Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Int Immunopharmacol ; 119: 110236, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37148772

ABSTRACT

Colorectal cancer (CRC) is currently recognized as the third most prevalent cancer worldwide. Vinpocetine is a synthetic derivative of the vinca alkaloid vincamine. It has been found effective in ameliorating the growth and progression of cancerous cells. However, its pharmacological effect on colon damage remains elusive. Hence, in this study, we have shown the role of vinpocetine in DMH-induced colon carcinogenesis. At first, male albino Wistar rats were administered with DMH consistently for four weeks to induce pre-neoplastic colon damage. Afterward, animals were treated with vinpocetine (4.2 and 8.4 mg/kg/day p.o.) for 15 days. Serum samples were collected to assess the physiological parameters, including ELISA and NMR metabolomics. Colon from all the groups was collected and processed separately for histopathology and western blot analysis. Vinpocetine attenuated the altered plasma parameters; lipid profile and showed anti-proliferative action as evidenced by suppressed COX-2 stimulation and decreased levels of IL-1ß, IL-2, IL-6, and IL-10. Vinpocetine is significantly effective in preventing CRC which may be associated with its anti-inflammatory and antioxidant potential. Accordingly, vinpocetine could serve as a potential anticancer agent for CRC treatment and thus be considered for future clinical and therapeutic research.


Subject(s)
Antineoplastic Agents , Vinca Alkaloids , Rats , Male , Animals , Cytokines/pharmacology , Vinca Alkaloids/therapeutic use , Vinca Alkaloids/pharmacology , Colon/pathology , Antineoplastic Agents/pharmacology , Rats, Wistar
2.
BBA Adv ; 2: 100046, 2022.
Article in English | MEDLINE | ID: mdl-37082584

ABSTRACT

Fluvoxamine's (FLX's) anticancer potential was investigated in pre-clinical research utilizing a DMH-induced colorectal cancer (CRC) rat model. qRT-PCR and immunoblotting validated the mechanistic investigation. The CRC condition was induced in response to COX-2 and IL-6, however, following FLX therapy, the condition returned to normal. FLX's anti-CRC potential may be attributable to COX-2 inhibition since this molecular activity was more apparent for COX-2 than IL-6. FLX repaired the altered metabolites linked to CRC rats, according to 1H-NMR analysis. FLX was shown to be similar to 5-FU in terms of tumor protection, which may be useful in future medication development.

3.
Redox Biol ; 43: 101965, 2021 07.
Article in English | MEDLINE | ID: mdl-33933881

ABSTRACT

Excessive ingestion of the common analgesic acetaminophen (APAP) leads to severe hepatotoxicity. Here we identify G protein ß5 (Gß5), elevated in livers from APAP overdose patients, as a critical regulator of cell death pathways and autophagic signaling in APAP-exposed liver. Liver-specific knockdown of Gß5 in mice protected the liver from APAP-dependent fibrosis, cell loss, oxidative stress, and inflammation following either acute or chronic APAP administration. Conversely, overexpression of Gß5 in liver was sufficient to drive hepatocyte dysfunction and loss. In hepatocytes, Gß5 depletion ameliorated mitochondrial dysfunction, allowed for maintenance of ATP generation and mitigated APAP-induced cell death. Further, Gß5 knockdown also reversed impacts of APAP on kinase cascades (e.g. ATM/AMPK) signaling to mammalian target of rapamycin (mTOR), a master regulator of autophagy and, as a result, interrupted autophagic flux. Though canonically relegated to nuclear DNA repair pathways, ATM also functions in the cytoplasm to control cell death and autophagy. Indeed, we now show that Gß5 forms a direct, stable complex with the FAT domain of ATM, important for autophosphorylation-dependent kinase activation. These data provide a viable explanation for these novel, G protein-independent actions of Gß5 in liver. Thus, Gß5 sits at a critical nexus in multiple pathological sequelae driving APAP-dependent liver damage.


Subject(s)
Acetaminophen , Chemical and Drug Induced Liver Injury , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Chemical and Drug Induced Liver Injury/metabolism , GTP-Binding Proteins/metabolism , Hepatocytes , Humans , Liver/metabolism , Mice , Mice, Inbred C57BL , Oxidative Stress
4.
J Liposome Res ; 31(3): 304-315, 2021 Sep.
Article in English | MEDLINE | ID: mdl-32901571

ABSTRACT

ß-sitosterol (BS), a phytosterol, exhibits ameliorative effects on hepatocellular carcinoma (HCC) due to its antioxidant activities. However, its poor aqueous solubility and negotiated bioavailability and short elimination half-life is a huge limitation for its therapeutic applications. To overcome these two shortcomings, BS-loaded niosomes were made to via, film hydration method and process parameters were optimized using a three-factor Box-Behnken design. The optimized formulation (BSF) was further surface-modified with polyethylene glycol (PEG). The resulting niosomes (BSMF) have spherical shapes, particle sizes, 219.6 ± 1.98 nm with polydispersity index (PDI) and zeta potential of 0.078 ± 0.04 and -19.54 ± 0.19 mV, respectively. The drug loading, entrapment efficiency, and drug release at 24 h of the BSMF were found to be 16.72 ± 0.09%, 78.04 ± 0.92%, and 75.10 ± 3.06%, respectively. Moreover, BSMF showed significantly greater cytotoxic potentials on Hep G2 cells with an enhanced cellular uptake relative to pure BS and BSF. The BSMF also displayed potentially improved curative property of HCC in albino wistar rat. Thus, the BSMF could be one of the promising therapeutic modalities for HCC treatment in terms of targeting potential resulting in enhanced therapeutic efficacy.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Drug Carriers , Liposomes/therapeutic use , Liver Neoplasms/drug therapy , Polyethylene Glycols , Rats , Sitosterols
5.
Front Pharmacol ; 12: 823285, 2021.
Article in English | MEDLINE | ID: mdl-35095533

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common tumors affecting a large population worldwide, with the fifth and seventh greatest mortality rates among men and women, respectively, and the third prime cause of mortality among cancer victims. Dimethyl itaconate (DI) has been reported to be efficacious in colorectal cancer by decreasing IL-1ß release from intestinal epithelial cells. In this study, diethylnitrosamine (DEN)-induced HCC in male albino Wistar rats was treated with DI as an anticancer drug. The function and molecular mechanism of DI against HCC in vivo were assessed using histopathology, enzyme-linked immunosorbent assay (ELISA), and Western blot studies. Metabolomics using 1H-NMR was used to investigate metabolic profiles. As per molecular insights, DI has the ability to trigger mitochondrial apoptosis through iNOS- and eNOS-induced activation of the NF-κB/Bcl-2 family of proteins, CytC, caspase-3, and caspase-9 signaling cascade. Serum metabolomics investigations using 1H-NMR revealed that aberrant metabolites in DEN-induced HCC rats were restored to normal following DI therapy. Furthermore, our data revealed that the DI worked as an anti-HCC agent. The anticancer activity of DI was shown to be equivalent to that of the commercial chemotherapeutic drug 5-fluorouracil.

6.
Antioxid Redox Signal ; 32(11): 766-784, 2020 04 10.
Article in English | MEDLINE | ID: mdl-31830804

ABSTRACT

Aims: Nonsteroidal anti-inflammatory drugs (NSAIDs), among the most commonly used drugs worldwide, are associated with gastrointestinal (GI) complications that severely limit the clinical utility of this essential class of pain medications. Here, we mechanistically dissect the protective impact of a natural product, malabaricone C (Mal C), on NSAID-induced gastropathy. Results: Mal C dose dependently diminished erosion of the stomach lining and inflammation in mice treated with NSAIDs with the protective impact translating to improvement in survival. By decreasing oxidative and nitrative stress, Mal C treatment prevented NSAID-induced mitochondrial dysfunction and cell death; nuclear factor κ-light-chain enhancer of activated B cell induction, release of proinflammatory cytokines and neutrophil infiltration; and disruptions in the vascular endothelial growth factor/endostatin balance that contributes to mucosal autohealing. Importantly, Mal C failed to impact the therapeutic anti-inflammatory properties of multiple NSAIDs in a model of acute inflammation. In all assays tested, Mal C proved as or more efficacious than the current first-line therapy for NSAID-dependent GI complications, the proton pump inhibitor omeprazole. Innovation: Given that omeprazole-mediated prophylaxis is, itself, associated with a shift in NSAID-driven GI complications from the upper GI to the lower GI system, there is a clear and present need for novel therapeutics aimed at ameliorating NSAID-induced gastropathy. Mal C provided significant protection against NSAID-induced gastric ulcerations impacting multiple critical signaling cascades contributing to inflammation, cell loss, extracellular matrix degradation, and angiogenic autohealing. Conclusion: Thus, Mal C represents a viable lead compound for the development of novel gastroprotective agents.


Subject(s)
Indomethacin/antagonists & inhibitors , Inflammation/drug therapy , Neovascularization, Pathologic/drug therapy , Protective Agents/pharmacology , Resorcinols/pharmacology , Stomach Ulcer/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Cell Death/drug effects , Dose-Response Relationship, Drug , Indomethacin/administration & dosage , Inflammation/chemically induced , Inflammation/pathology , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Structure , Neovascularization, Pathologic/chemically induced , Neovascularization, Pathologic/pathology , Oxidative Stress/drug effects , Protective Agents/administration & dosage , Protective Agents/metabolism , Resorcinols/administration & dosage , Resorcinols/metabolism , Stomach Ulcer/chemically induced , Stomach Ulcer/pathology , Wound Healing/drug effects
7.
Regul Toxicol Pharmacol ; 109: 104489, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31605713

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common cancers worldwideand haslimited treatment options. In view of this, zafirlukast (ZAF) was administered orally to DEN-induced HCC rats to evaluate its antineoplastic properties. ELISA, qRT-PCR and Western blot were used to determine the molecular mechanism associated with ZAF therapy for HCC. We found that HCC developed as a result of lower expression of caspases 3 and 9, but their levels returned to normal when the expression of eNOS, BAX, BAD, and Cyt C was decreased and when the expression of iNOS, Bcl-xl, and Bcl-2 was increased. Again, ZAF (80 mg/kg dose) treatment normalized the expression of caspase-mediated apoptotic factors, i.e. BAX and Bcl-2 proteins, as established through Western blot analysis. Later, 1H NMR-based serum metabolomics study revealed that levels of perturbed metabolites in DEN-induced rat serum returned to normal after ZAF administration. Altogether, the antineoplastic potential of ZAF was found to be comparable, and to some degree better, than the marketed chemotherapeutic 5-flurouracil, which may be beneficial for anti-HCC treatment from a future drug design perspective.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms, Experimental/drug therapy , Tosyl Compounds/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis Regulatory Proteins/blood , Apoptosis Regulatory Proteins/metabolism , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/metabolism , Cell Survival/drug effects , Diethylnitrosamine/toxicity , Drug Screening Assays, Antitumor , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Humans , Indoles , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Neoplasms, Experimental/blood , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/metabolism , Male , Metabolomics , Mitochondria/drug effects , Mitochondria/metabolism , Phenylcarbamates , Proton Magnetic Resonance Spectroscopy , Rats , Sulfonamides , Tosyl Compounds/therapeutic use
8.
Chem Biol Interact ; 280: 33-44, 2018 Jan 25.
Article in English | MEDLINE | ID: mdl-29223569

ABSTRACT

In this study, we isolated two compounds from the leaves of Salacia oblonga (SA1, mangiferin and SA2, naringenin), and their structures were confirmed by infrared spectroscopy, nuclear magnetic resonance (NMR) spectroscopy, and mass spectrometry. SA1 and SA2 were orally administered to streptozotocin-induced diabetic rats at 50 and 100 mg/kg daily for 15 days. Blood glucose level, serum lipid profile, oxidative stress parameters, histopathology, docking, molecular parameters, and NMR-based metabolic perturbation studies were performed to investigate the pharmacological activities of SA1 and SA2. Results suggested that both compounds reduced blood glucose level, restored body weight, and normalized lipid concentrations in the serum and oxidative stress biomarkers in the liver and pancreas. In addition, the docking study on several diabetes-associated targets revealed that both compounds had a strong binding affinity towards peroxisome proliferator-activated receptor gamma (PPARγ) and glucose transporter type 4 (GLUT4). Further real-time reverse transcription polymerase chain reaction and western blot analyses were performed to confirm the gene and protein expression levels of PPARγ and GLUT4 in the pancreatic tissues. Data obtained from the molecular studies showed that both compounds exhibited antidiabetic effects through dual activation of PPARγ/GLUT4 signaling pathways. Finally, the NMR-based metabolic studies showed that both compounds normalized the diabetogenic metabolites in the serum. Altogether, we concluded that SA1 and SA2 might be potential antidiabetic lead compounds for future drug development.


Subject(s)
Flavanones/pharmacology , Glucose Transporter Type 4/metabolism , Hypoglycemic Agents/pharmacology , PPAR gamma/metabolism , Xanthones/pharmacology , Animals , Blood Glucose/analysis , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/drug therapy , Flavanones/isolation & purification , Flavanones/therapeutic use , Glucose Transporter Type 4/agonists , Glucose Transporter Type 4/genetics , Glycogen/metabolism , Hypoglycemic Agents/isolation & purification , Hypoglycemic Agents/therapeutic use , Insulin/blood , Lipids/blood , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Molecular Docking Simulation , Oxidative Stress/drug effects , PPAR gamma/agonists , PPAR gamma/genetics , Pancreas/drug effects , Pancreas/metabolism , Pancreas/pathology , Protein Structure, Tertiary , Rats , Salacia/chemistry , Salacia/metabolism , Signal Transduction/drug effects , Streptozocin/toxicity , Xanthones/isolation & purification , Xanthones/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...