Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Mol Ther ; 32(6): 1895-1916, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38549376

RESUMO

Malignant tumors are often associated with an immunosuppressive tumor microenvironment (TME), rendering most of them resistant to standard-of-care immune checkpoint inhibitors (CPIs). Signal transducer and activator of transcription 3 (STAT3), a ubiquitously expressed transcription factor, has well-defined immunosuppressive functions in several leukocyte populations within the TME. Since the STAT3 protein has been challenging to target using conventional pharmaceutical modalities, we investigated the feasibility of applying systemically delivered RNA interference (RNAi) agents to silence its mRNA directly in tumor-associated immune cells. In preclinical rodent tumor models, chemically stabilized acylated small interfering RNAs (siRNAs) selectively silenced Stat3 mRNA in multiple relevant cell types, reduced STAT3 protein levels, and increased cytotoxic T cell infiltration. In a murine model of CPI-resistant pancreatic cancer, RNAi-mediated Stat3 silencing resulted in tumor growth inhibition, which was further enhanced in combination with CPIs. To further exemplify the utility of RNAi for cancer immunotherapy, this technology was used to silence Cd274, the gene encoding the immune checkpoint protein programmed death-ligand 1 (PD-L1). Interestingly, silencing of Cd274 was effective in tumor models that are resistant to PD-L1 antibody therapy. These data represent the first demonstration of systemic delivery of RNAi agents to the TME and suggest applying this technology for immuno-oncology applications.


Assuntos
Antígeno B7-H1 , Interferência de RNA , RNA Interferente Pequeno , Fator de Transcrição STAT3 , Microambiente Tumoral , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT3/genética , Animais , Camundongos , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Linhagem Celular Tumoral , Humanos , Microambiente Tumoral/imunologia , RNA Interferente Pequeno/genética , Imunoterapia/métodos , Resistencia a Medicamentos Antineoplásicos/genética , Inibidores de Checkpoint Imunológico/farmacologia , Modelos Animais de Doenças , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Neoplasias/terapia , Neoplasias/imunologia , Neoplasias/genética
2.
In Silico Pharmacol ; 12(1): 15, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38476933

RESUMO

The emergence of antibiotic-resistant pathogens generates impairment to human health. U1-SCTRX-lg1a is a peptide isolated from a phospholipase D extracted from the spider venom of Loxosceles gaucho with antimicrobial activity against Gram-negative bacteria (between 1.15 and 4.6 µM). The aim of this study was to suggest potential receptors associated with the antimicrobial activity of U1-SCTRX-lg1a using in silico bioinformatics tools. The search for potential targets of U1-SCRTX-lg1a was performed using the PharmMapper server. Molecular docking between U1-SCRTX-lg1a and the receptor was performed using PatchDock software. The prediction of ligand sites for each receptor was conducted using the PDBSum server. Chimera 1.6 software was used to perform molecular dynamics simulations only for the best dock score receptor. In addition, U1-SCRTX-lg1a and native ligand interactions were compared using AutoDock Vina software. Finally, predicted interactions were compared with the ligand site previously described in the literature. The bioprospecting of U1-SCRTX-lg1a resulted in the identification of three hundred (300) diverse targets (Table S1), forty-nine (49) of which were intracellular proteins originating from Gram-negative microorganisms (Table S2). Docking results indicate Scores (10,702 to 6066), Areas (1498.70 to 728.40) and ACEs (417.90 to - 152.8) values. Among these, NAD + NH3-dependent synthetase (PDB ID: 1wxi) showed a dock score of 9742, area of 1223.6 and ACE of 38.38 in addition to presenting a Normalized Fit score of 8812 on PharmMapper server. Analysis of the interaction of ligands and receptors suggests that the peptide derived from brown spider venom can interact with residues SER48 and THR160. Furthermore, the C terminus (- 7.0 score) has greater affinity for the receptor than the N terminus (- 7.7 score). The molecular dynamics assay shown that free energy value for the protein complex of - 214,890.21 kJ/mol, whereas with rigid docking, this value was - 29.952.8 sugerindo that after the molecular dynamics simulation, the complex exhibits a more favorable energy value compared to the previous state. The in silico bioprospecting of receptors suggests that U1-SCRTX-lg1a may interfere with NAD + production in Escherichia coli, a Gram-negative bacterium, altering the homeostasis of the microorganism and impairing growth. Supplementary Information: The online version contains supplementary material available at 10.1007/s40203-024-00190-8.

3.
In Silico Pharmacol, v. 12, n. 15, mar. 2024
Artigo em Inglês | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: bud-5285

RESUMO

The emergence of antibiotic-resistant pathogens generates impairment to human health. U1-SCTRX-lg1a is a peptide isolated from a phospholipase D extracted from the spider venom of Loxosceles gaucho with antimicrobial activity against Gram-negative bacteria (between 1.15 and 4.6 μM). The aim of this study was to suggest potential receptors associated with the antimicrobial activity of U1-SCTRX-lg1a using in silico bioinformatics tools. The search for potential targets of U1-SCRTX-lg1a was performed using the PharmMapper server. Molecular docking between U1-SCRTX-lg1a and the receptor was performed using PatchDock software. The prediction of ligand sites for each receptor was conducted using the PDBSum server. Chimera 1.6 software was used to perform molecular dynamics simulations only for the best dock score receptor. In addition, U1-SCRTX-lg1a and native ligand interactions were compared using AutoDock Vina software. Finally, predicted interactions were compared with the ligand site previously described in the literature. The bioprospecting of U1-SCRTX-lg1a resulted in the identification of three hundred (300) diverse targets (Table S1), forty-nine (49) of which were intracellular proteins originating from Gram-negative microorganisms (Table S2). Docking results indicate Scores (10,702 to 6066), Areas (1498.70 to 728.40) and ACEs (417.90 to – 152.8) values. Among these, NAD + NH3-dependent synthetase (PDB ID: 1wxi) showed a dock score of 9742, area of 1223.6 and ACE of 38.38 in addition to presenting a Normalized Fit score of 8812 on PharmMapper server. Analysis of the interaction of ligands and receptors suggests that the peptide derived from brown spider venom can interact with residues SER48 and THR160. Furthermore, the C terminus (– 7.0 score) has greater affinity for the receptor than the N terminus (– 7.7 score). The molecular dynamics assay shown that free energy value for the protein complex of – 214,890.21 kJ/mol, whereas with rigid docking, this value was – 29.952.8 sugerindo that after the molecular dynamics simulation, the complex exhibits a more favorable energy value compared to the previous state. The in silico bioprospecting of receptors suggests that U1-SCRTX-lg1a may interfere with NAD + production in Escherichia coli, a Gram-negative bacterium, altering the homeostasis of the microorganism and impairing growth.

4.
Adv Healthc Mater ; 11(12): e2102600, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35285167

RESUMO

Biomacromolecules have long been at the leading edge of academic and pharmaceutical drug development and clinical translation. With the clinical advances of new therapeutics, such as monoclonal antibodies and nucleic acids, the array of medical applications of biomacromolecules has broadened considerably. A major on-going effort is to expand therapeutic targets within intracellular locations. Owing to their large sizes, abundant charges, and hydrogen-bond donors and acceptors, advanced delivery technologies are required to deliver biomacromolecules effectively inside cells. In this review, strategies used for the intracellular delivery of three major forms of biomacromolecules: nucleic acids, proteins, and peptides, are highlighted. An emphasis is placed on synthetic delivery approaches and the major hurdles needed to be overcome for their ultimate clinical translation.


Assuntos
Ácidos Nucleicos , Sistemas de Liberação de Medicamentos , Ácidos Nucleicos/química , Peptídeos/química , Proteínas
5.
Bioorg Med Chem Lett ; 62: 128627, 2022 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-35158043

RESUMO

Prolonging duration of action of inhaled drugs is a challenging endeavor and guidance for medicinal chemistry teams is very limited, particularly if the site of action is intracellular. Herein, we identified recent literature reports of newly designed inhaled compounds with intracellular targets and summarized learnings from different approaches and strategies undertaken by medicinal chemistry teams. We highlight key properties that have shown to lead to longer duration of action and provide guidance on the best strategy to follow while designing a new inhaled drug with an intracellular target.


Assuntos
Química Farmacêutica
6.
Nano Lett ; 21(14): 6022-6030, 2021 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-34227381

RESUMO

Despite the well-recognized clinical success of therapeutic proteins, especially antibodies, their inability to penetrate the cell membrane restricts them to secretory extracellular or membrane-associated targets. Developing a direct cytosolic protein delivery system would offer unique opportunities for intracellular target-related therapeutic proteins. Here, we generated a supercharged polypeptide (SCP) with high cellular uptake efficiency, endosomal escape ability, and good biosafety and developed an SCP with an unnatural amino acid containing the phenylboronic acid (PBA) group, called PBA-SCP. PBA-SCP is capable of potently delivering proteins with various isoelectric points and molecular sizes into the cytosol of living cells without affecting their bioactivities. Importantly, cytosolically delivered antibodies remain functional and are capable of targeting, labeling, and manipulating diverse intracellular antigens. This study demonstrates an efficient and versatile intracellular protein delivery platform, especially for antibodies, and provides new possibilities for expanding protein-based therapeutics to intracellular "undruggable" targets.


Assuntos
Peptídeos , Proteínas , Transporte Biológico , Citosol/metabolismo , Endossomos/metabolismo , Peptídeos/metabolismo
7.
Eur J Med Chem ; 221: 113521, 2021 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-34082225

RESUMO

The anthraquinone scaffold has long been known as a source of efficacious antitumor drugs. In particular, the various chemical modifications of the side chains in this scaffold have yielded the compounds potent for the wild type tumor cells, their counterparts with molecular determinants of altered drug response, as well as in vivo settings. Further exploring the chemotype of anticancer heteroarene-fused anthraquinones, we herein demonstrate that derivative of anthra[2,3-b]thiophene-2-carboxamide, (compound 8) is highly potent against a panel of human tumor cell lines and their drug resistant variants. Treatment with submicromolar or low micromolar concentrations of 8 for only 30 min was sufficient to trigger lethal damage of K562 chronic myelogenous leukemia cells. Compound 8 (2.5 µM, 3-6 h) induced an apoptotic cell death as determined by concomitant activation of caspases 3 and 9, cleavage of poly(ADP-ribose) polymerase, increase of Annexin V/propidium iodide double stained cells, DNA fragmentation (subG1 fraction) and a decrease of mitochondrial membrane potential. Neither a significant interaction with double stranded DNA nor strong inhibition of the DNA dependent enzyme topoisomerase 1 by 8 were detectable in cell free systems. Laser scanning confocal microscopy revealed that some amount of 8 was detectable in mitochondria as early as 5 min after the addition to the cells; exposure for 1 h caused significant morphological changes and clustering of mitochondria. The bioisosteric analog 2 in which the thiophene ring was replaced with furan was less active although the patterns of cytotoxicity of both derivatives were similar. These results point at the specific role of the sulfur atom in the antitumor properties of carboxamide derivatives of heteroarene-fused anthraquinone.


Assuntos
Antraquinonas/farmacologia , Antineoplásicos/farmacologia , Tiofenos/farmacologia , Antraquinonas/síntese química , Antraquinonas/química , Antineoplásicos/síntese química , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Células K562 , Estrutura Molecular , Relação Estrutura-Atividade , Tiofenos/síntese química , Tiofenos/química , Células Tumorais Cultivadas
9.
Trends Cancer ; 6(4): 299-318, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32209445

RESUMO

The ideal cancer target antigen (Ag) is expressed at high copy numbers on neoplastic cells, absent on normal tissues, and contributes to the survival of cancer cells. Despite significant investments in the identification of cell surface Ags, there is a paucity of targets that meet such ideal cancer target criteria. Recent clinical trials in patients with cancer treated with immune checkpoint inhibitors (ICIs) indicate that cluster of differentiation (CD)8+ T cells, by means of their T cell receptors (TCRs) recognizing intracellular targets presented as peptides in the context of human leukocyte antigen (peptide-human leukocyte antigen complex; pHLA) molecules on tumor cells, can mediate deep and long-lasting antitumor responses in patients with solid tumors. Therefore, pHLA-target Ags may represent the long sought-after, ideal targets for solid tumor targeting by high-potency oncology compounds.


Assuntos
Antígenos de Neoplasias/metabolismo , Antineoplásicos Imunológicos/farmacologia , Descoberta de Drogas/métodos , Neoplasias/tratamento farmacológico , Receptores de Antígenos de Linfócitos T/metabolismo , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Antineoplásicos Imunológicos/uso terapêutico , Linhagem Celular Tumoral , Simulação por Computador , Reações Cruzadas/imunologia , Mapeamento de Epitopos/métodos , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Antígenos HLA/genética , Antígenos HLA/imunologia , Antígenos HLA/metabolismo , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Ligantes , Neoplasias/imunologia , Neoplasias/patologia , Biblioteca de Peptídeos , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo
10.
Theranostics ; 8(2): 549-562, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29290826

RESUMO

Rationale: Monoclonal antibodies (mAbs) mostly targeting extracellular or cell surface molecules have been widely used in the treatment of various diseases. However, mAbs cannot pass through the cell membrane as efficiently as small compounds, thus limiting their use against intracellular targets. Methods to shuttle antibodies into living cells may largely expand research and application in areas based on mAbs. Hepatitis B virus X protein (HBx) is an important intracellular multi-functional viral protein in the life cycle of hepatitis B virus (HBV). HBx plays essential roles in virus infection and replication and is strongly associated with HBV-related carcinogenesis. Methods: In this study, we developed a cell-penetrating whole molecule antibody targeting HBx (9D11-Tat) by the fusion of a cell penetrating peptide (CPP) on the C-terminus of the heavy chain of a potent mAb specific to HBx (9D11). The anti-HBV effect and mechanism of 9D11-Tat were investigated in cell and mouse models mimicking chronic HBV infection. Results: Our results demonstrated that the recombinant 9D11-Tat antibody could efficiently internalize into living cells and significantly suppress viral transcription, replication, and protein production both in vitro and in vivo. Further analyses suggested the internalized 9D11-Tat antibody could greatly reduce intracellular HBx via Fc binding receptor TRIM21-mediated protein degradation. This process simultaneously stimulated the activations of NF-κB, AP-1, and IFN-ß, which promoted an antiviral state of the host cell. Conclusion: In summary, our study offers a new approach to target intracellular pathogenesis-related protein by engineered cell-penetrating mAb expanding their potential for therapeutic applications. Moreover, the 9D11-Tat antibody may provide a novel therapeutic agent against human chronic HBV infection.


Assuntos
Anticorpos Monoclonais/farmacologia , Peptídeos Penetradores de Células/metabolismo , Vírus da Hepatite B/efeitos dos fármacos , Hepatite B Crônica/tratamento farmacológico , Hepatite B/tratamento farmacológico , Ribonucleoproteínas/metabolismo , Transativadores/metabolismo , Animais , Antivirais/metabolismo , Antivirais/farmacologia , Linhagem Celular Tumoral , Células Hep G2 , Hepatite B Crônica/metabolismo , Humanos , Interferon beta/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/metabolismo , Fator de Transcrição AP-1/metabolismo , Proteínas Virais Reguladoras e Acessórias
11.
Recent Pat Anticancer Drug Discov ; 13(2): 159-183, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29210664

RESUMO

BACKGROUND: The anthracene-9,10-dione (anthraquinone) derivatives represent an exceptionally valuable class in anticancer drug development. An outstanding antitumor potency of the anthracycline antibiotics attracted the attention of medicinal chemists since the discovery of these chemotypes. The prominent anthraquinone-based drugs doxorubicin, mitoxantrone as well as more recent epirubicin, idarubicin, and valrubicin are successfully used in chemotherapy of hematological malignancies and solid tumors. The anthraquinone core remains a promising scaffold for the search of new optimized drug candidates. OBJECTIVE: In this study, we analyze the progress in discovery and development of antitumor anthracene- 9,10-diones based on patent and journal publications in 2008-2017. The main goal is to dissect novel chemotypes of anthraquinone derivatives; other important issues such as the success in bioconjugate chemistry of anthraquinone containing agents as well as the patents on new applications of anthracyclines are beyond the scope of this review. CONCLUSION: A number of newly discovered natural products, the perspective directions for chemical modifications to optimize the anticancer properties, and novel intracellular targets demonstrate that anthracene- 9,10-diones deserve further in-depth investigation as an important source of drug candidates.


Assuntos
Antraquinonas/química , Antineoplásicos/química , Produtos Biológicos/química , Descoberta de Drogas/métodos , Animais , Antraquinonas/uso terapêutico , Antineoplásicos/uso terapêutico , Produtos Biológicos/uso terapêutico , Descoberta de Drogas/tendências , Humanos , Neoplasias/tratamento farmacológico
12.
Artigo em Inglês | MEDLINE | ID: mdl-29164074

RESUMO

Cathelicidin-BF, derived from the banded krait (Bungarus fasciatus), is a typically cationic, amphiphilic and α-helical antimicrobial peptide (AMP) with 30 amino acids that exerts powerful effects on multidrug-resistant (MDR) clinical isolates, including Pseudomonas aeruginosa, Acinetobacter baumannii, and Klebsiella pneumoniae, but whether it targets plasma membranes or intracellular targets to kill bacteria is still controversial. In the present study, we demonstrated that the disruption of bacterial membranes with high concentrations of cathelicidin-BF was the cause of bacterial death, as with conventional antibiotics at high concentrations. At lower concentrations, cathelicidin-BF did not cause bacterial plasma membrane disruption, but it was able to cross the membrane and aggregate at the nucleoid regions. Functional proteins of the transcription processes of P. aeruginosa and A. baumannii were affected by sublethal doses of cathelicidin-BF, as demonstrated by comparative proteomics using isobaric tags for relative and absolute quantification and subsequent gene ontology (GO) analysis. Analysis using the Kyoto Encyclopedia of Genes and Genomes showed that cathelicidin-BF mainly interferes with metabolic pathways related to amino acid synthesis, metabolism of cofactors and vitamins, metabolism of purine and energy supply, and other processes. Although specific targets of cathelicidin-BF must still be validated, our study offers strong evidence that cathelicidin-BF may act upon intracellular targets to kill superbugs, which may be helpful for further efforts to discover novel antibiotics to fight against them.


Assuntos
Acinetobacter baumannii/efeitos dos fármacos , Catelicidinas/farmacologia , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Pseudomonas aeruginosa/efeitos dos fármacos , Acinetobacter baumannii/genética , Acinetobacter baumannii/crescimento & desenvolvimento , Sequência de Aminoácidos , Aminoácidos/biossíntese , Animais , Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos , Proteínas de Bactérias , Catelicidinas/administração & dosagem , Membrana Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Coenzimas/metabolismo , Citoplasma , DNA Bacteriano , Farmacorresistência Bacteriana Múltipla/genética , Feminino , Humanos , Levofloxacino/farmacologia , Redes e Vias Metabólicas/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Testes de Sensibilidade Microbiana , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/crescimento & desenvolvimento , RNA Ribossômico 16S/genética , Vitaminas/metabolismo
13.
São Paulo; 2017. 146 p.
Tese em Português | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: bud-3383

RESUMO

Crotamine (nCrot) is a polypeptide composed of 42 amino acid residues from the venom of the South American rattlesnake (Crotalus durissus terrificus). Previously, our group described nCrot as a CPP (Cell-Penetrating Peptide), since it has the ability to rapidly penetrate into different cell types through the cell membrane lipid barrier. The ability of CPPs to cross plasma membranes has been utilized for several applications, including the delivery of bioactive molecules to inhibit cellular mechanisms involved in several diseases. The feature of nCrot as CPP is directed mainly by dividing cells, which proposes tumor cells as targets. Following this idea, the aim was to evaluate its ability to carry the pro-apoptotic BAX protein into tumor cells. Thus, seven peptides were synthesized from the nCrot sequence, six of these conjugated to BAX. The peptides carried BAX into the tumor cells tested (murine/ human melanoma and human mammary tumor), four of them were capable of significantly reducing cell viability, causing morphological changes, alteration in proapoptotic proteins expression, and inducing hyperpolarization of the mitochondrial membrane resulting in apoptosis and necrosis. By hypothesis, the actions of nCrot may be related to its inhibitory capacity on the K(V)1.3 channel. The high expression of this channel is observed in several cancers, including melanoma, breast cancer, prostate, pancreas, among others. In order to evaluate its action in a pancreatic cancer model, synthetic crotamine (sCrot) was used in different biochemical aspects of pancreatic tumor cells and it was observed that sCrot did not influence cell viability and did not induce phenotypic changes when analyzed HuR protein, which is related to tumor cell survival and resistance of chemotherapeutic drugs. Finally, nCrot and sCrot CPP activity was investigated in human melanoma cells, murine melanoma, breast tumor, human T lymphocytes from leukemia, human mononuclear cells, murine fibroblasts and human keratinocytes. sCrot and nCrot demonstrated the ability to rapidly penetrate into different cells, showing preference for tumor cells. The assay was performed at different time points and in real time on the living cells for 24 hours at time-lapse. In addition, co-localization of sCrot with internal membranes was evaluated, and the result demonstrated co-localization >80% in tumor cells, in contrast to <30% in non-tumor cells, suggesting selectivity by molecular targets in tumors. Also, low cytotoxicity was observed after treatment with sCrot and nCrot, which suggests the future use of sCrot as a molecular probe for labeling tumor cells in vivo, or to delivery molecules which present activity on selective intracellular targets.


A crotamina (nCrot) é um polipeptídio composto de 42 resíduos de aminoácidos, do veneno da cascavel da América do Sul (Crotalus durissus terrificus). Previamente, nosso grupo descreveu a nCrot como um CPP (do inglês, Cell-Penetrating Peptide), uma vez que esta possui capacidade de penetrar rapidamente em diferentes tipos de células atravessando a barreira lipídica da membrana celular. A capacidade dos CPPs de atravessar as membranas plasmáticas tem sido utilizada para várias aplicações, incluindo a entrega de moléculas bioativas para inibir mecanismos celulares envolvido em diversas doenças. A característica da nCrot como CPP é direcionada principalmente por células em divisão, o que propõe células tumorais como alvo. A partir desta ideia, o objetivo foi avaliar sua capacidade de carrear a proteína pró-apoptótica BAX para o interior de células tumorais. Assim, sete peptídeos foram sintetizados a partir da sequência da nCrot, sendo seis destes conjugados com BAX. Os peptídeos carrearam BAX para o interior das células tumorais testadas (melanoma murino/humano e tumor de mama humano) sendo 4 destes, capazes de diminuir significativamente a viabilidade celular, além de causarem mudanças morfológicas, alterações na expressão de proteínas pró-apoptóticas e induzirem hiperpolarização da membrana mitocondrial resultando em apoptose e necrose. Por hipótese, as ações da nCrot podem estar relacionadas à sua capacidade inibitória sobre o canal K(V)1.3. A alta expressão deste canal é observada em vários canceres, incluindo melanoma, câncer de mama, próstata, pâncreas, entre outros. A fim de avaliar sua ação em modelo de câncer de pâncreas, foi utilizada a crotamina sintética (sCrot) em diferentes aspectos bioquímicos das células tumorais de pâncreas e foi observado que a mesma não influenciou na viabilidade celular, e não induziu mudanças fenotípicas quando analisada a proteína HuR, que está relacionada com a sobrevivência da célula tumoral e a resistência de drogas quimioterápicas. Por fim, a atividade CPP da nCrot e sCrot foi investigada em células de melanoma humano, melanoma murino, tumor de mama, linfócitos T humanos provenientes de leucemia, células mononucleares humanas, fibroblastos murino e queratinócitos humanos. sCrot e nCrot demonstraram capacidade de penetrar rapidamente em diferentes celulares, mostrando preferência por células tumorais. O teste foi realizado em diferentes tempos e em tempo real nas células vivas por 24 horas em time-lapse. Ademais, foi avaliada a co-localização da sCrot com membranas internas, e o resultado demonstrou uma co-localização >80% em células tumorais, em contraste com <30% em células não-tumorais, sugerindo a seletividade por alvos moleculares em tumores. Também, foi observada baixa citotoxicidade após tratamento com sCrot e nCrot, o que sugere a futura utilização da sCrot como uma sonda molecular para a marcação das células tumorais in vivo, ou ainda para a entrega de moléculas que tenha ação sobre os alvos intracelulares seletivos.

14.
Proteomics ; 16(8): 1225-37, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26648572

RESUMO

Antimicrobial peptides have been considered well-deserving candidates to fight the battle against microorganisms due to their broad-spectrum antimicrobial activities. Several studies have suggested that membrane disruption is the basic mechanism of AMPs that leads to killing or inhibiting microorganisms. Also, AMPs have been reported to interact with macromolecules inside the microbial cells such as nucleic acids (DNA/RNA), protein synthesis, essential enzymes, membrane septum formation and cell wall synthesis. Proteins are associated with many intracellular mechanisms of cells, thus protein targets may be specifically involved in mechanisms of action of AMPs. AMPs like pyrrhocoricin, drosocin, apidecin and Bac 7 are documented to have protein targets, DnaK and GroEL. Moreover, the intracellular targeting AMPs are reported to influence more than one protein targets inside the cell, suggesting for the multiple modes of actions. This complex mechanism of intracellular targeting AMPs makes them more difficult for the development of resistance. Herein, we have summarized the current status of AMPs in terms of their mode of actions, entry to cytoplasm and inhibition of macromolecules. To reveal the mechanism of action, we have focused on AMPs with intracellular protein targets. We have also included the use of high-throughput proteome microarray to determine the unidentified AMP protein targets in this review.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Espaço Intracelular/metabolismo , Proteoma/metabolismo , Proteômica/métodos , Sequência de Aminoácidos , Peptídeos Catiônicos Antimicrobianos/genética , Peptídeos Catiônicos Antimicrobianos/farmacologia , Escherichia coli/efeitos dos fármacos , Proteínas de Escherichia coli/genética , Testes de Sensibilidade Microbiana , Modelos Biológicos , Proteoma/genética
15.
Biopolymers ; 104(2): 73-83, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25581614

RESUMO

AmyI-1-18, an octadecapeptide derived from α-amylase (AmyI-1) of rice (Oryza sativa L. japonica), is a novel cationic α-helical antimicrobial peptide (AMP) that contains two lysine and two arginine residues. The antimicrobial activity of AmyI-1-18 against human pathogens was quantitatively evaluated using a chemiluminescence method that measures ATP derived from viable cells. Of the ten kinds of human pathogens, AmyI-1-18 exhibited antimicrobial activity against nine. Its 50% growth-inhibitory concentrations (ICs50 ) against Porphyromonas gingivalis, Propionibacterium acnes, Pseudomonas aeruginosa, Candida albicans, and Streptococcus mutans were 13, 19, 50, 64, and 77 µM, respectively. AmyI-1-18 had little or no hemolytic activity even at 500 µM, and showed negligible cytotoxicity up to 1200 µM. The degree of 3,3'-dipropylthiadicarbocyanine iodide release from P. gingivalis cells induced by the addition of AmyI-1-18 was significantly lower than that induced by the addition of melittin. Flow cytometric analysis showed that the percentages of P. aeruginosa, S. mutans, and C. albicans cells stained with propidium iodide (PI), a DNA-intercalating dye, were 89%, 43%, and 3%, respectively, when AmyI-1-18 was added at a concentration equal to its 4×IC50 . Therefore, the antimicrobial activity of AmyI-1-18 against P. aeruginosa and S. mutans appears to be mainly attributable to its membrane-disrupting activity. In contrast, its antimicrobial activity against P. gingivalis and C. albicans most likely depends upon interactions with intracellular targets other than their cell membranes. Collectively, these results indicate that AmyI-1-18 is useful as a safe and potent AMP against the pathogens described above in many fields of healthcare.


Assuntos
Anti-Infecciosos/química , Anti-Infecciosos/farmacologia , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/farmacologia , Oryza/enzimologia , alfa-Amilases/química , Candida albicans/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Porphyromonas gingivalis/efeitos dos fármacos , Propionibacterium acnes/efeitos dos fármacos , Estrutura Secundária de Proteína , Pseudomonas aeruginosa/efeitos dos fármacos , Relação Estrutura-Atividade
16.
Cancer Genomics Proteomics ; 10(6): 239-50, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24336633

RESUMO

Antibody-based molecules can be delivered into cells either by intracellular expression or through cellular uptake. We describe technologies for identification and expression of intracellular antibodies for target validation, intracellular immunization and tumor therapy, such as intracellular antibody capture technology, suicide or silencing technology, antigen-antibody interaction dependent apoptosis and their application for inhibition of oncogenic intracellular proteins and induction of apoptosis. These strategies have to be viewed in the context that inhibition of protein-protein interactions by small molecules is often limited due to their large interaction surface. We summarize antibodies with the ability to penetrate cells and strategies to induce uptake of antibodies after modification with protein transduction domains. Interference in oncogenic pathways is described for moieties based on antibody 3E10, which translocates into the nucleus after extracellular administration. Finally, we discuss examples of tumor immunotherapy and vaccination against intracellular antigens, and possible interactions mediating their mode of action.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Antineoplásicos/imunologia , Imunoterapia/métodos , Neoplasias/imunologia , Neoplasias/terapia , Animais , Anticorpos Antineoplásicos/biossíntese , Humanos , Terapia de Alvo Molecular
17.
Front Microbiol ; 4: 353, 2013 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-24367355

RESUMO

Antimicrobial peptides (AMPs) are natural antibiotics produced by various organisms such as mammals, arthropods, plants, and bacteria. In addition to antimicrobial activity, AMPs can induce chemokine production, accelerate angiogenesis, and wound healing and modulate apoptosis in multicellular organisms. Originally, their antimicrobial mechanism of action was thought to consist solely of an increase in pathogen cell membrane permeability, but it has already been shown that several AMPs do not modulate membrane permeability in the minimal lethal concentration. Instead, they exert their effects by inhibiting processes such as protein and cell wall synthesis, as well as enzyme activity, among others. Although resistance to these molecules is uncommon several pathogens developed different strategies to overcome AMPs killing such as surface modification, expression of efflux pumps, and secretion of proteases among others. This review describes the various mechanisms of action of AMPs and how pathogens evolve resistance to them.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA