Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Cancer Ther ; 2024 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-38561023

RESUMO

CD33 (Siglec-3) is a cell surface receptor expressed in approximately 90% of AML blasts, making it an attractive target for therapy of acute myeloid leukemia (AML). While previous CD33-targeting antibody-drug conjugates (ADCs) like gemtuzumab ozogamicin (GO, Mylotarg) have shown efficacy in AML treatment, they have suffered from toxicity and narrow therapeutic window. This study aimed to develop a novel ADC with improved tolerability and a wider therapeutic window. GLK-33 consists of the anti-CD33 antibody lintuzumab and eight mavg-MMAU auristatin linker-payloads per antibody. The experimental methods included testing in cell cultures, patient-derived samples, mouse xenograft models, and rat toxicology studies. GLK-33 exhibited remarkable efficacy in reducing cell viability within CD33-positive leukemia cell lines and primary AML samples. Notably, GLK-33 demonstrated anti-tumor activity at single dose as low as 300 µg/kg in mice, while maintaining tolerability at single dose of 20 - 30 mg/kg in rats. In contrast to both GO and lintuzumab vedotin, GLK-33 exhibited a wide therapeutic window and activity against multidrug-resistant cells. The development of GLK-33 addresses the limitations of previous ADCs, offering a wider therapeutic window, improved tolerability, and activity against drug-resistant leukemia cells. These findings encourage further exploration of GLK-33 in AML through clinical trials.

2.
Mol Cancer Ther ; 2024 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-38324296

RESUMO

PURPOSE: Antibody-drug conjugates (ADCs) have shown impressive clinical activity with approval of many agents in hematological and solid tumors. However, challenges remain with both efficacy and safety of ADCs. This study describes novel trastuzumab-auristatin conjugates with the hydrophilic MMAE prodrug MMAU, and optimization of a glycopeptide linker leading to a wider therapeutic window. EXPERIMENTAL DESIGN: Trastuzumab was conjugated with auristatin payloads via a series of linkers using a stabilized maleimide handle. The ADCs were characterized in vitro and their relative in vivo anti-tumor efficacies were assessed in HER2+ xenograft models. Relative linker stabilities and the mechanism of linker cleavage were studied using in vitro assays. Toxicity and toxicokinetics of the best performing ADC were evaluated in cynomolgus monkey (cyno). RESULTS: The trastuzumab-MMAU ADC with stabilized glycopeptide linker showed maleimide stabilization and higher resistance to cleavage by serum and lysosomal enzymes compared to a valine-citrulline conjugated trastuzumab ADC (trastuzumab-vc-MMAE). A single dose of 1 or 2 mg/kg of trastuzumab-MMAU at drug-to-antibody ratios (DAR) of 8 and 4 respectively resulted in xenograft tumor growth inhibition, with superior efficacy to trastuzumab-vc-MMAE. Trastuzumab-MMAU DAR4 was tolerated at doses up to 12 mg/kg in cyno, which represents 2- to 4-fold higher dose than that observed with vedotin ADCs, and had increased terminal half-life and exposure. CONCLUSIONS: The optimized trastuzumab-MMAU ADC showed potent antitumor activity and was well tolerated with excellent pharmacokinetics in non-human primates, leading to a superior preclinical therapeutic window. The data supports potential utility of trastuzumab-MMAU for treatment of HER2+ tumors.

3.
Mol Pharm ; 17(10): 3885-3899, 2020 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-32787269

RESUMO

Boron neutron capture therapy (BNCT) for cancer is on the rise worldwide due to recent developments of in-hospital neutron accelerators which are expected to revolutionize patient treatments. There is an urgent need for improved boron delivery agents, and herein we have focused on studying the biochemical foundations upon which a successful GLUT1-targeting strategy to BNCT could be based. By combining synthesis and molecular modeling with affinity and cytotoxicity studies, we unravel the mechanisms behind the considerable potential of appropriately designed glucoconjugates as boron delivery agents for BNCT. In addition to addressing the biochemical premises of the approach in detail, we report on a hit glucoconjugate which displays good cytocompatibility, aqueous solubility, high transporter affinity, and, crucially, an exceptional boron delivery capacity in the in vitro assessment thereby pointing toward the significant potential embedded in this approach.


Assuntos
Terapia por Captura de Nêutron de Boro/métodos , Boro/administração & dosagem , Portadores de Fármacos/efeitos da radiação , Glucose/efeitos da radiação , Isótopos/administração & dosagem , Neoplasias/radioterapia , Boro/farmacocinética , Linhagem Celular Tumoral , Portadores de Fármacos/síntese química , Portadores de Fármacos/farmacocinética , Liberação Controlada de Fármacos/efeitos da radiação , Glucose/análogos & derivados , Glucose/síntese química , Glucose/farmacocinética , Transportador de Glucose Tipo 1/metabolismo , Humanos , Isótopos/farmacocinética , Simulação de Acoplamento Molecular
4.
Nutrients ; 12(7)2020 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-32640639

RESUMO

Commensal gut microbiota and probiotics have numerous effects on the host's metabolic and protective systems, which occur primarily through the intestinal epithelial cell interface. Prebiotics, like galacto-oligosaccharides (GOS) are widely used to modulate their function and abundance. However, important structure-function relations may exist, requiring a detailed structural characterization. Here, we detailed the structural characterization of bovine whey derived oligosaccharide preparations enriched with GOS or not, dubbed GOS-enriched milk oligosaccharides (GMOS) or MOS, respectively. We explore GMOS's and MOS's potential to improve intestinal epithelial barrier function, assessed in a model based on barrier disruptive effects of the Clostridioides difficile toxin A. GMOS and MOS contain mainly GOS species composed of ß1-6- and ß1-3-linked galactoses, and 3'- and 6'-sialyllactose. Both GMOS and MOS, combined with lactobacilli, like Lactobacillus rhamnosus (LPR, NCC4007), gave synergistic epithelial barrier protection, while no such effect was observed with Bifidobacterium longum (BL NCC3001), Escherichia coli (Nissle) or fructo-oligosaccharides. Mechanistically, for barrier protection with MOS, (i) viable LPR was required, (ii) acidification of growth medium was not enough, (iii) LPR did not directly neutralize toxin A, and (iv) physical proximity of LPR with the intestinal epithelial cells was necessary. This is the first study, highlighting the importance of structure-function specificity and the necessity of the simultaneous presence of prebiotic, probiotic and host cell interactions required for a biological effect.


Assuntos
Microbioma Gastrointestinal , Mucosa Intestinal , Oligossacarídeos , Simbióticos , Soro do Leite , Animais , Toxinas Bacterianas/efeitos adversos , Bovinos , Linhagem Celular Tumoral , Enterotoxinas/efeitos adversos , Galactose/química , Galactose/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/fisiologia , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Lactobacillus/metabolismo , Oligossacarídeos/química , Oligossacarídeos/metabolismo , Oligossacarídeos/farmacologia , Prebióticos , Probióticos/farmacologia , Substâncias Protetoras/química , Substâncias Protetoras/metabolismo , Substâncias Protetoras/farmacologia
5.
J Biol Chem ; 291(31): 16307-17, 2016 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-27268056

RESUMO

We show that a peptide from Chikungunya virus nsP3 protein spanning residues 1728-1744 binds the amphiphysin-2 (BIN1) Src homology-3 (SH3) domain with an unusually high affinity (Kd 24 nm). Our NMR solution complex structure together with isothermal titration calorimetry data on several related viral and cellular peptide ligands reveal that this exceptional affinity originates from interactions between multiple basic residues in the target peptide and the extensive negatively charged binding surface of amphiphysin-2 SH3. Remarkably, these arginines show no fixed conformation in the complex structure, indicating that a transient or fluctuating polyelectrostatic interaction accounts for this affinity. Thus, via optimization of such dynamic electrostatic forces, viral peptides have evolved a superior binding affinity for amphiphysin-2 SH3 compared with typical cellular ligands, such as dynamin, thereby enabling hijacking of amphiphysin-2 SH3-regulated host cell processes by these viruses. Moreover, our data show that the previously described consensus sequence PXRPXR for amphiphysin SH3 ligands is inaccurate and instead define it as an extended Class II binding motif PXXPXRpXR, where additional positive charges between the two constant arginine residues can give rise to extraordinary high SH3 binding affinity.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/química , Vírus Chikungunya/química , Proteínas Nucleares/química , Peptídeos/química , Proteínas Supressoras de Tumor/química , Proteínas não Estruturais Virais/química , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Motivos de Aminoácidos , Vírus Chikungunya/metabolismo , Humanos , Ressonância Magnética Nuclear Biomolecular , Proteínas Nucleares/metabolismo , Peptídeos/metabolismo , Ligação Proteica , Eletricidade Estática , Relação Estrutura-Atividade , Proteínas Supressoras de Tumor/metabolismo , Proteínas não Estruturais Virais/metabolismo , Domínios de Homologia de src
6.
Proc Natl Acad Sci U S A ; 107(50): 21743-8, 2010 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-21098279

RESUMO

Src homology 3 (SH3) domains are globular protein interaction modules that regulate cell behavior. The classic SH3 ligand-binding site accommodates a hydrophobic PxxP motif and a positively charged specificity-determining residue. We have determined the NMR structure of insulin receptor tyrosine kinase substrate (IRTKS) SH3 domain in complex with a repeat from Escherichia coli-secreted protein F-like protein encoded on prophage U (EspF(U)), a translocated effector of enterohemorrhagic E. coli that commandeers the mammalian actin assembly machinery. EspF(U)-IRTKS interaction is among the highest affinity natural SH3 ligands. Our complex structure reveals a unique type of SH3 interaction based on recognition of tandem PxxP motifs in the ligand. Strikingly, the specificity pocket of IRTKS SH3 has evolved to accommodate a polyproline type II helical peptide analogously to docking of the canonical PxxP by the conserved IRTKS SH3 proline-binding pockets. This cooperative binding explains the high-affinity SH3 interaction and is required for EspF(U)-IRTKS interaction in mammalian cells as well as the formation of localized actin "pedestals" beneath bound bacteria. Importantly, tandem PxxP motifs are also found in mammalian ligands and have been shown to contribute to IRTKS SH3 recognition similarly.


Assuntos
Actinas/metabolismo , Motivos de Aminoácidos , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Domínios de Homologia de src , Sequência de Aminoácidos , Animais , Sítios de Ligação , Proteínas de Transporte/química , Células Cultivadas , Proteínas de Escherichia coli/química , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Proteínas dos Microfilamentos/química , Modelos Moleculares , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Estrutura Terciária de Proteína , Alinhamento de Sequência
7.
J Biomol NMR ; 47(3): 171-81, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20437194

RESUMO

We propose a new alpha proton detection based approach for the sequential assignment of natively unfolded proteins. The proposed protocol superimposes on following features: HA-detection (1) enables assignment of natively unfolded proteins at any pH, i.e., it is not sensitive to rapid chemical exchange undergoing in natively unfolded proteins even at moderately high pH. (2) It allows straightforward assignment of proline-rich polypeptides without additional proline-customized experiments. (3) It offers more streamlined and less ambiguous assignment based on solely intraresidual (15)N(i)-(13)C'(i)-H(alpha)(i) (or (15)N(i)-(13)C(alpha)(i)-H(alpha)(i)) and sequential (15)N(i + 1)-(13)C'(i)-H(alpha)(i) (or (15)N(i + 1)-(13)C(alpha)(i)-H(alpha)(i)) correlation experiments together with efficient use of chemical shifts of (15)N and (13)C' nuclei, which show smaller dependence on residue type. We have tested the proposed protocol on two proteins, small globular 56-residue GB1, and highly disordered, proline-rich 47-residue fifth repeat of EspF(U). Using the proposed approach, we were able to assign 90% of (1)H(alpha), (13)C(alpha), (13)C', (15)N chemical shifts in EspF(U). We reckon that the HA-detection based strategy will be very useful in the assignment of natively unfolded proline-rich proteins or polypeptide chains.


Assuntos
Ressonância Magnética Nuclear Biomolecular/métodos , Proteínas/química , Proteínas de Bactérias/química , Isótopos de Carbono/química , Proteínas de Transporte/química , Deutério/química , Proteínas de Escherichia coli/química , Peptídeos e Proteínas de Sinalização Intracelular , Isótopos de Nitrogênio/química , Dobramento de Proteína , Proteínas/classificação
8.
Eur J Pharm Sci ; 40(5): 404-11, 2010 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-20433920

RESUMO

HIV-1 Tat-peptide is widely used as a vector for cargo delivery into intact cells. As a cationic, arginine-rich peptide it can readily penetrate the plasma membrane and facilitate the penetration of impermeable bioactive molecules such as proteins, peptides, nucleic acids and drugs. Although at first considered as an inert vector, recent studies have however shown that it might have effects on its own on various cellular processes. In the present study we have investigated the effects of the Tat-peptide(48-60) on two basic serine/threonine kinases, protein kinase C and A, since earlier studies have shown that certain arginine-rich peptides or proteins might have a modulatory effect on their activity. In in vitro studies, Tat-peptide inhibited PKC alpha in a concentration-dependent manner with an IC(50)-value of 22nM and PKA with an IC(50)-value of 1.2 microM. The mode of inhibition was studied in the presence of increasing concentrations of a substrate peptide or ATP. Tat-peptide competed with the kinase substrates, however it did not compete with ATP. In a panel of 70 kinases Tat-peptide showed inhibitory activity at least towards other AGC-family kinases (PKB, SGK1, S6K1, MSK1), CAMK-family kinases (CAMK1 and MELK) and a STE family kinase (MKK1). In HeLa cells Tat-peptide inhibited the phorbol ester-evoked ERK1/2 phosphorylation suggesting that Tat inhibited PKC also in intact cells. In thyroid cells Tat-peptide attenuated sphingosylphosphorylcholine-evoked Ca(2+)-fluxes, which have earlier been shown to be dependent on PKC. Taken together, these results indicate that the Tat-peptide(48-60) is a potent inhibitor which binds to the substrate binding site of the basophilic kinase domain.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Fragmentos de Peptídeos/farmacologia , Proteína Quinase C/antagonistas & inibidores , Produtos do Gene tat do Vírus da Imunodeficiência Humana/farmacologia , Trifosfato de Adenosina/metabolismo , Cálcio/metabolismo , Linhagem Celular Tumoral , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Células HeLa , Humanos , Cinética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fragmentos de Peptídeos/metabolismo , Peptídeos/metabolismo , Fosforilação , Proteína Quinase C/metabolismo , Glândula Tireoide/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo
9.
J Med Chem ; 52(13): 3969-81, 2009 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-19438240

RESUMO

Protein kinase C (PKC) is a widely studied molecular target for the treatment of cancer and other diseases. We have approached the issue of modifying PKC function by targeting the C1 domain in the regulatory region of the enzyme. Using the X-ray crystal structure of the PKC delta C1b domain, we have discovered conveniently synthesizable derivatives of dialkyl 5-(hydroxymethyl)isophthalate that can act as potential C1 domain ligands. Structure-activity studies confirmed that the important functional groups predicted by modeling were indispensable for binding to the C1 domain and that the modifications of these groups diminished binding. The most promising compounds were able to displace radiolabeled phorbol ester ([(3)H]PDBu) from PKC alpha and delta at K(i) values in the range of 200-900 nM. Furthermore, the active isophthalate derivatives could modify PKC activation in living cells either by inducing PKC-dependent ERK phosphorylation or by inhibiting phorbol-induced ERK phosphorylation. In conclusion, we report here, for the first time, that derivatives of isophthalic acid represent an attractive novel group of C1 domain ligands that can be used as research tools or further modified for potential drug development.


Assuntos
Ácidos Ftálicos/síntese química , Proteína Quinase C/antagonistas & inibidores , Animais , Sítios de Ligação , Ligação Competitiva , Linhagem Celular , Desenho de Fármacos , Ativação Enzimática , Células HeLa , Humanos , Insetos , Fosforilação , Ácidos Ftálicos/farmacologia , Proteína Quinase C/metabolismo , Proteína Quinase C-alfa/antagonistas & inibidores , Proteína Quinase C-alfa/metabolismo , Proteína Quinase C-delta/antagonistas & inibidores , Proteína Quinase C-delta/metabolismo , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/farmacologia , Relação Estrutura-Atividade
10.
Glycoconj J ; 26(3): 367-84, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19037724

RESUMO

Human mesenchymal stem cells (MSCs) are adult multipotent progenitor cells. They hold an enormous therapeutic potential, but at the moment there is little information on the properties of MSCs, including their surface structures. In the present study, we analyzed the mesenchymal stem cell glycome by using mass spectrometric profiling as well as a panel of glycan binding proteins. Structural verifications were obtained by nuclear magnetic resonance spectroscopy, mass spectrometric fragmentation, and glycosidase digestions. The MSC glycome was compared to the glycome of corresponding osteogenically differentiated cells. More than one hundred glycan signals were detected in mesenchymal stem cells and osteoblasts differentiated from them. The glycan profiles of MSCs and osteoblasts were consistently different in biological replicates, indicating that stem cells and osteoblasts have characteristic glycosylation features. Glycosylation features associated with MSCs rather than differentiated cells included high-mannose type N-glycans, linear poly-N-acetyllactosamine chains and alpha2-3-sialylation. Mesenchymal stem cells expressed SSEA-4 and sialyl Lewis x epitopes. Characteristic glycosylation features that appeared in differentiated osteoblasts included abundant sulfate ester modifications. The results show that glycosylation analysis can be used to evaluate MSC differentiation state.


Assuntos
Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Diferenciação Celular , Glicômica , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Configuração de Carboidratos , Sequência de Carboidratos , Linhagem Celular , Citometria de Fluxo , Humanos , Espectrometria de Massas , Dados de Sequência Molecular , Ácido N-Acetilneuramínico/metabolismo , Polissacarídeos/química , Ligação Proteica , Reprodutibilidade dos Testes
11.
J Mol Biol ; 382(1): 167-78, 2008 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-18644376

RESUMO

We have determined the solution structure of epidermal growth factor receptor pathway substrate 8 (Eps8) L1 Src homology 3 (SH3) domain in complex with the PPVPNPDYEPIR peptide from the CD3epsilon cytoplasmic tail. Our structure reveals the distinct structural features that account for the unusual specificity of the Eps8 family SH3 domains for ligands containing a PxxDY motif instead of canonical PxxP ligands. The CD3epsilon peptide binds Eps8L1 SH3 in a class II orientation, but neither adopts a polyproline II helical conformation nor engages the first proline-binding pocket of the SH3 ligand binding interface. Ile531 of Eps8L1 SH3, instead of Tyr or Phe residues typically found in this position in SH3 domains, renders this hydrophobic pocket smaller and nonoptimal for binding to conventional PxxP peptides. A positively charged arginine at position 512 in the n-Src loop of Eps8L1 SH3 plays a key role in PxxDY motif recognition by forming a salt bridge to D7 of the CD3epsilon peptide. In addition, our structural model suggests a hydrogen bond between the hydroxyl group of the aromatic ring of Y8 and the carboxyl group of E496, thus explaining the critical role of the PxxDY motif tyrosine residue in binding to Eps8 family SH3. These finding have direct implications also for understanding the atypical binding specificity of the amino-terminal SH3 of the Nck family proteins.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Domínios de Homologia de src , Motivos de Aminoácidos , Sequência de Aminoácidos , Complexo CD3/metabolismo , Calorimetria , Ligantes , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Mapeamento de Interação de Proteínas , Estrutura Secundária de Proteína , Alinhamento de Sequência , Relação Estrutura-Atividade
12.
Drug Metab Dispos ; 36(8): 1529-37, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18474681

RESUMO

Medetomidine is a chiral imidazole derivate whose dextroenantiomer is pharmacologically active. The major metabolic pathway of dexmedetomidine [(+)-4-(S)-[1-(2,3-dimethylphenyl)ethyl]-1H-imidazole] in humans is N-glucuronidation at the imidazolate nitrogens. We have purified the N3- and N1-glucuronides of dexmedetomidine, termed DG1 and DG2, respectively, according to their elution order in liquid chromatography and determined their structure by 1H nuclear magnetic resonance (NMR). Studying medetomidine glucuronidation by human liver microsomes (HLMs) and recombinant UDP glucuronosyltransferase (UGT) 1A4 indicated that another human UGT plays a major role in these activities. We now demonstrate that this enzyme is UGT2B10. HLMs catalyzed DG1 and DG2 formation, at a ratio of 3:1, with two-enzyme kinetics that contain both a high-affinity component, K(m1) values of 6.6 and 8.7 microM, and a low-affinity component, K(m2) values > 1 mM. The DG1/DG2 ratio in the case of UGT2B10 was lower, 1.4:1, whereas the substrate affinity for both reactions was high, K(m) values of 11 and 16 microM. UGT1A4 produced mainly DG1 (DG1/DG2 ratio of 6.6:1) at low substrate affinities, K(m) values above 0.6 mM, but superior expression-normalized V(max) values. Levomedetomidine [(-)-4-(R)-[1-(2,3-dimethylphenyl)ethyl]-1H-imidazole] glucuronidation by HLMs yielded mostly the N3-glucuronide (LG1, structure determined by NMR), with monophasic kinetics and a K(m) value of 14 microM. The activity of UGT1A4 toward levomedetomide was low and generated both LG1 and LG2, whereas UGT2B10 exhibited relatively high activity and sharp regioselectivity, yielding only LG1, with a K(m) value of 7.4 microM. The results highlight the contribution of UGT2B10 to medetomidine glucuronidation and its potential importance for other N-glucuronidation reactions within the human liver.


Assuntos
Agonistas alfa-Adrenérgicos/farmacocinética , Analgésicos não Narcóticos/farmacocinética , Glucuronídeos/metabolismo , Glucuronosiltransferase/metabolismo , Isoenzimas/metabolismo , Medetomidina/farmacocinética , Microssomos Hepáticos/enzimologia , Cromatografia Líquida , Humanos , Espectroscopia de Ressonância Magnética , Espectrometria de Massas , Proteínas Recombinantes/metabolismo , Espectrofotometria Ultravioleta
13.
Bioorg Chem ; 36(3): 148-55, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18378273

RESUMO

Three angiotensin II receptor antagonists--losartan, candesartan, and zolarsartan--were investigated. All the compounds, which are structural analogues, are metabolized via conjugation to glucuronic acid. Interestingly, both O- and N-glucuronidation take place, so that regioisomers are formed. One ether O-glucuronide, two acyl O-glucuronides, and five tetrazole-N-glucuronides were biosynthesized, in milligram scale, from the three sartan aglycones. Liver microsomes from bovine, moose, rat, and pig and recombinant human UDP-glucuronosyltransferases were used as catalysts. The synthesized compounds were identified as sartan glucuronides by mass spectrometry, while the sites of glucuronidation were determined by nuclear magnetic resonance spectroscopy. Drug metabolites are needed as standards for pharmaceutical research and, as the present study shows, they can easily be produced with enzymes as catalyst.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/síntese química , Antagonistas de Receptores de Angiotensina , Glucuronídeos/farmacologia , Animais , Benzimidazóis , Compostos de Bifenilo , Bovinos , Cervos , Ácido Glucurônico , Glucuronídeos/síntese química , Glucuronosiltransferase/metabolismo , Humanos , Losartan , Espectroscopia de Ressonância Magnética , Espectrometria de Massas , Microssomos Hepáticos/metabolismo , Ratos , Suínos , Tetrazóis
14.
Exp Hematol ; 35(8): 1279-92, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17662891

RESUMO

OBJECTIVE: Cell surface glycans contribute to the adhesion capacity of cells and are essential in cellular signal transduction. Yet, the glycosylation of hematopoietic stem and progenitor cells (HSPC), such as CD133+ cells, is poorly explored. MATERIALS AND METHODS: N-glycan structures of cord blood-derived CD133+ and CD133- cells were analyzed with mass spectrometric profiling and exoglycosidase digestion, cell surface glycan epitopes with lectin binding assay, and expression of N-glycan biosynthesis-related genes with microarray analysis. RESULTS: Over 10% difference was demonstrated in the N-glycan profiles of CD133+ and CD133- cells. Biantennary complex-type N-glycans were enriched in CD133+ cells. Of the genes regulating the synthesis of these structures, CD133+ cells overexpressed MGAT2 and underexpressed MGAT4. Moreover, the amount of high-mannose type N-glycans and terminal alpha2,3-sialylation was increased in CD133+ cells. Elevated alpha2,3-sialylation was supported by the overexpression of ST3GAL6. CONCLUSION: Our work presents new information on the characters of HSPCs. The new knowledge of HSPC-specific N-glycosylation advances their identification and provides tools to promote HSPC homing and mobilization or targeting to specific tissues.


Assuntos
Antígenos CD/genética , Regulação da Expressão Gênica , Glicoproteínas/genética , Células-Tronco Hematopoéticas/fisiologia , Peptídeos/genética , Polissacarídeos/química , Células-Tronco/fisiologia , Antígeno AC133 , Antígenos CD/biossíntese , Glicoproteínas/biossíntese , Glicoproteínas/deficiência , Glicosilação , Humanos , Recém-Nascido , Cinética , Análise de Sequência com Séries de Oligonucleotídeos , Peptídeos/deficiência
15.
Glycoconj J ; 24(2-3): 157-65, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17268859

RESUMO

Carbohydrates present on cell surfaces participate in numerous biological recognition phenomena including cell-cell interactions, cancer metastasis and pathogen invasion. Therefore, synthetic carbohydrates have a potential to act as pharmaceutical substances for treatment of various pathological phenomena by inhibiting specifically the interaction between cell surface carbohydrates and their protein receptors (lectins). However, the inherently low affinity of carbohydrate-protein interactions has often been an obstacle for successful generation of carbohydrate based pharmaceuticals. Multivalent glycoconjugates, i.e. structures carrying several copies of the active carbohydrate sequence in a carrier molecule, have been constructed to overcome this problem. Here we present two novel types of multivalent carbohydrate conjugates based on chondroitin oligomer and cyclodextrin carriers. These carriers were modified to express primary amino groups, and oligosaccharides were then bound to carrier molecules by reductive amination. Multivalent conjugates were produced using the human milk type oligosaccharides LNDFH I (Lewis-b hexasaccharide), LNnT, and GlcNAcbeta1-3Galbeta1-4GlcNAcbeta1-3Galbeta1-4Glc.


Assuntos
Condroitina/análogos & derivados , Glicoconjugados/química , Glicoconjugados/síntese química , Oligossacarídeos/química , Oligossacarídeos/síntese química , gama-Ciclodextrinas/química , Aminas/síntese química , Aminas/química , Sequência de Carboidratos , Condroitina/síntese química , Condroitina/química , Diaminas/química , Portadores de Fármacos/síntese química , Portadores de Fármacos/química , Glicoconjugados/biossíntese , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Oligossacarídeos/biossíntese , Oxirredução , Sialiltransferases/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , gama-Ciclodextrinas/síntese química , beta-D-Galactosídeo alfa 2-6-Sialiltransferase
16.
Biochemistry ; 45(17): 5567-75, 2006 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-16634638

RESUMO

Synaptic delivery of GluR-A (GluR1) subunit-containing glutamate receptors depends on a C-terminal type I PDZ binding motif in GluR-A. Synapse-associated protein 97 (SAP97) is the only PDZ domain protein known to associate with GluR-A. We have used NMR spectroscopy and a biotinylated peptide binding assay to characterize the interaction between synthetic GluR-A C-terminal peptides and the PDZ2 domain of SAP97 (SAP97(PDZ2)), previously determined to be the dominant factor responsible for the interaction. The binding mode appeared to be strongly influenced by redox conditions. Chemical shift changes observed in NMR spectra indicate that under reducing conditions, the last four residues of GluR-A peptides bind to PDZ2 in a fashion typical of class I PDZ interactions. The binding is weak and relatively nonselective as it occurs similarly with a PDZ2 domain derived from PSD-95, a related protein not believed to directly interact with GluR-A. In the absence of reducing agents, conserved cysteine residues in SAP97(PDZ2) and the GluR-A C-terminus gave rise to an anomalous behavior in a microplate assay with a biotinylated GluR-A 18-mer peptide. A covalent disulfide-linked complex between SAP97(PDZ2) and the GluR-A peptide was seen in the binding assay and in the NMR experiments performed under oxidizing conditions. The results are consistent with a two-step binding mechanism consisting of an initial PDZ interaction followed by stabilization of the complex by a disulfide bond. The possible physiological relevance of redox regulation of SAP97-GluR-A interaction remains to be established.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Dissulfetos/química , Proteínas de Membrana/metabolismo , Ligação Proteica/fisiologia , Receptores de AMPA/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Sequência de Aminoácidos , Animais , Sítios de Ligação , Biotinilação , Isótopos de Carbono , Dimerização , Proteínas de Membrana/química , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Oxirredução , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Ratos , Receptores de AMPA/química
17.
Biochemistry ; 45(9): 2862-71, 2006 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-16503641

RESUMO

Integrins are potential targets for the development of antiinflammatory agents. Here we develop a novel high-throughput assay by allowing a chemical library to compete with phage display peptide binding and identify a novel small-molecule ligand to the leukocyte-specific alpha(M)beta(2) integrin. The identified thioxothiazolidine-containing compound, IMB-10, had an unexpected activity in that it stabilized binding of alpha(M)beta(2) to its endogenous ligands proMMP-9 and fibrinogen. Single amino acid substitutions in the activity-regulating C-terminal helix and the underlying region in the ligand-binding I domain of the integrin suppressed the effect of IMB-10. A computational model indicated that IMB-10 occupies a distinct cavity present only in the activated form of the integrin I domain. IMB-10 inhibited alpha(M)beta(2)-dependent migration in vitro and inflammation-induced neutrophil emigration in vivo. Stabilization of integrin-mediated adhesion by a small molecule is a novel means to inhibit cell migration and may have a utility in treatment of inflammatory diseases involving leukocyte recruitment.


Assuntos
Anti-Inflamatórios/farmacologia , Movimento Celular/efeitos dos fármacos , Leucócitos/metabolismo , Antígeno de Macrófago 1/metabolismo , Tiazóis/farmacologia , Sequência de Aminoácidos , Animais , Anti-Inflamatórios/metabolismo , Feminino , Ligantes , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Dados de Sequência Molecular , Biblioteca de Peptídeos , Estrutura Terciária de Proteína/efeitos dos fármacos , Proteínas Recombinantes de Fusão/metabolismo , Tiazóis/metabolismo , Células Tumorais Cultivadas
18.
Drug Metab Dispos ; 33(5): 657-63, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15689500

RESUMO

Selected aspects of dobutamine glucuronidation were studied in detail. There are potentially four sites at which dobutamine can be conjugated to glucuronic acid. Three of the four dobutamine monoglucuronides that can be formed were enzymatically synthesized using pig liver microsomes, isolated, and characterized by tandem mass spectrometry, and (1)H and (13)C NMR spectroscopy. Analysis of dobutamine glucuronidation by liver microsomes from various sources revealed large variability in the ratios of the three regioisomers. Interestingly, catecholic dobutamine meta-O-glucuronide, by far the major product synthesized with human liver microsomes, was only a minor product for rat liver microsomes. Rabbit liver microsomes yielded diglucuronides, in addition to monoglucuronides. Activities of individual recombinant human UDP-glucuronosyltransferases (UGTs) were investigated, and the results suggested that dobutamine glucuronidation in the human liver is mainly carried out by UGTs 2B7 and 1A9. Among the extrahepatic UGTs, the formation of monoglucuronides, mainly catecholic meta-O-glucuronide, by UGTs 1A7 and 1A8 was similar to that by 1A9, whereas UGT1A10 also efficiently catalyzed the formation of catecholic dobutamine para-O-glucuronide.


Assuntos
Agonistas Adrenérgicos beta/metabolismo , Dobutamina/metabolismo , Glucuronídeos/metabolismo , Glucuronosiltransferase/metabolismo , Animais , Cromatografia Líquida de Alta Pressão , Humanos , Isomerismo , Espectroscopia de Ressonância Magnética , Espectrometria de Massas , Microssomos Hepáticos/enzimologia , Coelhos , Ratos , Proteínas Recombinantes/metabolismo , Padrões de Referência , Espectrofotometria Ultravioleta , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA