Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Anal Chem ; 95(37): 13829-13837, 2023 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-37642957

RESUMEN

Synthetic cannabinoids (SCs) make up a class of novel psychoactive substances (NPS), used predominantly in prisons and homeless communities in the U.K. SCs can have severe side effects, including psychosis, stroke, and seizures, with numerous reported deaths associated with their use. The chemical diversity of SCs presents the major challenge to their detection since approaches relying on specific molecular recognition become outdated almost immediately. Ideally one would have a generic approach to detecting SCs in portable settings. The problem of SC detection is more challenging still because the majority of SCs enter the prison estate adsorbed onto physical matrices such as paper, fabric, or herb materials. That is, regardless of the detection modality used, the necessary extraction step reduces the effectiveness and ability to rapidly screen materials on-site. Herein, we demonstrate a truly instant generic test for SCs, tested against real-world drug seizures. The test is based on two advances. First, we identify a spectrally silent region in the emission spectrum of most physical matrices. Second, the finding that background signals (including from autofluorescence) can be accurately predicted is based on tracking the fraction of absorbed light from the irradiation source. Finally, we demonstrate that the intrinsic fluorescence of a large range of physical substrates can be leveraged to track the presence of other drugs of interest, including the most recent iterations of benzodiazepines and opioids. We demonstrate the implementation of our presumptive test in a portable, pocket-sized device that will find immediate utility in prisons and law enforcement agencies around the world.


Asunto(s)
Analgésicos Opioides , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Humanos , Benzodiazepinas , Fluorescencia , Convulsiones
2.
Anal Chem ; 95(2): 703-713, 2023 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-36599091

RESUMEN

With synthetic cannabinoid receptor agonist (SCRA) use still prevalent across Europe and structurally advanced generations emerging, it is imperative that drug detection methods advance in parallel. SCRAs are a chemically diverse and evolving group, which makes rapid detection challenging. We have previously shown that fluorescence spectral fingerprinting (FSF) has the potential to provide rapid assessment of SCRA presence directly from street material with minimal processing and in saliva. Enhancing the sensitivity and discriminatory ability of this approach has high potential to accelerate the delivery of a point-of-care technology that can be used confidently by a range of stakeholders, from medical to prison staff. We demonstrate that a range of structurally distinct SCRAs are photochemically active and give rise to distinct FSFs after irradiation. To explore this in detail, we have synthesized a model series of compounds which mimic specific structural features of AM-694. Our data show that FSFs are sensitive to chemically conservative changes, with evidence that this relates to shifts in the electronic structure and cross-conjugation. Crucially, we find that the photochemical degradation rate is sensitive to individual structures and gives rise to a specific major product, the mechanism and identification of which we elucidate through density-functional theory (DFT) and time-dependent DFT. We test the potential of our hybrid "photochemical fingerprinting" approach to discriminate SCRAs by demonstrating SCRA detection from a simulated smoking apparatus in saliva. Our study shows the potential of tracking photochemical reactivity via FSFs for enhanced discrimination of SCRAs, with successful integration into a portable device.


Asunto(s)
Agonistas de Receptores de Cannabinoides , Drogas Ilícitas , Humanos , Agonistas de Receptores de Cannabinoides/química , Sistemas de Atención de Punto , Detección de Abuso de Sustancias/métodos
3.
Biochem J ; 477(7): 1241-1259, 2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-32195541

RESUMEN

Angiotensin-converting enzyme (ACE) is best known for its formation of the vasopressor angiotensin II that controls blood pressure but is also involved in other physiological functions through the hydrolysis of a variety of peptide substrates. The enzyme contains two catalytic domains (nACE and cACE) that have different affinities for ACE substrates and inhibitors. We investigated whether nACE inhibitor backbones contain a unique property which allows them to take advantage of the hinging of nACE. Kinetic analysis showed that mutation of unique nACE residues, in both the S2 pocket and around the prime subsites (S') to their C-domain counterparts, each resulted in a decrease in the affinity of nACE specific inhibitors (SG6, 33RE and ketoACE-13) but it required the combined S2_S' mutant to abrogate nACE-selectivity. However, this was not observed with the non-domain-selective inhibitors enalaprilat and omapatrilat. High-resolution structures were determined for the minimally glycosylated nACE with the combined S2_S' mutations in complex with the ACE inhibitors 33RE (1.8 Å), omapatrilat (1.8 Å) and SG6 (1.7 Å). These confirmed that the affinities of the nACE-selective SG6, 33RE and ketoACE-13 are not only affected by direct interactions with the immediate environment of the binding site, but also by more distal residues. This study provides evidence for a more general mechanism of ACE inhibition involving synergistic effects of not only the S2, S1' and S2' subsites, but also residues involved in the sub-domain interface that effect the unique ways in which the two domains stabilize active site loops to favour inhibitor binding.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/química , Inhibidores de la Enzima Convertidora de Angiotensina/metabolismo , Dominio Catalítico , Metaloendopeptidasas/química , Metaloendopeptidasas/metabolismo , Peptidil-Dipeptidasa A/química , Peptidil-Dipeptidasa A/metabolismo , Presión Sanguínea/fisiología , Cristalografía por Rayos X , Glicosilación , Humanos , Cinética , Ligandos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación , Peptidil-Dipeptidasa A/genética , Unión Proteica , Conformación Proteica en Lámina beta/genética , Sistema Renina-Angiotensina/fisiología
4.
Clin Sci (Lond) ; 134(21): 2851-2871, 2020 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-33146371

RESUMEN

Angiotensin converting enzyme (ACE) is well-known for its role in blood pressure regulation via the renin-angiotensin aldosterone system (RAAS) but also functions in fertility, immunity, haematopoiesis and diseases such as obesity, fibrosis and Alzheimer's dementia. Like ACE, the human homologue ACE2 is also involved in blood pressure regulation and cleaves a range of substrates involved in different physiological processes. Importantly, it is the functional receptor for severe acute respiratory syndrome (SARS)-coronavirus (CoV)-2 responsible for the 2020, coronavirus infectious disease 2019 (COVID-19) pandemic. Understanding the interaction between SARS-CoV-2 and ACE2 is crucial for the design of therapies to combat this disease. This review provides a comparative analysis of methodologies and findings to describe how structural biology techniques like X-ray crystallography and cryo-electron microscopy have enabled remarkable discoveries into the structure-function relationship of ACE and ACE2. This, in turn, has enabled the development of ACE inhibitors for the treatment of cardiovascular disease and candidate therapies for the treatment of COVID-19. However, despite these advances the function of ACE homologues in non-human organisms is not yet fully understood. ACE homologues have been discovered in the tissues, body fluids and venom of species from diverse lineages and are known to have important functions in fertility, envenoming and insect-host defence mechanisms. We, therefore, further highlight the need for structural insight into insect and venom ACE homologues for the potential development of novel anti-venoms and insecticides.


Asunto(s)
Betacoronavirus/patogenicidad , Infecciones por Coronavirus/enzimología , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/enzimología , Receptores Virales/metabolismo , Internalización del Virus , Enzima Convertidora de Angiotensina 2 , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Animales , Antivirales/uso terapéutico , Betacoronavirus/efectos de los fármacos , COVID-19 , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/virología , Interacciones Huésped-Patógeno , Humanos , Pandemias , Peptidil-Dipeptidasa A/química , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/virología , Conformación Proteica , Receptores Virales/química , SARS-CoV-2 , Relación Estructura-Actividad , Tratamiento Farmacológico de COVID-19
5.
Biochem J ; 476(22): 3505-3520, 2019 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-31682720

RESUMEN

The mosquitoes of the Anopheles and Aedes genus are some of the most deadly insects to humans because of their effectiveness as vectors of malaria and a range of arboviruses, including yellow fever, dengue, chikungunya, West Nile and Zika. The use of insecticides from different chemical classes is a key component of the integrated strategy against An. gambiae and Ae. aegypti, but the problem of insecticide resistance means that new compounds with different modes of action are urgently needed to replace chemicals that fail to control resistant mosquito populations. We have previously shown that feeding inhibitors of peptidyl dipeptidase A to both An. gambiae and Ae. aegypti mosquito larvae lead to stunted growth and mortality. However, these compounds were designed to inhibit the mammalian form of the enzyme (angiotensin-converting enzyme, ACE) and hence can have lower potency and lack selectivity as inhibitors of the insect peptidase. Thus, for the development of inhibitors of practical value in killing mosquito larvae, it is important to design new compounds that are both potent and highly selective. Here, we report the first structures of AnoACE2 from An. gambiae in its native form and with a bound human ACE inhibitor fosinoprilat. A comparison of these structures with human ACE (sACE) and an insect ACE homologue from Drosophila melanogaster (AnCE) revealed that the AnoACE2 structure is more similar to AnCE. In addition, important elements that differ in these structures provide information that could potentially be utilised in the design of chemical leads for selective mosquitocide development.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/química , Anopheles/enzimología , Proteínas de Insectos/química , Peptidil-Dipeptidasa A/química , Aedes/química , Aedes/enzimología , Aedes/genética , Animales , Anopheles/química , Anopheles/genética , Anopheles/crecimiento & desarrollo , Drosophila melanogaster/química , Drosophila melanogaster/enzimología , Fosinopril/análogos & derivados , Fosinopril/química , Humanos , Proteínas de Insectos/antagonistas & inhibidores , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Insecticidas/química , Larva/química , Larva/enzimología , Larva/genética , Larva/crecimiento & desarrollo , Modelos Moleculares , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo
6.
Biochem J ; 476(10): 1553-1570, 2019 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-31072910

RESUMEN

Angiotensin-converting enzyme (ACE) is a zinc metalloprotease best known for its role in blood pressure regulation. ACE consists of two homologous catalytic domains, the N- and C-domain, that display distinct but overlapping catalytic functions in vivo owing to subtle differences in substrate specificity. While current generation ACE inhibitors target both ACE domains, domain-selective ACE inhibitors may be clinically advantageous, either reducing side effects or having utility in new indications. Here, we used site-directed mutagenesis, an ACE chimera and X-ray crystallography to unveil the molecular basis for C-domain-selective ACE inhibition by the bradykinin-potentiating peptide b (BPPb), naturally present in Brazilian pit viper venom. We present the BPPb N-domain structure in comparison with the previously reported BPPb C-domain structure and highlight key differences in peptide interactions with the S4 to S9 subsites. This suggests the involvement of these subsites in conferring C-domain-selective BPPb binding, in agreement with the mutagenesis results where unique residues governing differences in active site exposure, lid structure and dynamics between the two domains were the major drivers for C-domain-selective BPPb binding. Mere disruption of BPPb interactions with unique S2 and S4 subsite residues, which synergistically assist in BPPb binding, was insufficient to abolish C-domain selectivity. The combination of unique S9-S4 and S2' subsite C-domain residues was required for the favourable entry, orientation and thus, selective binding of the peptide. This emphasizes the need to consider factors other than direct protein-inhibitor interactions to guide the design of domain-selective ACE inhibitors, especially in the case of larger peptides.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/química , Oligopéptidos/química , Peptidil-Dipeptidasa A/química , Animales , Células CHO , Catálisis , Cricetulus , Cristalografía por Rayos X , Humanos , Mutagénesis Sitio-Dirigida , Peptidil-Dipeptidasa A/genética , Dominios Proteicos
7.
J Biol Chem ; 293(39): 15330-15331, 2018 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-30266880

RESUMEN

Bacterial pathogens use several strategies to infect host cells, one of which involves blocking host defenses. During infection, the bacterial effector proteins GtgA, GogA, PipA, and NleC are injected into host cells by the type III secretion system (T3SS), where they suppress the proinflammatory NF-κB signaling pathway to dampen immune responses. The authors demonstrate that these effectors bind NF-κB via their DNA-mimicking regions and uncover differences in effector sequences and structures explaining the individual specificities of these effectors for distinct NF-κB subunits.


Asunto(s)
Proteínas de Escherichia coli , FN-kappa B , Proteínas Bacterianas , ADN , Metaloproteasas , Zinc
8.
Mol Pharmacol ; 93(4): 344-354, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29371233

RESUMEN

Angiotensin-converting enzyme (ACE) plays a central role in the renin-angiotensin system (RAS), which is primarily responsible for blood pressure homeostasis. Studies have shown that ACE inhibitors yield cardiovascular benefits that cannot be entirely attributed to the inhibition of ACE catalytic activity. It is possible that these benefits are due to interactions between ACE and RAS receptors that mediate the protective arm of the RAS, such as angiotensin II receptor type 2 (AT2R) and the receptor MAS. Therefore, in this study, we investigated the molecular interactions of ACE, including ACE homodimerization and heterodimerization with AT2R and MAS, respectively. Molecular interactions were assessed by fluorescence resonance energy transfer and bimolecular fluorescence complementation in human embryonic kidney 293 cells and Chinese hamster ovary-K1 cells transfected with vectors encoding fluorophore-tagged proteins. The specificity of dimerization was verified by competition experiments using untagged proteins. These techniques were used to study several potential requirements for the germinal isoform of angiotensin-converting enzyme expressed in the testes (tACE) dimerization as well as the effect of ACE inhibitors on both somatic isoforms of angiotensin-converting enzyme expressed in the testes (sACE) and tACE dimerization. We demonstrated constitutive homodimerization of sACE and of both of its domains separately, as well as heterodimerization of both sACE and tACE with AT2R, but not MAS. In addition, we investigated both soluble sACE and the sACE N domain using size-exclusion chromatography-coupled small-angle X-ray scattering and we observed dimers in solution for both forms of the enzyme. Our results suggest that ACE homo- and heterodimerization does occur under physiologic conditions.


Asunto(s)
Peptidil-Dipeptidasa A/química , Peptidil-Dipeptidasa A/metabolismo , Multimerización de Proteína/fisiología , Animales , Células CHO , Membrana Celular/metabolismo , Cricetinae , Cricetulus , Cristalización , Células HEK293 , Humanos , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
9.
FEBS Lett ; 598(2): 242-251, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37904282

RESUMEN

Human somatic angiotensin-1-converting enzyme (sACE) is composed of a catalytic N-(nACE) and C-domain (cACE) of similar size with different substrate specificities. It is involved in the regulation of blood pressure by converting angiotensin I to the vasoconstrictor angiotensin II and has been a major focus in the development of therapeutics for hypertension. Bioactive peptides from various sources, including milk, have been identified as natural ACE inhibitors. We report the structural basis for the role of two lacototripeptides, Val-Pro-Pro and Ile-Pro-Pro, in domain-specific inhibition of ACE using X-ray crystallography and kinetic analysis. The lactotripeptides have preference for nACE due to altered polar interactions distal to the catalytic zinc ion. Elucidating the mechanism of binding and domain selectivity of these peptides also provides important insights into the functional roles of ACE.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina , Peptidil-Dipeptidasa A , Humanos , Peptidil-Dipeptidasa A/química , Peptidil-Dipeptidasa A/metabolismo , Cinética , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/química , Inhibidores de la Enzima Convertidora de Angiotensina/metabolismo , Angiotensinas
10.
Sci Signal ; 16(789): eadd3184, 2023 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-37311034

RESUMEN

The activation of at least 23 different mammalian kinases requires the phosphorylation of their hydrophobic motifs by the kinase PDK1. A linker connects the phosphoinositide-binding PH domain to the catalytic domain, which contains a docking site for substrates called the PIF pocket. Here, we used a chemical biology approach to show that PDK1 existed in equilibrium between at least three distinct conformations with differing substrate specificities. The inositol polyphosphate derivative HYG8 bound to the PH domain and disrupted PDK1 dimerization by stabilizing a monomeric conformation in which the PH domain associated with the catalytic domain and the PIF pocket was accessible. In the absence of lipids, HYG8 potently inhibited the phosphorylation of Akt (also termed PKB) but did not affect the intrinsic activity of PDK1 or the phosphorylation of SGK, which requires docking to the PIF pocket. In contrast, the small-molecule valsartan bound to the PIF pocket and stabilized a second distinct monomeric conformation. Our study reveals dynamic conformations of full-length PDK1 in which the location of the linker and the PH domain relative to the catalytic domain determines the selective phosphorylation of PDK1 substrates. The study further suggests new approaches for the design of drugs to selectively modulate signaling downstream of PDK1.


Asunto(s)
Mamíferos , Polifosfatos , Animales , Especificidad por Sustrato , Fosforilación , Dominio Catalítico , Dimerización
11.
FEBS J ; 289(21): 6659-6671, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35653492

RESUMEN

Human angiotensin I-converting enzyme (ACE) has two isoforms, somatic ACE (sACE) and testis ACE (tACE). The functions of sACE are widespread, with its involvement in blood pressure regulation most extensively studied. sACE is composed of an N-domain (nACE) and a C-domain (cACE), both catalytically active but have significant structural differences, resulting in different substrate specificities. Even though ACE inhibitors are used clinically, they need much improvement because of serious side effects seen in patients (~ 25-30%) with long-term treatment due to nonselective inhibition of nACE and cACE. Investigation into the distinguishing structural features of each domain is therefore of vital importance for the development of domain-specific inhibitors with minimal side effects. Here, we report kinetic data and high-resolution crystal structures of both nACE (1.75 Å) and cACE (1.85 Å) in complex with fosinoprilat, a clinically used inhibitor. These structures allowed detailed analysis of the molecular features conferring domain selectivity by fosinoprilat. Particularly, altered hydrophobic interactions were observed to be a contributing factor. These experimental data contribute to improved understanding of the structural features that dictate ACE inhibitor domain selectivity, allowing further progress towards designing novel 2nd-generation domain-specific potent ACE inhibitors suitable for clinical administration, with a variety of potential future therapeutic benefits. DATABASE: The atomic coordinates and structure factors for nACE-fosinoprilat and cACE-fosinoprilat structures have been deposited with codes 7Z6Z and 7Z70, respectively, in the RCSB Protein Data Bank, www.pdb.org.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina , Peptidil-Dipeptidasa A , Humanos , Peptidil-Dipeptidasa A/química , Cristalografía por Rayos X , Inhibidores de la Enzima Convertidora de Angiotensina/química , Angiotensinas
12.
J Med Chem ; 65(4): 3371-3387, 2022 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-35113565

RESUMEN

Selective inhibition of the angiotensin-converting enzyme C-domain (cACE) and neprilysin (NEP), leaving the ACE N-domain (nACE) free to degrade bradykinin and other peptides, has the potential to provide the potent antihypertensive and cardioprotective benefits observed for nonselective dual ACE/NEP inhibitors, such as omapatrilat, without the increased risk of adverse effects. We have synthesized three 1-carboxy-3-phenylpropyl dipeptide inhibitors with nanomolar potency based on the previously reported C-domain selective ACE inhibitor lisinopril-tryptophan (LisW) to probe the structural requirements for potent dual cACE/NEP inhibition. Here we report the synthesis, enzyme kinetic data, and high-resolution crystal structures of these inhibitors bound to nACE and cACE, providing valuable insight into the factors driving potency and selectivity. Overall, these results highlight the importance of the interplay between the S1' and S2' subsites for ACE domain selectivity, providing guidance for future chemistry efforts toward the development of dual cACE/NEP inhibitors.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Neprilisina/farmacología , Peptidil-Dipeptidasa A/efectos de los fármacos , Inhibidores de la Enzima Convertidora de Angiotensina/síntesis química , Sitios de Unión/efectos de los fármacos , Bradiquinina/metabolismo , Simulación por Computador , Cristalografía por Rayos X , Humanos , Cinética , Lisinopril/farmacología , Peptidil-Dipeptidasa A/química , Piridinas/farmacología , Tiazepinas/farmacología
13.
FEBS J ; 288(7): 2238-2256, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33067882

RESUMEN

Angiotensin-1-converting enzyme (ACE) is a key enzyme in the renin-angiotensin-aldosterone and kinin systems where it cleaves angiotensin I and bradykinin peptides, respectively. However, ACE also participates in numerous other physiological functions, can hydrolyse many peptide substrates and has various exo- and endopeptidase activities. ACE achieves this complexity by containing two homologous catalytic domains (N- and C-domains), which exhibit different substrate specificities. Here, we present the first open conformation structures of ACE N-domain and a unique closed C-domain structure (2.0 Å) where the C terminus of a symmetry-related molecule is observed inserted into the active-site cavity and binding to the zinc ion. The open native N-domain structure (1.85 Å) enables comparison with ACE2, a homologue previously observed in open and closed states. An open S2 _S'-mutant N-domain structure (2.80 Å) includes mutated residues in the S2 and S' subsites that effect ligand binding, but are distal to the binding site. Analysis of these structures provides important insights into how structural features of the ACE domains are able to accommodate the wide variety of substrates and allow different peptidase activities. DATABASE: The atomic coordinates and structure factors for Open nACE, Open S2_S'-nACE and Native G13-cACE structures have been deposited with codes 6ZPQ, 6ZPT and 6ZPU, respectively, in the RCSB Protein Data Bank, www.pdb.org.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/química , Dominio Catalítico/genética , Peptidil-Dipeptidasa A/ultraestructura , Conformación Proteica , Sitios de Unión/genética , Cristalografía por Rayos X , Bases de Datos de Proteínas , Humanos , Peptidil-Dipeptidasa A/química , Peptidil-Dipeptidasa A/genética , Unión Proteica/genética , Especificidad por Sustrato/genética
14.
Biochemistry ; 49(16): 3464-76, 2010 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-20297840

RESUMEN

Carbonic anhydrase (CA) catalyzes the reversible hydration of carbon dioxide to hydrogen carbonate, and its role in maintaining pH balance has made it an attractive drug target. Steroidal sulfamate esters, inhibitors of the cancer drug target steroid sulfatase (STS), are sequestered in vivo by CA II in red blood cells, which may be the origin of their excellent drug properties. Understanding the structural basis of this is important for drug design. Structures of CA II complexed with 2-methoxyestradiol 3-O-sulfamate (3), 2-ethylestradiol 3,17-O,O-bis(sulfamate) (4), and 2-methoxyestradiol 17-O-sulfamate (5) are reported to 2.10, 1.85, and 1.64 A, respectively. Inhibitor 3 interacts with the active site Zn(II) ion through the 3-O-sulfamate, while inhibitors 4 and 5 bind through their 17-O-sulfamate. Comparison of the IC(50) values for CA II inhibition gave respective values of 56, 662, 2113, 169, 770, and 86 nM for estrone 3-O-sulfamate (1), 2-methoxyestradiol 3,17-O,O-bis(sulfamate) (2), 3, 4, 5, and 5'-((4H-1,2,4-triazol-4-yl)methyl)-3-chloro-2'-cyanobiphenyl-4-yl sulfamate (6), a nonsteroidal dual aromatase-sulfatase inhibitor. Inhibitors 2, 5, and 6 showed binding to a second adjacent site that is capable of binding both steroidal and nonsteroidal ligands. Examination of both IC(50) values and crystal structures suggests that 2-substituents on the steroid nucleus hinder binding via a 3-O-sulfamate, leading to coordination through a 17-O-sulfamate if present. These results underline the influence of small structural changes on affinity and mode of binding, the degree of flexibility in the design of sulfamate-based inhibitors, and suggest a strategy for inhibitors which interact with both the active site and the second adjacent binding site simultaneously that could be both potent and selective.


Asunto(s)
Anhidrasa Carbónica II/química , Bicarbonatos/metabolismo , Sitios de Unión , Dióxido de Carbono/metabolismo , Anhidrasa Carbónica II/antagonistas & inhibidores , Colorimetría , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Estrona/análogos & derivados , Estrona/química , Estrona/farmacología , Humanos , Cinética , Modelos Moleculares , Conformación Proteica
15.
J Med Chem ; 63(10): 5488-5500, 2020 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-32337993

RESUMEN

Neprilysin (NEP) and angiotensin-converting enzyme (ACE) are two key zinc-dependent metallopeptidases in the natriuretic peptide and kinin systems and renin-angiotensin-aldosterone system, respectively. They play an important role in blood pressure regulation and reducing the risk of heart failure. Vasopeptidase inhibitors omapatrilat and sampatrilat possess dual activity against these enzymes by blocking the ACE-dependent conversion of angiotensin I to the potent vasoconstrictor angiotensin II while simultaneously halting the NEP-dependent degradation of vasodilator atrial natriuretic peptide. Here, we report crystal structures of omapatrilat, sampatrilat, and sampatrilat-ASP (a sampatrilat analogue) in complex with NEP at 1.75, 2.65, and 2.6 Å, respectively. A detailed analysis of these structures and the corresponding structures of ACE with these inhibitors has provided the molecular basis of dual inhibitor recognition involving the catalytic site in both enzymes. This new information will be very useful in the design of safer and more selective vasopeptidase inhibitors of NEP and ACE for effective treatment in hypertension and heart failure.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/metabolismo , Diseño de Fármacos , Mesilatos/metabolismo , Neprilisina/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Piridinas/metabolismo , Tiazepinas/metabolismo , Tirosina/análogos & derivados , Inhibidores de la Enzima Convertidora de Angiotensina/química , Antihipertensivos/química , Antihipertensivos/metabolismo , Cristalografía por Rayos X/métodos , Mesilatos/química , Neprilisina/química , Peptidil-Dipeptidasa A/química , Unión Proteica/fisiología , Estructura Secundaria de Proteína , Piridinas/química , Tiazepinas/química , Tirosina/química , Tirosina/metabolismo
16.
J Med Chem ; 61(1): 344-359, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29206036

RESUMEN

Angiotensin-I converting enzyme (ACE) is a zinc metalloprotease consisting of two catalytic domains (N- and C-). Most clinical ACE inhibitor(s) (ACEi) have been shown to inhibit both domains nonselectively, resulting in adverse effects such as cough and angioedema. Selectively inhibiting the individual domains is likely to reduce these effects and potentially treat fibrosis in addition to hypertension. ACEi from the GVK Biosciences database were inspected for possible N-domain selective binding patterns. From this set, a diprolyl chemical series was modeled using docking simulations. The series was expanded based on key target interactions involving residues known to impart N-domain selectivity. In total, seven diprolyl compounds were synthesized and tested for N-domain selective ACE inhibition. One compound with an aspartic acid in the P2 position (compound 16) displayed potent inhibition (Ki = 11.45 nM) and was 84-fold more selective toward the N-domain. A high-resolution crystal structure of compound 16 in complex with the N-domain revealed the molecular basis for the observed selectivity.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/química , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Diseño de Fármacos , Peptidil-Dipeptidasa A/química , Peptidil-Dipeptidasa A/metabolismo , Prolina/química , Inhibidores de la Enzima Convertidora de Angiotensina/metabolismo , Animales , Células CHO , Dominio Catalítico , Cricetulus , Simulación del Acoplamiento Molecular , Prolina/metabolismo , Prolina/farmacología , Especificidad por Sustrato , Articulación Talocalcánea
17.
J Med Chem ; 61(22): 10141-10154, 2018 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-30372620

RESUMEN

Omapatrilat was designed as a vasopeptidase inhibitor with dual activity against the zinc metallopeptidases angiotensin-1 converting enzyme (ACE) and neprilysin (NEP). ACE has two homologous catalytic domains (nACE and cACE), which exhibit different substrate specificities. Here, we report high-resolution crystal structures of omapatrilat in complex with nACE and cACE and show omapatrilat has subnanomolar affinity for both domains. The structures show nearly identical binding interactions for omapatrilat in each domain, explaining the lack of domain selectivity. The cACE complex structure revealed an omapatrilat dimer occupying the cavity beyond the S2 subsite, and this dimer had low micromolar inhibition of nACE and cACE. These results highlight residues beyond the S2 subsite that could be exploited for domain selective inhibition. In addition, it suggests the possibility of either domain specific allosteric inhibitors that bind exclusively to the nonprime cavity or the potential for targeting specific substrates rather than completely inhibiting the enzyme.


Asunto(s)
Diseño de Fármacos , Peptidil-Dipeptidasa A/química , Peptidil-Dipeptidasa A/metabolismo , Piridinas/metabolismo , Tiazepinas/metabolismo , Secuencia de Aminoácidos , Dominio Catalítico , Humanos , Ligandos , Modelos Moleculares
18.
FEBS J ; 285(8): 1477-1490, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29476645

RESUMEN

Angiotensin-1-converting enzyme (ACE) is a zinc metallopeptidase that consists of two homologous catalytic domains (known as nACE and cACE) with different substrate specificities. Based on kinetic studies it was previously reported that sampatrilat, a tight-binding inhibitor of ACE, Ki = 13.8 nm and 171.9 nm for cACE and nACE respectively [Sharma et al., Journal of Chemical Information and Modeling (2016), 56, 2486-2494], was 12.4-fold more selective for cACE. In addition, samAsp, in which an aspartate group replaces the sampatrilat lysine, was found to be a nonspecific and lower micromolar affinity inhibitor. Here, we report a detailed three-dimensional structural analysis of sampatrilat and samAsp binding to ACE using high-resolution crystal structures elucidated by X-ray crystallography, which provides a molecular basis for differences in inhibitor affinity and selectivity for nACE and cACE. The structures show that the specificity of sampatrilat can be explained by increased hydrophobic interactions and a H-bond from Glu403 of cACE with the lysine side chain of sampatrilat that are not observed in nACE. In addition, the structures clearly show a significantly greater number of hydrophilic and hydrophobic interactions with sampatrilat compared to samAsp in both cACE and nACE consistent with the difference in affinities. Our findings provide new experimental insights into ligand binding at the active site pockets that are important for the design of highly specific domain selective inhibitors of ACE. DATABASE: The atomic coordinates and structure factors for N- and C-domains of ACE bound to sampatrilat and sampatrilat-Asp complexes (6F9V, 6F9R, 6F9T and 6F9U respectively) have been deposited in the Protein Data Bank, Research Collaboratory for Structural Bioinformatics, Rutgers University, New Brunswick, NJ (http://www.rcsb.org/).


Asunto(s)
Ácido Aspártico/metabolismo , Dominio Catalítico , Mesilatos/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Tirosina/análogos & derivados , Ácido Aspártico/química , Cristalografía por Rayos X , Humanos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Mesilatos/química , Peptidil-Dipeptidasa A/química , Inhibidores de Proteasas/química , Inhibidores de Proteasas/metabolismo , Unión Proteica , Conformación Proteica , Especificidad por Sustrato , Tirosina/química , Tirosina/metabolismo
19.
Sci Rep ; 7: 45409, 2017 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-28345667

RESUMEN

The control of mosquitoes is threatened by the appearance of insecticide resistance and therefore new control chemicals are urgently required. Here we show that inhibitors of mosquito peptidyl dipeptidase, a peptidase related to mammalian angiotensin-converting enzyme (ACE), are insecticidal to larvae of the mosquitoes, Aedes aegypti and Anopheles gambiae. ACE inhibitors (captopril, fosinopril and fosinoprilat) and two peptides (trypsin-modulating oostatic factor/TMOF and a bradykinin-potentiating peptide, BPP-12b) were all inhibitors of the larval ACE activity of both mosquitoes. Two inhibitors, captopril and fosinopril (a pro-drug ester of fosinoprilat), were tested for larvicidal activity. Within 24 h captopril had killed >90% of the early instars of both species with 3rd instars showing greater resistance. Mortality was also high within 24 h of exposure of 1st, 2nd and 3rd instars of An. gambiae to fosinopril. Fosinopril was also toxic to Ae. aegypti larvae, although the 1st instars appeared to be less susceptible to this pro-drug even after 72 h exposure. Homology models of the larval An. gambiae ACE proteins (AnoACE2 and AnoACE3) reveal structural differences compared to human ACE, suggesting that structure-based drug design offers a fruitful approach to the development of selective inhibitors of mosquito ACE enzymes as novel larvicides.


Asunto(s)
Aedes/efectos de los fármacos , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Anopheles/efectos de los fármacos , Larva/efectos de los fármacos , Animales , Captopril/farmacología , Vectores de Enfermedades , Fosinopril/análogos & derivados , Fosinopril/farmacología , Humanos , Resistencia a los Insecticidas/efectos de los fármacos , Insecticidas/farmacología , Mosquitos Vectores/efectos de los fármacos , Peptidil-Dipeptidasa A/metabolismo
20.
Methods Enzymol ; 407: 64-82, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16757315

RESUMEN

Two classes of proteins govern Ras activation. Guanine-nucleotide exchange factors (Ras GEFs) catalyze the activation of Ras by inducing the dissociation of GDP to allow association of the more abundant GTP, whereas GTPase-activating proteins (Ras GAPs), bind to the GTP-bound form and, by enhancing the intrinsic GTPase activity, catalyze Ras inactivation. A wide range of Ras GEFs and Ras GAPs have been identified from the various genome projects, and in a few instances, the mechanisms by which signals originating from activated receptors converge on specific GEFs and GAPs have been mapped. However, for most Ras GEFs and GAPs we have a poor understanding of their regulation. Here we focus on describing methods used to study the regulation of the GAP1 family of Ras GAPs. In particular, we emphasize how by combining biochemical, molecular, and imaging techniques, one can determine some of the complex array of mechanisms that have evolved to modulate the spatial and temporal dynamics of Ras regulation through these various Ras GAPs. By combining biochemical, molecular, and imaging techniques, we describe the visualization of the diverse and dynamic mechanisms through which stimulation of cell surface receptors leads to the regulation of these proteins. Thus, although each member of the GAP1 family performs the same basic biological function, that is, they function as Ras GAPs, each is designed to respond and decode signals from distinct second messenger pathways.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas Activadoras de ras GTPasa/metabolismo , Regulación de la Expresión Génica , Glutatión Transferasa/aislamiento & purificación , Células HeLa , Humanos , Liposomas/metabolismo , Proteínas Recombinantes de Fusión/aislamiento & purificación , Quinasas raf/metabolismo , Proteínas Activadoras de ras GTPasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA