Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.647
Filter
1.
Int J Mol Sci ; 25(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39125961

ABSTRACT

Garlic is a vegetable with numerous pro-health properties, showing high antioxidant capacity, and cytotoxicity for various malignant cells. The inhibition of cell proliferation by garlic is mainly attributed to the organosulfur compounds (OSCs), but it is far from obvious which constituents of garlic indeed participate in the antioxidant and cytotoxic action of garlic extracts. This study aimed to obtain insight into this question by examining the antioxidant activity and cytotoxicity of six OSCs and five phenolics present in garlic. Three common assays of antioxidant activity were employed (ABTS● decolorization, DPPH● decolorization, and FRAP). Cytotoxicity of both classes of compounds to PEO1 and SKOV-3 ovarian cancer cells, and MRC-5 fibroblasts was compared. Negligible antioxidant activities of the studied OSCs (alliin, allicin, S-allyl-D-cysteine, allyl sulfide, diallyl disulfide, and diallyl trisulfide) were observed, excluding the possibility of any significant contribution of these compounds to the total antioxidant capacity (TAC) of garlic extracts estimated by the commonly used reductive assays. Comparable cytotoxic activities of OSCs and phenolics (caffeic, p-coumaric, ferulic, gallic acids, and quercetin) indicate that both classes of compounds may contribute to the cytotoxic action of garlic.


Subject(s)
Allyl Compounds , Antioxidants , Disulfides , Garlic , Phenols , Plant Extracts , Sulfides , Sulfinic Acids , Garlic/chemistry , Humans , Antioxidants/pharmacology , Antioxidants/chemistry , Phenols/pharmacology , Phenols/chemistry , Disulfides/pharmacology , Disulfides/chemistry , Cell Line, Tumor , Plant Extracts/pharmacology , Plant Extracts/chemistry , Sulfinic Acids/pharmacology , Sulfinic Acids/chemistry , Sulfides/pharmacology , Sulfides/chemistry , Allyl Compounds/pharmacology , Allyl Compounds/chemistry , Sulfur Compounds/pharmacology , Sulfur Compounds/chemistry , Cysteine/analogs & derivatives , Cysteine/chemistry , Cysteine/pharmacology , Cell Survival/drug effects , Cell Proliferation/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism
2.
J Comput Aided Mol Des ; 38(1): 31, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39177727

ABSTRACT

Human Hippo signaling pathway is an evolutionarily conserved regulator network that controls organ development and has been implicated in various cancers. Transcriptional enhanced associate domain-4 (TEAD4) is the final nuclear effector of Hippo pathway, which is activated by Yes-associated protein (YAP) through binding to two separated YAP regions of α1-helix and Ω-loop. Previous efforts have all been addressed on deriving peptide inhibitors from the YAP to target TEAD4. Instead, we herein attempted to rationally design a so-called 'YAP helixα1-trap' based on the TEAD4 to target YAP by using dynamics simulation and energetics analysis as well as experimental assays at molecular and cellular levels. The trap represents a native double-stranded helical hairpin covering a specific YAP-binding site on TEAD4 surface, which is expected to form a three-helix bundle with the α1-helical region of YAP, thus competitively disrupting TEAD4-YAP interaction. The hairpin was further stapled by a disulfide bridge across its two helical arms. Circular dichroism characterized that the stapling can effectively constrain the trap into a native-like structured conformation in free state, thus largely minimizing the entropy penalty upon its binding to YAP. Affinity assays revealed that the stapling can considerably improve the trap binding potency to YAP α1-helix by up to 8.5-fold at molecular level, which also exhibited a good tumor-suppressing effect at cellular level if fused with TAT cell permeation sequence. In this respect, it is considered that the YAP helixα1-trap-mediated blockade of Hippo pathway may be a new and promising therapeutic strategy against cancers.


Subject(s)
Adaptor Proteins, Signal Transducing , Antineoplastic Agents , DNA-Binding Proteins , Molecular Dynamics Simulation , Muscle Proteins , TEA Domain Transcription Factors , Transcription Factors , YAP-Signaling Proteins , Transcription Factors/chemistry , Transcription Factors/metabolism , Transcription Factors/antagonists & inhibitors , Humans , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Muscle Proteins/chemistry , Muscle Proteins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Disulfides/chemistry , Disulfides/pharmacology , Protein Binding , Binding Sites , Cell Line, Tumor , Computer-Aided Design , Drug Design
3.
J Physiol Investig ; 67(4): 215-224, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-39206781

ABSTRACT

ABSTRACT: Diabetic retinopathy (DR) is one of the significant disabling outcomes of diabetes mellitus characterized by retinal microvascular damage, inflammation, and neuronal dysfunction. Allicin (Alc), a natural compound found in garlic, has garnered attention for its antioxidant and anti-inflammatory properties, positioning it as a potential therapeutic agent for DR. The aim of the present study was to investigate the therapeutic efficacy of Alc in DR management and elucidate its underlying mechanisms of action. We established a DR model in male Sprague-Dawley rats (n = 50, 200-250 g, 12 weeks old) using a high-fat diet for 8 weeks plus a low dose of streptozotocin administered at the start of the 4th week. The diabetic (Diab) animals were administered Alc (16 mg/kg/day, orally), either alone or in combination with mitochondrial division inhibitor-1 (Mdivi-1) as a mitophagy inhibitor, starting 28 days before tissue sampling. We evaluated histopathological changes, metabolic abnormalities associated with type 2 diabetes mellitus (T2DM), the expression of proteins regulating pyroptosis (NOD-like receptor family pyrin domain containing 3, cleaved-caspase 1, and gasdermin D-N terminal) and mitophagy (phosphatase and tensin homolog-induced kinase 1 [PINK1] and Parkin), as well as the levels of oxidative stress mediators and proinflammatory cytokines. Alc treatment effectively ameliorated histopathological changes and metabolic abnormalities associated with T2DM. It downregulated pyroptosis-related proteins, upregulated mitophagy-related proteins, reduced proinflammatory cytokine levels, and attenuated oxidative stress. Treatment with Mdivi-1 suppressed the beneficial effects of Alc. Our findings highlight the therapeutic potential of Alc in managing DR by targeting multiple pathophysiological pathways, including pyroptosis, inflammation, and oxidative stress. The observed antipyroptotic effects of Alc were partially mediated by the activation of the PINK1/parkin-mediated mitophagy pathway. Additional studies are necessary to thoroughly understand the therapeutic mechanisms of Alc and its viability as a treatment choice for DR.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Retinopathy , Disulfides , Inflammasomes , Mitophagy , Oxidative Stress , Rats, Sprague-Dawley , Sulfinic Acids , Ubiquitin-Protein Ligases , Animals , Male , Oxidative Stress/drug effects , Mitophagy/drug effects , Rats , Sulfinic Acids/pharmacology , Sulfinic Acids/therapeutic use , Inflammasomes/drug effects , Inflammasomes/metabolism , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/pathology , Diabetic Retinopathy/metabolism , Disulfides/pharmacology , Disulfides/therapeutic use , Ubiquitin-Protein Ligases/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Kinases/metabolism
4.
Langmuir ; 40(28): 14346-14354, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38953474

ABSTRACT

The issue of bacterial infectious diseases remains a significant concern worldwide, particularly due to the misuse of antibiotics, which has caused the emergence of antibiotic-resistant strains. Fortunately, the rapid development of nanomaterials has propelled significant progress in antimicrobial therapy, offering promising solutions. Among them, the utilization of nanoenzyme-based chemodynamic therapy (CDT) has become a highly hopeful approach to combating bacterial infectious diseases. Nevertheless, the application of CDT appears to be facing certain constraints for its low efficiency in the Fenton reaction at the infected site. In this study, we have successfully synthesized a versatile nanozyme, which was a composite of molybdenum sulfide (MoS2) and iron sulfide (FeS2), through the hydrothermal method. The results showed that iron/molybdenum sulfide nanozymes (Fe/Mo SNZs) with desirable peroxidase (POD) mimic activity can generate cytotoxic reactive oxygen species (ROS) by successfully triggering the Fenton reaction. The presence of MoS2 significantly accelerates the conversion of Fe2+/Fe3+ through a cocatalytic reaction that involves the participation of redox pairs of Mo4+/Mo6+, thereby enhancing the efficiency of CDT. Additionally, based on the excellent photothermal performance of Fe/Mo SNZs, a near-infrared (NIR) laser was used to induce localized temperature elevation for photothermal therapy (PTT) and enhance the POD-like nanoenzymatic activity. Notably, both in vitro and in vivo results demonstrated that Fe/Mo SNZs with good broad-spectrum antibacterial properties can help eradicate Gram-negative bacteria like Escherichia coli and Gram-positive bacteria like Staphylococcus aureus. The most exciting thing is that the synergistic PTT/CDT exhibited astonishing antibacterial ability and can achieve complete elimination of bacteria, which promoted wound healing after infection. Overall, this study presents a synergistic PTT/CDT strategy to address antibiotic resistance, providing avenues and directions for enhancing the efficacy of wound healing treatments and offering promising prospects for further clinical use in the near future.


Subject(s)
Anti-Bacterial Agents , Disulfides , Iron , Molybdenum , Sulfides , Wound Healing , Molybdenum/chemistry , Molybdenum/pharmacology , Wound Healing/drug effects , Sulfides/chemistry , Sulfides/pharmacology , Animals , Disulfides/chemistry , Disulfides/pharmacology , Iron/chemistry , Iron/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Catalysis , Staphylococcus aureus/drug effects , Mice , Escherichia coli/drug effects , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/pharmacology , Reactive Oxygen Species/metabolism , Nanostructures/chemistry , Phototherapy , Microbial Sensitivity Tests , Photothermal Therapy , Ferrous Compounds
5.
Neoplasma ; 71(3): 243-254, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38958714

ABSTRACT

Allicin (AL) is one of garlic-derived organosulfides and has a variety of pharmacological effects. Studies have reported that AL has notable inhibitory effects on liver cancer, gastric cancer, breast cancer, and other cancers. However, there are no relevant reports about its role in human nasopharyngeal carcinoma. Ferroptosis is an iron-dependent form of non-apoptotic regulated cell death. Increasing evidence indicates that induction of ferroptosis can inhibit the proliferation, migration, invasion, and survival of various cancer cells, which act as a tumor suppressor in cancer. In this study, we confirmed that AL can inhibit cell proliferation, migration, invasion, and survival in human nasopharyngeal carcinoma cells. Our finding shows that AL can induce the ferroptosis axis by decreasing the level of GSH and GPX4 and promoting the induction of toxic LPO and ROS. AL-mediated cytotoxicity in human nasopharyngeal carcinoma cells is dependent on ferroptosis. Therefore, AL has good anti-cancer properties and is expected to be a potential drug for the treatment of nasopharyngeal carcinoma.


Subject(s)
Cell Proliferation , Disulfides , Ferroptosis , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Reactive Oxygen Species , Sulfinic Acids , Humans , Ferroptosis/drug effects , Disulfides/pharmacology , Nasopharyngeal Carcinoma/drug therapy , Nasopharyngeal Carcinoma/pathology , Cell Proliferation/drug effects , Sulfinic Acids/pharmacology , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/pathology , Cell Line, Tumor , Reactive Oxygen Species/metabolism , Cell Movement/drug effects , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Glutathione/metabolism , Cell Survival/drug effects
6.
Eur J Med Chem ; 276: 116669, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39053189

ABSTRACT

The present study describes a small library of peptides derived from a potent and selective CXCR4 antagonist (3), wherein the native disulfide bond is replaced using a side-chain to tail macrolactamization technique to vary ring size and amino acid composition. The peptides were preliminary assessed for their ability to interfere with the interaction between the receptor and anti-CXCR4 PE-conjugated antibody clone 12G5. Two promising candidates (13 and 17) were identified and further evaluated in a125I-CXCL12 competition binding assay, exhibiting IC50 in the low-nanomolar range. Furthermore, both candidates displayed high selectivity towards CXCR4 with respect to the cognate receptor CXCR7, ability to block CXCL12-dependent cancer cell migration, and receptor internalization, albeit at a higher concentration compared to 3. Molecular modeling studies on 13 and 17 produced a theoretical model that may serve as a guide for future modifications, aiding in the development of analogs with improved affinity. Finally, the study provides valuable insights into developing therapeutic agents targeting CXCR4-mediated processes, demonstrating the adaptability of our lead peptide 3 to alternative cyclization approaches and offering prospects for comprehensive investigations into the receptor region's interaction with its C-terminal region.


Subject(s)
Disulfides , Peptides , Receptors, CXCR4 , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , Humans , Binding Sites/drug effects , Peptides/chemistry , Peptides/pharmacology , Peptides/chemical synthesis , Disulfides/chemistry , Disulfides/pharmacology , Structure-Activity Relationship , Molecular Structure , Dose-Response Relationship, Drug , Lactams/chemistry , Lactams/pharmacology , Lactams/chemical synthesis , Cell Movement/drug effects , Models, Molecular , Cell Line, Tumor
7.
Curr Microbiol ; 81(8): 245, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38940852

ABSTRACT

Garlic (Allium sativum L.), particularly its volatile essential oil, is widely recognized for medicinal properties. We have evaluated the efficacy of Indian Garlic Essential Oil (GEO) for antimicrobial and antibiofilm activity and its bioactive constituents. Allyl sulfur-rich compounds were identified as predominant phytochemicals in GEO, constituting 96.51% of total volatile oils, with 38% Diallyl trisulphide (DTS) as most abundant. GEO exhibited significant antibacterial activity against eleven bacteria, including three drug-resistant strains with minimum inhibitory concentrations (MICs) ranging from 78 to 1250 µg/mL. In bacterial growth kinetic assay GEO effectively inhibited growth of all tested strains at its ½ MIC. Antibiofilm activity was evident against two important human pathogens, S. aureus and P. aeruginosa. Mechanistic studies demonstrated that GEO disrupts bacterial cell membranes, leading to the release of nucleic acids, proteins, and reactive oxygen species. Additionally, GEO demonstrated potent antioxidant activity at IC50 31.18 mg/mL, while its isolated constituents, Diallyl disulphide (DDS) and Diallyl trisulphide (DTS), showed effective antibacterial activity ranging from 125 to 500 µg/mL and 250-1000 µg/mL respectively. Overall, GEO displayed promising antimicrobial and antibiofilm activity against enteric bacteria, suggesting its potential application in the food industry.


Subject(s)
Anti-Bacterial Agents , Antioxidants , Biofilms , Garlic , Microbial Sensitivity Tests , Oils, Volatile , Garlic/chemistry , Oils, Volatile/pharmacology , Oils, Volatile/chemistry , Antioxidants/pharmacology , Antioxidants/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Biofilms/drug effects , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology , Allyl Compounds/pharmacology , Allyl Compounds/chemistry , Phytochemicals/pharmacology , Phytochemicals/chemistry , Sulfides/pharmacology , Bacteria/drug effects , Pseudomonas aeruginosa/drug effects , Disulfides/pharmacology , India , Plant Oils/pharmacology , Plant Oils/chemistry , Humans , Plant Extracts/pharmacology , Plant Extracts/chemistry
8.
J Nanobiotechnology ; 22(1): 337, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38886712

ABSTRACT

BACKGROUND: Molybdenum disulfide (MoS2) has excellent physical and chemical properties. Further, chiral MoS2 (CMS) exhibits excellent chiroptical and enantioselective effects, and the enantioselective properties of CMS have been studied for the treatment of neurodegenerative diseases. Intriguingly, left- and right-handed materials have different effects on promoting the differentiation of neural stem cells into neurons. However, the effect of the enantioselectivity of chiral materials on peripheral nerve regeneration remains unclear. METHODS: In this study, CMS@bacterial cellulose (BC) scaffolds were fabricated using a hydrothermal approach. The CMS@BC films synthesized with L-2-amino-3-phenyl-1-propanol was defined as L-CMS. The CMS@BC films synthesized with D-2-amino-3-phenyl-1-propanol was defined as D-CMS. The biocompatibility of CMS@BC scaffolds and their effect on Schwann cells (SCs) were validated by cellular experiments. In addition, these scaffolds were implanted in rat sciatic nerve defect sites for three months. RESULTS: These chiral scaffolds displayed high hydrophilicity, good mechanical properties, and low cytotoxicity. Further, we found that the L-CMS scaffolds were superior to the D-CMS scaffolds in promoting SCs proliferation. After three months, the scaffolds showed good biocompatibility in vivo, and the nerve conducting velocities of the L-CMS and D-CMS scaffolds were 51.2 m/s and 26.8 m/s, respectively. The L-CMS scaffolds showed a better regenerative effect than the D-CMS scaffolds. Similarly, the sciatic nerve function index and effects on the motor and electrophysiological functions were higher for the L-CMS scaffolds than the D-CMS scaffolds. Finally, the axon diameter and myelin sheath thickness of the regenerated nerves were improved in the L-CMS group. CONCLUSION: We found that the CMS@BC can promote peripheral nerve regeneration, and in general, the L-CMS group exhibited superior repair performance. Overall, the findings of this study reveal that CMS@BC can be used as a chiral nanomaterial nerve scaffold for peripheral nerve repair.


Subject(s)
Cellulose , Disulfides , Molybdenum , Nerve Regeneration , Schwann Cells , Tissue Scaffolds , Nerve Regeneration/drug effects , Animals , Rats , Tissue Scaffolds/chemistry , Disulfides/chemistry , Disulfides/pharmacology , Schwann Cells/drug effects , Molybdenum/chemistry , Molybdenum/pharmacology , Cellulose/chemistry , Cellulose/pharmacology , Cellulose/analogs & derivatives , Rats, Sprague-Dawley , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Sciatic Nerve/drug effects , Sciatic Nerve/physiology , Cell Proliferation/drug effects , Tissue Engineering/methods , Male , Peripheral Nerve Injuries , Stereoisomerism
9.
ACS Nano ; 18(26): 16853-16866, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38896491

ABSTRACT

The precise modulation of electrical activity in specific neuronal populations is paramount for rectifying abnormal neurological functions and is a critical element in the therapeutic arsenal for neurological disorders. However, achieving a balance between minimal invasiveness and robust neuroprotection poses a considerable challenge. Herein, we present a nanoneuromodulation strategy integrating neuroprotective features to effectively address epilepsy with minimal invasiveness and enable wireless functionality. Strategically engineered nanotransducer, adorned with platinum (Pt) decoration with titanium disulfide (TiS2) (TiS2/Pt), enables precise modulation of neuronal electrical activity in vitro and in vivo, ensuring exceptional temporal fidelity under millisecond-precision near-infrared (NIR) light pulses irradiation. Concurrently, TiS2/Pt showcase a pronounced enhancement in enzyme-mimicking activity, offering a robust defense against oxidative neurological injury in vitro. Nanotransducer-enabled wireless neuromodulation with biocatalytic neuroprotective capacity is highly effective in alleviating epileptic high-frequency neural activity and diminishing oxidative stress levels, thereby restoring redox equilibrium. This integrated therapeutic approach reduces the severity of epilepsy, demonstrating minimal invasiveness and obviating the requirements for genetic manipulation and optical fiber implantation, while providing an alternative avenue for neurological disorder treatment.


Subject(s)
Epilepsy , Epilepsy/therapy , Animals , Titanium/chemistry , Titanium/pharmacology , Platinum/chemistry , Platinum/pharmacology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Neuroprotection/drug effects , Mice , Disulfides/chemistry , Disulfides/pharmacology , Oxidative Stress/drug effects , Humans , Neurons/drug effects , Neurons/metabolism , Infrared Rays , Rats
10.
Ecotoxicol Environ Saf ; 280: 116527, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38833978

ABSTRACT

Aflatoxin B1 (AFB1) is known to inhibit growth, and inflict hepatic damage by interfering with protein synthesis. Allicin, has been acknowledged as an efficacious antioxidant capable of shielding the liver from oxidative harm. This study aimed to examine the damage caused by AFB1 on bovine hepatic cells and the protective role of allicin against AFB1-induced cytotoxicity. In this study, cells were pretreated with allicin before the addition of AFB1 for co-cultivation. Our findings indicate that AFB1 compromises cellular integrity, suppresses the expression of nuclear factor erythroid 2-related factor 2 (Nrf2). In addition, allicin attenuates oxidative damage to bovine hepatic cells caused by AFB1 by promoting the expression of the Nrf2 pathway and reducing cell apoptosis. In conclusion, the results of this study will help advance clinical research and applications, providing new options and directions for the prevention and treatment of liver diseases.


Subject(s)
Aflatoxin B1 , Antioxidants , Apoptosis , Disulfides , Hepatocytes , NF-E2-Related Factor 2 , Oxidative Stress , Signal Transduction , Sulfinic Acids , Animals , Sulfinic Acids/pharmacology , Aflatoxin B1/toxicity , Cattle , Disulfides/pharmacology , NF-E2-Related Factor 2/metabolism , Signal Transduction/drug effects , Hepatocytes/drug effects , Oxidative Stress/drug effects , Apoptosis/drug effects , Antioxidants/pharmacology , Female
11.
Eur J Med Chem ; 275: 116541, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38851056

ABSTRACT

Psammaplin A (PsA), a symmetrical bromotyrosine-derived disulfide marine metabolite, has been reported could inhibit HDAC1/2/3 through its thiol monomer. Inspired by the disuflide bond structure of this marine natural product, we designed and synthesized a series of PsA analogues, in which the disulfide bond of PsA was replaced with diselenide bond or cyclic disulfide/diselenide/selenenylsulfide motifs. We also studied the HDAC inhibition, cell growth inhibition, and apoptosis induction of these PsA analogues. The results showed that, all the synthetic diselenide analogues and cyclic selenenyl sulfide compounds exhibited better antiproferative activity than their counterpart of disulfide analogues. Among the prepared analogues, diselenide analogue P-503 and P-116 significantly increased the ability of inhibiting HDAC6 and induced apoptosis and G2/M cell cycle arrest. However, cyclic selenenylsulfides analogues P-111 lost its HDAC inhibitory ability and exhibited no effect on cell cycle and apoptosis, indicating that the anti-proliferative mechanism of cyclic selenenylsulfides analogues has changed.


Subject(s)
Apoptosis , Cell Proliferation , Disulfides , Histone Deacetylase Inhibitors , Histone Deacetylases , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Disulfides/chemistry , Disulfides/pharmacology , Disulfides/chemical synthesis , Humans , Apoptosis/drug effects , Histone Deacetylases/metabolism , Cell Proliferation/drug effects , Structure-Activity Relationship , Molecular Structure , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Cell Line, Tumor , Tyrosine/analogs & derivatives
12.
Phytother Res ; 38(8): 4009-4021, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38863408

ABSTRACT

Environmental pollution, virus infection, allergens, and other factors may cause respiratory disease, which could be improved by dietary therapy. Allium species are common daily food seasoning and have high nutritional and medical value. Diallyl disulfide (DADS) is the major volatile oil compound of Allium species. The present study aims to explore the preventive effect and potential mechanism of DADS on pulmonary fibrosis. C57BL/6J mice were intratracheally injected with bleomycin (BLM) to establish pulmonary fibrosis and then administrated with DADS. Primary lung fibroblasts or A549 were stimulated with BLM, followed by DADS, farnesoid X receptor (FXR) agonist (GW4064), yes-associated protein 1 (YAP1) inhibitor (verteporfin), or silencing of FXR and YAP1. In BLM-stimulated mice, DADS significantly ameliorated histopathological changes and interleukin-1ß levels in bronchoalveolar lavage fluid. DADS decreased fibrosis markers, HIF-1α, inflammatory cytokines, and epithelial-mesenchymal transition in pulmonary mice and activated fibroblasts. DADS significantly enhanced FXR expression and inhibited YAP1 activation, which functions as GW4064 and verteporfin. A deficiency of FXR or YAP1 could result in the increase of these two protein expressions, respectively. DADS ameliorated extracellular matrix deposition, hypoxia, epithelial-mesenchymal transition, and inflammation in FXR or YAP1 knockdown A549. Taken together, targeting the crosstalk of FXR and YAP1 might be the potential mechanism for DADS against pulmonary fibrosis. DADS can serve as a potential candidate or dietary nutraceutical supplement for the treatment of pulmonary fibrosis.


Subject(s)
Allyl Compounds , Disulfides , Mice, Inbred C57BL , Pulmonary Fibrosis , Receptors, Cytoplasmic and Nuclear , Signal Transduction , YAP-Signaling Proteins , Animals , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/chemically induced , Mice , Disulfides/pharmacology , Humans , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/drug effects , Allyl Compounds/pharmacology , A549 Cells , Male , Allium/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Bleomycin , Lung/drug effects , Lung/pathology , Fibroblasts/drug effects , Fibroblasts/metabolism
13.
Biomater Adv ; 162: 213920, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38901063

ABSTRACT

Chronic refractory wounds caused by multidrug-resistant (MDR) bacterial and biofilm infections are a substantial threat to human health, which presents a persistent challenge in managing clinical wound care. We here synthesized a composite nanosheet AIPH/AMP/MoS2, which can potentially be used for combined therapy because of the photothermal effect induced by MoS2, its ability to deliver antimicrobial peptides, and its ability to generate alkyl free radicals independent of oxygen. The synthesized nanosheets exhibited 61 % near-infrared (NIR) photothermal conversion efficiency, marked photothermal stability and free radical generating ability. The minimal inhibitory concentrations (MICs) of the composite nanosheets against MDR Escherichia coli (MDR E. coli) and MDR Staphylococcus aureus (MDR S. aureus) were approximately 38 µg/mL and 30 µg/mL, respectively. The composite nanosheets (150 µg/mL) effectively ablated >85 % of the bacterial biofilm under 808-nm NIR irradiation for 6 min. In the wound model experiment, approximately 90 % of the wound healed after the 4-day treatment with the composite nanosheets. The hemolysis experiment, mouse embryonic fibroblast (MEFs) cytotoxicity experiment, and mouse wound healing experiment all unveiled the excellent biocompatibility of the composite nanosheets. According to the transcriptome analysis, the composite nanosheets primarily exerted a synergistic therapeutic effect by disrupting the cellular membrane function of S. aureus and inhibiting quorum sensing mediated by the two-component system. Thus, the synthesized composite nanosheets exhibit remarkable antibacterial and biofilm ablation properties and therefore can be used to improve wound healing in chronic biofilm infections.


Subject(s)
Biofilms , Disulfides , Escherichia coli , Molybdenum , Staphylococcus aureus , Wound Healing , Biofilms/drug effects , Wound Healing/drug effects , Animals , Mice , Disulfides/chemistry , Disulfides/pharmacology , Molybdenum/chemistry , Molybdenum/pharmacology , Escherichia coli/drug effects , Staphylococcus aureus/drug effects , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/chemistry , Free Radicals/chemistry , Free Radicals/metabolism , Nanostructures/chemistry , Microbial Sensitivity Tests , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Hot Temperature , Humans , Anti-Infective Agents/pharmacology , Anti-Infective Agents/chemistry
14.
J Mater Chem B ; 12(28): 6927-6939, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38904166

ABSTRACT

Radiotherapy is a pivotal means of cancer treatment, but it often leads to radiation dermatitis, a skin injury caused by radiation-induced excess reactive oxygen species (ROS). Scavenging free radicals in the course of radiation therapy will be an effective means to prevent radiation dermatitis. This study demonstrates a novel double network hydrogel doped with MoS2 nanosheets for the prevention of radiation-induced dermatitis. The resultant SPM hydrogel constructed from polyacrylamide (PAM) and sodium alginate (SA) nanofiber presented favorable mechanical and adhesion properties. It could conform well to the human body's irregular contours without secondary dressing fixation, making it suitable for skin protection applications. The in vitro and in vivo experiments showed that the antioxidant properties conferred by MoS2 nanosheets enable SPM to effectively mitigate excessive ROS and reduce oxidative stress, thereby preventing radiation dermatitis caused by oxidative damage. Biosafety assessments indicated good biocompatibility of the composite hydrogel, suggesting SPM's practicality and potential as an external dressing for skin radiation protection.


Subject(s)
Alginates , Antioxidants , Hydrogels , Radiodermatitis , Hydrogels/chemistry , Hydrogels/pharmacology , Antioxidants/chemistry , Antioxidants/pharmacology , Radiodermatitis/prevention & control , Radiodermatitis/drug therapy , Animals , Alginates/chemistry , Alginates/pharmacology , Humans , Acrylic Resins/chemistry , Acrylic Resins/pharmacology , Mice , Molybdenum/chemistry , Molybdenum/pharmacology , Disulfides/chemistry , Disulfides/pharmacology , Reactive Oxygen Species/metabolism , Adhesives/chemistry , Adhesives/pharmacology , Particle Size
15.
Colloids Surf B Biointerfaces ; 241: 114031, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38878661

ABSTRACT

The therapy of the clear cell renal cell carcinoma (ccRCC) is crucial for the human healthcare due to its easy metastasis and recurrence, as well as resistance to radiotherapy and chemotherapy. In this work, we propose the synthesis of MoS2@red phosphorus (MoS2@RP) heterojunction to induce synergistic photodynamic and photothermal therapy (PDT/PTT) of ccRCC. The MoS2@RP heterojunction exhibits enhanced spectra absorption in the NIR range and produce local heat-increasing under the NIR laser irradiation compared with pure MoS2 and RP. The high photocatalytic activity of the MoS2@RP heterojunction contributes to effective transferring of the photo-excited electrons from the RP to MoS2, which promotes the production of various types of radical oxygen species (ROS) to kill the ccRCC cells. After the NIR irradiation, the MoS2@RP can effectively induce the apoptosis in the ccRCC cells through localized hyperthermia and the generation of ROS, while exhibiting low cytotoxicity towards normal kidney cells. In comparison to MoS2, the MoS2@RP heterojunction shows an approximate increase of 22 % in the lethality rate of the ccRCC cells and no significant change in toxicity towards normal cells. Furthermore, the PDT/PTT treatment using the MoS2@RP heterojunction effectively eradicates a substantial number of deep-tissue ccRCC cells in vivo without causing significant damage to major organs. This study presents promising effect of the MoS2@RP heterojunction-based photo-responsive therapy for effective ccRCC treatment.


Subject(s)
Carcinoma, Renal Cell , Disulfides , Kidney Neoplasms , Molybdenum , Phosphorus , Photochemotherapy , Photothermal Therapy , Molybdenum/chemistry , Molybdenum/pharmacology , Humans , Disulfides/chemistry , Disulfides/pharmacology , Disulfides/chemical synthesis , Phosphorus/chemistry , Phosphorus/pharmacology , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Kidney Neoplasms/drug therapy , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/therapy , Carcinoma, Renal Cell/drug therapy , Animals , Apoptosis/drug effects , Mice , Cell Survival/drug effects , Reactive Oxygen Species/metabolism , Drug Screening Assays, Antitumor , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Particle Size , Cell Line, Tumor , Cell Proliferation/drug effects , Infrared Rays , Surface Properties
16.
ACS Nano ; 18(25): 16184-16198, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38864540

ABSTRACT

Drug-resistant bacterial infections pose a serious threat to human health; thus, there is an increasingly growing demand for nonantibiotic strategies to overcome drug resistance in bacterial infections. Mild photothermal therapy (PTT), as an attractive antibacterial strategy, shows great potential application due to its good biocompatibility and ability to circumvent drug resistance. However, its efficiency is limited by the heat resistance of bacteria. Herein, Cu2O@MoS2, a nanocomposite, was constructed by the in situ growth of Cu2O nanoparticles (NPs) on the surface of MoS2 nanosheets, which provided a controllable photothermal therapeutic effect of MoS2 and the intrinsic catalytic properties of Cu2O NPs, achieving a synergistic effect to eradicate multidrug-resistant bacteria. Transcriptome sequencing (RNA-seq) results revealed that the antibacterial process was related to disrupting the membrane transport system, phosphorelay signal transduction system, oxidative stress response system, as well as the heat response system. Animal experiments indicated that Cu2O@MoS2 could effectively treat wounds infected with methicillin-resistant Staphylococcus aureus. In addition, satisfactory biocompatibility made Cu2O@MoS2 a promising antibacterial agent. Overall, our results highlight the Cu2O@MoS2 nanocomposite as a promising solution to combating resistant bacteria without inducing the evolution of antimicrobial resistance.


Subject(s)
Anti-Bacterial Agents , Copper , Disulfides , Infrared Rays , Methicillin-Resistant Staphylococcus aureus , Microbial Sensitivity Tests , Molybdenum , Nanocomposites , Molybdenum/chemistry , Molybdenum/pharmacology , Disulfides/chemistry , Disulfides/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Copper/chemistry , Copper/pharmacology , Nanocomposites/chemistry , Methicillin-Resistant Staphylococcus aureus/drug effects , Animals , Mice , Photothermal Therapy , Humans
17.
PeerJ ; 12: e17363, 2024.
Article in English | MEDLINE | ID: mdl-38766487

ABSTRACT

Background: Promoter hypermethylation of the tumor suppressor gene is one of the well-studied causes of cancer development. The drugs that reverse the process by driving demethylation could be a candidate for anticancer therapy. This study was designed to investigate the effects of arsenic disulfide on PTPL1 methylation in diffuse large B cell lymphoma (DLBCL). Methods: We knocked down the expression of PTPL1 in two DLBCL cell lines (i.e., DB and SU-DHL-4 cells) using siRNA. Then the DLBCL proliferation was determined in the presence of PTPL1 knockdown. The methylation of PTPL1 in DLBCL cells was analyzed by methylation specific PCR (MSPCR). The effect of arsenic disulfide on the PTPL1 methylation was determined in DLBCL cell lines in the presence of different concentrations of arsenic disulfide (5 µM, 10 µM and 20 µM), respectively. To investigate the potential mechanism on the arsenic disulfide-mediated methylation, the mRNA expression of DNMT1, DNMT3B and MBD2 was determined. Results: PTPL1 functioned as a tumor suppressor gene in DLBCL cells, which was featured by the fact that PTPL1 knockdown promoted the proliferation of DLBCL cells. PTPL1 was found hypermethylated in DLBCL cells. Arsenic disulfide promoted the PTPL1 demethylation in a dose-dependent manner, which was related to the inhibition of DNMTs and the increase of MBD2. Conclusion: Experimental evidence shows that PTPL1 functions as a tumor suppressor gene in DLBCL progression. PTPL1 hyper-methylation could be reversed by arsenic disulfide in a dose-dependent manner.


Subject(s)
Arsenicals , DNA Methylation , Lymphoma, Large B-Cell, Diffuse , Humans , Arsenicals/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Disulfides/pharmacology , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/drug effects , DNA Methyltransferase 3B , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Promoter Regions, Genetic/drug effects
18.
Tissue Cell ; 88: 102398, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38728949

ABSTRACT

BACKGROUND: Allicin regulates macrophage autophagy and senescence, and inhibits hepatoma cell growth. This study investigated the mechanism by which allicin inhibits the growth of hepatoma cells. METHODS: Hepa1-6 mouse hepatoma cells were subcutaneously injected into C57BL/6 J mice to construct a tumor transplantation model. Macrophages were cultured with the supernatant of hepatoma cells to construct a cell model. The levels of mRNA and proteins and the level of Sestrin2 ubiquitination were measured by RTqPCR, immunofluorescence and Western blotting. The levels of autophagy-related factors and the activity of senescence-associated ß-galactosidase were determined by kits, and protein stability was detected by cycloheximide (CHX) tracking. RESULTS: Data analysis of clinical samples revealed that RBX1 was highly expressed in tumor tissues, while Sestrin2 was expressed at low levels in tumor tissues. Allicin can promote the expression of the autophagy-related proteins LC3 and Beclin-1 in tumor macrophages and inhibit the expression of the aging-related proteins p16 and p21, thus promoting autophagy in macrophages and inhibiting cell senescence. Moreover, allicin can inhibit the expression of RBX1, thereby reducing the ubiquitination of Sestrin2, enhancing the stability of Sestrin2, activating autophagy in tumor macrophages and inhibiting senescence. In addition, allicin treatment inhibited the proliferation and migration of hepatoma carcinoma cells cocultured with macrophages and significantly improved the development of liver cancer in mice. CONCLUSION: Allicin can affect the autophagy of macrophages and restrain the growth of hepatoma cells by regulating the ubiquitination of Sestrin2.


Subject(s)
Autophagy , Carcinoma, Hepatocellular , Cellular Senescence , Disulfides , Liver Neoplasms , Macrophages , Sulfinic Acids , Ubiquitination , Animals , Autophagy/drug effects , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/drug therapy , Disulfides/pharmacology , Cellular Senescence/drug effects , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/drug therapy , Sulfinic Acids/pharmacology , Macrophages/metabolism , Macrophages/drug effects , Mice , Cell Line, Tumor , Ubiquitination/drug effects , Humans , Cell Proliferation/drug effects , Mice, Inbred C57BL , Nuclear Proteins/metabolism , Male , Peroxidases/metabolism , Sestrins
19.
Immunopharmacol Immunotoxicol ; 46(3): 408-416, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38816179

ABSTRACT

BACKGROUND: Myelodysplastic syndrome (MDS) is a prevalent hematological neoplastic disorder in clinics and its immunopathogenesis has garnered growing interest. Oral and intravenous arsenic agents have long been used to treat hematological malignancies. The main component of oral arsenic is realgar (arsenic disulfide), while arsenic trioxide is the main component of intravenous arsenic. METHODS: This study aimed to assess the effects of ATO and Realgar on the enhancement of peripheral blood, drug safety, and T cell immune status in the NUP98-HOXD13 (NHD13) mice model of MDS, specifically in the peripheral blood, spleen, and liver. RESULTS: The study findings indicate that realgar and arsenic trioxide (ATO) can improve peripheral hemogram in mice, whereas realgar promotes higher peripheral blood cell production than ATO. Furthermore, the clinical administration method and dose did not cause significant toxicity or side effects and thus can be considered safe. Coexistence and interconversion of hyperimmune function and immunosuppression in mice were also observed in this study. In addition, there were interactions between immune cells in the peripheral blood, spleen, and liver to regulate the immune balance of the body and activate immunity via T-cell activation. CONCLUSION: In summary, oral and intravenous arsenic agents are beneficial in improving peripheral hemogram and immunity in mice.


Subject(s)
Arsenic Trioxide , Arsenicals , Disease Models, Animal , Myelodysplastic Syndromes , Animals , Arsenic Trioxide/administration & dosage , Arsenic Trioxide/pharmacology , Arsenicals/pharmacology , Arsenicals/administration & dosage , Mice , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/immunology , Sulfides/pharmacology , Sulfides/administration & dosage , Disulfides/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Spleen/drug effects , Spleen/immunology
20.
Pest Manag Sci ; 80(8): 4125-4136, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38578571

ABSTRACT

BACKGROUND: Maize stalk rot (MSR) caused by Fusarium graminearum is the primary factor contributing to the reduction in maize yield and quality. However, this soil-borne disease presents a significant challenge for sustainable control through field management and chemical agents. The screening of novel biocontrol agents can aid in developing innovative and successful strategies for MSR control. RESULTS: A total of 407 strains of bacteria were isolated from the rhizosphere soil of a resistant maize inbred line. One strain exhibited significant antagonistic activity in plate and pot experiments, and was identified as Burkholderia ambifaria H8. The strain could significantly inhibit the mycelial growth and spore germination of F. graminearum, induce resistance to stalk rot, and promote plant growth. The volatile compounds produced by strain H8 and its secondary metabolites in the sterile fermentation broth exhibited antagonistic activity. The primary volatile compound produced by strain H8 was identified as dimethyl disulfide (DMDS) using gas chromatography tandem mass spectrometry. Through in vitro antagonistic activity assays and microscopic observation, it was confirmed that DMDS was capable of inhibiting mycelial growth and disrupting the mycelial structure of F. graminearum, suggesting it may be the major active compound for strain H8. The transcriptome data of F. graminearum further indicated that strain H8 and its volatile compounds could alter pathogenic fungi metabolism, influence the related metabolic pathways, and potentially induce cell apoptosis within F. graminearum. CONCLUSION: Our results showed that B. ambifaria H8 was capable of producing the volatile substance dimethyl disulfide, which influenced the synthesis and permeability of cell membranes in pathogens. Thus, B. ambifaria H8 was found to be a promising biological control agent against MSR. © 2024 Society of Chemical Industry.


Subject(s)
Burkholderia , Disulfides , Fusarium , Plant Diseases , Volatile Organic Compounds , Zea mays , Fusarium/physiology , Zea mays/microbiology , Disulfides/pharmacology , Plant Diseases/microbiology , Plant Diseases/prevention & control , Burkholderia/physiology , Burkholderia/metabolism , Volatile Organic Compounds/pharmacology , Volatile Organic Compounds/metabolism , Pest Control, Biological , Biological Control Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL