Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Med Chem ; 66(13): 8767-8781, 2023 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-37352470

RESUMO

Macrophage migration inhibitory factor (MIF) is a multifunctional cytokine and essential signaling protein associated with inflammation and cancers. One of the newly described roles of MIF is binding to apoptosis-inducing factor (AIF) that "brings" cells to death in pathological conditions. The interaction between MIF and AIF and their nuclear translocation stands as a central event in parthanatos. However, classical competitive MIF tautomerase inhibitors do not interfere with MIF functions in parthanatos. In this study, we employed a pharmacophore-switch to provide allosteric MIF tautomerase inhibitors that interfere with the MIF/AIF co-localization. Synthesis and screening of a focused compound collection around the 1,2,3-triazole core enabled identification of the allosteric tautomerase MIF inhibitor 6y with low micromolar potency (IC50 = 1.7 ± 0.1 µM). This inhibitor prevented MIF/AIF nuclear translocation and protects cells from parthanatos. These findings indicate that alternative modes to target MIF hold promise to investigate MIF function in parthanatos-mediated diseases.


Assuntos
Fatores Inibidores da Migração de Macrófagos , Parthanatos , Humanos , Fatores Inibidores da Migração de Macrófagos/metabolismo , Fator de Indução de Apoptose , Inflamação/metabolismo , Oxirredutases Intramoleculares/metabolismo
2.
J Am Chem Soc ; 144(30): 13815-13822, 2022 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-35868012

RESUMO

In proteins, the amino acids Phe, Tyr, and especially Trp are frequently involved in π interactions such as π-π, cation-π, and CH-π bonds. These interactions are often crucial for protein structure and protein-ligand binding. A powerful means to study these interactions is progressive fluorination of these aromatic residues to modulate the electrostatic component of the interaction. However, to date no protein expression platform is available to produce milligram amounts of proteins labeled with such fluorinated amino acids. Here, we present a Lactococcus lactis Trp auxotroph-based expression system for efficient incorporation (≥95%) of mono-, di-, tri-, and tetrafluorinated, as well as a methylated Trp analog. As a model protein we have chosen LmrR, a dimeric multidrug transcriptional repressor protein from L. lactis. LmrR binds aromatic drugs, like daunomycin and riboflavin, between Trp96 and Trp96' in the dimer interface. Progressive fluorination of Trp96 decreased the affinity for the drugs 6- to 70-fold, clearly establishing the importance of electrostatic π-π interactions for drug binding. Presteady state kinetic data of the LmrR-drug interaction support the enthalpic nature of the interaction, while high resolution crystal structures of the labeled protein-drug complexes provide for the first time a structural view of the progressive fluorination approach. The L. lactis expression system was also used to study the role of Trp68 in the binding of riboflavin by the membrane-bound riboflavin transport protein RibU from L. lactis. Progressive fluorination of Trp68 revealed a strong electrostatic component that contributed 15-20% to the total riboflavin-RibU binding energy.


Assuntos
Lactococcus lactis , Triptofano , Aminoácidos/metabolismo , Cátions , Proteínas de Membrana Transportadoras/metabolismo , Riboflavina/metabolismo , Triptofano/química
3.
J Med Chem ; 65(3): 2059-2077, 2022 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-35041425

RESUMO

The homologous cytokines macrophage migration inhibitory factor (MIF) and d-dopachrome tautomerase (d-DT or MIF2) play key roles in cancers. Molecules binding to the MIF tautomerase active site interfere with its biological activity. In contrast, the lack of potent MIF2 inhibitors hinders the exploration of MIF2 as a drug target. In this work, screening of a focused compound collection enabled the identification of a MIF2 tautomerase inhibitor R110. Subsequent optimization provided inhibitor 5d with an IC50 of 1.0 µM for MIF2 tautomerase activity and a high selectivity over MIF. 5d suppressed the proliferation of non-small cell lung cancer cells in two-dimensional (2D) and three-dimensional (3D) cell cultures, which can be explained by the induction of cell cycle arrest via deactivation of the mitogen-activated protein kinase (MAPK) pathway. Thus, we discovered and characterized MIF2 inhibitors (5d) with improved antiproliferative activity in cellular models systems, which indicates the potential of targeting MIF2 in cancer treatment.


Assuntos
Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Oxirredutases Intramoleculares/metabolismo , Pirimidinonas/química , Antineoplásicos/química , Antineoplásicos/metabolismo , Sítios de Ligação , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Técnicas de Cultura de Células , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Desenho de Fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Oxirredutases Intramoleculares/antagonistas & inibidores , Cinética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Fatores Inibidores da Migração de Macrófagos/antagonistas & inibidores , Fatores Inibidores da Migração de Macrófagos/metabolismo , Simulação de Dinâmica Molecular , Fosforilação/efeitos dos fármacos , Pirimidinonas/metabolismo , Pirimidinonas/farmacologia , Relação Estrutura-Atividade
4.
Chemistry ; 28(1): e202103030, 2022 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-34724273

RESUMO

Macrophage migration inhibitory factor (MIF) and its homolog MIF2 (also known as D-dopachrome tautomerase or DDT) play key roles in cell growth and immune responses. MIF and MIF2 expression is dysregulated in cancers and neurodegenerative diseases. Accurate and convenient detection of MIF and MIF2 will facilitate research on their roles in cancer and other diseases. Herein, we report the development and application of a 4-iodopyrimidine based probe 8 for the selective labeling of MIF and MIF2. Probe 8 incorporates a fluorophore that allows in situ imaging of these two proteins. This enabled visualization of the translocation of MIF2 from the cytoplasm to the nucleus upon methylnitronitrosoguanidine stimulation of HeLa cells. This observation, combined with literature on nuclease activity for MIF, enabled the identification of nuclease activity for MIF2 on human genomic DNA.


Assuntos
Fatores Inibidores da Migração de Macrófagos , Células HeLa , Humanos , Oxirredutases Intramoleculares
5.
EBioMedicine ; 68: 103412, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34098338

RESUMO

BACKGROUND: Emphysematous COPD is characterized by aberrant alveolar repair. Macrophage migration inhibitory factor (MIF) contributes to alveolar repair, but for its structural and functional homolog D-dopachrome tautomerase (DDT) this is unknown. MIF mediates its effects through CD74 and/or C-X-C chemokine receptors 2 (CXCR2), 4(CXCR4), and possibly 7 (ACKR3). DDT can also signal through CD74, but interactions with other receptors have not been described yet. We therefore aimed at investigating if and how DDT contributes to epithelial repair in COPD. METHODS: We studied effects of recombinant DDT on cell proliferation and survival by clonogenic assay and annexin V-PI staining respectively. DDT-induced signaling was investigated by Western blot. Effects on epithelial growth and differentiation was studied using lung organoid cultures with primary murine or human epithelial cells and incubating with DDT or an ACKR3-blocking nanobody. DDT-ACKR3 interactions were identified by ELISA and co-immunoprecipitation. FINDINGS: We found that DDT promoted proliferation of and prevented staurosporine-induced apoptosis in A549 lung epithelial cells. Importantly, DDT also stimulated growth of primary alveolar epithelial cells as DDT treatment resulted in significantly more and larger murine and human alveolar organoids compared to untreated controls. The anti-apoptotic effect of DDT and DDT-induced organoid growth were inhibited in the presence of an ACKR3-blocking nanobody. Furthermore, ELISA assay and co-immunoprecipitation suggested DDT complexes with ACKR3. DDT could activate the PI3K-Akt pathway and this activation was enhanced in ACKR3-overexpressing cells. INTERPRETATION: In conclusion, DDT contributes to alveolar epithelial repair via ACKR3 and may thus augment lung epithelial repair in COPD. FUNDING: As stated in the Acknowledgments.


Assuntos
Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/metabolismo , Doença Pulmonar Obstrutiva Crônica/metabolismo , Receptores CXCR/metabolismo , Estaurosporina/efeitos adversos , Células A549 , Idoso , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Proteínas Proto-Oncogênicas c-akt/metabolismo , Doença Pulmonar Obstrutiva Crônica/induzido quimicamente , Doença Pulmonar Obstrutiva Crônica/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais
6.
Angew Chem Int Ed Engl ; 60(32): 17514-17521, 2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-34018657

RESUMO

Macrophage migration inhibitory factor (MIF) is involved in protein-protein interactions that play key roles in inflammation and cancer. Current strategies to develop small molecule modulators of MIF functions are mainly restricted to the MIF tautomerase active site. Here, we use this site to develop proteolysis targeting chimera (PROTAC) in order to eliminate MIF from its protein-protein interaction network. We report the first potent MIF-directed PROTAC, denoted MD13, which induced almost complete MIF degradation at low micromolar concentrations with a DC50 around 100 nM in A549 cells. MD13 suppresses the proliferation of A549 cells, which can be explained by deactivation of the MAPK pathway and subsequent induction of cell cycle arrest at the G2/M phase. MD13 also exhibits antiproliferative effect in a 3D tumor spheroid model. In conclusion, we describe the first MIF-directed PROTAC (MD13) as a research tool, which also demonstrates the potential of PROTACs in cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Benzoxazinas/farmacologia , Oxirredutases Intramoleculares/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Ftalimidas/farmacologia , Células A549 , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Antineoplásicos/síntese química , Benzoxazinas/síntese química , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Humanos , Oxirredutases Intramoleculares/química , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fatores Inibidores da Migração de Macrófagos/química , Ftalimidas/síntese química , Proteólise/efeitos dos fármacos , Esferoides Celulares/efeitos dos fármacos , Ubiquitina-Proteína Ligases/metabolismo
7.
J Med Chem ; 63(20): 11920-11933, 2020 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-32940040

RESUMO

Macrophage migration inhibitory factor (MIF) is a cytokine with key roles in inflammation and cancer, which qualifies it as a potential drug target. Apart from its cytokine activity, MIF also harbors enzyme activity for keto-enol tautomerization. MIF enzymatic activity has been used for identification of MIF binding molecules that also interfere with its biological activity. However, MIF tautomerase activity assays are troubled by irregularities, thus creating a need for alternative methods. In this study, we identified a 7-hydroxycoumarin fluorophore with high affinity for the MIF tautomerase active site (Ki = 18 ± 1 nM) that binds with concomitant quenching of its fluorescence. This property enabled development of a novel competition-based assay format to quantify MIF binding. We also demonstrated that the 7-hydroxycoumarin fluorophore interfered with the MIF-CD74 interaction and inhibited proliferation of A549 cells. Thus, we provide a high-affinity MIF binder as a novel tool to advance MIF-oriented research.


Assuntos
Inibidores Enzimáticos/farmacologia , Corantes Fluorescentes/farmacologia , Oxirredutases Intramoleculares/antagonistas & inibidores , Fatores Inibidores da Migração de Macrófagos/antagonistas & inibidores , Umbeliferonas/farmacologia , Ligação Competitiva/efeitos dos fármacos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Corantes Fluorescentes/síntese química , Corantes Fluorescentes/química , Humanos , Modelos Moleculares , Estrutura Molecular , Relação Estrutura-Atividade , Umbeliferonas/síntese química , Umbeliferonas/química
8.
Cancers (Basel) ; 11(7)2019 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-31248045

RESUMO

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a potent inducer of apoptosis that may be a promising agent in cancer therapy due to its selectivity toward tumor cells. However, many cancer cells are resistant to TRAIL due to defects in apoptosis signaling or activation of survival pathways. We hypothesized that a disruption of pro-survival signaling cascades with the multi-tyrosine kinase inhibitor sunitinib and would be an effective strategy to enhance TRAIL-mediated apoptosis. Here we demonstrate that sunitinib significantly augments the anticancer activity of TRAIL in models of colon cancer. The therapeutic benefit of the TRAIL/sunitinib combination was associated with increased apoptosis marked by enhanced caspase-3 cleavage and DNA fragmentation. Overexpression of the anti-apoptotic factor B-cell lymphoma 2 (BCL-2) in HCT116 cells reduced TRAIL/sunitinib-mediated apoptosis, further supporting that sunitinib enhances the anticancer activity of TRAIL via augmented apoptosis. Analysis of pro-survival factors identified that the combination of TRAIL and sunitinib significantly downregulated the anti-apoptotic protein X-linked inhibitor of apoptosis protein (XIAP) through a c-Jun N-terminal kinase (JNK)-mediated mechanism. Short hairpin RNA (shRNA)-mediated knockdown of JNK confirmed its key role in the regulation of sensitivity to this combination as cells with suppressed JNK expression exhibited significantly reduced TRAIL/sunitinib-mediated apoptosis. Importantly, the therapeutic benefit of the TRAIL/sunitinib combination was validated in the HCT116-Luc and HCT15 colon cancer xenograft models, which both demonstrated significant anti-tumor activity in response to combination treatment. Collectively, our data demonstrate that sunitinib enhances TRAIL-mediated apoptosis by heightened JNK activation, diminished XIAP levels, and augmented apoptosis.

9.
FEBS J ; 286(18): 3582-3593, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31081236

RESUMO

Fibrosis is characterized by the progressive alteration of the tissue structure due to the excessive production of extracellular matrix (ECM). The signaling system encompassing Receptor Activator of Nuclear factor NF-κB Ligand (RANKL)/RANK/Osteoprotegerin (OPG) was discovered to play an important role in the regulation of ECM formation and degradation in bone tissue. However, whether and how this signaling pathway plays a role in liver or pulmonary ECM degradation is unclear up to now. Interestingly, increased decoy receptor OPG levels are found in fibrotic tissues. We hypothesize that RANKL can stimulate RANK on macrophages and initiate the process of ECM degradation. This process may be inhibited by highly expressed OPG in fibrotic conditions. In this case, RANKL mutants that can bind to RANK without binding to OPG might become promising therapeutic candidates. In this study, we built a structure-based library containing 44 RANKL mutants and found that the Q236 residue of RANKL is important for OPG binding. We show that RANKL_Q236D can activate RAW cells to initiate the process of ECM degradation and is able to escape from the obstruction by exogenous OPG. We propose that the generation of RANKL mutants with reduced affinity for OPG is a promising strategy for the exploration of new therapeutics against fibrosis.


Assuntos
Fibrose/genética , Osteoprotegerina/química , Ligante RANK/química , Receptor Ativador de Fator Nuclear kappa-B/química , Animais , Matriz Extracelular/química , Matriz Extracelular/genética , Matriz Extracelular/ultraestrutura , Fibrose/patologia , Humanos , Fígado/metabolismo , Fígado/patologia , Pulmão/metabolismo , Pulmão/patologia , Macrófagos/química , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , NF-kappa B/genética , Osteoprotegerina/genética , Osteoprotegerina/ultraestrutura , Ligação Proteica/genética , Conformação Proteica , Ligante RANK/ultraestrutura , Receptor Ativador de Fator Nuclear kappa-B/genética , Receptor Ativador de Fator Nuclear kappa-B/ultraestrutura , Transdução de Sinais/genética
10.
Drug Discov Today ; 23(11): 1910-1918, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29936245

RESUMO

Macrophage migration inhibitory factor (MIF) is an important cytokine for which an increasing number of functions is being described in the pathogenesis of inflammation and cancer. Nevertheless, the availability of potent and druglike MIF inhibitors that are well-characterized in relevant disease models remains limited. Development of highly potent and selective small-molecule MIF inhibitors and validation of their use in relevant disease models will advance drug discovery. In this review, we provide an overview of recent advances in the identification of MIF as a pharmacological target in the pathogenesis of inflammatory diseases and cancer. We also give an overview of the current developments in the discovery and design of small-molecule MIF inhibitors and define future aims in this field.


Assuntos
Descoberta de Drogas/métodos , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Inflamação/tratamento farmacológico , Oxirredutases Intramoleculares/antagonistas & inibidores , Fatores Inibidores da Migração de Macrófagos/antagonistas & inibidores , Terapia de Alvo Molecular/métodos , Neoplasias/tratamento farmacológico , Desenho de Fármacos , Humanos
11.
Protein Expr Purif ; 148: 46-53, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29601965

RESUMO

The HLA class II histocompatibility antigen gamma chain, also known as HLA-DR antigen-associated invariant chain or CD74, has been shown to be involved in many biological processes amongst which antigen loading and transport of MHC class II molecules from the endoplasmic reticulum to the Golgi complex. It is also part of a receptor complex for macrophage migration inhibitory factor (MIF), and participates in inflammatory signaling. The inhibition of MIF-CD74 complex formation is regarded as a potentially attractive therapeutic target in inflammation, cancer and immune diseases. In order to be able to produce large quantities of the extracellular moiety of human CD74, which has been reported to be unstable and protease-sensitive, different constructs were made as fusions with two solubility enhancers: the well-known maltose-binding domain and Fh8, a small protein secreted by the parasite Fasciola hepatica. The fusion proteins could be purified with high yields from Escherichia coli and were demonstrated to be active in binding to MIF. Moreover, our results strongly suggest that the MIF binding site is located in the sequence between the transmembrane and the membrane-distal trimerisation domain of CD74, and comprises at least amino acids 113-125 of CD74.


Assuntos
Antígenos de Diferenciação de Linfócitos B/isolamento & purificação , Antígenos de Histocompatibilidade Classe II/isolamento & purificação , Oxirredutases Intramoleculares/isolamento & purificação , Fatores Inibidores da Migração de Macrófagos/isolamento & purificação , Peptídeos/química , Aminoácidos/genética , Animais , Antígenos de Diferenciação de Linfócitos B/química , Antígenos de Diferenciação de Linfócitos B/genética , Retículo Endoplasmático/genética , Escherichia coli/genética , Fasciola hepatica/química , Complexo de Golgi/genética , Antígenos HLA-DR/química , Antígenos HLA-DR/genética , Antígenos de Histocompatibilidade Classe II/química , Antígenos de Histocompatibilidade Classe II/genética , Humanos , Oxirredutases Intramoleculares/química , Oxirredutases Intramoleculares/genética , Fatores Inibidores da Migração de Macrófagos/química , Fatores Inibidores da Migração de Macrófagos/genética , Peptídeos/genética , Ligação Proteica , Solubilidade
12.
Bioorg Med Chem ; 26(5): 999-1005, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29428527

RESUMO

Macrophage migration inhibitory factor (MIF) is an essential signaling cytokine with a key role in the immune system. Binding of MIF to its molecular targets such as, among others, the cluster of differentiation 74 (CD74) receptor plays a key role in inflammatory diseases and cancer. Therefore, the identification of MIF binding compounds gained importance in drug discovery. In this study, we aimed to discover novel MIF binding compounds by screening of a focused compound collection for inhibition of its tautomerase enzyme activity. Inspired by the known chromen-4-one inhibitor Orita-13, a focused collection of compounds with a chromene scaffold was screened for MIF binding. The library was synthesized using versatile cyanoacetamide chemistry to provide diversely substituted chromenes. The screening provided inhibitors with IC50's in the low micromolar range. Kinetic evaluation suggested that the inhibitors were reversible and did not bind in the binding pocket of the substrate. Thus, we discovered novel inhibitors of the MIF tautomerase activity, which may ultimately support the development of novel therapeutic agents against diseases in which MIF is involved.


Assuntos
Benzopiranos/química , Oxirredutases Intramoleculares/antagonistas & inibidores , Fatores Inibidores da Migração de Macrófagos/antagonistas & inibidores , Benzopiranos/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Humanos , Concentração Inibidora 50 , Oxirredutases Intramoleculares/metabolismo , Cinética , Fatores Inibidores da Migração de Macrófagos/metabolismo , Conformação Molecular , Simulação de Dinâmica Molecular , Ligação Proteica , Relação Estrutura-Atividade
13.
FEBS J ; 284(15): 2501-2512, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28627025

RESUMO

Bone is a dynamic tissue that is maintained by continuous renewal. An imbalance in bone resorption and bone formation can lead to a range of disorders, such as osteoporosis. The receptor activator of NF-κB (RANK)-RANK-ligand (RANKL) pathway plays a major role in bone remodeling. Here, we investigated the effect of mutations at position I248 in the DE-loop of murine RANKL on the interaction of RANKL with RANK, and subsequent activation of osteoclastogenesis. Two single mutants, RANKL I248Y and I248K, were found to maintain binding and have the ability to reduce wild-type RANKL-induced osteoclastogenesis. The generation of RANK-antagonists is a promising strategy for the exploration of new therapeutics against osteoporosis.


Assuntos
Mutação , Osteoclastos/metabolismo , Osteogênese , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Substituição de Aminoácidos , Animais , Biologia Computacional , Transferência de Energia , Sistemas Especialistas , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas Imobilizadas/química , Proteínas Imobilizadas/metabolismo , Cinética , Camundongos , Mutagênese Sítio-Dirigida , Osteoclastos/citologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Multimerização Proteica , Ligante RANK/química , Ligante RANK/genética , Células RAW 264.7 , Receptor Ativador de Fator Nuclear kappa-B/química , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
14.
J Drug Target ; 25(4): 360-369, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27885847

RESUMO

Activated hepatic stellate cells (HSCs) are known to play a central role in liver fibrosis and their elimination is a crucial step toward the resolution and reversion of liver fibrosis. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a molecule that may contribute to the apoptotic removal of activated HSC through binding to its dedicated receptors. In the present study, we investigated the potential application of recombinant receptor-specific TRAIL proteins in the efficient elimination of activated HSCs. Our finding revealed differential contribution of TRAIL receptors among HSCs populations with activated hepatic stellate cells expresses more TRAIL receptors DR5. In vitro treatment of activated HSCs with DR5-specific or wild-type TRAIL variants induced a significant reduction in viability and extracellular matrix production, whereas no significant decrease in viability was associated with the treatment of cells by DR4-specific TRAIL. Our analysis indicate the successful application of the DR5 receptor-specific TRAIL variant in the targeted elimination of activated HSCs via interference with collagen production and simultaneous induction of apoptosis via activation of the caspase pathway. DR5 receptor-specific TRAIL may thus represent a new therapeutic compound for the treatment of liver fibrosis.


Assuntos
Células Estreladas do Fígado/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/fisiologia , Animais , Western Blotting , Linhagem Celular Transformada , Eletroforese em Gel de Poliacrilamida , Matriz Extracelular/metabolismo , Perfilação da Expressão Gênica , Humanos , Masculino , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética
15.
Cancer Biol Ther ; 15(12): 1658-66, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25482930

RESUMO

Current treatment modalities for pancreatic carcinoma afford only modest survival benefits. TRAIL, as a potent and specific inducer of apoptosis in cancer cells, would be a promising new treatment option. However, since not all pancreatic cancer cells respond to TRAIL, further improvements and optimizations are still needed. One strategy to improve the effectiveness of TRAIL-based therapies is to specifically target one of the 2 cell death inducing TRAIL-receptors, TRAIL-R1 or TRAIL-R2 to overcome resistance. To this end, we designed constructs expressing soluble TRAIL (sTRAIL) variants that were rendered specific for either TRAIL-R1 or TRAIL-R2 by amino acid changes in the TRAIL ectodomain. When we expressed these constructs, including wild-type sTRAIL (sTRAIL(wt)), TRAIL-R1 (sTRAIL(DR4)) and TRAIL-R2 (sTRAIL(DR5)) specific variants, in 293 producer cells we found all to be readily expressed and secreted into the supernatant. These supernatants were subsequently transferred onto target cancer cells and apoptosis measured. We found that the TRAIL-R1 specific variant had higher apoptosis-inducing activity in human pancreatic carcinoma Colo357 cells as well as PancTu1 cells that were additionally sensitized by targeting of XIAP. Finally, we tested TRAIL-R1 specific recombinant TRAIL protein (rTRAIL(DR4)) on Colo357 xenografts in nude mice and found them to be more efficacious than rTRAIL(wt). Our results demonstrate the benefits of synthetic biological approaches and show that TRAIL-R1 specific variants can potentially enhance the therapeutic efficacy of TRAIL-based therapies in pancreatic cancer, suggesting that they can possibly become part of individualized and tumor specific combination treatments in the future.


Assuntos
Variação Genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/genética , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Expressão Gênica , Humanos , Camundongos , Mutação , Neoplasias Pancreáticas/patologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/administração & dosagem , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Transfecção , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
16.
FEMS Microbiol Lett ; 356(1): 62-70, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24863934

RESUMO

Bacterial communication via the secretion of small diffusible compounds allows microorganisms to regulate gene expression in a coordinated manner. As many virulence traits are regulated in this fashion, disruption of chemical communication has been proposed as novel antimicrobial therapy. Quorum-quenching enzymes have been a promising discovery in this field as they interfere with the communication of Gram-negative bacteria. AHL-lactonases and AHL-acylases have been described in a variety of bacterial strains; however, usually only one of these two groups of enzymes has been described in a single species. We report here the presence of a member of each group of enzymes in the extremophile bacterium Deinococcus radiodurans. Co-occurrence of both enzymes in a single species increases the chance of inactivating foreign AHL signals under different conditions. We demonstrate that both enzymes are able to degrade the quorum-sensing molecules of various pathogens subsequently affecting virulence gene expression. These studies add the quorum-quenching enzymes of D. radiodurans to the list of potent quorum-quenchers and highlight the idea that quorum quenching could have evolved in some bacteria as a strategy to gain a competitive advantage by altering gene expression in other species.


Assuntos
Amidoidrolases/fisiologia , Proteínas de Bactérias/fisiologia , Hidrolases de Éster Carboxílico/fisiologia , Deinococcus/enzimologia , Percepção de Quorum , Acil-Butirolactonas/metabolismo , Amidoidrolases/química , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/química , Caenorhabditis elegans/microbiologia , Hidrolases de Éster Carboxílico/química , Dados de Sequência Molecular , Pseudomonas aeruginosa/fisiologia
17.
J Bacteriol ; 196(14): 2681-90, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24816606

RESUMO

The iron binding siderophore pyoverdine constitutes a major adaptive factor contributing to both virulence and survival in fluorescent pseudomonads. For decades, pyoverdine production has allowed the identification and classification of fluorescent and nonfluorescent pseudomonads. Here, we demonstrate that PvdP, a periplasmic enzyme of previously unknown function, is a tyrosinase required for the maturation of the pyoverdine chromophore in Pseudomonas aeruginosa. PvdP converts the nonfluorescent ferribactin, containing two iron binding groups, into a fluorescent pyoverdine, forming a strong hexadentate complex with ferrous iron, by three consecutive oxidation steps. PvdP represents the first characterized member of a small family of tyrosinases present in fluorescent pseudomonads that are required for siderophore maturation and are capable of acting on large peptidic substrates.


Assuntos
Proteínas de Bactérias/metabolismo , Regulação Enzimológica da Expressão Gênica/fisiologia , Monofenol Mono-Oxigenase/metabolismo , Oligopeptídeos/metabolismo , Pseudomonas aeruginosa/enzimologia , Proteínas de Bactérias/genética , Domínio Catalítico , Regulação Bacteriana da Expressão Gênica/fisiologia , Modelos Moleculares , Monofenol Mono-Oxigenase/genética , Oligopeptídeos/genética , Filogenia , Conformação Proteica , Transporte Proteico , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo
18.
Methods Mol Biol ; 1149: 681-8, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24818942

RESUMO

Pseudomonas aeruginosa is an opportunistic human pathogen responsible for severe to deadly infections in patients suffering from cystic fibrosis, AIDS, undergoing immune suppressing therapies or suffering from severe burns. In the recent years there has been an increasing interest in exploring animal infection models that, to a certain extent, could mimic human infections. Here we describe the use of the larvae of the greater wax moth Galleria mellonella as a non-expensive, easy-to-use, and easy-to-obtain animal model to study P. aeruginosa infections.


Assuntos
Bioensaio/métodos , Interações Hospedeiro-Patógeno , Mariposas/microbiologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/patogenicidade , Animais , Anti-Infecciosos/farmacologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Insulina , Mamíferos , Pseudomonas aeruginosa/efeitos dos fármacos , Virulência/efeitos dos fármacos
19.
Proc Natl Acad Sci U S A ; 111(4): 1568-73, 2014 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-24474783

RESUMO

The use of enzymes to interfere with quorum sensing represents an attractive strategy to fight bacterial infections. We used PvdQ, an effective quorum-quenching enzyme from Pseudomonas aeruginosa, as a template to generate an acylase able to effectively hydrolyze C8-HSL, the major communication molecule produced by the Burkholderia species. We discovered that the combination of two single mutations leading to variant PvdQ(Lα146W,Fß24Y) conferred high activity toward C8-HSL. Exogenous addition of PvdQ(Lα146W,Fß24Y) dramatically decreased the amount of C8-HSL present in Burkholderia cenocepacia cultures and inhibited a quorum sensing-associated phenotype. The efficacy of this PvdQ variant to combat infections in vivo was further confirmed by its ability to rescue Galleria mellonella larvae upon infection, demonstrating its potential as an effective agent toward Burkholderia infections. Kinetic analysis of the enzymatic activities toward 3-oxo-C12-L-HSL and C8-L-HSL corroborated a substrate switch. This work demonstrates the effectiveness of quorum-quenching acylases as potential novel antimicrobial drugs. In addition, we demonstrate that their substrate range can be easily switched, thereby paving the way to selectively target only specific bacterial species inside a complex microbial community.


Assuntos
Amidoidrolases/metabolismo , Burkholderia cenocepacia/patogenicidade , Percepção de Quorum , Amidoidrolases/química , Animais , Burkholderia cenocepacia/enzimologia , Cinética , Larva/microbiologia , Modelos Moleculares , Mariposas/crescimento & desenvolvimento , Mariposas/microbiologia , Especificidade por Substrato , Virulência
20.
Microbiol Mol Biol Rev ; 76(1): 46-65, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22390972

RESUMO

Cell-to-cell communication is a major process that allows bacteria to sense and coordinately react to the fluctuating conditions of the surrounding environment. In several pathogens, this process triggers the production of virulence factors and/or a switch in bacterial lifestyle that is a major determining factor in the outcome and severity of the infection. Understanding how bacteria control these signaling systems is crucial to the development of novel antimicrobial agents capable of reducing virulence while allowing the immune system of the host to clear bacterial infection, an approach likely to reduce the selective pressures for development of resistance. We provide here an up-to-date overview of the molecular basis and physiological implications of cell-to-cell signaling systems in Gram-negative bacteria, focusing on the well-studied bacterium Pseudomonas aeruginosa. All of the known cell-to-cell signaling systems in this bacterium are described, from the most-studied systems, i.e., N-acyl homoserine lactones (AHLs), the 4-quinolones, the global activator of antibiotic and cyanide synthesis (GAC), the cyclic di-GMP (c-di-GMP) and cyclic AMP (cAMP) systems, and the alarmones guanosine tetraphosphate (ppGpp) and guanosine pentaphosphate (pppGpp), to less-well-studied signaling molecules, including diketopiperazines, fatty acids (diffusible signal factor [DSF]-like factors), pyoverdine, and pyocyanin. This overview clearly illustrates that bacterial communication is far more complex than initially thought and delivers a clear distinction between signals that are quorum sensing dependent and those relying on alternative factors for their production.


Assuntos
Pseudomonas aeruginosa/patogenicidade , Animais , Comunicação Celular/genética , Comunicação Celular/fisiologia , Humanos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Virulência/genética , Virulência/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...