Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 469
Filter
1.
Nat Commun ; 13(1): 91, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013181

ABSTRACT

Pursuing and developing effective methodologies to construct highly active catalytic sites to maximize the atomic and energy efficiency by material engineering are attractive. Relative to the tremendous researches of carbon-based single atom systems, the construction of bio-applicable single atom materials is still in its infancy. Herein, we propose a facile and general interfacial-confined coordination strategy to construct high-quality single-atom nanotherapeutic agent with Fe single atoms being anchored on defective carbon dots confined in a biocompatible mesoporous silica nanoreactor. Furthermore, the efficient energy conversion capability of silica-based Fe single atoms system has been demonstrated on the basis of the exogenous physical photo irradiation and endogenous biochemical reactive oxygen species stimulus in the confined mesoporous network. More importantly, the highest photothermal conversion efficiency with the mechanism of increased electron density and narrow bandgap of this single atom structure in defective carbon was proposed by the theoretical DFT calculations. The present methodology provides a scientific paradigm to design and develop versatile single atom nanotherapeutics with adjustable metal components and tune the corresponding reactions for safe and efficient tumor therapeutic strategy.


Subject(s)
Carcinoma, Hepatocellular/therapy , Ferrosoferric Oxide/chemistry , Liver Neoplasms/therapy , Metal Nanoparticles/administration & dosage , Photothermal Therapy/methods , Theranostic Nanomedicine/methods , Animals , Carbon/chemistry , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Female , Glutathione/chemistry , Humans , Light , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Metal Nanoparticles/chemistry , Metal Nanoparticles/ultrastructure , Mice , Mice, Inbred BALB C , Quantum Theory , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Silicon Dioxide/chemistry , Xenograft Model Antitumor Assays
2.
Biochem Biophys Res Commun ; 587: 99-106, 2022 01 08.
Article in English | MEDLINE | ID: mdl-34872005

ABSTRACT

Colorectal cancer (CRC) is one of the most common malignant tumors in the digestive system, and Chinese herbal medicine plays an important role in tumor treatment. The in-depth study of auriculasin isolated from Flemingia philippinensis showed that auriculasin promoted reactive oxygen species (ROS) generation in a concentration-dependent manner; when ROS scavenger NAC was added, the effects of auriculasin in promoting ROS generation and inhibiting cell viability were blocked. Auriculasin induced CRC cell apoptosis, led to mitochondrial shrinkage, and increased the intracellular accumulation of Fe2+ and MDA. When auriculasin and NAC were added simultaneously, the levels of apoptosis, Fe2+ and MDA returned to the control group levels, indicating that auriculasin activated apoptosis and ferroptosis by inducing ROS generation. In addition, auriculasin promoted the expression of Keap1 and AIFM1, but significantly reduced the phosphorylation level of AIFM1, while NAC significantly blocked the regulation of Keap1 and AIFM1 by auriculasin, which indicates that auriculasin can also induce oxeiptosis through ROS. When Z-VAD-FMK, Ferrostatin-1, Keap1 siRNA, PGAM5 siRNA and AIFM1 siRNA were added respectively, the inhibitory effect of auriculasin on cell viability was significantly weakened, indicating that auriculasin inhibits cell viability by inducing apoptosis, ferroptosis and oxeiptosis. Auriculasin also inhibited the invasion and clone forming ability of CRC cells, while NAC blocked the above effects of auriculasin. Therefore, auriculasin can promote CRC cell apoptosis, ferroptosis and oxeiptosis by inducing ROS generation, thereby inhibiting cell viability, invasion and clone formation, indicating that auriculasin has a significant antitumor effect.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Ferroptosis/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Isoflavones/pharmacology , Reactive Oxygen Species/agonists , Antineoplastic Agents, Phytogenic/isolation & purification , Apoptosis/genetics , Apoptosis Inducing Factor/genetics , Apoptosis Inducing Factor/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Fabaceae/chemistry , Ferroptosis/genetics , HCT116 Cells , Humans , Iron/agonists , Iron/metabolism , Isoflavones/isolation & purification , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Malondialdehyde/agonists , Malondialdehyde/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Models, Biological , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Plant Extracts/chemistry , Reactive Oxygen Species/metabolism
3.
Arch Pharm Res ; 44(11): 1025-1036, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34751932

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most common tumor types globally. Despite the progress made in surgical procedures and therapeutic options, HCC remains a considerable cause of cancer-related mortality. In this study, we investigated the antitumor effects of sanguinarine (Sang) on HCC and its potential mechanisms. Our findings showed that Sang impairs the acidic environment of lysosomes by inhibiting cathepsin D maturation. In addition, Sang inhibited the formation of autolysosomes in RFP-GFP-LC3 transfected cells, subsequently suppressing late mitophagy. Sang also induced reactive oxygen species (ROS)-dependent autophagy and apoptosis in HCC cells, which was significantly attenuated following treatment with a ROS scavenger. Further investigation using autophagy inhibitors revealed that sanguinarine-induced mitochondrial dysfunction and mitophagy led to mitochondrial apoptosis in HCC cells. Immunohistochemical staining of sanguinarine-treated xenograft samples revealed that it initiated and blocked autophagy. In summary, our findings suggest that in HCC cells, Sang impairs lysosomal function and induces ROS-dependent mitophagy and apoptosis.


Subject(s)
Benzophenanthridines/pharmacology , Carcinoma, Hepatocellular/drug therapy , Isoquinolines/pharmacology , Liver Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Benzophenanthridines/therapeutic use , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Humans , Isoquinolines/therapeutic use , Liver Neoplasms/pathology , Lysosomes/drug effects , Mice , Mitochondria/drug effects , Mitochondria/pathology , Mitophagy/drug effects , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
4.
Int J Mol Sci ; 22(22)2021 Nov 18.
Article in English | MEDLINE | ID: mdl-34830314

ABSTRACT

The kidney is one of the most susceptible organs to age-related impairments. Generally, renal aging is accompanied by renal fibrosis, which is the final common pathway of chronic kidney diseases. Aristolochic acid (AA), a nephrotoxic agent, causes AA nephropathy (AAN), which is characterized by progressive renal fibrosis and functional decline. Although renal fibrosis is associated with renal aging, whether AA induces renal aging remains unclear. The aim of the present study is to investigate the potential use of AAN as a model of renal aging. Here, we examined senescence-related factors in AAN models by chronically administering AA to C57BL/6 mice. Compared with controls, the AA group demonstrated aging kidney phenotypes, such as renal atrophy, renal functional decline, and tubulointerstitial fibrosis. Additionally, AA promoted cellular senescence specifically in the kidneys, and increased renal p16 mRNA expression and senescence-associated ß-galactosidase activity. Furthermore, AA-treated mice exhibited proximal tubular mitochondrial abnormalities, as well as reactive oxygen species accumulation. Klotho, an antiaging gene, was also significantly decreased in the kidneys of AA-treated mice. Collectively, the results of the present study indicate that AA alters senescence-related factors, and that renal fibrosis is closely related to renal aging.


Subject(s)
Aging/drug effects , Aristolochic Acids/pharmacology , Collagen/genetics , Kidney/drug effects , Nephritis, Interstitial/chemically induced , Renal Insufficiency, Chronic/chemically induced , Aging/genetics , Animals , Collagen/agonists , Collagen/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Disease Models, Animal , Fibrosis , Gene Expression Regulation , Humans , Kidney/metabolism , Kidney/pathology , Klotho Proteins/genetics , Klotho Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Nephritis, Interstitial/genetics , Nephritis, Interstitial/metabolism , Nephritis, Interstitial/pathology , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Signal Transduction , Transforming Growth Factor beta/agonists , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
5.
Nat Commun ; 12(1): 5311, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34493724

ABSTRACT

Although some effective therapies have been available for cancer, it still poses a great threat to human health and life due to its drug resistance and low response in patients. Here, we develop a ferroptosis-based therapy by combining iron nanoparticles and cancer-specific gene interference. The expression of two iron metabolic genes (FPN and LCN2) was selectively knocked down in cancer cells by Cas13a or microRNA controlled by a NF-κB-specific promoter. Cells were simultaneously treated by iron nanoparticles. As a result, a significant ferroptosis was induced in a wide variety of cancer cells. However, the same treatment had little effect on normal cells. By transferring genes with adeno-associated virus and iron nanoparticles, the significant tumor growth inhibition and durable cure were obtained in mice with the therapy. In this work, we thus show a cancer therapy based on gene interference-enhanced ferroptosis.


Subject(s)
Cation Transport Proteins/antagonists & inhibitors , Ferroptosis/genetics , Iron/metabolism , Lipocalin-2/antagonists & inhibitors , Neoplasms/therapy , RNA Interference , Reactive Oxygen Species/agonists , Animals , CRISPR-Associated Proteins/genetics , CRISPR-Associated Proteins/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Line, Tumor , Dependovirus/genetics , Dependovirus/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lipocalin-2/genetics , Lipocalin-2/metabolism , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Neoplasms/genetics , Neoplasms/mortality , Neoplasms/pathology , Promoter Regions, Genetic , Reactive Oxygen Species/metabolism , Signal Transduction , Spleen/metabolism , Spleen/pathology , Survival Analysis , Tumor Burden , Xenograft Model Antitumor Assays
6.
Cells ; 10(7)2021 07 02.
Article in English | MEDLINE | ID: mdl-34359834

ABSTRACT

Astrocytes act as neural stem cells (NSCs) that have the potential to self-renew and differentiate into other neuronal cells. The protein expression of these astrocytes depends on the stage of differentiation, showing sequential expression of multiple proteins such as octamer-binding transcription factor 4 (Oct4), nestin, glial fibrillary acidic protein (GFAP), and aldehyde dehydrogenase 1 family member L1 (aldh1L1). Photobiomodulation (PBM) affects cell apoptosis, proliferation, migration, and adhesion. We hypothesized that astrocyte proliferation and differentiation would be modulated by PBM. We used an optimized astrocyte culture method and a 660-nanometer light-emitting diode (LED) to enhance the biological actions of many kinds of cells. We determined that the 660-nanometer LED promoted the biological actions of cultured astrocytes by increasing the reactive oxygen species levels. The overall viability of the cultured cells, which included various cells other than astrocytes, did not change after LED exposure; however, astrocyte-specific proliferation was observed by the increased co-expression of GFAP and bromodeoxyuridine (BrdU)/Ki67. Furthermore, the 660-nanometer LED provides evidence of differentiation, as shown by the decreased Oct4 and GFAP co-expression and increased nestin and aldh1L1 expression. These results demonstrate that a 660-nanometer LED can modify astrocyte proliferation, which suggests the efficacy of the therapeutic application of LED in various pathological states of the central nervous system.


Subject(s)
Astrocytes/radiation effects , Cell Proliferation/radiation effects , Gene Expression/radiation effects , Neurons/radiation effects , Animals , Apoptosis/genetics , Apoptosis/radiation effects , Astrocytes/cytology , Astrocytes/metabolism , Brain/cytology , Brain/metabolism , Cell Adhesion/radiation effects , Cell Differentiation/radiation effects , Cell Movement/radiation effects , Coculture Techniques , Embryo, Mammalian , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Lasers, Semiconductor , Light , Nestin/genetics , Nestin/metabolism , Neurons/cytology , Neurons/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Oxidoreductases Acting on CH-NH Group Donors/genetics , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism
7.
J Cell Biol ; 220(9)2021 09 06.
Article in English | MEDLINE | ID: mdl-34328510

ABSTRACT

Ferroptosis is a form of iron-dependent regulated cell death driven by uncontrolled lipid peroxidation. Mitochondria are double-membrane organelles that have essential roles in energy production, cellular metabolism, and cell death regulation. However, their role in ferroptosis has been unclear and somewhat controversial. In this Perspective, I summarize the diverse metabolic processes in mitochondria that actively drive ferroptosis, discuss recently discovered mitochondria-localized defense systems that detoxify mitochondrial lipid peroxides and protect against ferroptosis, present new evidence for the roles of mitochondria in regulating ferroptosis, and outline outstanding questions on this fascinating topic for future investigations. An in-depth understanding of mitochondria functions in ferroptosis will have important implications for both fundamental cell biology and disease treatment.


Subject(s)
Ferroptosis/genetics , GTP Cyclohydrolase/antagonists & inhibitors , Iron/metabolism , Mitochondria/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Amino Acid Transport System y+/antagonists & inhibitors , Amino Acid Transport System y+/genetics , Amino Acid Transport System y+/metabolism , Cells, Cultured , Ferroptosis/drug effects , GTP Cyclohydrolase/genetics , GTP Cyclohydrolase/metabolism , Gene Expression Regulation , Glutathione/antagonists & inhibitors , Glutathione/biosynthesis , Humans , Lipid Peroxidation/drug effects , Mitochondria/drug effects , Mitochondria/enzymology , Oxidants/pharmacology , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , S100 Calcium-Binding Protein A4/antagonists & inhibitors , S100 Calcium-Binding Protein A4/genetics , S100 Calcium-Binding Protein A4/metabolism , Signal Transduction , Ubiquinone/antagonists & inhibitors , Ubiquinone/metabolism
8.
Int J Mol Sci ; 22(9)2021 May 04.
Article in English | MEDLINE | ID: mdl-34064489

ABSTRACT

Melanoma represents one of the most aggressive and drug resistant skin cancers with poor prognosis in its advanced stages. Despite the increasing number of targeted therapies, novel approaches are needed to counteract both therapeutic resistance and the side effects of classic therapy. Betulinic acid (BA) is a bioactive phytocompound that has been reported to induce apoptosis in several types of cancers including melanomas; however, its effects on mitochondrial bioenergetics are less investigated. The present study performed in A375 human melanoma cells was aimed to characterize the effects of BA on mitochondrial bioenergetics and cellular behavior. BA demonstrated a dose-dependent inhibitory effect in both mitochondrial respiration and glycolysis in A375 melanoma cells and at sub-toxic concentrations (10 µM) induced mitochondrial dysfunction by eliciting a decrease in the mitochondrial membrane potential and changes in mitochondria morphology and localization. In addition, BA triggered a dose-dependent cytotoxic effect characterized by apoptotic features: morphological alterations (nuclear fragmentation, apoptotic bodies) and the upregulation of pro-apoptotic markers mRNA expression (Bax, Bad and Bak). BA represents a viable therapeutic option via a complex modulatory effect on mitochondrial metabolism that might be useful in advanced melanoma or as reliable strategy to counteract resistance to standard therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Melanocytes/drug effects , Mitochondria/drug effects , Pentacyclic Triterpenes/pharmacology , Reactive Oxygen Species/metabolism , Apoptosis/genetics , Cell Line, Tumor , Gene Expression Regulation , Glycolysis/drug effects , Glycolysis/genetics , Humans , Inhibitory Concentration 50 , Melanocytes/metabolism , Melanocytes/pathology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Oxidative Phosphorylation/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/agonists , Signal Transduction , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , bcl-Associated Death Protein/genetics , bcl-Associated Death Protein/metabolism , Betulinic Acid
9.
Cells ; 10(4)2021 04 13.
Article in English | MEDLINE | ID: mdl-33924665

ABSTRACT

Lithium hexafluorophosphate (LiPF6) is one of the leading electrolytes in lithium-ion batteries, and its usage has increased tremendously in the past few years. Little is known, however, about its potential environmental and biological impacts. In order to improve our understanding of the cytotoxicity of LiPF6 and the specific cellular response mechanisms to it, we performed a genome-wide screen using a yeast (Saccharomyces cerevisiae) deletion mutant collection and identified 75 gene deletion mutants that showed LiPF6 sensitivity. Among these, genes associated with mitochondria showed the most enrichment. We also found that LiPF6 is more toxic to yeast than lithium chloride (LiCl) or sodium hexafluorophosphate (NaPF6). Physiological analysis showed that a high concentration of LiPF6 caused mitochondrial damage, reactive oxygen species (ROS) accumulation, and ATP content changes. Compared with the results of previous genome-wide screening for LiCl-sensitive mutants, we found that oxidative phosphorylation-related mutants were specifically hypersensitive to LiPF6. In these deletion mutants, LiPF6 treatment resulted in higher ROS production and reduced ATP levels, suggesting that oxidative phosphorylation-related genes were important for counteracting LiPF6-induced toxicity. Taken together, our results identified genes specifically involved in LiPF6-modulated toxicity, and demonstrated that oxidative stress and ATP imbalance maybe the driving factors in governing LiPF6-induced toxicity.


Subject(s)
Fluorides/toxicity , Lithium/toxicity , Mitochondria/drug effects , Oxidative Phosphorylation/drug effects , Phosphates/toxicity , Saccharomyces cerevisiae/drug effects , Adaptation, Physiological/drug effects , Adenosine Triphosphate/antagonists & inhibitors , Adenosine Triphosphate/biosynthesis , Gene Expression Regulation, Fungal/drug effects , Gene Ontology , Genome-Wide Association Study , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Molecular Sequence Annotation , Oxidative Stress , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
10.
Biosci Biotechnol Biochem ; 85(5): 1097-1103, 2021 Apr 24.
Article in English | MEDLINE | ID: mdl-33784737

ABSTRACT

p62/sequestosome is a multifunctional adaptor protein that participates in a wide variety of cellular processes. 20(S)-Ginsenoside Rh2 (G-Rh2) has various biological effects, including anticancer activity. We found that G-Rh2 can induce apoptosis and autophagy in HeLa cells. G-Rh2 significantly enhanced the transcriptional level of p62. A siRNA was constructed to knock down p62 and assess its effect on apoptosis induced by G-Rh2. p62 protein levels were successfully downregulated in cells transfected with the p62-specific siRNA. Silencing of p62 further decreased cell viability while also enhancing cell apoptosis, reactive oxygen species generation, the ratio of Bax to Bcl-2, and the cleavage of PARP. p62 knockdown decreased expression levels of Nrf2. Moreover, silencing of p62 had no significant effect on autophagy induced by G-Rh2. These results suggest that combining G-Rh2 treatment with inhibition of p62 may be a potential treatment strategy for cervical cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Ginsenosides/pharmacology , Sequestosome-1 Protein/genetics , Apoptosis/genetics , Autophagy , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Dose-Response Relationship, Drug , HeLa Cells , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Sequestosome-1 Protein/antagonists & inhibitors , Sequestosome-1 Protein/metabolism , Signal Transduction , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
11.
Mol Biochem Parasitol ; 242: 111364, 2021 03.
Article in English | MEDLINE | ID: mdl-33639230

ABSTRACT

Inhibition of an imperative antioxidant enzyme with subsequent death is a victorious and widely accepted strategy to combat various infectious diseases. Among different antioxidant enzymes, thioredoxin reductase (TrxR) is an exclusive one. Studies have revealed that direct inhibition of TrxR by different classes of chemical moieties promptly results in the death of an organism. Especially the structural as well as biochemical modifications of the enzyme upon inhibition project serious threat towards the subject organism. Herein, an attempt was made to inhibit TrxR of filarial species by administering Auranofin, 1 chloro 2,4 dinitrobenzene (CDNB), Curcumin, and a novel carbamo dithioperoxo(thioate) derivative (4a). Our study has revealed that inhibition of TrxR resulted in the induction of the classical CED pathway of apoptosis along with the intrinsic and extrinsic pathways of apoptosis (Caspase mediated) routed through the ASK-1/p38 axis. Druggability analysis of filarial TrxR for the selected compounds was performed in silico through molecular docking studies. Therefore, this study attempts to decipher the mechanism of apoptosis induction following TrxR inhibition. The safety of those four compounds in terms of dose and toxicity was taken under consideration. Thitherto, the mechanism of TrxR mediated initiation of cell death in filarial parasite has remained undercover, and therefore, it is a maiden report on the characterization of apoptosis induction upon TrxR inhibition which will eventually help in generating effective antifilarial drugs in the future.


Subject(s)
Anthelmintics/pharmacology , Auranofin/pharmacology , Caspases/genetics , Curcumin/pharmacology , Dinitrochlorobenzene/pharmacology , Setaria Nematode/drug effects , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Animals , Anthelmintics/chemistry , Apoptosis/drug effects , Apoptosis/genetics , Auranofin/chemistry , Binding Sites , Caspases/metabolism , Cattle , Curcumin/chemistry , Dinitrochlorobenzene/chemistry , Gene Expression Regulation , Helminth Proteins/genetics , Helminth Proteins/metabolism , MAP Kinase Kinase Kinase 5/genetics , MAP Kinase Kinase Kinase 5/metabolism , Microfilariae/drug effects , Microfilariae/enzymology , Microfilariae/growth & development , Models, Molecular , Oxidative Stress , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Setaria Nematode/enzymology , Setaria Nematode/growth & development , Signal Transduction , Thioredoxin-Disulfide Reductase/chemistry , Thioredoxin-Disulfide Reductase/genetics , Thioredoxin-Disulfide Reductase/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Carbohydr Polym ; 256: 117521, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33483042

ABSTRACT

In our continuous searching for natural active polysaccharides with immunomodulatory activity, an arabinofuranan (AQP70-3) was isolated and purified from the fruits of Akebia quinata (Houtt.) Decne. by using ion-exchange chromatography and gel permeation chromatography for the first time. AQP70-3 contained both α-l-Araf and ß-l-Araf, and the absolute molecular weight was 1.06 × 104 g/mol. The backbone of AQP70-3 comprised →5)-α-l-Araf-(1→, →3,5)-α-l-Araf-(1→, and →2,5)-α-l-Araf-(1→, with branches of →1)-ß-l-Arafand →3)-α-l-Araf-(1→ residues. Biological assay suggested that AQP70-3 can stimulate phagocytic activity and promote the levels of nitric oxide (NO), interleukin (IL)-6, IL-1ß, and tumor necrosis factor-α (TNF-α) of RAW264.7 cells. Furthermore, AQP70-3 was found to increase the production of reactive oxygen species (ROS) and NO in zebrafish embryo model.


Subject(s)
Fruit/chemistry , Immunologic Factors/chemistry , Polysaccharides/chemistry , Ranunculales/chemistry , Reactive Oxygen Species/agonists , Animals , Carbohydrate Sequence , Embryo, Nonmammalian , Immunologic Factors/isolation & purification , Immunologic Factors/pharmacology , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Mice , Molecular Weight , Nitric Oxide/immunology , Nitric Oxide/metabolism , Phagocytosis/drug effects , Plant Extracts/chemistry , Polysaccharides/isolation & purification , Polysaccharides/pharmacology , RAW 264.7 Cells , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Stereoisomerism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Zebrafish
13.
Carbohydr Polym ; 255: 117490, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33436250

ABSTRACT

To deliver photosensitizers with PEGylated heparin (HP) into tumor cells for photodynamic therapy, we prepared two polyethylene glycol (PEG)-functionalized HP-based polymers conjugated with pyropheophorbide-a (Ppa): a non-GSH-responsive nanoagent (HP-Ppa-mPEG) with the mPEG moiety chemically attached to HP directly; and a GSH-responsive nanoagent (HP-Ppa-SS-mPEG) with the mPEG moiety conjugated to HP via a disulfide linkage. The Ppa-functionalized HP without PEGylation (HP-Ppa) was designed as another control. These amphiphilic polymers could aggregate into nanoparticles. Cellular uptake of three nanoparticles by 4T1 cells led to abundant production of reactive oxygen species after irradiation by a 660 nm laser, inducing cell apoptosis. HP-Ppa-SS-mPEG was found to achieve the highest tumor accumulation, the longest retention time and the best penetration into tumor tissues, resulting in the highest in vivo anticancer efficacy with 94.3 % tumor growth inhibition rate, suggesting that tumor microenvironment-responsive PEGylated HP-based nanomedicines may act as efficient anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Chlorophyll/analogs & derivatives , Heparin/chemistry , Mammary Neoplasms, Experimental/drug therapy , Photosensitizing Agents/pharmacology , Tumor Microenvironment/drug effects , Animals , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chlorophyll/chemistry , Female , Lasers , Light , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Nanoconjugates/chemistry , Photochemotherapy/methods , Photosensitizing Agents/chemical synthesis , Polyethylene Glycols/chemistry , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Treatment Outcome , Tumor Burden/drug effects
14.
Carbohydr Polym ; 254: 117459, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33357918

ABSTRACT

As a major therapeutic approach for cancer treatment, the effectiveness of chemotherapy is challenged by multidrug resistance (MDR). Herein, we fabricated novel redox-responsive, chondroitin sulfate-based nanoparticles that could simultaneously deliver quercetin (chemosensitizer), chlorin e6 (photosensitizer) and paclitaxel (chemotherapeutic agent) to exert enhanced chemo-photodynamic therapy for overcoming MDR and lung metastasis of breast cancer. In vitro cell study showed that nanoparticles down-regulated the expression of P-glycolprotein (P-gp) on MCF-7/ADR cells and thereby improved the anticancer efficacy of PTX against MCF-7/ADR cells. Moreover, NIR laser irradiation could induce nanoparticles to generate cellular reactive oxygen species (ROS), leading to mitochondrial membrane potential loss, and meanwhile facilitating lysosomal escape of drugs. Importantly, the novel nanoplatform exhibited effective in vivo MDR inhibition and anti-metastasis efficacy through enhanced chemo-photodynamic therapy. Thus, the study suggested that the multifunctional nanoplatform had good application prospect for effective breast cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/therapy , Drug Carriers , Drug Resistance, Neoplasm/radiation effects , Lung Neoplasms/therapy , Photosensitizing Agents/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Chlorophyllides , Chondroitin Sulfates/chemistry , Combined Modality Therapy , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Gene Expression , Humans , Infrared Rays/therapeutic use , Lasers , Lung Neoplasms/genetics , Lung Neoplasms/secondary , MCF-7 Cells , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Paclitaxel/pharmacology , Porphyrins/pharmacology , Quercetin/pharmacology , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
15.
FEBS J ; 288(1): 127-141, 2021 01.
Article in English | MEDLINE | ID: mdl-32338825

ABSTRACT

Most neurodegenerative disorders are associated with aggregation and accumulation of misfolded proteins. One of these proteins, tau, is involved in a number of pathologies including Alzheimer's disease and frontotemporal dementia. Aggregation and phosphorylation of tau have been shown to be a trigger for abnormal signal transduction and disruption of cellular homeostasis. Here, we have studied the effect of extracellular tau at different stages of aggregation in cortical co-cultures of neurons and astrocytes, to understand how this process affects tau pathogenicity. We found that the species formed after prolonged in vitro aggregation of tau (longer than 1 day) are able to stimulate reactive oxygen species (ROS) production through the activation of NADPH oxidase without decreasing the level of the endogenous antioxidant glutathione. The same late insoluble aggregates of tau induced calcium signals in neurons and a gradual increase in the ionic current of artificial membranes. Both tau-induced calcium signals and ROS production in NADPH oxidase were reduced in the presence of the inhibitor of voltage-gated calcium channels (VGCC) nifedipine. This suggests that insoluble aggregates of tau incorporate into the membrane and modify ionic currents, changing plasma membrane potential and activating VGCCs, which induces a calcium influx that triggers ROS production in NADPH oxidase. The combination of all these effects likely leads to toxicity, as only the same insoluble tau aggregates which demonstrated membrane-active properties produced neuronal cell death.


Subject(s)
Astrocytes/drug effects , Calcium Channels/genetics , Gene Expression Regulation/drug effects , NADPH Oxidases/genetics , Neurons/drug effects , tau Proteins/pharmacology , Amyloid beta-Peptides/agonists , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Animals, Newborn , Astrocytes/cytology , Astrocytes/metabolism , Calcium/metabolism , Calcium Channels/metabolism , Cell Death/drug effects , Coculture Techniques , Humans , NADPH Oxidases/metabolism , Neurons/cytology , Neurons/metabolism , Nifedipine/pharmacology , Oxidation-Reduction , Primary Cell Culture , Protein Aggregates , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Thapsigargin/pharmacology , Verapamil/pharmacology , tau Proteins/genetics , tau Proteins/metabolism
16.
J Gastrointest Cancer ; 52(1): 138-144, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32006341

ABSTRACT

BACKGROUND AND AIM: Sodium valproate (SV), a novel class of histone deacetylases (HDACs) inhibitors commonly used as an antiepileptic drug. HDAC inhibitors are known to possess anticancer potentials. In this study, we investigated the cytotoxic potential of SV in human hepatocellular carcinoma (HepG2 cells) cell line. METHODS: MTT assay was used to analyze cytotoxicity. Intracellular ROS and cytochrome c expression were analyzed by fluorescence microscopy. Morphology-related apoptosis was analyzed by dual staining with acridine orange/ethidium bromide. Caspase 3 protein expression was investigated by Western blotting analysis. RESULTS: Sodium valproate treatments in HepG2 cells caused significant and dose-dependent cytotoxicity. Intracellular ROS was remarkably increased in the cells which are treated with SV and caused early and late apoptosis as evidenced by dual staining. SV-treated cells expressed cytochrome c and caspase 3 protein expression. CONCLUSION: These results suggest the cytotoxic potentials of SV in HepG2 cells. This study may give an important clue for the inclusion of SV as an adjuvant along with standard anticancer agents after necessary in vivo and clinical studies.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Liver Neoplasms/drug therapy , Reactive Oxygen Species/agonists , Valproic Acid/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Hep G2 Cells , Histone Deacetylase Inhibitors/therapeutic use , Humans , Liver Neoplasms/pathology , Molecular Targeted Therapy/methods , Reactive Oxygen Species/metabolism , Valproic Acid/therapeutic use
17.
Int J Biochem Cell Biol ; 131: 105902, 2021 02.
Article in English | MEDLINE | ID: mdl-33309621

ABSTRACT

Fibrotic diseases account for more than 8 million deaths worldwide annually. Reactive oxygen species (ROS) has been shown to activate pyroptosis and promote the production of interleukin (IL)-1ß and IL-18, leading to fibrosis development. However, the role of dual oxidase 1 (DUOX1)-induced ROS production and pyroptosis in cardiac fibrosis remains largely unknown. Activin A was used to induce ROS and pyroptosis in cardiomyocytes. ROS level, pyroptosis, and cytokine production were detected using Active Oxygen Detection Kit, flow cytometry, and enzyme-linked immunosorbent assay, respectively. Western blotting analysis was used to measure expression changes of proteins. DUOX1 was silenced or overexpressed to investigate its role in fibrosis. We found that activin A induced ROS production and pyroptosis in cardiomyocytes, which was blocked by the ROS scavenger, N-acetyl-L-cysteine (NAC). Knockdown of DUOX1 reversed activin A-induced ROS production, pyroptosis, cytokine release, and the upregulation of proinflammatory proteins. Overexpression of DUOX1 resulted in opposite effects of knockdown DUOX1. Administration of an ROS scavenger blocked the effect of DUOX1 overexpression. Supplementation of IL-1ß and IL-18 caused significant fibrosis in human cardiac fibroblasts (hCFs). The knockdown of DUOX1 protected cardiomyocytes against activin A-induced fibrosis via the inhibition of ROS, cytokine release, and pyroptosis.


Subject(s)
Activins/pharmacology , Dual Oxidases/genetics , Myocytes, Cardiac/drug effects , Pyroptosis/drug effects , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Activins/antagonists & inhibitors , Caspase 1/genetics , Caspase 1/metabolism , Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type III/genetics , Collagen Type III/metabolism , Dual Oxidases/antagonists & inhibitors , Dual Oxidases/metabolism , Free Radical Scavengers/pharmacology , Gene Expression Regulation , Humans , Interleukin-18/genetics , Interleukin-18/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress/drug effects , Primary Cell Culture , Pyroptosis/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/agonists , Reactive Oxygen Species/antagonists & inhibitors , Signal Transduction , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism
18.
Article in English | MEDLINE | ID: mdl-33065288

ABSTRACT

We previously demonstrated that astaxanthin (ASTX), a xanthophyll carotenoid, repressed ethanol-induced inflammation and oxidative stress in macrophages. We explored the role of sirtuin 1 (SIRT1) and histone deacetylase 4 (HDAC4) in the inhibitory effect of ASTX on inflammation and oxidative stress in macrophages exposed to ethanol. Ethanol decreased mRNA and protein of SIRT1 while increasing those of HDAC4, which was attenuated by ASTX in RAW 264.7 macrophages and mouse bone marrow-derived macrophages (BMDMs). Inhibition of SIRT1 expression or activity augmented ethanol-induced Hdac4 expression, but SIRT1 activation elicited the opposite effect. Consistently, Hdac4 knockdown increased Sirt1 expression with decreases in ethanol-induced inflammatory gene expression, but its overexpression resulted in the opposite effects. Furthermore, BMDMs from mice with macrophage specific-deletion of Hdac4 (Hdac4MKO) showed significant decreases in ethanol-induced inflammatory genes and ROS accumulation but an increase in Sirt1 expression. Macrophage specific deletion of Hdac4 or ASTX abolished the changes in genes for mitochondrial biogenesis and glycolysis by ethanol. Ethanol increased mitochondrial respiration, ATP production, and proton leak, but decreased maximal respiration and spare respiratory capacity, all of which were abolished by ASTX in RAW 264.7 macrophages. The ethanol-induced alterations in mitochondrial respiration were abrogated in Hdac4MKO BMDMs. In conclusion, the anti-inflammatory and antioxidant properties of ASTX in ethanol-treated macrophages may be mediated, at least partly, by its opposite effect on SIRT1 and HDAC4 to empower SIRT1 to counteract ethanol-induced activation of HDAC4.


Subject(s)
Antioxidants/pharmacology , Ethanol/antagonists & inhibitors , Histone Deacetylases/genetics , Macrophages/drug effects , Sirtuin 1/genetics , Adenosine Triphosphate/biosynthesis , Animals , Ethanol/pharmacology , Gene Expression Regulation , Glycolysis/drug effects , Glycolysis/genetics , Histone Deacetylases/metabolism , Inflammation , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Knockout , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Organelle Biogenesis , Oxidative Phosphorylation/drug effects , Oxidative Stress , Primary Cell Culture , RAW 264.7 Cells , Reactive Oxygen Species/agonists , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Sirtuin 1/metabolism , Xanthophylls/pharmacology
19.
Sci Rep ; 10(1): 20348, 2020 11 23.
Article in English | MEDLINE | ID: mdl-33230153

ABSTRACT

The use of microorganisms that allows the recovery of critical high-tech elements such as gallium (Ga) and indium (In) has been considered an excellent eco-strategy. In this perspective, it is relevant to understand the strategies of Ga and In resistant strains to cope with these critical metals. This study aimed to explore the effect of these metals on two Ga/In resistant strains and to scrutinize the biological processes behind the oxidative stress in response to exposure to these critical metals. Two strains of Serratia fonticola, A3242 and B2A1Ga1, with high resistance to Ga and In, were submitted to metal stress and their protein profiles showed an overexpressed Superoxide Dismutase (SOD) in presence of In. Results of inhibitor-protein native gel incubations identified the overexpressed enzyme as a Fe-SOD. Both strains exhibited a huge increase of oxidative stress when exposed to indium, visible by an extreme high amount of reactive oxygen species (ROS) production. The toxicity induced by indium triggered biological mechanisms of stress control namely, the decrease in reduced glutathione/total glutathione levels and an increase in the SOD activity. The effect of gallium in cells was not so boisterous, visible only by the decrease of reduced glutathione levels. Analysis of the cellular metabolic viability revealed that each strain was affected differently by the critical metals, which could be related to the distinct metal uptakes. Strain A3242 accumulated more Ga and In in comparison to strain B2A1Ga1, and showed lower metabolic activity. Understanding the biological response of the two metal resistant strains of S. fonticola to stress induced by Ga and In will tackle the current gap of information related with bacteria-critical metals interactions.


Subject(s)
Environmental Pollutants/pharmacology , Gallium/pharmacology , Indium/pharmacology , Serratia/drug effects , Superoxide Dismutase/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Electronics/instrumentation , Environmental Pollutants/isolation & purification , Environmental Pollutants/metabolism , Gallium/isolation & purification , Gallium/metabolism , Humans , Indium/isolation & purification , Indium/metabolism , Microbial Sensitivity Tests , Oxidative Stress/drug effects , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Serratia/growth & development , Serratia/metabolism , Superoxide Dismutase/chemistry
20.
Crit Rev Ther Drug Carrier Syst ; 37(2): 161-182, 2020.
Article in English | MEDLINE | ID: mdl-32865904

ABSTRACT

Increasing drift in antimicrobial therapy failure against Mycobacterium tuberculosis, the causative agent of tuberculosis (TB), and the advent of extended resistant strains strongly demand discovery of mechanisms underlying development of drug resistance. The emergence of resistance against anti-TB drugs has reached an alarming level in various parts of the world, providing an active platform for the design of new targeted drug delivery. Reactive oxygen species (ROS) have an important role in controlling TB pathogenesis. At macrophage activation, ROS that are produced inside macrophages directly kill resident bacteria. These ROS possess a dual character because they can kill macrophages along with the resident bacteria. Targeting these ROS can play a remarkable part in overcoming resistance of conventional drugs. Nanoparticles (NPs) have evolved as a potential drug carrier for targeted delivery and elimination of various resistance mechanisms against antimicrobials. Receptor-mediated targeting of macrophages via different NPs may be a promising strategy for combating drug resistance and enhancing efficacy of old-fashioned antimycobacterial agents.


Subject(s)
Antitubercular Agents/pharmacology , Drug Carriers/chemistry , Mycobacterium tuberculosis/drug effects , Nanoparticles/chemistry , Tuberculosis/drug therapy , Antitubercular Agents/therapeutic use , Clinical Trials as Topic , Drug Resistance, Bacterial , Global Burden of Disease , Humans , Macrophage Activation/drug effects , Macrophages/immunology , Macrophages/metabolism , Mycobacterium tuberculosis/immunology , Reactive Oxygen Species/agonists , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Treatment Outcome , Tuberculosis/epidemiology , Tuberculosis/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...