Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 992
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Tissue Cell ; 88: 102371, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38593570

ABSTRACT

BACKGROUND: Paeonol is a representative active ingredient of the traditional Chinese medicinal herbs Cortex Moutan, which has a well-established cardioprotective effect on ischemic heart disease. However, there is little evidence of the protective effect of paeonol, and its pharmacological mechanism is also unclear. This study aims to explore the protective effect and mechanism of Paeonol on myocardial infarction rat and hypoxic H9c2 cells. METHODS: Myocardial ischemia/reperfusion (I/R) was induced by occlusion of the left anterior descending coronary artery for 1 h followed by 3 h of reperfusion, and then gavage with Paeonol for 7 days. H9c2 cells were applied for the in vitro experiments and hypoxia/reoxygenation (H/R) model was established. CKIP-1 expression was evaluated by qPCR and western blot. The expression of genes involved in apoptosis, inflammation and ion channel was measured by western blot. The currents levels of Nav1.5 and Kir2.1 were measured by whole-cell patch-clamp recording. RESULTS: CKIP-1 expression was decreased in H/R-induced H9c2 cells, which was inversely increased after Paeonol treatment. Paeonol treatment could increase the viability of H/R-induced H9c2 cells and diminish the apoptosis and inflammation of H/R-induced H9c2 cells, while si-CKIP-1 treatment inhibited the phenomena. Moreover, the currents levels of Nav1.5 and Kir2.1 were reduced in H/R-induced H9c2 cells, which were inhibited after Paeonol treatment. Intragastric Paeonol can reduce the ventricular arrhythmias in rats with myocardial infarction. CONCLUSIONS: The protective effects of Paeonol on myocardial infarction rats and hypoxic H9c2 cells were achieved by up-regulating CKIP-1.


Subject(s)
Acetophenones , Cell Hypoxia , Up-Regulation , Acetophenones/pharmacology , Animals , Rats , Up-Regulation/drug effects , Cell Hypoxia/drug effects , Cell Line , Ion Channels/metabolism , Ion Channels/genetics , Apoptosis/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Male , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/pathology , Myocardial Infarction/metabolism , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Rats, Sprague-Dawley
2.
Phytomedicine ; 109: 154570, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36610169

ABSTRACT

BACKGROUND: Lung cancer is one of the most common cancers worldwide and is by far the leading cause of cancer death attributed to its rapid metastasis and poor prognosis. Given that hypoxia-inducible factors (HIFs) are associated with cancer metastasis, discovering agents to inhibit HIF-mediated invasive cancer is highly desired. PURPOSE: This study aimed to investigate the natural acridone compounds isolated from Severinia buxifolia for the potential to delay hypoxia-induced lung cancer invasiveness by HIF inhibition. METHODS: Using a hypoxia-responsive element (HRE) luciferase reporter, cell migration and invasion assays, real-time PCR, Western blot, and DNA recombinant clones, compound effect on HIF activity, cancer metastasis, HIF-1α mRNA transcription, HIFs protein stability, and HIF-1α translation were observed under hypoxia conditions. RESULTS: Atalaphyllidine (Sbs-A) and atalaphyllinine (Sbs-B) were found to show the most potent effects on HIF transcriptional activity and HIF-1α protein expression in NSCLC cell line A549, although Sbs-A and Sbs-B might not attribute decreasing HIF-1α mRNA expression to potent inhibition of HIF activity. HIF-1α protein stability was not affected by Sbs-A; also, prolyl hydroxylase and proteasome inhibitors could not reverse the inhibitory effect from compounds. Furthermore, 3 - 10 µM low concentrations of Sbs-A inhibited HIF target gene expression, gelatin zymography activity, and A549 cancer invasion. Ultimately, Sbs-A inhibited HIF-1α 5'UTR-mediated translation independent of oxygen concentration, underlying the mechanism of compounds inhibiting HIF-1α protein expression. CONCLUSION: Our study proposed Severinia buxifolia-isolated acridone compounds inhibited 5'-mRNA HIFA-mediated translation and provided evidence supporting the ability of acridone compounds in targeting HIFα for delayed lung cancer metastasis.


Subject(s)
Hypoxia , Lung Neoplasms , Humans , Cell Line, Tumor , 5' Untranslated Regions , Cell Hypoxia , Lung Neoplasms/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
3.
Phytomedicine ; 108: 154513, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36332389

ABSTRACT

BACKGROUND: Hypoxia is a characteristic feature of many solid tumors. As an adaptive response to hypoxia, tumor cells activate hypoxia-inducible factor-1α (HIF-1α). Under hypoxic conditions, angiogenesis mediated by HIF-1α is involved in the growth and metastasis of tumor cells. During the angiogenic process, differentiated tip endothelial cells (ECs) characterized by high expression of DLL4 promote angiogenic germination through filopodia. Inhibitors of HIF-1α or DLL4 have been widely studied PURPOSE: We tried to find inhibitors targeting both HIF-1α and DLL4 in tumor which have not yet been developed. STUDY DESIGN: In this study, we examined a natural compound that inhibits sprouting angiogenesis and tumor growth by targeting both HIF-1α and DLL4 under hypoxic conditions. METHODS: After examining cell viability of 70 selected natural compounds, we assessed the effects of compounds on HIF-1α and DLL4 transcriptional activity using a dual-luciferase reporter assay. Western blot analysis, immunofluoresecnt assay and real-time qPCR were performed to identify expression of proteins, such as HIF-1α and DLL4, as well as HIF-1α target genes under hypoxic conditions. In vitro angiogenesis assay and in vivo allograft tumor experiment were performed to investigate inhibition of tumor growth through anti-angiogenic activity. RESULTS: Among these compounds, steppogenin, which is extracted from the root bark of Morus alba l, respectively inhibited the transcriptional activity of HIF-1α under hypoxic conditions in HEK293T cells and vascular endothelial growth factor (VEGF)-induced DLL4 expression in vascular ECs in a dose-dependent manner. In tumor cells and retinal pigment epithelial cells, steppogenin significantly suppressed HIF-1α protein levels under hypoxic conditions as well as VEGF-induced DLL4 expression in ECs. Furthermore, steppogenin suppressed hypoxia-induced vascular EC proliferation and migration as well as VEGF-induced sprouting of EC spheroids. CONCLUSION: These results suggest that the natural compound steppogenin could potentially be used to treat angiogenic diseases, such as those involving solid tumors, because of its dual inhibition of HIF-1α and DLL4.


Subject(s)
Neoplasms , Vascular Endothelial Growth Factor A , Humans , Adaptor Proteins, Signal Transducing/metabolism , Calcium-Binding Proteins/metabolism , Cell Hypoxia , Cell Line, Tumor , Endothelial Cells/metabolism , Endothelium/metabolism , Endothelium/pathology , HEK293 Cells , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/metabolism
4.
Toxicol Appl Pharmacol ; 448: 116092, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35654276

ABSTRACT

Gypenosides are major bioactive ingredients of G. pentaphyllum. In our previous study, we found that gypenosides had neuroprotective effects against hypoxia-induced injury. In the current study, we focused on the protective effects of gypenoside-14 (GP-14), which is one of the newly identified bioactive components, on neuronal injury caused by severe hypoxia (0.3% O2). The results showed that GP-14 pretreatment alleviated the cell viability damage and apoptosis induced by hypoxia in PC12 cells. Moreover, GP-14 pretreatment also attenuated primary neuron injuries under hypoxic conditions. Additionally, GP-14 pretreatment significantly ameliorated neuronal damage in the hippocampal region induced by high-altitude cerebral edema (HACE). At the molecular level, GP-14 pretreatment reversed the decreased activities of the AKT and ERK signaling pathways caused by hypoxia in PC12 cells and primary neurons. To comprehensively explore the possible mechanisms, transcriptome sequencing was conducted, and these results indicated that GP-14 could alter the transcriptional profiles of primary neuron. Taken together, our results suggest that GP-14 acts as a neuroprotective agent to protect against neuronal damage induced by severe hypoxia and it is a promising compound for the development of neuroprotective drugs.


Subject(s)
MAP Kinase Signaling System , Neurons , Neuroprotective Agents , Proto-Oncogene Proteins c-akt , Animals , Apoptosis/drug effects , Cell Hypoxia/drug effects , Gene Expression Profiling , Gynostemma/chemistry , MAP Kinase Signaling System/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Rats
5.
Molecules ; 27(11)2022 May 30.
Article in English | MEDLINE | ID: mdl-35684442

ABSTRACT

Background: This study comparatively assessed seven indigenous traditional tea plants on several attributes that included antioxidant, nutritional, caffeine contents, and cyclooxygenase activity. Methodology: Nutritional content of all tea plants were determined for energy, fat, carbohydrates, total sugars, dietary fiber and amino acids. Antioxidant potential and the antioxidant potentiating secondary metabolites were also measured and compared. Further, we investigated the tea plants for any role they would have on cyclooxygenase (COX) activity on cobalt chloride (CoCl2) induced human glioma cell lines (U87MG). Results: The tea plants were found non-cytotoxic at concentrations tested against the human Chang liver and HeK 293 kidney cells and were found to be naturally caffeine free. The lowest and highest extraction yield among the tea plants was 7.1% for B. saligna and 15.48% for L. scaberrimma respectively. On average, the flavonol content was 12 to 8 QE/g, ORAC 800 µmol TE/g, TEAC 150 µmol TE/g, FRAP 155 µmol AAE/g, polyphenols 40 mg GAE/g, flavanols 0.35 mg CE/g, flavonols 12 mg QE/g and total flavonoid content (TFC) 180 µg QE/mg. The COX activity has been found to be inhibited by a dose-dependent manner by L. scaberrimma, B. saligna and L. javanica. Conclusion: The results further support competitive value of tea plants and need for improved and further development.


Subject(s)
Antioxidants , Teas, Herbal , Antioxidants/chemistry , Caffeine , Cell Hypoxia , Cyclooxygenase Inhibitors , Flavonols , HEK293 Cells , Humans , Nutritive Value , Polyphenols/chemistry , Prostaglandin-Endoperoxide Synthases , South Africa
6.
J Nanobiotechnology ; 20(1): 187, 2022 Apr 12.
Article in English | MEDLINE | ID: mdl-35413842

ABSTRACT

BACKGROUND: Non-redundant properties such as hypoxia and acidosis promote tumor metabolic adaptation and limit anti-cancer therapies. The key to the adaptation of tumor cells to hypoxia is the transcriptional and stable expression of hypoxia-inducible factor-1 alpha (HIF-1α). The phosphorylation-activated tumorigenic signal PI3K/AKT/mTOR advances the production of downstream HIF-1α to adapt to tumor hypoxia. Studies have elucidated that acid favors inhibition of mTOR signal. Nonetheless, carbonic anhydrase IX (CAIX), overexpressed on membranes of hypoxia tumor cells with pH-regulatory effects, attenuates intracellular acidity, which is unfavorable for mTOR inhibition. Herein, a drug delivery nanoplatform equipped with dual PI3K/mTOR inhibitor Dactolisib (NVP-BEZ235, BEZ235) and CAIX inhibitor 4-(2-aminoethyl) benzene sulfonamide (ABS) was designed to mitigate hypoxic adaptation and improve breast cancer treatment. RESULTS: ABS and PEG-NH2 were successfully modified on the surface of hollow polydopamine (HPDA), while BEZ235 and Chlorin e6 (Ce6) were effectively loaded with the interior of HPDA to form HPDA-ABS/PEG-BEZ235/Ce6 (H-APBC) nanoparticles. The release of BEZ235 from H-APBC in acid microenvironment could mitigate PI3K/mTOR signal and resist HIF-1α-dependent tumor hypoxia adaptation. More importantly, ABS modified on the surface of H-APBC could augment intracellular acids and enhances the mTOR inhibition. The nanoplatform combined with phototherapy inhibited orthotopic breast cancer growth while reducing spontaneous lung metastasis, angiogenesis, based on altering the microenvironment adapted to hypoxia and extracellular acidosis. CONCLUSION: Taken together, compared with free BEZ235 and ABS, the nanoplatform exhibited remarkable anti-tumor efficiency, reduced hypoxia adaptation, mitigated off-tumor toxicity of BEZ235 and solved the limited bioavailability of BEZ235 caused by weak solubility.


Subject(s)
Breast Neoplasms , Carbonic Anhydrase IX , Nanoparticles , Phototherapy , Quinolines , TOR Serine-Threonine Kinases , Acidosis , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carbonic Anhydrase IX/antagonists & inhibitors , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation , Drug Delivery Systems , Humans , Imidazoles , Molecular Targeted Therapy , Nanoparticles/administration & dosage , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Quinolines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Tumor Hypoxia
8.
Oxid Med Cell Longev ; 2022: 3235102, 2022.
Article in English | MEDLINE | ID: mdl-35186183

ABSTRACT

Hypoxia-induced pulmonary arterial hypertension (HPAH) is due to hypoxia caused by vascular endothelial cell remolding and damage. Previous studies have suggested that CX3CL1 plays an important role in HPAH which is affected by oxidative stress. Ca2+ channel activation correlated with increasing NF-κB levels induced by ROS. Tanreqing injection (TRQ) is a traditional Chinese medicine (TCM) for acute upper respiratory tract infection and acute pneumonia. In the present study, we explored the effect of TRQ on human pulmonary artery smooth muscle cells (HPASMCs) undergoing hypoxia and feasible molecular mechanisms involved in. Cell proliferation was assayed using CCK8 kits. Immunofluorescence and western blotting along with ELISA assay were performed to investigate the effect of TRQ on hypoxia-induced ROS, Ca2+, hydroxyl free radicals, and the expression of Ca2+ channel protein TRPC1, CX3CR1, HIF-1α, NF-κBp65, and p-NF-κBp65 in HPASMCs. Human CX3CL1 and the inhibitor of TRPC1 as SKF96365 were used for further investigation. TRQ inhibited hypoxia-induced increasing cell adhesion, ROS, Ca2+, hydroxyl free radicals, CX3CR1, HIF-1α, NF-κBp65 activation, and even on TRPC1 expression in HPASMC which tended to be attenuated even reversed by CX3CL1. Our results suggested that TRQ might help to attenuate remodeling of HPASMC through inhibiting the ROS and TRPC1/CX3CL1 signaling pathway.


Subject(s)
Cell Hypoxia/drug effects , Chemokine CX3CL1/metabolism , Drugs, Chinese Herbal/therapeutic use , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/etiology , Myocytes, Smooth Muscle/drug effects , Pulmonary Artery/drug effects , TRPC Cation Channels/metabolism , Cell Proliferation , Drugs, Chinese Herbal/pharmacology , Humans , Hypertension, Pulmonary/physiopathology
9.
Am J Chin Med ; 50(2): 621-637, 2022.
Article in English | MEDLINE | ID: mdl-35114913

ABSTRACT

Hypoxic microenvironment and dysregulated endoplasmic reticulum stress/unfolded protein response (UPR) system are considered important factors that promote cancer progression. Although osthole extracted from Cnidium monnieri(Fructus Cnidii) has been confirmed to exhibit an anticancer activity in various cancers, the effects of osthole in hypoxic colon cancer cells have not been explored. Therefore, the aim of this study was to examine whether osthole has an inhibitory effect on hypoxic colon cancer HCT116 cells and further investigate the underlying molecular mechanisms. Treatment with osthole significantly attenuated the cell viability, proliferation, and migration in hypoxic HCT116 cells. Osthole also activated UPR signaling such as phospho-eukaryotic initiation factor 2 alpha (EIF2[Formula: see text]/ATF4/CHOP/DR5 cascade accompanied by upregulation of pro-apoptotic proteins. Moreover, the tubule-like formation of human umbilical vein endothelial cells, the secretion of vascular endothelial growth factor A, and the expression and activity of hypoxia-inducible factor-1[Formula: see text] (HIF-1[Formula: see text] in hypoxic HCT116 cells were markedly suppressed by osthole. However, suppressing EIF2[Formula: see text] phosphorylation with salubrinal or ISRIB markedly reversed the effects of osthole on the expressions of pro-apoptotic proteins and HIF-1[Formula: see text]. Co-treatment of hypoxic HCT116 cells with osthole greatly increased the sensitivity to cisplatin and the expressions of phospho-EIF2[Formula: see text] and cleaved caspase 3. Collectively, the inhibitory effect of osthole in hypoxic HCT116 cells may be associated with EIF2[Formula: see text] phosphorylation-mediated apoptosis and translational repression of HIF-1[Formula: see text]. Taken together, osthole may be a potential agent in the treatment of colon cancer.


Subject(s)
Colonic Neoplasms , Vascular Endothelial Growth Factor A , Apoptosis , Cell Hypoxia , Colonic Neoplasms/drug therapy , Coumarins , Eukaryotic Initiation Factor-2/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Phosphorylation , Tumor Microenvironment
10.
Mol Biol Rep ; 49(2): 885-894, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35001248

ABSTRACT

BACKGROUND: Anshen Buxin Liuwei pill (ABLP) is a Mongolian medicinal formula that is composed of six medicinal materials: the Mongolian medicine Bos taurus domesticus Gmelin, Choerospondias axillaris (Roxb.) Burtt et Hill, Myristica fragrans Houtt., Eugenia caryophµllata Thunb., Aucklandia lappa Decne., and Liqui dambar formosana Hance. ABLP is considered to have a therapeutic effect on symptoms such as coronary heart disease, angina pectoris, arrhythmia, depression and irritability, palpitation, and shortness of breath. METHODS: H9c2 cardiomyocytes were used to construct a hypoxia/reoxygenation (HR) injury model. CCK-8 assay and Annexin V-FITC cell apoptosis assays were used for cell viability and cell apoptosis determination. The LDH, SOD, MDA, CAT, CK, GSH-Px, Na+-K+-ATPase, and Ca2+-ATPase activities in cells were determined to assess the protective effects of ABLP. The mRNA levels of Sirtuin3 (Sirt3) and Cytochrome C (Cytc) in H9c2 cells were determined by quantitative real-time PCR. RESULTS: The results indicate that HR-treated cells began to shrink from the spindle in an irregular shape with some floated in the medium. By increasing the therapeutic dose of ABLP (5, 25, and 50 µg/mL), the cells gradually reconverted in a concentration-dependent manner. The release of CK in HR-treated cells was significantly increased, indicating that ABLP exerts a protective effect in H9c2 cells against HR injury and can improve mitochondrial energy metabolism and mitochondrial function integrity. The present study scrutinized the cardioprotective effects of ABLP against HR-induced H9c2 cell injury through antioxidant and mitochondrial pathways. CONCLUSIONS: ABLP could be a promising therapeutic drug for the treatment of myocardial ischemic cardiovascular disease. The results will provide reasonable information for the clinical use of ABLP.


Subject(s)
Medicine, East Asian Traditional/methods , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/metabolism , Animals , Apoptosis/drug effects , Cell Hypoxia/drug effects , Cell Line , Cell Survival/drug effects , Cytochromes c/metabolism , Hypoxia/metabolism , Mitochondria/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Rats , Sirtuin 3/metabolism
11.
J Nanobiotechnology ; 20(1): 42, 2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35062959

ABSTRACT

Despite considerable progress has been achieved in hypoxia-associated anti-tumor therapy, the efficacy of utilizing hypoxia-activated prodrugs alone is not satisfied owing to the inadequate hypoxia within the tumor regions. In this work, a mitochondrial targeted nanoplatform integrating photodynamic therapy, photothermal therapy and hypoxia-activated chemotherapy has been developed to synergistically treat cancer and maximize the therapeutic window. Polydopamine coated hollow copper sulfide nanoparticles were used as the photothermal nanoagents and thermosensitive drug carriers for loading the hypoxia-activated prodrug, TH302, in our study. Chlorin e6 (Ce6) and triphenyl phosphonium (TPP) were conjugated onto the surface of the nanoplatform. Under the action of TPP, the obtained nanoplatform preferentially accumulated in mitochondria to restore the drug activity and avoid drug resistance. Using 660 nm laser to excite Ce6 can generate ROS and simultaneously exacerbate the cellular hypoxia. While under the irradiation of 808 nm laser, the nanoplatform produced local heat which can increase the release of TH302 in tumor cells, ablate cancer cells as well as intensify the tumor hypoxia levels. The aggravated tumor hypoxia then significantly boosted the anti-tumor efficiency of TH302. Both in vitro and in vivo studies demonstrated the greatly improved anti-cancer activity compared to conventional hypoxia-associated chemotherapy. This work highlights the potential of using a combination of hypoxia-activated prodrugs plus phototherapy for synergistic cancer treatment.


Subject(s)
Cell Hypoxia/drug effects , Drug Delivery Systems/methods , Mitochondria/metabolism , Nanoparticles/chemistry , Photochemotherapy/methods , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Female , Mice , Mice, Inbred C57BL , Nitroimidazoles/chemistry , Nitroimidazoles/pharmacokinetics , Nitroimidazoles/pharmacology , Phosphoramide Mustards/chemistry , Phosphoramide Mustards/pharmacokinetics , Phosphoramide Mustards/pharmacology , Prodrugs/chemistry , Prodrugs/pharmacology , Tissue Distribution
12.
Pharmacol Res ; 175: 105989, 2022 01.
Article in English | MEDLINE | ID: mdl-34800628

ABSTRACT

Chronic cerebral ischemia (CCI) refers to long-term hypoperfusion of cerebral blood flow with the main clinical manifestations of progressive cognitive impairment. The pathological mechanism of CCI is complex, and there is a lack of effective treatments. Salvianolic acid A (SalA) is a neuroprotective extract of Salvia miltiorrhiza with the effects of anti-inflammation and anti-apoptosis. In this study, the effect of SalA on cognitive function and Drd2/Cryab/NF-κB signaling pathway in rats with CCI was investigated. Morris water maze and open field test were used to observe the effects of SalA on the cognitive function of CCI rats. The pathological changes in the brain were observed by HE, Nissl, and LFB staining. TUNEL staining, enzyme-linked immunosorbent assay, and western blot analysis were used to detect the inflammatory and apoptosis in the cortex and hippocampus. The expression of Drd2/Cryab/NF-κB pathway-related molecules and Drd2 localization were detected by western blotting and dual immunofluorescence, respectively. SH-SY5Y cells were exposed to chronic hypoglycemic and hypoxic injury in vitro, and Drd2 inhibitor haloperidol was used to verify the involved pathway. The results showed that SalA could improve the cognitive function of CCI rats, reduce pathological damage of cortex and hippocampus, inhibit neuroinflammation and apoptosis, and suppress the activation of NF-κB by regulating Drd2/Cryab pathway. And SalA inhibited NF-κB activation and nuclear translocation in SH-SY5Y cells by upregulating Drd2/Cryab pathway, which was reversed by haloperidol interference. In conclusion, SalA could relieve CCI-induced cognitive impairment in rats, at least partly through the Drd2/Cryab/NF-κB pathway.


Subject(s)
Brain Ischemia/drug therapy , Caffeic Acids/therapeutic use , Cognitive Dysfunction/drug therapy , Lactates/therapeutic use , Neuroinflammatory Diseases/drug therapy , Neuroprotective Agents/therapeutic use , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Caffeic Acids/pharmacology , Cell Hypoxia/drug effects , Cell Line, Tumor , Chronic Disease , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Crystallins/metabolism , Glucose/metabolism , Humans , Lactates/pharmacology , Male , Microtubule-Associated Proteins/metabolism , NF-kappa B/metabolism , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Neuroprotective Agents/pharmacology , Rats, Wistar , Receptors, Dopamine D2/metabolism
13.
Chem Biol Interact ; 352: 109783, 2022 Jan 25.
Article in English | MEDLINE | ID: mdl-34932953

ABSTRACT

This paper provides an identification and detailed assessment of hormetic dose responses of embryonic stem cells (ESCs) with particular emphasis on cell renewal (proliferation) and differentiation, underlying mechanistic foundations and potential therapeutic implications. Hormetic dose responses were commonly reported, being induced by a broad range of chemicals, including pharmaceuticals (e.g., atorvastatin, isoproterenol, lithium, nicotine, ouabain), dietary supplements (e.g., curcumin, multiple ginsenosides, resveratrol), endogenous agents (e.g., estrogen, hydrogen peroxide, melatonin), and physical stressor agents (e.g., hypoxia, ionizing radiation). ESC-hormetic dose responses are similar for other stem cell types (e.g., adipose-derived stem cells, apical papilla, bone marrow stem cells, dental pulp stem cells, endothelial stem cells, muscle stem cells, periodontal ligament stem cells, neural stem cells), indicating a high degree of generality for the hormetic-stem cells response. The widespread occurrence of hormetic dose responses shown by ESCs and other stem cells suggests that the hormetic dose response may represent a fundamental and highly conserved evolutionary strategy.


Subject(s)
Embryonic Stem Cells/drug effects , Hormesis , Animals , Biological Evolution , Cell Differentiation/drug effects , Cell Hypoxia/physiology , Cell Proliferation/drug effects , Cell Survival/drug effects , Dietary Supplements , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Fatty Acids/administration & dosage , Hormesis/physiology , Humans
14.
J Ethnopharmacol ; 283: 114734, 2022 Jan 30.
Article in English | MEDLINE | ID: mdl-34648900

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Previous studies have shown that the active fraction of Rhodiola tangutica (Maxim.) S.H. Fu (ACRT) dilates pulmonary arteries and thwarts pulmonary artery remodelling. The dilatation effect of ACRT on pulmonary artery vascular rings could be reduced by potassium (K+) channel blockers. However the exact mechanisms of ACRT on ion channels are still unclear. AIM OF THE STUDY: This study aimed to investigate whether the effect of ACRT on K+ channels inhibits cell proliferation after pulmonary artery smooth muscle cells (PASMCs) are exposed to hypoxia. MATERIALS AND METHODS: The whole-cell patch-clamp method was used to clarify the effect of ACRT on the K+ current (IK) of rat PASMCs exposed to hypoxia. The mRNA and protein expression levels were detected using real-time quantitative polymerase chain reaction (RT-qPCR) and western blotting, respectively. The intracellular calcium (Ca2+) concentration ([Ca2+]i) values in rat PASMCs were detected by laser scanning confocal microscopy. The cell cycle and cell proliferation were assessed using flow cytometry analysis and CCK-8 and EdU assays. RESULTS: ACRT pretreatment alleviated the inhibition of IK induced by hypoxia in rat PASMCs. Compared with hypoxia, ACRT upregulated voltage-dependent K+ channel (Kv) 1.5 and big-conductance calcium-activated K+ channel (BKCa) mRNA and protein expression and downregulated voltage-dependent Ca2+ channel (Cav) 1.2 mRNA and protein expression. ACRT decreased [Ca2+]i, inhibited the promotion of cyclin D1 and proliferating cell nuclear antigen (PCNA) expression, and prevented the proliferation of rat PASMCs exposed to hypoxia. CONCLUSION: In conclusion, the present study demonstrated that ACRT plays a key role in restoring ion channel function and then inhibiting the proliferation of PASMCs under hypoxia, ACRT has preventive and therapeutic potential in hypoxic pulmonary hypertension.


Subject(s)
Muscle, Smooth, Vascular/drug effects , Plant Extracts/pharmacology , Pulmonary Artery/drug effects , Rhodiola/chemistry , Animals , Calcium/metabolism , Cell Hypoxia , Cell Proliferation/drug effects , Hypertension, Pulmonary/drug therapy , Male , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Patch-Clamp Techniques , Potassium Channels/drug effects , Potassium Channels/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Pulmonary Artery/cytology , Rats , Rats, Sprague-Dawley
15.
Int J Mol Sci ; 22(24)2021 Dec 14.
Article in English | MEDLINE | ID: mdl-34948200

ABSTRACT

Combination therapy is becoming imperative for the treatment of many cancers, as it provides a higher chance of avoiding drug resistance and tumor recurrence. Among the resistance-conferring factors, the tumor microenvironment plays a major role, and therefore, represents a viable target for adjuvant therapeutic agents. Thus, hypoxia and extracellular acidosis are known to select for the most aggressive and resilient phenotypes and build poorly responsive regions of the tumor mass. Carbonic anhydrase (CA, EC 4.2.1.1) IX isoform is a surficial zinc metalloenzyme that is proven to play a central role in regulating intra and extracellular pH, as well as modulating invasion and metastasis processes. With its strong association and distribution in various tumor tissues and well-known druggability, this protein holds great promise as a target to pharmacologically interfere with the tumor microenvironment by using drug combination regimens. In the present review, we summarized recent publications revealing the potential of CA IX inhibitors to intensify cancer chemotherapy and overcome drug resistance in preclinical settings.


Subject(s)
Carbonic Anhydrase IX/antagonists & inhibitors , Carbonic Anhydrase Inhibitors/pharmacology , Neoplasms/drug therapy , Animals , Cell Hypoxia/drug effects , Combined Modality Therapy , Humans , Neoplasms/metabolism , Tumor Microenvironment/drug effects
16.
Int J Mol Sci ; 22(24)2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34948250

ABSTRACT

Hypoxia is a major obstacle to gastric cancer (GC) therapy and leads to chemoresistance as GC cells are frequently exposed to the hypoxia environment. Apigenin, a flavonoid found in traditional medicine, fruits, and vegetables and an HDAC inhibitor, is a powerful anti-cancer agent against various cancer cell lines. However, detailed mechanisms involved in the treatment of GC using APG are not fully understood. In this study, we investigated the biological activity of and molecular mechanisms involved in APG-mediated treatment of GC under hypoxia. APG promoted autophagic cell death by increasing ATG5, LC3-II, and phosphorylation of AMPK and ULK1 and down-regulating p-mTOR and p62 in GC. Furthermore, our results show that APG induces autophagic cell death via the activation of the PERK signaling, indicating an endoplasmic reticulum (ER) stress response. The inhibition of ER stress suppressed APG-induced autophagy and conferred prolonged cell survival, indicating autophagic cell death. We further show that APG induces ER stress- and autophagy-related cell death through the inhibition of HIF-1α and Ezh2 under normoxia and hypoxia. Taken together, our findings indicate that APG activates autophagic cell death by inhibiting HIF-1α and Ezh2 under hypoxia conditions in GC cells.


Subject(s)
Apigenin/metabolism , Stomach Neoplasms/metabolism , Adenylate Kinase/metabolism , Apigenin/pharmacology , Apoptosis , Autophagic Cell Death/drug effects , Autophagy , Autophagy-Related Protein 5/metabolism , Autophagy-Related Protein-1 Homolog/metabolism , Cell Death , Cell Hypoxia , Cell Line, Tumor , Cell Survival , Endoplasmic Reticulum Stress , Enhancer of Zeste Homolog 2 Protein/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Phosphorylation , RNA-Binding Proteins/metabolism , Signal Transduction , Stomach Neoplasms/physiopathology , TOR Serine-Threonine Kinases/metabolism
17.
Cell Rep ; 36(11): 109701, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34525352

ABSTRACT

Citrate lies at a critical node of metabolism, linking tricarboxylic acid metabolism and lipogenesis via acetyl-coenzyme A. Recent studies have observed that deficiency of the sodium-dependent citrate transporter (NaCT), encoded by SLC13A5, dysregulates hepatic metabolism and drives pediatric epilepsy. To examine how NaCT contributes to citrate metabolism in cells relevant to the pathophysiology of these diseases, we apply 13C isotope tracing to SLC13A5-deficient hepatocellular carcinoma (HCC) cells and primary rat cortical neurons. Exogenous citrate appreciably contributes to intermediary metabolism only under hypoxic conditions. In the absence of glutamine, citrate supplementation increases de novo lipogenesis and growth of HCC cells. Knockout of SLC13A5 in Huh7 cells compromises citrate uptake and catabolism. Citrate supplementation rescues Huh7 cell viability in response to glutamine deprivation or Zn2+ treatment, and NaCT deficiency mitigates these effects. Collectively, these findings demonstrate that NaCT-mediated citrate uptake is metabolically important under nutrient-limited conditions and may facilitate resistance to metal toxicity.


Subject(s)
Citrates/metabolism , Nutrients/metabolism , Symporters/metabolism , Acetyl Coenzyme A/metabolism , Adult , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Hypoxia , Cell Line, Tumor , Cell Survival/drug effects , Female , Gene Editing , Glutamine/metabolism , Glutamine/pharmacology , Humans , Lipogenesis , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Neurons/cytology , Neurons/metabolism , Nutrients/pharmacology , Rats , Symporters/deficiency , Symporters/genetics , Zinc/pharmacology
18.
Bioorg Med Chem ; 46: 116394, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34509160

ABSTRACT

The study aimed to isolate and characterize zinc ionophores from Terminalia bellirica fruit using a liposome assay and test its utility in H9c2 rat cardiomyoblasts cells subjected to hypoxia/reoxygenation. Ethyl acetate extract that exhibited zinc ionophore activity was resolved to yield three polyphenols that were characterized as epicatechin-3-gallate (ECG), epigallocatechin-3-gallate (EGCG) and epigallocatechin (EGC) by nuclear magnetic resonance and electrospray ionization-mass spectra. The polyphenols enhanced the uptake of zinc into the liposomes and increased FluoZin-3 fluorescence. These polyphenols in the presence of 10 µM ZnCl2 enhanced the zinc import into H9c2 cells, whose intracellular zinc levels were otherwise lowered upon hypoxia/reoxygenation. EGCG proved to be more potent than ECG, which indeed was more effective than EGC in improving cellular zinc levels and in attenuating the apoptosis of H9c2 cells after hypoxia/reoxygenation injury. The polyphenols required zinc for anti-apoptotic effect. The cardioprotective effect is indeed due to enhanced zinc uptake mediated by these polyphenols.


Subject(s)
Cell Hypoxia/drug effects , Ionophores/pharmacology , Myocytes, Cardiac/drug effects , Plant Extracts/pharmacology , Zinc/pharmacology , Animals , Antioxidants , Apoptosis/drug effects , Cell Line , Fruit/chemistry , Ionophores/chemistry , Ionophores/isolation & purification , Myocytes, Cardiac/pathology , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Protective Agents , Rats , Terminalia/chemistry , Zinc/chemistry , Zinc/isolation & purification
19.
Mol Med Rep ; 24(4)2021 Oct.
Article in English | MEDLINE | ID: mdl-34435647

ABSTRACT

The development of several retinal diseases is closely related to hypoxia. As a component of the Traditional Chinese medicine Salvia miltiorrhiza, the effects of cryptotanshinone (CT) on retinal cells under hypoxic conditions are not well understood. The aim of the present study was to explore how CT exerted its protective effects on retinal pigment epithelium (RPE) cells under hypoxic conditions induced by cobalt chloride (CoCl2). The effects of CT were investigated using a Cell Counting Kit­8 assay, Annexin V­FITC/PI staining, reverse transcription­quantitative PCR and western blotting in ARPE­19 cells. CT (10 and 20 µM) reduced the CoCl2­induced increase in vascular endothelial growth factor expression and hypoxia­inducible transcription factor­1α expression in ARPE­19 cells. Additionally, CT alleviated hypoxia­induced apoptosis by regulating Bcl­2 and Bax protein expression. CT treatment also reduced the increase in the mRNA levels of IL­6, IL­1ß and TNF­α induced by CoCl2. In summary, CT may protect RPE cells against apoptosis and inflammation in CoCl2­induced hypoxia, and these results warrant further in vivo study into its value as a drug for treating hypoxic eye diseases.


Subject(s)
Cell Hypoxia/drug effects , Phenanthrenes/pharmacology , Protective Agents/pharmacology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Cobalt/toxicity , Cytokines/genetics , Cytokines/metabolism , Humans , Proto-Oncogene Proteins c-bcl-2/metabolism , Vascular Endothelial Growth Factor A/metabolism , bcl-2-Associated X Protein/metabolism
20.
Biomed Pharmacother ; 142: 112052, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34426261

ABSTRACT

BACKGROUND: In breast cancer, low oxygen availability is associated with a more aggressive phenotype and with malignant evolution of non-invasive cells. Natural compounds have long attracted attention in cancer treatment, and in recent years garlic (Allium sativum) organosulfur derivatives have been shown to negatively affect growth and invasion of tumor cells. METHODS: Homemade ethanol-based garlic extract (GE) was administered to MCF7 and MCF10DCIS breast tumor cell lines grown under moderate hypoxia. Cell cycle, epithelial-to-mesenchymal transition and cancer stem cell markers were evaluated. RESULTS: We revealed that, in the non-invasive MCF10DCIS cells but not in the post-EMT MCF7 cells, low oxygen availability induced the decrease of E-cadherin and the increase of vimentin and motility, that were prevented by GE administration. In both cell lines, treatment with GE counteracted the up-modulation of CD133 positive cells induced by hypoxia. CONCLUSIONS: Overall, our data firstly revealed anti-cancer properties of garlic in non-invasive breast cancer cells. In particular, they demonstrated a protective role of this natural product against the hypoxia-induced increase of molecules that play crucial roles in tumor evolution, suggesting that garlic derivatives can be considered in new approaches for preventing progression of breast tumors from non-invasive to infiltrating lesions.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/drug therapy , Cell Hypoxia/drug effects , Garlic/chemistry , Plant Extracts/pharmacology , AC133 Antigen/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Epithelial-Mesenchymal Transition/drug effects , Ethanol/chemistry , Humans , Neoplastic Stem Cells/metabolism , Vimentin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL