Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
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.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
Pathog Dis ; 76(7)2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30184071

RESUMEN

Due to emergence of multidrug resistance in pathogens, the attention of the scientific community is now directed towards strengthening the reservoir of antimicrobial compounds. Prior to in vivo studies, the interaction and penetration of a hybrid peptide K11 in bacterial cells using confocal microscopy was assessed which was observed as early as 10 min after incubation with the peptide. Cell lysis along with leakage of cytoplasmic content was confirmed by electron microscopy. To evaluate the in vivo performance of the peptide, it was contained in carbopol hydrogel. Efficacy of the hydrogel formulation was then evaluated against Acinetobacter baumannii-infected wounds using a murine excision model. Treatment resulted in restoration of body weight, complete clearance of infection from the wound by day 7 and 99% wound enclosure by day 21, in contrast to the persistence of infection and 70% wound enclosure in the infected group. Further, this treatment resulted in a 2.6-fold decrease in the levels of malondialdehyde along with a 4.5-fold increase in the levels of catalase on day 3. Appearance of normal histo-architecture was observed in the treatment group. Based on these results, the peptide hydrogel can be exploited in future as one of the strategies for developing a topical anti-infective therapeutic agent.


Asunto(s)
Infecciones por Acinetobacter/tratamiento farmacológico , Antiinfecciosos/administración & dosificación , Péptidos Catiónicos Antimicrobianos/administración & dosificación , Magaininas/administración & dosificación , Meliteno/administración & dosificación , Proteínas Recombinantes de Fusión/administración & dosificación , Infección de Heridas/tratamiento farmacológico , Proteínas de Xenopus/administración & dosificación , Infecciones por Acinetobacter/microbiología , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/ultraestructura , Animales , Antiinfecciosos/farmacología , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/farmacología , Modelos Animales de Enfermedad , Hidrogel de Polietilenoglicol-Dimetacrilato/administración & dosificación , Magaininas/genética , Magaininas/farmacología , Meliteno/genética , Meliteno/farmacología , Ratones , Microscopía Confocal , Microscopía Electrónica , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Factores de Tiempo , Resultado del Tratamiento , Cicatrización de Heridas , Infección de Heridas/microbiología , Proteínas de Xenopus/genética , Proteínas de Xenopus/farmacología
12.
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
13.
Pesqui. vet. bras ; 38(4): 595-604, abr. 2018. graf
Artículo en Portugués | LILACS, VETINDEX | ID: biblio-955370

RESUMEN

A busca por alternativa aos fármacos sintéticos têm revelado descobertas no campo da farmacologia e, nesse sentido, melitina e apamina, dois constituintes do veneno de abelhas, foram descritas com várias ações farmacológicas. Este estudo objetivou avaliar in vitro as capacidades antiviral e virucida destes componentes. Para tanto, células MDBK (Madin Darby Bovine Kidney), após verificação das respectivas doses tóxicas por ensaio MTT ((3-(4,5 dimetiltiazol-2yl)-2-5-difenil-2H tetrazolato de bromo), foram cultivadas em microplacas e tratadas com diferentes concentrações de apamina, melitina e sua associação. Esse tratamento ocorreu antes e após a infecção com 0,1 MOI (multiplicidade de infecção) de cepas citopatogênicas de herpesvírus bovino tipo 1 (BoHV-1) cepa Los Angeles e vírus da diarreia viral bovina (BVDV) cepa NADL. Após incubação por 72 horas, 37oC, as células foram submetidas ao ensaio MTT para estimativa da viabilidade celular. Em experimento paralelo, placas que foram submetidas ao mesmo procedimento sofreram ciclo de congelamento e descongelamento das células, para rompimento das mesmas e mensuração dos títulos virais. O ensaio virucida foi realizado incubando-se suspensões de BoHV-1 e BVDV com as soluções de apamina, melitina e associação por 24 horas a 37oC e 22oC. O título viral foi avaliado às 0 horas, 1, 2, 4, 8 e 24 horas de incubação. A concentração citotóxica para 50% das células (CC50) de melitina foi 2,32 μg/ml e apamina não demonstrou toxicidade à maior concentração testada (100μg/ml). Houve efeito antiviral da melitina sobre BoHV-1, especialmente na concentração de 2μg/ml, onde observou-se 85,96% de viabilidade celular quando o tratamento foi realizado antes da infecção e 86,78% de viabilidade quando o tratamento foi realizado após a infecção. Houve ainda redução de 90% das partículas virais de BoHV-1. Em menores concentrações (1 e 1,5μg/ml) de melitina não houve atividade antiviral, pois a viabilidade celular foi baixa, demonstrando efeito citopático do vírus. Na associação das duas substâncias houve queda no título de BVDV e observou-se maior viabilidade celular quando comparados à ação isolada dos composto sobre este vírus. Isso se confirma na atividade virucida, uma vez que houve decréscimo de 90% das partículas virais de BVDV com a associação dos dois compostos do veneno de abelhas. Atuando individualmente, melitina apresentou efeito antiviral e virucida frente ao BoHV-1, zerando seu título em apenas 2 horas a 37oC. Conclui-se que melitina tem ação antiviral e virucida frente ao BoHV-1 e sua associação com apamina potencializou seus efeitos frente ao BVDV.(AU)


The search for an alternative to synthetic drugs have revealed discoveries in the field of pharmacology and, according to melittin and apamin, two components of bee venom which have been described were with various pharmacological actions.This study aimed to evaluate the in vitro antiviral and virucidal capabilities of these components. Therefore, after verification of their toxic doses by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay, MDBK cells (Madin Darby Bovine Kidney) have been cultivated in microplates and treated with different concentrations of apamin, melittin and its association. This treatment occurred before and after infection with MOI (multiplicity of infection) 0.1 of cytopathogenic strains of bovine herpesvirus type 1 (BoHV-1) strain Los Angeles and bovine viral diarrhea virus (BVDV) strain NADL. After incubation for 72 hours, 37°C, the cells were submitted to MTT assay to estimate cell viability. In parallel experiments, plates were subjected to the same procedure suffered freezing and thawing cycle the cells to rupture the same and measurement of viral titers. The virucidal assay was performed by incubating suspension of bovine herpesvirus type-1 and BVDV with apamin solutions, melittin and association for 24 hours at 37°C and 22°C. The viral titer was evaluated at 0 hours, 1, 2, 4, 8 and 24 hours of incubation. The cytotoxic concentration to 50% of the cells (CC50) of melittin was 2.32g/mL and apamin did not show toxicity at the greater concentration tested (100μg/mL). There was antiviral effect of melittin on bovine herpesvirus type-1, especially at a concentration of 2μg/mL, where was observed 85.96% cell viability when treatment was performed before the infection and 86.78% viability when the treatment was carried out after infection. There was also a 90% reduction of viral particles of bovine herpesvirus type-1. In lower concentrations (1 and 1.5μg/mL) melittin no antiviral activity because cell viability was low, showing cytopathic effect of the virus. At the association two substances there were a decrease in the title of BVDV and there was higher cell viability when compared to the isolated action of the compounds of this virus. This is confirmed in the virucidal activity, since there was a decrease of 90% of the viral particles of BVDV with the combination of the two compounds of bee venom. Acting individually, melittin showed antiviral effect and virucidal against for BoHV-1, zeroing its title in just 2 hours at 37°C. It is concluded that melittin has antiviral and virucidal action against the BoHV-1 and its association with apamin potentiate its effects against BVDV.(AU)


Asunto(s)
Apamina/administración & dosificación , Bovinos/anomalías , Bovinos/virología , Herpesvirus Bovino 1/inmunología , Meliteno/administración & dosificación
14.
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
15.
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
16.
J Zhejiang Univ Sci B ; 18(6): 522-531, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28585428

RESUMEN

Hepatocarcinoma is one of the malignant cancers with significant morbidity and mortality. Immunotherapy has emerged in clinical treatment, owing to the limitation and severe side effects of chemotherapy. In the immune system, natural killer (NK) cells are important effectors required to eliminate malignant tumor cells without the limitation of major histocompatibility complex (MHC) molecule issues. Hence, treatment which could stimulate NK cells is of great interest. Here, we investigated the efficacy of the combined therapy of TT-1 (a mutant of melittin) and interferon-α (IFN-α) on NK cells and human liver cancer HepG-2/Huh7 cells in vitro and in vivo, as well as the mechanism involved. The combination therapy significantly inhibited the growth of HepG-2/Huh7 cells in vivo, but this effect was impaired after depleting NK cells. TT-1 not only up-regulated MHC class I-related chain molecules A (MICA) expression, but also prevented the secretion of soluble MICA (sMICA). Both the mRNA and protein of a disintegrin and metallopeptidase 10 (ADAM 10) in HepG-2/Huh7 cells were decreased after TT-1 treatment. The combined therapy of TT-1 and IFN-α could suppress the growth of HepG-2/Huh7 xenografted tumor effectively via promoting the interaction of NK group 2, member D (NKG2D) and MICA, indicating that TT-1+IFN-α would be a potential approach in treating liver cancer.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Antígenos de Histocompatibilidad Clase I/metabolismo , Interferón-alfa/administración & dosificación , Neoplasias Hepáticas/tratamiento farmacológico , Meliteno/análogos & derivados , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Adyuvantes Inmunológicos/administración & dosificación , Secuencia de Aminoácidos , Animales , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/patología , Proliferación Celular/efectos de los fármacos , Células Hep G2 , Humanos , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Neoplasias Hepáticas Experimentales/tratamiento farmacológico , Neoplasias Hepáticas Experimentales/inmunología , Neoplasias Hepáticas Experimentales/patología , Meliteno/administración & dosificación , Meliteno/genética , Ratones , Ratones Desnudos , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/genética , Ensayos Antitumor por Modelo de Xenoinjerto
17.
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
18.
J Hematol Oncol ; 10(1): 71, 2017 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-28320480

RESUMEN

Melittin is the main effective component of bee venom and has extensive biological functions; however, serious side effects have restricted its clinical application. Preclinical and clinical studies showed that the main adverse events were allergic reaction and pain at the administration site. To decrease the toxicity, we prepared melittin nano-liposomes by encapsulating melittin with poloxamer 188 and explored the inhibitory activities on liver cancer together with biological safety. Here, we showed that melittin nano-liposomes significantly inhibited the survival of hepatocellular carcinoma (HCC) cells in vitro and prominently suppressed the growth of subcutaneous and orthotopic HCC transplantation tumors in vivo. It was important that it induced less inflammation and allergy in mice compared with melittin. Overall, melittin nano-liposomes would have a better application in HCC therapy due to its significant anti-tumor activity and better biological safety.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Meliteno/administración & dosificación , Poloxámero/uso terapéutico , Animales , Abejas , Cápsulas/química , Carcinoma Hepatocelular/complicaciones , Xenoinjertos , Humanos , Hipersensibilidad/prevención & control , Inflamación/inducido químicamente , Inflamación/prevención & control , Liposomas , Neoplasias Hepáticas/complicaciones , Meliteno/efectos adversos , Meliteno/toxicidad , Ratones , Nanopartículas
19.
Eur J Pharm Sci ; 99: 209-218, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-28024889

RESUMEN

Prostate cancer is the second leading cause of death due to cancer in men. Owing to shortcomings in the current treatments, other therapies are being considered. Toxic gene delivery is one of the most effective methods for cancer therapy. Cationic polymers are able to form stable nanoparticles via interaction with nucleic acids electrostatically. Branched polyethylenimine that contains amine groups has notable buffering capacity and the ability to escape from endosome through the proton sponge effect. However, the cytotoxicity of this polymer is high, and modification is one of the applicable strategies to overcome this problem. In this study, PEI was targeted with chlorotoxin (CTX) via N-succinimidyl 3-(2-pyridyldithio) propionate (SPDP) cross-linker. CTX can bind specifically to matrix metalloproteinase-2 that is overexpressed in certain cancers. Melittin as the major component of bee venom has been reported to have anti-cancer activity. This was thus selected to deliver to PC3 cell line. Flow cytometry analysis revealed that transfection efficiency of targeted nanoparticles is significantly higher compared to non-targeted nanoparticles. Targeted nanoparticles carrying the melittin gene also decreased cell viability of PC3 cells significantly while no toxic effects were observed on NIH3T3 cell line. Therefore, CTX-targeted nanoparticles carrying the melittin gene could serve as an appropriate gene delivery system for prostate and other MMP-2 positive cancer cells.


Asunto(s)
Meliteno/administración & dosificación , Meliteno/química , Nanopartículas/química , Neoplasias de la Próstata/terapia , Venenos de Escorpión/administración & dosificación , Venenos de Escorpión/química , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Humanos , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Células 3T3 NIH , Polietileneimina/química , Polímeros/química , Transfección/métodos
20.
PLoS One ; 11(3): e0151675, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26985663

RESUMEN

The Formosan subterranean termite, Coptotermes formosanus Shiraki, is a costly invasive urban pest in warm and humid regions around the world. Feeding workers of the Formosan subterranean termite genetically engineered yeast strains that express synthetic protozoacidal lytic peptides has been shown to kill the cellulose digesting termite gut protozoa, which results in death of the termite colony. In this study, we tested if Melittin, a natural lytic peptide from bee venom, could be delivered into the termite gut via genetically engineered yeast and if the expressed Melittin killed termites via lysis of symbiotic protozoa in the gut of termite workers and/or destruction of the gut tissue itself. Melittin expressing yeast did kill protozoa in the termite gut within 56 days of exposure. The expressed Melittin weakened the gut but did not add a synergistic effect to the protozoacidal action by gut necrosis. While Melittin could be applied for termite control via killing the cellulose-digesting protozoa in the termite gut, it is unlikely to be useful as a standalone product to control insects that do not rely on symbiotic protozoa for survival.


Asunto(s)
Venenos de Abeja/genética , Isópteros/efectos de los fármacos , Isópteros/parasitología , Meliteno/genética , Control Biológico de Vectores/métodos , Animales , Antiprotozoarios/administración & dosificación , Venenos de Abeja/administración & dosificación , Bioensayo , Ingeniería Genética , Meliteno/administración & dosificación , Saccharomyces cerevisiae/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA