Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 50(3): 668-676.e5, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30824324

RESUMEN

Human polyomaviruses cause a common childhood infection worldwide and typically elicit a neutralizing antibody and cellular immune response, while establishing a dormant infection in the kidney with minimal clinical manifestations. However, viral reactivation can cause severe pathology in immunocompromised individuals. We developed a high-throughput, functional antibody screen to examine the humoral response to BK polyomavirus. This approach enabled the isolation of antibodies from all peripheral B cell subsets and revealed the anti-BK virus antibody repertoire as clonally complex with respect to immunoglobulin sequences and isotypes (both IgM and IgG), including a high frequency of monoclonal antibodies that broadly neutralize BK virus subtypes and the related JC polyomavirus. Cryo-electron microscopy of a broadly neutralizing IgG single-chain variable fragment complexed with BK virus-like particles revealed the quaternary nature of a conserved viral epitope at the junction between capsid pentamers. These features unravel a potent modality for inhibiting polyomavirus infection in kidney transplant recipients and other immunocompromised patients.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Linfocitos B/inmunología , Virus BK/inmunología , Memoria Inmunológica/inmunología , Virus JC/inmunología , Infecciones por Polyomavirus/inmunología , Poliomavirus/inmunología , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Cápside/inmunología , Línea Celular , Epítopos/inmunología , Células HEK293 , Humanos , Inmunidad Celular/inmunología , Riñón/inmunología
2.
Blood ; 133(13): 1507-1516, 2019 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-30692123

RESUMEN

A large unmet medical need exists for safer antithrombotic drugs because all currently approved anticoagulant agents interfere with hemostasis, leading to an increased risk of bleeding. Genetic and pharmacologic evidence in humans and animals suggests that reducing factor XI (FXI) levels has the potential to effectively prevent and treat thrombosis with a minimal risk of bleeding. We generated a fully human antibody (MAA868) that binds the catalytic domain of both FXI (zymogen) and activated FXI. Our structural studies show that MAA868 traps FXI and activated FXI in an inactive, zymogen-like conformation, explaining its equally high binding affinity for both forms of the enzyme. This binding mode allows the enzyme to be neutralized before entering the coagulation process, revealing a particularly attractive anticoagulant profile of the antibody. MAA868 exhibited favorable anticoagulant activity in mice with a dose-dependent protection from carotid occlusion in a ferric chloride-induced thrombosis model. MAA868 also caused robust and sustained anticoagulant activity in cynomolgus monkeys as assessed by activated partial thromboplastin time without any evidence of bleeding. Based on these preclinical findings, we conducted a first-in-human study in healthy subjects and showed that single subcutaneous doses of MAA868 were safe and well tolerated. MAA868 resulted in dose- and time-dependent robust and sustained prolongation of activated partial thromboplastin time and FXI suppression for up to 4 weeks or longer, supporting further clinical investigation as a potential once-monthly subcutaneous anticoagulant therapy.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticoagulantes/uso terapéutico , Coagulación Sanguínea/efectos de los fármacos , Factor XI/antagonistas & inhibidores , Trombosis/tratamiento farmacológico , Adolescente , Adulto , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Anticoagulantes/farmacología , Femenino , Humanos , Inmunoglobulina G/farmacología , Inmunoglobulina G/uso terapéutico , Macaca fascicularis , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Simulación del Acoplamiento Molecular , Trombosis/sangre , Adulto Joven
3.
Proc Natl Acad Sci U S A ; 114(47): 12448-12453, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29109273

RESUMEN

The TGF-ß family ligands myostatin, GDF11, and activins are negative regulators of skeletal muscle mass, which have been reported to primarily signal via the ActRIIB receptor on skeletal muscle and thereby induce muscle wasting described as cachexia. Use of a soluble ActRIIB-Fc "trap," to block myostatin pathway signaling in normal or cachectic mice leads to hypertrophy or prevention of muscle loss, perhaps suggesting that the ActRIIB receptor is primarily responsible for muscle growth regulation. Genetic evidence demonstrates however that both ActRIIB- and ActRIIA-deficient mice display a hypertrophic phenotype. Here, we describe the mode of action of bimagrumab (BYM338), as a human dual-specific anti-ActRIIA/ActRIIB antibody, at the molecular and cellular levels. As shown by X-ray analysis, bimagrumab binds to both ActRIIA and ActRIIB ligand binding domains in a competitive manner at the critical myostatin/activin binding site, hence preventing signal transduction through either ActRII. Myostatin and the activins are capable of binding to both ActRIIA and ActRIIB, with different affinities. However, blockade of either single receptor through the use of specific anti-ActRIIA or anti-ActRIIB antibodies achieves only a partial signaling blockade upon myostatin or activin A stimulation, and this leads to only a small increase in muscle mass. Complete neutralization and maximal anabolic response are achieved only by simultaneous blockade of both receptors. These findings demonstrate the importance of ActRIIA in addition to ActRIIB in mediating myostatin and activin signaling and highlight the need for blocking both receptors to achieve a strong functional benefit.


Asunto(s)
Receptores de Activinas Tipo II/antagonistas & inhibidores , Anticuerpos Bloqueadores/farmacología , Anticuerpos Monoclonales/farmacología , Hipertrofia/inducido químicamente , Músculo Esquelético/efectos de los fármacos , Receptores de Activinas Tipo II/metabolismo , Activinas/metabolismo , Animales , Anticuerpos Bloqueadores/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados , Proteínas Morfogenéticas Óseas/metabolismo , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Factores de Diferenciación de Crecimiento/metabolismo , Células HEK293 , Humanos , Hipertrofia/patología , Masculino , Ratones , Ratones SCID , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Miostatina/metabolismo , Ratas , Ratas Wistar , Proteínas Recombinantes/metabolismo , Transducción de Señal/efectos de los fármacos , Síndrome Debilitante/tratamiento farmacológico , Síndrome Debilitante/patología
4.
Chembiochem ; 17(11): 1012-20, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-26991759

RESUMEN

FimH is a bacterial lectin found at the tips of type 1 pili of uropathogenic Escherichia coli (UPEC). It mediates shear-enhanced adhesion to mannosylated surfaces. Binding of UPEC to urothelial cells initiates the infection cycle leading to urinary tract infections (UTIs). Antiadhesive glycomimetics based on α-d-mannopyranose offer an attractive alternative to the conventional antibiotic treatment because they do not induce a selection pressure and are therefore expected to have a reduced resistance potential. Genetic variation of the fimH gene in clinically isolated UPEC has been associated with distinct mannose binding phenotypes. For this reason, we investigated the mannose binding characteristics of four FimH variants with mannose-based ligands under static and hydrodynamic conditions. The selected FimH variants showed individually different binding behavior under both sets of conditions as a result of the conformational variability of FimH. Clinically relevant FimH variants typically exist in a dynamic conformational equilibrium. Additionally, we evaluated inhibitory potencies of four FimH antagonists representing different structural classes. Inhibitory potencies of three of the tested antagonists were dependent on the binding phenotype and hence on the conformational equilibrium of the FimH variant. However, the squarate derivative was the notable exception and inhibited FimH variants irrespective of their binding phenotype. Information on antagonist affinities towards various FimH variants has remained largely unconsidered despite being essential for successful antiadhesion therapy.


Asunto(s)
Adhesinas de Escherichia coli/metabolismo , Proteínas Fimbrias/metabolismo , Adhesinas de Escherichia coli/genética , Escherichia coli/aislamiento & purificación , Escherichia coli/metabolismo , Proteínas Fimbrias/antagonistas & inhibidores , Proteínas Fimbrias/genética , Humanos , Manosa/química , Manosa/metabolismo , Mutación , Fenotipo , Unión Proteica , Estructura Terciaria de Proteína , Infecciones Urinarias/microbiología , Infecciones Urinarias/patología , Uroplaquina Ia/química , Uroplaquina Ia/metabolismo
5.
Sci Transl Med ; 15(681): eabq5241, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36724238

RESUMEN

In October 2019, Novartis launched brolucizumab, a single-chain variable fragment molecule targeting vascular endothelial growth factor A, for the treatment of neovascular age-related macular degeneration. In 2020, rare cases of retinal vasculitis and/or retinal vascular occlusion (RV/RO) were reported, often during the first few months after treatment initiation, consistent with a possible immunologic pathobiology. This finding was inconsistent with preclinical studies in cynomolgus monkeys that demonstrated no drug-related intraocular inflammation, or RV/RO, despite the presence of preexisting and treatment-emergent antidrug antibodies (ADAs) in some animals. In this study, the immune response against brolucizumab in humans was assessed using samples from clinical trials and clinical practice. In the brolucizumab-naïve population, anti-brolucizumab ADA responses were detected before any treatment, which was supported by the finding that healthy donors can harbor brolucizumab-specific B cells. This suggested prior exposure of the immune system to proteins with structural similarity. Experiments on samples showed that naïve and brolucizumab-treated ADA-positive patients developed a class-switched, high-affinity immune response, with several linear epitopes being recognized by ADAs. Only patients with RV/RO showed a meaningful T cell response upon recall with brolucizumab. Further studies in cynomolgus monkeys preimmunized against brolucizumab with adjuvant followed by intravitreal brolucizumab challenge demonstrated that high ADA titers were required to generate ocular inflammation and vasculitis/vascular thrombosis, comparable to RV/RO in humans. Immunogenicity therefore seems to be a prerequisite to develop RV/RO. However, because only 2.1% of patients with ADA develop RV/RO, additional factors must play a role in the development of RV/RO.


Asunto(s)
Vasculitis Retiniana , Animales , Humanos , Adyuvantes Inmunológicos , Inhibidores de la Angiogénesis , Inflamación , Inyecciones Intravítreas , Macaca fascicularis , Factor A de Crecimiento Endotelial Vascular
6.
Cell Mol Life Sci ; 68(24): 4091-100, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21598021

RESUMEN

The glycolipid specific Drosophila melanogaster ß1,4-N-acetylgalactosaminyltransferase B (ß4GalNAcTB) depends on a zinc finger DHHC protein family member named GalNAcTB pilot (GABPI) for activity and translocation to the Golgi. The six-membrane spanning protein actually lacks the cysteine in the cytoplasmic DHHC motif, displaying DHHS instead. Here we show that the whole conserved region around the DHHS sequence, which is essential for palmitoylation in DHHC proteins, is not required for GABPI to interact with ß4GalNAcTB. In contrast, the two luminal loops between transmembrane domain 3-4 and 5-6 contain conserved amino acids, which are crucial for activity. Besides the dependence on GABPI, ß4GalNAcTB requires its exceptional short stem region for activity. A few hydrophobic amino acids positioned close to the transmembrane domain are essential for the interaction with GABPI. Along with its catalytic domain, ß4GalNAcTB, thus, requires an area in its own stem region and two small luminal loops of GABPI as "add-on" domains. Moreover, some inactive GABPI mutants could be rescued by fusion with ß4GalNAcTB, indicating their importance in direct GABPI-ß4GalNAcTB interaction.


Asunto(s)
Proteínas de Drosophila/química , Drosophila melanogaster/metabolismo , Proteínas de la Membrana/química , N-Acetilgalactosaminiltransferasas/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiología , Drosophila melanogaster/enzimología , Aparato de Golgi/metabolismo , Células HEK293 , Humanos , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/fisiología , Datos de Secuencia Molecular , N-Acetilgalactosaminiltransferasas/metabolismo , N-Acetilgalactosaminiltransferasas/fisiología , Estructura Terciaria de Proteína , Alineación de Secuencia , Análisis de Secuencia de Proteína
7.
Mol Cancer Ther ; 19(10): 2089-2104, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32847974

RESUMEN

The sole inhibitory Fcγ receptor CD32b (FcγRIIb) is expressed throughout B and plasma cell development and on their malignant counterparts. CD32b expression on malignant B cells is known to provide a mechanism of resistance to rituximab that can be ameliorated with a CD32b-blocking antibody. CD32b, therefore, represents an attractive tumor antigen for targeting with a monoclonal antibody (mAb). To this end, two anti-CD32b mAbs, NVS32b1 and NVS32b2, were developed. Their complementarity-determining regions (CDR) bind the CD32b Fc binding domain with high specificity and affinity while the Fc region is afucosylated to enhance activation of FcγRIIIa on immune effector cells. The NVS32b mAbs selectively target CD32b+ malignant cells and healthy B cells but not myeloid cells. They mediate potent killing of opsonized CD32b+ cells via antibody-dependent cellular cytotoxicity and phagocytosis (ADCC and ADCP) as well as complement-dependent cytotoxicity (CDC). In addition, NVS32b CDRs block the CD32b Fc-binding domain, thereby minimizing CD32b-mediated resistance to therapeutic mAbs including rituximab, obinutuzumab, and daratumumab. NVS32b mAbs demonstrate robust antitumor activity against CD32b+ xenografts in vivo and immunomodulatory activity including recruitment of macrophages to the tumor and enhancement of dendritic cell maturation in response to immune complexes. Finally, the activity of NVS32b mAbs on CD32b+ primary malignant B and plasma cells was confirmed using samples from patients with B-cell chronic lymphocytic leukemia (CLL) and multiple myeloma. The findings indicate the promising potential of NVS32b mAbs as a single agent or in combination with other mAb therapeutics for patients with CD32b+ malignant cells.


Asunto(s)
Linfoma de Células B/genética , Neoplasias de Células Plasmáticas/genética , Receptores de IgG/inmunología , Animales , Células CHO , Cricetulus , Humanos
8.
Sci Rep ; 6: 27996, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27302108

RESUMEN

During bacterial pathogenesis extensive contacts between the human and the bacterial extracellular proteomes take place. The identification of novel host-pathogen interactions by standard methods using a case-by-case approach is laborious and time consuming. To overcome this limitation, we took advantage of large libraries of human and bacterial recombinant proteins. We applied a large-scale protein microarray-based screening on two important human pathogens using two different approaches: (I) 75 human extracellular proteins were tested on 159 spotted Staphylococcus aureus recombinant proteins and (II) Neisseria meningitidis adhesin (NadA), an important vaccine component against serogroup B meningococcus, was screened against ≈2300 spotted human recombinant proteins. The approach presented here allowed the identification of the interaction between the S. aureus immune evasion protein FLIPr (formyl-peptide receptor like-1 inhibitory protein) and the human complement component C1q, key players of the offense-defense fighting; and of the interaction between meningococcal NadA and human LOX-1 (low-density oxidized lipoprotein receptor), an endothelial receptor. The novel interactions between bacterial and human extracellular proteins here presented might provide a better understanding of the molecular events underlying S. aureus and N. meningitidis pathogenesis.


Asunto(s)
Interacciones Huésped-Patógeno , Neisseria meningitidis/fisiología , Análisis por Matrices de Proteínas/métodos , Staphylococcus aureus/fisiología , Adhesinas Bacterianas/química , Adhesinas Bacterianas/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Sitios de Unión , Células CHO , Complemento C1q/metabolismo , Cricetulus , Humanos , Unión Proteica , Proteínas Recombinantes/metabolismo , Receptores Depuradores de Clase E/metabolismo
9.
Nat Commun ; 7: 13027, 2016 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-27708334

RESUMEN

Type 1 diabetes (T1D) is characterized by a chronic, progressive autoimmune attack against pancreas-specific antigens, effecting the destruction of insulin-producing ß-cells. Here we show interleukin-2 (IL-2) is a non-pancreatic autoimmune target in T1D. Anti-IL-2 autoantibodies, as well as T cells specific for a single orthologous epitope of IL-2, are present in the peripheral blood of non-obese diabetic (NOD) mice and patients with T1D. In NOD mice, the generation of anti-IL-2 autoantibodies is genetically determined and their titre increases with age and disease onset. In T1D patients, circulating IgG memory B cells specific for IL-2 or insulin are present at similar frequencies. Anti-IL-2 autoantibodies cloned from T1D patients demonstrate clonality, a high degree of somatic hypermutation and nanomolar affinities, indicating a germinal centre origin and underscoring the synergy between cognate autoreactive T and B cells leading to defective immune tolerance.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Tolerancia Inmunológica , Interleucina-2/inmunología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Autoanticuerpos/inmunología , Epítopos/inmunología , Femenino , Humanos , Inmunoglobulina G/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Persona de Mediana Edad , Páncreas/inmunología , Péptidos/inmunología , Linfocitos T/citología , Adulto Joven
10.
J Med Chem ; 58(5): 2221-39, 2015 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-25666045

RESUMEN

Urinary tract infections (UTIs), predominantly caused by uropathogenic Escherichia coli (UPEC), belong to the most prevalent infectious diseases worldwide. The attachment of UPEC to host cells is mediated by FimH, a mannose-binding adhesin at the tip of bacterial type 1 pili. To date, UTIs are mainly treated with antibiotics, leading to the ubiquitous problem of increasing resistance against most of the currently available antimicrobials. Therefore, new treatment strategies are urgently needed. Here, we describe the development of an orally available FimH antagonist. Starting from the carboxylate substituted biphenyl α-d-mannoside 9, affinity and the relevant pharmacokinetic parameters (solubility, permeability, renal excretion) were substantially improved by a bioisosteric approach. With 3'-chloro-4'-(α-d-mannopyranosyloxy)biphenyl-4-carbonitrile (10j) a FimH antagonist with an optimal in vitro PK/PD profile was identified. Orally applied, 10j was effective in a mouse model of UTI by reducing the bacterial load in the bladder by about 1000-fold.


Asunto(s)
Antibacterianos/farmacología , Compuestos de Bifenilo/farmacología , Infecciones por Escherichia coli/tratamiento farmacológico , Proteínas Fimbrias/antagonistas & inhibidores , Manósidos/farmacología , Infecciones Urinarias/tratamiento farmacológico , Escherichia coli Uropatógena/efectos de los fármacos , Adhesinas de Escherichia coli , Administración Oral , Animales , Antibacterianos/administración & dosificación , Antibacterianos/química , Antibacterianos/farmacocinética , Compuestos de Bifenilo/química , Compuestos de Bifenilo/farmacocinética , Células CACO-2 , Permeabilidad de la Membrana Celular/efectos de los fármacos , Cristalografía por Rayos X , Diseño de Fármacos , Infecciones por Escherichia coli/microbiología , Femenino , Citometría de Flujo , Humanos , Técnicas In Vitro , Manósidos/administración & dosificación , Manósidos/química , Manósidos/farmacocinética , Ratones , Ratones Endogámicos C3H , Estereoisomerismo , Relación Estructura-Actividad , Distribución Tisular , Infecciones Urinarias/microbiología
11.
ChemMedChem ; 9(1): 78-83, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24357503

RESUMEN

The lectin FimH is terminally expressed on type 1 pili of uropathogenic Escherichia coli (UPEC), which is the main cause of urinary tract infections (UTIs). FimH enables bacterial adhesion to urothelial cells, the initial step of infection. Various mannose derivatives have been shown to antagonize FimH and are therefore considered to be promising therapeutic agents for the treatment of UTIs. As part of the preclinical development process, when the kinetic properties of FimH antagonists were examined by surface plasmon resonance, extremely low dissociation rates (k(off)) were found, which is uncommon for carbohydrate-lectin interactions. As a consequence, the corresponding half-lives (t1/2) of the FimH antagonist complexes are above 3.6 h. For a therapeutic application, extended t1/2 values are a prerequisite for success, since the target occupancy time directly influences the in vivo drug efficacy. The long t1/2 value of the tested FimH antagonists further confirms their drug-like properties and their high therapeutic potential.


Asunto(s)
Proteínas Fimbrias/antagonistas & inhibidores , Lectinas/metabolismo , Manosa/metabolismo , Adhesinas de Escherichia coli/metabolismo , Sitios de Unión , Escherichia coli/metabolismo , Proteínas Fimbrias/metabolismo , Semivida , Cinética , Lectinas/química , Manosa/química , Unión Proteica
12.
J Med Chem ; 55(22): 9810-6, 2012 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-23088608

RESUMEN

Mannose-based FimH antagonists are considered new therapeutics for the treatment of urinary tract infections (UTIs). They prevent the adhesion of uropathogenic Escherichia coli (UPEC) to urothelial cell surfaces triggered by the lectin FimH, which is located at the tip of bacterial type 1 pili. Because all reported FimH antagonists are α-d-mannosides, they are also potential ligands of mannose receptors of the human host system. We therefore investigated the selectivity range of five FimH antagonists belonging to different compound families by comparing their affinities for FimH and eight human mannose receptors. On the basis of the detected selectivity range of approximately 5 orders of magnitude, no adverse side effects resulting from nonselective binding to the human receptors have to be expected. FimH antagonists can therefore be further considered as potential therapeutics for the treatment of UTI.


Asunto(s)
Adhesinas de Escherichia coli/efectos de los fármacos , Proteínas Fimbrias/antagonistas & inhibidores , Interacciones Huésped-Patógeno/efectos de los fármacos , Manosa/metabolismo , Manósidos/farmacología , Infecciones Urinarias/tratamiento farmacológico , Escherichia coli Uropatógena/efectos de los fármacos , Humanos , Lectinas Tipo C/metabolismo , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Estructura Molecular , Proteína Básica de Mielina/metabolismo , Receptores de Superficie Celular/metabolismo , Relación Estructura-Actividad
13.
ChemMedChem ; 7(8): 1404-22, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22644941

RESUMEN

Urinary tract infections (UTIs) are caused primarily by uropathogenic Escherichia coli (UPEC), which encode filamentous surface-adhesive organelles called type 1 pili. FimH is located at the tips of these pili. The initial attachment of UPEC to host cells is mediated by the interaction of the carbohydrate recognition domain (CRD) of FimH with oligomannosides on urothelial cells. Blocking these lectins with carbohydrates or analogues thereof prevents bacterial adhesion to host cells and therefore offers a potential therapeutic approach for prevention and/or treatment of UTIs. Although numerous FimH antagonists have been developed so far, few of them meet the requirement for clinical application due to poor pharmacokinetics. Additionally, the binding mode of an antagonist to the CRD of FimH can switch from an in-docking mode to an out-docking mode, depending on the structure of the antagonist. In this communication, biphenyl α-D-mannosides were modified to improve their binding affinity, to explore their binding mode, and to optimize their pharmacokinetic properties. The inhibitory potential of the FimH antagonists was measured in a cell-free competitive binding assay, a cell-based flow cytometry assay, and by isothermal titration calorimetry. Furthermore, pharmacokinetic properties such as log D, solubility, and membrane permeation were analyzed. As a result, a structure-activity and structure-property relationships were established for a series of biphenyl α-D-mannosides.


Asunto(s)
Compuestos de Bifenilo/química , Proteínas Fimbrias/antagonistas & inhibidores , Manosa/química , Adhesinas de Escherichia coli/metabolismo , Sitios de Unión , Calorimetría , Línea Celular Tumoral , Cristalografía por Rayos X , Escherichia coli/metabolismo , Proteínas Fimbrias/metabolismo , Semivida , Humanos , Manosa/síntesis química , Manosa/farmacocinética , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Termodinámica , Infecciones Urinarias/microbiología
14.
J Med Chem ; 55(10): 4700-13, 2012 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-22519985

RESUMEN

The initial step for the successful establishment of urinary tract infections (UTIs), predominantly caused by uropathogenic Escherichia coli, is the adhesion of bacteria to urothelial cells. This attachment is mediated by FimH, a mannose-binding adhesin, which is expressed on the bacterial surface. To date, UTIs are mainly treated with antibiotics, leading to the ubiquitous problem of increasing resistance against most of the currently available antimicrobials. Therefore, new treatment strategies are urgently needed, avoiding selection pressure and thereby implying a reduced risk of resistance. Here, we present a new class of highly active antimicrobials, targeting the virulence factor FimH. When the most potent representative, an indolinylphenyl mannoside, was administered in a mouse model at the low dosage of 1 mg/kg (corresponding to approximately 25 µg/mouse), the minimal therapeutic concentration to prevent UTI was maintained for more than 8 h. In a treatment study, the colony-forming units in the bladder could be reduced by almost 4 orders of magnitude, comparable to the standard antibiotic treatment with ciprofloxacin (8 mg/kg, sc).


Asunto(s)
Antiinfecciosos Urinarios/farmacología , Adhesión Bacteriana/efectos de los fármacos , Derivados del Benceno/farmacología , Infecciones por Escherichia coli/tratamiento farmacológico , Proteínas Fimbrias/antagonistas & inhibidores , Indoles/farmacología , Manósidos/farmacología , Infecciones Urinarias/tratamiento farmacológico , Adhesinas de Escherichia coli , Animales , Antiinfecciosos Urinarios/síntesis química , Antiinfecciosos Urinarios/farmacocinética , Derivados del Benceno/síntesis química , Derivados del Benceno/farmacocinética , Línea Celular , Infecciones por Escherichia coli/enzimología , Indoles/síntesis química , Indoles/farmacocinética , Riñón/efectos de los fármacos , Riñón/microbiología , Manósidos/síntesis química , Manósidos/farmacocinética , Ratones , Pruebas de Sensibilidad Microbiana , Relación Estructura-Actividad , Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/microbiología , Infecciones Urinarias/enzimología , Escherichia coli Uropatógena/efectos de los fármacos , Escherichia coli Uropatógena/enzimología , Escherichia coli Uropatógena/aislamiento & purificación , Urotelio/citología , Factores de Virulencia/antagonistas & inhibidores
15.
Assay Drug Dev Technol ; 9(5): 455-64, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21675870

RESUMEN

Urinary tract infections (UTIs), including cystitis and pyelonephritis, affect a large proportion of the population and account for significant medical costs. In more than 80% of UTIs, uropathogenic Escherichia coli (UPEC) is the causative pathogen. The initial step in the pathogenesis of the infection is the adherence of UPEC to the human bladder epithelium, enabling the invasion into the host cells and the development of UTIs. This process is mediated by the lectin FimH located on type I pili and enables UPECs to attach to oligomannosides of the glycoprotein uroplakin Ia presented on uroepithelial cells. FimH antagonists such as α-d-mannopyranosides have been shown to interfere with the attachment of UPEC to their host cells, thus providing a novel therapeutic opportunity for the treatment and prevention of UTIs. In this article, we report a flow cytometry-based assay to evaluate the potential of FimH antagonists for the prevention of the infection of the human urinary bladder cell line 5637 by UPEC strain UTI89. The assay was optimized and validated, and the inhibitory potency of different α-d-mannopyranosides was determined. Finally, the IC(50) values measured by the flow cytometry-based assay were compared with those reported for other assay formats.


Asunto(s)
Adhesinas de Escherichia coli/metabolismo , Infecciones por Escherichia coli/tratamiento farmacológico , Proteínas Fimbrias/metabolismo , Citometría de Flujo/métodos , Manosa/metabolismo , Infecciones Urinarias/tratamiento farmacológico , Escherichia coli Uropatógena/fisiología , Animales , Unión Competitiva , Línea Celular Tumoral , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos , Células Epiteliales/efectos de los fármacos , Agregación Eritrocitaria/efectos de los fármacos , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Proteínas Fimbrias/antagonistas & inhibidores , Cobayas , Humanos , Immunoblotting/métodos , Concentración 50 Inhibidora , Manosa/análogos & derivados , Terapia Molecular Dirigida , Reproducibilidad de los Resultados , Vejiga Urinaria/efectos de los fármacos , Sistema Urinario/efectos de los fármacos , Infecciones Urinarias/metabolismo , Infecciones Urinarias/microbiología
16.
J Med Chem ; 53(24): 8627-41, 2010 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-21105658

RESUMEN

Urinary tract infection (UTI) by uropathogenic Escherichia coli (UPEC) is one of the most common infections, particularly affecting women. The interaction of FimH, a lectin located at the tip of bacterial pili, with high mannose structures is critical for the ability of UPEC to colonize and invade the bladder epithelium. We describe the synthesis and the in vitro/in vivo evaluation of α-D-mannosides with the ability to block the bacteria/host cell interaction. According to the pharmacokinetic properties, a prodrug approach for their evaluation in the UTI mouse model was explored. As a result, an orally available, low molecular weight FimH antagonist was identified with the potential to reduce the colony forming units (CFU) in the urine by 2 orders of magnitude and in the bladder by 4 orders of magnitude. With FimH antagonist, the great potential for the effective treatment of urinary tract infections with a new class of orally available antiinfectives could be demonstrated.


Asunto(s)
Antibacterianos/síntesis química , Benzoatos/síntesis química , Infecciones por Escherichia coli/tratamiento farmacológico , Proteínas Fimbrias/antagonistas & inhibidores , Manósidos/síntesis química , Profármacos/síntesis química , Infecciones Urinarias/tratamiento farmacológico , Escherichia coli Uropatógena/efectos de los fármacos , Adhesinas de Escherichia coli , Administración Oral , Animales , Antibacterianos/farmacocinética , Antibacterianos/farmacología , Benzoatos/farmacocinética , Benzoatos/farmacología , Unión Competitiva , Proteínas Sanguíneas/metabolismo , Células CACO-2 , Permeabilidad de la Membrana Celular , Recuento de Colonia Microbiana , Diseño de Fármacos , Infecciones por Escherichia coli/microbiología , Femenino , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Absorción Intestinal , Manósidos/farmacocinética , Manósidos/farmacología , Ratones , Profármacos/farmacocinética , Profármacos/farmacología , Unión Proteica , Solubilidad , Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/microbiología , Infecciones Urinarias/microbiología , Escherichia coli Uropatógena/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA