed significant differences in these parameters from the model controls (P<0.05). CONCLUSION: RSV can enhance the local antioxidant capacity of the ovary, reduce the levels of AMH and IGF-1, and improve the morphology of the ovarian tissue in rats with PCOS, indicating its potential value in the treatment of PCOS.


Subject(s)
Antioxidants , Insulin-Like Growth Factor I , Ovary , Oxidative Stress , Polycystic Ovary Syndrome , Resveratrol , Stilbenes , Animals , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/metabolism , Female , Resveratrol/pharmacology , Rats , Ovary/drug effects , Ovary/metabolism , Oxidative Stress/drug effects , Insulin-Like Growth Factor I/metabolism , Stilbenes/pharmacology , Stilbenes/therapeutic use , Anti-Mullerian Hormone/blood , Superoxide Dismutase/metabolism , Glutathione Peroxidase/metabolism , Catalase/metabolism , Rats, Sprague-Dawley , Disease Models, Animal , Rosiglitazone/pharmacology
7.
Biochem Biophys Res Commun ; 729: 150358, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-38981401

ABSTRACT

Pterostilbene (PTE), a naturally occurring phenolic compound primarily found in blueberries, demonstrates neuroprotective properties. However, the role of PTE in Parkinson's disease (PD) remains unclear. This study aimed to investigate the neuroprotective role of PTE in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD animal model. Our findings demonstrate that administering PTE effectively reversed the diminished levels of dopamine in the striatum, thereby ameliorating motor impairments in the MPTP model. Moreover, PTE administration mitigated the loss of dopaminergic (DA) neurons and reduced the upregulation of α-synuclein (α-syn) induced by MPTP. Mechanistic analysis revealed that PTE administration inhibited the activation of microglia and astrocytes, as well as pro-inflammatory factors such as TNF-α and IL-1ß in the MPTP model. Additionally, PTE administration decreased MPTP-induced levels of reactive oxygen species (ROS) and malondialdehyde (MDA), while increasing total antioxidant capacity (TAOC) and superoxide dismutase (SOD) activity, thereby attenuating oxidative stress. Collectively, these findings demonstrate that PTE exerts neuroprotective effects in the MPTP mouse model of PD by suppressing neuroinflammation and oxidative stress. Thus, PTE holds promise as a therapeutic agent for PD.


Subject(s)
Mice, Inbred C57BL , Neuroinflammatory Diseases , Neuroprotective Agents , Oxidative Stress , Stilbenes , Animals , Oxidative Stress/drug effects , Stilbenes/pharmacology , Stilbenes/therapeutic use , Male , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Mice , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Reactive Oxygen Species/metabolism , MPTP Poisoning/drug therapy , MPTP Poisoning/metabolism , MPTP Poisoning/pathology , alpha-Synuclein/metabolism
8.
Chem Biol Drug Des ; 104(1): e14597, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39044124

ABSTRACT

Early detection of lung squamous cell carcinoma (LUSC) has a significant impact on clinical outcomes, and pterostilbene (PT) is a natural compound with promising anti-oncogenic activities. This study aimed to identify potential LUSC biomarkers through a series of bioinformatic analyses and clinical verification and explored the interaction between PT and selected biomarkers during the treatment of LUSC. The analysis of the expression profile of the clinical samples of LUSC was performed to identify dysexpressed genes (DEGs) and validated by IHC. The role of KANK3 in the anti-LUSC effects of PT was assessed with a series of in vitro and in vivo assays. 4335 DEGs were identified, including 1851 upregulated genes and 2484 downregulated genes. Survival analysis showed that KANK3 was significantly higher in patients with LUSC with an advanced tumor stage. In in vitro assays, PT suppressed cell viability, induced apoptosis, and inhibited migration and invasion in LUSC cell lines, which was associated with downregulation of KANK3. After the reinduction of the KANK3 level in LUSC cells, the anti-LUSC function of PT was impaired. In mice model, reinduction of KANK3 increased tumor growth and metastasis even under the treatment of PT. The findings outlined in the current study indicated that PT exerted anti-LUSC function in a KANK3 inhibition-dependent manner.


Subject(s)
Carcinoma, Squamous Cell , Lung Neoplasms , Stilbenes , Stilbenes/pharmacology , Stilbenes/chemistry , Stilbenes/therapeutic use , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Animals , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Mice , Cell Line, Tumor , Apoptosis/drug effects , Cell Movement/drug effects , Mice, Nude , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Male , Female , Mice, Inbred BALB C , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Cell Survival/drug effects , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/antagonists & inhibitors , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Cell Proliferation/drug effects
9.
PeerJ ; 12: e17687, 2024.
Article in English | MEDLINE | ID: mdl-39056050

ABSTRACT

Background: Cisplatin (CIS) is a broad-spectrum anticancer drug, with cytotoxic effects on either malignant or normal cells. We aimed to evaluate the hepatotoxicity in rats caused by CIS and its amelioration by the co-administration of either curcumin or resveratrol. Materials and Methods: Forty adult male rats divided into four equal groups: (control group): rats were given a saline solution (0.9%) once intraperitoneally, daily for the next 28 days; (cisplatin group): rats were given a daily oral dose of saline solution (0.9%) for 28 days after receiving a single dose of cisplatin (3.3 mg/kg) intraperitoneally for three successive days; (CIS plus curcumin/resveratrol groups): rats received the same previous dose of cisplatin (3.3 mg/kg) daily for three successive days followed by oral administration of either curcumin/resveratrol solution at a dose of (20 mg/kg) or (10 mg/kg) consequently daily for 28 days. Different laboratory tests (ALT, AST, ALP, bilirubin, oxidative stress markers) and light microscopic investigations were done. Results: Administration of CIS resulted in hepatotoxicity in the form of increased liver enzymes, oxidative stress markers; degenerative and apoptotic changes, the co-administration of CIS with either curcumin or resveratrol improved hepatotoxicity through improved microscopic structural changes, reduction in liver enzymes activity, decreased oxidative stress markers, improved degenerative, and apoptotic changes in liver tissues. Conclusion: Co-administration of either curcumin or resveratrol with cisplatin treatment could ameliorate hepatotoxicity caused by cisplatin in rats via anti-inflammatory and oxidative stress-apoptotic pathways.


Subject(s)
Apoptosis , Chemical and Drug Induced Liver Injury , Cisplatin , Curcumin , Oxidative Stress , Resveratrol , Animals , Resveratrol/pharmacology , Resveratrol/administration & dosage , Cisplatin/toxicity , Cisplatin/administration & dosage , Curcumin/pharmacology , Curcumin/administration & dosage , Oxidative Stress/drug effects , Male , Rats , Apoptosis/drug effects , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , Chemical and Drug Induced Liver Injury/etiology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/toxicity , Liver/drug effects , Liver/metabolism , Liver/pathology , Antioxidants/pharmacology , Antioxidants/administration & dosage , Stilbenes/administration & dosage , Stilbenes/pharmacology , Stilbenes/therapeutic use , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/administration & dosage , Rats, Wistar
11.
Biomater Adv ; 163: 213962, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39032435

ABSTRACT

Solid tumors create a hypoxic microenvironment and this character can be utilized for cancer therapy, but the hypoxia levels are insufficient to achieve satisfactory therapeutic benefits. Some tactics have been used to improve hypoxia, which however will cause side effects due to the uncontrolled drug release. We herein report near-infrared (NIR) photoactivatable three-in-one nanoagents (PCT) to aggravate tumor hypoxia and enable amplified photo-combinational chemotherapy. PCT are formed based on a thermal-responsive liposome nanoparticle containing three therapeutic agents: a hypoxia responsive prodrug tirapazamine (TPZ) for chemotherapy, a vascular targeting agent combretastatin A-4 (CA4) for vascular disturbance and a semiconducting polymer for both photodynamic therapy (PDT) and photothermal therapy (PTT). With NIR laser irradiation, PCT generate heat for PTT and destructing thermal-responsive liposomes to achieve activatable releases of TPZ and CA4. Moreover, PCT produce singlet oxygen (1O2) for PDT via consuming tumor oxygen. CA4 can disturb the blood vessels in tumor microenvironment to aggravate the hypoxic microenvironment, which results in the activation of TPZ for amplified chemotherapy. PCT thus enable PTT, PDT and hypoxia-amplified chemotherapy to afford a high therapeutic efficacy to almost absolutely eradicate subcutaneous 4 T1 tumors and effectively inhibit tumor metastases in lung and liver. This work presents an activatable three-in-one therapeutic nanoplatform with remotely controllable and efficient therapeutic actions to treat cancer.


Subject(s)
Infrared Rays , Liposomes , Nanoparticles , Photochemotherapy , Tirapazamine , Animals , Humans , Photochemotherapy/methods , Tirapazamine/pharmacology , Tirapazamine/chemistry , Tirapazamine/therapeutic use , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Mice , Tumor Microenvironment/drug effects , Cell Line, Tumor , Photothermal Therapy/methods , Stilbenes/pharmacology , Stilbenes/therapeutic use , Stilbenes/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Prodrugs/pharmacology , Prodrugs/chemistry , Prodrugs/therapeutic use , Photosensitizing Agents/therapeutic use , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Tumor Hypoxia/drug effects
12.
Biochem Pharmacol ; 226: 116373, 2024 08.
Article in English | MEDLINE | ID: mdl-38885772

ABSTRACT

Diabetic nephropathy (DN) is a complication of diabetes and is mainly characterized by renal fibrosis, which could be attributed to chronic kidney inflammation. Stimulator of interferon genes (STING), a linker between immunity and metabolism, could ameliorate various metabolic and inflammatory diseases. However, the regulatory role of STING in DN remains largely unexplored. In this study, knockdown of STING decreased extracellular matrix (ECM), pro-inflammatory, and fibrotic factors in high glucose (HG)-induced glomerular mesangial cells (GMCs), whereas overexpression of STING triggered the inflammatory fibrosis process, suggesting that STING was a potential target for DN. Polydatin (PD) is a glucoside of resveratrol and has been reported to ameliorate DN by inhibiting inflammatory responses. Nevertheless, whether PD improved DN via STING remains unclear. Here, transcriptomic profiling implied that the STING/NF-κB pathway might be an important target for PD. We further found that PD decreased the protein expression of STING, and subsequently suppressed the activation of downstream targets including TBK1 phosphorylation and NF-κB nuclear translocation, and eventually inhibited the production of ECM, pro-inflammatory and fibrotic factors in HG-induced GMCs. Notably, results of molecular docking, molecular dynamic simulations, surface plasmon resonance, cellular thermal shift assay and Co-immunoprecipitation assay indicated that PD directly bound to STING and restored the declined proteasome-mediated degradation of STING induced by HG. In diabetic mice, PD also inhibited the STING pathway and improved the pathological changes of renal inflammatory fibrosis. Our study elucidated the regulatory role of STING in DN, and the novel mechanism of PD treating DN via inhibiting STING expression.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Nephropathies , Fibrosis , Glucosides , Membrane Proteins , Mice, Inbred C57BL , Stilbenes , Glucosides/pharmacology , Glucosides/therapeutic use , Animals , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/pathology , Mice , Membrane Proteins/metabolism , Membrane Proteins/genetics , Fibrosis/drug therapy , Male , Stilbenes/pharmacology , Stilbenes/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Signal Transduction/drug effects , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Mesangial Cells/pathology , Humans
13.
BMC Cancer ; 24(1): 737, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38879516

ABSTRACT

BACKGROUND: Bladder cancer (BC) is among the most prevalent malignant urothelial tumors globally, yet the prognosis for patients with muscle-invasive bladder cancer (MIBC) remains dismal, with a very poor 5-year survival rate. Consequently, identifying more effective and less toxic chemotherapeutic alternatives is critical for enhancing clinical outcomes for BC patients. Isorhapontigenin (ISO), a novel stilbene isolated from a Gnetum found in certain provinces of China, has shown potential as an anticancer agent due to its diverse anticancer activities. Despite its promising profile, the specific anticancer effects of ISO on BC and the underlying mechanisms are still largely unexplored. METHODS: The anchorage-independent growth, migration and invasion of BC cells were assessed by soft agar and transwell invasion assays, respectively. The RNA levels of SOX2, miR-129 and SNHG1 were quantified by qRT-PCR, while the protein expression levels were validated through Western blotting. Furthermore, methylation-specific PCR was employed to assess the methylation status of the miR-129 promoter. Functional assays utilized siRNA knockdown, plasmid-mediated overexpression, and chemical inhibition approaches. RESULTS: Our study demonstrated that ISO treatment significantly reduced SNHG1 expression in a dose- and time-dependent manner in BC cells, leading to the inhibition of anchorage-independent growth and invasion in human basal MIBC cells. This effect was accompanied by the downregulation of MMP-2 and MMP-9 and the upregulation of the tumor suppressor PTEN. Further mechanistic investigations revealed that SOX2, a key upstream regulator of SNHG1, played a crucial role in mediating the ISO-induced transcriptional suppression of SNHG1. Additionally, we found that ISO treatment led to a decrease in DNMT3b protein levels, which in turn mediated the hypomethylation of the miR-129 promoter and the subsequent suppression of SOX2 mRNA 3'-UTR activity, highlighting a novel pathway through which ISO exerts its anticancer effects. CONCLUSIONS: Collectively, our study highlights the critical role of SNHG1 downregulation as well as its upstream DNMT3b/miR-129/SOX2 axis in mediating ISO anticancer activity. These findings not only elucidate the mechanism of action of ISO but also suggest novel targets for BC therapy.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases , DNA Methyltransferase 3B , Down-Regulation , Gene Expression Regulation, Neoplastic , RNA, Long Noncoding , Stilbenes , Urinary Bladder Neoplasms , Humans , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/metabolism , RNA, Long Noncoding/genetics , Cell Line, Tumor , Stilbenes/pharmacology , Stilbenes/therapeutic use , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Neoplasm Invasiveness , Cell Movement/drug effects , Cell Proliferation/drug effects , DNA Methylation/drug effects , MicroRNAs/genetics
14.
Int J Oncol ; 65(2)2024 Aug.
Article in English | MEDLINE | ID: mdl-38873997

ABSTRACT

Non­small cell lung cancer (NSCLC) is one of the major causes of cancer­related death worldwide. Cisplatin is a front­line chemotherapeutic agent in NSCLC. Nevertheless, subsequent harsh side effects and drug resistance limit its further clinical application. Polydatin (PD) induces apoptosis in various cancer cells by generating reactive oxygen species (ROS). However, underlying molecular mechanisms of PD and its effects on cisplatin­mediated antitumor activity in NSCLC remains unknown. MTT, colony formation, wound healing analyses and flow cytometry was employed to investigate the cell phenotypic changes and ROS generation. Relative gene and protein expressions were evaluated by reverse transcription­quantitative PCR and western blot analyses. The antitumor effects of PD, cisplatin and their combination were evaluated by mouse xenograft model. In the present study, it was found that PD in combination with cisplatin synergistically enhances the antitumor activity in NSCLC by stimulating ROS­mediated endoplasmic reticulum stress, and the C­Jun­amino­terminal kinase and p38 mitogen­activated protein kinase signaling pathways. PD treatment elevated ROS generation by promoting expression of NADPH oxidase 5 (NOX5), and NOX5 knockdown attenuated ROS­mediated cytotoxicity of PD in NSCLC cells. Mice xenograft model further confirmed the synergistic antitumor efficacy of combined therapy with PD and cisplatin. The present study exhibited a superior therapeutic strategy for some patients with NSCLC by combining PD and cisplatin.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Cisplatin , Drug Synergism , Glucosides , Lung Neoplasms , NADPH Oxidase 5 , Oxidative Stress , Reactive Oxygen Species , Stilbenes , Xenograft Model Antitumor Assays , Cisplatin/pharmacology , Cisplatin/therapeutic use , Glucosides/pharmacology , Glucosides/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Animals , Humans , Stilbenes/pharmacology , Stilbenes/therapeutic use , Mice , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Apoptosis/drug effects , A549 Cells , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Proliferation/drug effects , Endoplasmic Reticulum Stress/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Male
15.
Chem Biol Interact ; 398: 111110, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38876248

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is a prevalent chronic liver disorder that is linked to metabolic syndrome, mitochondrial dysfunction and impaired autophagy. Polydatin (PD), a natural polyphenol from Polygonum cuspidatum, exhibits various pharmacological effects and protects against NAFLD. The aim of this study was to reveal the molecular mechanisms and therapeutic potential of PD for NAFLD, with a focus on the role of mitochondrial autophagy mediated by sirtuin 3 (SIRT3), fork-head box O3 (FOXO3) and BCL2/adenovirus E1B 19 kDa protein-interacting protein 3 (BNIP3), and by PTEN-induced putative kinase 1 (PINK1) and parkin (PRKN). We combined network pharmacology analysis, animal models and cell culture experiments to show that PD could regulate the mitochondrial autophagy pathway by modulating several key genes related to mitochondrial function, and ameliorate the liver function, histopathology and mitochondrial biogenesis of NAFLD mice and hepatocytes by activating the SIRT3-FOXO3-BNIP3 axis and the PINK1-PRKN-dependent mechanism of mitochondrial autophagy. We also identified the core targets of PD, including SIRT3, FOXO3A, CASP3, PARKIN, EGFR, STAT3, MMP9 and PINK, and confirmed that silencing SIRT3 could significantly attenuate the beneficial effect of PD. This study provided novel theoretical and experimental support for PD as a promising candidate for NAFLD treatment, and also suggested new avenues and methods for investigating the role of mitochondrial autophagy in the pathogenesis and intervention of NAFLD.


Subject(s)
Forkhead Box Protein O3 , Glucosides , Mice, Inbred C57BL , Mitochondria , Non-alcoholic Fatty Liver Disease , Protein Kinases , Sirtuin 3 , Stilbenes , Ubiquitin-Protein Ligases , Animals , Forkhead Box Protein O3/metabolism , Sirtuin 3/metabolism , Sirtuin 3/genetics , Glucosides/pharmacology , Glucosides/therapeutic use , Glucosides/chemistry , Stilbenes/pharmacology , Stilbenes/therapeutic use , Mice , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Protein Kinases/metabolism , Male , Mitochondria/drug effects , Mitochondria/metabolism , Humans , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Autophagy/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Hepatocytes/drug effects , Hepatocytes/metabolism , Membrane Proteins
16.
Biomolecules ; 14(6)2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38927082

ABSTRACT

New furan, thiophene, and triazole oximes were synthesized through several-step reaction paths to investigate their potential for the development of central nervous systems (CNS)-active and cholinesterase-targeted therapeutics in organophosphorus compound (OP) poisonings. Treating patients with acute OP poisoning is still a challenge despite the development of a large number of oxime compounds that should have the capacity to reactivate acetylcholinesterase (AChE) and butyrylcholinesterase (BChE). The activity of these two enzymes, crucial for neurotransmission, is blocked by OP, which has the consequence of disturbing normal cholinergic nerve signal transduction in the peripheral and CNS, leading to a cholinergic crisis. The oximes in use have one or two pyridinium rings and cross the brain-blood barrier poorly due to the quaternary nitrogen. Following our recent study on 2-thienostilbene oximes, in this paper, we described the synthesis of 63 heterostilbene derivatives, of which 26 oximes were tested as inhibitors and reactivators of AChE and BChE inhibited by OP nerve agents-sarin and cyclosarin. While the majority of oximes were potent inhibitors of both enzymes in the micromolar range, we identified several oximes as BChE or AChE selective inhibitors with the potential for drug development. Furthermore, the oximes were poor reactivators of AChE; four heterocyclic derivatives reactivated cyclosarin-inhibited BChE up to 70%, and cis,trans-5 [2-((Z)-2-(5-((E)-(hydroxyimino)methyl)thiophen-2-yl)vinyl)benzonitrile] had a reactivation efficacy comparable to the standard oxime HI-6. In silico analysis and molecular docking studies, including molecular dynamics simulation, connected kinetic data to the structural features of these oximes and confirmed their productive interactions with the active site of cyclosarin-inhibited BChE. Based on inhibition and reactivation and their ADMET properties regarding lipophilicity, CNS activity, and hepatotoxicity, these compounds could be considered for further development of CNS-active reactivators in OP poisoning as well as cholinesterase-targeted therapeutics in neurodegenerative diseases such as Alzheimer's and Parkinson's.


Subject(s)
Acetylcholinesterase , Butyrylcholinesterase , Cholinesterase Inhibitors , Molecular Docking Simulation , Oximes , Triazoles , Oximes/chemistry , Oximes/pharmacology , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/chemical synthesis , Butyrylcholinesterase/metabolism , Butyrylcholinesterase/chemistry , Acetylcholinesterase/metabolism , Acetylcholinesterase/chemistry , Humans , Triazoles/chemistry , Triazoles/pharmacology , Triazoles/chemical synthesis , Stilbenes/chemistry , Stilbenes/pharmacology , Stilbenes/therapeutic use , Stilbenes/chemical synthesis , Cholinesterase Reactivators/chemistry , Cholinesterase Reactivators/pharmacology , Cholinesterase Reactivators/chemical synthesis , Cholinesterase Reactivators/therapeutic use , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacology , Central Nervous System/drug effects , Central Nervous System/metabolism
17.
Exp Neurol ; 378: 114835, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38789024

ABSTRACT

Cerebral Palsy (CP) is the main motor disorder in childhood resulting from damage to the developing brain. Treatment perspectives are required to reverse the primary damage caused by the early insult and consequently to recover motor skills. Resveratrol has been shown to act as neuroprotection with benefits to skeletal muscle. This study aimed to investigate the effects of neonatal resveratrol treatment on neurodevelopment, skeletal muscle morphology, and cerebellar damage in CP model. Wistar rat pups were allocated to four experimental groups (n = 15/group) according CP model and treatment: Control+Saline (CS), Control+Resveratrol (CR), CP + Saline (CPS), and CP + Resveratrol (CPR). CP model associated anoxia and sensorimotor restriction. CP group showed delay in the disappearance of the palmar grasp reflex (p < 0.0001) and delay in the appearance of reflexes of negative geotaxis (p = 0.01), and free-fall righting (p < 0.0001), reduced locomotor activity and motor coordination (p < 0.05) than CS group. These motor skills impairments were associated with a reduction in muscle weight (p < 0.001) and area and perimeter of soleus end extensor digitorum longus muscle fibers (p < 0.0001), changes in muscle fibers typing pattern (p < 0.05), and the cerebellum showed signs of neuroinflammation due to elevated density and percentage of activated microglia in the CPS group compared to CS group (p < 0.05). CP animals treated with resveratrol showed anticipation of the appearance of negative geotaxis and free-fall righting reflexes (p < 0.01), increased locomotor activity (p < 0.05), recovery muscle fiber types pattern (p < 0.05), and reversal of the increase in density and the percentage of activated microglia in the cerebellum (p < 0.01). Thus, we conclude that neonatal treatment with resveratrol can contribute to the recovery of the delay neurodevelopment resulting from experimental CP due to its action in restoring the skeletal muscle morphology and reducing neuroinflammation from cerebellum.


Subject(s)
Animals, Newborn , Cerebellum , Cerebral Palsy , Microglia , Muscle, Skeletal , Rats, Wistar , Resveratrol , Resveratrol/pharmacology , Animals , Cerebellum/drug effects , Cerebellum/pathology , Rats , Microglia/drug effects , Microglia/pathology , Cerebral Palsy/drug therapy , Cerebral Palsy/pathology , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Disease Models, Animal , Stilbenes/pharmacology , Stilbenes/therapeutic use , Male , Recovery of Function/drug effects , Female
19.
Physiol Res ; 73(2): 239-251, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38710061

ABSTRACT

Oxygen therapy provides an important treatment for preterm and low-birth-weight neonates, however, it has been shown that prolonged exposure to high levels of oxygen (hyperoxia) is one of the factors contributing to the development of bronchopulmonary dysplasia (BPD) by inducing lung injury and airway hyperreactivity. There is no effective therapy against the adverse effects of hyperoxia. Therefore, this study was undertaken to test the hypothesis that natural phytoalexin resveratrol will overcome hyperoxia-induced airway hyperreactivity, oxidative stress, and lung inflammation. Newborn rats were exposed to hyperoxia (fraction of inspired oxygen - FiO2>95 % O2) or ambient air (AA) for seven days. Resveratrol was supplemented either in vivo (30 mg·kg-1·day-1) by intraperitoneal administration or in vitro to the tracheal preparations in an organ bath (100 mikroM). Contractile and relaxant responses were studied in tracheal smooth muscle (TSM) using the in vitro organ bath system. To explain the involvement of nitric oxide in the mechanisms of the protective effect of resveratrol against hyperoxia, a nitric oxide synthase inhibitor - Nomega-nitro-L-arginine methyl ester (L-NAME), was administered in some sets of experiments. The superoxide dismutase (SOD) and glutathione peroxidase (GPx) activities and the tumor necrosis factor-alpha (TNF-alpha) and interleukin-1beta (IL-1beta) levels in the lungs were determined. Resveratrol significantly reduced contraction and restored the impaired relaxation of hyperoxia-exposed TSM (p<0.001). L-NAME reduced the inhibitory effect of resveratrol on TSM contractility, as well as its promotion relaxant effect (p<0.01). Resveratrol preserved the SOD and GPx activities and decreased the expression of TNF-alpha and IL-1beta in hyperoxic animals. The findings of this study demonstrate the protective effect of resveratrol against hyperoxia-induced airway hyperreactivity and lung damage and suggest that resveratrol might serve as a therapy to prevent the adverse effects of neonatal hyperoxia. Keywords: Bronchopulmonary dysplasia, Hyperoxia, Airway hyperreactivity, Resveratrol, Pro-inflammatory cytokines.


Subject(s)
Animals, Newborn , Bronchopulmonary Dysplasia , Disease Models, Animal , Oxidative Stress , Pneumonia , Resveratrol , Animals , Resveratrol/pharmacology , Oxidative Stress/drug effects , Bronchopulmonary Dysplasia/prevention & control , Bronchopulmonary Dysplasia/metabolism , Pneumonia/prevention & control , Pneumonia/metabolism , Pneumonia/chemically induced , Rats , Hyperoxia/complications , Hyperoxia/metabolism , Stilbenes/pharmacology , Stilbenes/therapeutic use , Antioxidants/pharmacology , Bronchial Hyperreactivity/prevention & control , Bronchial Hyperreactivity/metabolism , Bronchial Hyperreactivity/physiopathology , Bronchial Hyperreactivity/chemically induced , Rats, Sprague-Dawley , Male
20.
Chem Biol Interact ; 396: 111058, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38761877

ABSTRACT

Pterostilbene (PTE), a natural phenolic compound, has exhibited promising anticancer properties in the preclinical treatment of cervical cancer (CC). This study aims to comprehensively investigate the potential targets and mechanisms underlying PTE's anticancer effects in CC, thereby providing a theoretical foundation for its future clinical application and development. To accomplish this, we employed a range of methodologies, including network pharmacology, bioinformatics, and computer simulation, with specific techniques such as WGCNA, PPI network construction, ROC curve analysis, KM survival analysis, GO functional enrichment, KEGG pathway enrichment, molecular docking, MDS, and single-gene GSEA. Utilizing eight drug target prediction databases, we have identified a total of 532 potential targets for PTE. By combining CC-related genes from the GeneCards disease database with significant genes derived from WGCNA analysis of the GSE63514 dataset, we obtained 7915 unique CC-related genes. By analyzing the intersection of the 7915 CC-related genes and the 2810 genes that impact overall survival time in CC, we identified 690 genes as crucial for CC. Through the use of a Venn diagram, we discovered 36 overlapping targets shared by PTE and CC. We have constructed a PPI network and identified 9 core candidate targets. ROC and KM curve analyses subsequently revealed IL1B, EGFR, IL1A, JUN, MYC, MMP1, MMP3, and ANXA5 as the key targets modulated by PTE in CC. GO and KEGG pathway enrichment analyses indicated significant enrichment of these key targets, primarily in the MAPK and IL-17 signaling pathways. Molecular docking analysis verified the effective binding of PTE to all nine key targets. MDS results showed that the protein-ligand complex between MMP1 and PTE was the most stable among the nine targets. Additionally, GSEA enrichment analysis suggested a potential link between elevated MMP1 expression and the activation of the IL-17 signaling pathway. In conclusion, our study has identified key targets and uncovered the molecular mechanism behind PTE's anticancer activity in CC, establishing a firm theoretical basis for further exploration of PTE's pharmacological effects in CC therapy.


Subject(s)
Computational Biology , Molecular Docking Simulation , Network Pharmacology , Stilbenes , Uterine Cervical Neoplasms , Humans , Stilbenes/pharmacology , Stilbenes/chemistry , Stilbenes/therapeutic use , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/genetics , Female , Protein Interaction Maps/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL