Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.714
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Biochem ; 91: 449-473, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35303792

RESUMEN

Metals are essential components in life processes and participate in many important biological processes. Dysregulation of metal homeostasis is correlated with many diseases. Metals are also frequently incorporated into diagnosis and therapeutics. Understanding of metal homeostasis under (patho)physiological conditions and the molecular mechanisms of action of metallodrugs in biological systems has positive impacts on human health. As an emerging interdisciplinary area of research, metalloproteomics involves investigating metal-protein interactions in biological systems at a proteome-wide scale, has received growing attention, and has been implemented into metal-related research. In this review, we summarize the recent advances in metalloproteomics methodologies and applications. We also highlight emerging single-cell metalloproteomics, including time-resolved inductively coupled plasma mass spectrometry, mass cytometry, and secondary ion mass spectrometry. Finally, we discuss future perspectives in metalloproteomics, aiming to attract more original research to develop more advanced methodologies, which could be utilized rapidly by biochemists or biologists to expand our knowledge of how metal functions in biology and medicine.


Asunto(s)
Investigación Biomédica , Metaloproteínas , Humanos , Metaloproteínas/análisis , Metaloproteínas/química , Metaloproteínas/genética , Metales/análisis , Metales/química , Proteoma/genética , Proteómica/métodos
2.
Cell ; 185(12): 2013-2015, 2022 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-35688131

RESUMEN

Zinc is an essential element in living organisms, yet little is known about how cells ensure that zinc is allocated to the correct metalloproteins. Papers in Cell and Cell Reports demonstrate that the ZNG1 family of GTPases have metallochaperone functions: they directly transfer zinc to, and thereby activate, methionine aminopeptidases that are crucial for protein modification during or after translation.


Asunto(s)
Metaloproteínas , Zinc , Metaloproteínas/metabolismo , Chaperonas Moleculares/metabolismo , Zinc/metabolismo
3.
Cell ; 185(12): 2148-2163.e27, 2022 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-35584702

RESUMEN

Zinc (Zn) is an essential micronutrient and cofactor for up to 10% of proteins in living organisms. During Zn limitation, specialized enzymes called metallochaperones are predicted to allocate Zn to specific metalloproteins. This function has been putatively assigned to G3E GTPase COG0523 proteins, yet no Zn metallochaperone has been experimentally identified in any organism. Here, we functionally characterize a family of COG0523 proteins that is conserved across vertebrates. We identify Zn metalloprotease methionine aminopeptidase 1 (METAP1) as a COG0523 client, leading to the redesignation of this group of COG0523 proteins as the Zn-regulated GTPase metalloprotein activator (ZNG1) family. Using biochemical, structural, genetic, and pharmacological approaches across evolutionarily divergent models, including zebrafish and mice, we demonstrate a critical role for ZNG1 proteins in regulating cellular Zn homeostasis. Collectively, these data reveal the existence of a family of Zn metallochaperones and assign ZNG1 an important role for intracellular Zn trafficking.


Asunto(s)
Metaloendopeptidasas/metabolismo , Zinc , Animales , GTP Fosfohidrolasas/metabolismo , Homeostasis , Metalochaperonas/metabolismo , Metaloproteínas/genética , Ratones , Pez Cebra/metabolismo , Zinc/metabolismo
4.
Cell ; 175(5): 1418-1429.e9, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30454649

RESUMEN

We report here a simple and global strategy to map out gene functions and target pathways of drugs, toxins, or other small molecules based on "homomer dynamics" protein-fragment complementation assays (hdPCA). hdPCA measures changes in self-association (homomerization) of over 3,500 yeast proteins in yeast grown under different conditions. hdPCA complements genetic interaction measurements while eliminating the confounding effects of gene ablation. We demonstrate that hdPCA accurately predicts the effects of two longevity and health span-affecting drugs, the immunosuppressant rapamycin and the type 2 diabetes drug metformin, on cellular pathways. We also discovered an unsuspected global cellular response to metformin that resembles iron deficiency and includes a change in protein-bound iron levels. This discovery opens a new avenue to investigate molecular mechanisms for the prevention or treatment of diabetes, cancers, and other chronic diseases of aging.


Asunto(s)
Hierro/metabolismo , Metaloproteínas/metabolismo , Metformina/farmacología , Saccharomyces cerevisiae/metabolismo , Sirolimus/farmacología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Prueba de Complementación Genética , Humanos , Metaloproteínas/genética , Saccharomyces cerevisiae/genética
5.
Genes Dev ; 35(3-4): 177-179, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33526584

RESUMEN

Molybdenum cofactor (Moco) is synthesized endogenously in humans and is essential for human development. Supplementation of Moco or its precursors has been explored as a therapy to treat Moco-deficient patients but with significant limitations. By using the nematode C. elegans as a model, Warnhoff and colleagues (pp. 212-217) describe the beneficial impact of protein-bound Moco supplementation to treat Moco deficiency. If such an effect is conserved, this advance from basic research in worms may have significant clinical implications as a novel therapy for molybdenum cofactor deficiency.


Asunto(s)
Caenorhabditis elegans , Pteridinas , Animales , Coenzimas , Humanos , Errores Innatos del Metabolismo de los Metales , Metaloproteínas , Cofactores de Molibdeno
6.
Genes Dev ; 35(3-4): 212-217, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33446569

RESUMEN

The molybdenum cofactor (Moco) is a 520-Da prosthetic group that is synthesized in all domains of life. In animals, four oxidases (among them sulfite oxidase) use Moco as a prosthetic group. Moco is essential in animals; humans with mutations in genes that encode Moco biosynthetic enzymes display lethal neurological and developmental defects. Moco supplementation seems a logical therapy; however, the instability of Moco has precluded biochemical and cell biological studies of Moco transport and bioavailability. The nematode Caenorhabditis elegans can take up Moco from its bacterial diet and transport it to cells and tissues that express Moco-requiring enzymes, suggesting a system for Moco uptake and distribution. Here we show that protein-bound Moco is the stable, bioavailable species of Moco taken up by C. elegans from its diet and is an effective dietary supplement, rescuing a Celegans model of Moco deficiency. We demonstrate that diverse Moco:protein complexes are stable and bioavailable, suggesting a new strategy for the production and delivery of therapeutically active Moco to treat human Moco deficiency.


Asunto(s)
Caenorhabditis elegans/metabolismo , Coenzimas/administración & dosificación , Errores Innatos del Metabolismo de los Metales/terapia , Metaloproteínas/administración & dosificación , Pteridinas/administración & dosificación , Animales , Bacterias/metabolismo , Transporte Biológico , Coenzimas/deficiencia , Coenzimas/farmacocinética , Humanos , Metaloproteínas/deficiencia , Metaloproteínas/farmacocinética , Cofactores de Molibdeno , Unión Proteica , Pteridinas/farmacocinética
7.
Nature ; 603(7901): 522-527, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35236987

RESUMEN

Selective metal coordination is central to the functions of metalloproteins:1,2 each metalloprotein must pair with its cognate metallocofactor to fulfil its biological role3. However, achieving metal selectivity solely through a three-dimensional protein structure is a great challenge, because there is a limited set of metal-coordinating amino acid functionalities and proteins are inherently flexible, which impedes steric selection of metals3,4. Metal-binding affinities of natural proteins are primarily dictated by the electronic properties of metal ions and follow the Irving-Williams series5 (Mn2+ < Fe2+ < Co2+ < Ni2+ < Cu2+ > Zn2+) with few exceptions6,7. Accordingly, metalloproteins overwhelmingly bind Cu2+ and Zn2+ in isolation, regardless of the nature of their active sites and their cognate metal ions1,3,8. This led organisms to evolve complex homeostatic machinery and non-equilibrium strategies to achieve correct metal speciation1,3,8-10. Here we report an artificial dimeric protein, (AB)2, that thermodynamically overcomes the Irving-Williams restrictions in vitro and in cells, favouring the binding of lower-Irving-Williams transition metals over Cu2+, the most dominant ion in the Irving-Williams series. Counter to the convention in molecular design of achieving specificity through structural preorganization, (AB)2 was deliberately designed to be flexible. This flexibility enabled (AB)2 to adopt mutually exclusive, metal-dependent conformational states, which led to the discovery of structurally coupled coordination sites that disfavour Cu2+ ions by enforcing an unfavourable coordination geometry. Aside from highlighting flexibility as a valuable element in protein design, our results illustrate design principles for constructing selective metal sequestration agents.


Asunto(s)
Metaloproteínas , Metales , Proteínas , Aminoácidos , Dominio Catalítico , Iones , Metaloproteínas/química , Metales/química , Metales/metabolismo , Proteínas/química
8.
Proc Natl Acad Sci U S A ; 121(33): e2405836121, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39116128

RESUMEN

The 2011 discovery of the first rare earth-dependent enzyme in methylotrophic Methylobacterium extorquens AM1 prompted intensive research toward understanding the unique chemistry at play in these systems. This enzyme, an alcohol dehydrogenase (ADH), features a La3+ ion closely associated with redox-active coenzyme pyrroloquinoline quinone (PQQ) and is structurally homologous to the Ca2+-dependent ADH from the same organism. AM1 also produces a periplasmic PQQ-binding protein, PqqT, which we have now structurally characterized to 1.46-Å resolution by X-ray diffraction. This crystal structure reveals a Lys residue hydrogen-bonded to PQQ at the site analogously occupied by a Lewis acidic cation in ADH. Accordingly, we prepared K142A- and K142D-PqqT variants to assess the relevance of this site toward metal binding. Isothermal titration calorimetry experiments and titrations monitored by UV-Vis absorption and emission spectroscopies support that K142D-PqqT binds tightly (Kd = 0.6 ± 0.2 µM) to La3+ in the presence of bound PQQ and produces spectral signatures consistent with those of ADH enzymes. These spectral signatures are not observed for WT- or K142A-variants or upon addition of Ca2+ to PQQ ⸦ K142D-PqqT. Addition of benzyl alcohol to La3+-bound PQQ ⸦ K142D-PqqT (but not Ca2+-bound PQQ ⸦ K142D-PqqT, or La3+-bound PQQ ⸦ WT-PqqT) produces spectroscopic changes associated with PQQ reduction, and chemical trapping experiments reveal the production of benzaldehyde, supporting ADH activity. By creating a metal binding site that mimics native ADH enzymes, we present a rare earth-dependent artificial metalloenzyme primed for future mechanistic, biocatalytic, and biosensing applications.


Asunto(s)
Methylobacterium extorquens , Methylobacterium extorquens/enzimología , Methylobacterium extorquens/metabolismo , Metaloproteínas/química , Metaloproteínas/metabolismo , Alcohol Deshidrogenasa/metabolismo , Alcohol Deshidrogenasa/química , Cristalografía por Rayos X , Cofactor PQQ/metabolismo , Cofactor PQQ/química , Materiales Biomiméticos/química , Materiales Biomiméticos/metabolismo , Metales de Tierras Raras/química , Metales de Tierras Raras/metabolismo , Modelos Moleculares , Lantano/química , Lantano/metabolismo
9.
Nucleic Acids Res ; 52(D1): D483-D493, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-37941148

RESUMEN

MESPEUS is a freely accessible database which uses carefully selected metal coordination groups found in metalloprotein structures taken from the Protein Data Bank. The database contains geometrical information of metal sites within proteins, including 40 metal types. In order to completely determine the metal coordination, the symmetry-related units of a given protein structure are taken into account and are generated using the appropriate space group symmetry operations. This permits a more complete description of the metal coordination geometry by including all coordinating atoms. The user-friendly web interface allows users to directly search for a metal site of interest using several useful options, including searching for metal elements, metal-donor distances, coordination number, donor residue group, and structural resolution. These searches can be carried out singly or in combination. The details of a metal site and the metal site(s) in the whole structure can be graphically displayed using the interactive web interface. MESPEUS is automatically updated monthly by synchronizing with the PDB database. An investigation for the Mg-ATP interaction is given to demonstrate how MESPEUS can be used to extract information about metal sites by selecting structure and coordination features. MESPEUS is available at http://mespeus.nchu.edu.tw/.


Asunto(s)
Metaloproteínas , Metaloproteínas/química , Metales/química , Bases de Datos de Proteínas , Interfaz Usuario-Computador , Internet
10.
Nucleic Acids Res ; 52(3): 1090-1106, 2024 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-38055834

RESUMEN

Exonic sequences contain both protein-coding and RNA splicing information but the interplay of the protein and splicing code is complex and poorly understood. Here, we have studied traditional and auxiliary splicing codes of human exons that encode residues coordinating two essential divalent metals at the opposite ends of the Irving-Williams series, a universal order of relative stabilities of metal-organic complexes. We show that exons encoding Zn2+-coordinating amino acids are supported much less by the auxiliary splicing motifs than exons coordinating Ca2+. The handicap of the former is compensated by stronger splice sites and uridine-richer polypyrimidine tracts, except for position -3 relative to 3' splice junctions. However, both Ca2+ and Zn2+ exons exhibit close-to-constitutive splicing in multiple tissues, consistent with their critical importance for metalloprotein function and a relatively small fraction of expendable, alternatively spliced exons. These results indicate that constraints imposed by metal coordination spheres on RNA splicing have been efficiently overcome by the plasticity of exon-intron architecture to ensure adequate metalloprotein expression.


Asunto(s)
Calcio , Metaloproteínas , Empalme del ARN , Zinc , Humanos , Empalme Alternativo , Exones , Intrones , Metaloproteínas/genética , Sitios de Empalme de ARN
11.
Nucleic Acids Res ; 52(11): 6459-6471, 2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38726868

RESUMEN

CRISPR-Cas systems serve as adaptive immune systems in bacteria and archaea, protecting against phages and other mobile genetic elements. However, phages and archaeal viruses have developed countermeasures, employing anti-CRISPR (Acr) proteins to counteract CRISPR-Cas systems. Despite the revolutionary impact of CRISPR-Cas systems on genome editing, concerns persist regarding potential off-target effects. Therefore, understanding the structural and molecular intricacies of diverse Acrs is crucial for elucidating the fundamental mechanisms governing CRISPR-Cas regulation. In this study, we present the structure of AcrIIA28 from Streptococcus phage Javan 128 and analyze its structural and functional features to comprehend the mechanisms involved in its inhibition of Cas9. Our current study reveals that AcrIIA28 is a metalloprotein that contains Zn2+ and abolishes the cleavage activity of Cas9 only from Streptococcus pyrogen (SpyCas9) by directly interacting with the REC3 domain of SpyCas9. Furthermore, we demonstrate that the AcrIIA28 interaction prevents the target DNA from being loaded onto Cas9. These findings indicate the molecular mechanisms underlying AcrIIA28-mediated Cas9 inhibition and provide valuable insights into the ongoing evolutionary battle between bacteria and phages.


Asunto(s)
Proteína 9 Asociada a CRISPR , Sistemas CRISPR-Cas , Fagos de Streptococcus , Streptococcus , Proteína 9 Asociada a CRISPR/metabolismo , Proteína 9 Asociada a CRISPR/genética , Proteína 9 Asociada a CRISPR/química , ADN/metabolismo , ADN/genética , Edición Génica , Metaloproteínas/metabolismo , Metaloproteínas/genética , Metaloproteínas/química , Modelos Moleculares , Unión Proteica , Dominios Proteicos , Streptococcus/genética , Streptococcus/virología , Fagos de Streptococcus/genética , Fagos de Streptococcus/metabolismo , Proteínas Virales/metabolismo , Proteínas Virales/genética , Proteínas Virales/química , Zinc/metabolismo
12.
Proc Natl Acad Sci U S A ; 120(16): e2300137120, 2023 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-37036998

RESUMEN

Heme-containing integral membrane proteins are at the heart of many bioenergetic complexes and electron transport chains. The importance of these electron relay hubs across biology has inspired the design of de novo proteins that recreate their core features within robust, versatile, and tractable protein folds. To this end, we report here the computational design and in-cell production of a minimal diheme membrane cytochrome which successfully integrates into the cellular membrane of live bacteria. This synthetic construct emulates a four-helix bundle found in modern respiratory complexes but has no sequence homology to any polypeptide sequence found in nature. The two b-type hemes, which appear to be recruited from the endogenous heme pool, have distinct split redox potentials with values close to those of natural membrane-spanning cytochromes. The purified protein can engage in rapid biomimetic electron transport with small molecules, with other redox proteins, and with biologically relevant diffusive electron carriers. We thus report an artificial membrane metalloprotein with the potential to serve as a functional electron transfer module in both synthetic protocells and living systems.


Asunto(s)
Citocromos , Metaloproteínas , Citocromos/metabolismo , Oxidación-Reducción , Transporte de Electrón , Metaloproteínas/metabolismo , Hemo/metabolismo
13.
Proc Natl Acad Sci U S A ; 120(44): e2314788120, 2023 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-37871225

RESUMEN

Nitrogenase is an active target of heterologous expression because of its importance for areas related to agronomy, energy, and environment. One major hurdle for expressing an active Mo-nitrogenase in Escherichia coli is to generate the complex metalloclusters (P- and M-clusters) within this enzyme, which involves some highly unique bioinorganic chemistry/metalloenzyme biochemistry that is not generally dealt with in the heterologous expression of proteins via synthetic biology; in particular, the heterologous synthesis of the homometallic P-cluster ([Fe8S7]) and M-cluster core (or L-cluster; [Fe8S9C]) on their respective protein scaffolds, which represents two crucial checkpoints along the biosynthetic pathway of a complete nitrogenase, has yet to be demonstrated by biochemical and spectroscopic analyses of purified metalloproteins. Here, we report the heterologous formation of a P-cluster-containing NifDK protein upon coexpression of Azotobacter vinelandii nifD, nifK, nifH, nifM, and nifZ genes, and that of an L-cluster-containing NifB protein upon coexpression of Methanosarcina acetivorans nifB, nifS, and nifU genes alongside the A. vinelandii fdxN gene, in E. coli. Our metal content, activity, EPR, and XAS/EXAFS data provide conclusive evidence for the successful synthesis of P- and L-clusters in a nondiazotrophic host, thereby highlighting the effectiveness of our metallocentric, divide-and-conquer approach that individually tackles the key events of nitrogenase biosynthesis prior to piecing them together into a complete pathway for the heterologous expression of nitrogenase. As such, this work paves the way for the transgenic expression of an active nitrogenase while providing an effective tool for further tackling the biosynthetic mechanism of this important metalloenzyme.


Asunto(s)
Azotobacter vinelandii , Metaloproteínas , Nitrogenasa/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Fijación del Nitrógeno/genética , Oxidorreductasas/metabolismo , Metaloproteínas/metabolismo , Proteínas Bacterianas/metabolismo
14.
Brief Bioinform ; 24(1)2023 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-36642411

RESUMEN

Accurately predicting the interaction modes for metalloproteins remains extremely challenging in structure-based drug design and mechanism analysis of enzymatic catalysis due to the complexity of metal coordination in metalloproteins. Here, we report a docking method for metalloproteins based on geometric probability (GPDOCK) with unprecedented accuracy. The docking tests of 10 common metal ions with 9360 metalloprotein-ligand complexes demonstrate that GPDOCK has an accuracy of 94.3% in predicting binding pose. What is more, it can accurately realize the docking of metalloproteins with ligand when one or two water molecules are engaged in the metal ion coordination. Since GPDOCK only depends on the three-dimensional structure of metalloprotein and ligand, structure-based machine learning model is employed for the scoring of binding poses, which significantly improves computational efficiency. The proposed docking strategy can be an effective and efficient tool for drug design and further study of binding mechanism of metalloproteins. The manual of GPDOCK and the code for the logistical regression model used to re-rank the docking results are available at https://github.com/wangkai-zhku/GPDOCK.git.


Asunto(s)
Metaloproteínas , Metaloproteínas/química , Metaloproteínas/metabolismo , Unión Proteica , Ligandos , Aprendizaje Automático , Catálisis , Simulación del Acoplamiento Molecular , Sitios de Unión
15.
Nat Chem Biol ; 19(5): 548-555, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36593274

RESUMEN

Metal ions have various important biological roles in proteins, including structural maintenance, molecular recognition and catalysis. Previous methods of predicting metal-binding sites in proteomes were based on either sequence or structural motifs. Here we developed a co-evolution-based pipeline named 'MetalNet' to systematically predict metal-binding sites in proteomes. We applied MetalNet to proteomes of four representative prokaryotic species and predicted 4,849 potential metalloproteins, which substantially expands the currently annotated metalloproteomes. We biochemically and structurally validated previously unannotated metal-binding sites in several proteins, including apo-citrate lyase phosphoribosyl-dephospho-CoA transferase citX, an Escherichia coli enzyme lacking structural or sequence homology to any known metalloprotein (Protein Data Bank (PDB) codes: 7DCM and 7DCN ). MetalNet also successfully recapitulated all known zinc-binding sites from the human spliceosome complex. The pipeline of MetalNet provides a unique and enabling tool for interrogating the hidden metalloproteome and studying metal biology.


Asunto(s)
Metaloproteínas , Proteoma , Humanos , Secuencia de Aminoácidos , Proteoma/química , Metales/metabolismo , Metaloproteínas/metabolismo , Sitios de Unión , Escherichia coli/metabolismo , Aprendizaje Automático
16.
Nature ; 566(7742): 94-99, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30728519

RESUMEN

Small molecules containing the N-nitroso group, such as the bacterial natural product streptozotocin, are prominent carcinogens1,2 and important cancer chemotherapeutics3,4. Despite the considerable importance of this functional group to human health, enzymes dedicated to the assembly of the N-nitroso unit have not been identified. Here we show that SznF, a metalloenzyme from the biosynthesis of streptozotocin, catalyses an oxidative rearrangement of the guanidine group of Nω-methyl-L-arginine to generate an N-nitrosourea product. Structural characterization and mutagenesis of SznF reveal two separate active sites that promote distinct steps in this transformation using different iron-containing metallocofactors. This biosynthetic reaction, which has little precedent in enzymology or organic synthesis, expands the catalytic capabilities of non-haem-iron-dependent enzymes to include N-N bond formation. We find that biosynthetic gene clusters that encode SznF homologues are widely distributed among bacteria-including environmental organisms, plant symbionts and human pathogens-which suggests an unexpectedly diverse and uncharacterized microbial reservoir of bioactive N-nitroso metabolites.


Asunto(s)
Metaloproteínas/metabolismo , Estreptozocina/biosíntesis , Estreptozocina/química , Arginina/análogos & derivados , Dominio Catalítico/genética , Coenzimas/metabolismo , Cristalografía por Rayos X , Guanidina/metabolismo , Hierro/metabolismo , Metaloproteínas/química , Metaloproteínas/genética , Modelos Moleculares , Familia de Multigenes , Compuestos de Nitrosourea/metabolismo , Streptomyces/enzimología , Streptomyces/genética
17.
Nucleic Acids Res ; 51(D1): D593-D602, 2023 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-36243971

RESUMEN

Metalloenzymes are attractive research targets in fields of chemistry, biology, and medicine. Given that metalloenzymes can manifest conservation of metal-coordination and ligand binding modes, the excavation and expansion of metalloenzyme-specific knowledge is of interest in bridging metalloenzyme-related fields. Building on our previous metalloenzyme-ligand association database, MeLAD, we have expanded the scope of metalloenzyme-specific knowledge and services, by forming a versatile platform, termed the Metalloenzyme Data Bank and Analysis (MeDBA). The MeDBA provides: (i) manual curation of metalloenzymes into different categories, that this M-I, M-II and M-III; (ii) comprehensive information on metalloenzyme activities, expression profiles, family and disease links; (iii) structural information on metalloenzymes, in particular metal binding modes; (iv) metalloenzyme substrates and bioactive molecules acting on metalloenzymes; (v) excavated metal-binding pharmacophores and (vi) analysis tools for structure/metal active site comparison and metalloenzyme profiling. The MeDBA is freely available at https://medba.ddtmlab.org.


Asunto(s)
Bases de Datos de Proteínas , Metaloproteínas , Dominio Catalítico , Ligandos , Metaloproteínas/metabolismo , Metales , Enzimas
18.
Chem Soc Rev ; 53(1): 137-162, 2024 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-38018371

RESUMEN

Natural metalloenzymes with astonishing reaction activity and specificity underpin essential life transformations. Nevertheless, enzymes only operate under mild conditions to keep sophisticated structures active, limiting their potential applications. Artificial metalloenzymes that recapitulate the catalytic activity of enzymes can not only circumvent the enzymatic fragility but also bring versatile functions into practice. Among them, metal-organic frameworks (MOFs) featuring diverse and site-isolated metal sites and supramolecular structures have emerged as promising candidates for metalloenzymes to move toward unparalleled properties and behaviour of enzymes. In this review, we systematically summarize the significant advances in MOF-based metalloenzyme mimics with a special emphasis on active pocket engineering at the atomic level, including primary catalytic sites and secondary coordination spheres. Then, the deep understanding of catalytic mechanisms and their advanced applications are discussed. Finally, a perspective on this emerging frontier research is provided to advance bioinspired catalysis.


Asunto(s)
Estructuras Metalorgánicas , Metaloproteínas , Estructuras Metalorgánicas/química , Metaloproteínas/química , Catálisis , Metales/química , Dominio Catalítico
19.
Biochemistry ; 63(16): 2040-2050, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39088332

RESUMEN

Hydrogen-bonding (H-bonding) interactions in metalloprotein active sites can critically regulate enzyme function. Changes in the protein structure triggered by interplay with substrates, products, and partner proteins are often translated to the metallocofactor by way of specific changes in H-bond networks connected to the active site. However, the complexities of metalloprotein architecture and mechanism often preclude our ability to define the precise molecular interactions giving rise to these intricate regulatory pathways. To address this shortcoming, we have developed conformationally switchable artificial metalloproteins (swArMs) in which allosteric Gln-binding triggers protein conformational changes that impact the microenvironment surrounding an installed metallocofactor. Herein, we report a combined structural, spectroscopic, and computational approach to enhance the conformation-dependent changes in H-bond interactions surrounding the metallocofactor site of a swArM. Structure-informed molecular dynamics simulations were employed to predict point mutations that could enhance active site H-bond interactions preferentially in the Gln-bound holo-conformation of the swArM. Testing our predictions via the unique infrared spectral signals associated with the metallocofactor site, we have identified three key residues capable of imparting conformational control over the metallocofactor microenvironment. The resultant swArMs not only model biologically relevant structural regulation but also provide an enhanced Gln-responsive biological probe to be leveraged in future biosensing applications.


Asunto(s)
Enlace de Hidrógeno , Metaloproteínas , Simulación de Dinámica Molecular , Conformación Proteica , Metaloproteínas/química , Metaloproteínas/metabolismo , Metaloproteínas/genética , Dominio Catalítico
20.
Biochemistry ; 63(3): 339-347, 2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38232298

RESUMEN

Metalloproteins play fundamental roles in organisms and are utilized as starting points for the directed evolution of artificial enzymes. Knowing the strategies of metalloproteins, by which they exquisitely tune their activities, will not only lead to an understanding of biochemical phenomena but also contribute to various applications. The blue copper protein (BCP) has been a renowned model system to understand the biology, chemistry, and physics of metalloproteins. Pseudoazurin (Paz), a blue copper protein, mediates electron transfer in the bacterial anaerobic respiratory chain. Its redox potential is finely tuned by hydrogen (H) bond networks; however, difficulty in visualizing H atom positions in the protein hinders the detailed understanding of the protein's structure-function relationship. We here used neutron and sub-ångström resolution X-ray crystallography to directly observe H atoms in Paz. The 0.86-Å-resolution X-ray structure shows that the peptide bond between Pro80 and the His81 Cu ligand deviates from the ideal planar structure. The 1.9-Å-resolution neutron structure confirms a long-overlooked H bond formed by the amide of His81 and the S atom of another Cu ligand Cys78. Quantum mechanics/molecular mechanics calculations show that this H bond increases the redox potential of the Cu site and explains the experimental results well. Our study demonstrates the potential of neutron and sub-ångström resolution X-ray crystallography to understand the chemistry of metalloproteins at atomic and quantum levels.


Asunto(s)
Cobre , Metaloproteínas , Cobre/metabolismo , Cristalografía por Rayos X , Enlace de Hidrógeno , Ligandos , Modelos Moleculares , Metaloproteínas/metabolismo , Neutrones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA