Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
J Pineal Res ; 74(3): e12855, 2023 Apr.
Article En | MEDLINE | ID: mdl-36692032

Autophagy deficiency in macrophages exacerbates inflammation in atherosclerosis (AS), and recently, galectin-3 (Gal-3) has been implicated as a critical promoter of inflammation in AS. Further, melatonin (Mel) exerts an autophagy-promoting effect in many chronic inflammatory diseases. In this study, we aimed to investigate whether Mel inhibits AS progression by downregulating Gal-3 to enhance autophagy and inhibit inflammation. Thus, we performed in vivo and in vitro experiments using high-fat diet (HFD)-fed ApoE-/-  mice and THP-1 macrophages, respectively. Smart-seq of AS plaque macrophages revealed that the differentially expressed genes (DEGs) downregulated by Mel were enriched in immune-related processes, and changes in inflammation status were confirmed based on lower levels of proinflammatory factors in Mel-treated HFD-fed ApoE-/-  mice and THP-1 macrophages. Further, via transcriptome-based multiscale network pharmacology platform (TMNP), the upstream target genes of the smart-seq DEGs were identified, and Gal-3 showed a high score. Gal-3 was downregulated both in vivo and in vitro by Mel treatment. Besides, the enrichment of the target genes predicted via the TMNP method indicated that autophagy considerably affected the DEGs. Mel treatment as well as Gal-3 knockdown downregulated most inflammatory response-related proteins could attribute to enhancing autophagy. Mechanistically, Mel treatment inhibited Gal-3 leading to lowering the activity of the nuclear transcription factor-kappa B (NF-κB) pathway, and promoting the nuclear localization of transcription factor EB (TFEB). However, increased secretion of Gal-3 activated the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) pathway and impaired autophagy via binding to CD98. Thus, Mel promoted autophagy and restrained inflammation by downregulating Gal-3, implying that it holds promise as a treatment for AS.


Atherosclerosis , Melatonin , Animals , Mice , Galectin 3/genetics , Galectin 3/metabolism , Galectin 3/pharmacology , Melatonin/pharmacology , Down-Regulation , Signal Transduction , Phosphatidylinositol 3-Kinases/metabolism , Atherosclerosis/drug therapy , Atherosclerosis/genetics , Inflammation/metabolism , NF-kappa B/metabolism , Autophagy , Apolipoproteins E/genetics , Apolipoproteins E/metabolism
2.
Aging (Albany NY) ; 13(5): 7096-7119, 2021 02 26.
Article En | MEDLINE | ID: mdl-33639613

Atherosclerosis is a chronic inflammatory disease that commonly affects the elderly and is characterized by vascular damage, macrophage infiltration, and plaque formation. Moreover, it increases the risk of cardiovascular disease. The pathogenesis of atherosclerosis involves an interplay between macrophage autophagy and apoptosis. A recently discovered transcription factor, transcription factor EB (TFEB) is known to activate autophagy in macrophages. Sirtuin deacetylase 1 (SIRT1), a nicotinamide adenine dinucleotide (NAD+)-dependent histone deacetylase, activates several transcription factors, including TFEB. We studied the effects of berberine on the NAD+ synthesis pathway and interactions between SIRT1 and TFEB. We also studied the effects of berberine-induced TFEB activation via SIRT1 on autophagy and apoptosis of peritoneal macrophages. We found that berberine promoted autophagy of peritoneal macrophages by activating SIRT1 via the NAD+ synthesis pathway and, in turn, promoting TFEB nuclear translocation and deacetylation. The functional regulation of SIRT1 and TFEB by berberine could be exploited as a potential therapeutic strategy for atherosclerosis.


Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Berberine/pharmacology , Macrophages, Peritoneal/drug effects , Sirtuin 1/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Flow Cytometry , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred C57BL , NAD/metabolism
3.
Pancreas ; 49(10): 1335-1341, 2020.
Article En | MEDLINE | ID: mdl-33122522

OBJECTIVES: Pancreatic acinar necrosis is a typical feature in the early phase of severe acute pancreatitis (SAP). Muscarinic acetylcholine receptor M3 (CHRM3) has been reported to play important roles in promoting insulin secretion and tumor cell proliferation, but its effect on necrosis remains unknown. This study revealed the important role of CHRM3 in regulating L-arginine-induced SAP and the molecular mechanisms. METHODS: To verify the function of CHRM3, pancreatic tissues and primary acinar cells of CRISPR/Cas9-mediated Chrm3 knockout mice were used in CHRM3 knockdown experiments, and to ascertain the CHRM3 overexpression, PLV-EGFP-Chrm3 plasmids were transfected in acinar cells in vitro. RESULTS: In L-arginine-induced SAP, CHRM3 is activated and regulates SAP through the mitogen-activated protein kinase/p38 pathway. Moreover, the expression of miR-31-5p decreased in the SAP model both in vitro and in vivo. Mir-31-5p effects the necrosis of acinar cells in SAP by upregulating the target gene RIP3, and miR-31-5p is a downstream miRNA of CHRM3. CONCLUSIONS: Necrosis in L-arginine-induced SAP is promoted by CHRM3 through the mitogen-activated protein kinase-p38/miR-31-5p/RIP3 axis.


Acinar Cells/enzymology , MicroRNAs/metabolism , Pancreas/enzymology , Pancreatitis/prevention & control , Receptor, Muscarinic M3/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Acinar Cells/pathology , Animals , Arginine , Cells, Cultured , Disease Models, Animal , Male , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Necrosis , Pancreas/pathology , Pancreatitis/chemically induced , Pancreatitis/enzymology , Pancreatitis/pathology , Phosphorylation , Receptor, Muscarinic M3/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction
4.
Aging (Albany NY) ; 12(9): 8622-8639, 2020 05 11.
Article En | MEDLINE | ID: mdl-32392535

The lemon essential oil (LEO), extracted from the fruit of lemon, has been used to treat multiple pathological diseases, such as diabetes, inflammation, cardiovascular diseases, depression and hepatobiliary dysfunction. The study was designed to study the effects of LEO on cognitive dysfunction induced by Alzheimer's disease (AD). We used APP/PS1 double transgene (APP/PS1) AD mice in the experiment; these mice exhibit significant deficits in synaptic density and hippocampal-dependent spatial related memory. The effects of LEO on learning and memory were examined using the Morris Water Maze (MWM) test, Novel object recognition test, and correlative indicators, including a neurotransmitter (acetylcholinesterase, AChE), a nerve growth factor (brain-derived neurotrophic factor, BDNF), a postsynaptic marker (PSD95), and presynaptic markers (synapsin-1, and synaptophysin), in APP/PS1 mice. Histopathology was performed to estimate the effects of LEO on AD mice. A significantly lowered brain AChE depression in APP/PS1 and wild-type C57BL/6L (WT) mice. PSD95/ Synaptophysin, the index of synaptic density, was noticeably improved in histopathologic changes. Hence, it can be summarized that memory-enhancing activity might be associated with a reduction in the AChE levels and is elevated by BDNF, PSD95, and synaptophysin through enhancing synaptic plasticity.


Acetylcholinesterase/metabolism , Cognition/drug effects , Cognitive Dysfunction/drug therapy , Hippocampus/drug effects , Neuronal Plasticity/drug effects , Plant Oils/pharmacology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Cognitive Dysfunction/psychology , Disease Models, Animal , Disks Large Homolog 4 Protein/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Morris Water Maze Test , Neuroprotective Agents/pharmacology , Oils, Volatile/pharmacology , Spatial Memory/drug effects
5.
Food Funct ; 11(4): 3549-3562, 2020 Apr 30.
Article En | MEDLINE | ID: mdl-32270151

A high-fat diet is recognized as an important factor in the development of cardiovascular diseases including cardiomyopathy. Besides high-fat diets, large quantities of ethanol also induce cardiomyopathy in both animals and humans. Emerging evidence suggests that low ethanol intake may have a protective effect on the cardiovascular system. This study aimed to clarify whether low-dose ethanol intake could prevent high-fat diet-induced adverse effects on cardiomyocytes in mice. After 6-8 weeks of feeding, the heart weight significantly decreased in ethanol + HFD mice compared to HFD mice. In addition, cardiac triglycerides and lipid droplets also decreased, but no statistically significant difference in cholesterol level was found between the two groups. Expression of the fatty acid transporters Cd36, Slc27a1 and Got2 was downregulated in the ethanol + HFD group. According to echocardiography, the mass and volume of the left ventricle were reduced, and the ejection fraction (EF) and fractional shortening (FS) were increased in mice fed with alcohol. Low doses of ethanol reduced the cardiomyocytes' cross-sectional area and the expression of the hypertrophic markers ANP and BNP. Moreover, Col1a1, the main collagen type expressed in the heart, was also reduced by low-dose ethanol consumption. Also, the expression of Rgs5, a crucial component of the signaling pathway involved in cardiac remodeling and heart failure, was upregulated in response to ethanol intake. The data suggest that low ethanol intake prevents adverse effects induced by a high-fat diet, such as lipid accumulation, cardiac dysfunction, hypertrophy and fibrosis. Furthermore, low ethanol intake upregulates Rgs5, which suggests it plays a role in cardiac remodeling and heart failure.


Cardiomyopathies/prevention & control , Diet, High-Fat , Ethanol/administration & dosage , Protective Agents/administration & dosage , Administration, Oral , Animals , Cardiomyopathies/diagnostic imaging , Disease Models, Animal , Echocardiography , Ethanol/pharmacology , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Protective Agents/pharmacology , Random Allocation , Rats
6.
Gene ; 729: 144319, 2020 Mar 01.
Article En | MEDLINE | ID: mdl-31884108

In previous study, we have found that microRNA-23a is down regulated in atherosclerotic tissues. Here we demonstrate that miR-23a directly binds to 3'UTR of HSP90 mRNA to suppress the expression of HSP90. To investigate the potential roles of miR-23a in macrophage, THP-1 macrophages were transfected with miR-23a mimics or inhibitors. Our results showed inflammatory factors IL-6 and MCP-1 concentrations in cell culture medium of macrophage and foam cell transfected with miR-23a mimics were decreased. Furthermore, we find that apoptosis of macrophage and foam cells transfected with miR-23a mimics were inhibited. Over expression of miR-23a in foam cells could reduced lipid intake and accumulation in foam cells. Meanwhile, we found that in inflammatory macrophages and foam cells transfected with miR-23a mimcs, HSP90 and NF-κB proteins are significantly decreased. Our results have suggested a promising and potential therapeutic target for atherosclerosis.


Atherosclerosis/genetics , Atherosclerosis/pathology , Foam Cells/pathology , HSP90 Heat-Shock Proteins/genetics , Macrophages/pathology , MicroRNAs/genetics , 3' Untranslated Regions , Apoptosis/genetics , Atherosclerosis/metabolism , Cell Proliferation/genetics , Foam Cells/metabolism , Humans , Inflammation/genetics , Macrophages/metabolism , MicroRNAs/metabolism , NF-kappa B/metabolism , THP-1 Cells
7.
J Cell Biochem ; 121(3): 2618-2631, 2020 03.
Article En | MEDLINE | ID: mdl-31692054

Acinar cells in acute pancreatitis (AP) die through apoptosis and necrosis, the impacts of which are quite different. Early clinical interference strategies on preventing the progress of AP to severe acute pancreatitis (SAP) are the elimination of inflammation response and inhibition of necrosis. Muscarinic acetylcholine receptor M3 was encoded by Chrm3 gene. It is one of the best-characterized receptors of pancreatic ß cells and regulates insulin secretion, but its function in AP remains unclear. In this study, we explored the function of Chrm3 gene in the regulation of cell death in l-arginine-induced SAP animal models. We found that Chrm3 was upregulated in pancreatitis, and we further confirmed the localization of Chrm3 resided in both pancreatic islets and acinar cell membranes. The reduction of Chrm3 decreased the pathological lesion of SAP and reduced amylase activities in serum. Consistently, Chrm3 can suppress acinar cells necrosis markedly, but has no effect on regulating apoptosis after l-arginine treatment. It was shown that Chrm3 attenuated acinar cells necrosis at least in part by stabilizing caspase-8. Thus, this study indicates that Chrm3 is critical participants in SAP, and regulation of Chrm3 expression might be a useful therapeutic strategy for preventing pathologic necrosis.


Acinar Cells/pathology , Caspase 8/metabolism , Necrosis , Pancreatitis/prevention & control , Protective Agents/pharmacology , Receptor, Muscarinic M3/physiology , Transcriptome , Acinar Cells/metabolism , Animals , Arginine/toxicity , Caspase 8/chemistry , Caspase 8/genetics , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatitis/chemically induced , Pancreatitis/pathology
8.
Cell Physiol Biochem ; 52(6): 1325-1338, 2019.
Article En | MEDLINE | ID: mdl-31050281

BACKGROUND/AIMS: Atherosclerosis is a chronic inflammatory cardiovascular disease. Macrophages are major components of atherosclerotic plaques and play a key role in the development of atherosclerosis by secreting a variety of pro-inflammatory factors. Our previous studies have confirmed that upconversion nanoparticles encapsulating chlorin e6 (UCNPs-Ce6) mediated photodynamic therapy (PDT) can promote cholesterol efflux and induce apoptosis in THP-1 macrophages. In this study, we investigated whether reactive oxygen species (ROS) generated by UCNPs-Ce6-mediated PDT can induce autophagy to inhibit the expression of pro-inflammatory factor in M1 peritoneal macrophages. METHODS: Peritoneal macrophages were collected from C57/BL6 mice injected with 3% thioglycollate broth medium and induced by lipopolysaccharides and interferon-γ. Intracellular ROS production was assessed by 2'-7'-dichloroflorescein diacetate and flow cytometry. Autophagy was assayed by western blot, transmission electron microscopy and immunofluorescence. Pro-inflammatory cytokines were detected by enzyme-linked immunosorbent assay and western blot. RESULTS: Model M1 peritoneal macrophages were established after 24 h induction. Protein expression levels of LC3 II and Beclin1, and degradation of p62 increased and peaked at 2 h in the PDT group. Meanwhile, levels of inflammatory cytokines iNOS, IL-12, and TNF-α markedly decreased after PDT. The increase in autophagy levels and decrease in pro-inflammatory cytokines were significantly inhibited by 3-methyladenine. Furthermore, ROS generated by UCNPs- Ce6 mediated PDT activated autophagy. The expression of autophagy related-protein and inflammatory cytokines iNOS, IL-12, and TNF-α were inhibited by the ROS inhibitor N-acetyl cysteine. CONCLUSION: ROS generated by UCNPs-Ce6-mediated PDT activated autophagy and inhibited the expression of pro-inflammatory factors of M1 peritoneal macrophage via the PI3K/AKT/mTOR signaling pathway.


Autophagy/drug effects , Metal Nanoparticles/chemistry , Porphyrins/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Beclin-1/metabolism , Interleukin-12/metabolism , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Nitric Oxide Synthase Type II/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Photochemotherapy , Porphyrins/chemistry , Porphyrins/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/metabolism
9.
Cell Death Dis ; 10(3): 202, 2019 Feb 27.
Article En | MEDLINE | ID: mdl-30814492

The authors wish to point out that in Figure 1f, the picture of DAPI in the ATG5 siRNA group is incorrect. During the process of image synthesis, the authors mixed the pictures of DAPI in the control group and ATG5 siRNA group, leading to the duplicate between them of DAPI. Furthermore, the AMPK blot and the AKT blot in Figure 2a were inadvertently duplicated with the third ß-actin in Figure 2a and AKT in Figure 4e, respectively. The authors would like to apologize for any inconvenience this may have caused.

10.
Cell Physiol Biochem ; 48(4): 1616-1627, 2018.
Article En | MEDLINE | ID: mdl-30071509

BACKGROUND/AIMS: Atherosclerosis is a chronic inflammatory cardiovascular disease. Macrophages are major components of atherosclerotic plaques and play a key role in the development of atherosclerosis by secreting a variety of pro-inflammatory factors. Our previous studies have confirmed that upconversion nanoparticles encapsulating chlorin e6 (UCNPs-Ce6) mediated photodynamic therapy (PDT) can promote cholesterol efflux and induce apoptosis in THP-1 macrophages. In this study, we investigated whether reactive oxygen species (ROS) generated by UCNPs-Ce6-mediated PDT can induce autophagy to inhibit the expression of pro-inflammatory factor in M1 peritoneal macrophages. METHODS: Peritoneal macrophages were collected from C57/BL6 mice injected with 3% thioglycollate broth medium and induced by lipopolysaccharides and interferon-γ. Intracellular ROS production was assessed by 2'-7'-dichloroforescein diacetate and flow cytometry. Autophagy was assayed by western blot, transmission electron microscopy and immunofluorescence. Pro-inflammatory cytokines were detected by enzyme-linked immunosorbent assay and western blot. RESULTS: Model M1 peritoneal macrophages were established after 24 h induction. Protein expression levels of LC3 II and Beclin1, and degradation of p62 increased and peaked at 2 h in the PDT group. Meanwhile, levels of inflammatory cytokines iNOS, IL-12, and TNF-α markedly decreased after PDT. The increase in autophagy levels and decrease in pro-inflammatory cytokines were significantly inhibited by 3-methyladenine. Furthermore, ROS generated by UCNPs-Ce6 mediated PDT activated autophagy. The expression of autophagy related-protein and inflammatory cytokines iNOS, IL-12, and TNF-α were inhibited by the ROS inhibitor N-acetyl cysteine. CONCLUSIONS: ROS generated by UCNPs-Ce6-mediated PDT activated autophagy and inhibited the expression of pro-inflammatory factors of M1 peritoneal macrophage via the PI3K/AKT/mTOR signaling pathway.


Autophagy/drug effects , Magnetite Nanoparticles/toxicity , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Animals , Cell Survival/drug effects , Enzyme Inhibitors/pharmacology , Interleukin-12/metabolism , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Magnetite Nanoparticles/chemistry , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Nitric Oxide Synthase Type II/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Photochemotherapy , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Receptors, IgG/metabolism , TOR Serine-Threonine Kinases/metabolism , Transcription Factor TFIIH , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/metabolism
11.
Oncotarget ; 8(46): 81591-81603, 2017 Oct 06.
Article En | MEDLINE | ID: mdl-29113417

In 1841, the extraction of hematoporphyrin from dried blood by removing iron marked the birth of the photosensitizer. The last twenty years has witnessed extensive research in the application of photodynamic therapy (PDT) in tumor-bearing (or other diseases) animal models and patients. The period has seen development of photosensitizers from the first to the third generation, and their evolution from simple to more complex entities. This review focuses on porphyrin photosensitizers and their effect on tumors, mediated via several pathways involved in cell necrosis, apoptosis or autophagic cell death, and the preventive and therapeutic application of PDT against atherosclerosis.

12.
Cell Death Dis ; 8(6): e2864, 2017 06 08.
Article En | MEDLINE | ID: mdl-28594401

Macrophage-derived foam cells are a major component of atherosclerotic plaques and have an important role in the progression of atherosclerotic plaques, thus posing a great threat to human health. Photodynamic therapy (PDT) has emerged as a therapeutic strategy for atherosclerosis. Here, we investigated the effect of PDT mediated by upconversion fluorescent nanoparticles encapsulating chlorin e6 (UCNPs-Ce6) on the cholesterol efflux of THP-1 macrophage-derived foam cells and explored the possible mechanism of this effect. First, we found that PDT notably enhanced the cholesterol efflux and the induction of autophagy in both THP-1 and peritoneal macrophage-derived foam cells. The autophagy inhibitor 3-methyladenine and an ATG5 siRNA significantly attenuated PDT-induced autophagy, which subsequently suppressed the ABCA1-mediated cholesterol efflux. Furthermore, the reactive oxygen species (ROS) produced by PDT were responsible for the induction of autophagy, which could be blocked by the ROS inhibitor N-acetyl cysteine (NAC). NAC also reversed the PDT-induced suppression of p-mTOR and p-Akt. Therefore, our findings demonstrate that PDT promotes cholesterol efflux by inducing autophagy, and the autophagy was mediated in part through the ROS/PI3K/Akt/mTOR signaling pathway in THP-1 and peritoneal macrophage-derived foam cells.


Atherosclerosis , Autophagy/drug effects , Cholesterol/metabolism , Foam Cells/metabolism , Macrophages, Peritoneal/metabolism , Nanoparticles/chemistry , Photochemotherapy/methods , Reactive Oxygen Species/metabolism , Animals , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Atherosclerosis/pathology , Foam Cells/pathology , Humans , Macrophages, Peritoneal/pathology , Mice , THP-1 Cells
13.
Oxid Med Cell Longev ; 2017: 8519169, 2017.
Article En | MEDLINE | ID: mdl-28191279

Monocyte-derived macrophages participate in infaust inflammatory responses by secreting various types of proinflammatory factors, resulting in further inflammatory reactions in atherosclerotic plaques. Autophagy plays an important role in inhibiting inflammation; thus, increasing autophagy may be a therapeutic strategy for atherosclerosis. In the present study, hydroxysafflor yellow A-mediated sonodynamic therapy was used to induce autophagy and inhibit inflammation in THP-1 macrophages. Following hydroxysafflor yellow A-mediated sonodynamic therapy, autophagy was induced as shown by the conversion of LC3-II/LC3-I, increased expression of beclin 1, degradation of p62, and the formation of autophagic vacuoles. In addition, inflammatory factors were inhibited. These effects were blocked by Atg5 siRNA, the autophagy inhibitor 3-methyladenine, and the reactive oxygen species scavenger N-acetyl cysteine. Moreover, AKT phosphorylation at Ser473 and mTOR phosphorylation at Ser2448 decreased significantly after HSYA-SDT. These effects were inhibited by the PI3K inhibitor LY294002, the AKT inhibitor triciribine, the mTOR inhibitor rapamycin, mTOR siRNA, and N-acetyl cysteine. Our results demonstrate that HSYA-SDT induces an autophagic response via the PI3K/Akt/mTOR signaling pathway and inhibits inflammation by reactive oxygen species in THP-1 macrophages.


Autophagy/drug effects , Chalcone/analogs & derivatives , Macrophages/drug effects , Quinones/pharmacology , Reactive Oxygen Species/metabolism , Ultrasonic Therapy/methods , Blotting, Western , Cell Line , Cell Survival/drug effects , Chalcone/pharmacology , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Microscopy, Electron, Transmission , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , TOR Serine-Threonine Kinases/metabolism
14.
Cell Death Dis ; 8(1): e2558, 2017 01 19.
Article En | MEDLINE | ID: mdl-28102849

Impaired autophagy in macrophages accompanies the progression of atherosclerosis and contributes to lipid loading in plaques and ineffective lipid degradation. Therefore, evoking autophagy and its associated cholesterol efflux may provide a therapeutic treatment for atherosclerosis. In the present study, berberine-mediated sonodynamic therapy (BBR-SDT) was used to induce autophagy and cholesterol efflux in THP-1 macrophages and derived foam cells. Following BBR-SDT, autophagy was increased in the macrophages, autophagy resistance in the foam cells was prevented, and cholesterol efflux was induced. The first two effects were blocked by the reactive oxygen species scavenger, N-acetyl cysteine. BBR-SDT also reduced the phosphorylation of Akt and mTOR, two key molecules in the PI3K/AKT/mTOR signaling pathway, which is responsible for inducing autophagy. Correspondingly, treatment with the autophagy inhibitor, 3-methyladenine, or the PI3K inhibitor, LY294002, abolished the autophagy-induced effects of BBR-SDT. Furthermore, induction of cholesterol efflux by BBR-SDT was reversed by an inhibition of autophagy by 3-methyladenine or by a small interfering RNA targeting Atg5. Taken together, these results demonstrate that BBR-SDT effectively promotes cholesterol efflux by increasing reactive oxygen species generation, and this subsequently induces autophagy via the PI3K/AKT/mTOR signaling pathway in both 'normal' macrophages and lipid-loaded macrophages (foam cells). Thus, BBR-SDT may be a promising atheroprotective therapy to inhibit the progression of atherosclerosis and should be further studied.


Atherosclerosis/drug therapy , Autophagy/drug effects , Cholesterol/metabolism , Atherosclerosis/genetics , Atherosclerosis/pathology , Autophagy/genetics , Berberine/administration & dosage , Cell Survival/drug effects , Cholesterol/genetics , Chromones/administration & dosage , Humans , Lipids/biosynthesis , Macrophages/drug effects , Macrophages/metabolism , Morpholines/administration & dosage , Oncogene Protein v-akt , Phosphatidylinositol 3-Kinases/genetics , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/genetics , Ultrasonography
15.
Cell Death Dis ; 7(12): e2527, 2016 12 22.
Article En | MEDLINE | ID: mdl-28005078

Lipid catabolism disorder is the primary cause of atherosclerosis. Transcription factor EB (TFEB) prevents atherosclerosis by activating macrophage autophagy to promote lipid degradation. Hypericin-mediated sonodynamic therapy (HY-SDT) has been proved non-invasively inducing THP-1-derived macrophage apoptosis; however, it is unknown whether macrophage autophagy could be triggered by HY-SDT to influence cellular lipid catabolism via regulating TFEB. Here, we report that HY-SDT resulted in the time-dependent THP-1-derived macrophage autophagy activation through AMPK/AKT/mTOR pathway. Besides, TFEB nuclear translocation in macrophage was triggered by HY-SDT to promote autophagy activation and lysosome regeneration which enhanced lipid degradation in response to atherogenic lipid stressors. Moreover, following HY-SDT, the ABCA1 expression level was increased to promote lipid efflux in macrophage, and the expression levels of CD36 and SR-A were decreased to inhibit lipid uptake, both of which were prevented by TFEB knockdown. These results indicated that TFEB nuclear translocation activated by HY-SDT was not only the key regulator of autophagy activation and lysosome regeneration in macrophage to promote lipolysis, but also had a crucial role in reverse cholesterol transporters to decrease lipid uptake and increase lipid efflux. Reactive oxygen species (ROS) were adequately generated in macrophage by HY-SDT. Further, ROS scavenger N-acetyl-l-cysteine abolished HY-SDT-induced TFEB nuclear translocation and autophagy activation, implying that ROS were the primary upstream factors responsible for these effects during HY-SDT. In summary, our data indicate that HY-SDT decreases lipid content in macrophage by promoting ROS-dependent nuclear translocation of TFEB to influence consequent autophagy activation and cholesterol transporters. Thus, HY-SDT may be beneficial for atherosclerosis via TFEB regulation to ameliorate lipid overload in atherosclerotic plaques.


Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Lipids/chemistry , Macrophages/cytology , Macrophages/metabolism , Perylene/analogs & derivatives , Reactive Oxygen Species/metabolism , Ultrasonics , ATP Binding Cassette Transporter 1/metabolism , Adenylate Kinase/metabolism , Anthracenes , Apoptosis/drug effects , CD36 Antigens/metabolism , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytoprotection/drug effects , Humans , Lipid Metabolism/drug effects , Lysosomes/drug effects , Lysosomes/metabolism , Macrophages/drug effects , Macrophages/ultrastructure , Perylene/pharmacology , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Scavenger Receptors, Class A/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
16.
Cell Physiol Biochem ; 39(5): 1746-1760, 2016.
Article En | MEDLINE | ID: mdl-27744450

BACKGROUND/AIMS: Sonodynamic therapy (SDT) is considered a new approach for the treatment of atherosclerosis. We previously confirmed that hydroxyl acetylated curcumin (HAC) was a sonosensitizer. In this study, we investigated the mechanism of THP-1 macrophage apoptosis and autophagy induced by HAC mediated SDT (HAC-SDT). METHODS: Cell viability was measured using a CCK-8 assay. Laser scanning confocal microscopy was used to measure the levels of intracellular reactive oxygen species (ROS), sub-cellular HAC localization, BAX and cytochrome C translocation, LC3 expression, monodansylcadaverine staining and Dil-labeled oxidized low density lipoprotein (Dil-ox-LDL) uptake. Flow cytometry was used to analyze apoptosis and autophagy via Annexin V/propidium iodide and acridine orange staining, respectively. The expression levels of apoptosis- and autophagy-related proteins were detected by Western blot. Oil red O was used to measure intracellular lipid accumulation. RESULTS: We identified HAC (5.0 µg/mL) located in lysosomes, endoplasmic reticulum, Golgi apparatus and mitochondria after 4 h of incubation. Compared with other sonosensitizers (e.g., curcumin and emodin), HAC had a more obvious sonodynamic effect on macrophages. Furthermore, the mitochondrial-caspase pathway was confirmed to play a crucial role in the HAC-SDT-induced apoptosis; BAX translocated from the cytosol to the mitochondria during HAC-SDT. Subsequently, mitochondrial cytochrome C was released into the cytosol, activating the caspase cascade in a time-dependent manner. Furthermore, HAC-SDT could induce PI3K/AKT/mTOR pathway dependent autophagy, accompanied by a decrease in the lipid uptake of THP-1 macrophages. This mechanism was demonstrated by the formation of acidic vesicular organelles, the conversion of LC3 I to LC3 II, the expression of related proteins, and the attenuation of both Dil-ox-LDL and oil red O staining. Moreover, pre-treatment with the autophagy inhibitor 3-methyladenine enhanced the HAC-SDT-induced apoptosis. Additionally, HAC-SDT-induced autophagy and apoptosis were both blocked by ROS scavenger N-acetyl-l-cysteine. CONCLUSION: The results suggested that autophagy not only played an inhibitory role in the process of apoptosis but also could effectively attenuate lipid aggregation in THP-1 macrophages during HAC-SDT. As important intracellular mediators, the ROS generated by HAC-SDT also played a crucial role in initiating apoptosis and autophagy.


Autophagy/drug effects , Curcumin/pharmacology , Endoplasmic Reticulum/drug effects , Golgi Apparatus/drug effects , Lysosomes/drug effects , Macrophages/drug effects , Mitochondria/drug effects , Acetylation , Autophagy/genetics , Autophagy/radiation effects , Cell Line , Cell Survival/drug effects , Cell Survival/radiation effects , Curcumin/analogs & derivatives , Cytochromes c/genetics , Cytochromes c/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/radiation effects , Endoplasmic Reticulum/ultrastructure , Gene Expression , Golgi Apparatus/metabolism , Golgi Apparatus/radiation effects , Golgi Apparatus/ultrastructure , Humans , Hydroxylation , Lipids/chemistry , Lipoproteins, LDL/metabolism , Lysosomes/metabolism , Lysosomes/radiation effects , Lysosomes/ultrastructure , Macrophages/cytology , Macrophages/metabolism , Macrophages/radiation effects , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Mitochondria/radiation effects , Mitochondria/ultrastructure , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Ultrasonic Waves , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
17.
Cell Physiol Biochem ; 38(2): 545-57, 2016.
Article En | MEDLINE | ID: mdl-26828938

BACKGROUND/AIMS: Pseudohypericin (P-HY) and its congener hypericin are the major hydroxylated phenanthroperylenediones present in Hypericum species. Our previous study indicated that hypericin was able to function as a sonosensitizer. The potential use of P-HY as a sonosensitizer for sonodynamic therapy (SDT) requires further exploration. Thus, this study aimed to investigate the effects of P-HY-SDT on THP-1 macrophages. METHODS: THP-1 macrophages were incubated with P-HY, and cell viability was measured using a CCK-8 assay. Fluorescence microscopy assessed the intracellular reactive oxygen species (ROS), mitochondrial membrane potential (ΔΨm ) and mitochondrial permeability transition pore (mPTP) opening. Apoptotic and necrotic cell levels were measured by the flow cytometry analysis. Western blots were employed to assay BAX, Cytochrome C expression and apoptosis-related proteins. RESULTS: P-HY-SDT induced THP-1 macrophage apoptosis. The levels of ROS were significantly increased in the SDT group, resulting in both mPTP opening and ΔΨm loss, which led to apoptosis. In addition, the translocation of BAX, release of Cytochrome C and the upregulated expression of apoptosis-related proteins after P-HY-SDT were observed, all of which were reversed by N-acetyl cysteine (NAC). CONCLUSION: P-HY-SDT induced THP-1 macrophage apoptosis through the mitochondria-caspase pathway via ROS generation, the translocation of BAX and the release of Cytochrome C to regulate the mPTP opening.


Apoptosis/drug effects , Caspases/metabolism , Macrophages/drug effects , Mitochondria/drug effects , Perylene/analogs & derivatives , Cell Line , Cell Survival/drug effects , Cytochromes c/metabolism , Humans , Hypericum/chemistry , Macrophages/metabolism , Macrophages/pathology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Perylene/chemistry , Perylene/pharmacology , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Plaque, Atherosclerotic/therapy , Reactive Oxygen Species/metabolism , Ultrasonic Therapy
18.
Int J Nanomedicine ; 10: 3719-36, 2015.
Article En | MEDLINE | ID: mdl-26045663

Atherosclerosis (AS) is the most vital cardiovascular disease, which poses a great threat to human health. Macrophages play an important role in the progression of AS. Photodynamic therapy (PDT) has emerged as a useful therapeutic modality not only in the treatment of cancer but also in the treatment of AS. The purpose of this study was to determine the molecular mechanisms underlying the activity of PDT, using mesoporous-silica-coated upconversion fluorescent nanoparticles encapsulating chlorin e6 (UCNPs-Ce6) in the induction of apoptosis in THP-1 macrophages. Here, we investigated the ability of UCNPs-Ce6-mediated PDT to induce THP-1 macrophage apoptosis by facilitating the induction of reactive oxygen species (ROS) and regulation of mitochondrial permeability transition pore (MPTP) to depolarize mitochondrial membrane potential (MMP). Both Bax translocation and the release of cytochrome C were examined using immunofluorescence and Western blotting. Our results indicated that the levels of ROS were significantly increased in the PDT group, resulting in both MPTP opening and MMP depolarization, which led to apoptosis. In addition, immunofluorescence and Western blotting revealed that PDT induced both Bax translocation and the release of cytochrome C, as well as upregulation of cleaved caspase-9, cleaved caspase-3, and cleaved poly(ADP-ribose) polymerase. Therefore, we demonstrated that UCNPs-Ce6-mediated PDT induces apoptosis in THP-1 macrophages via ROS bursts. The proapoptotic factor Bax subsequently translocates from the cytosol to the mitochondria, resulting in the MPTP opening and cytochrome C release. This study demonstrated the great potential of UCNPs-Ce6-mediated PDT in the treatment of AS.


Apoptosis/drug effects , Mitochondria/drug effects , Nanoparticles/chemistry , Photochemotherapy/methods , Porphyrins/chemistry , Reactive Oxygen Species/metabolism , Caspases/metabolism , Cell Line/drug effects , Cell Line/metabolism , Chlorophyllides , Cytochromes c/metabolism , Cytosol/drug effects , Cytosol/metabolism , Humans , Macrophages/drug effects , Macrophages/metabolism , Membrane Potential, Mitochondrial/drug effects , Metabolic Networks and Pathways/drug effects , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/drug effects , Mitochondrial Permeability Transition Pore , Nanoparticles/therapeutic use , Poly(ADP-ribose) Polymerases/metabolism , Porphyrins/pharmacology , Radiation-Sensitizing Agents/chemistry , Radiation-Sensitizing Agents/pharmacology
19.
Int J Nanomedicine ; 10: 821-38, 2015.
Article En | MEDLINE | ID: mdl-25653524

PURPOSE: To investigate the sonoactivity of hypericin (HY), together with its sonodynamic effect on THP-1 macrophages and the underlying mechanism. MATERIALS AND METHODS: CCK-8 was used to examine cell viability. Confocal laser scanning microscopy was performed to assess the localization of HY in cells, reactive oxygen species (ROS) generation, and opening of the mitochondrial permeability transition pore (mPTP) after different treatments. Apoptosis was analyzed using Hoechst-propidium iodide and transmission electron microscopy. Mitochondrial membrane potential (ΔΨm) collapse was detected via fluorescence microscopy. Lipoprotein oxidation was determined in malondialdehyde (MDA) assays. Western blotting was conducted to determine the translocation of BAX and cytochrome C and the expression of apoptosis-related proteins. RESULTS: HY was sublocalized among the nuclei and the mitochondria, endoplasmic reticulum, Golgi apparatus, and lysosome in the cytosol of THP-1 macrophages. Under low-intensity ultrasound irradiation, HY significantly decreased cell viability and induced apoptosis. Furthermore, greater ROS generation, higher MDA levels, and greater ΔΨm loss were observed in the sonodynamic therapy (SDT) group. Both ROS generation and MDA levels were significantly reduced by the ROS scavenger N-acetyl cysteine (NAC) and the singlet oxygen scavenger sodium azide. Most of the loss of ΔΨm was inhibited by pretreatment with NAC, sodium azide, and the mPTP inhibitor cyclosporin A (CsA). mPTP opening was induced upon SDT but was reduced by pretreatment with bongkrekic acid, 4,4'-diisothiocyanatostilbene-2,2'-disulfonic acid disodium, CsA, and NAC. Western blot analyses revealed translocation of BAX and cytochrome C, downregulated expression of Bcl-2, and upregulated expression of cleaved caspase-9, cleaved caspase-3, and cleaved poly(ADP-ribose) polymerase in the SDT group, which were reversed by NAC. CONCLUSION: HY mediated SDT-induced apoptosis in THP-1 macrophages via ROS generation. Then, the proapoptotic factor BAX translocated from the cytosol to the mitochondria, increasing the ratio of BAX/Bcl-2, and the mPTP opened to release cytochrome C. This study demonstrated the great potential of HY-mediated SDT for treating atherosclerosis.


Apoptosis/drug effects , Macrophages/pathology , Mitochondria/pathology , Perylene/analogs & derivatives , Radiation-Sensitizing Agents/pharmacology , Ultrasonic Therapy , Anthracenes , Apoptosis/radiation effects , Blotting, Western , Caspase 3/metabolism , Caspase 9/metabolism , Cell Survival/drug effects , Cell Survival/radiation effects , Cytochromes c/metabolism , Cytosol/metabolism , Humans , Macrophages/drug effects , Macrophages/radiation effects , Membrane Potential, Mitochondrial/drug effects , Microscopy, Electron, Transmission , Mitochondria/metabolism , Perylene/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Reactive Oxygen Species/metabolism
...