Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 13(1): 1833, 2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-35383169

RESUMEN

Presequence protease (PreP), a 117 kDa mitochondrial M16C metalloprotease vital for mitochondrial proteostasis, degrades presequence peptides cleaved off from nuclear-encoded proteins and other aggregation-prone peptides, such as amyloid ß (Aß). PreP structures have only been determined in a closed conformation; thus, the mechanisms of substrate binding and selectivity remain elusive. Here, we leverage advanced vitrification techniques to overcome the preferential denaturation of one of two ~55 kDa homologous domains of PreP caused by air-water interface adsorption. Thereby, we elucidate cryoEM structures of three apo-PreP open states along with Aß- and citrate synthase presequence-bound PreP at 3.3-4.6 Å resolution. Together with integrative biophysical and pharmacological approaches, these structures reveal the key stages of the PreP catalytic cycle and how the binding of substrates or PreP inhibitor drives a rigid body motion of the protein for substrate binding and catalysis. Together, our studies provide key mechanistic insights into M16C metalloproteases for future therapeutic innovations.


Asunto(s)
Péptidos beta-Amiloides , Mitocondrias , Péptidos beta-Amiloides/metabolismo , Microscopía por Crioelectrón , Humanos , Metaloproteasas/metabolismo , Mitocondrias/metabolismo , Conformación Molecular , Conformación Proteica , Especificidad por Sustrato
2.
J Mol Biol ; 433(13): 166993, 2021 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-33865867

RESUMEN

It is known that insulin-degrading-enzyme (IDE) plays a crucial role in the clearance of Alzheimer's amyloid-ß (Aß). The cysteine-free IDE mutant (cf-E111Q-IDE) is catalytically inactive against insulin, but its effect on Aß degradation is unknown that would help in the allosteric modulation of the enzyme activity. Herein, the degradation of Aß(1-40) by cf-E111Q-IDE via a non-chaperone mechanism is demonstrated by NMR and LC-MS, and the aggregation of fragmented peptides is characterized using fluorescence and electron microscopy. cf-E111Q-IDE presented a reduced effect on the aggregation kinetics of Aß(1-40) when compared with the wild-type IDE. Whereas LC-MS and diffusion ordered NMR spectroscopy revealed the generation of Aß fragments by both wild-type and cf-E111Q-IDE. The aggregation propensities and the difference in the morphological phenotype of the full-length Aß(1-40) and its fragments are explained using multi-microseconds molecular dynamics simulations. Notably, our results reveal that zinc binding to Aß(1-40) inactivates cf-E111Q-IDE's catalytic function, whereas zinc removal restores its function as evidenced from high-speed AFM, electron microscopy, chromatography, and NMR results. These findings emphasize the catalytic role of cf-E111Q-IDE on Aß degradation and urge the development of zinc chelators as an alternative therapeutic strategy that switches on/off IDE's function.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Insulisina/metabolismo , Proteínas Mutantes/metabolismo , Secuencia de Aminoácidos , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/ultraestructura , Biocatálisis , Cromatografía Líquida de Alta Presión , Humanos , Insulisina/química , Insulisina/genética , Espectrometría de Masas , Microscopía Electrónica de Transmisión , Simulación de Dinámica Molecular , Proteínas Mutantes/química , Proteínas Mutantes/genética , Mutación Missense , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/ultraestructura , Unión Proteica , Proteolisis , Especificidad por Sustrato , Zinc/química , Zinc/metabolismo
3.
Structure ; 29(7): 709-720.e3, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33378640

RESUMEN

Zinc metalloprotease 1 (Zmp1), a Mycobacterium tuberculosis 75 kDa secreted enzyme, mediates key stages of tuberculosis disease progression. The biological activity of Zmp1 presumably stems from its ability to degrade bacterium- and/or host-derived peptides. The crystal structures of Zmp1 and related M13 metalloproteases, such as neprilysin and endothelin-converting enzyme-1 were determined only in the closed conformation, which cannot capture substrates or release proteolytic products. Thus, the mechanisms of substrate binding and selectivity remain elusive. Here we report two open-state cryo-EM structures of Zmp1, revealed by our SAXS analysis to be the dominant states in solution. Our structural analyses reveal how ligand binding induces a conformational switch in four linker regions to drive the rigid body motion of the D1 and D2 domains, which form the sizable catalytic chamber. Furthermore, they offer insights into the catalytic cycle and mechanism of substrate recognition of M13 metalloproteases for future therapeutic innovations.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Metaloproteasas/química , Metaloproteasas/metabolismo , Mycobacterium tuberculosis/enzimología , Microscopía por Crioelectrón , Ligandos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Dominios Proteicos , Dispersión del Ángulo Pequeño , Difracción de Rayos X
4.
J Biol Chem ; 295(11): 3506-3517, 2020 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-32019868

RESUMEN

Pseudomonas aeruginosa uses a type III secretion system (T3SS) to inject cytotoxic effector proteins into host cells. The promiscuous nucleotidyl cyclase, exoenzyme Y (ExoY), is one of the most common effectors found in clinical P. aeruginosa isolates. Recent studies have revealed that the nucleotidyl cyclase activity of ExoY is stimulated by actin filaments (F-actin) and that ExoY alters actin cytoskeleton dynamics in vitro, via an unknown mechanism. The actin cytoskeleton plays an important role in numerous key biological processes and is targeted by many pathogens to gain competitive advantages. We utilized total internal reflection fluorescence microscopy, bulk actin assays, and EM to investigate how ExoY impacts actin dynamics. We found that ExoY can directly bundle actin filaments with high affinity, comparable with eukaryotic F-actin-bundling proteins, such as fimbrin. Of note, ExoY enzymatic activity was not required for F-actin bundling. Bundling is known to require multiple actin-binding sites, yet small-angle X-ray scattering experiments revealed that ExoY is a monomer in solution, and previous data suggested that ExoY possesses only one actin-binding site. We therefore hypothesized that ExoY oligomerizes in response to F-actin binding and have used the ExoY structure to construct a dimer-based structural model for the ExoY-F-actin complex. Subsequent mutational analyses suggested that the ExoY oligomerization interface plays a crucial role in mediating F-actin bundling. Our results indicate that ExoY represents a new class of actin-binding proteins that modulate the actin cytoskeleton both directly, via F-actin bundling, and indirectly, via actin-activated nucleotidyl cyclase activity.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Proteínas Bacterianas/metabolismo , Glucosiltransferasas/metabolismo , Pseudomonas aeruginosa/enzimología , Citoesqueleto de Actina/ultraestructura , Factores Despolimerizantes de la Actina/metabolismo , Actinas/química , Actinas/metabolismo , Actinas/ultraestructura , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/ultraestructura , Glucosiltransferasas/química , Glucosiltransferasas/genética , Glucosiltransferasas/ultraestructura , Glicoproteínas de Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Modelos Moleculares , Mutación/genética , Unión Proteica , Multimerización de Proteína
5.
Eur J Med Chem ; 184: 111746, 2019 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-31610373

RESUMEN

Presequence protease (PreP) is a proteostatic enzyme that plays a key role in the maintenance of mitochondrial health. Defects in PreP stability are associated with neurological disorders in humans, and altered activity of this enzyme modulates the progress of Alzheimer's disease-like pathology in mice. As agonists that boost PreP proteolytic activity represent a promising therapeutic avenue, we sought to determine the structural basis for the action of benzimidazole derivatives (3c and 4c), first reported by Vangavaragu et al. (Eur. J. Med. Chem. 76 (2014) 506-516) that enhance the activity of PreP. However, we found the published procedure for the synthesis of 3c yielded aldimine A instead. We then developed an alternative synthesis and obtained 3c, termed compound C, and an alternative benzimidazole derivative, termed compound B. We tested compounds A, B and C for their ability to enhance the activities of human PreP. In contrast to the previous report, we observed that none of the compounds A, B, or C (3c) modulated the catalytic activity of human PreP. Here we report our findings on the mis-identification of the reported benzimidazoles and the lack of biological activity of such compounds on human PreP. Thus, PreP modulators for PreP-based therapies remain to be discovered.


Asunto(s)
Bencimidazoles/farmacología , Proteínas Mitocondriales/metabolismo , Serina Endopeptidasas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Bencimidazoles/síntesis química , Bencimidazoles/química , Relación Dosis-Respuesta a Droga , Humanos , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/aislamiento & purificación , Estructura Molecular , Serina Endopeptidasas/genética , Serina Endopeptidasas/aislamiento & purificación , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad
6.
Eur J Med Chem ; 179: 557-566, 2019 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-31276900

RESUMEN

Insulin-degrading enzyme, IDE, is a metalloprotease implicated in the metabolism of key peptides such as insulin, glucagon, ß-amyloid peptide. Recent studies have pointed out its broader role in the cell physiology. In order to identify new drug-like inhibitors of IDE with optimal pharmacokinetic properties to probe its multiple roles, we ran a high-throughput drug repurposing screening. Ebselen, cefmetazole and rabeprazole were identified as reversible inhibitors of IDE. Ebselen is the most potent inhibitor (IC50(insulin) = 14 nM). The molecular mode of action of ebselen was investigated by biophysical methods. We show that ebselen induces the disorder of the IDE catalytic cleft, which significantly differs from the previously reported IDE inhibitors. IDE inhibition by ebselen can explain some of its reported activities in metabolism as well as in neuroprotection.


Asunto(s)
Azoles/farmacología , Reposicionamiento de Medicamentos , Inhibidores Enzimáticos/farmacología , Insulisina/antagonistas & inhibidores , Compuestos de Organoselenio/farmacología , Azoles/química , Biocatálisis , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/química , Ensayos Analíticos de Alto Rendimiento , Humanos , Insulisina/metabolismo , Isoindoles , Estructura Molecular , Compuestos de Organoselenio/química , Relación Estructura-Actividad
7.
Biochemistry ; 58(27): 2996-3004, 2019 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-31243996

RESUMEN

Anthrax, a lethal, weaponizable disease caused by Bacillus anthracis, acts through exotoxins that are primary mediators of systemic toxicity and also targets for neutralization by passive immunotherapy. The ease of engineering B. anthracis strains resistant to established therapy and the historic use of the microbe in bioterrorism present a compelling test case for platforms that permit the rapid and modular development of neutralizing agents. In vitro antigen-binding fragment (Fab) selection offers the advantages of speed, sequence level molecular control, and engineering flexibility compared to traditional monoclonal antibody pipelines. By screening an unbiased, chemically synthetic phage Fab library and characterizing hits in cell-based assays, we identified two high-affinity neutralizing Fabs, A4 and B7, against anthrax edema factor (EF), a key mediator of anthrax pathogenesis. Engineered homodimers of these Fabs exhibited potency comparable to that of the best reported neutralizing monoclonal antibody against EF at preventing EF-induced cyclic AMP production. Using internalization assays in COS cells, B7 was found to block steps prior to EF internalization. This work demonstrates the efficacy of synthetic alternatives to traditional antibody therapeutics against anthrax while also demonstrating a broadly generalizable, rapid, and modular screening pipeline for neutralizing antibody generation.


Asunto(s)
Carbunco/tratamiento farmacológico , Anticuerpos Neutralizantes/farmacología , Bacillus anthracis/efectos de los fármacos , Toxinas Bacterianas/antagonistas & inhibidores , Fragmentos Fab de Inmunoglobulinas/farmacología , Secuencia de Aminoácidos , Animales , Carbunco/metabolismo , Carbunco/microbiología , Anticuerpos Neutralizantes/química , Antígenos Bacterianos/metabolismo , Bacillus anthracis/fisiología , Toxinas Bacterianas/metabolismo , Células CHO , Células COS , Línea Celular , Chlorocebus aethiops , Cricetulus , AMP Cíclico/metabolismo , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Ratones , Multimerización de Proteína
8.
J Chem Inf Model ; 58(9): 1926-1934, 2018 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-30133282

RESUMEN

Insulin degrading enzyme (IDE), a metalloprotease that degrades amyloid-ß (Aß) peptides and insulin, is associated with Alzheimer's disease and diabetes. The mechanism of IDE catalyzed degrading of Aß peptides, which is of fundamental importance in the design of therapeutic methods for Alzheimer's disease, has not been fully understood. In this work, combined quantum mechanics and molecular mechanics (QM/MM) style Møller-Plesset second order perturbation theory (MP2) geometry optimization calculations are performed to investigate the catalytic mechanism of the Aß40 Phe19-Phe20 peptide bond cleavage by human IDE. The analyses using QM/MM MP2 optimization suggest that a neutral water molecule is at the active site of the enzyme-substrate (ES) complex. The water molecule is in hydrogen bonding with the nearby anionic Glu111 of IDE but not directly bound to the catalytic Zn ion. This is confirmed by QM/MM DFTB3 molecular dynamics simulation. Our studies also reveal that the hydrolysis of the Aß40 Phe19-Phe20 peptide bond by IDE consists of four key steps. The neutral water is first activated by moving toward and binding to the Zn ion. A gem-diol intermediate is then formed by the activated neutral water molecule attacking the C atom of the Phe19-Phe20 peptide bond. The next is the protonation of the N atom of Phe19-Phe20 peptide bond to form an intermediate with an elongated C-N bond. The final step is the breaking of the Phe19-Phe20 C-N bond. The final step is the rate-determining step with a calculated Gibbs free energy of activation of 17.34 kcal/mol, in good agreement with the experimental value 16.7 kcal/mol. This mechanism provides the basis for the design of biochemical methods to modulate the activity of IDE in humans.


Asunto(s)
Péptidos beta-Amiloides/química , Insulisina/metabolismo , Teoría Cuántica , Catálisis , Cinética , Modelos Moleculares , Unión Proteica , Conformación Proteica , Proteolisis , Programas Informáticos , Agua , Zinc
9.
Elife ; 72018 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-29596046

RESUMEN

Insulin degrading enzyme (IDE) plays key roles in degrading peptides vital in type two diabetes, Alzheimer's, inflammation, and other human diseases. However, the process through which IDE recognizes peptides that tend to form amyloid fibrils remained unsolved. We used cryoEM to understand both the apo- and insulin-bound dimeric IDE states, revealing that IDE displays a large opening between the homologous ~55 kDa N- and C-terminal halves to allow selective substrate capture based on size and charge complementarity. We also used cryoEM, X-ray crystallography, SAXS, and HDX-MS to elucidate the molecular basis of how amyloidogenic peptides stabilize the disordered IDE catalytic cleft, thereby inducing selective degradation by substrate-assisted catalysis. Furthermore, our insulin-bound IDE structures explain how IDE processively degrades insulin by stochastically cutting either chain without breaking disulfide bonds. Together, our studies provide a mechanism for how IDE selectively degrades amyloidogenic peptides and offers structural insights for developing IDE-based therapies.


Asunto(s)
Insulina/química , Insulina/metabolismo , Insulisina/química , Insulisina/metabolismo , Microscopía por Crioelectrón , Cristalografía por Rayos X , Humanos , Espectrometría de Masas , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Proteolisis , Dispersión del Ángulo Pequeño
10.
J Mol Biol ; 430(3): 337-347, 2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29273204

RESUMEN

Antibody Fab fragments have been exploited with significant success to facilitate the structure determination of challenging macromolecules as crystallization chaperones and as molecular fiducial marks for single particle cryo-electron microscopy approaches. However, the inherent flexibility of the "elbow" regions, which link the constant and variable domains of the Fab, can introduce disorder and thus diminish their effectiveness. We have developed a phage display engineering strategy to generate synthetic Fab variants that significantly reduces elbow flexibility, while maintaining their high affinity and stability. This strategy was validated using previously recalcitrant Fab-antigen complexes where introduction of an engineered elbow region enhanced crystallization and diffraction resolution. Furthermore, incorporation of the mutations appears to be generally portable to other synthetic antibodies and may serve as a universal strategy to enhance the success rates of Fabs as structure determination chaperones.


Asunto(s)
Antígenos/química , Microscopía por Crioelectrón/métodos , Fragmentos Fab de Inmunoglobulinas/química , Antígenos/ultraestructura , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/ultraestructura , Cristalización/métodos , Humanos , Fragmentos Fab de Inmunoglobulinas/genética , Fragmentos Fab de Inmunoglobulinas/ultraestructura , Modelos Moleculares , Chaperonas Moleculares/química , Chaperonas Moleculares/ultraestructura , Biblioteca de Péptidos , Conformación Proteica , Ingeniería de Proteínas , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/ultraestructura , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/ultraestructura
12.
Nat Commun ; 8: 15375, 2017 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-28541346

RESUMEN

In the intestinal epithelium, the aberrant regulation of cell/cell junctions leads to intestinal barrier defects, which may promote the onset and enhance the severity of inflammatory bowel disease (IBD). However, it remains unclear how the coordinated behaviour of cytoskeletal network may contribute to cell junctional dynamics. In this report, we identified ACF7, a crosslinker of microtubules and F-actin, as an essential player in this process. Loss of ACF7 leads to aberrant microtubule organization, tight junction stabilization and impaired wound closure in vitro. With the mouse genetics approach, we show that ablation of ACF7 inhibits intestinal wound healing and greatly increases susceptibility to experimental colitis in mice. ACF7 level is also correlated with development and progression of ulcerative colitis (UC) in human patients. Together, our results reveal an important molecular mechanism whereby coordinated cytoskeletal dynamics contributes to cell adhesion regulation during intestinal wound repair and the development of IBD.


Asunto(s)
Colitis/etiología , Proteínas de Microfilamentos/fisiología , Animales , Células CACO-2 , Adhesión Celular/fisiología , Colitis/patología , Colitis/fisiopatología , Colitis Ulcerosa/etiología , Colitis Ulcerosa/patología , Colitis Ulcerosa/fisiopatología , Cristalografía por Rayos X , Modelos Animales de Enfermedad , Femenino , Humanos , Enfermedades Inflamatorias del Intestino/etiología , Enfermedades Inflamatorias del Intestino/patología , Enfermedades Inflamatorias del Intestino/fisiopatología , Mucosa Intestinal/patología , Mucosa Intestinal/fisiopatología , Masculino , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/deficiencia , Proteínas de Microfilamentos/genética , Microtúbulos/fisiología , Modelos Moleculares , Uniones Estrechas/patología , Uniones Estrechas/fisiología , Cicatrización de Heridas/fisiología
13.
Oncotarget ; 7(42): 68833-68841, 2016 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-27626698

RESUMEN

Ibrutinib (ibr), a first-in-class Bruton tyrosine kinase (BTK) inhibitor, has demonstrated high response rates in both relapsed/refractory and treatment naïve chronic lymphocytic leukemia (CLL). However, about 25% of patients discontinue ibrutinib therapy at a median follow-up of 20 months and many patients discontinue the treatment due to leukemia progression or Richter transformation. Mutations affecting the C481 residue of BTK disrupt ibrutinib binding and have been characterized by us and others as the most common mechanism of ibrutinib resistance. Thus far, all described BTK mutations are located in its kinase domain and mutations outside this domain have never been described. Herein, we report a patient whose CLL progressed, was salvaged with ibrutinib and then relapsed. Serial analysis of samples throughout patient's clinical course identified a structurally novel mutation (BTKT316A) in the SH2 domain, but not kinase domain, of Bruton tyrosine kinase which was associated with disease relapse. Functionally, cells carrying BTKT316A show resistance to ibrutinib at both cellular and molecular levels to a similar extent as BTKC481S. Our study lends further insight into the diverse mechanisms of ibrutinib resistance that has important implications for the development of next-generation BTK inhibitors as well as mutation detection in relapsed patients.


Asunto(s)
Resistencia a Antineoplásicos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Proteínas Tirosina Quinasas/genética , Pirazoles/uso terapéutico , Pirimidinas/uso terapéutico , Adenina/análogos & derivados , Agammaglobulinemia Tirosina Quinasa , Antineoplásicos/uso terapéutico , Transformación Celular Neoplásica , Análisis Mutacional de ADN , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Leucemia Linfocítica Crónica de Células B/genética , Persona de Mediana Edad , Mutación , Recurrencia Local de Neoplasia , Piperidinas , Inhibidores de Proteínas Quinasas/uso terapéutico , Dominios Homologos src
14.
Nat Commun ; 7: 11692, 2016 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-27216888

RESUMEN

Turnover of focal adhesions allows cell retraction, which is essential for cell migration. The mammalian spectraplakin protein, ACF7 (Actin-Crosslinking Factor 7), promotes focal adhesion dynamics by targeting of microtubule plus ends towards focal adhesions. However, it remains unclear how the activity of ACF7 is regulated spatiotemporally to achieve focal adhesion-specific guidance of microtubule. To explore the potential mechanisms, we resolve the crystal structure of ACF7's NT (amino-terminal) domain, which mediates F-actin interactions. Structural analysis leads to identification of a key tyrosine residue at the calponin homology (CH) domain of ACF7, whose phosphorylation by Src/FAK (focal adhesion kinase) complex is essential for F-actin binding of ACF7. Using skin epidermis as a model system, we further demonstrate that the phosphorylation of ACF7 plays an indispensable role in focal adhesion dynamics and epidermal migration in vitro and in vivo. Together, our findings provide critical insights into the molecular mechanisms underlying coordinated cytoskeletal dynamics during cell movement.


Asunto(s)
Movimiento Celular/fisiología , Epidermis/fisiología , Adhesiones Focales/metabolismo , Proteínas de Microfilamentos/fisiología , Actinas/metabolismo , Animales , Técnicas de Cultivo de Célula/métodos , Cristalografía por Rayos X , Células Epidérmicas , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Células HEK293 , Humanos , Queratinocitos , Ratones , Ratones Desnudos , Proteínas de Microfilamentos/química , Microtúbulos/metabolismo , Modelos Animales , Fosforilación , Cultivo Primario de Células , Unión Proteica , Dominios Proteicos , Imagen de Lapso de Tiempo , Tirosina/metabolismo , Cicatrización de Heridas/fisiología , Familia-src Quinasas/metabolismo
15.
Proc Natl Acad Sci U S A ; 113(18): 5000-5, 2016 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-27091995

RESUMEN

CC chemokine ligand 5 (CCL5) and CCL3 are critical for immune surveillance and inflammation. Consequently, they are linked to the pathogenesis of many inflammatory conditions and are therapeutic targets. Oligomerization and glycosaminoglycan (GAG) binding of CCL5 and CCL3 are vital for the functions of these chemokines. Our structural and biophysical analyses of human CCL5 reveal that CCL5 oligomerization is a polymerization process in which CCL5 forms rod-shaped, double-helical oligomers. This CCL5 structure explains mutational data and offers a unified mechanism for CCL3, CCL4, and CCL5 assembly into high-molecular-weight, polydisperse oligomers. A conserved, positively charged BBXB motif is key for the binding of CC chemokines to GAG. However, this motif is partially buried when CCL3, CCL4, and CCL5 are oligomerized; thus, the mechanism by which GAG binds these chemokine oligomers has been elusive. Our structures of GAG-bound CCL5 and CCL3 oligomers reveal that these chemokine oligomers have distinct GAG-binding mechanisms. The CCL5 oligomer uses another positively charged and fully exposed motif, KKWVR, in GAG binding. However, residues from two partially buried BBXB motifs along with other residues combine to form a GAG-binding groove in the CCL3 oligomer. The N termini of CC chemokines are shown to be involved in receptor binding and oligomerization. We also report an alternative CCL3 oligomer structure that reveals how conformational changes in CCL3 N termini profoundly alter its surface properties and dimer-dimer interactions to affect GAG binding and oligomerization. Such complexity in oligomerization and GAG binding enables intricate, physiologically relevant regulation of CC chemokine functions.


Asunto(s)
Quimiocina CCL3/química , Quimiocina CCL3/ultraestructura , Quimiocina CCL5/química , Quimiocina CCL5/ultraestructura , Glicosaminoglicanos/química , Sitios de Unión , Dimerización , Humanos , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad
16.
Trends Endocrinol Metab ; 27(1): 24-34, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26651592

RESUMEN

Insulin-degrading enzyme (IDE) selectively degrades peptides, such as insulin, amylin, and amyloid ß (Aß) that form toxic aggregates, to maintain proteostasis. IDE defects are linked to the development of type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD). Structural and biochemical analyses revealed the molecular basis for IDE-mediated destruction of amyloidogenic peptides and this information has been exploited to develop promising inhibitors of IDE to improve glucose homeostasis. However, the inhibition of IDE can also lead to glucose intolerance. In this review, I focus on recent advances regarding our understanding of the structure and function of IDE and the discovery of IDE inhibitors, as well as challenges in developing IDE-based therapy for human diseases, particularly T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2/enzimología , Diabetes Mellitus Tipo 2/metabolismo , Insulina/metabolismo , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Diabetes Mellitus Tipo 2/genética , Intolerancia a la Glucosa , Humanos , Insulisina/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo
17.
Nat Commun ; 6: 8250, 2015 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-26394692

RESUMEN

Insulin-degrading enzyme (IDE) is a protease that cleaves insulin and other bioactive peptides such as amyloid-ß. Knockout and genetic studies have linked IDE to Alzheimer's disease and type-2 diabetes. As the major insulin-degrading protease, IDE is a candidate drug target in diabetes. Here we have used kinetic target-guided synthesis to design the first catalytic site inhibitor of IDE suitable for in vivo studies (BDM44768). Crystallographic and small angle X-ray scattering analyses show that it locks IDE in a closed conformation. Among a panel of metalloproteases, BDM44768 selectively inhibits IDE. Acute treatment of mice with BDM44768 increases insulin signalling and surprisingly impairs glucose tolerance in an IDE-dependent manner. These results confirm that IDE is involved in pathways that modulate short-term glucose homeostasis, but casts doubt on the general usefulness of the inhibition of IDE catalytic activity to treat diabetes.


Asunto(s)
Ácidos Hidroxámicos/síntesis química , Insulisina/antagonistas & inhibidores , Triazoles/síntesis química , Animales , Células CACO-2 , Dominio Catalítico , Diabetes Mellitus/tratamiento farmacológico , Evaluación Preclínica de Medicamentos , Prueba de Tolerancia a la Glucosa , Humanos , Ácidos Hidroxámicos/farmacología , Ácidos Hidroxámicos/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Microsomas Hepáticos , Terapia Molecular Dirigida , Distribución Aleatoria , Relación Estructura-Actividad , Triazoles/farmacología , Triazoles/uso terapéutico
18.
Toxins (Basel) ; 7(7): 2598-614, 2015 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-26184312

RESUMEN

Bacillus anthracis adenylyl cyclase toxin edema factor (EF) is one component of the anthrax toxin and is essential for establishing anthrax disease. EF activation by the eukaryotic Ca2+-sensor calmodulin (CaM) leads to massive cAMP production resulting in edema. cAMP also inhibits the nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase, thus reducing production of reactive oxygen species (ROS) used for host defense in activated neutrophils and thereby facilitating bacterial growth. Methionine (Met) residues in CaM, important for interactions between CaM and its binding partners, can be oxidized by ROS. We investigated the impact of site-specific oxidation of Met in CaM on EF activation using thirteen CaM-mutants (CaM-mut) with Met to leucine (Leu) substitutions. EF activation shows high resistance to oxidative modifications in CaM. An intact structure in the C-terminal region of oxidized CaM is sufficient for major EF activation despite altered secondary structure in the N-terminal region associated with Met oxidation. The secondary structures of CaM-mut were determined and described in previous studies from our group. Thus, excess cAMP production and the associated impairment of host defence may be afforded even under oxidative conditions in activated neutrophils.


Asunto(s)
Adenilil Ciclasas/metabolismo , Antígenos Bacterianos/metabolismo , Bacillus anthracis/metabolismo , Toxinas Bacterianas/metabolismo , Calmodulina/metabolismo , Adenilil Ciclasas/genética , Sustitución de Aminoácidos , Antígenos Bacterianos/genética , Toxinas Bacterianas/genética , Calmodulina/química , Calmodulina/genética , AMP Cíclico/metabolismo , Escherichia coli/genética , Leucina/química , Leucina/genética , Leucina/metabolismo , Metionina/química , Metionina/genética , Metionina/metabolismo , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Oxidación-Reducción , Estructura Terciaria de Proteína
19.
J Mol Biol ; 427(6 Pt B): 1345-1358, 2015 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-25636406

RESUMEN

CC chemokine ligands (CCLs) are 8- to 14-kDa signaling proteins involved in diverse immune functions. While CCLs share similar tertiary structures, oligomerization produces highly diverse quaternary structures that protect chemokines from proteolytic degradation and modulate their functions. CCL18 is closely related to CCL3 and CCL4 with respect to both protein sequence and genomic location, yet CCL18 has distinct biochemical and biophysical properties. Here, we report a crystal structure of human CCL18 and its oligomerization states in solution based on crystallographic and small-angle X-ray scattering analyses. Our data show that CCL18 adopts an α-helical conformation at its N-terminus that weakens its dimerization, explaining CCL18's preference for the monomeric state. Multiple contacts between monomers allow CCL18 to reversibly form a unique open-ended oligomer different from those of CCL3, CCL4, and CCL5. Furthermore, these differences hinge on proline 8, which is conserved in CCL3 and CCL4 but is replaced by lysine in human CCL18. Our structural analyses suggest that a mutation of proline 8 to alanine stabilizes a type 1 ß-turn at the N-terminus of CCL4 to prevent dimerization but prevents dimers from making key contacts with each other in CCL3. Thus, the P8A mutation induces depolymerization of CCL3 and CCL4 by distinct mechanisms. Finally, we used structural, biochemical, and functional analyses to unravel why insulin-degrading enzyme degrades CCL3 and CCL4 but not CCL18. Our results elucidate the molecular basis for the oligomerization of three closely related CC chemokines and suggest how oligomerization shapes CCL chemokine function.


Asunto(s)
Quimiocina CCL3/química , Quimiocina CCL4/química , Quimiocinas CC/metabolismo , Insulisina/metabolismo , Secuencia de Aminoácidos , Quimiocina CCL3/genética , Quimiocina CCL3/metabolismo , Quimiocina CCL4/genética , Quimiocina CCL4/metabolismo , Quimiocinas CC/química , Quimiocinas CC/genética , Quimiotaxis , Cristalografía por Rayos X , Humanos , Insulisina/química , Insulisina/genética , Datos de Secuencia Molecular , Mutación/genética , Estructura Cuaternaria de Proteína , Dispersión del Ángulo Pequeño , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
20.
Eur J Med Chem ; 90: 547-67, 2015 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-25489670

RESUMEN

Insulin degrading enzyme (IDE) is a zinc metalloprotease that degrades small amyloid peptides such as amyloid-â and insulin. So far the dearth of IDE-specific pharmacological inhibitors impacts the understanding of its role in the physiopathology of Alzheimer's disease, amyloid-â clearance, and its validation as a potential therapeutic target. Hit 1 was previously discovered by high-throughput screening. Here we describe the structure-activity study, that required the synthesis of 48 analogues. We found that while the carboxylic acid, the imidazole and the tertiary amine were critical for activity, the methyl ester was successfully optimized to an amide or a 1,2,4-oxadiazole. Along with improving their activity, compounds were optimized for solubility, lipophilicity and stability in plasma and microsomes. The docking or co-crystallization of some compounds at the exosite or the catalytic site of IDE provided the structural basis for IDE inhibition. The pharmacokinetic properties of best compounds 44 and 46 were measured in vivo. As a result, 44 (BDM43079) and its methyl ester precursor 48 (BDM43124) are useful chemical probes for the exploration of IDE's role.


Asunto(s)
Carbamatos/farmacología , Ácidos Carboxílicos/química , Inhibidores Enzimáticos/farmacología , Imidazoles/química , Insulisina/antagonistas & inhibidores , Insulisina/metabolismo , Oxadiazoles/farmacología , Carbamatos/síntesis química , Carbamatos/química , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Modelos Moleculares , Estructura Molecular , Oxadiazoles/síntesis química , Oxadiazoles/química , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...