Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Biochem Biophys Res Commun ; 503(3): 1968-1972, 2018 09 10.
Article in English | MEDLINE | ID: mdl-30078676

ABSTRACT

Multiple sclerosis (MS) is a disabling inflammatory and demyelinating disorder of the central nervous system. Tetramethylpyrazine (TMP) has been demonstrated to ameliorate cerebral ischemic injury and spinal cord injury by inhibiting inflammatory cell activation and pro-inflammatory cytokine production. However, the effects of TMP on MS have not been studied. In this study, we evaluated the effects of TMP on the inflammatory response in experimental autoimmune encephalomyelitis (EAE), which is an animal model of MS. TMP (30 mg/kg) treatment significantly reduced the expression levels of NLR Family, Pyrin Domain-Containing 3 Protein inflammasome and caspase-1and decreased inflammatory infiltration and glial activation. Moreover, TMP (30 mg/kg) suppressed the expression of pro-inflammatory cytokines (interleukin-18 [IL-18] and IL-17) and promoted the expression of an anti-inflammatory cytokine (IL-10). The reduced inflammatory response resulted in improvement in clinical scores and decreased demyelination in EAE mice. Therefore, our results demonstrate that TMP (30 mg/kg) improved functional recovery in part by reducing inflammation in EAE mice. TMP may be a potential therapeutic agent for MS therapy.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Inflammation/drug therapy , Pyrazines/therapeutic use , Animals , Cytokines/analysis , Cytokines/immunology , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/pathology , Enzyme-Linked Immunosorbent Assay , Inflammation/pathology , Mice , Mice, Inbred C57BL
2.
Toxicol Appl Pharmacol ; 272(3): 895-904, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23933165

ABSTRACT

Inflammation response and oxidative stress play important roles in acute lung injury (ALI). Activation of the cAMP/protein kinase A (PKA) signaling pathway may attenuate ALI by suppressing immune responses and inhibiting the generation of reactive oxygen species (ROS). Hydroxysafflor yellow A (HSYA) is a natural flavonoid compound that reduces oxidative stress and inflammatory cytokine-mediated damage. In this study, we examined whether HSYA could protect the lungs from oleic acid (OA)-induced injury, which was used to mimic ALI, and determined the role of the cAMP/PKA signaling pathway in this process. Arterial oxygen tension (PaO2), carbon dioxide tension, pH, and the PaO2/fraction of inspired oxygen ratio in the blood were detected using a blood gas analyzer. We measured wet/dry lung weight ratio and evaluated tissue morphology. The protein and inflammatory cytokine levels in the bronchoalveolar lavage fluid and serum were determined using enzyme-linked immunoassay. The activities of superoxide dismutase, glutathione peroxidase, PKA, and nicotinamide adenine dinucleotide phosphate oxidase, and the concentrations of cAMP and malondialdehyde in the lung tissue were detected using assay kits. Bcl-2, Bax, caspase 3, and p22(phox) levels in the lung tissue were analyzed using Western blotting. OA increased the inflammatory cytokine and ROS levels and caused lung dysfunction by decreasing cAMP synthesis, inhibiting PKA activity, stimulating caspase 3, and reducing the Bcl-2/Bax ratio. H-89 increased these effects. HSYA significantly increased the activities of antioxidant enzymes, inhibited the inflammatory response via cAMP/PKA pathway activation, and attenuated OA-induced lung injury. Our results show that the cAMP/PKA signaling pathway is required for the protective effect of HSYA against ALI.


Subject(s)
Acute Lung Injury/enzymology , Acute Lung Injury/prevention & control , Chalcone/analogs & derivatives , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/physiology , Oleic Acid/toxicity , Quinones/therapeutic use , Acute Lung Injury/chemically induced , Animals , Chalcone/pharmacology , Chalcone/therapeutic use , Enzyme Activation/drug effects , Enzyme Activation/physiology , Male , Protein Kinase Inhibitors/toxicity , Quinones/pharmacology , Random Allocation , Rats , Rats, Sprague-Dawley
3.
Oncol Lett ; 25(6): 260, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37205919

ABSTRACT

1,4,5,6,7,8-Hexahydropyrido[4,3-d]pyrimidine (PPM) promotes apoptosis of HepG2 cells and serves a role in tumor suppression. However, the role of microRNA (miRNA) regulation in initiating apoptosis remains unclear. Therefore, the present study performed reverse transcription-quantitative PCR to investigate the association between PPM and miRNA, which demonstrated that PPM upregulated the expression of miR-26b-5p. Wound healing and Transwell assays showed that PPM inhibited the migration and invasion of HepG2 cells, and EdU staining experiments showed that PPM inhibited the proliferation of HepG2 cells. Transfection with miR-26b-5p inhibitor reversed the effects of PPM on HepG2 cells. Flow cytometry results showed that PPM promoted apoptosis of HepG2 cells by upregulating miRNA (miR)-26b-5p, and Western blotting results showed that PPM promoted the expression of apoptosis-associated protein Bax and inhibited the expression of Bcl-2 by upregulating miR-26b-5p. Using a proteomic approach combined with bioinformatics analysis, CDK8 was identified as a potential target of miR-26b-5p and was downregulated by miR-26b-5p overexpression. However, PPM induced HepG2 cell cycle arrest without the involvement of miR-26b-5p. Western blotting results showed that PPM upregulation of miR-26b-5p suppresses NF-κB/p65 signaling pathway in HepG2 cells by targeting of CDK8. The present results suggested that miR-26b-5p may function as a target gene of PPM and may serve a role in hepatocellular carcinoma treatment.

4.
Biomed Pharmacother ; 136: 111227, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33485070

ABSTRACT

Hydroxysafflor yellow A (HSYA) extracted from the herb Cathartics tinctorius L. negatively regulates liver cancer growth. However, the exact mechanism of HSYA action in liver cancer remains largely unknown. In this study, HSYA inhibited liver cancer cell growth in vivo and in vitro, evidenced by cell proliferation inhibition detected by CCK8, numerous apoptotic cells shown by flow cytometry assay, and expression of apoptosis-related proteins determined by western blot. Importantly, our data revealed that HSYA triggered autophagic response and autophagosome accumulation considering the increased levels of LC3II-conversion examined by western blot, LC3 puncta visualized by immunofluorescence, and expression of autophagy-related genes shown by quantitative real-time PCR. Furthermore, HSYA blocked the late-phase of autophagic flux via impairing the lysosomal acidification and downregulating LAMP1 expression, thereby likely inducing apoptosis. In addition, HSYA inhibited PI3K/AKT/mTOR signaling pathway. Taken together, as HSYA might inhibit cell proliferation and promote apoptosis via blocking autophagic flux in liver cancer, it may be considered a promising candidate for liver cancer therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Chalcone/analogs & derivatives , Liver Neoplasms/drug therapy , Quinones/pharmacology , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagosomes/pathology , Cell Proliferation/drug effects , Chalcone/pharmacology , Hep G2 Cells , Humans , Hydrogen-Ion Concentration , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/pathology , Microtubule-Associated Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
5.
Eur J Med Chem ; 225: 113796, 2021 Dec 05.
Article in English | MEDLINE | ID: mdl-34450496

ABSTRACT

In order to obtain new anti-hepatoma drugs with low toxicity, some 1,4,5,6,7,8-hexahydropyrido[4,3-d]pyrimidines (PPMs, 4a-t) were synthesized in this study. Many of them showed significant anti-hepatoma effects against HCC cells and low toxicity toward HHL-5 cells. Combined with their anti-hepatoma activity and toxicity, 4-CF3-substituted 4k was selected as an effective lead compound. Preliminary mechanistic studies revealed that 4k could up-regulate the expression levels of Bax and caspase-3 proteins, down-regulate the expression levels of Bcl-2 protein, promote significant apoptosis of HepG2, and block cells in G2-M phase to prevent cells from completing mitosis. Also, 4k could significantly inhibit the activation of PI3K/AKT/NF-κB pathway by blocking the phosphorylation of PI3K, AKT, NF-κB/p65 and IFN-γ-induced nuclear transport. Docking analysis showed that 4k could reasonably bind to the active sites of Bcl-2, NF-κB/p65, PI3K and AKT. This result suggested that 4k could be used as a new type of NF-κB inhibitor, which provides a scientific basis for further research into the treatment of hepatoma.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Drug Discovery , Liver Neoplasms/drug therapy , NF-kappa B/antagonists & inhibitors , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Cycle/drug effects , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Molecular Structure , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Structure-Activity Relationship
6.
Biomed Pharmacother ; 109: 806-814, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30551534

ABSTRACT

The antitumor effect of hydroxysafflor yellow A (HSYA), an active ingredient of the herb Carthamus tinctorius L. (Asteraceae) (safflower), was investigated in the current work. Researches of HSYA on vasculogenesis inhibition, along with the related molecular mechanisms, including the expression of MMP-2, MMP-9, and p38MAPK (COX-2, ATF-2, p-p38MAPK, and p38MAPK) signaling pathway in H22 tumor-bearing mice or HepG2 cells were performed. The animal experiments proved the level of MMP-2 and MMP-9 in H22-transplanted tumor tissue in mice markedly decreased by HSYA, and results both in vivo and in vitro confirmed that COX-2 expression was reduced significantly via p38MAPK|ATF-2 signaling pathway. According to the outcomes, HSYA suppressed p38MAPK phosphorylation in a concentration-dependent manner, while exerting no effect on the total p38MAPK protein expression. It was also showed that suppression of p38 activation by SB203580 decreased the HepG2 cell viability, proliferation, and migration, wherein HSYA exhibited a similar effect. Furthermore, Western blot analysis on caspase-3 and cleaved-caspase-3 revealed that HSYA could induce apoptosis of HepG2 cells. These findings provided experimental evidences that HSYA might be a promising anticancer agent for HCC.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Chalcone/analogs & derivatives , Liver Neoplasms/enzymology , Neovascularization, Pathologic/enzymology , Quinones/therapeutic use , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Chalcone/pharmacology , Chalcone/therapeutic use , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Hep G2 Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Male , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Phosphorylation/drug effects , Phosphorylation/physiology , Pigments, Biological/pharmacology , Pigments, Biological/therapeutic use , Quinones/pharmacology
7.
Acta Crystallogr C Struct Chem ; 74(Pt 10): 1171-1179, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30284984

ABSTRACT

3,5-Bis(arylidene)-4-piperidone (BAP) derivatives display good antitumour and anti-inflammatory activities because of their double α,ß-unsaturated ketone structural characteristics. If N-benzenesulfonyl substituents are introduced into BAPs, the configuration of the BAPs would change significantly and their anti-inflammatory activities should improve. Four N-benzenesulfonyl BAPs, namely (3E,5E)-1-(4-methylbenzenesulfonyl)-3,5-bis[4-(trifluoromethyl)benzylidene]piperidin-4-one dichloromethane monosolvate, C28H21F6NO3S·CH2Cl2, (4), (3E,5E)-1-(4-fluorobenzenesulfonyl)-3,5-bis[4-(trifluoromethyl)benzylidene]piperidin-4-one, C27H18F7NO3S, (5), (3E,5E)-1-(4-nitrobenzenesulfonyl)-3,5-bis[4-(trifluoromethyl)benzylidene]piperidin-4-one, C27H18F6N2O5S, (6), and (3E,5E)-1-(4-cyanobenzenesulfonyl)-3,5-bis[4-(trifluoromethyl)benzylidene]piperidin-4-one dichloromethane monosolvate, C28H18F6N2O3S·CH2Cl2, (7), were prepared by Claisen-Schmidt condensation and N-sulfonylation. They were characterized by NMR, FT-IR and HRMS (high resolution mass spectrometry). Single-crystal structure analysis reveals that the two 4-(trifluoromethyl)phenyl rings on both sides of the piperidone ring in (4)-(7) adopt an E stereochemistry of the olefinic double bonds. Molecules of both (4) and (6) are connected by hydrogen bonds into one-dimensional chains. In (5) and (7), pairs of adjacent molecules embrace through intermolecular hydrogen bonds to form a bimolecular combination, which are further extended into a two-dimensional sheet. The anti-inflammatory activity data reveal that (4)-(7) significantly inhibit LPS-induced interleukin (IL-6) and tumour necrosis factor (TNF-α) secretion. Most importantly, (6) and (7), with strong electron-withdrawing substituents, display more potential inhibitory effects than (4) and (5).


Subject(s)
Anti-Inflammatory Agents/chemistry , Benzene Derivatives/chemistry , Piperidones/chemistry , Animals , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/pharmacology , Benzene Derivatives/chemical synthesis , Benzene Derivatives/pharmacology , Crystallography, X-Ray , Hydrogen Bonding , Interleukin-6/immunology , Macrophages/drug effects , Macrophages/immunology , Mice , Piperidones/chemical synthesis , Piperidones/pharmacology , RAW 264.7 Cells , Sulfones/chemical synthesis , Sulfones/chemistry , Sulfones/pharmacology , Tumor Necrosis Factor-alpha/immunology
8.
Mol Med Rep ; 15(1): 47-56, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27909717

ABSTRACT

Hepatic fibrosis is a reversible pathological process, in which fibrotic tissue is excessively deposited in the liver during the repair process that follows hepatic injury. Early prevention or treatment of hepatic fibrosis has great significance on the treatment of chronic hepatic diseases. Hydroxysafflor yellow A (HSYA) is a water-soluble monomer extracted from safflower, which serves numerous pharmacological roles. However, it remains to be elucidated how HSYA regulates hepatic fibrogenesis. The aim of the present study was to reveal the possible mechanisms underlying the effects of HSYA on the prevention and treatment of hepatic fibrosis. A rat model of hepatic fibrosis was established in the present study, and the rats were administered various doses of HSYA. The effects of HSYA on pathological alterations of the liver tissue in rats with hepatic fibrosis were observed using hematoxylin-eosin staining and Masson staining. In order to explore the anti­hepatic fibrosis effects and underlying mechanisms of HSYA, serum levels, and hepatic function and hepatic fibrosis indices were evaluated. The results demonstrated that HSYA can improve the general condition of rats with hepatic fibrosis and relieve cellular swelling of the liver, fatty degeneration, necrosis, inflammatory cell infiltration and fibroplastic proliferation. Subsequent to administration of HSYA, globulin was increased during hepatic fibrosis caused by tetrachloromethane. However, total cholesterol, triglyceride, alanine aminotransferase, aspartate aminotransferase and levels of hyaluronic acid, laminin, procollagen â…¢ N­terminal peptide, collagen type â…£ and hydroxyproline were significantly reduced. The results additionally demonstrated that HSYA could enhance superoxide dismutase activity and reduce malondialdehyde levels, inhibiting lipid peroxidation caused by free radicals.


Subject(s)
Chalcone/analogs & derivatives , Liver Cirrhosis, Experimental/drug therapy , Liver Cirrhosis, Experimental/pathology , Liver/drug effects , Liver/pathology , Quinones/therapeutic use , Alanine Transaminase/blood , Alanine Transaminase/metabolism , Animals , Aspartate Aminotransferases/blood , Aspartate Aminotransferases/metabolism , Carbon Tetrachloride , Carthamus tinctorius/chemistry , Chalcone/chemistry , Chalcone/therapeutic use , Lipid Metabolism/drug effects , Liver/metabolism , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/metabolism , Male , Quinones/chemistry , Rats, Sprague-Dawley
9.
Neuroreport ; 27(4): 213-9, 2016 Mar 02.
Article in English | MEDLINE | ID: mdl-26684398

ABSTRACT

The prognosis of patients exposed to a sub-threshold dose of a proconvulsant is difficult to establish. In this study, we investigated the effect of a single sub-threshold dose of the proconvulsant pilocarpine (PILO) on the progression of seizures that were subsequently induced by daily electrical stimulation (kindling) of the amygdaloid formation. Male Sprague­Dawley rats were each implanted with an electrode in the right basolateral amygdala and an indwelling cannula in the right ventricle. The animals were randomized into groups and were administered one of the following treatments: saline, PILO, saline+L-α-aminoadipic acid (L-AAA; one dosage tested), PILO+L-AAA, or PILO+L-methionine sulfoximine (three dosages tested). Amygdaloid stimulation and electroencephalography were performed once daily. We performed immunohistochemistry and western blot for glial fibrillary acidic protein and glutamine synthetase (GS). We also assayed the enzymic activity of GS in discrete brain regions. An intraperitoneal injection of a sub-threshold PILO dose enhanced the progression of amygdaloid-kindling seizures and was accompanied by an increase in reactive-astrocyte and GS (content and activity) in the hippocampus and piriform cortex. L-AAA and L-methionine sulfoximine, inhibitors of astrocytic and GS function, respectively, abolished the effect of PILO on amygdaloid-kindling seizures. We conclude that one sub-threshold dose of a proconvulsant may enhance the progression of subsequent epilepsy and astrocytic GS may play a role in this phenomenon. Thus, a future therapy for epilepsy could be inhibition of astrocytes and/or GS.


Subject(s)
Astrocytes/drug effects , Basolateral Nuclear Complex/drug effects , Glutamate-Ammonia Ligase/metabolism , Kindling, Neurologic/drug effects , Muscarinic Agonists/toxicity , Pilocarpine/toxicity , 2-Aminoadipic Acid/pharmacology , Animals , Astrocytes/enzymology , Basolateral Nuclear Complex/enzymology , Catheters, Indwelling , Disease Models, Animal , Electric Stimulation , Electrodes, Implanted , Fluorescent Antibody Technique , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/drug effects , Hippocampus/enzymology , Kindling, Neurologic/metabolism , Lithium Chloride , Male , Methionine Sulfoximine/pharmacology , Rats, Sprague-Dawley , Temporal Lobe/drug effects , Temporal Lobe/enzymology
10.
Eur J Pharmacol ; 754: 105-14, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25720342

ABSTRACT

Hydroxysafflor yellow A (HSYA), a flavonoid derived and isolated from traditional Chinese medicine Carthamus tinctorius L., possesses anti-tumor activity. However, its effects on hepatocellular carcinoma (HCC) have not been investigated. The proliferation and metastasis of HCC are dependent on angiogenesis, which also strongly links with several signal transduction pathways associated with cell proliferation and apoptosis. This study aimed to explore the effect of HSYA on vasculogenesis and to determine its molecular mechanism by investigating the expression of ERK/MAPK (p-c-Raf, c-Raf, p-ERK1/2, ERK1/2) and NF-κB (p65, IκB and p-IκB) signaling pathway in H22 tumor-bearing mice. The results showed that HSYA could considerably suppress tumor growth by inhibiting secretion of angiogenesis factors (vascular endothelial growth factor A, basic fibroblast growth factor) and vascular endothelial growth factor receptor1. At the moleculcould block ERK1/2 phosphorylation and then restrain the activation of NF-κB and its nuclear translocation by down-regulating the expression of p65 in the nucleus, up-regulating p65 level in the cytoplasm, inhibiting IκB phosphorylation and cytoplasmic degradation of IκB-α. Finally, we demonstrate that HSYA could suppress mRNA expression levels of cell proliferation-related genes (cyclinD1, c-myc, c-Fos) compared with negative control group. And best of all, HSYA could improve spleen/thymus indexes, which was evaluated as the marker of protective effect on the immune system. Our findings support HSYA as a promising candidate for the prevention and treatment of HCC.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Chalcone/analogs & derivatives , Liver Neoplasms/drug therapy , MAP Kinase Signaling System/drug effects , NF-kappa B/antagonists & inhibitors , Neovascularization, Pathologic/drug therapy , Quinones/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/blood supply , Cell Line, Tumor , Cell Proliferation/drug effects , Chalcone/pharmacology , Chalcone/therapeutic use , Liver Neoplasms/blood supply , Male , Mice , NF-kappa B/metabolism , Neoplasm Transplantation , Quinones/therapeutic use
11.
Am J Chin Med ; 42(3): 543-59, 2014.
Article in English | MEDLINE | ID: mdl-24871650

ABSTRACT

To discover and develop novel natural compounds, active ingredients, single herbs and combination formulas or prescriptions in traditional Chinese medicine (TCM) with therapeutic selectivity that can preferentially kill cancer cells and inhibit the amplification of cancer without significant toxicity is an important area in cancer therapy. A lot of valuable TCMs were applied as alternative or complementary medicines in the United States and Europe. But these TCMs, as one of the main natural resources, were widely used to research and develop new drugs in Asia. In TCMs, some specific herbs, animals, minerals and combination formulas were recorded and exploited due to their active ingredients and specific natural compounds with antitumor activities. The article focused on the antitumor properties of natural compounds and combination formulas or prescriptions in TCMs, described its influence on tumor progression, angiogenesis, metastasis, and revealed its mechanisms of antitumor and inhibitory action. Among the nature compounds, triptolide, berberine, matrine, oxymatrine, kurarinone and deoxypodophyllotoxin (DPT) with specific molecular structures have been separated, purified, and evaluated their antitumor properties in vitro and in vivo. Cancer is a multifactorial and multistep disease, so the treatment effect of combination formulas and prescriptions in TCMs involving multi-targets and multi-signal pathways on tumor may be superior than that of agents targeting a single molecular target alone. Shi Quan Da Bu Tang and Yanshu injection, as well known combination formulas and prescriptions in TCMs, have shown an excellent therapeutic effect on cancer.


Subject(s)
Antineoplastic Agents, Phytogenic , Drug Discovery , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Medicine, Chinese Traditional , Neoplasms/drug therapy , Plants, Medicinal/chemistry , Alkaloids/isolation & purification , Alkaloids/pharmacology , Alkaloids/therapeutic use , Amphibian Venoms/chemistry , Amphibian Venoms/isolation & purification , Amphibian Venoms/pharmacology , Amphibian Venoms/therapeutic use , Animals , Berberine/isolation & purification , Berberine/pharmacology , Berberine/therapeutic use , Diterpenes/isolation & purification , Diterpenes/pharmacology , Diterpenes/therapeutic use , Drug Combinations , Drugs, Chinese Herbal/therapeutic use , Epoxy Compounds/isolation & purification , Epoxy Compounds/pharmacology , Epoxy Compounds/therapeutic use , Flavonoids/isolation & purification , Flavonoids/pharmacology , Flavonoids/therapeutic use , Humans , Molecular Conformation , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/pathology , Phenanthrenes/isolation & purification , Phenanthrenes/pharmacology , Phenanthrenes/therapeutic use , Podophyllotoxin/analogs & derivatives , Podophyllotoxin/isolation & purification , Podophyllotoxin/pharmacology , Podophyllotoxin/therapeutic use , Quinolizines/isolation & purification , Quinolizines/pharmacology , Quinolizines/therapeutic use , Matrines
12.
Phytomedicine ; 21(3): 199-206, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24192212

ABSTRACT

Long-term inhalation of gasoline engine exhaust (GEE) increases the risk of respiratory disease. Studies have suggested involvement of platelets in the development of some lung diseases. Hydroxysafflor yellow A (HSYA), a flavonoid compound, prevents hemostasis. Therefore, we investigated its effects on GEE-induced lung injury, and role of platelets in injury. Sixty-week-old male Sprague-Dawley rats were exposed to GEE for 4h/day for 6 weeks, and then grouped as follows: control, GEE, GEE+HSYA, GEE+HSYA+GW9662, and GEE+GW9662. Arterial oxygen tension (PaO2), carbon dioxide tension (PaCO2), pH, and the PaO2/fraction of inspired oxygen ratio (PaO2/FiO2) in the blood were detected using a blood gas analyzer. Wet/dry lung weight ratio, total protein in bronchoalveolar lavage fluid (BALF), and cytokine concentrations in serum and BALF were determined. Furthermore, cyclic adenosine monophosphate (cAMP) level and expression levels of target proteins were analyzed. Platelets were counted and their state was evaluated. HSYA attenuated GEE-mediated decreases in PaO2, PaO2/FiO2, platelet cAMP level, protein kinase A (PKA) activity, and peroxisome proliferator-activated receptor γ (PPARγ) expression. HSYA also attenuated GEE-mediated increases in lung permeability, cytokine levels in serum and BALF, plasma platelet count, and ADP-mediated platelet aggregation. Moreover, it suppressed GEE-induced increases in the expression of adhesion molecules and proinflammatory cytokines in platelets and lung tissue. Therefore, HSYA is therapeutically effective for GEE-mediated lung injury and acts by enhancing PKA activity and inhibiting platelet activation.


Subject(s)
Carthamus tinctorius/chemistry , Chalcone/analogs & derivatives , Gasoline , Lung Injury/prevention & control , Phytotherapy , Platelet Activation/drug effects , Quinones/therapeutic use , Vehicle Emissions , Animals , Bronchoalveolar Lavage Fluid , Chalcone/pharmacology , Chalcone/therapeutic use , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytokines/blood , Down-Regulation , Inhalation Exposure/adverse effects , Lung Injury/chemically induced , Lung Injury/metabolism , Male , Motor Vehicles , PPAR gamma/metabolism , Permeability , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Platelet Aggregation/drug effects , Platelet Count , Quinones/pharmacology , Rats , Rats, Sprague-Dawley
13.
Int J Clin Exp Pathol ; 7(5): 2595-608, 2014.
Article in English | MEDLINE | ID: mdl-24966974

ABSTRACT

Hydroxysafflor yellow A (HSYA), a major constituent in the hydrophilic fraction of the safflower plant, can retard the progress of hepatic fibrosis. However, the anti-inflammatory properties and the underlying mechanisms of HSYA on I/R-induced acute liver injury are unknown. Inhibiting macrophage activation is a potential strategy to treat liver ischemia/reperfusion (I/R) injury. In this study, we investigated the therapeutic effect of HSYA on liver I/R injury and the direct effect of HSYA on macrophage activation following inflammatory conditions. The therapeutic effects of HSYA on I/R injury were tested in vivo using a mouse model of segmental (70%) hepatic ischemia. The mechanisms of HSYA were examined in vitro by evaluating migration and the cytokine expression profile of the macrophage cell line RAW264.7 exposed to acute hypoxia and reoxygenation (H/R). Results showed that mice pretreated with HSYA had reduced serum transaminase levels, attenuated inflammation and necrosis, reduced expression of inflammatory cytokines, and less macrophage recruitment following segmental hepatic ischemia. In vitro HSYA pretreated RAW264.7 macrophages displayed reduced migratory response and produced less inflammatory cytokines. In addition, HSYA pretreatment down-regulated the expression of matrix matalloproteinase-9 and reactive oxygen species, and inhibited NF-κB activation and P38 phosphorylation in RAW264.7 cells. Thus, these data suggest that HSYA can reduce I/R-induced acute liver injury by directly attenuating macrophage activation under inflammatory conditions.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Chalcone/analogs & derivatives , Liver Diseases/prevention & control , Liver/drug effects , Macrophage Activation/drug effects , Macrophages/drug effects , Quinones/pharmacology , Reperfusion Injury/prevention & control , Animals , Cell Line , Chalcone/pharmacology , Chemotaxis/drug effects , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Inflammation Mediators/metabolism , Liver/immunology , Liver/metabolism , Liver/pathology , Liver Diseases/immunology , Liver Diseases/metabolism , Liver Diseases/pathology , Macrophages/immunology , Macrophages/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , NF-kappa B/metabolism , Phosphorylation , Reactive Oxygen Species/metabolism , Reperfusion Injury/immunology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction/drug effects , Time Factors , p38 Mitogen-Activated Protein Kinases/metabolism
14.
J Clin Neurosci ; 17(4): 495-500, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20171893

ABSTRACT

Monosialoganglioside GM1 is a known neurotrophic factor. Nerve growth factor (NGF), a member of the neurotrophin family, is important for the survival, differentiation and maturation of neurons. The aim of this study was to test whether administration of GM1 and NGF can ameliorate glutamate (Glu) neurotoxicity in primary cultured embryonic rat dorsal root ganglia (DRG) and spinal cord neurons, and to investigate the mechanism underlying any effect. DRG and spinal cord neurons were exposed to the following treatments: Glu (2 mmol/L); Glu (2 mmol/L) plus GM1 (10mg/mL); Glu (2 mmol/l) plus NGF (10 ng/mL); Glu (2 mmol/L) plus GM1 (5mg/mL) and NGF (5 ng/mL). Cell viability was assessed using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, ultrastructural alterations were examined using inverse phase contrast microscopy and electron microscopy, mitochondrial membrane potential was measured using rhodamine 123 labeling and flow cytometry, and neurofilament light (NF-L) mRNA expression was detected by reverse transcription-polymerase chain reaction. It was found that GM1 and NGF can increase the viability of neurons incubated with Glu, which, after GM1 and NGF treatment, were almost morphologically normal. The mitochondrial membrane potential of neurons was lowest for neurons treated with Glu alone, and that for neurons treated with Glu plus GM1 and NGF was higher than that for treatment with GM1 or NGF alone. The mRNA of NF-L was expressed at the highest level in neurons treated with Glu plus GM1 and NGF. Our findings indicate that NGF and GM1 act synergistically to protect DRG and spinal cord neurons from Glu cytotoxicity. NGF and GM1 may function by maintaining normal mitochondrial membrane potential or by promoting NF-L mRNA expression.


Subject(s)
G(M1) Ganglioside/pharmacology , Membrane Potential, Mitochondrial/drug effects , Nerve Growth Factor/pharmacology , Neurofilament Proteins/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Cell Separation , Cell Survival/drug effects , Cells, Cultured , Drug Synergism , Flow Cytometry , Ganglia, Spinal/drug effects , Ganglia, Spinal/ultrastructure , Glutamic Acid/toxicity , Microscopy, Electron, Transmission , Neurofilament Proteins/biosynthesis , Neurons/ultrastructure , RNA, Messenger , Rats , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/drug effects
15.
Brain Res Bull ; 79(1): 85-8, 2009 Apr 06.
Article in English | MEDLINE | ID: mdl-19133317

ABSTRACT

Monosialoganglioside (GM1) has been considered to have a neurotrophic factor-like activity. Nerve growth factor (NGF), a member of the neurotrophin family, is essential for neuronal survival, differentiation and maturation. The aim of the present study was to investigate whether co-administration of GM1 and NGF reverses glutamate (Glu) neurotoxicity in primary cultured rat embryonic spinal cord neurons. Spinal cord neurons were exposed to Glu (2 mmol/l), Glu (2 mmol/l) plus GM1 (10 mg/ml), Glu (2 mmol/l) plus NGF (10 ng/ml), Glu (2 mmol/l) plus GM1 (5 mg/ml) and NGF (5 ng/ml) and then processed for detecting intracellular concentrations of Ca2+([Ca2+]i) by confocal laser scanning microscopy and growth associated protein 43 (GAP43) mRNA by RT-PCR. The fluorescent intensity in Glu plus GM1 and NGF incubated neurons was the lowest as compared with that in other groups. The expression of GAP43 mRNA in Glu plus GM1 and NGF incubated neurons was the highest as compared with that in other groups. These results implicated that GM1 and NGF have synergistic neuroprotective effects on spinal cord neurons with excitotoxicity induced by Glu in vitro.


Subject(s)
G(M1) Ganglioside/administration & dosage , Nerve Growth Factor/administration & dosage , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Spinal Cord/cytology , Analysis of Variance , Animals , Calcium/metabolism , Cells, Cultured , Drug Therapy, Combination , GAP-43 Protein/metabolism , Microscopy, Confocal , Neurons/physiology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL