Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
Mol Pharm ; 21(5): 2148-2162, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38536949

RESUMEN

Triple-negative breast cancer (TNBC) is an aggressive type of breast cancer for which effective therapies are lacking. Targeted remodeling of the immunosuppressive tumor microenvironment (TME) and activation of the body's immune system to fight tumors with well-designed nanoparticles have emerged as pivotal breakthroughs in tumor treatment. To simultaneously remodel the immunosuppressive TME and trigger immune responses, we designed two potential therapeutic nanodelivery systems to inhibit TNBC. First, the bromodomain-containing protein 4 (BRD4) inhibitor JQ1 and the cyclooxygenase-2 (COX-2) inhibitor celecoxib (CXB) were coloaded into chondroitin sulfate (CS) to obtain CS@JQ1/CXB nanoparticles (NPs). Then, the biomimetic nanosystem MM@P3 was prepared by coating branched polymer poly(ß-amino ester) self-assembled NPs with melittin embedded macrophage membranes (MM). Both in vitro and in vivo, the CS@JQ1/CXB and MM@P3 NPs showed excellent immune activation efficiencies. Combination treatment exhibited synergistic cytotoxicity, antimigration ability, and apoptosis-inducing and immune activation effects on TNBC cells and effectively suppressed tumor growth and metastasis in TNBC tumor-bearing mice by activating the tumor immune response and inhibiting angiogenesis. In summary, this study offers a novel combinatorial immunotherapeutic strategy for the clinical TNBC treatment.


Asunto(s)
Azepinas , Celecoxib , Triazoles , Neoplasias de la Mama Triple Negativas , Microambiente Tumoral , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/patología , Microambiente Tumoral/efectos de los fármacos , Animales , Femenino , Ratones , Humanos , Celecoxib/administración & dosificación , Línea Celular Tumoral , Sulfatos de Condroitina/química , Sulfatos de Condroitina/administración & dosificación , Nanopartículas/química , Nanopartículas/administración & dosificación , Meliteno/administración & dosificación , Meliteno/química , Apoptosis/efectos de los fármacos , Sistema de Administración de Fármacos con Nanopartículas/química , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones Endogámicos BALB C , Inhibidores de la Ciclooxigenasa 2/administración & dosificación , Inhibidores de la Ciclooxigenasa 2/farmacología , Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Polímeros/química , Ratones Desnudos , Sistemas de Liberación de Medicamentos/métodos
2.
Pharmacol Res ; 175: 105993, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34801680

RESUMEN

A dysregulation of the wound healing process can lead to the development of various intractable ulcers or excessive scar formation. Therefore it is essential to identify novel pharmacological strategies to promote wound healing and restore the mechanical integrity of injured tissue. The goal of the present study was to formulate a nano-complex containing melittin (MEL) and diclofenac (DCL) with the aim to evaluate their synergism and preclinical efficacy in an in vivo model of acute wound. After its preparation and characterization, the therapeutic potential of the combined nano-complexes was evaluated. MEL-DCL nano-complexes exhibited better regenerated epithelium, keratinization, epidermal proliferation, and granulation tissue formation, which in turn showed better wound healing activity compared to MEL, DCL, or positive control. The nano-complexes also showed significantly enhanced antioxidant activity. Treatment of wounded skin with MEL-DCL nano-complexes showed significant reduction of interleukin-6 (IL-6), IL-1ß, and tumor necrosis factor-α (TNF-α) pro-inflammatory markers that was paralleled by a substantial increase in mRNA expression levels of collagen, type I, alpha 1 (Col1A1) and collagen, type IV, alpha 1 (Col4A1), and hydroxyproline content as compared to individual drugs. Additionally, MEL-DCL nano-complexes were able to significantly increase hypoxia-inducible factor 1-alpha (HIF-1α) and transforming growth factor beta 1 (TGF-ß1) proteins expression compared to single drugs or negative control group. SB431542, a selective inhibitor of type-1 TGF-ß receptor, significantly prevented in our in vitro assay the wound healing process induced by the MEL-DCL nano-complexes, suggesting a key role of TGF-ß1 in the wound closure. In conclusion, the nano-complex of MEL-DCL represents a novel pharmacological tool that can be topically applied to improve wound healing.


Asunto(s)
Antiinflamatorios no Esteroideos/administración & dosificación , Citocinas/metabolismo , Diclofenaco/administración & dosificación , Hidrogeles/administración & dosificación , Meliteno/administración & dosificación , Nanoestructuras/administración & dosificación , Cicatrización de Heridas/efectos de los fármacos , Animales , Células Cultivadas , Sinergismo Farmacológico , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Queratinocitos/efectos de los fármacos , Masculino , Ratas Wistar , Piel/efectos de los fármacos , Piel/metabolismo
3.
J Mater Sci Mater Med ; 32(7): 75, 2021 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-34156547

RESUMEN

Methicillin-resistant and Vancomycin-resistant Staphylococcus aureus bacteria (MRSA and VRSA, respectively) can seriously jeopardizes bone implants. This research aimed to examine the potential synergistic effects of Melittin and vancomycin in preventing MRSA and VRSA associated bone implant infections. Chitosan/bioactive glass nanoparticles/vancomycin composites were coated on hydrothermally etched titanium substrates by casting method. The composite coatings were coated by Melittin through drop casting technique. Melittin raised the proliferation of MC3T3 cells, making it an appropriate option as osteoinductive and antibacterial substance in coatings of orthopedic implants. Composite coatings having combined vancomycin and Melittin eliminated both planktonic and adherent MRSA and VRSA bacteria, whereas coatings containing one of them failed to kill the whole VRSA bacteria. Therefore, chitosan/bioactive glass/vancomycin/Melittin coating can be used as a bone implant coating because of its anti-infective properties.


Asunto(s)
Biopelículas , Cerámica/química , Meliteno/administración & dosificación , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Ortopedia , Prótesis e Implantes , Staphylococcus aureus/efectos de los fármacos , Resistencia a la Vancomicina/efectos de los fármacos , Vancomicina/administración & dosificación , Vancomicina/química , Células 3T3 , Animales , Antibacterianos/farmacología , Péptidos Antimicrobianos/farmacología , Adhesión Bacteriana/efectos de los fármacos , Sustitutos de Huesos/química , Proliferación Celular , Quitosano/química , Materiales Biocompatibles Revestidos/química , Meliteno/química , Ratones , Microscopía Electrónica de Rastreo , Nanopartículas/química , Polvos , Propiedades de Superficie , Titanio/química
4.
Microb Pathog ; 127: 60-69, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30513367

RESUMEN

Bacterial infections caused by antibiotic resistant bacteria are the leading cause of morbidity and mortality after burn injuries. This issue has driven the need for promising antimicrobial drugs to eradication of bacterial pathogens. Accordingly, we aimed to determine the therapeutic value of melittin, as a natural Antimicrobial peptide (AMP), in eradication of extensively drug-resistant (XDR) Acinetobacter spp. on a mouse model of third degree burn infection. Melittin killed all examined XDR isolates at 4 µg/mL up to 3 h. Melittin caused significant fluorescence release from XDR isolates at the minimum dose of 0.062 µg/mL. Vesicle formation on the membrane and squeezing of bacteria followed by cell lysis indicated the membranolytic effect of melittin. Melittin at 32 µg/mL completely eradicated the colonized XDR bacteria on infected burn mice during 2 h. No toxicity was observed on injured or healthy derma, as well as circulating Red Blood Cells (RBCs) in the examined mice. Potent promising antibacterial activity of melittin and the lack of toxicity at the therapeutic dose can clarify that melittin can be implemented as a topical drug lead in a preclinical trial of third degree burn infections.


Asunto(s)
Infecciones por Acinetobacter/tratamiento farmacológico , Acinetobacter/efectos de los fármacos , Antiinfecciosos/administración & dosificación , Quemaduras/complicaciones , Farmacorresistencia Bacteriana Múltiple , Meliteno/administración & dosificación , Infección de Heridas/tratamiento farmacológico , Infecciones por Acinetobacter/microbiología , Animales , Antiinfecciosos/efectos adversos , Bacteriólisis/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Meliteno/efectos adversos , Ratones , Viabilidad Microbiana/efectos de los fármacos , Resultado del Tratamiento
5.
Nano Lett ; 18(8): 4762-4770, 2018 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-30028623

RESUMEN

Specific drug delivery to metastatic tumors remains a great challenge for antimetastasis therapy. We herein report a bioengineered macrophage-based delivery system (LD-MDS) that can be preferentially delivered to lung metastases and intelligently transformed into nanovesicles and secondary nanovesicles for antimetastasis therapy. LD-MDS was prepared by anchoring a legumain-specific propeptide of melittin (legM) and cytotoxic soravtansine (DM4) prodrug onto the membrane of living macrophages. LD-MDS is responsively activated by legumain protease and converted into DM4-loaded exosome-like nanovesicles (DENs), facilitating efficient internalization by metastatic 4T1 cancer cells and considerable cell death. Afterward, the damaged 4T1 cells can release secondary nanovesicles and free drug molecules to destroy neighboring cancer cells. In vivo, LD-MDS displays superior targeting efficiency for lung metastatic lesions with diameters less than 100 µm and remarkably inhibits lung metastasis. This study provides a new opportunity to explore endogenous macrophages as living drug delivery vehicles with controlled drug release to target metastatic lung tumors.


Asunto(s)
Antineoplásicos/química , Portadores de Fármacos/química , Neoplasias Pulmonares/tratamiento farmacológico , Macrófagos/química , Nanopartículas/química , Animales , Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cisteína Endopeptidasas/metabolismo , Liberación de Fármacos , Humanos , Neoplasias Pulmonares/patología , Macrófagos/citología , Maitansina/administración & dosificación , Maitansina/química , Meliteno/administración & dosificación , Meliteno/química , Ratones Desnudos , Metástasis de la Neoplasia , Profármacos/administración & dosificación , Profármacos/química
6.
Drug Dev Ind Pharm ; 44(6): 982-987, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29325460

RESUMEN

Melittin, a small water-soluble cationic amphipathic α-helical linear peptide, consisted of 26 amino acids, is the honeybee venom major constituent. Several reports have proved the lytic and apoptotic effects of melittin in several cancerous cell lines. In this study, we aimed to fabricate an AS1411 aptamer-melittin to specifically deliver melittin to nucleolin positive cells (A549). Melittin was covalently attached to antinucleolin aptamer (AS1411) and its toxicity in A549 (nucleolin positive) and L929 (nucleolin negative) was studied using MTT and Annexin V flow cytometry methods. Aptamer-melittin conjugate formation was confirmed by gel electrophoresis. Hemolytic effect of aptamer-melittin conjugate was compared to melittin alone. The aptamer-melittin conjugate showed efficient cell uptake and was more cytotoxic in A549 cells than melittin (p < .001). This complex was less toxic in control cells. Competitive inhibition assay confirmed that aptamer-melittin complex delivery occurred through receptor-ligand interaction on the cell surface. Moreover, aptamer-melittin showed a significantly less hemolytic activity as compared with free melittin. This study showed that melittin could be specifically delivered to A549 cells when it was covalently conjugated to antinucleolin aptamer (AS1411) in vitro. This system can reduce the cytotoxic effects of melittin on cells with no nucleolin receptor overexpression which comprise most of normal cells such as L929 cells.


Asunto(s)
Antineoplásicos/farmacología , Sistemas de Liberación de Medicamentos/métodos , Meliteno/administración & dosificación , Oligodesoxirribonucleótidos/farmacología , Aptámeros de Nucleótidos , Línea Celular Tumoral , Humanos , Fosfoproteínas , Proteínas de Unión al ARN , Nucleolina
7.
Nanomedicine ; 13(7): 2325-2328, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28712916

RESUMEN

Issues concerning non-specificity, degradation and hemolysis severely hamper the development of membranolytic amphiphilic peptides into safe and efficient anticancer agents. To increase the therapeutic potential, we have previously developed a strategy based on formulation of the peptides in biocompatible nanosized lipodisks. Studies using melittin as model peptide show that the proteolytic degradation and hemolytic effect of the peptide are substantially reduced upon loading in lipodisks. Here, we explored the possibilities to increase the specificity and boost the cytotoxicity of melittin to tumor cells by use of targeting lipodisk. We demonstrate that small (~20 nm) EGF-targeted lipodisks can be produced and loaded with substantial amounts of peptide (lipid/peptide molar ratio >7) by means of a simple and straightforward preparation protocol. In vitro cell studies confirm specific binding of the peptide-loaded disks to tumor cells and suggest that cellular internalization of the disks results in a significantly improved cell-killing effect.


Asunto(s)
Antineoplásicos/administración & dosificación , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Meliteno/administración & dosificación , Fosfatidilcolinas/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Línea Celular Tumoral , Humanos , Meliteno/farmacocinética , Meliteno/farmacología , Neoplasias/tratamiento farmacológico , Fosfatidiletanolaminas/química , Polietilenglicoles/química
8.
Gen Physiol Biophys ; 36(4): 373-381, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28653651

RESUMEN

Melittin is the main component of bee venom consisting of 26 amino acids that has multiple effects, including antibacterial, antiviral and anti-inflammatory in various cell types. This peptide forms pores in biological membranes and triggers cell death. Therefore it has potential as an anti-cancer therapy. However, the therapeutic application of melittin is limited due to its main side effect, hemolysis, which is especially pronounced following intravenous administration. In the present study, we formulated tetrameric melittin-carrying poly-D,L-lactic-co-glycolic acid nanoparticles (PLGA-NPs) and analyzed the lytic activity of this system on liposomes that resembles breast cancer cells. Tetrameric melittin binds avidly to PLGA-NPs with an encapsulation efficiency of 97% and retains its lytic activity demonstrating the effectiveness of PLGA-NPs as nanocarriers for this cytolytic peptide.


Asunto(s)
Preparaciones de Acción Retardada/química , Liposomas/química , Meliteno/química , Fluidez de la Membrana/efectos de los fármacos , Nanocápsulas/química , Nanocápsulas/ultraestructura , Implantes Absorbibles , Difusión , Diseño de Fármacos , Meliteno/administración & dosificación
9.
Biomed Pharmacother ; 179: 117371, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39216447

RESUMEN

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.


Asunto(s)
Síndrome de Liberación de Citoquinas , Citocinas , Macrófagos , Meliteno , Nanopartículas , Animales , Meliteno/farmacología , Meliteno/administración & dosificación , Ratones , Nanopartículas/química , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Citocinas/metabolismo , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Células RAW 264.7 , Masculino , Lipopolisacáridos , Inflamación/tratamiento farmacológico , Inflamación/patología , Lípidos/química , Ácido Fólico/química , Ratones Endogámicos C57BL , Mediadores de Inflamación/metabolismo , Liposomas
10.
J Pept Sci ; 19(10): 639-50, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24014474

RESUMEN

A random phage 12-mer peptide library and a whole-cell subtractive biopanning protocol against HepG2 cells were used to select a novel peptide-specific binding to hepatocellular carcinoma cells. As a result, peptide SLSLITMLKISR (AM-2) was screened as a novel homing peptide to hepatocellular carcinoma cells, tested by immunofluorescence and immunochemistry assays. Subsequently, peptide AM-2 was linked to melittin by A(EAAAK)2A, and the antitumor effect of this ligation product was detected by MTT assay, fluorescence-activated cell sorting, and scanning electron microscopy methods. Results of cell growth inhibition tests confirmed that the affinity of melittin was increased after being incorporated into AM-2, and AM-2-melittin specifically targeted and killed HepG2 cells in vitro. Thus, AM-2 is a valuable ligand for tumor targeting, which leads to increased binding and killing effect of hepatocellular carcinoma cells in vitro when ligated to melittin, and AM-2-melittin has a clinical potential application as target agents for the treatment of human hepatocellular carcinoma.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Meliteno/administración & dosificación , Péptidos/administración & dosificación , Secuencia de Aminoácidos , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Neoplasias Hepáticas/patología , Biblioteca de Péptidos , Péptidos/química , Unión Proteica
11.
Apoptosis ; 17(1): 61-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21928088

RESUMEN

Acute hepatic failure remains an extremely poor prognosis and still results in high mortality. Therefore, better treatment is urgently needed. Melittin, a major component of bee venom, is known to inhibit inflammatory reactions induced by lipopolysaccharide (LPS) or tumor necrosis factor (TNF)-α in various cell types. However, there is no evidence of the anti-inflammatory and anti-apoptotic effect of melittin on liver cells. In the present study, we investigated the effects of melittin on D: -galactosamine (GalN)/lipopolysaccharide (LPS)-induced acute hepatic failure. Acute liver injury was induced with GalN/LPS to determine in vivo efficacy of melittin. Mice were randomly divided into four groups: sterile saline treated group (NC), melittin only treated group (NM), GalN/LPS-treated group (GalN/LPS), and GalN/LPS treated with melittin group (M+GalN/LPS). Mice were given intraperitoneal GalN/LPS with or without melittin treatment. Liver injury was assessed biochemically and histologically. Inflammatory cytokines in the serum, apoptosis of hepatocytes, and cleavage of caspase-3 in the liver were determined. The expression of TNF-α and interleukin (IL)-1ß were increased in the GalN/LPS group. However, treatment of melittin attenuated the increase of inflammatory cytokines. The M+GalN/LPS group showed significantly fewer apoptotic cells compared to the GalN/LPS group. Melittin significantly inhibited the expression of caspase and bax protein levels as well as cytochrome c release in vivo. In addition, melittin prevented the activation of the transcription factor nuclear factor-kappa B (NF-κB) induced by GalN/LPS. These results clearly indicate that melittin provided protection against GalN/LPS-induced acute hepatic failure through the inhibition of inflammatory cytokines and apoptosis.


Asunto(s)
Antiinflamatorios/administración & dosificación , Apoptosis/efectos de los fármacos , Fallo Hepático Agudo/inmunología , Meliteno/administración & dosificación , Sustancias Protectoras/administración & dosificación , Animales , Citocinas/inmunología , Modelos Animales de Enfermedad , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Humanos , Fallo Hepático Agudo/tratamiento farmacológico , Fallo Hepático Agudo/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL
12.
Neurochem Res ; 37(10): 2222-8, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22851351

RESUMEN

Intradermal injection of an active compound of European honeybee toxin, melittin, into the forearm in humans produces temporary pain and evokes sustained increase of local skin temperature. This increase of skin temperature is suppressed by the pretreatment of a voltage gated sodium channel blocker, lidocaine, suggesting that neurogenic inflammation is involved in the skin temperature increase after the melittin treatment. In this study, we tested a hypothesis that the melittin-induced skin temperature increase is augmented by an N-methyl-D-aspartate (NMDA) glutamate receptor that is present on the peripheral terminals of cutaneous primary afferents. Skin temperature was examined after the local application of incremental doses of melittin by a computer-assisted-thermography in pentobarbital-anesthetized rats. Local subcutaneous glutamate was collected through a microdialysis probe and glutamate levels were measured by a high pressure liquid chromatography with electrochemical detection method. Intraplantar injection of melittin resulted in the increase of subcutaneous glutamate levels and the increase of local skin temperature, which was partially attenuated by co-injection of an NMDA receptor antagonist, MK-801. In addition, intraplantar injection of NMDA itself increased the local skin temperature. Our data suggest that melittin-induced increase of skin temperature is enhanced through the activation of peripheral NMDA receptors by locally released glutamate. We suggest that topical administration of NMDA receptor antagonists could be an effective treatment of neuro-inflammatory pain.


Asunto(s)
Meliteno/toxicidad , Receptores de N-Metil-D-Aspartato/fisiología , Termografía , Animales , Cromatografía Líquida de Alta Presión , Masculino , Meliteno/administración & dosificación , Ratas , Ratas Sprague-Dawley
13.
Biomed Pharmacother ; 147: 112670, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35123230

RESUMEN

Methicillin-resistant Staphylococcus epidermidis (MRSE) bacteria are being recognized as true pathogens as they are able to resist methicillin and commonly form biofilms. Recent studies have shown that antimicrobial peptides (AMPs) are promising agents against biofilm-associated bacterial infections. In this study, we aimed to explore the antibiofilm activity of melittin, either alone or in combination with vancomycin and rifampin, against biofilm-producing MRSE strains. Minimum biofilm preventive concentration (MBPC), minimum biofilm inhibition concentration (MBIC), and minimum biofilm eradication concentration (MBEC), as well as fractional biofilm preventive-, inhibitory-, and eradication concentrations (FBPCi, FBICi, and FBECi), were determined for the antimicrobial agents tested. Cytotoxicity and hemolytic activity of melittin at its synergistic concentration were examined on human embryonic kidney cells (HEK-293) and Red Blood Cells (RBCs), respectively. The effect of melittin on the downregulation of biofilm-associated genes was explored using Real-Time PCR. MBPC, MBIC, and MBEC values for melittin were in the range of 0.625-20, 0.625-20, and 10-40 µg/µL, respectively. Melittin showed high synergy (FBPCi, FBICi and FBECi < 0.5). The synergism resulted in a 64-512-fold, 2-16 and 2-8-fold reduction in melittin, rifampicin and vancomycin concentrations, respectively. The synergistic melittin concentration found to be effective did not manifest either cytotoxicity on HEK-293 or hemolytic activity on RBCs. Results showed that melittin downregulated the expression of biofilm-associated icaA, aap, and psm genes in all isolates tested, ranging from 0.04-folds to 2.11-folds for icaA and from 0.05 to 3.76-folds for aap and psm. The preventive and therapeutic indexes of melittin were improved 8-fold when combined with vancomycin and rifampin. Based on these findings, the combination of melittin with conventional antibiotics could be proposed for treating or preventing biofilm-associated MRSE infections.


Asunto(s)
Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Meliteno/farmacología , Resistencia a la Meticilina , Staphylococcus epidermidis/efectos de los fármacos , Antibacterianos/administración & dosificación , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Farmacorresistencia Bacteriana Múltiple , Sinergismo Farmacológico , Quimioterapia Combinada , Genes Bacterianos , Células HEK293 , Humanos , Meliteno/administración & dosificación , Pruebas de Sensibilidad Microbiana , Rifampin/administración & dosificación , Rifampin/farmacología , Vancomicina/administración & dosificación , Vancomicina/farmacología
14.
Pharmazie ; 66(5): 362-7, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21699070

RESUMEN

Melittin liposomes surface modified with poloxamer 188 were developed, and the effect of poloxamer 188 was investigated with regard to anti-cancer effect and vascular stimulation. Melittin liposomes surface modified with poloxamer 188 at different concentrations (0%, 2%, and 5%) were prepared using the adsorption method, followed by in vitro characterization, including entrapment efficiency, zeta potential, particle size, and morphology. Subsequently, the influence of repeated freeze-thawing on the liposomes was investigated, and the effect of poloxamer 188 on the repeated freeze-thawing process was explored. Vascular stimulation effects of MLT, and MLT liposome that surface coated with or without poloxamer were all studied. Pharmacokinetics of the different MLT preparations were determined and the anticancer activity of the MLT formulations was investigated. The particle size of the liposomes gradually increased with increasing poloxamer 188 content, while the entrapment efficiency did not change significantly. After the first freeze-thaw cycle, size and PDI were both markedly reduced, entrapment efficiency rose, and there was no significant change of zeta potential. The vascular irritation caused by MLT could be reduced to an extent by encapsulation in liposome, but not completely eliminated, while liposomes coated with poloxamer 188 can effectively abolish the phenomenon. Melittin liposomes with surface modified by poloxamer exhibit enhanced bioavailability, effective anticancer activity, and reduced side effects compared with melittin solution. Poloxamer plays an important role in melittin liposomes.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Liposomas/química , Meliteno/química , Meliteno/farmacología , Poloxámero/química , Tensoactivos/química , Animales , Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Química Farmacéutica , Portadores de Fármacos , Composición de Medicamentos , Sistemas de Liberación de Medicamentos , Ensayo de Inmunoadsorción Enzimática , Técnicas In Vitro , Masculino , Meliteno/administración & dosificación , Ratones , Ratones Endogámicos ICR , Microscopía Electrónica de Transmisión , Tamaño de la Partícula , Conejos , Ratas , Ratas Sprague-Dawley , Vasculitis/inducido químicamente , Venas/patología
15.
ACS Appl Mater Interfaces ; 13(15): 17158-17173, 2021 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-33847113

RESUMEN

Melittin (MLT) has been studied preclinically as an anticancer agent based on its broad lytic effects in multiple tumor types. However, unsatisfactory tissue distribution, hemolysis, rapid metabolism, and limited specificity are critical obstacles that limit the translation of MLT. Emerging drug delivery strategies hold promise for targeting, controlled drug release, reduced side effects, and ultimately improved treatment efficiency. In this review, we discuss recent advances in the use of diverse carriers to deliver MLT, with an emphasis on the design and mechanisms of action. We further outline the opportunities for MLT-based cancer immunotherapy.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Meliteno/administración & dosificación , Meliteno/farmacología , Neoplasias/tratamiento farmacológico , Animales , Humanos , Meliteno/uso terapéutico
16.
Int J Biol Sci ; 17(14): 3818-3836, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34671201

RESUMEN

Rationale: In intracranial arterial dolichoectasia (IADE) development, the feedback loop between inflammatory cytokines and macrophages involves TNF-α and NF-κB signaling pathways and leads to subsequent MMP-9 activation and extracellular matrix (ECM) degeneration. In this proof-of-concept study, melittin-loaded L-arginine-coated iron oxide nanoparticle (MeLioN) was proposed as the protective measure of IADE formation for this macrophage-mediated inflammation and ECM degeneration. Methods: IADE was created in 8-week-old C57BL/6J male mice by inducing hypertension and elastase injection into a basal cistern. Melittin was loaded on the surface of ION as a core-shell structure (hydrodynamic size, 202.4 nm; polydispersity index, 0.158). Treatment of MeLioN (2.5 mg/kg, five doses) started after the IADE induction, and the brain was harvested in the third week. In the healthy control, disease control, and MeLioN-treated group, the morphologic changes of the cerebral arterial wall were measured by diameter, thickness, and ECM composition. The expression level of MMP-9, CD68, MCP-1, TNF-α, and NF-κB was assessed from immunohistochemistry, polymerase chain reaction, and Western blot assay. Results: MeLioN prevented morphologic changes of cerebral arterial wall related to IADE formation by restoring ECM alterations and suppressing MMP-9 expression. MeLioN inhibited MCP-1 expression and reduced CD68-positive macrophage recruitments into cerebral arterial walls. MeLioN blocked TNF-α activation and NF-κB signaling pathway. In the Sylvian cistern, co-localization was found between the CD68-positive macrophage infiltrations and the MeLioN distributions detected on Prussian Blue and T2* gradient-echo MRI, suggesting the role of macrophage harboring MeLioN. Conclusions: The macrophage infiltration into the arterial wall plays a critical role in the MMP-9 secretion. MeLioN, designed for ION-mediated melittin delivery, effectively prevents IADE formation by suppressing macrophage-mediated inflammations and MMP activity. MeLioN can be a promising strategy preventing IADE development in high-risk populations.


Asunto(s)
Arterias Cerebrales/patología , Trastornos Cerebrovasculares/prevención & control , Inflamación/prevención & control , Macrófagos/fisiología , Nanopartículas de Magnetita/uso terapéutico , Meliteno/administración & dosificación , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Trastornos Cerebrovasculares/patología , Quimiocina CCL2/antagonistas & inhibidores , Quimiocina CCL2/metabolismo , Modelos Animales de Enfermedad , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo
17.
Int J Pharm ; 577: 119071, 2020 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-31991184

RESUMEN

Gating modifier toxins (GMTs) from animal venom have shown great potential in controlling blood glucose levels in type II diabetes (T2D), but their high acute toxicity and quick clearance in the body hamper their potential therapeutic use. Inspired by their highly positive charge, we have developed a nanocomplex system based on polyelectrolytes, in which strong interactions form between positively charged GMTs and negatively charged dextran sulfate (DS). Using melittin as a model GMT and adapting flash nanocomplexation (FNC) technology for complex preparation, uniform nanocomplexes (polydispersity index: ~0.1) with high melittin encapsulation efficiency (~100%), high payload capacity (~30%), and tunable release profiles were formulated. In contrast to the high acute liver toxicity and low survival rate (60% after 8 days) observed after a single intraperitoneal (i.p.) injection of 3 mg/kg free melittin, melittin-loaded nanocomplexes displayed improved safety (100% survival after 8 days) due to prolonged melittin release. In a mouse model of T2D, a single i.p. injection of nanocomplexes decreased the blood glucose level to 12 mmol/L within 12 h and maintained it within the therapeutic range (<15 mmol/L) for 48 h. In addition, body weight decreased following treatment. This GMT/DS binary system shows great promise due to its simple components, facile preparation method, and enhanced potential druggability, including a decreased dosing frequency, decreased acute toxicity, and improved pathological indicators.


Asunto(s)
Glucemia/efectos de los fármacos , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Meliteno/administración & dosificación , Animales , Preparaciones de Acción Retardada , Sulfato de Dextran/química , Portadores de Fármacos/química , Liberación de Fármacos , Femenino , Masculino , Meliteno/farmacología , Meliteno/toxicidad , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Nanopartículas , Polielectrolitos/química , Pruebas de Toxicidad Aguda
18.
Nat Commun ; 11(1): 1110, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-32111828

RESUMEN

Targeted delivery of a nanovaccine loaded with a tumor antigen and adjuvant to the lymph nodes (LNs) is an attractive approach for improving cancer immunotherapy outcomes. However, the application of this technique is restricted by the paucity of suitable tumor-associated antigens (TAAs) and the sophisticated technology required to identify tumor neoantigens. Here, we demonstrate that a self-assembling melittin-lipid nanoparticle (α-melittin-NP) that is not loaded with extra tumor antigens promotes whole tumor antigen release in situ and results in the activation of antigen-presenting cells (APCs) in LNs. Compared with free melittin, α-melittin-NPs markedly enhance LN accumulation and activation of APCs, leading to a 3.6-fold increase in antigen-specific CD8+ T cell responses. Furthermore, in a bilateral flank B16F10 tumor model, primary and distant tumor growth are significantly inhibited by α-melittin-NPs, with an inhibition rate of 95% and 92%, respectively. Thus, α-melittin-NPs induce a systemic anti-tumor response serving as an effective LN-targeted whole-cell nanovaccine.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Sistemas de Liberación de Medicamentos , Ganglios Linfáticos/inmunología , Meliteno/administración & dosificación , Nanopartículas/administración & dosificación , Neoplasias/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/metabolismo , Línea Celular Tumoral , Citocinas/inmunología , Femenino , Inmunoterapia , Lípidos/administración & dosificación , Lípidos/química , Ganglios Linfáticos/metabolismo , Meliteno/química , Meliteno/inmunología , Meliteno/metabolismo , Ratones , Ratones Endogámicos C57BL , Nanopartículas/química , Nanopartículas/metabolismo , Neoplasias/terapia , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Theranostics ; 10(7): 3049-3063, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32194854

RESUMEN

Simultaneously targeted treatment of tumor cells and their surrounding growth-supporting immune cells is a promising strategy to reshape immunosuppressive tumor microenvironment (TME) and potentiate host innate and adaptive antitumor immune responses. Methods: We designed a series of melittin-(RADA)n hybrid peptide sequences with varying self-assembling motifs of RADA and screened out a melittin-(RADA)6 peptide that has an optimal gel-formation ability and in vitro antitumor activity. Results: The formed melittin-(RADA)6 (MR52) hydrogel scaffold could be loaded with a specific Ca2+/calmodulin-dependent protein kinase II (CAMKII) inhibitor, KN93, originally found to have both direct tumoricidal activity and macrophages-reprogramming ability, for potent immunotherapy against melanoma and hepatoma ascites in mice models. Our MR52 hydrogel has an interweaving nanofiber-like structure, possesses direct antitumor and controlled drug release properties, and promotes the enhanced intracellular uptake of loaded cargo. Compared to free KN93, the MR52-KN93 hydrogel (MRK) improved the killing effects and levels of immunogenic cell death (ICD) on tumor cells significantly. Due to the dual role of KN93, the injection of the MRK hydrogel retarded the growth of subcutaneous melanoma tumors dramatically and resulted in a high number of mature dendritic cells of draining lymph nodes, significantly enhancing the portion of cytotoxic T cells and reduced number of M2-like tumor-associated macrophages (TAMs) in tumors. Using a mouse model of malignant ascites (MAs), where traditional therapy was ineffective, we demonstrated that the MRK hydrogel treatment offered a significantly prolonged survival compared to controls. Following treatment with the MRK hydrogel, macrophages had elevated programmed cell death protein ligand-1 (PD-L1) expression, promising follow-up combined anti-PD-1 therapy that confers a cure rate of approximately 30% against MAs in mice models. Conclusion: Thus, the MRK hydrogel may serve as a prospective platform for antitumor applications.


Asunto(s)
Antineoplásicos/uso terapéutico , Ascitis/terapia , Bencilaminas/uso terapéutico , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/antagonistas & inhibidores , Hidrogeles/administración & dosificación , Inmunoterapia/métodos , Neoplasias Hepáticas Experimentales/terapia , Melanoma Experimental/terapia , Meliteno/administración & dosificación , Terapia Molecular Dirigida/métodos , Proteínas de Neoplasias/antagonistas & inhibidores , Oligopéptidos/administración & dosificación , Inhibidores de Proteínas Quinasas/uso terapéutico , Sulfonamidas/uso terapéutico , Macrófagos Asociados a Tumores/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Antineoplásicos/administración & dosificación , Ascitis/etiología , Ascitis/inmunología , Antígeno B7-H1/biosíntesis , Bencilaminas/administración & dosificación , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Técnicas de Reprogramación Celular , Composición de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Inyecciones Intraperitoneales , Neoplasias Hepáticas Experimentales/complicaciones , Neoplasias Hepáticas Experimentales/inmunología , Activación de Macrófagos , Masculino , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas de Neoplasias/fisiología , Inhibidores de Proteínas Quinasas/administración & dosificación , Distribución Aleatoria , Proteínas Recombinantes de Fusión/administración & dosificación , Sulfonamidas/administración & dosificación , Escape del Tumor/efectos de los fármacos , Macrófagos Asociados a Tumores/clasificación , Macrófagos Asociados a Tumores/enzimología
20.
Drug Dev Ind Pharm ; 35(8): 959-68, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19274512

RESUMEN

The water-soluble peptide, melittin, was modified with an anionic agent, sodium dodecyl sulfate by hydrophobic ion-pairing. Investigations showed that the formed complex was very soluble in organic solvent, especially, in dimethylsulfoxide and dehydrated alcohol. Furthermore, the physiochemical properties of the complex in the solid state or in an aqueous medium were characterized using octanol/water partition measurement, Fourier transform infrared spectroscopy, and differential scanning calorimetry. The complex was formulated into poly(D,L-lactide-co-glycolide acid) nanoparticles by an emulsion solvent diffusion method. It was found that the nanoparticles of about 130 nm in size can be produced with a high encapsulation efficiency, and the entrapment of nanoparticles prepared with the formed complex increased from about 50% to nearly 100% compared with that for pure melittin. Moreover, the growth inhibitory effects of modified melittin and melittin-loaded nanoparticles in breast cancer MCF-7 cells were not changed comparing with free melittin as determined by (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) assay.


Asunto(s)
Ácido Láctico/química , Meliteno/administración & dosificación , Ácido Poliglicólico/química , Dodecil Sulfato de Sodio/química , Tensoactivos/química , Neoplasias de la Mama/metabolismo , Rastreo Diferencial de Calorimetría , Línea Celular Tumoral , Química Farmacéutica/métodos , Dimetilsulfóxido/química , Portadores de Fármacos/química , Femenino , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Nanopartículas , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Solubilidad , Solventes/química , Espectroscopía Infrarroja por Transformada de Fourier
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda