Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Molecules ; 28(21)2023 Oct 27.
Article in English | MEDLINE | ID: mdl-37959722

ABSTRACT

OBJECTIVE: Our study aims to assess Ardisia japonica (AJ)'s anti-blood-stasis effect and its underlying action mechanisms. METHODS: The primary components of AJ were determined using liquid chromatography-mass spectrometry (LC-MS). The blood stasis model was used to investigate the anti-blood-stasis effect of AJ extract. The underlying mechanisms of AJ against blood stasis were investigated via network pharmacology, molecular docking, and plasma non-targeted metabolomics. RESULTS: In total, 94 compounds were identified from an aqueous extract of AJ, including terpenoids, phenylpropanoids, alkaloids, and fatty acyl compounds. In rats with blood stasis, AJ reduced the area of stasis, decreased the inflammatory reaction in the liver and lungs of rats, lowered the plasma viscosity, increased the index of erythrocyte deformability, and decreased the index of erythrocyte aggregation, suggesting that AJ has an anti-blood-stasis effect. Different metabolites were identified via plasma untargeted metabolomics, and it was found that AJ exerts its anti-blood-stasis effect by reducing inflammatory responses through the cysteine and methionine metabolism, linolenic acid metabolism, and sphingolipid metabolism. For the effect of AJ on blood stasis syndrome, the main active ingredients predicted via network pharmacology include sinensetin, galanin, isorhamnetin, kaempferol, wogonin, quercetin, and bergenin, and their targets were TP53, HSP90AA1, VEGFA, AKT1, EGFR, and PIK3CA that were mainly enriched in the PI3K/AKT and MAPK signaling pathways, which modulate the inflammatory response. Molecular docking was also performed, and the binding energies of these seven compounds to six proteins were less than -5, indicating that the chemical components bind to the target proteins. CONCLUSIONS: This study suggests AJ effectively prevents blood stasis by reducing inflammation.


Subject(s)
Ardisia , Drugs, Chinese Herbal , Rats , Animals , Network Pharmacology , Molecular Docking Simulation , Phosphatidylinositol 3-Kinases , Metabolomics/methods , Drugs, Chinese Herbal/pharmacology , Inflammation/drug therapy
2.
J Hepatol ; 76(1): 148-159, 2022 01.
Article in English | MEDLINE | ID: mdl-34689996

ABSTRACT

BACKGROUND & AIMS: Regulatory T cell (Treg) depletion increases antitumor immunity. However, severe autoimmunity can occur following systemic loss of Tregs, which could be avoided by selectively depleting intratumoral Tregs. Herein, we aimed to investigate the role of tumor-infiltrating CCR4+ Tregs in hepatocellular carcinoma (HCC) and to provide a potential target strategy for immunotherapy. METHODS: CCR4+ Tregs were analyzed by flow cytometry in murine models and clinical samples. The function of tumor-infiltrating and induced CCR4+ Tregs was interrogated by genetic and epigenetic approaches. To block CCR4+ Treg chemotaxis, we developed an N-terminus recombinant protein of CCR4 (N-CCR4-Fc) as a neutralizing pseudo-receptor that effectively bound to its ligand CCL22. The efficacy of CCR4 antagonism as an immunotherapeutic agent was evaluated by tumor weights, growth kinetics and survival curves. RESULTS: CCR4+ Tregs were the predominant type of Tregs recruited to hepatitis B-associated HCC (HBV+ HCC), correlating with sorafenib resistance and HBV load titers. Compared with CCR4- Tregs, CCR4+ Tregs exhibited increased IL-10 and IL-35 expression, and enhanced functionality in suppressing CD8+ T cells. CCR4+ Tregs also displayed PD-1+TCF1+ stem-like properties. ATAC-seq data revealed substantial chromatin remodeling between tumor-infiltrating Tregs (TIL-Tregs) and induced Tregs, suggesting that long-term chromatin reprogramming accounted for the acquisition of enhanced immunosuppressive stem-like specificity by CCR4+ TIL-Tregs. Treatment with a CCR4 antagonist or N-CCR4-Fc blocked intratumoral Treg accumulation, overcame sorafenib resistance, and sensitized tumors to PD-1 checkpoint blockade. CONCLUSIONS: Intratumoral stem-like CCR4+ Tregs orchestrated immunosuppressive resource cells in the tumor microenvironment. CCR4 could be targeted to enhance antitumor immunity by specifically blocking infiltration of Tregs into the tumor microenvironment and inhibiting maintenance of the TIL-Treg pool. LAY SUMMARY: Targeting regulatory T cells is a promising approach in cancer immunotherapy; however, severe autoimmunity can occur following systemic regulatory T cell loss. This could be avoided by selectively depleting intratumoral regulatory T cells. Herein, targeting intratumoral stem-like CCR4+ regulatory T cells helped to overcome sorafenib resistance and sensitize tumors to immune checkpoint blockade in mouse models of liver cancer. This approach could have wide clinical applicability.


Subject(s)
Carcinoma, Hepatocellular/etiology , Hepatitis B/complications , Immunocompromised Host/drug effects , Receptors, CCR4/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , China , Disease Models, Animal , Hepatitis B/immunology , Hepatitis B virus/drug effects , Hepatitis B virus/pathogenicity , Immunocompromised Host/genetics , Immunocompromised Host/immunology , Liver Neoplasms/etiology , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Mice , Receptors, CCR4/immunology , T-Lymphocytes, Regulatory/immunology
3.
J Virol ; 94(22)2020 10 27.
Article in English | MEDLINE | ID: mdl-32847856

ABSTRACT

The ongoing coronavirus disease 2019 (COVID-19) pandemic has caused >20 million infections and >750,000 deaths. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiological agent of COVID-19, has been found closely related to the bat coronavirus strain RaTG13 (Bat-CoV RaTG13) and a recently identified pangolin coronavirus (Pangolin-CoV-2020). Here, we first investigated the ability of SARS-CoV-2 and three related coronaviruses to utilize animal orthologs of angiotensin-converting enzyme 2 (ACE2) for cell entry. We found that ACE2 orthologs of a wide range of domestic and wild mammals, including camels, cattle, horses, goats, sheep, cats, rabbits, and pangolins, were able to support cell entry of SARS-CoV-2, suggesting that these species might be able to harbor and spread this virus. In addition, the pangolin and bat coronaviruses, Pangolin-CoV-2020 and Bat-CoV RaTG13, were also found able to utilize human ACE2 and a number of animal-ACE2 orthologs for cell entry, indicating risks of spillover of these viruses into humans in the future. We then developed potently anticoronavirus ACE2-Ig proteins that are broadly effective against the four distinct coronaviruses. In particular, through truncating ACE2 at its residue 740 but not 615, introducing a D30E mutation, and adopting an antibody-like tetrameric-ACE2 configuration, we generated an ACE2-Ig variant that neutralizes SARS-CoV-2 at picomolar range. These data demonstrate that the improved ACE2-Ig variants developed in this study could potentially be developed to protect from SARS-CoV-2 and some other SARS-like viruses that might spillover into humans in the future.IMPORTANCE The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the etiological agent of the currently uncontrolled coronavirus disease 2019 (COVID-19) pandemic. It is important to study the host range of SARS-CoV-2, because some domestic species might harbor the virus and transmit it back to humans. In addition, insight into the ability of SARS-CoV-2 and SARS-like viruses to utilize animal orthologs of the SARS-CoV-2 receptor ACE2 might provide structural insight into improving ACE2-based viral entry inhibitors. In this study, we found that ACE2 orthologs of a wide range of domestic and wild animals can support cell entry of SARS-CoV-2 and three related coronaviruses, providing insights into identifying animal hosts of these viruses. We also developed recombinant ACE2-Ig proteins that are able to potently block these viral infections, providing a promising approach to developing antiviral proteins broadly effective against these distinct coronaviruses.


Subject(s)
Antibodies, Neutralizing/genetics , Betacoronavirus/physiology , Coronavirus/classification , Angiotensin-Converting Enzyme 2 , Animals , Antibodies, Neutralizing/chemistry , Betacoronavirus/genetics , Coronavirus/genetics , Coronavirus/physiology , Disease Models, Animal , HEK293 Cells , Humans , Immunoglobulins/chemistry , Immunoglobulins/genetics , Models, Chemical , Peptidyl-Dipeptidase A/chemistry , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Receptors, Virus/chemistry , Receptors, Virus/genetics , Recombinant Proteins/genetics , SARS-CoV-2 , Virus Internalization/drug effects
4.
Pharmacol Res ; 157: 104800, 2020 07.
Article in English | MEDLINE | ID: mdl-32278046

ABSTRACT

The HBV-initiated hepatocellular carcinoma (HCC) frequently develops from or accompanies long-term chronic hepatitis, inflammation, and cirrhosis, and has a poor prognosis. Sorafenib, an orally active multi-kinase inhibitor, currently the most common approved drug for first-line systemic treatment of advanced HCC, only improves overall survival of three months, suggesting the need for new therapeutic strategies. In this study, we identified that sorafenib selectively resisted in immune competent C57BL/6 mice but not nude mice. The chemokines CCL22 and CCL17 were upregulated by sorafenib, which elevated dramatically higher in HBV-associated HCC. Mechanically, sorafenib accelerates CCL22 expression via TNF-α-RIP1-NF-κB signaling pathway. Blocking CCL22 signaling with antagonist C-021 and sorafenib treated in combination can inhibit tumor growth and enhance the antitumor response, whereas no significant differences in tumor burden were observed in nude mice upon addition of C-021. These findings strongly suggest that CCL22 signaling pathway strongly contributes to sorafenib resistance in HBV-associated HCC, indicating a potential therapeutic strategy for immunological chemotherapy complementing first-line agents against HBV-associated HCC.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Chemokine CCL22/metabolism , Drug Resistance, Neoplasm , Liver Neoplasms/drug therapy , Lymphocytes, Tumor-Infiltrating/drug effects , Protein Kinase Inhibitors/pharmacology , Sorafenib/pharmacology , T-Lymphocytes/drug effects , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Chemokine CCL22/antagonists & inhibitors , Chemokine CCL22/genetics , Hep G2 Cells , Hepatitis B/complications , Hepatitis B/virology , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/virology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Quinazolines/pharmacology , Receptors, CCR4/antagonists & inhibitors , Receptors, CCR4/genetics , Receptors, CCR4/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Tumor Burden/drug effects
5.
Materials (Basel) ; 17(16)2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39203213

ABSTRACT

The electrochemical corrosion behaviors of Ti3SiC2/Cu composites in harsh media including dilute HNO3 and concentrated H2SO4 were studied in detail and the related corrosion mechanisms were explored. Under open-circuit potential, the corrosion resistance of Ti3SiC2/Cu in dilute HNO3 was worse than that in concentrated H2SO4. In dilute HNO3, Ti3SiC2/Cu exhibited a typical passivation character with a narrow passivation interval. During the corrosion process, the dissolution of Cu-Si compounds resulted in the destruction of the passivation layer on the surface. Additionally, with the increasing of the potentials, the oxidation of Cu and Si atoms led to the generation of the oxide film again on the surface. In concentrated H2SO4, the Ti3SiC2/Cu composite was covered by a double-layered passivation layer, which was composed of an internal layer of TiO2 and an external layer of Cu2O and SiO2. This was because Cu diffused into the surface and was oxidized into Cu2O, which formed a denser oxidized film with SiO2. In addition, it was found that Ti3SiC2/Cu has better corrosion resistance in concentrated H2SO4.

6.
Microbiol Spectr ; 11(6): e0267623, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37943512

ABSTRACT

IMPORTANCE: Spike-receptor interaction is a critical determinant for the host range of coronaviruses. In this study, we investigated the SARS-CoV-2 WHU01 strain and five WHO-designated SARS-CoV-2 variants of concern (VOCs), including Alpha, Beta, Gamma, Delta, and the early Omicron variant, for their Spike interactions with ACE2 proteins of 18 animal species. First, the receptor-binding domains (RBDs) of Alpha, Beta, Gamma, and Omicron were found to display progressive gain of affinity to mouse ACE2. More interestingly, these RBDs were also found with progressive loss of affinities to multiple ACE2 orthologs. The Omicron RBD showed decreased or complete loss of affinity to eight tested animal ACE2 orthologs, including that of some livestock animals (horse, donkey, and pig), pet animals (dog and cat), and wild animals (pangolin, American pika, and Rhinolophus sinicus bat). These findings shed light on potential host range shift of SARS-CoV-2 VOCs, especially that of the Omicron variant.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 , Cat Diseases , Chiroptera , Dog Diseases , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Animals , Cats , Dogs , Mice , Angiotensin-Converting Enzyme 2/metabolism , Animals, Wild/virology , Cat Diseases/virology , Chiroptera/virology , COVID-19/metabolism , Dog Diseases/virology , Horses/virology , Mutation , Protein Binding , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Swine/virology , Spike Glycoprotein, Coronavirus/genetics
7.
Cancer Res ; 81(9): 2386-2398, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33653774

ABSTRACT

Hepatitis B-associated hepatocellular carcinoma (HCC) is often accompanied by severe vascular invasion and portal vein tumor thrombus, leading to a poor prognosis. However, the underlying mechanism of this disease remains obscure. In this study, we demonstrate that the hepatitis B virus (HBV)-encoded gene HBx induces high IL8 production through MEK-ERK signal activation, leading to enhanced endothelial permeability to facilitate tumor vascular invasion. In a vascular metastatic model using a tail vein injection in a transgenic mouse with selective expression of human CXCR1 in the endothelium, activation of the IL8-CXCR1 cascade by overexpression of IL8 in tumor cells dramatically enhanced liver metastasis. Mechanistically, IL8 selectively induced GARP-latent-TGFß in liver sinusoidal endothelial cells and subsequently provoked preferential regulatory T-cell polarization to suppress antitumor immunity. Collectively, these findings reveal a hepatitis B-associated IL8-CXCR1 signaling axis that mediates vascular invasion and local microenvironmental immune escape of HCC to induce intrahepatic metastasis, which may serve as potential therapeutic targets for HBV-associated HCC. SIGNIFICANCE: This study identifies a hepatitis B-induced IL8/CXCR1/TGFß signaling cascade that suppresses antitumor immunity and enhances metastasis in hepatocellular carcinoma, providing new potential targets for therapeutic intervention.


Subject(s)
Carcinoma, Hepatocellular/secondary , Carcinoma, Hepatocellular/virology , Hepatitis B virus/metabolism , Hepatitis B/complications , Interleukin-8/metabolism , Liver Neoplasms/secondary , Liver Neoplasms/virology , Liver/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Disease Models, Animal , HEK293 Cells , Hep G2 Cells , Hepatitis B/virology , Hepatitis B virus/genetics , Human Umbilical Vein Endothelial Cells , Humans , Interleukin-8/genetics , Interleukin-8/pharmacology , Liver Neoplasms/metabolism , MAP Kinase Signaling System/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Interleukin-8A/genetics , Receptors, Interleukin-8A/metabolism , Recombinant Proteins/pharmacology , Trans-Activators/genetics , Trans-Activators/metabolism , Transfection , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/metabolism
8.
Oncotarget ; 8(7): 11555-11565, 2017 Feb 14.
Article in English | MEDLINE | ID: mdl-28030791

ABSTRACT

PURPOSE: Dexamethasone (Dex), a glucocorticoid (GC), is used as a pretreatment drug in cancer patients undergoing chemotherapy. Dex functions by binding to the glucocorticoid receptor (GR) to prevent allergic reactions and severe chemotherapeutic side effects such as nausea and vomiting. However, the mechanisms by which Dex causes chemoresistance remain unknown. METHODS: We used docetaxel and cisplatin to treat triple-negative breast cancer (TNBC) cells with or without Dex and assessed cell proliferation using a sulforhodamine B colorimetric (SRB) assay. Additionally, Western blotting was employed to measure Krüppel-like factor 5 (KLF5), GR and several apoptosis-related proteins. To determine how the GR regulates KLF5, we used qRT-PCR, luciferase reporter assays and ChIP assays. Finally, we detected the involvement of Dex in TNBC chemotherapeutic resistance using HCC1806 xenograft model in vivo. RESULTS: In this study, we demonstrated that Dex induces docetaxel and cisplatin resistance in TNBC cells in vitro and in vivo. Dex up-regulates pro-survival transcription factor KLF5 expression at both mRNA and protein levels dependent on GR. Importantly, Dex failed to promote cancer cell survival and tumor growth when KLF5 induction was blocked. CONCLUSIONS: We conclude that KLF5 is a Dex-induced gene that contributes to Dex-mediated drug chemoresistance, providing a potential novel target for TNBC treatment.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Kruppel-Like Transcription Factors/metabolism , Taxoids/pharmacology , Anti-Inflammatory Agents/pharmacology , Dexamethasone/pharmacology , Docetaxel , Female , Humans , Up-Regulation
9.
PLoS One ; 9(8): e104261, 2014.
Article in English | MEDLINE | ID: mdl-25101695

ABSTRACT

The Tongshu Capsule (TSC) is a prevalent form of traditional Chinese medicine widely used for its purported effects in treating mammary gland hyperplasia and inflammation. Though successful in several clinical studies, there is no clear evidence as to why TSC has a positive treatment effect, and little known about underlying mechanism that may account for it. In this study, we examined the effects of TSC and found that it has a comparatively strong growth inhibition on ERα positive breast cancer cells. TSC seems to cause G1 cell cycle arrest instead of apoptosis. Interestingly, TSC also down-regulated the expression of ERα and Cyclin D1. Consistently, TSC suppressed E2 mediated ERα downstream gene expression and cell proliferation in ERα positive breast cancer cell lines MCF7 and T47D. Depletion of ERα partially abolished the effects of TSC on the decrease of Cyclin D1 and cell viability. Our findings suggest that TSC may have therapeutic effects on ERα positive breast cancers and moreover that TSC may suppress breast epithelial cell proliferation by inhibiting the estrogen pathway.


Subject(s)
Breast Neoplasms , Estrogen Receptor alpha/biosynthesis , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation/drug effects , Medicine, Chinese Traditional , Signal Transduction/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cyclin D1/metabolism , Estrogens/metabolism , Female , Humans
10.
PLoS One ; 9(5): e95307, 2014.
Article in English | MEDLINE | ID: mdl-24798046

ABSTRACT

PURPOSE: Hepatocellular carcinoma (HCC) is the sixth most common solid tumor worldwide and the third leading cause of cancer-related death. HCC is a particularly serious threat to the Chinese population. Although many molecular alterations are known to be involved in the tumorigenesis of hepatocytes, no systemic survey has examined the somatic mutations in HCC samples from Chinese patients. Our goal was to elucidate somatic mutations in Chinese HCC patients and investigate the possible molecular mechanisms involved in tumorigenesis. EXPERIMENTAL DESIGN: A total of 110 hepatitis B virus (HBV)-positive HCC samples and 46 HBV-negative HCC samples were genotyped for hot-spot mutations in the CSF1R, CTNNB1, KRAS, BRAF, NRAS, ERBB2, MET, PIK3CA, JAK1, and SMO genes. The transcriptomes of the CTNNB1 mutation-positive HCC samples from the HBV-positive patients (CB+ HCC) were compared to adjacent non-cancerous livers, and significantly altered genes were functionally validated in vitro. RESULTS: CTNNB1 mutations accounted for the majority of the mutations detected in our study. A slightly higher mutation rate was found in the HBV-positive patients than in their negative counterparts. A distinct pattern of CTNNB1 mutation was detected in these two populations, and drastic changes at the transcriptomic level were detected in the CB+ tumors compared to adjacent non-cancerous livers. Potential tumor suppressors (FoxA3 and Onecut1) and oncogenes (MAFG and SSX1) were functionally validated. CONCLUSIONS: Our work is the first systemic characterization of oncogenic mutations in HCC samples from Chinese patients. Targeting the Wnt-ß-catenin pathway may represent a valid treatment option for Chinese HCC patients. Our work also suggests that targeting ONECUT1, FOXA3, SSX1, and MAFG may be a valid treatment option for CTNNB1 mutation positive HCC patients.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Mutation , Neoplasm Proteins/biosynthesis , Transcriptome/genetics , beta Catenin , Adult , Aged , Asian People , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , China , Female , Gene Expression Profiling , Hepatitis B/genetics , Hepatitis B/metabolism , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Proteins/genetics , Wnt Signaling Pathway/genetics , beta Catenin/biosynthesis , beta Catenin/genetics
11.
Biol Trace Elem Res ; 152(1): 50-6, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23279943

ABSTRACT

This study aimed to assess whether maifanite can improve the learning and memory, and antioxidant abilities of Alzheimer's disease (AD) rats. The 70 rats were divided into seven groups: [A] normal control group, [B] AD model group, [C] sham group, [D] positive control group (donepezil), [E] low-dose maifanite group, [F] middle-dose maifanite group, [G] high-dose maifanite group. For [B], [D], [E], [F], and [G] groups, Aß(25-35) ventricle injection was carried out, then respective medicine were administered once a day for 60 consecutive days. The step-down and step-through test were used to measure learning and memory ability. The hippocampus levels of superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), and malondialdehyde (MDA) were assayed. The hippocampus contents of Al, Fe, Cu, Zn, Se, and Mn were analyzed by inductively coupled plasma-atomic emission spectrometer. Maifanite decreased the acquisition errors and the retention errors while prolonging the step-down latency, and decreased the number of electric shocks while prolonging the first latency of AD rats. Aß(25-35) ventricle injection initiated the decrease of SOD and GSH-Px activities and the increase of MDA content, and triggered the rise of Al, Fe, and Cu levels and the decline of Mn, Zn, and Se levels. The SOD and GSH-Px activities were enhanced followed by reduced MDA level, and the levels of Mn, Zn, and Se increased accompanied by Al, Fe, and Cu decreased in the maifanite treat groups. Maifanite could improve the learning and memory, and the antioxidant abilities of AD rats. Maifanite had the potential prevention and treatment for AD.


Subject(s)
Alzheimer Disease/prevention & control , Avoidance Learning/drug effects , Hippocampus/drug effects , Oxidative Stress/drug effects , Trace Elements/pharmacology , Alzheimer Disease/chemically induced , Alzheimer Disease/physiopathology , Amyloid beta-Peptides , Animals , Glutathione Peroxidase/metabolism , Hippocampus/metabolism , Male , Malondialdehyde/metabolism , Medicine, Chinese Traditional , Memory/drug effects , Peptide Fragments , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism , Trace Elements/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL