Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
J Cell Mol Med ; 28(19): e70132, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39350724

ABSTRACT

Aging is a risk factor for various human disorders, including cancer. Current literature advocates that the primary principles of aging depend on the endogenous stress-induced DNA damage caused by reactive oxygen species 50 Hz low-frequency magnetic field was suggested to induce DNA damage and chromosomal instability. NF-kB, activated by DNA damage, is upregulated in age-related cancers and inhibition of NF-kB results in aging-related delayed pathologies. Metformin (Met), an NF-kB inhibitor, significantly reduces both NF-kB activation and expression in aging and cancer. This in vitro study, therefore, was set out to assess the effects of 5mT MF in 50 Hz frequency and Met treatment on the viability and proliferation of aged mouse NIH/3T3 fibroblasts and expression of RELA/p65, matrix metalloproteinases MMP2 and MMP9, and E-cadherin (CDH1) genes. The trypan blue exclusion assay was used to determine cell viability and the BrdU incorporation assay to determine cell proliferation. The MMP-2/9 protein analysis was carried out by immunocytochemistry, NF-kB activity by ELISA and the expressions of targeted genes by qRT-PCR methods. Four doses of Met (500 uM, 1 mM, 2 mM and 10 mM) suppressed both the proliferation and viability of fibroblasts exposed to the MF in a dose-dependent pattern, and the peak inhibition was recorded at the 10 mM dose. Met reduced the expression of NF-kB, and MMP2/9, elevated CDH1 expression and suppressed NF-kB activity. These findings suggest that Met treatment suppresses the carcinogenic potential of 50 Hz MFs in aged mouse fibroblasts, possibly through modulation of NF-kB activation and epithelial-mesenchymal transition modulation.


Subject(s)
Cell Proliferation , Cell Survival , Fibroblasts , Magnetic Fields , Metformin , NF-kappa B , Animals , Metformin/pharmacology , Mice , Fibroblasts/metabolism , Fibroblasts/drug effects , NIH 3T3 Cells , NF-kappa B/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/pathology , Transcription Factor RelA/metabolism , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 2/genetics , Cadherins/metabolism , Cadherins/genetics , Cellular Senescence/drug effects
2.
BMC Cardiovasc Disord ; 24(1): 354, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38992615

ABSTRACT

BACKGROUND: Hyperlipidemia damages vascular wall and serves as a foundation for diseases such as atherosclerosis, hypertension and stiffness. The NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome is implicated in vascular dysfunction associated with hyperlipidemia-induced vascular injury. Sodium tanshinone IIA sulfonate (STS), a well-established cardiovascular protective drug with recognized anti-inflammatory, antioxidant, and vasodilatory properties, is yet to be thoroughly investigated for its impact on vascular relaxant imbalance induced by hyperlipidemia. METHODS: In this study, we treated ApoE-knockout (ApoE-/-) mouse with STS and assessed the activation of the NLRP3 inflammasome, expression of MMP2/9, integrity of elastic fibers, and vascular constriction and relaxation. RESULTS: Our findings reveal that STS intervention effectively preserves elastic fibers, significantly restores aortic relaxation function in ApoE-/- mice, and reduces their excessive constriction. Furthermore, STS inhibits the phosphorylation of spleen tyrosine kinase (SYK), suppresses NLRP3 inflammasome activation, and reduces MMP2/9 expression. CONCLUSIONS: These results demonstrate that STS protects vascular relaxation against hyperlipidemia-induced damage through modulation of the SYK-NLRP3 inflammasome-MMP2/9 pathway. This research provides novel insights into the mechanisms underlying vascular relaxation impairment in a hyperlipidemic environment and uncovers a unique mechanism by which STS preserves vascular relaxation, offering valuable foundational research evidence for its clinical application in promoting vascular health.


Subject(s)
Disease Models, Animal , Inflammasomes , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 9 , Mice, Inbred C57BL , Mice, Knockout, ApoE , NLR Family, Pyrin Domain-Containing 3 Protein , Phenanthrenes , Signal Transduction , Syk Kinase , Vasodilation , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes/metabolism , Syk Kinase/metabolism , Matrix Metalloproteinase 2/metabolism , Phenanthrenes/pharmacology , Male , Matrix Metalloproteinase 9/metabolism , Vasodilation/drug effects , Hyperlipidemias/drug therapy , Hyperlipidemias/physiopathology , Vasodilator Agents/pharmacology , Phosphorylation , Mice , Aorta/drug effects , Aorta/physiopathology , Aorta/metabolism , Aorta/enzymology , Apolipoproteins E
3.
Pharmacol Res ; 194: 106846, 2023 08.
Article in English | MEDLINE | ID: mdl-37414199

ABSTRACT

Malignant proliferation and metastasis are the main causes of breast cancer death. The transcription factor high mobility group (HMG) box-containing protein 1 (HBP1) is an important tumor suppressor whose deletion or mutation is closely related to the appearance of tumors. Here, we investigated the role of HBP1 in breast cancer suppression. HBP1 enhances the activity of the tissue inhibitors of metalloproteinases 3 (TIMP3) promoter, thereby increasing protein and mRNA levels of TIMP3. TIMP3 increases the phosphatase and tensin homolog (PTEN) protein level by inhibiting its degradation and acts as a metalloproteinase inhibitor to inhibit the protein levels of MMP2/9. In this study, we demonstrated that the HBP1/TIMP3 axis plays a crucial role in inhibiting the tumorigenesis of breast cancer. HBP1 deletion interferes with the regulation of the axis and induces the occurrence and malignant progression of breast cancer. In addition, the HBP1/TIMP3 axis promotes the sensitivity of breast cancer to radiation therapy and hormone therapy. Our study opens new perspectives on the treatment and prognosis of breast cancer.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , RNA, Messenger/genetics , Prognosis , Promoter Regions, Genetic , High Mobility Group Proteins/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tissue Inhibitor of Metalloproteinase-3/genetics , Tissue Inhibitor of Metalloproteinase-3/metabolism
4.
Eur J Appl Physiol ; 2023 Dec 03.
Article in English | MEDLINE | ID: mdl-38043087

ABSTRACT

OBJECTIVES: The objectives of this study were to assess the effect of 8 weeks of moderate-intensity aerobic training on permeability inflammatory indicators of matrix metalloproteinases (MMPs) and specific tissue inhibitors of MMPs in female migraineurs. METHODS: Female migraineurs (n = 28, age 32 ± 6) were randomised into two groups: migraine with exercise training (EXE + Mig, n = 13) and migraine without exercise training (NON-EXE + Mig, n = 15). Matched healthy women were also recruited as a healthy control group (CON, n = 15). The EXE-Mig group performed 8 weeks of aerobic training. Pre and post intervention, serum matrix metalloproteinases (MMP-2 and 9) and specific tissue inhibitors of MMPs (TIMP-1 and 2) were measured. In addition, body composition indices and VO2max were determined. RESULTS: Exercise training reduced serum MMP-9 in female migraineurs with between-group changes and a time x group interaction (p < 0.05). In addition, exercise training reduced the serum MMP-9/TIMP-1 ratio in female migraineurs with between-group changes and time x group interaction (p < 0.05). However, no training-induced effect was observed in serum TIMP-1, TIMP-2, MMP-2 contents (p > 0.05) and MMP-2/TIMP-2 ratio (p > 0.05). Finally, exercise training reduced body fat content, WHR and BMI, and improved VO2max (p < 0.01). CONCLUSIONS: Our results demonstrated beneficial effects of aerobic exercise training on some circulatory inflammation factors (MMP9, MMP-9/TIMP-1) and some health indicators in female migraineurs, suggesting that such training can be employed as a non-pharmacological therapeutic method.

5.
Int J Mol Sci ; 24(6)2023 Mar 08.
Article in English | MEDLINE | ID: mdl-36982278

ABSTRACT

Metabolic disorders characterized by elevated blood glucose levels are a recognized risk factor for hepatocellular carcinoma (HCC). Lipid dysregulation is critically involved in the HCC progression, regulating energy storage, metabolism, and cell signaling. There is a clear link between de novo lipogenesis in the liver and activation of the NF-κB pathway, which is involved in cancer metastasis via regulation of metalloproteinases MMP-2/9. As conventional therapies for HCC reach their limits, new effective and safe drugs need to be found for the prevention and/or adjuvant therapy of HCC. The marine plant Posidonia oceanica (L.) Delile is endemic to the Mediterranean and has traditionally been used to treat diabetes and other health disorders. The phenol-rich leaf extract of Posidonia oceanica (POE) is known to have cell-safe bioactivities. Here, high glucose (HG) conditions were used to study lipid accumulation and fatty acid synthase (FASN) expression in human HepG2 hepatoma cells using Oil Red O and Western blot assays. Under HG conditions, the activation status of MAPKs/NF-κB axis and MMP-2/9 activity were determined by Western blot and gelatin zymography assays. The potential ameliorative role of POE against HG-related stress in HepG2 cells was then investigated. POE reduced lipid accumulation and FASN expression with an impact on de novo lipogenesis. Moreover, POE inhibited the MAPKs/NF-κB axis and, consequently, MMP-2/9 activity. Overall, these results suggest that P. oceanica may be a potential weapon in the HCC additional treatment.


Subject(s)
Alismatales , Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Hep G2 Cells , Matrix Metalloproteinase 2 , NF-kappa B , Glucose , Lipids
6.
BMC Cancer ; 22(1): 778, 2022 Jul 16.
Article in English | MEDLINE | ID: mdl-35840921

ABSTRACT

A major challenge in the treatment of liver cancer is that a large proportion of patients fail to achieve long-term disease control, with death from liver cancer cell migration and invasion. Acid-sensitive ion channel 1α (ASIC1α) is involved in the migration, invasion, and proliferation of liver cancer cells. Therefore, we explored the mechanism of ASIC1α-mediated liver cancer cell migration and invasion. We determined the levels of ASIC1α by western blotting and immunofluorescence in HepG2 and SK-Hep1 cells cultured in various acidic conditions. In addition, wound healing assay, transwell invasion assay, and MTT assay were conducted to assess the migration, invasion, and proliferation abilities of liver cancer cells. Western blotting was conducted to determine the levels of MMP2, MMP9, ASIC1α, p-PI3Kp85, t-PI3Kp85, p-AKT(Ser473), t-AKT, p-mTOR (Ser2448), t-mTOR. We first found that the levels of ASIC1α in the HepG2 and SK-Hep1 cells in acidic conditions (pH 6.5) were significantly increased. Inhibition and knockdown of ASIC1α down-regulated MMP-2/9 expression and inhibited the migration, invasion, and proliferation of HepG2 and SK-Hep1 cells; overexpression of ASIC1α had the opposite effect. We further demonstrated that ASIC1α up-regulates MMP-2/9 via activation of the PI3K/AKT/mTOR pathway, thereby promoting migration, invasion, and proliferation of liver cancer cells. Overexpression of MMP-2/9 and activation of AKT reversed these effects on liver cancer cells caused by inhibition of ASIC1α. We conclude that ASIC1α can regulate migration, invasion, and proliferation of liver cancer cells through the MMP-2/9/PI3K/AKT/mTOR pathway. These observations may provide a new reference for liver cancer chemotherapy.


Subject(s)
Acid Sensing Ion Channels , Liver Neoplasms , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 9 , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , TOR Serine-Threonine Kinases , Acid Sensing Ion Channels/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
7.
Cell Mol Neurobiol ; 42(8): 2715-2725, 2022 Nov.
Article in English | MEDLINE | ID: mdl-34345995

ABSTRACT

Prevention of the nuclear translocation of ANXA1 with Tat-NTS was recently reported to alleviate neuronal injury and protect against cerebral stroke. However, the role that Tat-NTS plays in the occurrence and development of gliomas still needs to be elucidated. Therefore, human glioblastoma (GB) cells were treated with various concentrations of Tat-NTS for 24 h, and cell proliferation, migration and invasion were assessed with CCK-8 and Transwell assays. The nuclear translocation of ANXA1 was evaluated by subcellular extraction and immunofluorescence, and protein expression levels were detected by Western blot analysis. In addition, the activity of MMP-2/9 was measured by gelatin zymography. The results revealed that Tat-NTS significantly inhibited the nuclear translocation of ANXA1 in U87 cells and inhibited the proliferation, migration and invasion of GB cells. Tat-NTS also suppressed cell cycle regulatory proteins and MMP-2/-9 activity and expression. Moreover, Tat-NTS reduced the level of p-p65 NF-κB in U87 cells. These results suggest that the Tat-NTS-induced inhibition of GB cell proliferation, migration and invasion is closely associated with the induction of cell cycle arrest, downregulation of MMP-2/-9 expression and activity and suppression of the NF-κB signaling pathway. Thus, Tat-NTS may be a potential chemotherapeutic agent for the treatment of GB.


Subject(s)
Annexin A1 , Glioblastoma , Annexin A1/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gelatin , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Matrix Metalloproteinase 2/metabolism , NF-kappa B/metabolism , Neoplasm Invasiveness , Sincalide/metabolism
8.
Pharmacol Res ; 176: 106051, 2022 02.
Article in English | MEDLINE | ID: mdl-34973467

ABSTRACT

Aortic dissection (AD) is a disease with high mortality and lacks effective drug treatment. Recent studies have shown that the development of AD is closely related to glucose metabolism. Lactate dehydrogenase A (LDHA) is a key glycolytic enzyme and plays an important role in cardiovascular disease. However, the role of LDHA in the progression of AD remains to be elucidated. Here, we found that the level of LDHA was significantly elevated in AD patients and the mouse model established by BAPN combined with Ang II. In vitro, the knockdown of LDHA reduced the growth of human aortic vascular smooth muscle cells (HAVSMCs), glucose consumption, and lactate production induced by PDGF-BB. The overexpression of LDHA in HAVSMCs promoted the transformation of HAVSMCs from contractile phenotype to synthetic phenotype, and increased the expression of MMP2/9. Mechanistically, LDHA promoted MMP2/9 expression through the LDHA-NDRG3-ERK1/2-MMP2/9 pathway. In vivo, Oxamate, LDH and lactate inhibitor, reduced the degradation of elastic fibers and collagen deposition, inhibited the phenotypic transformation of HAVSMCs from contractile phenotype to synthetic phenotype, reduced the expression of NDRG3, p-ERK1/2, and MMP2/9, and delayed the progression of AD. To sum up, the increase of LDHA promotes the production of MMP2/9, stimulates the degradation of extracellular matrix (ECM), and promoted the transformation of HAVSMCs from contractile phenotype to synthetic phenotype. Oxamate reduced the progression of AD in mice. LDHA may be a therapeutic target for AD.


Subject(s)
Aortic Dissection/drug therapy , Lactate Dehydrogenase 5/antagonists & inhibitors , Oxamic Acid/therapeutic use , Adult , Aged , Aortic Dissection/metabolism , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Female , Glucose/metabolism , Humans , Lactate Dehydrogenase 5/genetics , Lactate Dehydrogenase 5/metabolism , Lactic Acid/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , Middle Aged , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Oxamic Acid/pharmacology
9.
Biotechnol Appl Biochem ; 69(5): 1929-1941, 2022 Oct.
Article in English | MEDLINE | ID: mdl-34554598

ABSTRACT

Human papillomavirus (HPV) infection-dependent cervical cancer is one of the most common gynecological cancers and often becomes aggressive, with rapid proliferation, invasion/migration, and drug resistance. Here, 135 fresh human cervical squamous cell carcinoma (CSCC) tissue specimens, comprising 21 adjacent normal (AN), 30 cervical intraepithelial neoplasia (CIN1-3 ), 45 CSCC, and 39 drugs (chemo-radiation)-resistant cervical tumor (DRCT) tissues were included. HPV-positive (HeLa, SiHa), HPV-negative (C33A), and cisplatin-resistant (CisR-HeLa/-SiHa/-C33A) cell lines were used for in vitro studies. HPV16/18 oncoproteins E6/E7, pERK1/2, and glycogen synthase kinase-3 (GSK3) and the matrix metalloproteinases (MMPs) MMP-9/-2 were assessed using immunohistochemistry, WB, and gelatin zymography. HPV16/18 infection was observed in 16.7% of the CIN1-3 , 77.8% of the CSCC, and 89.7% of DRCT samples. Total and inactive GSK3ß expressions were associated with overall CSCC progression (p = 0.039 and p = 0.024, respectively) and chemoresistance (p = 0.004 and p = 0.014, respectively). Positive correlations were observed, between the expression of E6 and pGSK3ß expression (p = 0.013); E6 and CSCC progression (p < 0.0001)/drug resistance (p = 0.0001). CisR-HeLa/-SiHa was more dependent on pGSK3ß, and activation of GSK3 by SMIs (iAkt), treatment with nimbolide, or knockdown of E6/E7 reduced cisplatin resistance and promoted apoptosis. Hence, the activation of GSK3ß with nimbolide and iAkt can be exploited for therapeutic interventions of cervical cancer.


Subject(s)
Oncogene Proteins, Viral , Papillomavirus Infections , Uterine Cervical Neoplasms , Female , Humans , Uterine Cervical Neoplasms/metabolism , Papillomavirus Infections/drug therapy , Human papillomavirus 16/genetics , Human papillomavirus 16/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Cisplatin/metabolism , Glycogen Synthase Kinase 3/metabolism , Human papillomavirus 18/metabolism , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/metabolism , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Matrix Metalloproteinase 2/metabolism , Drug Resistance , Cell Line, Tumor
10.
Neurochem Res ; 46(9): 2297-2306, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34086144

ABSTRACT

As a serious trauma of the neurological system, spinal cord injury (SCI) results in permanent disability, gives rise to immediate vascular damage and a wide range of matters that induce the breakage of blood spinal cord barrier (BSCB). SCI activates the expression of MMP-2/9, which are considered to accelerate the disruption of BSCB. Recent research shows that Dl-3-n-butylphthalide (NBP) exerted protective effects on blood spinal cord barrier in animals after SCI, but the underlying molecular mechanism of NBP on the BSCB undergoing SCI is unknown. Here, our research show that NBP inhibited the expression of MMP-2/9, then improved the permeability of BSCB following SCI. After the T9 level of spinal cord performed with a moderate injury, NBP was managed by intragastric administration and further performed once a day. NBP remarkably improved the permeability of BSCB and junction proteins degration, then promoted locomotion recovery. The protective effect of NBP on BSCB destruction is related to the regulation of MMP-2/9 induced by SCI. Moreover, NBP obviously inhibited the MMP-2/9 expression and junction proteins degradation in microvascular endothelial cells. In conclusion, our results indicate that MMP-2/9 are relevant to the breakdown of BSCB, NBP impairs BSCB destruction through inhibiting MMP-2/9 and promotes functional recovery subjected to SCI. NBP is likely to become a new nominee as a therapeutic to treat SCI via a transigent BSCB.


Subject(s)
Benzofurans/therapeutic use , Matrix Metalloproteinase Inhibitors/therapeutic use , Neuroprotective Agents/therapeutic use , Spinal Cord Injuries/drug therapy , Tight Junction Proteins/metabolism , Tight Junctions/drug effects , Animals , Cell Hypoxia/drug effects , Claudin-5/metabolism , Female , Glucose/deficiency , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Occludin/metabolism , Oxygen/metabolism , Rats, Sprague-Dawley , Recovery of Function/drug effects , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord Injuries/enzymology
11.
J Nanobiotechnology ; 19(1): 68, 2021 Mar 04.
Article in English | MEDLINE | ID: mdl-33663548

ABSTRACT

It was shown that some nanomaterials may have anticancer properties, but lack of selectivity is one of challenges, let alone selective suppression of cancer growth by regulating the cellular microenvironment. Herein, we demonstrated for the first time that carbon quantum dots/Cu2O composite (CQDs/Cu2O) selectively inhibited ovarian cancer SKOV3 cells by targeting cellular microenvironment, such as matrix metalloproteinases, angiogenic cytokines and cytoskeleton. The result was showed CQDs/Cu2O possessed anticancer properties against SKOV3 cells with IC50 = 0.85 µg mL-1, which was approximately threefold lower than other tested cancer cells and approximately 12-fold lower than normal cells. Compared with popular anticancer drugs, the IC50 of CQDs/Cu2O was approximately 114-fold and 75-fold lower than the IC50 of commercial artesunate (ART) and oxaliplatin (OXA). Furthermore, CQDs/Cu2O possessed the ability to decrease the expression of MMP-2/9 and induced alterations in the cytoskeleton of SKOV3 cells by disruption of F-actin. It also exhibited stronger antiangiogenic effects than commercial antiangiogenic inhibitor (SU5416) through down-regulating the expression of VEGFR2. In addition, CQDs/Cu2O has a vital function on transcriptional regulation of multiple genes in SKOV3 cells, where 495 genes were up-regulated and 756 genes were down-regulated. It is worth noting that CQDs/Cu2O also regulated angiogenesis-related genes in SKOV3 cells, such as Maspin and TSP1 gene, to suppress angiogenesis. Therefore, CQDs/Cu2O selectively mediated of ovarian cancer SKOV3 cells death mainly through decreasing the expression of MMP-2, MMP-9, F-actin, and VEGFR2, meanwhile CQDs/Cu2O caused apoptosis of SKOV3 via S phase cell cycle arrest. These findings reveal a new application for the use of CQDs/Cu2O composite as potential therapeutic interventions in ovarian cancer SKOV3 cells.


Subject(s)
Carbon/pharmacology , Cell Death/drug effects , Cytokines/metabolism , Cytoskeleton/metabolism , Matrix Metalloproteinases/metabolism , Nanocomposites/chemistry , Ovarian Neoplasms/drug therapy , Quantum Dots/chemistry , Angiogenesis Inducing Agents , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Copper/chemistry , Copper/pharmacology , Down-Regulation/drug effects , Female , Humans , Matrix Metalloproteinase 9/metabolism , Mice , Vascular Endothelial Growth Factor Receptor-2/metabolism
12.
Biochem Cell Biol ; 98(5): 575-582, 2020 10.
Article in English | MEDLINE | ID: mdl-32298598

ABSTRACT

MicroRNA (miR)-518-3p has been shown to function as a tumor suppressor. This study was conducted to investigate the effects of miR-518-3p in colorectal cancer (CRC). The miR-518-3p mimic, mimic negative control (NC), miR-518-3p inhibitor, inhibitor-NC, ShRNA-TRIP4, and ShRNA-NC vectors were transfected into SW480 cells using Lipofectamine 2000. Cell viability was detected using CCK-8. Colony formation, cell invasiveness, and cell migration were assessed by plate colony formation, Transwell assays, and wound healing assays, respectively. Relative mRNA and protein levels were detected using RT-qPCR and Western blot, respectively. The target gene thyroid hormone receptor interactor 4 (TRIP4) of miR-518-3p was identified and further verified using dual-luciferase reporter assay. Compared with normal tissues, levels of miR-518-3p were decreased and TRIP4 was significantly increased in the tissues from patients with CRC. Following transfection with a miR-518-3p mimic or ShRNA-TRIP4, cell viability decreased in a time-dependent manner, and colony formation rate, wound closure rate, and the number of invasive cells were much lower for the transfected cells than in the corresponding NC and control groups. miR-518-3p overexpression or silencing of TRIP4 significantly down-regulated the expression of MMP-2 and MMP-9. Knockdown of miR-518-3p had the opposite effects, and TRIP4 was identified as a target of miR-518-3p. The inhibitory effects of miR-518-3p on the progressions of CRC are associated with TRIP4.


Subject(s)
Colorectal Neoplasms/metabolism , MicroRNAs/metabolism , Transcription Factors/metabolism , Cell Movement , Cell Proliferation , Cell Survival , Cells, Cultured , Colorectal Neoplasms/pathology , Humans , MicroRNAs/genetics , Transcription Factors/genetics
13.
Bioorg Chem ; 101: 103992, 2020 08.
Article in English | MEDLINE | ID: mdl-32554279

ABSTRACT

Thiazole derivatives are known to possess various biological activities such as antiparasitic, antifungal, antimicrobial and antiproliferative activities. Matrix metalloproteinases (MMPs) are important protease target involved in tumor progression including angiogenesis, tissue invasion, and migration. Therefore, MMPs have also been reported as potential diagnostic and prognostic biomarkers in many types of cancer. Herein, new aryl thiazoles were synthesized and evaluated for their anticancer effects on a panel of cancer cell lines including the invasive MDA-MB-231 line. Some of these compounds showed IC50 values in the submicromolar range in anti-proliferative assays. In order to examine the relationship between their anticancer activity and MMPs targets, the compounds were evaluated for their inhibitory effects on MMP-2 and 9. That data obtained revealed that most of these compounds were potent dual MMP-2/9 inhibitors at nanomolar concentrations. Among these, 2-(1-(2-(2-((E)-4-iodobenzylidene)hydrazineyl)-4-methylthiazol-5-yl)ethylidene)hydrazine-1-carboximidamide (4a) was the most potent non-selective dual MMP-2/9 inhibitor with inhibitory concentrations of 56 and 38 nM respectively. When compound 4a was tested in an MDA-MB-231, HCT-116, MCF-7 model, it effectively inhibited tumor growth, strongly induced cancer cell apoptosis, inhibit cell migration, and suppressed cell cycle progression leading to DNA fragmentation. Taken together, the results of our studies indicate that the newly discovered thiazole-based MMP-2/9 inhibitors have significant potential for anticancer treatment.


Subject(s)
Cell Cycle Checkpoints/drug effects , DNA Fragmentation/drug effects , Drug Discovery , Matrix Metalloproteinase 2/drug effects , Matrix Metalloproteinase 9/drug effects , Protease Inhibitors/pharmacology , Triazoles/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Humans , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacokinetics , Structure-Activity Relationship , Wound Healing/drug effects
14.
Int J Mol Sci ; 21(4)2020 Feb 14.
Article in English | MEDLINE | ID: mdl-32074974

ABSTRACT

In this study, we aimed to elucidate the anti-invasive effects of Cudrania tricuspidata root-gold nanoparticles (CTR-GNPs) using glioblastoma cells. We demonstrated the rapid synthesis of CTR-GNPs using UV-vis spectra. The surface morphology, crystallinity, reduction, capsulation, and stabilization of CTR-GNPs were analyzed using high resolution transmission electron microscopy (HR-TEM), energy dispersive spectroscopy (EDS), X-ray diffraction (XRD), and Fourier-transform infrared spectroscopy (FT-IR). Furthermore, CTR-GNPs displayed excellent photocatalytic activity as shown by the photo-degradation of methylene blue and rhodamine B. Cell migration and invasion assays with human glioblastoma cells were performed to investigate the anti-invasive effect of CTR-GNPs on U87 cells that were treated with phorbol 12-myristate 13-acetate. The results show that CTR-GNPs can significantly inhibit both basal and phorbol 12-myristate 13-acetate (PMA)-induced migration and invasion ability. Importantly, treatment with CTR-GNPs significantly decreased the levels of metalloproteinase (MMP)-2/-9 and phospholipase D1 (PLD1) and protein but not PLD2, which is involved in the modulation of migration and the invasion of glioblastoma cells. These results present a novel mechanism showing that CTR-GNPs can attenuate the migration and invasion of glioblastoma cells induced by PMA through transcriptional and translational regulation of MMP-2/-9 and PLD1. Taken together, our results suggest that CTR-GNPs might be an excellent therapeutic alternative for wide range of glioblastomas.


Subject(s)
Down-Regulation/drug effects , Gold/chemistry , Metal Nanoparticles/toxicity , Moraceae/chemistry , Plant Extracts/chemistry , Cell Line, Tumor , Cell Movement/drug effects , Glioblastoma/metabolism , Glioblastoma/pathology , Green Chemistry Technology , Humans , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Metal Nanoparticles/chemistry , Moraceae/metabolism , Phospholipase D/genetics , Phospholipase D/metabolism , Plant Roots/chemistry , Tetradecanoylphorbol Acetate/pharmacology
15.
Pharm Biol ; 58(1): 231-238, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32202448

ABSTRACT

Context: The relationship between resveratrol and histone acetylation in renal cell carcinoma (RCC) has not yet been reported.Objective: To explore the functional role of resveratrol in RCC.Materials and methods: Functional experiments were performed to determine proliferatio n of ACHN cells with treatment of resveratrol (0, 7.8125, 15.625, 31.25 and 62.5 µg/mL, for 12, 24 and 48 h of culture) or 0.1 µM SAHA. The enzyme activities of MMP-2/-9 were measured by gelatine zymography and histone acetylation by Western blot.Results: When the cells were treated with 15.625, 31.25 and 62.5 µg/mL resveratrol, ACHN cells viability was 73.2 ± 3.5%, 61.4 ± 3.1%, 50.2 ± 4.7% for 12 h, 62.7 ± 4.5%, 52.4 ± 5.5%, 40.2 ± 3.8% for 24 h, and 60.8 ± 3.7%, 39.4 ± 5.1%, 37.6 ± 2.7% for 48 h, and the wound closure (%) of migration was increased from 0.6 to 0.7, 0.85, 0.9 for 12 h and from 0.23 to 0.3, 0.48, 0.59 for 24 h. The invasion rate was 8.5 ± 0.9%, 7.4 ± 0.3% and 5.8 ± 0.6%, and cell cycle was arrested at G1 from 42.5 ± 2.9% to 55.3 ± 5.7%, 59.8 ± 3.4%, 68.7 ± 4.6%. MMP-2/-9 expression (p < 0.05) was inhibited by resveratrol. The protein levels of histone acetylation (p < 0.01) was increased by resveratrol.Discussion and conclusions: Our results suggest that these effects might be related to a high level of histone acetylation, and resveratrol can be considered as an alternative treatment for RCC.


Subject(s)
Carcinoma, Renal Cell/metabolism , Cell Movement/drug effects , Histones/metabolism , Kidney Neoplasms/metabolism , Resveratrol/pharmacology , Acetylation , Carcinoma, Renal Cell/pathology , Cell Cycle , Cell Line, Tumor , Cell Survival/drug effects , Humans , Kidney Neoplasms/pathology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism
16.
J Cell Biochem ; 120(3): 3611-3620, 2019 03.
Article in English | MEDLINE | ID: mdl-30417585

ABSTRACT

Overexpression of leucine aminopeptidase 3 (LAP3) is involved in proliferation, migration, and invasion of several tumor cells and plays a crucial role in tumor metastasis. However, the related mechanism remains unknown. In this study, we used MDA-MB-231 and MCF7 breast cancer cell lines to explore the role of LAP3 in the regulation of cancer cell migration and invasion by employing the natural LAP3 inhibitor bestatin and a lentivirus vector that overexpresses or knocks down LAP3. Bestatin inhibited tumor cell migration and invasion in a dose-dependent manner. Western blot assay showed that bestatin and knockdown of LAP3 upregulated phosphorylation of Hsp27 and downregulated expression of fascin. Phosphorylation of Akt and expression of matrix metalloproteinase-2/9 can also be downregulated. LAP3 overexpression showed the opposite results. Immunohistochemistry analysis was conducted to detect expression levels of LAP3 in breast cancer tissues. High LAP3 expression was correlated with the grade of malignancy. Findings of this study uncovered the molecular mechanism of LAP3 on breast cancer metastasis and indicated that LAP3 may act as a potential antimetastasis therapeutic target.


Subject(s)
Breast Neoplasms/metabolism , Carrier Proteins/blood , Cell Movement , Gene Expression Regulation, Neoplastic , Leucyl Aminopeptidase/metabolism , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Microfilament Proteins/blood , Neoplasm Proteins/metabolism , Up-Regulation , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carrier Proteins/genetics , Female , Humans , Leucyl Aminopeptidase/genetics , MCF-7 Cells , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Microfilament Proteins/genetics , Neoplasm Invasiveness , Neoplasm Proteins/genetics
17.
J Nanobiotechnology ; 17(1): 2, 2019 Jan 07.
Article in English | MEDLINE | ID: mdl-30616599

ABSTRACT

BACKGROUND: Previous studies have demonstrated that exposure to nickel nanoparticles (Nano-Ni) causes oxidative stress and severe, persistent lung inflammation, which are strongly associated with pulmonary toxicity. However, few studies have investigated whether surface modification of Nano-Ni could alter Nano-Ni-induced lung injury, inflammation, and fibrosis in vivo. Here, we propose that alteration of physicochemical properties of Nano-Ni through modification of Nano-Ni surface may change Nano-Ni-induced lung injury, inflammation, and fibrosis. METHODS: At first, dose-response and time-response studies were performed to observe lung inflammation and injury caused by Nano-Ni. In the dose-response studies, mice were intratracheally instilled with 0, 10, 20, 50, and 100 µg per mouse of Nano-Ni and sacrificed at day 3 post-exposure. In the time-response studies, mice were intratracheally instilled with 50 µg per mouse of Nano-Ni and sacrificed at days 1, 3, 7, 14, 28, and 42 post-instillation. At the end of the experiment, mice were bronchoalveolar lavaged (BAL) and the neutrophil count, CXCL1/KC level, LDH activity, and concentration of total protein in the BAL fluid (BALF) were determined. In the comparative studies, mice were intratracheally instilled with 50 µg per mouse of Nano-Ni or with the same molar concentration of Ni as Nano-Ni of either partially [O]-passivated Nano-Ni (Nano-Ni-P) or carbon-coated Nano-Ni (Nano-Ni-C). At day 3 post-exposure, BAL was performed and the above cellular and biochemical parameters in the BALF were analyzed. The MMP-2/9 protein levels and activities in the BALF and mouse lung tissues were also determined. Mouse lung tissues were also collected for H&E staining, and measurement of thiobarbituric acid reactive substances (TBARS) and 8-hydroxy-2'-deoxyguanosine (8-OHdG) in the genomic DNA. At day 42 post-exposure, mouse right lung tissues were collected for H&E and Trichrome stainings, and left lung tissues were collected to determine the hydroxyproline content. RESULTS: Exposure of mice to Nano-Ni resulted in a dose-response increase in acute lung inflammation and injury reflected by increased neutrophil count, CXCL1/KC level, LDH activity, and concentration of total protein in the BALF. The time-response study showed that Nano-Ni-induced acute lung inflammation and injury appeared as early as day 1, peaked at day 3, and attenuated at day 7 post-instillation. Although the neutrophil count, CXCL1/KC level, LDH activity, and concentration of total protein in the BALF dramatically decreased over the time, their levels were still higher than those of the controls even at day 42 post-exposure. Based on the results of the dose- and time-response studies, we chose a dose of 50 µg per mouse of Nano-Ni, and day 3 post-exposure as short-term and day 42 post-exposure as long-term to compare the effects of Nano-Ni, Nano-Ni-P, and Nano-Ni-C on mouse lungs. At day 3 post-exposure, 50 µg per mouse of Nano-Ni caused acute lung inflammation and injury that were reflected by increased neutrophil count, CXCL1/KC level, LDH activity, concentration of total protein, and MMP-2/9 protein levels and activities in the BALF. Nano-Ni exposure also caused increased MMP-2/9 activities in the mouse lung tissues. Histologically, infiltration of large numbers of neutrophils and macrophages in the alveolar space and interstitial tissues was observed in mouse lungs exposed to Nano-Ni. Nano-Ni-P exposure caused similar acute lung inflammation and injury as Nano-Ni. However, exposure to Nano-Ni-C only caused mild acute lung inflammation and injury. At day 42 post-exposure, Nano-Ni caused extensive interstitial fibrosis and proliferation of interstitial cells with inflammatory cells infiltrating the alveolar septa and alveolar space. Lung fibrosis was also observed in Nano-Ni-P-exposed lungs, but to a much lesser degree. Only slight or no lung fibrosis was observed in Nano-Ni-C-exposed lungs. Nano-Ni and Nano-Ni-P, but not Nano-Ni-C, caused significantly elevated levels of TBARS in mouse lung tissues and 8-OHdG in mouse lung tissue genomic DNA, suggesting that Nano-Ni and Nano-Ni-P induce lipid peroxidation and oxidative DNA damage in mouse lung tissues, while Nano-Ni-C does not. CONCLUSION: Our results demonstrate that short-term Nano-Ni exposure causes acute lung inflammation and injury, while long-term Nano-Ni exposure causes chronic lung inflammation and fibrosis. Surface modification of Nano-Ni alleviates Nano-Ni-induced pulmonary effects; partially passivated Nano-Ni causes similar effects as Nano-Ni, but the chronic inflammation and fibrosis were at a much lesser degree. Carbon coating significantly alleviates Nano-Ni-induced acute and chronic lung inflammation and injury.


Subject(s)
Lung Injury/chemically induced , Metal Nanoparticles/toxicity , Nickel/chemistry , Animals , Bronchoalveolar Lavage Fluid , Chemokine CXCL1/metabolism , DNA Damage , L-Lactate Dehydrogenase/metabolism , Male , Metal Nanoparticles/chemistry , Mice, Inbred C57BL , Neutrophils/metabolism , Oxidation-Reduction , Oxidative Stress , Particle Size , Pneumonia/chemically induced , Surface Properties
18.
J Obstet Gynaecol Res ; 45(5): 1035-1044, 2019 May.
Article in English | MEDLINE | ID: mdl-30779263

ABSTRACT

AIM: To analyze the effects of high ovarian response on endometrial collagen synthesis and related gene expression during the peri-implantation period in patients undergoing in vitro fertilization-embryo transfer. METHODS: Peripheral blood and endometrial biopsies were obtained from infertile women on day 6 after oocytes retrieval or ovulation in 16 stimulated cycles (SC) and 16 natural cycles (NC) respectively. Serum estrogen (E2 ), progesterone (P4 ), histological staging, endometrial collagen, matrix metalloproteinases (MMP2, 9) and tissue inhibitors of metalloproteinases (TIMP1, 3) were assayed. RESULTS: Serum levels of both E2 and P4 were significantly higher in the SC group than those in the NC group. All endometrial samples were in the secretory phase. The collagen in the stroma of the SC group was more dense and higher than that in the NC group. MMP2 and MMP9 were detected significantly lower in the SC group than those in the NC group, while TIMP1 and TIMP3 were significantly higher. MMP2, 9 expressions are increased by estrogen and reduced by progesterone in dose-dependent manner through estrogen receptor and progesterone receptor. Correspondingly, TIMP1, 3 expressions decreased by estrogen dose-dependently while progesterone played the opposite role. CONCLUSION: High levels of P4 could stimulate excessive synthesis of collagen in peri-implantation endometrium of women with high ovarian response, and the mechanisms may be related to the decrease of MMP2, 9 and the increase of TIMP1, 3 through P4 receptor.


Subject(s)
Collagen/metabolism , Embryo Implantation , Embryo Transfer , Endometrium/metabolism , Fertilization in Vitro , Infertility, Female/metabolism , Infertility, Female/therapy , Ovulation Induction , Adult , Female , Humans
19.
Phytother Res ; 32(7): 1373-1381, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29532526

ABSTRACT

Tumour metastasis is the major cause of breast cancer mortality. Myricetin, a natural polyphenol, is found in teas, wines, and berries. The pharmacodynamic action and molecular mechanism of myricetin on breast cancer metastasis remain unknown. Here, we investigated the effect of myricetin on MDA-Mb-231Br cell viability, migration, invasion, and 4T1 mouse lung metastasis mouse models. MMP-2/9 protein expression and ST6GALNAC5 expression were analysed using western blot assays and quantitative real-time polymerase chain reaction, respectively. Cell migration and invasion were detected by wound-healing and Boyden transwell assays. The antimetastatic effect in vivo was evaluated by lung metastasis model. Myricetin significantly decreased the activities of MMP-2/9 and mRNA levels of ST6GALNAC5. In addition, the migration, invasion, and adhesion were effectively inhibited in a concentration-dependent manner. On the other hand, mice treated with myricetin exhibited smaller tumour nodules compared with the vehicle mice, with only 17.78 ± 15.41% after treatment with 50 mg/kg myricetin. In conclusion, myricetin could significantly block invasion of MDA-Mb-231Br cells through suppressing the protein expression of MMP-2/9 and the expression of ST6GALNAC5, as well as lung metastasis in a mouse model, which suggests that myricetin should be developed as a potential therapeutic candidate for breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Flavonoids/therapeutic use , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Down-Regulation , Female , Flavonoids/pharmacology , Humans , Mice , Mice, Inbred BALB C , Neoplasm Metastasis
20.
Biol Reprod ; 97(5): 660-670, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-29036448

ABSTRACT

Accumulating evidence has suggested an interaction between endometriotic cells and macrophages in the endometriotic microenvironment and the potential role of this interaction in the pathogenesis of endometriosis. However, how endometriotic cells communicate with macrophages to influence their function is poorly understood. In the present study, we found that the mRNA expression and production of CC chemokine ligand 2 (CCL2) were much higher in human endometriotic epithelial cells (11Z and 12Z) than those in human endometrial epithelial cells (HES). The inhibition of CCL2 action using neutralizing antibodies substantially suppressed macrophage migration induced by endometriotic epithelial cells. The endometriosis-associated macrophages (EAMs), which are the macrophages that are stimulated by the conditioned medium (CM) of human endometriotic cells, highly expressed the M2 phenotype markers (MRC1 and TREM2). In addition, the CM of EAMs significantly increased cell migration in 12Z cells, but no significant change was observed in cell growth. RT-PCR and antibody array analyses revealed that EAMs highly express and produce interleukin (IL) 6 compared to macrophages stimulated by the CM of HES cells. Moreover, the EAM-CM-induced migration and MMP2/9 expression in endometriotic cells were significantly attenuated by IL6 signaling inhibition. These results suggest a reciprocal activation of macrophages and endometriotic cells via the soluble factors CCL2 and IL6, which may contribute to the development of endometriosis.


Subject(s)
Cell Movement/physiology , Epithelial Cells/physiology , Interleukin-6/pharmacology , Macrophage Activation/physiology , Macrophages/physiology , Cell Line , Cell Movement/drug effects , Cell Survival , Chemokine CCL2/metabolism , Endometrium/cytology , Epithelial Cells/drug effects , Female , Gene Expression Regulation/physiology , Humans , Interleukin-6/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL