Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
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
2.
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
3.
Biochem Biophys Res Commun ; 481(1-2): 111-116, 2016 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-27818199

RESUMEN

Somatic angiotensin converting enzyme (sACE) is comprised of two homologous domains (N and C domains), whereas the smaller germinal isoform (tACE) is identical to the C domain. Both isozymes share an identical stalk, transmembrane and cytoplasmic domain, and undergo ectodomain shedding by an as yet unknown protease. Here we present evidence for the role of regions distal and proximal to the cleavage site in human ACE shedding. First, because of intrinsic differences between the N and C domains, discrete secondary structures (α-helix 7 and 8) on the surface of tACE were replaced with their N domain counterparts. Surprisingly, neither α-helix 7 nor α-helix 8 proved to be an absolute requirement for shedding. In the proximal ectodomain of tACE residues H610-L614 were mutated to alanines and this resulted in a decrease in ACE shedding. An N-terminal extension of this mutation caused a reduction in cellular ACE activity. More importantly, it affected the processing of the protein to the membrane, resulting in expression of an underglycosylated form of ACE. When E608-H614 was mutated to the homologous region of the N domain, processing was normal and shedding only moderately decreased suggesting that this region is more crucial for the processing of ACE than it is for regulating shedding. Finally, to determine whether glycosylation of the asparagine proximal to the Pro1199-Leu polymorphism in sACE affected shedding, the equivalent P623L mutation in tACE was investigated. The P623L tACE mutant showed an increase in shedding and MALDI MS analysis of a tryptic digest indicated that N620WT was glycosylated. The absence of an N-linked glycan at N620, resulted in an even greater increase in shedding. Thus, the conformational flexibility that the leucine confers to the stalk, is increased by the lack of glycosylation reducing access of the sheddase to the cleavage site.


Asunto(s)
Micropartículas Derivadas de Células/química , Micropartículas Derivadas de Células/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Peptidil-Dipeptidasa A/química , Peptidil-Dipeptidasa A/metabolismo , Sustitución de Aminoácidos , Animales , Sitios de Unión , Células CHO , Membrana Celular/química , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Micropartículas Derivadas de Células/ultraestructura , Cricetulus , Activación Enzimática , Humanos , Peptidil-Dipeptidasa A/ultraestructura , Unión Proteica , Conformación Proteica , Dominios Proteicos , Relación Estructura-Actividad
4.
J Biol Chem ; 289(3): 1798-814, 2014 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-24297181

RESUMEN

Somatic angiotensin-converting enzyme (sACE), a key regulator of blood pressure and electrolyte fluid homeostasis, cleaves the vasoactive angiotensin-I, bradykinin, and a number of other physiologically relevant peptides. sACE consists of two homologous and catalytically active N- and C-domains, which display marked differences in substrate specificities and chloride activation. A series of single substitution mutants were generated and evaluated under varying chloride concentrations using isothermal titration calorimetry. The x-ray crystal structures of the mutants provided details on the chloride-dependent interactions with ACE. Chloride binding in the chloride 1 pocket of C-domain ACE was found to affect positioning of residues from the active site. Analysis of the chloride 2 pocket R522Q and R522K mutations revealed the key interactions with the catalytic site that are stabilized via chloride coordination of Arg(522). Substrate interactions in the S2 subsite were shown to affect chloride affinity in the chloride 2 pocket. The Glu(403)-Lys(118) salt bridge in C-domain ACE was shown to stabilize the hinge-bending region and reduce chloride affinity by constraining the chloride 2 pocket. This work demonstrated that substrate composition to the C-terminal side of the scissile bond as well as interactions of larger substrates in the S2 subsite moderate chloride affinity in the chloride 2 pocket of the ACE C-domain, providing a rationale for the substrate-selective nature of chloride dependence in ACE and how this varies between the N- and C-domains.


Asunto(s)
Cloruros/química , Peptidil-Dipeptidasa A/química , Sustitución de Aminoácidos , Sitios de Unión , Cloruros/metabolismo , Cristalografía por Rayos X , Humanos , Mutación Missense , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Termodinámica
5.
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
6.
Biochem J ; 436(1): 53-9, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21352096

RESUMEN

Human ACE (angiotensin-I-converting enzyme) has long been regarded as an excellent target for the treatment of hypertension and related cardiovascular diseases. Highly potent inhibitors have been developed and are extensively used in the clinic. To develop inhibitors with higher therapeutic efficacy and reduced side effects, recent efforts have been directed towards the discovery of compounds able to simultaneously block more than one zinc metallopeptidase (apart from ACE) involved in blood pressure regulation in humans, such as neprilysin and ECE-1 (endothelin-converting enzyme-1). In the present paper, we show the first structures of testis ACE [C-ACE, which is identical with the C-domain of somatic ACE and the dominant domain responsible for blood pressure regulation, at 1.97Å (1 Å=0.1 nm)] and the N-domain of somatic ACE (N-ACE, at 2.15Å) in complex with a highly potent and selective dual ACE/ECE-1 inhibitor. The structural determinants revealed unique features of the binding of two molecules of the dual inhibitor in the active site of C-ACE. In both structures, the first molecule is positioned in the obligatory binding site and has a bulky bicyclic P(1)' residue with the unusual R configuration which, surprisingly, is accommodated by the large S(2)' pocket. In the C-ACE complex, the isoxazole phenyl group of the second molecule makes strong pi-pi stacking interactions with the amino benzoyl group of the first molecule locking them in a 'hand-shake' conformation. These features, for the first time, highlight the unusual architecture and flexibility of the active site of C-ACE, which could be further utilized for structure-based design of new C-ACE or vasopeptidase inhibitors.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/química , Oligopéptidos/química , Péptidos/química , Peptidil-Dipeptidasa A/química , Ácidos Fosfínicos/química , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/metabolismo , Sitios de Unión , Enzimas Convertidoras de Endotelina , Humanos , Metaloendopeptidasas/antagonistas & inhibidores , Metaloendopeptidasas/metabolismo , Modelos Moleculares , Oligopéptidos/farmacología , Péptidos/metabolismo , Péptidos/farmacología , Peptidil-Dipeptidasa A/metabolismo , Ácidos Fosfínicos/farmacología , Relación Estructura-Actividad
7.
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
8.
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
9.
J Biol Chem ; 285(46): 35685-93, 2010 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-20826823

RESUMEN

Angiotensin-I-converting enzyme (ACE) plays a critical role in the regulation of blood pressure through its central role in the renin-angiotensin and kallikrein-kinin systems. ACE contains two domains, the N and C domains, both of which are heavily glycosylated. Structural studies of ACE have been fraught with severe difficulties because of surface glycosylation of the protein. In order to investigate the role of glycosylation in the N domain and to create suitable forms for crystallization, we have investigated the importance of the 10 potential N-linked glycan sites using enzymatic deglycosylation, limited proteolysis, and mass spectrometry. A number of glycosylation mutants were generated via site-directed mutagenesis, expressed in CHO cells, and analyzed for enzymatic activity and thermal stability. At least eight of 10 of the potential glycan sites are glycosylated; three C-terminal sites were sufficient for expression of active N domain, whereas two N-terminal sites are important for its thermal stability. The minimally glycosylated Ndom389 construct was highly suitable for crystallization studies. The structure in the presence of an N domain-selective phosphinic inhibitor RXP407 was determined to 2.0 Å resolution. The Ndom389 structure revealed a hinge region that may contribute to the breathing motion proposed for substrate binding.


Asunto(s)
Oligopéptidos/química , Peptidil-Dipeptidasa A/química , Ácidos Fosfínicos/química , Estructura Terciaria de Proteína , Animales , Sitios de Unión , Biocatálisis/efectos de los fármacos , Western Blotting , Células CHO , Cricetinae , Cricetulus , Cristalografía por Rayos X , Estabilidad de Enzimas , Glicosilación , Humanos , Espectrometría de Masas , Modelos Moleculares , Estructura Molecular , Mutación , Oligopéptidos/metabolismo , Oligopéptidos/farmacología , Peptidil-Dipeptidasa A/metabolismo , Ácidos Fosfínicos/metabolismo , Ácidos Fosfínicos/farmacología , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato , Temperatura
10.
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
11.
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
12.
J Mol Biol ; 357(3): 964-74, 2006 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-16476442

RESUMEN

Human somatic angiotensin I-converting enzyme (sACE) is a key regulator of blood pressure and an important drug target for combating cardiovascular and renal disease. sACE comprises two homologous metallopeptidase domains, N and C, joined by an inter-domain linker. Both domains are capable of cleaving the two hemoregulatory peptides angiotensin I and bradykinin, but differ in their affinities for a range of other substrates and inhibitors. Previously we determined the structure of testis ACE (C domain); here we present the crystal structure of the N domain of sACE (both in the presence and absence of the antihypertensive drug lisinopril) in order to aid the understanding of how these two domains differ in specificity and function. In addition, the structure of most of the inter-domain linker allows us to propose relative domain positions for sACE that may contribute to the domain cooperativity. The structure now provides a platform for the design of "domain-specific" second-generation ACE inhibitors.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/síntesis química , Inhibidores de la Enzima Convertidora de Angiotensina/metabolismo , Peptidil-Dipeptidasa A/química , Peptidil-Dipeptidasa A/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Dominio Catalítico , Cristalización , Cristalografía por Rayos X , Diseño de Fármacos , Humanos , Datos de Secuencia Molecular , Peptidil-Dipeptidasa A/genética , Estructura Terciaria de Proteína
13.
Biochem J ; 389(Pt 3): 739-44, 2005 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-15813703

RESUMEN

sACE (somatic angiotensin-converting enzyme) consists of two homologous, N and C domains, whereas the testis isoenzyme [tACE (testis ACE)] consists of a single C domain. Both isoenzymes are shed from the cell surface by a sheddase activity, although sACE is shed much less efficiently than tACE. We hypothesize that the N domain of sACE plays a regulatory role, by occluding a recognition motif on the C domain required for ectodomain shedding and by influencing the catalytic efficiency. To test this, we constructed two mutants: CNdom-ACE and CCdom-ACE. CNdom-ACE was shed less efficiently than sACE, whereas CCdom-ACE was shed as efficiently as tACE. Notably, cleavage occurred both within the stalk and the interdomain bridge in both mutants, suggesting that a sheddase recognition motif resides within the C domain and is capable of directly cleaving at both positions. Analysis of the catalytic properties of the mutants and comparison with sACE and tACE revealed that the k(cat) for sACE and CNdom-ACE was less than or equal to the sum of the kcat values for tACE and the N-domain, suggesting negative co-operativity, whereas the kcat value for the CCdom-ACE suggested positive co-operativity between the two domains. Taken together, the results provide support for (i) the existence of a sheddase recognition motif in the C domain and (ii) molecular flexibility of the N and C domains in sACE, resulting in occlusion of the C-domain recognition motif by the N domain as well as close contact of the two domains during hydrolysis of peptide substrates.


Asunto(s)
Peptidil-Dipeptidasa A/química , Peptidil-Dipeptidasa A/metabolismo , Animales , Células CHO , Dominio Catalítico , Cricetinae , Expresión Génica , Mutación , Estructura Terciaria de Proteína
14.
FEBS J ; 283(6): 1060-76, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26748546

RESUMEN

UNLABELLED: Angiotensin-1-converting enzyme (ACE), a zinc metallopeptidase, consists of two homologous catalytic domains (N and C) with different substrate specificities. Here we report kinetic parameters of five different forms of human ACE with various amyloid beta (Aß) substrates together with high resolution crystal structures of the N-domain in complex with Aß fragments. For the physiological Aß(1-16) peptide, a novel ACE cleavage site was found at His14-Gln15. Furthermore, Aß(1-16) was preferentially cleaved by the individual N-domain; however, the presence of an inactive C-domain in full-length somatic ACE (sACE) greatly reduced enzyme activity and affected apparent selectivity. Two fluorogenic substrates, Aß(4-10)Q and Aß(4-10)Y, underwent endoproteolytic cleavage at the Asp7-Ser8 bond with all ACE constructs showing greater catalytic efficiency for Aß(4-10)Y. Surprisingly, in contrast to Aß(1-16) and Aß(4-10)Q, sACE showed positive domain cooperativity and the double C-domain (CC-sACE) construct no cooperativity towards Aß(4-10)Y. The structures of the Aß peptide-ACE complexes revealed a common mode of peptide binding for both domains which principally targets the C-terminal P2' position to the S2' pocket and recognizes the main chain of the P1' peptide. It is likely that N-domain selectivity for the amyloid peptide is conferred through the N-domain specific S2' residue Thr358. Additionally, the N-domain can accommodate larger substrates through movement of the N-terminal helices, as suggested by the disorder of the hinge region in the crystal structures. Our findings are important for the design of domain selective inhibitors as the differences in domain selectivity are more pronounced with the truncated domains compared to the more physiological full-length forms. DATABASE: The atomic coordinates and structure factors for N-domain ACE with Aß peptides 4-10 (5AM8), 10-16 (5AM9), 1-16 (5AMA), 35-42 (5AMB) and (4-10)Y (5AMC) complexes have been deposited in the Protein Data Bank, Research Collaboratory for Structural Bioinformatics, Rutgers University, New Brunswick, NJ, USA (http://www.rcsb.org/).


Asunto(s)
Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Peptidil-Dipeptidasa A/química , Peptidil-Dipeptidasa A/metabolismo , Secuencia de Aminoácidos , Péptidos beta-Amiloides/genética , Sitios de Unión , Cristalografía por Rayos X , Variación Genética , Humanos , Hidrólisis , Técnicas In Vitro , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Peptidil-Dipeptidasa A/genética , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato
15.
ACS Med Chem Lett ; 5(4): 346-51, 2014 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-24900839

RESUMEN

The K-26 family of bacterial secondary metabolites are N-modified tripeptides terminated by an unusual phosphonate analog of tyrosine. These natural products, produced via three different actinomycetales, are potent inhibitors of human angiotensin-I converting enzyme (ACE). Herein we investigate the interkingdom pharmacology of the K-26 family by synthesizing these metabolites and assessing their potency as inhibitors of both the N-terminal and C-terminal domains of human ACE. In most cases, selectivity for the C-terminal domain of ACE is displayed. Co-crystallization of K-26 in both domains of human ACE reveals the structural basis of the potent inhibition and has shown an unusual binding motif that may guide future design of domain-selective inhibitors. Finally, the activity of K-26 is assayed against a cohort of microbially produced ACE relatives. In contrast to the synthetic ACE inhibitor captopril, which demonstrates broad interkingdom inhibition of ACE-like enzymes, K-26 selectively targets the eukaryotic family.

16.
FEBS J ; 281(3): 943-56, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24289879

RESUMEN

Human somatic angiotensin-I converting enzyme (ACE) is a zinc-dependent dipeptidyl carboxypeptidase and a central component of the renin angiotensin aldosterone system (RAAS). Its involvement in the modulation of physiological actions of peptide hormones has positioned ACE as an important therapeutic target for the treatment of hypertension and cardiovascular disorders. Here, we report the crystal structures of the two catalytic domains of human ACE (N- and C-) in complex with FI, the S enantiomer of the phosphinic ACE/ECE-1 (endothelin converting enzyme) dual inhibitor FII, to a resolution of 1.91 and 1.85 Å, respectively. In addition, we have determined the structure of AnCE (an ACE homologue from Drosophila melanogaster) in complex with both isomers. The inhibitor FI (S configuration) can adapt to the active site of ACE catalytic domains and shows key differences in its binding mechanism mostly through the reorientation of the isoxazole phenyl side group at the P1' position compared with FII (R configuration). Differences in binding are also observed between FI and FII in complex with AnCE. Thus, the new structures of the ACE-inhibitor complexes presented here provide useful information for further exploration of ACE inhibitor pharmacophores involving phosphinic peptides and illustrate the role of chirality in enhancing drug specificity.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/química , Modelos Moleculares , Oligopéptidos/química , Peptidil-Dipeptidasa A/química , Ácidos Fosfínicos/química , Inhibidores de la Enzima Convertidora de Angiotensina/metabolismo , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Dominio Catalítico , Proteínas de Drosophila , Drosophila melanogaster/enzimología , Humanos , Proteínas de Insectos/antagonistas & inhibidores , Proteínas de Insectos/química , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Ligandos , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Oligopéptidos/metabolismo , Oligopéptidos/farmacología , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Ácidos Fosfínicos/metabolismo , Ácidos Fosfínicos/farmacología , Conformación Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Estereoisomerismo , Especificidad por Sustrato , Difracción de Rayos X
17.
Eur J Pharm Sci ; 56: 113-9, 2014 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-24561703

RESUMEN

Angiotensin-converting enzyme (ACE, EC 3.4.15.1) is a metallopeptidase comprised of two homologous catalytic domains (N- and C-domains). The C-domain cleaves the vasoactive angiotensin II precursor, angiotensin I, more efficiently than the N-domain. Thus, C-domain-selective ACE inhibitors have been designed to investigate the pharmacological effects of blocking the C-terminal catalytic site of the enzyme and improve the side effect profile of current ACE inhibitors. Lisinopril-tryptophan (LisW-S), an analogue of the ACE inhibitor lisinopril, is highly selective for the C-domain. In this study, we have analysed the ex vivo domain selectivity and pharmacokinetic profile of LisW-S. The IC50 value of LisW-S was 38.5 nM in rat plasma using the fluorogenic substrate Abz-FRKP(Dnp)P-OH. For the pharmacokinetics analysis of LisW-S, a sensitive and selective LC-MS/MS method was developed and validated to determine the concentration of LisW-S in rat plasma. LisW-S was administered to Wistar rats at a dose of 1 mg/kg bodyweight intravenously, 5 mg/kg bodyweight orally. The Cmax obtained following oral administration of the drug was 0.082 µM and LisW-S had an apparent terminal elimination half-life of around 3.1 h. The pharmacokinetic data indicate that the oral bioavailability of LisW-S was approximately 5.4%. These data provide a basis for better understanding the absorption mechanism of LisW-S and evaluating its clinical application.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/farmacocinética , Lisinopril/farmacocinética , Triptófano/farmacocinética , Inhibidores de la Enzima Convertidora de Angiotensina/química , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Células CACO-2 , Dominio Catalítico , Humanos , Lisinopril/química , Lisinopril/farmacología , Masculino , Peptidil-Dipeptidasa A/metabolismo , Ratas Wistar , Triptófano/química , Triptófano/farmacología
18.
PLoS One ; 9(2): e88001, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24505347

RESUMEN

BACKGROUND: Angiotensin I-converting enzyme (ACE) has two functional N- and C-domain active centers that display differences in the metabolism of biologically-active peptides including the hemoregulatory tetrapeptide, Ac-SDKP, hydrolysed preferentially by the N domain active center. Elevated Ac-SDKP concentrations are associated with reduced tissue fibrosis. RESULTS: We identified a patient of African descent exhibiting unusual blood ACE kinetics with reduced relative hydrolysis of two synthetic ACE substrates (ZPHL/HHL ratio) suggestive of the ACE N domain center inactivation. Inhibition of blood ACE activity by anti-catalytic mAbs and ACE inhibitors and conformational fingerprint of blood ACE suggested overall conformational changes in the ACE molecule and sequencing identified Ser333Trp substitution in the N domain of ACE. In silico analysis demonstrated S333W localized in the S1 pocket of the active site of the N domain with the bulky Trp adversely affecting binding of ACE substrates due to steric hindrance. Expression of mutant ACE (S333W) in CHO cells confirmed altered kinetic properties of mutant ACE and conformational changes in the N domain. Further, the S333W mutant displayed decreased ability (5-fold) to cleave the physiological substrate AcSDKP compared to wild-type ACE. CONCLUSIONS AND SIGNIFICANCE: A novel Ser333Trp ACE mutation results in dramatic changes in ACE kinetic properties and lowered clearance of Ac-SDKP. Individuals with this mutation (likely with significantly increased levels of the hemoregulatory tetrapeptide in blood and tissues), may confer protection against fibrosis.


Asunto(s)
Fibrosis/genética , Mutación/genética , Oligopéptidos/genética , Oligopéptidos/metabolismo , Péptidos/genética , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Secuencia de Aminoácidos , Animales , Células CHO , Línea Celular , Cricetulus , Fibrosis/metabolismo , Humanos , Cinética , Datos de Secuencia Molecular , Péptidos/metabolismo , Alineación de Secuencia
19.
J Clin Hypertens (Greenwich) ; 15(6): 413-9, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23730990

RESUMEN

Angiotensin-converting enzyme (ACE) inhibitors are first-line therapy for the treatment of hypertension, congestive heart failure, and diabetic nephropathy. ACE inhibitors are associated with adverse side effects such as persistent dry cough (ACE-cough) and, rarely, life-threatening angioedema (ACE-AE). The authors investigated the influence of ACE I/D polymorphism in combination with serum ACE activity, B2 receptor -9/+9 polymorphism, and B2 receptor C-58T single nucleotide polymorphism (SNP) on the development of ACE-AE and ACE-cough. The frequencies of ACE I/D as well as B2 receptor +9/-9 and C-58T polymorphisms were compared in patients with ACE-AE, ACE-cough, and ACE inhibitor-exposed controls, and serum ACE activity was measured. There were 52 cases of ACE-AE, 36 cases of ACE-cough, and 77 controls. The genotyping revealed a significant association between the B2 -9 allele and ACE inhibitor-induced AE (62% vs 38%, P=.008), and ACE inhibitor-induced cough (61% vs 38%, P=.02) when compared with controls. There was no significant association between ACE I/D polymorphism as well as the B2 C-58T SNP with both ACE-induced AE and cough. ACE activity was significantly higher in controls compared with patients with ACE-AE (34.5 ± 1.14 mU/mL vs 17.8 ± 0.86 mU/mL, P=.0001) and ACE-cough (34.5 ± 1.14 mU/mL vs 23.3 ± 1.88 mU/mL, P=.0001). Thus, our data suggest that the B2 -9 allele and reduced ACE activity are associated with both ACE-AE and ACE-cough.


Asunto(s)
Angioedema/inducido químicamente , Inhibidores de la Enzima Convertidora de Angiotensina/efectos adversos , Población Negra/genética , Tos/inducido químicamente , Hipertensión/tratamiento farmacológico , Hipertensión/genética , Peptidil-Dipeptidasa A/genética , Polimorfismo Genético , Receptor de Bradiquinina B2/genética , Anciano , Angioedema/genética , Comorbilidad , Tos/genética , Femenino , Genotipo , Humanos , Hipertensión/etnología , Masculino , Persona de Mediana Edad , Peptidil-Dipeptidasa A/sangre , Reacción en Cadena de la Polimerasa , Sudáfrica
20.
S Afr Med J ; 102(6): 461-4, 2012 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-22668937

RESUMEN

A number of membrane proteins are enzymatically cleaved or 'shed' from the cell surface, resulting in the modulation of biological events and opening novel pharmaceutical approaches to diverse diseases by targeting shedding. Our focus has been on understanding the shedding of angiotensin-converting enzyme (ACE), an enzyme that plays a pivotal role in blood pressure regulation. The identification of novel hereditary ACE mutations that result in increased ACE shedding has advanced our understanding of the role of ACE shedding in health and disease. Extensive biochemical and molecular analysis has helped to elucidate the mechanism of ACE shedding. These findings point to the potential therapeutic role of targeting shedding in regulating tissue ACE levels in cardiovascular disease.


Asunto(s)
Hipertensión/enzimología , Proteínas de la Membrana/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Proteínas ADAM/metabolismo , Humanos , Hipertensión/tratamiento farmacológico , Metaloproteinasas de la Matriz/metabolismo , Proteínas de la Membrana/genética , Terapia Molecular Dirigida , Peptidil-Dipeptidasa A/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA