Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 405
Filtrar
1.
Mol Med Rep ; 31(1)2025 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39450531

RESUMO

The present study aimed to explore the effect of melittin (MLT) on the growth of Schwann cells (SCs) in high glucose conditions and to understand the mechanisms involved. The goal was to provide a theoretical basis for using MLT in the treatment of diabetic peripheral neuropathy (DPN). The CCK­8 assay was used to measure cell activity at different concentrations of glucose and MLT. Flow cytometry was employed to analyze the effect of MLT on cell cycle phases and apoptosis in SCs under high glucose conditions. To identify differentially expressed proteins, 4D label­free quantitative proteomics with liquid chromatography­mass spectrometry was used, followed by biological analysis to explore potential mechanisms. PCR, western blotting and immunofluorescence were conducted to confirm these mechanisms. Melittin (0.2 µg/ml) increased the proliferation of SCs in a high glucose environment. Flow cytometry showed that after MLT treatment, the proportion of cells in the G2/M+S phase increased and the combined ratio of early and late apoptosis decreased under high glucose conditions. Proteomics identified 1,784 proteins with significant changes in expression; 725 were upregulated, and 1,059 were downregulated. Kyoto Encyclopedia of Genes and Genomes analysis indicated that the differentially expressed proteins were mainly involved in metabolic pathways and neurodegenerative disease pathways. PCR, western blotting and immunofluorescence confirmed the increase in Crabp2, Wnt3a, C­Jun, CDK4, CyclinD1 and proliferating cell nuclear antigen. In high glucose conditions, MLT protects SCs from glucose toxicity by upregulating the Crabp2/Wnt/ß­catenin signaling pathway, potentially providing a new treatment for DPN.


Assuntos
Proliferação de Células , Glucose , Meliteno , Células de Schwann , Via de Sinalização Wnt , Células de Schwann/metabolismo , Células de Schwann/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Via de Sinalização Wnt/efeitos dos fármacos , Animais , Glucose/metabolismo , Meliteno/farmacologia , Apoptose/efeitos dos fármacos , Ratos , Hiperglicemia/metabolismo , Proteômica/métodos , Regulação para Cima/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos
2.
Int J Mol Sci ; 25(19)2024 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-39408837

RESUMO

The interaction forces and mechanical properties of the interaction between melittin (Mel) and lipopolysaccharide (LPS) are considered to be crucial driving forces for Mel when killing Gram-negative bacteria (GNB). However, how their interaction forces perform at the single-molecule level and the dissociation kinetic characteristics of the Mel/LPS complex remain poorly understood. In this study, the single-molecule-level interaction forces between Mel and LPSs from E. coli K-12, O55:B5, O111:B4, and O128:B12 were explored using atomic force microscopy (AFM)-based single-molecule force spectroscopy (SMFS). AFM-based dynamic force spectroscopy (DFS) and an advanced analytical model were employed to investigate the kinetic characteristics of the Mel/LPS complex dissociation. The results indicated that Mel could interact with both rough (R)-form LPS (E. coli K-12) and smooth (S)-form LPSs (E. coli O55:B5, O111:B4, and O128:B12). The S-form LPS showed a more robust interaction with Mel than the R-form LPS, and a slight difference existed in the interaction forces between Mel and the diverse S-form LPS. Mel interactions with the S-form LPSs showed greater specific and non-specific interaction forces than the R-form LPS (p < 0.05), as determined by AFM-based SMFS. However, there was no significant difference in the specific and non-specific interaction forces among the three samples of S-form LPSs (p > 0.05), indicating that the variability in the O-antigen did not affect the interaction between Mel and LPSs. The DFS result showed that the Mel/S-form LPS complexes had a lower dissociation rate constant, a shorter energy barrier width, a longer bond lifetime, and a higher energy barrier height, demonstrating that Mel interacted with S-form LPS to form more stable complexes. This research enhances the existing knowledge of the interaction micromechanics and kinetic characteristics of Mel and LPS at the single-molecule level. Our research may help with the design and evaluation of new anti-GNB drugs.


Assuntos
Lipopolissacarídeos , Meliteno , Microscopia de Força Atômica , Imagem Individual de Molécula , Meliteno/química , Meliteno/metabolismo , Microscopia de Força Atômica/métodos , Lipopolissacarídeos/química , Lipopolissacarídeos/metabolismo , Imagem Individual de Molécula/métodos , Escherichia coli/metabolismo , Bactérias Gram-Negativas/metabolismo , Cinética
3.
Biomed Pharmacother ; 180: 117573, 2024 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-39426282

RESUMO

Melittin is preclinically investigated as anticancer agent in multiple tumor types. But its regulation role and regulatory mechanism regarding NSCLC is unknown. In our investigation, Proteomic test was employed to identify proteins that expressed abnormally in cancer cells and that with Melittin treatmented. The results showed CTSB was one of the Top proteins with different expression levels in the lysosomes of Melittin-treatmented cancer cells and showed an up-regulation trend. CTSB expression was increased in NSCLC cancer tissues compared to adjacent normal tissues, as demonstrated in lung cancer tissue chips experiment. However, Melittin treatment increased the CTSB level in lysosomes, which inhibited the malignant progression of NSCLC. We hypothesized that the relative homeostasis of CTSB in cancer cells was destroyed, and CTSB exerts its hydrolytic effect excessively, resulting in excessive autophagy of cancer cells, thus inhibiting the malignant progression of cancer cells. The direct combination of Melittin and CTSB was proposed by molecular docking technique, LiP-SMap was used to analyze the target genes and active components extracted from high-throughput sequencing proteomic data, and successfully verified that melittin was successfully demonstrated to directly target CTSB-binding. In vivo and in vitro studies have shown that Melittin treatment inhibits the malignant progression of A549 and HCC1833 cells and animal tumors, namely non-small cell lung cancer, by promoting CTSB-mediated hyperautophagy. CTSB-specific inhibitor CA-074 Me and autophagy inhibitor 3-MA treatment reversed the inhibit effect of Melittin to the malignant progression of NSCLC. Taken together, Melittin treatment inhibited malignant progression regarding NSCLC through enhancing CTSB-mediated hyperautophagy.

4.
Front Pharmacol ; 15: 1443497, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39434904

RESUMO

Introduction: Melittin is a potent antimicrobial peptide from bee venom that is effective against both Gram-positive and Gram-negative bacteria. However, it is extremely toxic to mammalian cells and, as yet, has no clinical use. Modifications to its amino acid sequence, cyclization, truncation, and dimerization have been attempted in order to reduce its toxicity whilst maintaining its antimicrobial activity. Methods: In this study, we targeted the three lysine residues present in melittin and substituted them with lysine homologs containing shorter side chains (ornithine, Orn, diaminobutyric acid, Dab, and diaminopropanoic acid, Dap) and made both parallel and antiparallel melittin dimers to observe how lysine substitution and dimerization affects its activity and toxicity. The antibacterial activity of melittin and its analogs was tested against S. aureus (Gram-positive bacteria) and E. coli (Gram-negative bacteria), and cytotoxicity was tested against the mammalian cell lines HEK293 and H4IIE. Results: Overall, dimerization and lysine substitution exhibited improved antimicrobial activity toward E. coli and limited improvement toward S. aureus. However, mammalian cell toxicity was only marginally reduced compared to native melittin. Interestingly, the parallel dimer was found to be marginally more active than the antiparallel dimer, indicating orientation maybe important for activity, although both dimers were less effective than the native and Lys-analog peptides toward S. aureus. Of the Lys substitutions, Dab and Dap improved melittin's activity toward E. coli. Discussion: Dimerization and Lys substitution of melittin improved the antimicrobial activity toward Gram-negative bacteria but did not significantly improve its activity toward Gram-positive bacteria. Some analogs also displayed reduced toxicity toward HEK293 and H4IIE cells but overall remained toxic at bactericidal concentrations. Our data indicates that although highly antibacterial, melittin's toxicity is the major drawback in its potential use.

5.
Chem Biol Drug Des ; 104(4): e14637, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39396919

RESUMO

Apitherapy has started to gain tremendous recognition because of extraordinary pharmacological importance of honeybee-related ingredients and their derivatives. There has been a renewed interest in the bee venom-based therapies. Interdisciplinary researchers are studying the chemistry and translational value of venom for effective cancer treatment. Bee venom and its major component, melittin, are cytotoxic in cancer cells. In this study, MTT and scratch assays were performed for analysis of melittin-mediated antimetastatic effects. QPCR was used for expression profiling of metastasis-related genes. Three anti-metastatic genes (BRMS1, DRG1, and KAI1/CD82) were studied for the first time after bee venom and melittin treatment in MDA-MB-231 breast cancer cells compared with normal breast cells, and two prometastatic genes (EGFR and WNT7B) were also examined. KAI1/CD82 and BRMS1 are the negative regulators of EGFR. WNT7B is a negative regulator of KAI1/CD82. Selective cytotoxicity of bee venom and melittin was found to be higher as compared to cisplatin. Melittin induced an increase in the expression of BRMS1 and DRG1, whereas bee venom upregulated DRG1 and KAI1/CD82 expression in breast cancer. WNT7B was downregulated in bee venom-treated breast cancer cells. Results suggested that bee venom/melittin exerted antimetastatic effects primarily through upregulation of BRMS1, DRG1, and KAI1/CD82, and downregulation of WNT7B.


Assuntos
Venenos de Abelha , Neoplasias da Mama , Proteína Kangai-1 , Meliteno , Regulação para Cima , Meliteno/farmacologia , Meliteno/uso terapêutico , Humanos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Venenos de Abelha/farmacologia , Feminino , Regulação para Cima/efeitos dos fármacos , Proteína Kangai-1/metabolismo , Proteína Kangai-1/genética , Antineoplásicos/farmacologia , Antineoplásicos/química , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas Wnt/metabolismo , Receptores ErbB/metabolismo , Receptores ErbB/genética , Animais , Metástase Neoplásica , Movimento Celular/efeitos dos fármacos
6.
Front Microbiol ; 15: 1416995, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39252832

RESUMO

Introduction: Staphylococcus aureus is a prominent cause of postoperative infections, often persisting within host cells, leading to chronic infections. Conventional antibiotics struggle to eliminate intracellular S. aureus due to poor cell penetration. Antimicrobial peptides are a new hope for tackling intracellular bacteria. Accordingly, this study examines the antimicrobial peptide MDP1, derived from melittin, for its efficacy against intracellular S. aureus. Methods: In this study, the physiochemical properties (Prediction of three-dimensional structure, circular dichroism and helical wheel projection analysis) were investigated. Extracellular antibacterial activity and cytotoxicity of MDP1 were also assessed. The mechanism of interaction of MDP1 with S. aureus was evaluated by molecular dynamic simulation, atomic force and confocal microscopy. Bacterial internalization into an endothelial cell model was confirmed through culture and transmission electron microscopy. The effect of the peptide on intracellular bacteria was investigated by culture and epi-fluorescence microscopy. Results and discussion: 3D structural prediction proved the conformation of MDP1 as an α-helix peptide. Helical-wheel projection analysis indicated the proper orientation of hydrophobic amino acid residues for membrane interaction. CD spectroscopy of MDP1 showed that MDP1 in SDS 10 and 30 mM adopted 87 and 91% helical conformation. Atomic force and confocal microscopy assessments as well as molecular dynamics studies revealed the peptide-bacterial membrane interaction. MDP1, at the concentration of 0.32 µg mL-1, demonstrated a fold reduction of 21.7 ± 1.8, 1.7 ± 0.2, and 7.3 ± 0.8 in intracellular bacterial load for ATCC, VRSA, and MRSA, respectively. Molecular dynamics results demonstrate a preferential interaction of MDP1 with POPG/POPE membranes, primarily driven by electrostatic forces and hydrogen bonding. In POPC systems, two out of four MDP1 interacted effectively, while all four MDP1 engaged with POPG/POPE membranes. Gathering all data together, MDP1 is efficacious in the reduction of intracellular VRSA and MRSA proved by culture and epi-fluorescent microscopy although further studies should be performed to increase the intracellular activity of MDP1.

7.
Pharmacol Res ; 209: 107430, 2024 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-39332751

RESUMO

The therapeutic potential of bee venom-derived peptides, particularly apamin and melittin, in cancer treatment has garnered significant attention as a promising avenue for advancing oncology. This systematic review examines preclinical studies highlighting the emerging role of these peptides in enhancing cancer therapies. Melittin and apamin, when conjugated with other therapeutic agents or formulated into novel delivery systems, have demonstrated improved efficacy in targeting tumor cells. Key findings indicate that melittin-based conjugates, such as polyethylene glycol (PEG)ylated versions, show potential in enhancing therapeutic outcomes and minimizing toxicity across various cancer models. Similarly, apamin-conjugated formulations have improved the efficacy of established anti-cancer drugs, contributing to enhanced targeting and reduced systemic toxicity. These developments underscore a growing interest in leveraging bee venom-derived peptides as adjuncts in cancer therapy. The integration of these peptides into treatment regimens offers a promising strategy to address current limitations in cancer treatment, such as drug resistance and off-target effects. However, comprehensive validation through clinical trials is essential to confirm their safety and effectiveness in human patients. This review highlights the global emergence of bee venom-derived peptides in cancer treatment, advocating for continued research and development to fully realize their therapeutic potential.

8.
Int J Biol Macromol ; 281(Pt 1): 136119, 2024 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-39343259

RESUMO

Apitherapy has a long history in treating Parkinson's disease (PD) in humans, with evidence suggesting that bee venom (BV) can mitigate Parkinson's symptoms. Central to BV's effects is melittin (MLT), a principal peptide whose neuroprotective mechanisms in PD are not fully understood. The study investigated the effects of MLT on an experimental PD model in mice and dopaminergic neuron cells, induced by MPTP or MPP+. We concentrate on the autophagic response elicited by MLT during PD pathogenesis. The findings showed that MLT was shown to protect against MPP+/MPTP cytotoxicity and preserve tyrosine hydroxylase (TH) levels, indicating neuronal safeguarding. Remarkably, MLT instigated mitophagy, enhancing mitochondrial homeostasis in MPP+-exposed SH-SY5Y cells. Further, MLT's promotion of mitophagy was confirmed to be AMPK/mTOR signaling-dependent. Validation using Bafilomycin A1, an autophagy inhibitor, confirmed MLT's neuroprotective role, with autophagy inhibition negating MLT's benefits and reducing TH preservation. These findings illuminate MLT's therapeutic potential, particularly its modulation of mitochondrial dysfunction in PD pathology. Our research advances the understanding of MLT's mechanistic action, emphasizing its role in mitochondrial autophagy and AMPK/mTOR signaling, offering a novel perspective beyond the symptomatic relief associated with BV.

9.
Biomolecules ; 14(9)2024 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-39334885

RESUMO

Antimicrobial peptides (AMPs) are believed to be a prominent alternative to the common antibiotics. However, despite decades of research, there are still no good clinical examples of peptide-based antimicrobial drugs for system application. The main reasons are loss of activity in the human body, cytotoxicity, and low selectivity. To overcome these challenges, a well-established structure-function relationship for AMPs is critical. In the present study, we focused on the well-known examples of melittin and magainin to investigate in detail the initial stages of AMP interaction with lipid membranes at low peptide-to-lipid ratio. By combining the patch-clamp technique with the bioelectrochemical method of intramembrane field compensation, we showed that these peptides interact with the membrane in different ways: melittin inserts deeper into the lipid bilayer than magainin. This difference led to diversity in pore formation. While magainin, after a threshold concentration, formed the well-known toroidal pores, allowing the translocation of the peptide through the membrane, melittin probably induced predominantly pure lipidic pores with a very low rate of peptide translocation. Thus, our results shed light on the early stages of peptide-membrane interactions and suggest new insights into the structure-function relationship of AMPs based on the depth of their membrane insertion.


Assuntos
Bicamadas Lipídicas , Magaininas , Meliteno , Meliteno/química , Meliteno/metabolismo , Meliteno/farmacologia , Magaininas/química , Magaininas/farmacologia , Magaininas/metabolismo , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Membrana Celular/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/química , Humanos
10.
ACS Appl Mater Interfaces ; 16(37): 49148-49163, 2024 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-39240583

RESUMO

Intracellular infections are difficult to treat, as pathogens can take advantage of intracellular hiding, evade the immune system, and persist and multiply in host cells. One such intracellular parasite, Leishmania, is the causative agent of leishmaniasis, a neglected tropical disease (NTD), which disproportionately affects the world's most economically disadvantaged. Existing treatments have relied mostly on chemotherapeutic compounds that are becoming increasingly ineffective due to drug resistance, while the development of new therapeutics has been challenging due to the variety of clinical manifestations caused by different Leishmania species. The antimicrobial peptide melittin has been shown to be effective in vitro against a broad spectrum of Leishmania, including species that cause the most common form, cutaneous leishmaniasis, and the most deadly, visceral leishmaniasis. However, melittin's high hemolytic and cytotoxic activity toward host cells has limited its potential for clinical translation. Herein, we report a design strategy for producing a melittin-containing antileishmanial agent that not only enhances melittin's leishmanicidal potency but also abrogates its hemolytic and cytotoxic activity. This therapeutic construct can be directly produced in bacteria, significantly reducing its production cost critical for a NTD therapeutic. The designed melittin-containing fusion crystal incorporates a bioresponsive cathepsin linker that enables it to specifically release melittin in the phagolysosome of infected macrophages. Significantly, this targeted approach has been demonstrated to be efficacious in treating macrophages infected with L. amazonensis and L. donovani in cell-based models and in the corresponding cutaneous and visceral mouse models.


Assuntos
Leishmaniose Cutânea , Leishmaniose Visceral , Meliteno , Meliteno/química , Meliteno/farmacologia , Leishmaniose Visceral/tratamento farmacológico , Animais , Camundongos , Leishmaniose Cutânea/tratamento farmacológico , Antiprotozoários/farmacologia , Antiprotozoários/química , Camundongos Endogâmicos BALB C , Humanos , Leishmania/efeitos dos fármacos , Feminino , Macrófagos/efeitos dos fármacos , Macrófagos/parasitologia , Macrófagos/metabolismo
11.
Biomed Pharmacother ; 179: 117385, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39241571

RESUMO

As the research on cancer-related treatment deepens, integrating traditional therapies with emerging interventions reveals new therapeutic possibilities. Melittin and phospholipase A2, the primary anti-cancer components of bee venom, are currently gaining increasing attention. This article reviews the various formulations of melittin in cancer therapy and its potential applications in clinical treatments. The reviewed formulations include melittin analogs, hydrogels, adenoviruses, fusion toxins, fusion peptides/proteins, conjugates, liposomes, and nanoparticles. The article also explored the collaborative therapeutic effects of melittin with natural products, synthetic drugs, radiotherapy, and gene expression regulatory strategies. Phospholipase A2 plays a key role in bee venom anti-cancer strategy due to its unique biological activity. Using an extensive literature review and the latest scientific results, this paper explores the current state and challenges of this field, with the aim to provide new perspectives that guide future research and potential clinical applications. This will further promote the application of bee venom in cancer therapy.


Assuntos
Antineoplásicos , Venenos de Abelha , Meliteno , Neoplasias , Fosfolipases A2 , Meliteno/farmacologia , Humanos , Fosfolipases A2/metabolismo , Fosfolipases A2/farmacologia , Venenos de Abelha/farmacologia , Animais , Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico
12.
Pharmaceuticals (Basel) ; 17(9)2024 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-39338374

RESUMO

Apitoxin therapy (BVT-bee venom therapy) is an emerging complementary treatment utilizing bee venom for various medical conditions. This review explores the potential and therapeutic mechanisms of bee venom, focusing on its chemical composition and the methods for its extraction and purification to enhance safety while maintaining bioactivity. Bee venom contains amphipathic peptides such as melittin and apamin, enzymes like phospholipase A2, and bioamines including histamine and catecholamines, contributing to its pleiotropic effects. The therapeutic applications of bee venom span anti-inflammatory, analgesic, antimicrobial, antiviral, neuroprotective, anti-arthritic, and anti-cancer activities. Clinical and laboratory studies have demonstrated its efficacy in treating chronic and autoimmune diseases, pain management, and improving quality of life. The immunogenic properties of bee venom necessitate ongoing research to mitigate allergic reactions, ensuring its safe and effective use in medical practice. This review summarizes the current state of research on bee venom therapy, highlighting its potential benefits and future research directions.

13.
Front Microbiol ; 15: 1419917, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39091304

RESUMO

The prevalent life-threatening microbial and cancer diseases and lack of effective pharmaceutical therapies created the need for new molecules with antimicrobial and anticancer potential. Bee venom (BV) was collected from honeybee workers, and melittin (NM) was extracted from BV and analyzed by urea-polyacrylamide gel electrophoresis (urea-PAGE). The isolated melittin was hydrolyzed with alcalase into new bioactive peptides and evaluated for their antimicrobial and anticancer activity. Gel filtration chromatography fractionated melittin hydrolysate (HM) into three significant fractions (F1, F2, and F3), that were characterized by electrospray ionization mass spectrometry (ESI-MS) and evaluated for their antimicrobial, anti-biofilm, antitumor, and anti-migration activities. All the tested peptides showed antimicrobial and anti-biofilm activities against Gram-positive and Gram-negative bacteria. Melittin and its fractions significantly inhibited the proliferation of two types of cancer cells (Huh-7 and HCT 116). Yet, melittin and its fractions did not affect the viability of normal human lung Wi-38 cells. The IC50 and selectivity index data evidenced the superiority of melittin peptide fractions over intact melittin. Melittin enzymatic hydrolysate is a promising novel product with high potential as an antibacterial and anticancer agent.

14.
Cureus ; 16(7): e65721, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39211643

RESUMO

BACKGROUND AND OBJECTIVES: In ancient China, bee venom was widely used to treat various diseases. Although using bee venom is not currently a mainstream medical method, some have applied it to treat certain conditions, including idiopathic facial paralysis (IFP). Recently, melittin (Mel), the main active component of bee venom, has been shown strong anti-inflammatory and analgesic effects. However, how bee venom improves neurological dysfunction in facial paralysis remains unknown. This study aimed to investigate the anti-neurotraumatic effect of Mel on Schwann cells (SCs), the main cells of the neuron sheath, injured by oxidative stress. METHODS: A model of hypoxic SCs was established, and CCK-8 assay, siRNA transfection, enzyme-linked immunosorbent assay, quantitative reverse transcription-polymerase chain reaction, western blot, immunofluorescence, and cell ultrastructure analyses were conducted to investigate the mitigation of hypoxia-induced damage to SCs in vitro, revealing the effects of Mel on oxidative stress injury in SCs. RESULTS: The overexpression of HIF-1α in CoCl2-induced SCs (p < 0.05) indicated the establishment of an SCs hypoxia model. The proliferation and regeneration process of the hypoxic SCs enhanced in the Mel-treated group compared to the CoCl2 group has been proven through the CCK-8 experiment (p < 0.0001) and S-100 mRNA expression detection (p < 0.0001). The increased level of reactive oxygen species (ROS) (p < 0.001) and decreased superoxide dismutase (SOD) levels (p < 0.05) in the CoCl2-induced SCs indicated that Mel can alleviate the oxidative stress damage to SCs induced by CoCl2. Mel alleviated oxidative stress and inflammation in hypoxic SCs by reducing pro-inflammatory cytokines IL-1ß (p < 0.0001) and TNF-α (p < 0.0001). In addition, Mel augmented cellular vitality and regulated indicators related to oxygen metabolism, cell repair, neurometabolism, and vascular endothelial formation after hypoxia, such as C-JUN (p < 0.05), glial cell line-derived neurotrophic factor (GDNF; p < 0.001), vascular endothelial growth factor (VEGF; p < 0.05), hypoxia-inducible factor 1-alpha (HIF-1α; p < 0.05), interleukin-1 receptor type 1 (IL-1R1; p < 0.05), enolase1 (ENO1; p < 0.05), aldose reductase (AR; p < 0.01), SOD (p < 0.05), nerve growth factor (NGF; p < 0.05), and inducible nitric oxide synthase (iNOS; p < 0.05). In terms of its mechanism, Mel inhibited the expression of proteins associated with the NF-κB pathway such as IKK (p < 0.01), p65 (p < 0.05), p60 (p < 0.001), IRAK1 (p < 0.05), and increased IKB-α (p < 0.0001). Moreover, knocking out of IL-1R1 in the si-IL-1R1 group enhanced the therapeutic effect of Mel compared to the Mel-treated group (all of which p < 0.05). CONCLUSION: This research provided evidence of the substantial involvement of IL-1R1 in oxidative stress damage caused by hypoxia in SCs and proved that Mel alleviated oxidative stress injury in SCs by targeting IL-1R1 to downregulate the NF-κB-mediated inflammatory response. Mel could potentially serve as an innovative therapeutic approach for the treatment of IFP.

15.
Toxicon ; 249: 108071, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39134227

RESUMO

Various animal toxins pose a significant threat to human safety, necessitating urgent attention to their treatment and research. The clinical potential of programmed cell death (PCD) is widely regarded as a target for envenomation, given its crucial role in regulating physiological and pathophysiological processes. Current research on animal toxins examines their specific components in pathomechanisms and injuries, as well as their clinical applications. This review explores the relationship between various toxins and several types of PCD, such as apoptosis, necroptosis, autophagy, ferroptosis, and pyroptosis, to provide a reference for future understanding of the pathophysiology of toxins and the development of their potential clinical value.


Assuntos
Apoptose , Autofagia , Toxinas Biológicas , Animais , Apoptose/efeitos dos fármacos , Humanos , Autofagia/efeitos dos fármacos , Toxinas Biológicas/toxicidade , Necroptose/efeitos dos fármacos , Piroptose/efeitos dos fármacos , Ferroptose/efeitos dos fármacos
16.
Neuropeptides ; 107: 102462, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39197274

RESUMO

OBJECTIVE: In this study, we explored the neuroprotective effect of melittin (MEL) after brain ischemia using a rat model. METHODS: The rats underwent middle cerebral artery occlusion (MCAO) for 60 min and were randomly divided into the control group, saline group, and MEL group. Rats in each group were injected intraperitoneally with MEL one day before MCAO until sacrificed. Morris water maze and rotation test were used to assess locomotor function and cognitive ability. The 9.4 Tesla MRI was used to scan and assess the infarct volume of the rat brains. Immunohistochemistry was used to detect the sites of action of MEL on microglia. Western blot and ELISA were used to measure the effect of MEL on the production of pro-inflammatory cytokines. The effect of MEL on neuronal cell apoptosis was observed by flow cytometry. RESULTS: Compared with the saline group, MEL treatment significantly increased the density of neurons in the cerebral cortical and reduced the cerebral infarct size after MCAO (33.9 ± 8.8% vs. 15.8 ± 3.9%, P < 0.05). Meanwhile, the time for MEL-treated rats to complete the water maze task on the 11th day after MCAO was significantly shorter than that of rats in the saline group (P < 0.05). MEL treatment also prolonged the rotarod retention time on day 14 after MCAO. Immunohistochemistry analysis showed that MEL inhibited the activation of microglia and suppressed the expression of TNF-α, IL-6, and IL-1ß in the brain after ischemia. MEL treatment resulted in a significant decrease in TLR4, MyD88, and NF-κB p65 levels in extracts from the ischemic cerebral cortex. Finally, MEL reduced neuronal apoptosis induced by ischemic stroke (P < 0.05). CONCLUSION: MEL treatment promotes neurological function recovery after cerebral ischemia in rats. These effects are potentially mediated through anti-inflammatory and anti-apoptotic mechanisms.


Assuntos
AVC Isquêmico , Meliteno , Neurônios , Fármacos Neuroprotetores , Ratos Sprague-Dawley , Animais , Masculino , Fármacos Neuroprotetores/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , AVC Isquêmico/tratamento farmacológico , AVC Isquêmico/metabolismo , Ratos , Meliteno/farmacologia , Meliteno/uso terapêutico , Infarto da Artéria Cerebral Média/tratamento farmacológico , Apoptose/efeitos dos fármacos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Aprendizagem em Labirinto/efeitos dos fármacos , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Modelos Animais de Doenças
17.
J Control Release ; 375: 285-299, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39216597

RESUMO

Immunotherapy is a rapidly developing and effective strategy for cancer therapy. Among various immunotherapy approaches, peptides have garnered significant attention due to their potent immunomodulatory effects. In particular, melittin emerged as a promising candidate to enhance cancer immunotherapy by inducing immunogenic cell death, promoting the maturation of antigen-presenting cells, activating T cells, enhancing the infiltration and cytotoxicity of effector lymphocytes, and modulating macrophage phenotypes for relieving immunosuppression. However, the clinical application of melittin is limited by poor targeting and systemic toxicity. To overcome these challenges, melittin has been incorporated into biomaterials and related nanotechnologies, resulting in extended circulation time in vivo, improved targeting, reduced adverse effects, and enhanced anti-cancer immunological action. This review provides an in-depth analysis of the immunomodulatory effects of melittin-incorporated nanomedicines and examines their development and challenges for clinical cancer immunotherapy.


Assuntos
Imunoterapia , Meliteno , Nanomedicina , Neoplasias , Meliteno/administração & dosagem , Humanos , Neoplasias/terapia , Neoplasias/imunologia , Neoplasias/tratamento farmacológico , Imunoterapia/métodos , Animais , Nanomedicina/métodos , Fatores Imunológicos/administração & dosagem , Fatores Imunológicos/uso terapêutico
18.
Biomed Pharmacother ; 179: 117371, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39216447

RESUMO

The continuous activation of macrophages play a critical role in the pathogenesis of cytokine storm (CS). Considering that CS results from the participation of multiple cytokines, the therapeutic effect of a single cytokine or its receptor-targeted blockade therapy remains uncertain. Melittin, which can systematically suppress the overexpression of proinflammatory mediators via inhibiting the mitogen-activated protein kinase and nuclear factor kappa-B pathways in activated macrophages, shows great potential in alleviating CS and acute inflammatory injury (AII). However, its clinical application is limited by its hemolytic activity, non-specific cytotoxicity and lack of targeting. In this study, a folic acid-modified and melittin stable-loaded solid lipid nanoparticle (Fa-MpG@LNP) with a core-shell structure was developed for CS control via targeted inhibition of the overproduction of proinflammatory mediators in activated macrophages with specific expression of folate receptor-ß. The resultant Fa-MpG@LNP showed ideal physicochemical properties and stability, low hemolytic activity and non-specific cytotoxicity, and it can specifically bind to lipopolysaccharide (LPS)-stimulated macrophages and effectively reduce the elevated levels of proinflammatory mediators. After intravenous administration, the Fa-MpG@LNP accumulated at inflamed tissue and significantly downregulate the overproduction of proinflammatory cytokines in tissue-infiltrated macrophages, resulting in a significant decrease of cytokine concentration in inflamed tissue and serum in LPS-induced acute pneumonia mice, and finally alleviate AII with undetectable toxic side effects. These results indicate the clinical application potential of Fa-MpG@LNP in alleviating CS and its related symptoms.


Assuntos
Síndrome da Liberação de Citocina , Citocinas , Macrófagos , Meliteno , Nanopartículas , Animais , Meliteno/farmacologia , Meliteno/administração & dosagem , Camundongos , Nanopartículas/química , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Citocinas/metabolismo , Síndrome da Liberação de Citocina/tratamento farmacológico , Células RAW 264.7 , Masculino , Lipopolissacarídeos , Inflamação/tratamento farmacológico , Inflamação/patologia , Lipídeos/química , Ácido Fólico/química , Camundongos Endogâmicos C57BL , Mediadores da Inflamação/metabolismo , Lipossomos
19.
J Nanobiotechnology ; 22(1): 432, 2024 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-39034393

RESUMO

Rheumatoid arthritis (RA) involves chronic joint inflammation. Combining acupuncture and medication for RA treatment faces challenges like spatiotemporal variability, limited drug loading in acupuncture needles, and premature or untargeted drug release. Here, we designed a new type of tubular acupuncture needles, with an etched hollow honeycomb-like structure to enable the high loading of therapeutics, integrating the traditional acupuncture and drug repository into an all-in-one therapeutic platform. In these proof-of-concept experiments, we fabricated injectable hollow honeycomb electroacupuncture needles (HC-EA) loaded with melittin hydrogel (MLT-Gel), enabling the combination treatment of acupuncture stimulation and melittin therapy in a spatiotemporally synchronous manner. Since the RA microenvironment is mildly acidic, the acid-responsive chitosan (CS)/sodium beta-glycerophosphate (ß-GP)/ hyaluronic acid (HA) composited hydrogel (CS/GP/HA) was utilized to perform acupuncture stimulation and achieve the targeted release of injected therapeutics into the specific lesion site. Testing our therapeutic platform involved a mouse model of RA and bioinformatics analysis. MLT-Gel@HC-EA treatment restored Th17/Treg-mediated immunity balance, reduced inflammatory factor release (TNF-α, IL-6, IL-1ß), and alleviated inflammation at the lesion site. This novel combination of modified acupuncture needle and medication, specifically melittin hydrogel, holds promise as a therapeutic strategy for RA treatment.


Assuntos
Terapia por Acupuntura , Artrite Reumatoide , Hidrogéis , Meliteno , Agulhas , Animais , Meliteno/farmacologia , Meliteno/química , Camundongos , Artrite Reumatoide/terapia , Artrite Reumatoide/tratamento farmacológico , Hidrogéis/química , Terapia por Acupuntura/métodos , Quitosana/química , Ácido Hialurônico/química , Masculino , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL
20.
Artigo em Inglês | MEDLINE | ID: mdl-38963508

RESUMO

Listeria monocytogenes is a notable food-borne pathogen that has the ability to create biofilms on different food processing surfaces, making it more resilient to disinfectants and posing a greater risk to human health. This study assessed melittin peptide's anti-biofilm and anti-pathogenicity effects on L. monocytogenes ATCC 19115. Melittin showed minimum inhibitory concenteration (MIC) of 100 µg/mL against this strain and scanning electron microscopy images confirmed its antimicrobial efficacy. The OD measurement demonstrated that melittin exhibited a strong proficiency in inhibiting biofilms and disrupting pre-formed biofilms at concentrations ranging from 1/8MIC to 2MIC and this amount was 92.59 ± 1.01% to 7.17 ± 0.31% and 100% to 11.50 ± 0.53%, respectively. Peptide also reduced hydrophobicity and self-aggregation of L. monocytogenes by 35.25% and 14.38% at MIC. Melittin also significantly reduced adhesion to HT-29 and Caco-2 cells by 61.33% and 59%, and inhibited invasion of HT-29 and Caco-2 cells by 49.33% and 40.66% for L. monocytogenes at the MIC value. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) revealed melittin's impact on gene expression, notably decreasing inlB (44%) and agrA (45%) gene expression in L. monocytogenes. flaA and hly genes also exhibited reduced expression. Also, significant changes were observed in sigB and prfA gene expression. These results underscore melittin's potential in combating bacterial infections and biofilm-related challenges in the food industry.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA