Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Life Sci ; 337: 122349, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38128755

ABSTRACT

Liver fibrosis involves several signalling pathways working in concert regulating the deposition of extracellular matrix. In this study, we evaluated the effect of quercetin and simvastatin alone and their combination on the treatment of experimentally induced hepatic fibrosis in rats. To decipher the potential mechanisms involved, liver fibrosis was induced in rats by administration of 40 % carbon tetrachloride (CCl4) (1 µl/g rat, i.p., twice weekly) for 6 weeks. Quercetin (50 mg/kg, orally), simvastatin (40 mg/kg, orally) either individually or combined were administered for another 4 weeks. The three treatment groups ameliorated hepatic dysfunction and altered parameters of sphingolipid and pyroptosis pathways. Yet, the combined group showed a more pronounced effect. Treatments lowered serum levels of GOT, GPT, ALP and elevated albumin and total protein levels. Histopathological and electron microscope examination of liver tissue revealed diminished fibrosis and inflammation. Protein expression levels of α-SMA, IL-1ß, PPAR-γ, TGF-ß1, caspase-1 and caspase-3 expression in liver tissues were reduced. Additionally, hepatic mRNA levels of SphK1 and NLRP3 decreased after treatment. Furthermore, the three groups lowered MDA levels and elevated total antioxidant capacity, GSH and Nrf2 expression levels. Treatments downregulated sphingolipid pathway and NLRP3-mediated pyroptosis and stimulated an anti-apoptotic, anti-proliferative and antioxidant activity. This suggests that targeting the SphK1/NLRP3 pathway could be a prospective therapeutic strategy against liver fibrosis.


Subject(s)
NLR Family, Pyrin Domain-Containing 3 Protein , Quercetin , Rats , Animals , Quercetin/therapeutic use , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Simvastatin/pharmacology , Simvastatin/therapeutic use , Liver Cirrhosis/pathology , Liver/metabolism , Antioxidants/metabolism , Carbon Tetrachloride/pharmacology , Sphingolipids/metabolism
3.
Int Immunopharmacol ; 118: 110074, 2023 May.
Article in English | MEDLINE | ID: mdl-36989898

ABSTRACT

AIM: New therapeutic strategies are required to enhance the anticancer efficacy of chemotherapeutic drugs and to reduce their cytotoxicity. The purpose of this study was to assess the anti-tumor, antimetastatic and anti-apoptotic activities of sinapic acid (SA) and 3,3'-diindolylmethane (DIM) in solid Ehrlich carcinoma (SEC) induced in mice and combining SA or DIM compounds with cyclophosphamide (CYP). METHODS: For induction of solid tumor, the right hind limbs of mice were inoculated subcutaneously with Ehrlich carcinoma cells. After 5 days of tumor inoculation, mice were treated with SA (56 mg/kg), DIM (40 mg/kg), CYP (10 mg/kg), and their combinations (SA/CYP) and (SA/DIM) for 21 days. The mRNA levels of Elabela, Serpina3, caspase-3, MMP-2 and MMP-9 were assessed by qPCR. Tumor and liver tissues were stained with hematoxylin and eosin for histological examination. Serum was investigated for ALT and AST activities. MAIN FINDINGS: Treatment of SEC mice with SA and DIM significantly reduced solid tumor weight by 45.6% and 33.2%, respectively. They also reduced tumor size and increased life span of SEC mice. SA and DIM diminished area of metastatic nodules of tumor cells in the liver by 54.1% and 47.4%, respectively. They also reduced serum aminotransferases activities. Both SA and DIM were found to upregulate caspase 3 and downregulate MMP-2 and MMP-9. Furthermore, SA and DIM reduced gene expression of Elabela by (44.8% and 35.1%) and Serpina3 by (30.7% and 23.5%), respectively. SA and DIM were also shown to potentiate the anti-tumor activity CYP. SA and DIM showed promising antitumor effects and enhanced CYP antitumor activity mostly through upregulation of apoptotic caspase 3 and suppressing metastatic enzymes MMP-2 and MMP-9. Additionally, SA and DIM exhibited a hepatoprotective effect. Our results suggest that these natural compounds may be used to improve the efficacy and reduce the adverse effects of chemotherapeutic drugs in the treatment of solid malignancies.


Subject(s)
Carcinoma , Matrix Metalloproteinase 2 , Animals , Mice , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Caspase 3 , Cell Proliferation , Cell Line, Tumor , Apoptosis , Cyclophosphamide/therapeutic use , Cyclophosphamide/pharmacology
4.
Life Sci ; 261: 118458, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32961231

ABSTRACT

AIM: Niclosamide (NIC) is an anthelmintic agent repurposed as a potent anticancer agent. However, its use is hindered by its poor solubility. We investigated the underlying mechanisms of NIC anticancer activity employing a novel oral NIC pluronic-based nanoformulation and tested its effect in thioacetamide-induced hepatocellular carcinoma (HCC) in rats. We evaluated its antitumor effect through regulating Wnt/ß-catenin and Notch signaling pathways and apoptosis. MAIN METHODS: Niclosamide-loaded pluronic nanoparticles (NIC-NPs) were optimally developed and characterized with sustained release properties up to 7 days. Sixteen weeks after HCC induction, NIC (70 mg/kg) and an equivalent dose of NIC-NPs were administered orally for 3 consecutive weeks. Hepatocyte integrity was assessed by measuring serum levels of aminotransferases, ALP, GGT, bilirubin, albumin and total protein. HCC development was detected by measuring AFP expression. Necroinflammation and fibrosis were scored by histopathological examination. Wnt/ß-catenin and Notch signaling were evaluated by measuring hepatic mRNA levels of Wnt3A, Lrp5 and Lrp6 Co-receptors, Dvl-2, Notch1 and Hes1 and ß-catenin protein levels. Apoptosis was assessed by measuring mRNA and protein levels of cyclin D1 and caspase-3. KEY FINDING: The novel NIC-NPs restored liver integrity, reduced AFP levels and showed improved anticancer and proapoptotic activities compared to drug alone. The inhibitory effect of NIC on Wnt/ß-catenin and Notch signaling pathways was potentiated by the NIC-NPs formulation. SIGNIFICANCE: We conclude that NIC acts by inhibiting Wnt/ß-catenin and Notch signaling and inducing apoptosis in HCC. Developing pluronic-based nanoformulations may be a promising approach to improve NIC solubility and offer the possibility of controlled release.


Subject(s)
Anthelmintics/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Niclosamide/therapeutic use , Receptors, Notch/metabolism , Wnt Signaling Pathway/drug effects , Animals , Anthelmintics/administration & dosage , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Drug Carriers/chemistry , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Micelles , Nanoparticles/chemistry , Niclosamide/administration & dosage , Rats, Sprague-Dawley , Signal Transduction/drug effects
5.
Naunyn Schmiedebergs Arch Pharmacol ; 393(3): 303-313, 2020 03.
Article in English | MEDLINE | ID: mdl-31612257

ABSTRACT

Renal fibrosis is a progressive process resulting from a sustained injury that may ultimately cause renal failure. Cisplatin is an antitumor drug that induces renal injury and nephrotoxicity and is widely employed as a model for acute and chronic renal injury. Several signaling pathways are implicated in fibrogenic cell activation among which is Hedgehog (Hh) signaling. We here investigated the effects of arsenic trioxide (Ars) and curcumin in ameliorating cisplatin-induced kidney fibrosis via regulating Hh signaling. Cisplatin (4.5 mg/kg) was administered in Sprague-Dawley rats for two consecutive days and renal fibrosis was induced after 21 days. Once renal fibrosis was confirmed, Ars (3.5 mg/kg/day, orally) and curcumin (200 mg/kg/day, orally) were administered daily for another 21 days. Ars and curcumin corrected kidney function markers as creatinine clearance and urea nitrogen. Both agents ameliorated fibrosis as shown by lowered TGF-ß1 mRNA levels, α-SMA protein levels, and hydroxylproline content. Cisplatin-activated Hh signaling which was blocked by both Ars and curcumin as demonstrated by decreased mRNA levels of Shh, Smo, and Ptch and suppressed renal Gli1 and Gli2 protein levels. Our results indicate new therapeutic roles for Ars and curcumin and suggest that blocking Hh signaling may be a promising approach for alleviating renal fibrosis. Symbols indicate α-SMA, alpha-smooth muscle actin; TGF-ß, transforming growth factor-beta; Ptch, patched; Smo, smoothened; Shh, sonic hedgehog; Ihh, Indian hedgehog; Dhh, desert hedgehog; and SUFU, suppressor of fused.


Subject(s)
Arsenic Trioxide/administration & dosage , Cisplatin/toxicity , Curcumin/administration & dosage , Hedgehog Proteins/antagonists & inhibitors , Kidney Diseases/chemically induced , Kidney Diseases/prevention & control , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/toxicity , Drug Delivery Systems/methods , Drug Therapy, Combination , Fibrosis , Hedgehog Proteins/metabolism , Kidney Diseases/metabolism , Male , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology
6.
Life Sci ; 236: 116933, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31614146

ABSTRACT

AIMS: Hepatocellular carcinoma (HCC) pathogenesis involves the interplay of multiple signalling pathways. Notch and Hedgehog (Hh) are two major developmental pathways that act in concert to regulate adult cell repair. CK2α -serine-threonine kinase-down-regulation enhanced apoptotic activity and was proven beneficial for HCC patients. Quercetin is a bioactive flavonoid and has been shown to protect against HCC through its antioxidant activity. This study was carried out to elucidate the antineoplastic effect of quercetin through regulating both Notch and Hh pathways, apoptosis, cell proliferation and CK2α activity. MAIN METHODS: Hepatocellular carcinoma was induced in male Sprague Dawley rats by thioacetamide. Quercetin was administered in both protective and curative doses. Parameters of liver function and oxidative stress were assessed. CK2α, Notch and Hh pathways were evaluated using RT-PCR and ELISA. Apoptosis was investigated by detecting caspase-3, caspase-8 and p53. Proliferative and cell cycle markers as cyclin D1 and Ki-67 were detected immunohistochemically. KEY FINDINGS: Quercetin inhibited CK2α and downregulated mRNA and protein expression of Notch1 and Gli2. Quercetin also suppressed caspase-3 expression but not caspase-8. Quercetin elevated p53 expression whereas proliferative and cell cycle markers cyclin D1 and Ki-67 were downregulated. Markers of hepatic cellular integrity such as AST, ALT, ALP, GGT, albumin and bilirubin were significantly ameliorated. This was confirmed by histological examination. Quercetin also alleviated oxidative stress as shown by SOD, GSH, MDA and NO levels. SIGNIFICANCE: We can conclude that in addition to its antioxidant power, quercetin blocked Notch, Hedgehog, regulated the apoptotic and proliferative pathways and inhibited CK2α in HCC.


Subject(s)
Antioxidants/pharmacology , Carcinoma, Hepatocellular/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Liver Neoplasms/metabolism , Quercetin/pharmacology , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Casein Kinase II/antagonists & inhibitors , Casein Kinase II/metabolism , Cell Proliferation/drug effects , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Male , Oxidative Stress , Rats , Rats, Sprague-Dawley , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/metabolism
7.
Toxicol Appl Pharmacol ; 380: 114697, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31394158

ABSTRACT

Liver cholestasis is a complex disease of broad etiologies. Hedgehog (Hh) signaling has been shown to play a pivotal role in modulating liver repair post cholestatic liver injury. We here investigated the role of vitamin D in experimental liver cholestasis induced by bile-duct ligation (BDL) in rats. Male Sprague Dawley rats underwent BDL surgery and two weeks post-surgery; vitamin D was administered daily for two weeks. Serum markers of hepatocellular integrity were measured and fibrosis was detected by measuring α-SMA and hepatic TGF-ß1 as well as hepatic collagen content. Hh signaling was evaluated by measuring Smo and Gli1 levels. Histopathological examination of hepatic tissue was performed. Vitamin D alleviated obstructive cholestatic damage as illustrated by total bilirubin as well as gamma glutamyl transferase (γ-GT) serum levels. It also alleviated hepatocellular damage as suggested by reducing elevated serum aminotransferases induced by BDL. Antifibrotic activity of vitamin D was confirmed by decrease in mRNA and protein expression of α-SMA, as well as collagen content in hepatic tissue. Furthermore, vitamin D suppressed BDL-induced elevated hepatic TGF-ß1 mRNA and protein levels. BDL activated Hh signaling and upregulated its upstream component smoothened (Smo) and downstream target gene Gli1. Treatment with vitamin D reduced Smo mRNA levels and suppressed hepatic Gli1 mRNA and protein levels. Molecular docking of vitamin D to Smo revealed that vitamin D binds similarly to its endogenous cholesterol ligand and blocks its activation. These results demonstrate that vitamin D mitigated cholestatic liver injury through inhibiting Hh signaling.


Subject(s)
Cholecalciferol/therapeutic use , Cholestasis/drug therapy , Liver Cirrhosis/drug therapy , Vitamins/therapeutic use , Animals , Bile Ducts , Cholecalciferol/pharmacology , Cholestasis/genetics , Cholestasis/metabolism , Cholestasis/pathology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Hedgehog Proteins/metabolism , Ligation , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Molecular Docking Simulation , Rats, Sprague-Dawley , Signal Transduction , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Vitamins/pharmacology , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism
8.
Life Sci ; 217: 251-260, 2019 Jan 15.
Article in English | MEDLINE | ID: mdl-30550888

ABSTRACT

Drug-induced liver toxicity is the most frequent cause of acute liver failure worldwide. Hepatotoxicity caused by acetaminophen (ACT) overdose is mediated by its metabolic product promoting oxidative stress and activation of inflammatory mediators. Nuclear factor erythroid-related factor-2 (Nrf-2) induces the release of cytoprotective enzymes in response to electrophilic or oxidative stress and is considered a promising therapeutic target. Dimethyl fumarate (DMF) is a potent activator of (Nrf-2), its anti-inflammatory and antioxidant properties of DMF have been highlighted recently. We designed this study to explore the effect of DMF (100 mg/kg, orally) administered once and twice on hepatotoxicity induced by acetaminophen (ACT, 500 mg/kg, i.p.) in mice. DMF administration enhanced ACT-induced parameters in liver function, inhibited apoptosis and ameliorated the antioxidant machinery and inflammatory markers in a Nrf-2-dependent fashion. DMF elevated Nrf-2 and HO-1 levels and ameliorated liver injury as indicated by lowered levels of serum aminotransferases, ALP, GGT and bilirubin levels. Hepatic (Bcl-2) was elevated whereas hepatic caspase-3, NFκ-B, TNF-α and MPO were reduced. Hepatic levels of GSH, SOD, MDA and NO were altered promoting the antioxidant machinery. Histological examination of liver has further supported these results. These findings suggest that DMF can be employed in the treatment ACT-induced liver injury acting primarily through targeting Nrf-2/HO-1 pathway.


Subject(s)
Acetaminophen/adverse effects , Analgesics, Non-Narcotic/adverse effects , Antioxidants/therapeutic use , Chemical and Drug Induced Liver Injury/drug therapy , Dimethyl Fumarate/therapeutic use , Heme Oxygenase-1/metabolism , Liver/drug effects , Membrane Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Animals , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Liver/metabolism , Liver/pathology , Mice, Inbred BALB C , Oxidative Stress/drug effects , Signal Transduction/drug effects
9.
Life Sci ; 211: 245-251, 2018 Oct 15.
Article in English | MEDLINE | ID: mdl-30243650

ABSTRACT

Liver fibrosis is a global health issue that causes morbidity and mortality with no currently available treatment. It has been shown that low dose paclitaxel (PTX) can stabilize microtubules and inhibit the profibrotic transforming growth factor-beta 1 (TGF-ß1) signaling pathway. In this study the effect of treatment with low dose PTX was examined using a model of cholestatic liver fibrosis. Bile-duct ligation (BDL) was induced in rats for 2 weeks then PTX (0.3 mg/kg/ip) was administered three times a week for 2 weeks. Administration of PTX ameliorated BDL-induced elevation in biomarkers of hepatocellular damage (alanine transaminase; ALT and aspartate transaminase; AST) and obstructive cholestatic injury (total bilirubin and gamma glutamyl transferase; γ-GT). PTX was able to correct the increase in liver weight to body weight ratio and the bile duct proliferation induced by BDL. Additionally, PTX treatment corrected the BDL-induced fibrosis of portal tracts, elevation of hydroxyproline content and increased alpha smooth muscle actin (α-SMA) mRNA and protein expression. This antifibrotic effect of PTX was further examined through its inhibitory effect on TGF-ß1 mRNA and protein expression in addition to c-Myc mRNA expression. Furthermore, PTX rectified the BDL-induced decrease in interleukin-10 (IL-10) mRNA and protein expression. In conclusion, this study suggests that PTX at low dose has the potential to treat BDL-induced liver fibrosis in rats possibly through suppression of TGF-ß1 and c-Myc and activation of IL-10 pathways.


Subject(s)
Bile Ducts/surgery , Gene Expression Regulation/drug effects , Interleukin-10/metabolism , Liver Cirrhosis/prevention & control , Paclitaxel/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Interleukin-10/genetics , Ligation , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Proto-Oncogene Proteins c-myc/genetics , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta1/genetics
10.
Sci Rep ; 8(1): 10836, 2018 Jul 18.
Article in English | MEDLINE | ID: mdl-30022069

ABSTRACT

Targeted cancer immunotherapy offers increased efficacy concomitantly with reduced side effects. One antibody with promising clinical potential is 14F7, which specifically recognises the NeuGc GM3 ganglioside. This antigen is found in the plasma membrane of a range of tumours, but is essentially absent from healthy human cells. 14F7 can discriminate NeuGc GM3 from the very similar NeuAc GM3, a common component of cell membranes. The molecular basis for this unique specificity is poorly understood. Here we designed and expressed 14F7-derived single-chain Fvs (scFvs), which retained the specificity of the parent antibody. Detailed expression and purification protocols are described as well as the synthesis of the NeuGc GM3 trisaccharide. The most successful scFv construct, which comprises an alternative variable light chain (VLA), allowed structure determination to 2.2 Å resolution. The structure gives insights into the conformation of the important CDR H3 loop and the suspected antigen binding site. Furthermore, the presence of VLA instead of the original VL elucidates how this subdomain indirectly stabilises the CDR H3 loop. The current work may serve as a guideline for the efficient production of scFvs for structure determination.


Subject(s)
Antibodies, Monoclonal/chemistry , G(M3) Ganglioside/chemistry , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Light Chains/chemistry , Immunoglobulin Variable Region/chemistry , Neoplasms/drug therapy , Single-Chain Antibodies/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Crystallography, X-Ray , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/metabolism , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/metabolism , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/metabolism , Protein Conformation , Single-Chain Antibodies/genetics , Single-Chain Antibodies/metabolism
11.
Article in English | MEDLINE | ID: mdl-28716464

ABSTRACT

Toll-like receptor 4 (TLR-4) plays important roles in innate immunity. Changes in the reduction-oxidation balance of tissues can lead to a pro-inflammatory state and insulin resistance. An action thought to be mediated by TLRs. Omega-3 fatty acids and Peroxisome Proliferator Activated Receptor gamma (PPAR-γ) agonists as pioglitazone are used for decreasing inflammation. The aim of this study is to investigate the anti-diabetic effects of combining omega -3 fatty acid with pioglitazone on type 2 diabetes, and the modifying effects on TLR-4. Type 2 diabetes was induced in male Sprague-Dawley rats by combination of high fat diet and low dose streptozotocin (35mg/kg). Diabetic rats were treated with omega-3 fatty acids (10% W/W diet), pioglitazone (20mg/kg), and their combination for 4 weeks. Omega-3 fatty acids and the combination treatment significantly decreased TLR-4 activation. Omega-3 fatty acids, pioglitazone, and their combination significantly decreased TLR-4 mRNA expression, hepatic malondialdehyde, total cholesterol and triglycerides levels, compared to diabetic group. Pioglitazone and the combination significantly decreased blood glucose levels and improved insulin resistance. In conclusion, combining omega-3 fatty acids with pioglitazone showed potential effects in lowering blood glucose levels and improving lipid profile and insulin resistance. Such effects are mediated through modulation of TLR-4.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diet, High-Fat/adverse effects , Fatty Acids, Omega-3/administration & dosage , Pioglitazone/administration & dosage , Toll-Like Receptor 4/genetics , Animals , Blood Glucose/drug effects , Cholesterol/metabolism , Combined Modality Therapy , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/genetics , Down-Regulation , Fatty Acids, Omega-3/pharmacology , Insulin Resistance , Male , Malondialdehyde/metabolism , Pioglitazone/pharmacology , Rats , Rats, Sprague-Dawley , Streptozocin
12.
Tumour Biol ; 39(6): 1010428317707376, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28651490

ABSTRACT

Angiogenesis plays important roles in progression of hepatocellular carcinoma. The antiangiogenic mechanisms of vitexicarpine are not fully defined. Therefore, we conducted the following study to evaluate the antiangiogenic mechanism and antitumor activity of vitexicarpine in vivo model of hepatocellular carcinoma through modulation of vascular endothelial growth factor signaling pathway. Hepatocellular carcinoma was induced in Sprague Dawley rats by thioacetamide. Hepatocellular carcinoma was assessed by measuring serum alpha-fetoprotein and investigating liver sections stained with hematoxylin/eosin. Hepatocellular carcinoma rats were injected with vitexicarpine (150 mg/kg) for 2 weeks. Hepatic vascular endothelial growth factor was measured by enzyme-linked immunosorbent assay. Protein and expression of hepatic phospho-Ser473-AKT (p-AKT) and phospho-Tyr419-Src (p-Src) were determined. The apoptotic pathway was evaluated by assessment of protein expression of caspase-3. Vitexicarpine increased rats' survival time and decreased serum alpha-fetoprotein as well as it ameliorated fibrosis and massive hepatic tissue breakdown. It attenuated hepatocellular carcinoma-induced protein and gene expression of vascular endothelial growth factor, p-AKT, p-Src, and caspase-3. In conclusion, this study suggests that vitexicarpine possesses both antiangiogenic and antitumor activities through inhibition of vascular endothelial growth factor, p-AKT/AKT, and p-Src with subsequent inhibition of apoptotic pathway.


Subject(s)
Apigenin/administration & dosage , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Vascular Endothelial Growth Factor A/genetics , Angiogenesis Inhibitors/administration & dosage , Animals , Apoptosis/drug effects , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neovascularization, Pathologic/chemically induced , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Rats , Signal Transduction/drug effects , Thioacetamide/toxicity , Xenograft Model Antitumor Assays , alpha-Fetoproteins/metabolism
13.
PLoS Comput Biol ; 10(7): e1003745, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25079355

ABSTRACT

Hydrophobins represent an important group of proteins from both a biological and nanotechnological standpoint. They are the means through which filamentous fungi affect their environment to promote growth, and their properties at interfaces have resulted in numerous applications. In our study we have combined protein docking, molecular dynamics simulation, and electron cryo-microscopy to gain atomistic level insight into the surface structure of films composed of two class II hydrophobins: HFBI and HFBII produced by Trichoderma reesei. Together our results suggest a unit cell composed of six proteins; however, our computational results suggest P6 symmetry, while our experimental results show P3 symmetry with a unit cell size of 56 Å. Our computational results indicate the possibility of an alternate ordering with a three protein unit cell with P3 symmetry and a smaller unit cell size, and we have used a Monte Carlo simulation of a spin model representing the hydrophobin film to show how this alternate metastable structure may play a role in increasing the rate of surface coverage by hydrophobin films, possibly indicating a mechanism of more general significance to both biology and nanotechnology.


Subject(s)
Fungal Proteins/chemistry , Fungal Proteins/metabolism , Molecular Dynamics Simulation , Microscopy, Electron , Protein Binding , Protein Conformation , Static Electricity , Surface Properties , Trichoderma/chemistry
14.
Biochem Soc Trans ; 39(5): 1327-33, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21936810

ABSTRACT

In order to achieve greater selectivity in drug discovery, researchers in both academia and industry are targeting cell regulatory systems. This often involves targeting the protein-protein interactions of regulatory multiprotein assemblies. Protein-protein interfaces are widely recognized to be challenging targets as they tend to be large and relatively flat, and therefore usually do not have the concave binding sites that characterize the so-called 'druggable genome'. One such prototypic multiprotein target is the Notch transcription complex, where an extensive network of protein-protein interactions stabilize the ternary complex comprising the ankyrin domain, CSL (CBF1/suppressor of Hairless/Lag-1) and MAML (Mastermind-like). Enhanced Notch activity is implicated in the development of T-ALL (T-cell acute lymphoblastic leukaemia) and selective inhibitors of Notch would be useful cancer medicines. In the present paper, we describe a fragment-based approach to explore the druggability of the ankyrin domain. Using biophysical methods and X-ray crystal structure analyses, we demonstrate that molecules can bind to the surface of the ankyrin domain at the interface region with CSL and MAML. We show that they probably represent starting points for designing larger compounds that can inhibit important protein-protein interactions that stabilize the Notch complex. Given the relatively featureless topography of the ankyrin domain, this unexpected development should encourage others to explore the druggability of such challenging multiprotein systems using fragment-based approaches.


Subject(s)
Ankyrins/chemistry , Ankyrins/metabolism , Drug Design , Peptide Fragments/chemistry , Protein Interaction Domains and Motifs , Receptor, Notch1/chemistry , Receptor, Notch1/metabolism , Amino Acid Sequence , Ankyrins/genetics , Binding Sites , Crystallography, X-Ray , Dimerization , Models, Molecular , Molecular Sequence Data , Molecular Structure , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Binding , Protein Interaction Mapping , Protein Structure, Tertiary , Receptor, Notch1/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...