Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
J Appl Toxicol ; 44(2): 301-312, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37770382

ABSTRACT

Interleukin-12 (IL-12) is known to exert antitumor immune effects by promoting the activation and proliferation of T cells and NK cells within the immune system. However, clinical trials have observed systemic toxicity associated with the administration of IL-12. This has shelved development plans for its use as a cancer therapeutic drug. Therefore, it is critical that we perform a systematic evaluation of the toxicity and safety of repeated IL-12 administration. In this study, we conducted a comprehensive evaluation of the toxicity and safety of repeated rhIL-12 (recombinant human interleukin-12) administration in rhesus monkeys by assessing its effects on the immune system, organ function, and vital signs. Rhesus monkeys were subcutaneously injected with 0.5, 2.5, and 12.5 µg/kg of rhIL-12 for up to for 14 consecutive weeks. The low dose exhibited no signs of toxicity, whereas animals receiving higher doses displayed symptoms such as loose stools, reduced activity, anemia, and elevated liver function indicators (AST and TBIL). Following three administrations of 12.5 µg/kg, high dosing was adjusted to 7.5 µg/kg due to manifestations of symptoms like loose stools, decreased activity, and huddling in the cage. Furthermore, rhesus monkeys exhibited marked immunogenic responses to recombinant human interleukin-12 (rhIL-12). However, based on overall study findings, the No Observed Adverse Effect Level (NOAEL) for the subcutaneous injection of rhIL-12, when repeatedly administered for 3 months in rhesus monkeys, was considered to be 0.5 µg/kg. The Highest Non-Severely Toxic Dose (HNSTD) was considered to be 7.5 µg/kg.


Subject(s)
Antineoplastic Agents , Interleukin-12 , Animals , Humans , Macaca mulatta , Recombinant Proteins/toxicity , Interleukin-12/toxicity , Killer Cells, Natural
2.
Mar Drugs ; 21(3)2023 Feb 25.
Article in English | MEDLINE | ID: mdl-36976201

ABSTRACT

Cancer-derived small extracellular vesicles (sEVs) serve as critical mediators of cell-to-cell communication. Manzamine A (MA), a unique marine-derived alkaloid with various bioactivities, exerts anticancer effects against several kinds of tumors, but it remains unclear whether it has the same activity against breast cancer. Here, we proved that MA inhibits MDA-MB-231 and MCF-7 cell proliferation, migration, and invasion in a time- and dose-dependent manner. In addition, MA promotes autophagosome formation but suppresses autophagosome degradation in breast cancer cells. Importantly, we also found that MA stimulates sEVs secretion and increases autophagy-related protein accumulation in secreted sEVs, further potentiated by autophagy inhibitor chloroquine (CQ). Mechanistically, MA decreases the expression level of RIP1, the key upstream regulator of the autophagic pathway, and reduces the acidity of lysosome. Overexpression of RIP1 activated AKT/mTOR signaling, thus attenuating MA-induced autophagy and the corresponding secretion of autophagy-associated sEVs. Collectively, these data suggested that MA is a potential inhibitor of autophagy by preventing autophagosome turnover, and RIP1 mediates MA-induced secretory autophagy, which may be efficacious for breast cancer treatment.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/pathology , Autophagy , Signal Transduction , Cell Line, Tumor , Cell Proliferation , Apoptosis
3.
Molecules ; 28(14)2023 Jul 12.
Article in English | MEDLINE | ID: mdl-37513222

ABSTRACT

Considering the resistance and toxicity of traditional chemotherapeutic drugs, seeking potential candidate for treating breast cancer effectively is a clinical problem that should be solved urgently. Natural products have attracted extensive attention, owing to their multi-target advantages and low toxicity. In the current study, the effects of XK-81, a novel bromophenol compound extracted from Leathesia nana, on breast cancer, and its underlying mechanisms, were explored. Firstly, data from in vitro experiments indicated that 4T-1, one of common mouse breast cancer cell lines, was a XK-81-susceptible cell line, and ferroptosis was the major death manner in response to XK-81 treatment, which was evidenced by increasing intracellular Fe2+ and ROS level with condensed mitochondrial membrane densities, as well as decreasing the protein expressions of SLC7A11 and GPX4. In vivo, XK-81 suppressed the growth of 4T-1 breast-tumor in both BALB/C mice and zebrafish. Obviously, XK-81 decreased the protein expression of SLC7A11 and GPX4 in tumor tissues, hinting at the occurrence of ferroptosis. Moreover, XK-81 increased CD8+ T cells and NK cells numbers and regulated M1/M2 macrophage ratio in tumor tissues, indicating XK-81's immunotherapeutic effect. Additionally, the secretions of immune-related cytokines, including TNF-α, IL-1ß, and IL-12, were elevated with XK-81 stimulation in RAW 264.7 cells. Intriguingly, compared with doxorubicin-induced heart damage, XK-81 demonstrated the therapeutic advantage of little cardiotoxicity on the heart. XK-81 demonstrated potential antitumor advantage by both directly inducing ferroptosis-mediated death of tumor cells and immunization.


Subject(s)
Mammary Neoplasms, Animal , Zebrafish , Mice , Animals , Mice, Inbred BALB C , Immunotherapy , Immunization
4.
Anticancer Drugs ; 32(10): 1003-1010, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34145179

ABSTRACT

Surgical tumor removing is the most common procedure after a confirmed cancer diagnosis with no detected metastasis. Surgery can reduce tumor burden and address pathologic changes caused by local compression of tissues by the tumor. This lowers the chances of tumor cell spreading and creates more favorable conditions for further treatment. However, not all tumor cells can be eliminated through surgery. Even in the early stages of the disease, tumor cells often metastasize and cannot be identified by current detection methods. These tiny, disseminated tumors are often the cause of tumor recurrence. There is currently a lack of effective treatment options that can completely prevent tumor recurrence after surgery. To simulate the actual clinical situation, we selected murine-derived tumor cell lines S180 and Kcc853 to establish a post-transplantation residual tumor model in mice. Surgery was performed on mice inoculated with tumors. Tumor tissue was partially excised to set up the postsurgical residual tumor models. The model simulated the clinical situation where tumor cells were not completely eliminated or there were small tumors that had metastasized before surgery. IL-12 was injected to observe its effect on residual tumors or metastatic microtumors. The administration of IL-12 after surgery can significantly inhibit the growth of residual tumors and metastasis, improve the postoperative tumor-free rate and address the problem of tumor recurrence caused by the growth of residual tumors and micro-metastasis. Therefore, the use of IL-12 antitumor cytokine combined with surgery can effectively inhibit tumor recurrence. Low-dose IL-12 (1-10 ng/kg in humans) can inhibit residual tumor growth.


Subject(s)
Antineoplastic Agents/pharmacology , Interleukin-12/pharmacology , Kidney Neoplasms/drug therapy , Sarcoma/drug therapy , Animals , Body Weight/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Humans , Kidney Neoplasms/surgery , Mice , Mice, Nude , Sarcoma/surgery , Vincristine/pharmacology , Xenograft Model Antitumor Assays
5.
Toxicol Appl Pharmacol ; 409: 115271, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33065153

ABSTRACT

With the growth of the aging population, osteoporosis is becoming a global health problem. Ursolic acid (UA) is an active ingredient existed in a variety of foods and nature plants and owns plenty of pharmacological effects especially in treating metabolic disease. Our predication from network pharmacology hinted that UA has potential for ameliorating osteoporosis. Firstly through in vivo experiment, we confirmed that UA administration obviously protected against ovariectomy (OVX)-induced osteoporosis in rats by improving microarchitectural deterioration of trabecular bone (P < 0.001), decreasing numbers of TRAP positive osteoclast in vertebra (P < 0.001), as well as decreasing serum osteoclast-specific cytokines release (P < 0.001). Besides, UA ameliorated kidney damage secondary to OVX-induced osteoporosis by ameliorating glomerular atrophy, decreasing BUN and creatinine levels in OVX rats. In vitro, UA noticeably decreased osteoclastic-special marker proteins c-Fos and NFATc1 expressions (P < 0.001) in response to RANKL stimulation in macrophagy. Importantly, autophagy pathway was activated in the process of osteoclast differentiation and blocked by UA pretreatment. Furthermore, autophagy inhibitors suppressed osteoclast differentiation (P < 0.001). Collectively, UA may ameliorate osteoporosis by suppressing osteoclast differentiation mediated by autophagy. Our research provides scientific support for UA treating osteoporosis and offers an optimal dose for daily intake of UA safely to prevent bone diseases.


Subject(s)
Autophagy/drug effects , Biological Products/pharmacology , Osteoporosis/drug therapy , Osteoporosis/prevention & control , Triterpenes/pharmacology , Animals , Cell Differentiation/drug effects , Cell Line , Female , Mice , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteogenesis/drug effects , Osteoporosis/metabolism , Ovariectomy/methods , RAW 264.7 Cells , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Ursolic Acid
6.
Biosci Biotechnol Biochem ; 84(7): 1460-1466, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32195627

ABSTRACT

Chitooligosaccharides (COS) are derived from chitosan, which can be used as nutraceuticals and functional foods. Because of their various biological activities, COS are widely used in the food, medicine, agriculture, and other fields. COS were prepared by chitosanase  from Pseudoalteromonas sp. SY39 and their anti-obesity activity was researched in mice in this study. The effects of hydrolysis time, temperature, the ratio of enzyme to chitosan, and pH on the productivity of COS were discussed. Preparation process of COS was established in a 5-L fermenter. COS were characterized and their anti-obesity activity was studied in animal experiments. The results showed that COS could effectively reduce serum lipid levels and obesity in mice, and have a good anti-obesity activity. The preparation technology and remarkable anti-obesity activity of COS further expand their applications in the food and pharmaceutical industries.


Subject(s)
Anti-Obesity Agents/administration & dosage , Anti-Obesity Agents/chemical synthesis , Chitin/analogs & derivatives , Chitosan/chemistry , Glycoside Hydrolases/chemistry , Obesity/drug therapy , Pseudoalteromonas/enzymology , Animals , Anti-Obesity Agents/pharmacology , Chitin/administration & dosage , Chitin/chemical synthesis , Chitin/pharmacology , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Diet, High-Fat/adverse effects , Disease Models, Animal , Fatty Acids, Nonesterified/blood , Hydrogen-Ion Concentration , Hydrolysis , Male , Mice , Obesity/blood , Obesity/etiology , Oligosaccharides , Temperature , Triglycerides/blood
7.
Mar Drugs ; 18(6)2020 Jun 14.
Article in English | MEDLINE | ID: mdl-32545859

ABSTRACT

As prebiotics, galacto-oligosaccharides (GOSs) can improve the intestinal flora and have important applications in medicine. ß-galactosidases could promote the synthesis of GOSs in lactose and catalyze the hydrolysis of lactose. In this study, a new ß-galactosidase gene (gal2A), which belongs to the glycoside hydrolase family 2, was cloned from marine bacterium Alteromonas sp. QD01 and expressed in Escherichia coli. The molecular weight of Gal2A was 117.07 kDa. The optimal pH and temperature of Gal2A were 8.0 and 40 °C, respectively. At the same time, Gal2A showed wide pH stability in the pH range of 6.0-9.5, which is suitable for lactose hydrolysis in milk. Most metal ions promoted the activity of Gal2A, especially Mn2+ and Mg2+. Importantly, Gal2A exhibited high transglycosylation activity, which can catalyze the formation of GOS from milk and lactose. These characteristics indicated that Gal2A may be ideal for producing GOSs and lactose-reducing dairy products.


Subject(s)
Alteromonas/chemistry , Lactose/chemistry , Milk , Prebiotics , beta-Galactosidase/chemistry , Alteromonas/genetics , Animals , Cloning, Molecular , Dairying , Oligosaccharides/chemistry , beta-Galactosidase/genetics
8.
Phytother Res ; 34(10): 2639-2648, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32302031

ABSTRACT

The incidence of ulcerative colitis (UC) is increasing in recent years. The protective effect of cryptotanshinone, a natural compound from Salvia miltiorrhiza Bunge, on UC was investigated both in vivo and in vitro models. UC model was established by dextran sulfate sodium administration in drinking water and cryptotanshinone was orally administrated. RAW264.7 cells were stimulated by lipopolysaccharide (LPS) with or without cryptotanshinone pretreatment. The body weights and disease activity index (DAI) were recorded. The pathological alterations were evaluated by H&E staining. The levels of pro-inflammatory cytokines in colon tissues and cell culture medium were determined with enzyme-linked immune sorbent assay (ELISA) kits. The protein expression was detected by Western blotting and immunohistochemistry. Results showed that cryptotanshinone significantly increased the body weight and colon length, reduced the score of DAI, and improved pathological changes. Furthermore, the expression of inducible nitric oxide synthase, cyclooxygenase-2, receptor-interacting protein kinase 3, NF-κB p65 and the secretion of tumor necrosis factor-α, IL-6 in colon tissues and LPS-stimulated cells were significantly inhibited by cryptotanshinone. Besides, cryptotanshinone significantly inhibited LPS-triggered toll-like receptor 4 luciferase reporter activity with an IC50 at 7.2 µM. In conclusion, cryptotanshinone ameliorated experimental UC possibly by inhibiting intestinal inflammation.


Subject(s)
Colitis, Ulcerative/drug therapy , Dextran Sulfate/adverse effects , Drugs, Chinese Herbal/therapeutic use , Inflammation/drug therapy , Phenanthrenes/therapeutic use , Animals , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/pathology , Disease Models, Animal , Drugs, Chinese Herbal/pharmacology , Male , Mice , Phenanthrenes/chemistry
9.
Int J Mol Sci ; 21(5)2020 Feb 27.
Article in English | MEDLINE | ID: mdl-32121010

ABSTRACT

The recent emergence of antibiotic-resistant bacteria requires the development of new antibiotics or new agents capable of enhancing antibiotic activity. Lysozyme degrades bacterial cell wall without involving antibiotic resistance and has become a new antibacterial strategy. However, direct use of native, active proteins in clinical settings is not practical as it is fragile under various conditions. In this study, lysozyme was integrated into chitosan nanoparticles (CS-NPs) by the ionic gelation technique to obtain lysozyme immobilized chitosan nanoparticles (Lys-CS-NPs) and then characterized by dynamic light scattering (DLS) and transmission electron microscopy (TEM), which showed a small particle size (243.1 ± 2.1 nm) and positive zeta potential (22.8 ± 0.2 mV). The immobilization significantly enhanced the thermal stability and reusability of lysozyme. In addition, compared with free lysozyme, Lys-CS-NPs exhibited superb antibacterial properties according to the results of killing kinetics in vitro and measurement of the minimum inhibitory concentration (MIC) of CS-NPs and Lys-CS-NPs against Pseudomonas aeruginosa (P. aeruginosa), Klebsiella pneumoniae (K. pneumoniae), Escherichia coli (E. coli), and Staphylococcus aureus (S. aureus). These results suggest that the integration of lysozyme into CS-NPs will create opportunities for the further potential applications of lysozyme as an anti-bacterium agent.


Subject(s)
Anti-Bacterial Agents/pharmacology , Chitosan/chemistry , Drug Carriers/chemistry , Muramidase/pharmacology , Nanoparticles/chemistry , Dynamic Light Scattering , Enzyme Stability , Enzymes, Immobilized/pharmacology , Escherichia coli/drug effects , Klebsiella pneumoniae/drug effects , Microbial Sensitivity Tests , Microscopy, Electron, Transmission , Particle Size , Pseudomonas aeruginosa/drug effects , Staphylococcus aureus/drug effects , Temperature
10.
Acta Pharmacol Sin ; 40(5): 666-676, 2019 May.
Article in English | MEDLINE | ID: mdl-30218072

ABSTRACT

Hispidulin (4',5,7-trihydroxy-6-methoxyflavone) is a phenolic flavonoid isolated from the medicinal plant S. involucrata, which exhibits anti-neoplastic activity against several types of cancer. However, the mechanism underlying its anti-cancer activity against hepatocellular carcinoma (HCC) has not been fully elucidated. In this study, we investigated whether and how hispidulin-induced apoptosis of human HCC cells in vitro and in vivo. We showed that hispidulin (10, 20 µmol/L) dose-dependently inhibited cell growth and promoted apoptosis through mitochondrial apoptosis pathway in human HCC SMMC7721 cells and Huh7 cells. More importantly, we revealed that its pro-apoptotic effects depended on endoplasmic reticulum stress (ERS) and unfolded protein response (UPR), as pretreatment with salubrinal, a selective ERS inhibitor, or shRNA targeting a UPR protein CHOP effectively abrogated hispidulin-induced cell apoptosis. Furthermore, we showed that hispidulin-induced apoptosis was mediated by activation of AMPK/mTOR signaling pathway as pretreatment with Compound C, an AMPK inhibitor, or AMPK-targeting siRNA reversed the pro-apoptotic effect of hispidulin. In HCC xenograft nude mice, administration of hispidulin (25, 50 mg/kg every day, ip, for 27 days) dose-dependently suppressed the tumor growth, accompanied by inducing ERS and apoptosis in tumor tissue. Taken together, our results demonstrate that hispidulin induces ERS-mediated apoptosis in HCC cells via activating the AMPK/mTOR pathway. This study provides new insights into the anti-tumor activity of hispidulin in HCC.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Flavones/therapeutic use , Signal Transduction/drug effects , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Flavones/pharmacology , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Male , Mice, Inbred BALB C , Mice, Nude , Unfolded Protein Response/drug effects , Xenograft Model Antitumor Assays
11.
Mar Drugs ; 17(1)2019 Jan 21.
Article in English | MEDLINE | ID: mdl-30669556

ABSTRACT

Chitooligosaccharides (CHOS) have gained increasing attention because of their important biological activities. Enhancing the efficiency of CHOS production essentially requires screening of novel chitosanase with unique characteristics. Therefore, a rapid and efficient one-step affinity purification procedure plays important roles in screening native chitosanases. In this study, we report the design and synthesis of affinity resin for efficient purification of native chitosanases without any tags, using chitodisaccharides (CHDS) as an affinity ligand, to couple with Sepharose 6B via a spacer, cyanuric chloride. Based on the CHDS-modified affinity resin, a one-step affinity purification method was developed and optimized, and then applied to purify three typical glycoside hydrolase (GH) families: 46, 75, and 80 chitosanase. The three purified chitosanases were homogeneous with purities of greater than 95% and bioactivity recovery of more than 40%. Moreover, we also developed a rapid and efficient affinity purification procedure, in which tag-free chitosanase could be directly purified from supernatant of bacterial culture. The purified chitosanases samples using such a procedure had apparent homogeneity, with more than 90% purity and 10⁻50% yield. The novel purification methods established in this work can be applied to purify native chitosanases in various scales, such as laboratory and industrial scales.


Subject(s)
Chitin/analogs & derivatives , Chromatography, Affinity/instrumentation , Glycoside Hydrolases/isolation & purification , Chitin/chemical synthesis , Chitosan , Chromatography, Affinity/methods , Ligands , Oligosaccharides , Sepharose/chemistry , Triazines/chemistry
12.
Mar Drugs ; 17(5)2019 May 24.
Article in English | MEDLINE | ID: mdl-31137685

ABSTRACT

Alginate oligosaccharides (AOS) show versatile bioactivities. Although various alginate lyases have been characterized, enzymes with special characteristics are still rare. In this study, a polysaccharide lyase family 7 (PL7) alginate lyase-encoding gene, aly08, was cloned from the marine bacterium Vibrio sp. SY01 and expressed in Escherichia coli. The purified alginate lyase Aly08, with a molecular weight of 35 kDa, showed a specific activity of 841 U/mg at its optimal pH (pH 8.35) and temperature (45 °C). Aly08 showed good pH-stability, as it remained more than 80% of its initial activity in a wide pH range (4.0-10.0). Aly08 was also a thermo-tolerant enzyme that recovered 70.8% of its initial activity following heat shock treatment for 5 min. This study also demonstrated that Aly08 is a polyG-preferred enzyme. Furthermore, Aly08 degraded alginates into disaccharides and trisaccharides in an endo-manner. Its thermo-tolerance and pH-stable properties make Aly08 a good candidate for further applications.


Subject(s)
Aquatic Organisms/enzymology , Polysaccharide-Lyases/metabolism , Temperature , Vibrio/enzymology , Aquatic Organisms/genetics , Enzyme Stability , Escherichia coli/genetics , Hydrogen-Ion Concentration , Polysaccharide-Lyases/chemistry , Polysaccharide-Lyases/genetics , Polysaccharide-Lyases/isolation & purification , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Vibrio/genetics
13.
Molecules ; 24(1)2019 Jan 06.
Article in English | MEDLINE | ID: mdl-30621320

ABSTRACT

Chitosanases play an important role in chitosan degradation, forming enzymatic degradation products with several biological activities. Although many chitosanases have been discovered and studied, the enzymes with special characteristics are still rather rare. In this study, a new chitosanase, CsnM, with an apparent molecular weight of 28 kDa was purified from the marine bacterium Pseudoalteromonas sp. SY39. CsnM is a cold-adapted enzyme, which shows highest activity at 40 °C and exhibits 30.6% and 49.4% of its maximal activity at 10 and 15 °C, respectively. CsnM is also a thermo-tolerant enzyme that recovers 95.2%, 89.1% and 88.1% of its initial activity after boiling for 5, 10 and 20 min, respectively. Additionally, CsnM is an endo-type chitosanase that yields chitodisaccharide as the main product (69.9% of the total product). It's cold-adaptation, thermo-tolerance and high chitodisaccharide yield make CsnM a superior candidate for biotechnological application to produce chitooligosaccharides.


Subject(s)
Bacterial Proteins/metabolism , Glycoside Hydrolases/metabolism , Pseudoalteromonas/enzymology , Bacterial Proteins/isolation & purification , Chitin/analogs & derivatives , Chitin/metabolism , Chitosan , Glycoside Hydrolases/isolation & purification , Molecular Weight , Oligosaccharides , Temperature
14.
Toxicol Appl Pharmacol ; 352: 68-76, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29800642

ABSTRACT

Perfluorooctanoic acid (PFOA), a wide spread environmental pollutant, was associated with developmental cardiotoxicity in chicken embryo, while the underlying molecular mechanism had not been fully elucidated. In the current study, 2 mg/kg (egg weight) PFOA and/or 100 mg/kg (egg weight) l-carnitine were exposed to embryonic day zero (ED0) chicken embryo via air cell injection, and then bone morphogenic protein 2 (BMP2) silencing lentivirus or BMP2 recombinant protein were introduced into ED2 embryo. Electrocardiography and histological methods were utilized to assess the cardiac function and morphology in hatchling chickens, respectively. Consistent with previous results, 2 mg/kg PFOA exposure at ED0 significantly elevated heart rate and thinned right ventricular wall in hatchling chickens, while l-carnitine co-treatment reverted such changes. BMP2 silencing induced very similar changes in hatchling chicken hearts as PFOA exposure, while co-exposure of recombinant BMP2 protein alleviated PFOA-induced changes. l-carnitine exposure alleviated the BMP2-silencing induced changes as well. Western blotting revealed that PFOA exposure enhanced BMP2 expression and suppressed pSMAD1 expression in ED15 chicken embryo hearts, while both changes were reverted by l-carnitine co-exposure. Furthermore, silencing of BMP2 significantly increased the expression level of PPAR alpha in ED15 chicken embryo hearts, while silencing of PPAR alpha did not have significant impact on BMP2 expression. In conclusion, BMP2/pSMAD1 signaling participates in the PFOA-induced developmental cardiotoxicity in chicken embryo, which is likely located upstream of PPAR alpha for this particular endpoint. Protection of BMP2 signaling might contribute to l-carnitine mediated protection against PFOA-induced developmental cardiotoxicity.


Subject(s)
Bone Morphogenetic Protein 2/pharmacology , Caprylates/toxicity , Carnitine/pharmacology , Environmental Pollutants/toxicity , Fluorocarbons/toxicity , Heart Diseases/prevention & control , Heart/drug effects , Animals , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Cardiotoxicity , Chick Embryo , Cytoprotection , Gene Expression Regulation, Developmental , Gene Silencing , Heart/embryology , Heart/physiopathology , Heart Diseases/chemically induced , Heart Diseases/embryology , Heart Diseases/physiopathology , Heart Rate/drug effects , PPAR alpha/genetics , PPAR alpha/metabolism , Phosphorylation , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Smad1 Protein/genetics , Smad1 Protein/metabolism , Ventricular Function, Right/drug effects , Ventricular Remodeling/drug effects
15.
Cell Biol Int ; 41(12): 1296-1306, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28792088

ABSTRACT

Vascular smooth muscle cell (VSMC) proliferation is a major contributor to atherosclerosis. This study investigated the inhibitory effects of oleanolic acid (OA) against oxidized low-density lipoprotein (ox-LDL)-induced VSMC proliferation in A7r5 cells and explored underlying molecular mechanism. The cell proliferation was quantified with cell counting kit-8 (CCK-8), in which ox-LDL significantly increased A7r5 cells proliferation, while OA pretreatment effectively alleviated such changes without inducing overt cytotoxicity, as indicated by lactate dehydrogenase (LDH) assay. Quantitative real-time RT-PCR (qRT-PCR) and Western blotting revealed increased UCP2 and FGF-2 expression levels as well as decreased p53 and TSP-1 expression levels in A7r5 cells following ox-LDL exposure, while OA pretreatment reversed such changes. Furthermore, inhibiting UCP2 with genipin remarkably reversed the changes in the expression levels of FGF-2, p53, and TSP-1 induced by ox-LDL exposure; silencing FGF-2 with siRNA did not significantly change the expression levels of UCP2 but effectively reversed the changes in the expression levels of p53 and TSP-1, and activation of p53 with PRIMA-1 only significantly affected the changes in the expression levels of TSP-1, but not in UCP2 or FGF-2, suggesting a UCP-2/FGF-2/p53/TSP-1 signaling in A7r5 cells response to ox-LDL exposure. Additionally, co-treatment of OA and genipin exhibited similar effects to the expression levels of UCP2, FGF-2, p53, and TSP-1 as OA or genipin solo treatment in ox-LDL-exposed A7r5 cells, suggesting the involvement of UCP-2/FGF-2/p53/TSP-1 in the mechanism of OA. In conclusion, OA inhibits ox-LDL-induced VSMC proliferation in A7r5 cells, the mechanism involves the changes in UCP-2/FGF-2/p53/TSP-1.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Oleanolic Acid/pharmacology , Uncoupling Protein 2/metabolism , Apoptosis/drug effects , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cell Proliferation/drug effects , Cells, Cultured , Fibroblast Growth Factor 1/metabolism , Humans , L-Lactate Dehydrogenase/metabolism , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , Muscle, Smooth, Vascular/cytology , Thrombospondin 1/metabolism , Tumor Suppressor Protein p53/metabolism
17.
Acta Pharmacol Sin ; 38(12): 1618-1631, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29119970

ABSTRACT

Hispidulin, a polyphenolic flavonoid extracted from the traditional Chinese medicinal plant S involucrata, exhibits anti-tumor effects in a wide array of human cancer cells, mainly through growth inhibition, apoptosis induction and cell cycle arrest. However, its precise anticancer mechanisms remain unclear. In this study, we investigated the molecular mechanisms that contribute to hispidulin-induced apoptosis of human clear-cell renal cell carcinoma (ccRCC) lines Caki-2 and ACHN. Hispidulin (10, 20 µmol/L) decreased the viability of ccRCC cells in dose- and time-dependent manners without affecting that of normal tubular epithelial cells. Moreover, hispidulin treatment dose-dependently increased the levels of cleaved caspase-8 and caspase-9, but the inhibitors of caspase-8 and caspase-9 only partly abrogated hispidulin-induced apoptosis, suggesting that hispidulin triggered apoptosis via both extrinsic and intrinsic pathways. Moreover, hispidulin treatment significantly inhibited the activity of sphingosine kinase 1 (SphK1) and consequently promoted ceramide accumulation, thus leading to apoptosis of the cancer cells, whereas pretreatment with K6PC-5, an activator of SphK1, or overexpression of SphK1 significantly attenuated the anti-proliferative and pro-apoptotic effects of hispidulin. In addition, hispidulin treatment dose-dependently activated ROS/JNK signaling and led to cell apoptosis. We further demonstrated in Caki-2 xenograft nude mice that injection of hispidulin (20, 40 mg·kg-1·d-1, ip) dose-dependently suppressed tumor growth accompanied by decreased SphK1 activity and increased ceramide accumulation in tumor tissues. Our findings reveal a new explanation for the anti-tumor mechanisms of hispidulin, and suggest that SphK1 and ceramide may serve as potential therapeutic targets for the treatment of ccRCC.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Ceramides/metabolism , Flavones/pharmacology , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Animals , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Kidney Neoplasms/drug therapy , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured
19.
Int J Mol Sci ; 18(6)2017 Jun 08.
Article in English | MEDLINE | ID: mdl-28594376

ABSTRACT

Perfluorooctanoic acid (PFOA) is an environmental contaminant that could induce developmental cardiotoxicity in a chicken embryo, which may be alleviated by l-carnitine. To explore the roles of reactive oxygen species (ROS) and nitric oxide (NO) in such changes and the potential effects of l-carnitine, fertile chicken eggs were exposed to PFOA via an air cell injection, with or without l-carnitine co-treatment. The ROS and NO levels in chicken embryo hearts were determined with electron spin resonance (ESR), and the protein levels of the nuclear factor κ-light chain-enhancer of activated B cells (NF-κB) p65 and inducible nitric oxide synthase (iNOS) in chicken embryo hearts were assessed with western blotting. The results of ESR indicated that PFOA exposure induced an elevation in the ROS levels in ED19 chicken embryo hearts and hatchling chicken hearts, while l-carnitine could alleviate such changes. Meanwhile, increased NO levels were observed in ED19 embryo hearts and hatchling hearts following PFOA exposure, while l-carnitine co-treatment exerted modulatory effects. Western blotting revealed that p65 translocation in ED19 embryo hearts and hatchling hearts was enhanced by PFOA, while l-carnitine co-treatment alleviated such changes. iNOS expression levels in ED19 embryo hearts followed the same pattern as NO levels, while a suppression of expression was observed in hatchling hearts exposed to PFOA. ROS/NF-κB p65 and iNOS/NO seem to be involved in the late stage (ED19 and post hatch) of PFOA-induced developmental cardiotoxicity in a chicken embryo. l-carnitine could exert anti-oxidant and NO modulatory effects in the developing chicken embryo hearts, which likely contribute to its cardioprotective effects.


Subject(s)
Caprylates/adverse effects , Cardiotonic Agents/pharmacology , Cardiotoxicity/etiology , Cardiotoxicity/metabolism , Carnitine/pharmacology , Fluorocarbons/adverse effects , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism , Animals , Cardiotoxicity/prevention & control , Chick Embryo , Heart/drug effects , Heart Rate , Nitric Oxide Synthase Type II/metabolism , Transcription Factor RelA/metabolism
20.
J Cardiovasc Pharmacol ; 67(4): 344-50, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26771154

ABSTRACT

The aim of this study is to assess the potential protective effect of oleanolic acid (OA) against ox-LDL induced damage in human umbilical vascular endothelial cells (HUVECs) and investigate potential mechanism of action including antioxidative effects and inhibition of mitochondria apoptosis pathway. Cell counting kit 8 was used to evaluate the viability of HUVECs. 2', 7'-DCFH-DA staining and flow cytometry was used to assess the levels of intracellular reactive oxygen species in HUVECs. The protein expression levels of uncoupling protein 2, cytochrome C, and apoptosis induction factors were measured by western blotting. The results indicated that OA treatment alleviated ox-LDL induced cytotoxicity in HUVECs and ameliorated the reactive oxygen species levels. Western blotting results demonstrated that OA treatment increased the expression level of uncoupling protein 2 and decreased the release of cytochrome C and apoptosis induction factors from mitochondria to cytoplasm, suggesting inhibition of mitochondria apoptosis pathway. In conclusion, OA could protect HUVECs from ox-LDL-induced cytotoxicity; its antioxidant property and inhibition of mitochondria apoptosis are likely crucial contributors.


Subject(s)
Antioxidants/pharmacology , Oleanolic Acid/pharmacology , Reactive Oxygen Species/metabolism , Apoptosis/drug effects , Apoptosis Inducing Factor , Blotting, Western , Cytochromes c/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Lipoproteins, LDL/administration & dosage , Mitochondria/drug effects , Uncoupling Protein 2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL