1.Chemical constituents from Alternaria alternata and their activity of down-regulating TMSB10 expression.
Lan WANG ; Nuan ZHANG ; Tao YUAN
China Journal of Chinese Materia Medica 2025;50(1):139-156
This study investigated the secondary metabolites in the product of rice fermentation with Alternaria alternata and their activity of down-regulating thymosin beta 10(TMSB10) protein expression. The secondary metabolites of A. alternata were separated and purified by various chromatographic separation techniques, including silica gel column chromatography, Sephadex LH-20 column chromatography, and semi-preparative high-performance liquid chromatography. Their structures were identified by spectral techniques such as nuclear magnetic resonance spectroscopy(NMR), infrared spectroscopy(IR), and high-resolution electrospray ionization mass spectrometry(HR-ESI-MS) and comparison with literature data. A total of 10 compounds were isolated and identified, including two new compounds(1S,3S)-2,3-dihydro-3,6-dihydroxy-8-methoxy-1-methylcyclopenta[c][2]benzopyran-5(1H)-one(1), 3,3a, 6-trihydroxy-8-methoxy-1-methyl-2,3,3a, 9b-tetrahydrocyclopenta[c]isochromen-5(1H)-one(2), and the eight known compounds are alternariol 9-methyl ether(3), 1-deoxyrubralactone(4),(3aR)-3,3a-dihydro-1,6-dihydroxy-8-methoxy-3a-methylcyclopenta[c][2]benzopyran-2,5-dione(5), altechromone A(6), alternariol(7), altenuene(8), altenusin(9), and 3'-hydroxyalternariol 5-O-methyl ether(10). The effects of these compounds on TMSB10 expression were tested. Compound 7 showed a significant down-regulation effect on TMSB10 expression with an inhibition rate of 40.5%. The results showed that benzopyrone compounds of A. alternata have the activity of down-regulating the expression of TMSB10 protein, providing theoretical basis and research value for the study of non-small cell lung cancer.
Alternaria/metabolism*
;
Down-Regulation/drug effects*
;
Humans
;
Molecular Structure
2.Effects of Down-regulation of NCL Expression on the Biological Behavior of Acute Myeloid Leukemia Kasumi-1 Cells.
Hui-Li LIU ; Wen-Xin XU ; Yang-Yan CAI ; Hong-Mei LI
Journal of Experimental Hematology 2025;33(5):1312-1317
OBJECTIVE:
To investigate the role of nucleolin (NCL) in acute myeloid leukemia (AML) Kasumi-1 cells and its underlying mechanism.
METHODS:
The Kasumi-1 cells were infected with lentivirus carrying shRNA to downregulate NCL expression. Cell proliferation was detected by CCK-8 assay, and cell apoptosis and cell cycle were determined by flow cytometry. Transcriptome next-generation sequencing (NGS) was performed to predict associated signaling pathways, the expression levels of related genes were measured by RT-PCR.
RESULTS:
Down-regulation of NCL expression significantly inhibited the proliferation of Kasumi-1 cells (P <0.01) and markedly increased the apoptosis rate (P <0.001). Cell cycle analysis showed significant changes in the distribution of cells in the G1 and S phases after NCL knockdown (P <0.05), while no significant difference was observed in the G2 phase (P >0.05). Transcriptome sequencing analysis demonstrated that differentially expressed genes in Kasumi-1 cells with low expression of NCL were primarily enriched in key signaling pathways, including ribosome, spliceosome, RNA transport, cell cycle, and amino acid biosynthesis. qPCR validation showed that the expression of BAX, CASP3, CYCS, PMAIP1, TP53 , and CDKN1A was significantly upregulated after NCL downregulation (P <0.05), with CDKN1A exhibiting the most pronounced difference.
CONCLUSION
NCL plays a critical role in regulating the proliferation, apoptosis, and cell cycle progression of Kasumi-1 cells. The mechanism likely involves suppressing cell cycle progression through activation of the TP53-CDKN1A pathway and promoting apoptosis by upregulating apoptosis-related genes.
Humans
;
Leukemia, Myeloid, Acute/pathology*
;
Down-Regulation
;
Cell Proliferation
;
Apoptosis
;
RNA-Binding Proteins/genetics*
;
Nucleolin
;
Cell Line, Tumor
;
Phosphoproteins/metabolism*
;
Cell Cycle
;
Signal Transduction
;
RNA, Small Interfering
3.Hypertension exacerbates postoperative learning and memory impairment in rats possibly due to UCP2 downregulation-mediated mitochondrial dysfunction.
Luyu LIU ; Maowei GONG ; Guosong LIAO ; Weixing ZHAO ; Qiang FU
Journal of Southern Medical University 2025;45(4):725-735
OBJECTIVES:
To explore the correlation of hypertension with postoperative cognitive dysfunction and its possible mechanism.
METHODS:
Twelve-week-old spontaneously hypertensive rats (SHRs) and Wistar-Kyoto (WKY) rats were both randomized into control group and surgical group (n=8). In the latter group, the rats received carotid artery exposure surgery under sevoflurane anesthesia to establish models of postoperative learning and memory impairment. Postoperative cognitive function changes of the rats were evaluated using behavioral tests. The hippocampus of the rats were collected for determining ATP level and mitochondrial membrane potential (MMP) and for detecting expressions of UCP2 and astrocyte markers (GFAP and NOX4) using Western blotting and immunofluorescence staining. Serum levels of ROS, IL-6, IL-1β and TNF‑α were detected using ELISA. Nissl staining was used to examine hippocampal neuronal loss in the CA1 region.
RESULTS:
The SHRs exhibited exacerbated learning and memory deficits following the surgery as shown by significantly reduced performance in novel object recognition tests and context-related and tone-related fear conditioning experiments. Compared with WKY rats, the SHRs had significantly decreased mitochondrial UCP2 expression and MMP in the hippocampus, increased hippocampal ATP level, and markedly increased serum levels of ROS and inflammatory factors, showing also increased activation of hippocampal astrocytes and microglia and reduced number of neurons positive for Nissl staining.
CONCLUSIONS
Hypertension can exacerbate major postoperative learning and memory impairment in rats possibly as a result of UCP2-mediated mitochondrial dysfunction and oxidative stress damage, which further leads to astrocyte overactivation and neuronal damage.
Animals
;
Rats, Inbred SHR
;
Rats
;
Uncoupling Protein 2
;
Rats, Inbred WKY
;
Hypertension/physiopathology*
;
Hippocampus/metabolism*
;
Mitochondria/metabolism*
;
Down-Regulation
;
Male
;
Memory Disorders/etiology*
;
Mitochondrial Proteins/metabolism*
4.Curcumin inhibits lipid metabolism in non-small cell lung cancer by downregulating the HIF-1α pathway.
Dandan LI ; Jiaxin CHU ; Yan YAN ; Wenjun XU ; Xingchun ZHU ; Yun SUN ; Haofeng DING ; Li REN ; Bo ZHU
Journal of Southern Medical University 2025;45(5):1039-1046
OBJECTIVES:
To investigate the effect of curcumin on lipid metabolism in non-small cell lung cancer (NSCLC) and its molecular mechanism.
METHODS:
The inhibitory effect of curcumin (0-70 μmol/L) on proliferation of A549 and H1299 cells was assessed using MTT assay, and 20 and 40 μmol/L curcumin was used in the subsequent experiments. The effect of curcumin on lipid metabolism was evaluated using cellular uptake assay, wound healing assay, triglyceride (TG)/free fatty acid (NEFA) measurements, and Oil Red O staining. Western blotting was performed to detect the expressions of PGC-1α, PPAR-α, and HIF-1α in curcumin-treated cells. Network pharmacology was used to predict the metabolic pathways, and the results were validated by Western blotting. In a nude mouse model bearing A549 cell xenograft, the effects of curcumin (20 mg/kg) on tumor growth and lipid metabolism were assessed by measuring tumor weight and observing the changes in intracellular lipid droplets.
RESULTS:
Curcumin concentration-dependently inhibited the proliferation of A549 and H1299 cells and significantly reduced TG and NEFA levels and intracellular lipid droplets. Western blotting revealed that curcumin significantly upregulated PGC-1α and PPAR‑α expressions in the cells. KEGG pathway enrichment analysis predicted significant involvement of the HIF-1 signaling pathway in curcumin-treated NSCLC, suggesting a potential interaction between HIF-1α and PPAR‑α. Western blotting confirmed that curcumin downregulated the expression of HIF-1α. In the tumor-bearing mice, curcumin treatment caused significant reduction of the tumor weight and the number of lipid droplets in the tumor cells.
CONCLUSIONS
Curcumin inhibits NSCLC cell proliferation and lipid metabolism by downregulating the HIF-1α pathway.
Curcumin/pharmacology*
;
Humans
;
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism*
;
Animals
;
Lipid Metabolism/drug effects*
;
Carcinoma, Non-Small-Cell Lung/pathology*
;
Lung Neoplasms/pathology*
;
Mice, Nude
;
Down-Regulation
;
Mice
;
Cell Proliferation/drug effects*
;
Cell Line, Tumor
;
Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha
;
PPAR alpha/metabolism*
;
Signal Transduction/drug effects*
;
A549 Cells
5.Down-regulation of ACADM-mediated lipotoxicity inhibits invasion and metastasis of estrogen receptor-positive breast cancer cells.
Jiahao LI ; Ruiting XIAN ; Rong LI
Journal of Southern Medical University 2025;45(6):1163-1173
OBJECTIVES:
To investigate the effect of downregulation of medium-chain acyl-coenzyme A dehydrogenase (ACADM) on invasion and migration of estrogen receptor-positive breast cancer cells and the underlying mechanism.
METHODS:
The Kaplan-Meier Plotter database was used to analyze the ACADM expression levels in breast cancer and normal tissues and their association with patient prognosis. Human breast cancer MCF-7 and T47D cell lines with lentivirus-mediated ACADM knockdown were established, and their in situ tumor formation and metastasis after tail vein injection were evaluated in nude mice. The MCF-7 and T47D cells with ACADM knockdown and their unmodified parental cells were examined with oil-red O staining assay, ROS assay, mitochondrial respiratory chain function assay before and after treatments with ROS scavenger, Elamipretide (a cardiolipin oxidation inhibitor) or SC79 (an AKT activator), and the changes in migration and invasion abilities of the treated cells were analyzed with Transwell invasion assay and Boyden chamber assay. Western blotting was used to detect protein expression levels of related signaling pathways in the treated cells.
RESULTS:
ACADM overexpression was associated with a significantly shorter overall survival of breast cancer patients. In MCF-7 and T47D cells, ACADM knockdown resulted in downregulation of N calnexin, vimentin, p-P13K and p-AKT proteins, increased levels of free fatty acids and reactive oxygen species, lowered activities of mitochondrial respiratory chain complex III and V, and reduced mitochondrial inner phospholipids. ACADM knockdown significantly decreased the invasive capacity of the cells, which were obviously reversed by treatment with ROS scavenger, Elamipretide, and SC79.
CONCLUSIONS
Down-regulation of ACADM inhibits migration and invasion ability of estrogen receptor-positive breast cancer cells by lowering lipotoxicity and impairing mitochondrial function through the ROS/PI3K/AKT pathway.
Humans
;
Breast Neoplasms/metabolism*
;
Female
;
Mice, Nude
;
Down-Regulation
;
Neoplasm Invasiveness
;
Animals
;
Mice
;
Receptors, Estrogen/metabolism*
;
MCF-7 Cells
;
Cell Movement
;
Cell Line, Tumor
;
Reactive Oxygen Species/metabolism*
;
Acyl-CoA Dehydrogenase/genetics*
;
Signal Transduction
;
Neoplasm Metastasis
;
Proto-Oncogene Proteins c-akt/metabolism*
6.Danzhi Jiangtang Capsule improves renal vascular endothelial function in rats with diabetic nephropathy by downregulating the Notch1/NICD/MAML1 signaling pathway.
Sijia ZHU ; Jingcheng MA ; Yujiao ZHENG ; Chuanyun WU ; Jiangen ZHAO ; Lingxiu LI ; Li WANG ; Xuemei ZHOU
Journal of Southern Medical University 2025;45(10):2250-2257
OBJECTIVES:
To investigate the therapeutic mechanism of Danzhi Jiangtang Capsule (DZJTC) for repairing renal vascular endothelial injury in rats with diabetic nephropathy (DN).
METHODS:
Fifty male SD rat models of DN, established by left nephrectomy, high-sugar and high-fat diet and streptozotocin injection, were randomized into DN model group, low-, medium-, and high-dose DZJTC treatment groups, and DAPT (a γ-secretase inhibitor) treatment group, with 10 rats with normal feeding as the control group. DZJTC was administered by daily gavage at 0.315, 0.63, or 1.26 g/kg, and DAPT (20 mg/kg, dissolved in 50% CMC-Na solution) was given by gavage every other day for 4 weeks; normal saline was given in the control and model groups. After treatment, the levels of creatinine (CRE), blood urea nitrogen (BUN), and microalbuminuria (mALB) were detected with ELISA, and renal pathologies were observed by transmission electron microscopy. Renal expressions of vascular endothelial growth factor (VEGF) and endothelin-1 (ET-1) were measured by immunohistochemistry, and the protein expressions of CD31 and Notch signaling pathway components were detected using Western blotting.
RESULTS:
The rat models of DN showed significantly increased CRE, BUN, and mALB levels, obvious renal pathologies under electron microscopy, increased renal VEGF, ET-1 and CD31 expressions, and upregulated Notch1, NICD, and MAML1 protein levels. Treatment with DZJTC at the 3 doses and DAPT significantly reduced CRE, BUN, and mALB levels, improved renal pathology, decreased VEGF, ET-1 and CD31 expressions, and lowered Notch1, NICD and MAML1 levels, and the effects were the most pronounced with high-dose DZJTC.
CONCLUSIONS
DZJTC ameliorates hyperproliferation and dysfunction of renal vascular endothelium in DN rats possibly by regulating renal VEGF and ET-1 levels via inhibiting NICD- and MAML1-mediated Notch signaling pathway.
Animals
;
Male
;
Drugs, Chinese Herbal/therapeutic use*
;
Rats
;
Rats, Sprague-Dawley
;
Signal Transduction/drug effects*
;
Diabetic Nephropathies/drug therapy*
;
Receptor, Notch1/metabolism*
;
Kidney/blood supply*
;
Diabetes Mellitus, Experimental
;
Down-Regulation
;
Endothelium, Vascular/metabolism*
;
Nuclear Proteins/metabolism*
7.Lycium barbarum polysaccharides alleviates cisplatin-induced granulosa cell injury by downregulating miR-23a.
Liuqing LIU ; Kun WANG ; Xueqing WANG ; Bingxin DU
Journal of Southern Medical University 2025;45(11):2340-2349
OBJECTIVES:
To evaluate the protective effect of Lycium barbarum polysaccharides (LBP) against cisplatin-induced ovarian granulosa cell injury and investigate its possible mechanisms.
METHODS:
Human granulosa-like tumor cell line (KGN) were treated with 2.5 µg/mL cisplatin for 24 h, followed by treatment with 100, 500, and 1000 mg/L LBP, and the changes in cell viability, apoptosis, level of anti-Müllerian hormone (AMH), and cell ultrastructure were detected with CCK-8 assay, flow cytometry, ELISA and transmission electron microscopy. The cellular expressions of Bax, caspase-3, Bcl-2, and the PI3K/AKT pathway proteins were analyzed using Western blotting, and the expression of miR-23a was detected with RT-qPCR. KGN cell models with lentivirus-mediated miR-23a overexpression or knockdown were used to verify the therapeutic mechanism of LBP.
RESULTS:
Cisplatin treatment significantly inhibited cell viability, induced apoptosis, decreased AMH level, caused ultrastructural abnormalities, increased Bax and caspase-3 expression, and lowered Bcl-2 expression in KGN cells. Cisplatin also suppressed the activation of the PI3K/AKT signaling pathway and upregulated miR-23a expression in the cells. LBP intervention obviously alleviated cisplatin-induced injuries in KGN cells, and in particular, LBP treatment at the medium dose for 24 h significantly improved KGN cell viability, reduced apoptosis, enhanced their endocrine function, and ameliorated ultrastructural abnormalities. Mechanistically, medium-dose LBP obviously activated the PI3K/AKT pathway by downregulating miR-23a in cisplatin-treated cells, subsequently inhibiting Bax and caspase-3 while upregulating Bcl-2. Overexpression of miR-23a weakened while knockdown of miR-23a significantly enhanced the protective effects of LBP.
CONCLUSIONS
LBP alleviates cisplatin-induced apoptosis in KGN cells by inhibiting miR-23a expression and activating the PI3K/AKT pathway, suggesting a potential therapeutic strategy for ovarian function preservation.
Humans
;
Cisplatin/adverse effects*
;
MicroRNAs/genetics*
;
Female
;
Granulosa Cells/cytology*
;
Apoptosis/drug effects*
;
Drugs, Chinese Herbal/pharmacology*
;
Down-Regulation
;
Signal Transduction/drug effects*
;
Proto-Oncogene Proteins c-akt/metabolism*
;
Phosphatidylinositol 3-Kinases/metabolism*
;
Cell Line, Tumor
;
Cell Survival/drug effects*
8.Downregulation of Neuralized1 in the Hippocampal CA1 Through Reducing CPEB3 Ubiquitination Mediates Synaptic Plasticity Impairment and Cognitive Deficits in Neuropathic Pain.
Yan GAO ; Yiming QIAO ; Xueli WANG ; Manyi ZHU ; Lili YU ; Haozhuang YUAN ; Liren LI ; Nengwei HU ; Ji-Tian XU
Neuroscience Bulletin 2025;41(12):2233-2253
Neuropathic pain is frequently comorbidity with cognitive deficits. Neuralized1 (Neurl1)-mediated ubiquitination of CPEB3 in the hippocampus is critical in learning and memory. However, the role of Neurl1 in the cognitive impairment in neuropathic pain remains elusive. Herein, we found that lumbar 5 spinal nerve ligation (SNL) in male rat-induced neuropathic pain was followed by learning and memory deficits and LTP impairment in the hippocampus. The Neurl1 expression in the hippocampal CA1 was decreased after SNL. And this decrease paralleled the reduction of ubiquitinated-CPEB3 level and reduced production of GluA1 and GluA2. Overexpression of Neurl1 in the CA1 rescued cognitive deficits and LTP impairment, and reversed the reduction of ubiquitinated-CPEB3 level and the decrease of GluA1 and GluA2 production following SNL. Specific knockdown of Neurl1 or CPEB3 in bilateral hippocampal CA1 in naïve rats resulted in cognitive deficits and impairment of synaptic plasticity. The rescued cognitive function and synaptic plasticity by the treatment of overexpression of Neurl1 before SNL were counteracted by the knockdown of CPEB3 in the CA1. Collectively, the above results suggest that the downregulation of Neurl1 through reducing CPEB3 ubiquitination and, in turn, repressing GluA1 and GluA2 production and mediating synaptic plasticity impairment in hippocampal CA1 leads to the genesis of cognitive deficits in neuropathic pain.
Animals
;
Male
;
Neuralgia/metabolism*
;
Rats
;
Down-Regulation/physiology*
;
Ubiquitination/physiology*
;
Neuronal Plasticity/physiology*
;
Rats, Sprague-Dawley
;
CA1 Region, Hippocampal/metabolism*
;
Cognitive Dysfunction/metabolism*
;
RNA-Binding Proteins/metabolism*
;
Receptors, AMPA/metabolism*
9.TSZAF monomer combination downregulates the Wnt/β-catenin signaling pathway and inhibits neutrophil recruitment to prevent lung cancer metastasis.
Pan YU ; Jialiang YAO ; Long ZHANG ; Yanhong WANG ; Xinyi LU ; Jiajun LIU ; Zujun QUE ; Yao LIU ; Qian BA ; Jiwei LIU ; Yan WU ; Jianhui TIAN
Chinese Journal of Natural Medicines (English Ed.) 2025;23(9):1069-1079
Metastasis remains the primary cause of cancer-related mortality worldwide. Circulating tumor cells (CTCs) represent critical targets for metastasis prevention and treatment. Traditional Chinese medicine may prevent lung cancer metastasis through long-term intervention in CTC activity. Tiao-Shen-Zhi-Ai Formular (TSZAF) represents a Chinese medicine compound prescription utilized clinically for lung cancer treatment. This study combined three principal active ingredients from TSZAF into a novel TSZAF monomer combination (TSZAF mc) to investigate its anti-metastatic effects and mechanisms. TSZAF mc demonstrated significant inhibition of proliferation, migration, and invasion in CTC-TJH-01 and LLC cells, while inducing cellular apoptosis in vitro. Moreover, TSZAF mc substantially inhibited LLC cell growth and metastasis in vivo. Mechanistically, TAZSF mc significantly suppressed the Wnt/β-catenin signaling pathway and CXCL5 expression in lung cancer cells and tissues. Additionally, TAZSF mc notably reduced neutrophil infiltration in metastatic lesions. These findings indicate that TSZAF mc inhibits lung cancer growth and metastasis by suppressing the Wnt/β-catenin signaling pathway and reducing CXCL5 secretion, thereby decreasing neutrophil recruitment and infiltration. TSZAF mc demonstrates potential as an effective therapeutic agent for lung cancer metastasis.
Lung Neoplasms/genetics*
;
Wnt Signaling Pathway/drug effects*
;
Animals
;
Humans
;
Drugs, Chinese Herbal/pharmacology*
;
Mice
;
Neoplasm Metastasis/prevention & control*
;
Cell Proliferation/drug effects*
;
Cell Line, Tumor
;
Neutrophil Infiltration/drug effects*
;
Down-Regulation/drug effects*
;
Cell Movement/drug effects*
;
beta Catenin/genetics*
;
Apoptosis/drug effects*
;
Mice, Inbred C57BL
;
Male
;
Neoplastic Cells, Circulating/drug effects*
10.Morin, a matrix metalloproteinase 9 inhibitor, attenuates endothelial-to-mesenchymal transition in atherosclerosis by downregulating Notch-1 signaling.
Yuan HE ; Xiao-Xuan QIN ; Ming-Wei LIU ; Wei SUN
Journal of Integrative Medicine 2024;22(6):683-695
OBJECTIVE:
Atherosclerotic cardiovascular disease poses a significant health challenge globally. Recent findings highlight the pivotal role of the endothelial-to-mesenchymal transition (EndMT) in atherosclerosis. Morin is a bioflavonoid mainly extracted from white mulberry, a traditional Chinese herbal medicine with anti-inflammatory and antioxidant properties. This study examines whether morin can alleviate atherosclerosis by suppressing EndMT and seeks to elucidate the underlying mechanism.
METHODS:
We induced an in vitro EndMT model in human umbilical vein endothelial cells (HUVECs) by stimulating the cells with transforming growth factor-β1 (TGF-β1) (10 ng/mL) for 48 h. The in vivo experiments were performed in an atherosclerosis model using apolipoprotein E (ApoE)-/- mice fed with a high-fat diet (HFD). Mice in the intervention group were given morin (50 mg/kg) orally for 4 weeks. Molecular docking and microscale thermophoresis were assayed to understand the interactions between morin and matrix metalloproteinase-9 (MMP-9).
RESULTS:
Morin inhibited the expression of EndMT markers in a dose-dependent manner in TGF-β1-treated HUVECs. Administering 50 μmol/L morin suppressed the upregulation of MMP-9 and Notch-1 signaling in TGF-β1-induced EndMT. Moreover, the overexpression of MMP-9 activated Notch-1 signaling, thereby reversing morin's inhibitory effect on EndMT. In the HFD-induced atherosclerotic ApoE-/- mice, morin notably reduced aortic intimal hyperplasia and plaque formation by suppressing EndMT. Furthermore, morin demonstrated a strong binding affinity for MMP-9.
CONCLUSION
Morin acts as an MMP-9 inhibitor to disrupt EndMT in atherosclerosis by limiting the activation of Notch-1 signaling. This study underscores morin's potential utility in the development of anti-atherosclerotic medication. Please cite this article as: He Y, Qin XX, Liu MW, Sun W. Morin, a matrix metalloproteinase 9 inhibitor, attenuates endothelial-to-mesenchymal transition in atherosclerosis by downregulating Notch-1 Signaling. J Integr Med. 2024; 22(6): 684-696.
Flavonoids/pharmacology*
;
Animals
;
Atherosclerosis/metabolism*
;
Humans
;
Receptor, Notch1/genetics*
;
Signal Transduction/drug effects*
;
Matrix Metalloproteinase 9/genetics*
;
Human Umbilical Vein Endothelial Cells/drug effects*
;
Mice
;
Epithelial-Mesenchymal Transition/drug effects*
;
Down-Regulation/drug effects*
;
Male
;
Transforming Growth Factor beta1/genetics*
;
Matrix Metalloproteinase Inhibitors/pharmacology*
;
Mice, Inbred C57BL
;
Flavones

Result Analysis
Print
Save
E-mail