Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Am J Ther ; 30(5): e447-e453, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37713689

RESUMO

BACKGROUND: Deposition of wild-type or mutant transthyretin (TTR) amyloid fibrils in the myocardium causes TTR amyloid cardiomyopathy (ATTR-CM). Targeted therapeutics for ATTR-CM include TTR stabilizers (tafamidis and diflunisal) and oligonucleotide drugs (revusiran, patisiran, and inotersen). TTR stabilizers prevent dissociation of transthyretin tetramers. Transthyretin monomers can misfold and form amyloid fibrils. TTR stabilizers thereby limit amyloid fibrils development and deposition. Oligonucleotide drugs inhibit hepatic synthesis of transthyretin, which decreases transthyretin protein levels and thus the amyloid fibril substrate. AREAS OF UNCERTAINTY: To study the safety and efficacy of targeted therapeutics in patients with ATTR-CM, we performed a pooled analysis. A random-effects model with the Mantel-Haenszel method was used to pool the data. DATA SOURCES: A literature search was performed using PubMed, Cochrane CENTRAL, and Embase databases using the search terms "cardiac amyloidosis" AND "tafamidis" OR "patisiran" OR "inotersen" OR "revusiran" OR "diflunisal." THERAPEUTIC ADVANCES: We identified 6 studies that compared targeted therapeutics with placebo. One study was stopped prematurely because of increased mortality in the targeted therapeutics arm. Pooled analysis included 1238 patients, of which 738 patients received targeted therapeutics and 500 patients received placebo. When compared with placebo, targeted therapeutics significantly reduced all-cause mortality [OR 0.39, 95% confidence interval (CI): 0.16-0.97, P = 0.04]. Only 2 studies reported the effect on cardiovascular-related hospitalizations. There was a trend toward an improvement in global longitudinal strain (mean difference -0.69, 95% CI: -1.44 to 0.05, P = 0.07). When compared with placebo, there was no increase in serious adverse events with targeted therapeutics (OR 1.06, 95% CI: 0.78-1.44, P = 0.72). CONCLUSION: Evidence from the pooled analysis revealed targeted therapeutics improve survival and are well-tolerated. These findings suggest a potential role for targeted therapeutics in the treatment of patients with ATTR-CM.


Assuntos
Neuropatias Amiloides Familiares , Cardiomiopatias , Diflunisal , Humanos , Neuropatias Amiloides Familiares/tratamento farmacológico , Pré-Albumina/metabolismo , Pré-Albumina/uso terapêutico , Diflunisal/farmacologia , Diflunisal/uso terapêutico , Oligonucleotídeos/farmacologia , Oligonucleotídeos/uso terapêutico , Cardiomiopatias/tratamento farmacológico
2.
ChemMedChem ; 18(4): e202200599, 2023 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-36533570

RESUMO

Here we report the encapsulation of an osteosarcoma stem cell (OSC) potent gallium(III)-diflunisal complex 1 into polymeric nanoparticles, and its delivery into osteosarcoma cells. At the optimum feed (20 %, 1 NP20 ), nanoparticle encapsulation of 1 enhances potency towards bulk osteosarcoma cells and OSCs (cultured in monolayer and three-dimensional systems). Strikingly, the nanoparticle formulation exhibits up to 5645-fold greater potency towards OSCs than frontline anti-osteosarcoma drugs, doxorubicin and cisplatin. The nanoparticle formulation evokes a similar mechanism of action as the payload, which bodes well for future translation. Specifically, the nanoparticle formulation induces nuclear DNA damage, cyclooxygenase-2 downregulation, and caspase-dependent apoptosis. To the best of our knowledge, this is the first study to demonstrate that polymeric nanoparticles can be used to effectively deliver an OSC-active metal complex into osteosarcoma cells.


Assuntos
Neoplasias Ósseas , Diflunisal , Gálio , Nanopartículas , Osteossarcoma , Humanos , Diflunisal/farmacologia , Micelas , Gálio/farmacologia , Linhagem Celular Tumoral , Osteossarcoma/tratamento farmacológico , Polímeros/farmacologia , Células-Tronco Neoplásicas
3.
Amyloid ; 28(1): 24-29, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32811187

RESUMO

Transthyretin (TTR) tetramer dissociation is rate limiting for aggregation and subunit exchange. Slowing of TTR tetramer dissociation via kinetic stabiliser binding slows cardiomyopathy progression. Quadruplicate subunit exchange comparisons of the drug candidate AG10, and the drugs tolcapone, diflunisal, and tafamidis were carried out at 1, 5, 10, 20 and 30 µM concentrations in 4 distinct pooled wild type TTR (TTRwt) human plasma samples. These experiments reveal that the concentration dependence of the efficacy of each compound at inhibiting TTR dissociation was primarily determined by the ratio between the stabiliser's dissociation constants from TTR and albumin, which competes with TTR to bind kinetic stabilisers. The best stabilisers, tafamidis (80 mg QD), AG10 (800 mg BID), and tolcapone (3 x 100 mg over 12 h), exhibit very similar kinetic stabilisation at the plasma concentrations resulting from these doses. At a 10 µM plasma concentration, AG10 is slightly more potent as a kinetic stabiliser vs. tolcapone and tafamidis (which are similar), which are substantially more potent than diflunisal. Dissociation of TTR can be limited to 10% of its normal rate at concentrations of 5.7 µM AG10, 10.3 µM tolcapone, 12.0 µM tafamidis, and 188 µM diflunisal. The potency similarities revealed by our study suggest that differences in safety, adsorption and metabolism, pharmacokinetics, and tissue distribution become important for kinetic stabiliser clinical use decisions.


Assuntos
Neuropatias Amiloides Familiares/tratamento farmacológico , Amiloide/genética , Cardiomiopatias/tratamento farmacológico , Pré-Albumina/genética , Amiloide/antagonistas & inibidores , Amiloide/sangue , Amiloide/química , Neuropatias Amiloides Familiares/sangue , Neuropatias Amiloides Familiares/genética , Neuropatias Amiloides Familiares/patologia , Benzoatos/farmacologia , Benzoxazóis/farmacologia , Cardiomiopatias/sangue , Cardiomiopatias/genética , Cardiomiopatias/patologia , Diflunisal/farmacologia , Humanos , Cinética , Pré-Albumina/química , Agregados Proteicos/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Multimerização Proteica/efeitos dos fármacos , Subunidades Proteicas/antagonistas & inibidores , Subunidades Proteicas/sangue , Subunidades Proteicas/química , Subunidades Proteicas/genética , Pirazóis/farmacologia , Tolcapona/farmacologia
4.
EMBO Rep ; 20(10): e47788, 2019 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-31418171

RESUMO

Extracellular HMGB1 triggers inflammation following infection or injury and supports tumorigenesis in inflammation-related malignancies. HMGB1 has several redox states: reduced HMGB1 recruits inflammatory cells to injured tissues forming a heterocomplex with CXCL12 and signaling via its receptor CXCR4; disulfide-containing HMGB1 binds to TLR4 and promotes inflammatory responses. Here we show that diflunisal, an aspirin-like nonsteroidal anti-inflammatory drug (NSAID) that has been in clinical use for decades, specifically inhibits in vitro and in vivo the chemotactic activity of HMGB1 at nanomolar concentrations, at least in part by binding directly to both HMGB1 and CXCL12 and disrupting their heterocomplex. Importantly, diflunisal does not inhibit TLR4-dependent responses. Our findings clarify the mode of action of diflunisal and open the way to the rational design of functionally specific anti-inflammatory drugs.


Assuntos
Quimiocina CXCL12/metabolismo , Diflunisal/farmacologia , Proteína HMGB1/metabolismo , Leucócitos/metabolismo , Células 3T3 , Animais , Quimiotaxia/efeitos dos fármacos , Diflunisal/química , Dissulfetos/metabolismo , Ácido Glicirrízico/farmacologia , Humanos , Inflamação/patologia , Leucócitos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Espectroscopia de Ressonância Magnética , Camundongos
5.
Biol Pharm Bull ; 41(7): 979-984, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29962408

RESUMO

Hereditary transthyretin (TTR)-related amyloidosis is caused by mutations in the TTR gene. The mutations destabilize the tetramer and/or monomer of TTR, and thus the stabilization of TTR is a key strategy for the treatment of TTR-related amyloidosis. In this review, we summarized the natural products and synthetic compounds that have been shown to inhibit the amyloidogenesis of TTR. The stabilizers and/or the amyloid fibril disrupters isolated from natural sources may become lead compounds for the treatment of TTR-related amyloidosis.


Assuntos
Amiloide/antagonistas & inibidores , Amiloidose/tratamento farmacológico , Anti-Inflamatórios não Esteroides/farmacologia , Produtos Biológicos/farmacologia , Pré-Albumina/metabolismo , Amiloide/metabolismo , Amiloidose/genética , Anti-Inflamatórios não Esteroides/uso terapêutico , Benzoxazóis/farmacologia , Benzoxazóis/uso terapêutico , Produtos Biológicos/uso terapêutico , Diflunisal/farmacologia , Diflunisal/uso terapêutico , Humanos , Mutação , Pré-Albumina/genética
6.
FEBS Lett ; 591(15): 2311-2322, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28656718

RESUMO

The ß-clamp is the processivity-promoting factor for most of the enzymes in prokaryotic DNA replication; hence, it is a crucial drug target. In the present study, we investigated the ß-clamp from Helicobacter pylori, aiming to seek potential drug molecules against this gastric-cancer-causing bacterium. An in silico screening of Food and Drug Administration (FDA) approved drugs against the H. pylori ß-clamp, followed by its in vitro inhibition using a surface competition approach, yielded the drug diflunisal as a positive initial hit. Diflunisal inhibits the growth of H. pylori in the micromolar range. We determined the structure of diflunisal in complex with the ß-clamp to show that the drug binds at subsite I, which is a protein-protein interaction site. Successful identification of FDA-approved molecules against H. pylori may lead to better and faster drug development.


Assuntos
Antibacterianos/farmacologia , DNA Polimerase III/antagonistas & inibidores , DNA Polimerase III/química , Diflunisal/farmacologia , Helicobacter pylori/efeitos dos fármacos , Antibacterianos/química , Sítios de Ligação , Cristalografia por Raios X , DNA Ligases/metabolismo , DNA Polimerase III/metabolismo , Diflunisal/química , Aprovação de Drogas , Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Helicobacter pylori/enzimologia , Concentração Inibidora 50 , Simulação de Acoplamento Molecular , Conformação Proteica , Estados Unidos , United States Food and Drug Administration
7.
CNS Neurosci Ther ; 23(7): 605-619, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28570028

RESUMO

BACKGROUND: The absence of transthyretin (TTR) in AD mice decreases brain Aß clearance and reduces the low-density lipoprotein receptor-related protein 1 (LRP1). It is possible that neuroprotection by TTR is dependent on its tetramer structural stability, as studies using TTR mutants showed that unstable L55P TTR has low affinity for Aß, and TTR tetrameric stabilizers such as iododiflunisal ameliorate AD features in vivo. METHODS: We firstly investigated TTR folding status in human plasma measuring the resistance to urea denaturation. The importance of TTR stability on Aß internalization was studied in human cerebral microvascular endothelial (hCMEC/D3) and hepatoma cells (HepG2), by flow cytometry. To investigate the fate of Aß at the blood-brain barrier, Aß efflux from hCMEC/D3 cells seeded on transwells was measured using ELISA. Further, to assess Aß colocalization with lysosomes, Lysotracker was used. Moreover, levels of LRP1 were assessed in the liver and plasma of mice with different TTR backgrounds or treated with iododiflunisal. RESULTS: We showed that TTR stability is decreased in AD and that WT TTR and drug-stabilized L55P TTR are able to increase uptake of Aß. Furthermore, measurement of Aß efflux showed that stable or stabilized TTR increased Aß efflux from the basolateral to the apical side. Moreover, HepG2 cells incubated with Aß in the presence of WT TTR, but not L55P TTR, showed an increased number of lysosomes. Further, in the presence of WT TTR, Aß peptide colocalized with lysosomes, indicating that only stable TTR assists Aß internalization, leading to its degradation. Finally, we demonstrated that only stable TTR can increase LRP1 levels. CONCLUSION: TTR stabilization exerts a positive effect on Aß clearance and LRP1 levels, suggesting that TTR protective role in AD is dependent on its stability. These results provide relevant information for the design of TTR-based therapeutic strategies for AD.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Pré-Albumina/metabolismo , Peptídeos beta-Amiloides/antagonistas & inibidores , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Linhagem Celular , Diflunisal/análogos & derivados , Diflunisal/farmacologia , Escherichia coli , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Lisossomos/metabolismo , Camundongos Transgênicos , Pré-Albumina/química , Presenilina-1/genética , Presenilina-1/metabolismo , Desnaturação Proteica , Multimerização Proteica , Estabilidade Proteica , Receptores de LDL/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Ureia/metabolismo
8.
J Microencapsul ; 33(5): 475-86, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27499126

RESUMO

Diflunisal (DIF) is non-steroidal anti-inflammatory drug used in the treatment of rheumatoid arthritis, osteoarthritis. The current engrossment was aimed at formulation and assessment of DIF-loaded solid lipid nanoparticles (SLNs) for topical/dermal application. SLNs formulated by hot homogenisation method based on microemulsification technique were spherical with a mean size of 124.0 ± 2.07 nm; PDI 0.294 ± 0.15. The cumulative amount permeated/area was 109.99 ± 0.008 µg/cm(2), along with permeation flux (6.30 ± 0.09 µg/cm(2)/h) and skin retention (11.74 ± 0.155 µg/cm(2)) across mice skin. The SLNs of DIF showed significant decrease in fluid volume, granuloma tissue weight, leukocyte count/mm(3) after application of SLN formulation in mice air pouch model. Similarly, in mice ear oedema and rat paw oedema model, there was 2.30 and 1.29 time increase in percentage inhibition of oedema after SLN formulation application, respectively, as compared with conventional cream. Hence, the SLNs of DIF may prove to be a potential nanocarrier to effectively treat the local inflammatory conditions associated with arthritis.


Assuntos
Artrite Experimental/tratamento farmacológico , Diflunisal , Portadores de Fármacos , Nanopartículas/química , Absorção Cutânea , Animais , Diflunisal/química , Diflunisal/farmacologia , Portadores de Fármacos/química , Portadores de Fármacos/farmacologia , Feminino , Camundongos , Ratos
9.
Elife ; 52016 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-27244239

RESUMO

Salicylate and acetylsalicylic acid are potent and widely used anti-inflammatory drugs. They are thought to exert their therapeutic effects through multiple mechanisms, including the inhibition of cyclo-oxygenases, modulation of NF-κB activity, and direct activation of AMPK. However, the full spectrum of their activities is incompletely understood. Here we show that salicylate specifically inhibits CBP and p300 lysine acetyltransferase activity in vitro by direct competition with acetyl-Coenzyme A at the catalytic site. We used a chemical structure-similarity search to identify another anti-inflammatory drug, diflunisal, that inhibits p300 more potently than salicylate. At concentrations attainable in human plasma after oral administration, both salicylate and diflunisal blocked the acetylation of lysine residues on histone and non-histone proteins in cells. Finally, we found that diflunisal suppressed the growth of p300-dependent leukemia cell lines expressing AML1-ETO fusion protein in vitro and in vivo. These results highlight a novel epigenetic regulatory mechanism of action for salicylate and derivative drugs.


Assuntos
Antineoplásicos/farmacologia , Diflunisal/farmacologia , Inibidores Enzimáticos/farmacologia , Regulação Leucêmica da Expressão Gênica , Leucemia Mieloide Aguda/tratamento farmacológico , Ácido Salicílico/farmacologia , Fatores de Transcrição de p300-CBP/antagonistas & inibidores , Acetilcoenzima A/antagonistas & inibidores , Acetilcoenzima A/metabolismo , Acetilação/efeitos dos fármacos , Animais , Antineoplásicos/química , Ligação Competitiva , Domínio Catalítico , Linhagem Celular Tumoral , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Diflunisal/química , Inibidores Enzimáticos/química , Células HEK293 , Humanos , Leucemia Mieloide Aguda/enzimologia , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Leucócitos/efeitos dos fármacos , Leucócitos/enzimologia , Leucócitos/patologia , Camundongos , Camundongos SCID , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Ligação Proteica , Proteína 1 Parceira de Translocação de RUNX1/genética , Proteína 1 Parceira de Translocação de RUNX1/metabolismo , Ácido Salicílico/química , Transdução de Sinais , Relação Estrutura-Atividade , Ensaios Antitumorais Modelo de Xenoenxerto , Fatores de Transcrição de p300-CBP/genética , Fatores de Transcrição de p300-CBP/metabolismo
10.
PLoS One ; 11(4): e0153529, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27093678

RESUMO

Amyloid formation of the human plasma protein transthyretin (TTR) is associated with several human disorders, including familial amyloidotic polyneuropathy (FAP) and senile systemic amyloidosis. Dissociation of TTR's native tetrameric assembly is the rate-limiting step in the conversion into amyloid, and this feature presents an avenue for intervention because binding of an appropriate ligand to the thyroxin hormone binding sites of TTR stabilizes the native tetrameric assembly and impairs conversion into amyloid. The desired features for an effective TTR stabilizer include high affinity for TTR, high selectivity in the presence of other proteins, no adverse side effects at the effective concentrations, and a long half-life in the body. In this study we show that the commonly used flame retardant tetrabromobisphenol A (TBBPA) efficiently stabilizes the tetrameric structure of TTR. The X-ray crystal structure shows TBBPA binding in the thyroxine binding pocket with bromines occupying two of the three halogen binding sites. Interestingly, TBBPA binds TTR with an extremely high selectivity in human plasma, and the effect is equal to the recently approved drug tafamidis and better than diflunisal, both of which have shown therapeutic effects against FAP. TBBPA consequently present an interesting scaffold for drug design. Its absorption, metabolism, and potential side-effects are discussed.


Assuntos
Excipientes/química , Bifenil Polibromatos/química , Pré-Albumina/química , Amiloide/metabolismo , Amiloidose/metabolismo , Benzoxazóis/farmacologia , Sítios de Ligação/fisiologia , Linhagem Celular Tumoral , Cristalografia por Raios X/métodos , Diflunisal/farmacologia , Desenho de Fármacos , Meia-Vida , Humanos , Ligantes , Bifenil Polibromatos/metabolismo , Pré-Albumina/metabolismo , Ligação Proteica/fisiologia , Tiroxina/farmacologia
11.
Anticancer Agents Med Chem ; 16(10): 1266-74, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26320814

RESUMO

A series of diflunisal 4-thiazolidinones were synthesized. Some selected compounds were determined at one dose towards the full panel of 60 human cancer cell lines by National Cancer Institute. 2',4'-Difluoro-4-hydroxy-N-[4-oxo-2-(thiophen-2-yl)-1,3-thiazolidin-3-yl]biphenyl- 3-carboxamide (4a) demonstrated the most marked effect on K-562 cancer cell line with 58.59 % growth inhibition at 10 µM. Compound 4a was evaluated in vitro using the MTT colorimetric method against human leukemia cell line K-562 and mouse embryonic fibroblasts cell line NIH- 3T3 at different doses for cell viability and growth inhibition. Compound 4a exhibited anticancer activity with IC50 value of 5.2 µM against K-562 cells and did not display cytotoxicity towards NIH-3T3 cells compared with diflunisal. In addition, this compound could be an interesting prototype as an antiproliferative agent.


Assuntos
Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Diflunisal/química , Animais , Antineoplásicos/química , Linhagem Celular Tumoral , Técnicas de Química Sintética , Diflunisal/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Células K562/efeitos dos fármacos , Camundongos , Células NIH 3T3
12.
Eur J Med Chem ; 108: 301-308, 2016 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-26695731

RESUMO

Hepatitis C virus (HCV) infection is a main cause of chronic liver disease, leading to liver cirrhosis and hepatocellular carcinoma (HCC). The objective of our research was to develop effective agents against viral replication. We have previously identified the hydrazide-hydrazone scaffold as a promising hepatitis C virus (HCV) and hepatocelluler inhibitor. Herein we describe the design a number of 2',4'-difluoro-4-hydroxy-N'-(arylmethylidene) biphenyl-3-carbohydrazide (3a-t) as anti-HCV and anticancer agents. Results from evaluation of anti-HCV activity indicated that most of the synthesized hydrazone derivatives inhibited viral replication in the Huh7/Rep-Feo1b and Huh 7.5-FGR-JCI-Rluc2A reporter systems. Antiproliferative activities of increasing concentrations of 2',4'-difluoro-4-hydroxy-N'-(2-pyridyl methylidene)biphenyl-3-carbohydrazide 3b and diflunisal (2.5-40 µM) were assessed in liver cancer cell lines (Huh7, HepG2, Hep3B, Mahlavu, FOCUS and SNU-475) with sulforhodamine B assay for 72 h. Compound 3b with 2-pyridinyl group in the hydrazone part exhibited promising cytotoxic activity against all cell lines with IC50 values of 10, 10.34 16.21 4.74, 9.29 and 8.33 µM for Huh7, HepG2, Hep3B, Mahlavu, FOCUS and SNU-475 cells, respectively, and produced dramatic cell cycle arrest at SubG1/G0 phase as an indicator of apoptotic cell death induction.


Assuntos
Antineoplásicos/farmacologia , Antivirais/síntese química , Antivirais/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Diflunisal/análogos & derivados , Hepacivirus/efeitos dos fármacos , Hidrazinas/farmacologia , Hidrazonas/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Antineoplásicos/síntese química , Antineoplásicos/química , Antivirais/química , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/patologia , Proliferação de Células/efeitos dos fármacos , Diflunisal/química , Diflunisal/farmacologia , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Hidrazinas/química , Hidrazonas/síntese química , Hidrazonas/química , Neoplasias Hepáticas/patologia , Testes de Sensibilidade Microbiana , Estrutura Molecular , Relação Estrutura-Atividade , Células Tumorais Cultivadas
13.
Biochemistry ; 54(2): 268-78, 2015 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-25478940

RESUMO

The circulating protein transthyretin (TTR) can unfold, oligomerize, and form highly structured amyloid fibrils that are deposited in tissues, causing organ damage and disease. This pathogenic process is caused by a heritable TTR point mutation in cases of familial TTR-related amyloidosis or wild-type TTR in cases of age-associated amyloidosis (previously called senile systemic amyloidosis). The TTR amyloid cascade is hypothesized to begin with the dissociation of the TTR native tetrameric structure into folded but unstable monomeric TTR subunits. Unfolding of monomeric TTR initiates an oligomerization process leading to aggregation and fibril formation. Numerous proteostatic mechanisms for regulating the TTR amyloid cascade exist. Extracellular chaperones provide an innate defense against misfolded proteins. Clusterin (CLU), a plasma protein, has the capacity to recognize exposed hydrophobic regions of misfolded proteins, shielding them from aggregation. We have previously demonstrated that CLU is associated with the amyloid fibrils in cardiac tissues from patients with TTR amyloidosis. In this study, we have used tetrameric and monomeric TTR structural variants to determine the ability of CLU to inhibit TTR amyloid fibril formation. Using circular dichroism spectroscopy, we determined that CLU preferentially stabilizes monomeric TTR and generates increasingly stable conformations under acid stress. Moreover, studies using surface plasmon resonance showed a direct interaction of CLU with high-molecular weight TTR oligomers. The interactions of CLU with monomeric and aggregated TTR proceed in a cooperative manner in the presence of diflunisal, a small molecule drug used to stabilize TTR tetramers.


Assuntos
Amiloide/antagonistas & inibidores , Anti-Inflamatórios não Esteroides/farmacologia , Clusterina/metabolismo , Diflunisal/farmacologia , Pré-Albumina/metabolismo , Estabilidade Proteica/efeitos dos fármacos , Amiloide/metabolismo , Humanos , Masculino , Mutação , Pré-Albumina/química , Pré-Albumina/genética , Estrutura Secundária de Proteína/efeitos dos fármacos , Desdobramento de Proteína/efeitos dos fármacos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
14.
Biochem Pharmacol ; 89(4): 464-76, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24695359

RESUMO

We previously identified salicylate as a novel catalytic inhibitor of human DNA topoisomerase II (topo II; EC 5.99.1.3) that preferentially targets the alpha isoform by interfering with topo II-mediated DNA cleavage. Many pharmaceuticals and compounds found in foods are salicylate-based. We have now investigated whether these are also catalytic inhibitors of topo II and the structural determinants modulating these effects. We have determined that a number of hydroxylated benzoic acids attenuate doxorubicin-induced DNA damage signaling mediated by the ATM protein kinase and inhibit topo II decatenation activity in vitro with varying potencies. Based on the chemical structures of these and other derivatives, we identified unique properties influencing topo II inhibition, including the importance of substitutions at the 2'- and 5'-positions. We extended our findings to a number of salicylate-based pharmaceuticals including sulfasalazine and diflunisal and found that both were effective at attenuating doxorubicin-induced DNA damage signaling, topo II DNA decatenation and they blocked stabilization of doxorubicin-induced topo II cleavable complexes in cells. In a manner similar to salicylate, we determined that these agents inhibit topo II-mediated DNA cleavage. This was accompanied by a concomitant decrease in topo II-mediated ATP-hydrolysis. Taken together, these findings reveal a novel function for the broader class of salicylate-related compounds and highlight the need for additional studies into whether they may impact the efficacy of chemotherapy regimens that include topo II poisons.


Assuntos
Antineoplásicos/química , DNA de Neoplasias/química , Proteínas de Ligação a DNA/antagonistas & inibidores , Inibidores Enzimáticos/química , Modelos Moleculares , Proteínas de Neoplasias/antagonistas & inibidores , Salicilatos/química , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Antígenos de Neoplasias/química , Antígenos de Neoplasias/metabolismo , Antineoplásicos/antagonistas & inibidores , Antineoplásicos/farmacologia , Biocatálise/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , DNA Topoisomerases Tipo II/química , DNA Topoisomerases Tipo II/metabolismo , DNA Catenado/química , DNA Catenado/metabolismo , DNA de Cinetoplasto/química , DNA de Cinetoplasto/metabolismo , DNA de Neoplasias/metabolismo , DNA Super-Helicoidal/química , DNA Super-Helicoidal/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Diflunisal/química , Diflunisal/farmacologia , Doxorrubicina/antagonistas & inibidores , Doxorrubicina/farmacologia , Inibidores Enzimáticos/farmacologia , Humanos , Hidrólise/efeitos dos fármacos , Células MCF-7 , Conformação Molecular/efeitos dos fármacos , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Plasmídeos/química , Plasmídeos/metabolismo , Salicilatos/farmacologia , Salicilato de Sódio/análogos & derivados , Salicilato de Sódio/química , Salicilato de Sódio/farmacologia , Sulfassalazina/química , Sulfassalazina/farmacologia
15.
PLoS One ; 7(9): e45368, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23028965

RESUMO

Transthyretin (TTR) protects against A-Beta toxicity by binding the peptide thus inhibiting its aggregation. Previous work showed different TTR mutations interact differently with A-Beta, with increasing affinities correlating with decreasing amyloidogenecity of the TTR mutant; this did not impact on the levels of inhibition of A-Beta aggregation, as assessed by transmission electron microscopy. Our work aimed at probing differences in binding to A-Beta by WT, T119M and L55P TTR using quantitative assays, and at identifying factors affecting this interaction. We addressed the impact of such factors in TTR ability to degrade A-Beta. Using a dot blot approach with the anti-oligomeric antibody A11, we showed that A-Beta formed oligomers transiently, indicating aggregation and fibril formation, whereas in the presence of WT and T119M TTR the oligomers persisted longer, indicative that these variants avoided further aggregation into fibrils. In contrast, L55PTTR was not able to inhibit oligomerization or to prevent evolution to aggregates and fibrils. Furthermore, apoptosis assessment showed WT and T119M TTR were able to protect against A-Beta toxicity. Because the amyloidogenic potential of TTR is inversely correlated with its stability, the use of drugs able to stabilize TTR tetrameric fold could result in increased TTR/A-Beta binding. Here we showed that iododiflunisal, 3-dinitrophenol, resveratrol, [2-(3,5-dichlorophenyl)amino] (DCPA) and [4-(3,5-difluorophenyl)] (DFPB) were able to increase TTR binding to A-Beta; however only DCPA and DFPB improved TTR proteolytic activity. Thyroxine, a TTR ligand, did not influence TTR/A-Beta interaction and A-Beta degradation by TTR, whereas RBP, another TTR ligand, not only obstructed the interaction but also inhibited TTR proteolytic activity. Our results showed differences between WT and T119M TTR, and L55PTTR mutant regarding their interaction with A-Beta and prompt the stability of TTR as a key factor in this interaction, which may be relevant in AD pathogenesis and for the design of therapeutic TTR-based therapies.


Assuntos
Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Pré-Albumina/metabolismo , Precursor de Proteína beta-Amiloide/genética , Linhagem Celular Tumoral , Diflunisal/análogos & derivados , Diflunisal/farmacologia , Dinitrofenóis/farmacologia , Humanos , Mutação , Ácidos Ftálicos/farmacologia , Pré-Albumina/genética , Ligação Proteica/efeitos dos fármacos , Resveratrol , Estilbenos/farmacologia
16.
Rinsho Shinkeigaku ; 49(11): 953-5, 2009 Nov.
Artigo em Japonês | MEDLINE | ID: mdl-20030258

RESUMO

Familial amyloid polyneuropathy (FAP) was long considered to be an incurable disease, but a new therapeutic approach was developed 15 years ago. As the liver produces most of the transthyretin (TTR) in serum, it was assumed that the replacement of a liver expressing an abnormal TTR gene should stop the production of the variant TTR, the serum amyloid precursor in FAP. Until now about 1,500 FAP patients underwent liver transplantation, and the 10-year-survival rate is about 77%. After operation the progression of FAP symptoms certainly stopped, and patients who were in an early stage of the disease and underwent successful operations showed considerable improvement in their quality of life. Electrophysiological study of peripheral nerve function has demonstrated that liver transplantation can halt the progression of peripheral neuropathy in FAP patients, and histopathological regression of amyloid deposits was seen on the patients with long post-transplatation courses. Pharmacological therapies have been considered for FAP patients and among them, diflunisal, one of non-steroidal antiinflammatory drugs, is very promising. TTR tetramer dissociation is an initial step for the process of TTR-derived amyloid fibril formation associated with FAP and diflinisal can inhibit this process by stabilization of the TTR tetramer. Clinical trial of this drug for FAP patients is now going worldwide.


Assuntos
Neuropatias Amiloides Familiares/terapia , Transplante de Fígado , Substituição de Aminoácidos/genética , Amiloide/genética , Amiloide/metabolismo , Neuropatias Amiloides Familiares/classificação , Neuropatias Amiloides Familiares/genética , Neuropatias Amiloides Familiares/fisiopatologia , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/farmacologia , Ensaios Clínicos como Assunto , Diflunisal/administração & dosagem , Diflunisal/farmacologia , Feminino , Humanos , Fígado/metabolismo , Masculino , Mutação , Pré-Albumina/genética , Pré-Albumina/metabolismo
17.
Bioorg Med Chem Lett ; 19(15): 4399-402, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19515562

RESUMO

To discover the new medicinal activity, the structure of diflunisal has been modified. Forty amide derivatives of diflunisal were synthesized starting from diflunisal in three steps. Their inhibition growth rate of human lung cancer cell (A549) and human endometrial adenocarcinoma cell (Ishikawa) in vitro was evaluated. The preliminary assay results showed that compounds 6j, 7o and 8c exhibited good anti-tumor activities and excellent selectivity for the Ishikawa cell, may be potential anti-tumor agents.


Assuntos
Amidas/química , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Química Farmacêutica/métodos , Diflunisal/análogos & derivados , Diflunisal/síntese química , Neoplasias/tratamento farmacológico , Anti-Infecciosos/síntese química , Anti-Infecciosos/farmacologia , Linhagem Celular Tumoral , Cristalografia por Raios X/métodos , Diflunisal/farmacologia , Desenho de Fármacos , Humanos , Concentração Inibidora 50 , Modelos Químicos , Estrutura Molecular
18.
PLoS One ; 4(1): e4124, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19125186

RESUMO

The thyroid hormone and retinol transporter protein known as transthyretin (TTR) is in the origin of one of the 20 or so known amyloid diseases. TTR self assembles as a homotetramer leaving a central hydrophobic channel with two symmetrical binding sites. The aggregation pathway of TTR into amiloid fibrils is not yet well characterized but in vitro binding of thyroid hormones and other small organic molecules to TTR binding channel results in tetramer stabilization which prevents amyloid formation in an extent which is proportional to the binding constant. Up to now, TTR aggregation inhibitors have been designed looking at various structural features of this binding channel others than its ability to host iodine atoms. In the present work, greatly improved inhibitors have been designed and tested by taking into account that thyroid hormones are unique in human biochemistry owing to the presence of multiple iodine atoms in their molecules which are probed to interact with specific halogen binding domains sitting at the TTR binding channel. The new TTR fibrillogenesis inhibitors are based on the diflunisal core structure because diflunisal is a registered salicylate drug with NSAID activity now undergoing clinical trials for TTR amyloid diseases. Biochemical and biophysical evidence confirms that iodine atoms can be an important design feature in the search for candidate drugs for TTR related amyloidosis.


Assuntos
Amiloide , Anti-Inflamatórios não Esteroides , Diflunisal , Iodo/química , Pré-Albumina , Multimerização Proteica/efeitos dos fármacos , Amiloide/química , Amiloide/metabolismo , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/farmacologia , Sítios de Ligação , Diflunisal/química , Diflunisal/farmacologia , Desenho de Fármacos , Humanos , Modelos Moleculares , Estrutura Molecular , Pré-Albumina/química , Pré-Albumina/metabolismo , Estrutura Quaternária de Proteína , Hormônios Tireóideos/química , Hormônios Tireóideos/metabolismo
19.
J Biol Chem ; 283(18): 11887-96, 2008 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-18326041

RESUMO

Transthyretin (TTR) is normally a stable plasma protein. However, in cases of familial TTR-related amyloidosis and senile systemic amyloidosis (SSA), TTR is deposited as amyloid fibrils, leading to organ dysfunction and possibly death. The mechanism by which TTR undergoes the transition from stable, soluble precursor to insoluble amyloid fibril and the factors that promote this process are largely undetermined. Most models involve the dissociation of the native TTR tetramer as the initial step. It is largely accepted that the TTR gene mutations associated with TTR-related amyloidosis lead to the expression of variant proteins that are intrinsically unstable and prone to aggregation. It has been suggested that amyloidogenicity may be conferred to wild-type TTR (the form deposited in SSA) by chemical modification of the lone cysteine residue (Cys(10)) through mixed disulfide bonds. S-Sulfonation and S-cysteinylation are prevalent TTR modifications physiologically, and studies have suggested their ability to modulate the structure of TTR under denaturing conditions. In the present study, we have used fluorescence-detected sedimentation velocity to determine the effect of S-sulfonate and S-cysteine on the quaternary structural stability of fluorophore-conjugated recombinant TTR under nondenaturing conditions. We determined that S-sulfonation stabilized TTR tetramer stability by a factor of 7, whereas S-cysteinylation enhanced dissociation by 2-fold with respect to the unmodified form. In addition, we report the direct observation of tetramer stabilization by the potential therapeutic compound diflunisal. Finally, as proof of concept, we report the sedimentation of TTR in serum and the qualitative assessment of the resulting data.


Assuntos
Cisteína/química , Diflunisal/farmacologia , Pré-Albumina/química , Fluoresceína-5-Isotiocianato/metabolismo , Fluorescência , Humanos , Concentração de Íons de Hidrogênio , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Termodinâmica , Ultracentrifugação
20.
Proc Natl Acad Sci U S A ; 104(12): 4808-13, 2007 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-17360344

RESUMO

Carboranes represent a potentially rich but underutilized class of inorganic and catabolism-inert pharmacophores. The regioselectivity and ease of derivatization of carboranes allows for facile syntheses of a wide variety of novel structures. The steric bulk, rigidity, and ease of B- and C-derivatization and lack of pi-interactions associated with hydrophobic carboranes may be exploited to enhance the selectivity of previously identified bioactive molecules. Transthyretin (TTR) is a thyroxine-transport protein found in the blood that has been implicated in a variety of amyloid related diseases. Previous investigations have identified a variety of nonsteroidal antiinflammatory drugs (NSAIDs) and structurally related derivatives that imbue kinetic stabilization to TTR, thus inhibiting its dissociative fragmentation and subsequent aggregation to form putative toxic amyloid fibrils. However, the cyclooxygenase (COX) activity associated with these pharmaceuticals may limit their potential as long-term therapeutic agents for TTR amyloid diseases. Here, we report the synthesis and evaluation of carborane-containing analogs of the promising NSAID pharmaceuticals previously identified. The replacement of a phenyl ring in the NSAIDs with a carborane moiety greatly decreases their COX activity with the retention of similar efficacy as an inhibitor of TTR dissociation. The most promising of these compounds, 1-carboxylic acid-7-[3-fluorophenyl]-1,7-dicarba-closo-dodecaborane, showed effectively no COX-1 or COX-2 inhibition at a concentration more than an order of magnitude larger than the concentration at which TTR dissociation is nearly completely inhibited. This specificity is indicative of the potential for the exploitation of the unique properties of carboranes as potent and selective pharmacophores.


Assuntos
Amiloide/efeitos dos fármacos , Boranos/síntese química , Boranos/farmacologia , Pré-Albumina/antagonistas & inibidores , Amiloide/biossíntese , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/farmacologia , Boranos/química , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Diflunisal/química , Diflunisal/farmacologia , Ácido Flufenâmico/química , Ácido Flufenâmico/farmacologia , Humanos , Concentração de Íons de Hidrogênio
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA