Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
J Am Chem Soc ; 146(33): 23449-23456, 2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-39133525

RESUMEN

Natural products are important precursors for antibiotic drug design. These chemical scaffolds serve as synthetic inspiration for chemists who leverage their structures to develop novel antibacterials and chemical probes. We have previously studied carolacton, a natural product macrolactone fromSorangium cellulosum, and discovered a simplified derivative, A2, that maintained apparent biofilm inhibitory activity, although the biological target was unknown. Herein, we utilize affinity-based protein profiling (AfBPP) in situ during biofilm formation to identify the protein target using a photoexcitable cross-linking derivative of A2. From these studies, we identified glucan binding protein B (GbpB), a peptidoglycan hydrolase, as the primary target of A2. Further characterization of the interaction between A2 and GbpB, as well as PcsB, a closely related homologue from the more pathogenic S. pneumoniae, revealed binding to the catalytic CHAP (cysteine, histidine, aminopeptidase) domain. To the best of our knowledge, this is the first report of a small-molecule binder of a conserved and essential bacterial CHAP hydrolase, revealing its potential as an antibiotic target. This work also highlights A2 as a useful tool compound for streptococci and as an initial scaffold for the design of more potent CHAP binders.


Asunto(s)
Biopelículas , Biopelículas/efectos de los fármacos , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/química , Antibacterianos/farmacología , Antibacterianos/química , Sondas Moleculares/química , Sondas Moleculares/metabolismo , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , N-Acetil Muramoil-L-Alanina Amidasa/química , Lactonas/química , Lactonas/metabolismo , Lactonas/farmacología , Hidrolasas/metabolismo , Hidrolasas/química , Hidrolasas/antagonistas & inhibidores
2.
Nat Commun ; 15(1): 6734, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39112491

RESUMEN

Staphylococcus aureus is responsible for a substantial number of invasive infections globally each year. These infections are problematic because they are frequently recalcitrant to antibiotic treatment. Antibiotic tolerance, the ability of bacteria to persist despite normally lethal doses of antibiotics, contributes to antibiotic treatment failure in S. aureus infections. To understand how antibiotic tolerance is induced, S. aureus biofilms exposed to multiple anti-staphylococcal antibiotics are examined using both quantitative proteomics and transposon sequencing. These screens indicate that arginine metabolism is involved in antibiotic tolerance within a biofilm and support the hypothesis that depletion of arginine within S. aureus communities can induce antibiotic tolerance. Consistent with this hypothesis, inactivation of argH, the final gene in the arginine synthesis pathway, induces antibiotic tolerance. Arginine restriction induces antibiotic tolerance via inhibition of protein synthesis. In murine skin and bone infection models, an argH mutant has enhanced ability to survive antibiotic treatment with vancomycin, highlighting the relationship between arginine metabolism and antibiotic tolerance during S. aureus infection. Uncovering this link between arginine metabolism and antibiotic tolerance has the potential to open new therapeutic avenues targeting previously recalcitrant S. aureus infections.


Asunto(s)
Antibacterianos , Arginina , Biopelículas , Infecciones Estafilocócicas , Staphylococcus aureus , Arginina/metabolismo , Antibacterianos/farmacología , Animales , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Ratones , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Vancomicina/farmacología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Femenino , Farmacorresistencia Bacteriana/genética , Farmacorresistencia Bacteriana/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Hidrolasas/metabolismo , Hidrolasas/genética , Proteómica
3.
J Hazard Mater ; 477: 135380, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39088944

RESUMEN

The enzymatic degradation of plastic offers a green, sustainable strategy and scalable circular carbon route for solving polyester waste. Among the earlies discovered plastic-degrading enzymes are PET hydrolase (PETase) and MHET hydrolase (MHETase), which act synergistically. To promote the adsorption of enzymes on PET surfaces, increase their robustness, and enable directly depolymerization, we designed hydrophobin HFBI fused-PETase and MHETase. A customized self-assembled synergistic biocatalyst (MC@CaZn-MOF) was further developed to promote the two-step depolymerization process. The tailored catalysts showed better adhesion to the PET surface and desirable durability, retaining over 70% relative activity after incubation at pH 8.0 and 60 °C for 120 h. Importantly, MC@CaZn-MOF could directly decompose untreated AGf-PET to generate 9.5 mM TPA with weight loss over 90%. The successful implementation of a bifunctional customized catalyst makes the large-scale biocatalytic degradation of PET feasible, contributing to polymer upcycling and environmental sustainability.


Asunto(s)
Biocatálisis , Polimerizacion , Plásticos/química , Hidrolasas/metabolismo , Hidrolasas/química , Biodegradación Ambiental , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Estructuras Metalorgánicas/química
4.
Nat Commun ; 15(1): 7068, 2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-39152129

RESUMEN

Laboratory evolution studies have demonstrated that parallel evolutionary trajectories can lead to genetically distinct enzymes with high activity towards a non-preferred substrate. However, it is unknown whether such enzymes have convergent conformational dynamics and mechanistic features. To address this question, we use as a model the wild-type Homo sapiens kynureninase (HsKYNase), which is of great interest for cancer immunotherapy. Earlier, we isolated HsKYNase_66 through an unusual evolutionary trajectory, having a 410-fold increase in the kcat/KM for kynurenine (KYN) and reverse substrate selectivity relative to HsKYNase. Here, by following a different evolutionary trajectory we generate a genetically distinct variant, HsKYNase_93D9, that exhibits KYN catalytic activity comparable to that of HsKYNase_66, but instead it is a "generalist" that accepts 3'-hydroxykynurenine (OH-KYN) with the same proficiency. Pre-steady-state kinetic analysis reveals that while the evolution of HsKYNase_66 is accompanied by a change in the rate-determining step of the reactions, HsKYNase_93D9 retains the same catalytic mechanism as HsKYNase. HDX-MS shows that the conformational dynamics of the two enzymes are markedly different and distinct from ortholog prokaryotic enzymes with high KYN activity. Our work provides a mechanistic framework for understanding the relationship between evolutionary mechanisms and phenotypic traits of evolved generalist and specialist enzyme species.


Asunto(s)
Evolución Molecular , Hidrolasas , Quinurenina , Especificidad por Sustrato , Hidrolasas/química , Hidrolasas/metabolismo , Hidrolasas/genética , Humanos , Quinurenina/metabolismo , Quinurenina/química , Cinética , Conformación Proteica
5.
Arch Microbiol ; 206(9): 380, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-39143366

RESUMEN

Haloalkane dehalogenase, LinB, is a member of the α/ß hydrolase family of enzymes. It has a wide range of halogenated substrates, but, has been mostly studied in context of degradation of hexachlorocyclohexane (HCH) isomers, especially ß-HCH (5-12% of total HCH isomers), which is the most recalcitrant and persistent among all the HCH isomers. LinB was identified to directly act on ß-HCH in a one or two step transformation which decreases its toxicity manifold. Thereafter, many studies focused on LinB including its structure determination using X-ray crystallographic studies, structure comparison with other haloalkane dehalogenases, substrate specificity and kinetic studies, protein engineering and site-directed mutagenesis studies in search of better catalytic activity of the enzyme. LinB was mainly identified and characterized in bacteria belonging to sphingomonads. Detailed sequence comparison of LinB from different sphingomonads further revealed the residues critical for its activity and ability to catalyze either one or two step transformation of ß-HCH. Association of LinB with IS6100 elements is also being discussed in detail in sphingomonads. In this review, we summarized vigorous efforts done by different research groups on LinB for developing better bioremediation strategies against HCH contamination. Also, kinetic studies, protein engineering and site directed mutagenesis studies discussed here forms the basis of further exploration of LinB's role as an efficient enzyme in bioremediation projects.


Asunto(s)
Hexaclorociclohexano , Hidrolasas , Hidrolasas/metabolismo , Hidrolasas/genética , Hidrolasas/química , Hexaclorociclohexano/metabolismo , Especificidad por Sustrato , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/química , Cinética , Biodegradación Ambiental , Cristalografía por Rayos X , Mutagénesis Sitio-Dirigida , Sphingomonas/enzimología , Sphingomonas/genética , Sphingomonas/metabolismo
6.
Molecules ; 29(15)2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-39125019

RESUMEN

Identifying the catalytic regioselectivity of enzymes remains a challenge. Compared to experimental trial-and-error approaches, computational methods like molecular dynamics simulations provide valuable insights into enzyme characteristics. However, the massive data generated by these simulations hinder the extraction of knowledge about enzyme catalytic mechanisms without adequate modeling techniques. Here, we propose a computational framework utilizing graph-based active learning from molecular dynamics to identify the regioselectivity of ginsenoside hydrolases (GHs), which selectively catalyze C6 or C20 positions to obtain rare deglycosylated bioactive compounds from Panax plants. Experimental results reveal that the dynamic-aware graph model can excellently distinguish GH regioselectivity with accuracy as high as 96-98% even when different enzyme-substrate systems exhibit similar dynamic behaviors. The active learning strategy equips our model to work robustly while reducing the reliance on dynamic data, indicating its capacity to mine sufficient knowledge from short multi-replica simulations. Moreover, the model's interpretability identified crucial residues and features associated with regioselectivity. Our findings contribute to the understanding of GH catalytic mechanisms and provide direct assistance for rational design to improve regioselectivity. We presented a general computational framework for modeling enzyme catalytic specificity from simulation data, paving the way for further integration of experimental and computational approaches in enzyme optimization and design.


Asunto(s)
Ginsenósidos , Simulación de Dinámica Molecular , Ginsenósidos/química , Ginsenósidos/metabolismo , Especificidad por Sustrato , Hidrolasas/química , Hidrolasas/metabolismo , Panax/química , Panax/enzimología
7.
Int J Mol Sci ; 25(15)2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39125688

RESUMEN

Polyethylene terephthalate (PET) degradation by enzymatic hydrolysis is significant for addressing plastic pollution and fostering sustainable waste management practices. Identifying thermophilic and thermostable PET hydrolases is particularly crucial for industrial bioprocesses, where elevated temperatures may enhance enzymatic efficiency and process kinetics. In this study, we present the discovery of a novel thermophilic and thermostable PETase enzyme named Sis, obtained through metagenomic sequence-based analysis. Sis exhibits robust activity on nanoPET substrates, demonstrating effectiveness at temperatures up to 70 °C and displaying exceptional thermal stability with a melting temperature (Tm) of 82 °C. Phylogenetically distinct from previously characterised PET hydrolases, Sis represents a valuable addition to the repertoire of enzymes suitable for PET degradation.


Asunto(s)
Estabilidad de Enzimas , Tereftalatos Polietilenos , Tereftalatos Polietilenos/química , Tereftalatos Polietilenos/metabolismo , Hidrólisis , Filogenia , Temperatura , Especificidad por Sustrato , Cinética , Hidrolasas/química , Hidrolasas/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética
8.
Molecules ; 29(14)2024 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-39064965

RESUMEN

The ability of Mycobacterium tuberculosis to derive lipids from the host, store them intracellularly, and then break them down into energy requires a battery of serine hydrolases. Serine hydrolases are a large, diverse enzyme family with functional roles in dormant, active, and reactivating mycobacterial cultures. To rapidly measure substrate-dependent shifts in mycobacterial serine hydrolase activity, we combined a robust mycobacterial growth system of nitrogen limitation and variable carbon availability with nimble in-gel fluorogenic enzyme measurements. Using this methodology, we rapidly analyzed a range of ester substrates, identified multiple hydrolases concurrently, observed functional enzyme shifts, and measured global substrate preferences. Within every growth condition, mycobacterial hydrolases displayed the full, dynamic range of upregulated, downregulated, and constitutively active hydrolases independent of the ester substrate. Increasing the alkyl chain length of the ester substrate also allowed visualization of distinct hydrolase activity likely corresponding with lipases most responsible for lipid breakdown. The most robust expression of hydrolase activity was observed under the highest stress growth conditions, reflecting the induction of multiple metabolic pathways scavenging for energy to survive under this high stress. The unique hydrolases present under these high-stress conditions could represent novel drug targets for combination treatment with current front-line therapeutics. Combining diverse fluorogenic esters with in-gel activity measurements provides a rapid, customizable, and sensitive detection method for mycobacterial serine hydrolase activity.


Asunto(s)
Hidrolasas , Mycobacterium tuberculosis , Mycobacterium tuberculosis/enzimología , Hidrolasas/metabolismo , Especificidad por Sustrato , Proteínas Bacterianas/metabolismo , Serina/metabolismo , Pruebas de Enzimas/métodos
9.
Anal Biochem ; 693: 115598, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38964700

RESUMEN

The widespread use of polyamides such as nylons has led to the accumulation of nylon waste, which is particularly resistant to decomposition due to the intrinsic stability of the amide bond. New methods are required for the true recycling of these waste materials by depolymerization. Enzymes that are capable of hydrolyzing polyamides have been proposed as biocatalysts that may be suitable for this application. NylC is an enzyme that can mediate the hydrolysis of aminohexanoic acid oligomers, and to some extent, bulk polymers. However, current assays to characterize the activity of this enzyme require long reaction times and/or rely on secondary reactions to quantify hydrolysis. Herein, we have designed structurally-optimized small molecule chromogenic esters that serve as substrate analogues for monitoring NylC acyltransferase activity in a continuous manner. This assay can be performed in minutes at room temperature, and the substrate N-acetyl-GABA-pNP ester (kcat = 0.37 s-1, KM = 256 µM) shows selectivity for NylC in complex biological media. We also demonstrate that activity towards this substrate analogue correlates with amide hydrolysis, which is the primary activity of this enzyme. Furthermore, our screening of substrate analogues provides insight into the substrate specificity of NylC, which is relevant to biocatalytic applications.


Asunto(s)
Nylons , Nylons/química , Nylons/metabolismo , Hidrólisis , Especificidad por Sustrato , Hidrolasas/metabolismo , Hidrolasas/química , Aciltransferasas/metabolismo , Aciltransferasas/química , Aciltransferasas/análisis , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/química
10.
J Agric Food Chem ; 72(32): 18146-18154, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-39075026

RESUMEN

Zearalenone (ZEN) is an estrogenic mycotoxin causing reproductive toxicity in livestock. Currently, lactone hydrolases are used in the enzymatic degradation of ZEN. However, most lactone hydrolases suffer from low degradation efficiency and poor thermal stability. ZHD518, as a documented neutral enzyme for ZEN degradation, exhibits high enzymatic activity under neutral conditions. In this study, a multifunctional peptide S1v1-(AEAEAHAH)2 was fused to the N-terminus of ZHD518. Compared with the wild-type enzyme, the peptide fusion significantly enhanced protein expression by 1.28 times, enzyme activity by 9.27 times, thermal stability by 37.08 times after incubation at 45 °C for 10 min and enzyme stability during long-term storage. Moreover, ZEN concentrations in corn bran, corn germ meal, and corn gluten powder decreased from 5.29 ± 0.04, 5.31 ± 0.03, and 5.30 ± 0.01 µg/g to 0.48 ± 0.05, 0.48 ± 0.06, and 0.21 ± 0.04 µg/g, respectively, following a 60 min treatment with S1v1-GS-ZHD518, resulting in degradation rates of 90.98, 91.00, and 95.32%, respectively. In conclusion, the properties of S1v1-GS-ZHD518, such as its efficient degradability, high temperature resistance and storage resistance, offer the possibility of its application in food or feed.


Asunto(s)
Estabilidad de Enzimas , Péptidos , Zea mays , Zearalenona , Zearalenona/química , Zearalenona/metabolismo , Zea mays/química , Zea mays/metabolismo , Zea mays/genética , Péptidos/química , Péptidos/metabolismo , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Hidrolasas/genética , Hidrolasas/metabolismo , Hidrolasas/química , Lactonas/química , Lactonas/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/genética
11.
Anal Biochem ; 694: 115631, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39084336

RESUMEN

Cyclic 3-phosphosphoglyceric anhydride (cPGA), a side product of glycolysis, acylates cellular amines and thiols to form amides and thioesters, respectively. Since these acylation reactions are harmful, organisms rely on a protein, known as DJ-1 in humans, to inactivate cPGA. Inactivation of cPGA likely plays a significant role in cytoprotection by DJ-1, but further progress in this direction is hampered by the lack of quantitative assays to measure the cPGA hydrolase activity of DJ-1 in biological samples. Here we report an optimized procedure for preparation of cPGA which is then used as a substrate to quantify enzymatic activity of DJ-1. The end-point assay for cPGA hydrolase uses dilute cell lysates to hydrolyze cPGA for 0.5-3.5 min followed by conversion of the remaining cPGA into thioester for spectrophotometric quantitation. We illustrate the utility of this assay by showing that higher levels of cPGA hydrolase activity result in better protection from acylation by cPGA. Moreover, the decrease of cPGA hydrolase activity due to oxidation of the catalytic cysteine of DJ-1 under oxidative stress and its subsequent recovery can be monitored using the assay. This relatively simple assay allows functional characterization of DJ-1 in biological samples through quantitative assessment of its cPGA hydrolase activity.


Asunto(s)
Estrés Oxidativo , Proteína Desglicasa DJ-1 , Proteína Desglicasa DJ-1/metabolismo , Humanos , Hidrolasas/metabolismo , Pruebas de Enzimas/métodos
12.
Nature ; 631(8022): 884-890, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39020178

RESUMEN

Plastic production reached 400 million tons in 2022 (ref. 1), with packaging and single-use plastics accounting for a substantial amount of this2. The resulting waste ends up in landfills, incineration or the environment, contributing to environmental pollution3. Shifting to biodegradable and compostable plastics is increasingly being considered as an efficient waste-management alternative4. Although polylactide (PLA) is the most widely used biosourced polymer5, its biodegradation rate under home-compost and soil conditions remains low6-8. Here we present a PLA-based plastic in which an optimized enzyme is embedded to ensure rapid biodegradation and compostability at room temperature, using a scalable industrial process. First, an 80-fold activity enhancement was achieved through structure-based rational engineering of a new hyperthermostable PLA hydrolase. Second, the enzyme was uniformly dispersed within the PLA matrix by means of a masterbatch-based melt extrusion process. The liquid enzyme formulation was incorporated in polycaprolactone, a low-melting-temperature polymer, through melt extrusion at 70 °C, forming an 'enzymated' polycaprolactone masterbatch. Masterbatch pellets were integrated into PLA by melt extrusion at 160 °C, producing an enzymated PLA film (0.02% w/w enzyme) that fully disintegrated under home-compost conditions within 20-24 weeks, meeting home-composting standards. The mechanical and degradation properties of the enzymated film were compatible with industrial packaging applications, and they remained intact during long-term storage. This innovative material not only opens new avenues for composters and biomethane production but also provides a feasible industrial solution for PLA degradation.


Asunto(s)
Plásticos Biodegradables , Biodegradación Ambiental , Enzimas Inmovilizadas , Hidrolasas , Poliésteres , Ingeniería de Proteínas , Plásticos Biodegradables/química , Plásticos Biodegradables/metabolismo , Enzimas Inmovilizadas/química , Enzimas Inmovilizadas/metabolismo , Hidrolasas/metabolismo , Hidrolasas/química , Poliésteres/química , Poliésteres/metabolismo , Suelo/química , Temperatura , Estabilidad de Enzimas , Compostaje
13.
Appl Microbiol Biotechnol ; 108(1): 404, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38953996

RESUMEN

Polyethylene terephthalate (PET) is a major component of plastic waste. Enzymatic PET hydrolysis is the most ecofriendly recycling technology. The biorecycling of PET waste requires the complete depolymerization of PET to terephthalate and ethylene glycol. The history of enzymatic PET depolymerization has revealed two critical issues for the industrial depolymerization of PET: industrially available PET hydrolases and pretreatment of PET waste to make it susceptible to full enzymatic hydrolysis. As none of the wild-type enzymes can satisfy the requirements for industrialization, various mutational improvements have been performed, through classical technology to state-of-the-art computational/machine-learning technology. Recent engineering studies on PET hydrolases have brought a new insight that flexibility of the substrate-binding groove may improve the efficiency of PET hydrolysis while maintaining sufficient thermostability, although the previous studies focused only on enzymatic thermostability above the glass transition temperature of PET. Industrial biorecycling of PET waste is scheduled to be implemented, using micronized amorphous PET. Next stage must be the development of PET hydrolases that can efficiently degrade crystalline parts of PET and expansion of target PET materials, not only bottles but also textiles, packages, and microplastics. This review discusses the current status of PET hydrolases, their potential applications, and their profespectal goals. KEY POINTS: • PET hydrolases must be thermophilic, but their operation must be below 70 °C • Classical and state-of-the-art engineering approaches are useful for PET hydrolases • Enzyme activity on crystalline PET is most expected for future PET biorecycling.


Asunto(s)
Hidrolasas , Tereftalatos Polietilenos , Tereftalatos Polietilenos/metabolismo , Tereftalatos Polietilenos/química , Hidrolasas/metabolismo , Hidrolasas/química , Hidrolasas/genética , Hidrólisis , Ingeniería de Proteínas/métodos , Biodegradación Ambiental , Reciclaje
14.
MAbs ; 16(1): 2375798, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38984665

RESUMEN

Monoclonal antibodies (mAb) and other biological drugs are affected by enzymatic polysorbate (PS) degradation that reduces product stability and jeopardizes the supply of innovative medicines. PS represents a critical surfactant stabilizing the active pharmaceutical ingredients, which are produced by recombinant Chinese hamster ovary (CHO) cell lines. While the list of potential PS-degrading CHO host cell proteins (HCPs) has grown over the years, tangible data on industrially relevant HCPs are still scarce. By means of a highly sensitive liquid chromatography-tandem mass spectrometry method, we investigated seven different mAb products, resulting in the identification of 12 potentially PS-degrading hydrolases, including the strongly PS-degrading lipoprotein lipase (LPL). Using an LPL knockout CHO host cell line, we were able to stably overexpress and purify the remaining candidate hydrolases through orthogonal affinity chromatography methods, enabling their detailed functional characterization. Applying a PS degradation assay, we found nine mostly secreted, PS-active hydrolases with varying hydrolytic activity. All active hydrolases showed a serine-histidine-aspartate/glutamate catalytical triad. Further, we subjected the active hydrolases to pH-screenings and revealed a diverse range of activity optima, which can facilitate the identification of residual hydrolases during bioprocess development. Ultimately, we compiled our dataset in a risk matrix identifying PAF-AH, LIPA, PPT1, and LPLA2 as highly critical hydrolases based on their cellular expression, detection in purified antibodies, active secretion, and PS degradation activity. With this work, we pave the way toward a comprehensive functional characterization of PS-degrading hydrolases and provide a basis for a future reduction of PS degradation in biopharmaceutical drug products.


Asunto(s)
Anticuerpos Monoclonales , Cricetulus , Hidrolasas , Células CHO , Animales , Anticuerpos Monoclonales/química , Hidrolasas/metabolismo , Polisorbatos/química , Productos Biológicos/metabolismo , Humanos
15.
Biochemistry ; 63(15): 1901-1912, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-38995238

RESUMEN

Significant attention has been shifted toward the use and development of biodegradable polymeric materials to mitigate environmental accumulation and potential health impacts. One such material, poly(aspartic acid) (PAA), is a biodegradable alternative to superabsorbent poly(carboxylates), like poly(acrylate). Three enzymes are known to hydrolyze PAA: PahZ1KT-1 and PahZ2KT-1 from Sphingomonas sp. KT-1 and PahZ1KP-2 from Pedobacter sp. KP-2. We previously reported the X-ray crystal structure for PahZ1KT-1, which revealed a homodimer complex with a strongly cationic surface spanning one side of each monomer. Here, we report the first characterization of any polymer hydrolase binding to DNA, where modeling data predict binding of the polyanionic DNA near the cationic substrate binding surface. Our data reveal that PahZ1 homologues from Sphingomonas sp. KT-1 and Pedobacter sp. KP-2 bind ssDNA and dsDNA with nanomolar binding affinities. PahZ1KT-1 binds ssDNA and dsDNA with an apparent dissociation constant, KD,app = 81 ± 14 and 19 ± 1 nM, respectively, and these estimates are similar to the same behaviors exhibited by PahZ1KP-2. Gel permeation chromatography data reveal that dsDNA binding promotes inhibition of PahZ1-catalyzed PAA biodegradation for each homologue. We propose a working model wherein binding of PahZ1 to extracellular biofilm DNA aids in the localization of the hydrolase to the environment in which PAA would first be encountered, thereby providing a mechanism to degrade extracellular PAA and potentially harvest aspartic acid for nutritional uptake.


Asunto(s)
Sphingomonas , Sphingomonas/enzimología , Pedobacter/enzimología , ADN/metabolismo , Hidrolasas/metabolismo , Hidrolasas/química , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Péptidos/metabolismo , Péptidos/química , ADN de Cadena Simple/metabolismo , Modelos Moleculares , Unión Proteica , Ácido Aspártico/metabolismo , Ácido Aspártico/química
16.
Nat Commun ; 15(1): 6121, 2024 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-39033196

RESUMEN

The biguanide drug metformin is a first-line blood glucose-lowering medication for type 2 diabetes, leading to its presence in the global environment. However, little is known about the fate of metformin by microbial catabolism. Here, we characterize a Ni2+-dependent heterohexameric enzyme (MetCaCb) from the ureohydrolase superfamily, catalyzing the hydrolysis of metformin into guanylurea and dimethylamine. Either subunit alone is catalytically inactive, but together they work as an active enzyme highly specific for metformin. The crystal structure of the MetCaCb complex shows the coordination of the binuclear metal cluster only in MetCa, with MetCb as a protein binder of its active cognate. An in-silico search and functional assay discover a group of MetCaCb-like protein pairs exhibiting metformin hydrolase activity in the environment. Our findings not only establish the genetic and biochemical foundation for metformin catabolism but also provide additional insights into the adaption of the ancient enzymes toward newly occurred substrate.


Asunto(s)
Hidrolasas , Metformina , Níquel , Metformina/metabolismo , Metformina/química , Níquel/metabolismo , Níquel/química , Hidrolasas/metabolismo , Hidrolasas/química , Hidrolasas/genética , Cristalografía por Rayos X , Hidrólisis , Especificidad por Sustrato , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Modelos Moleculares
17.
Methods Mol Biol ; 2837: 199-206, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39044086

RESUMEN

Chimeric mouse models with a humanized liver (Hu-HEP mice) provide a unique tool to study human hepatotropic virus diseases, including viral infection, viral pathogenesis, and anti-viral therapy. Here, we describe a detailed protocol for studying hepatitis B infection in NRG-derived fumarylacetoacetate hydrolase (FAH) knockout mice repopulated with human hepatocytes (FRG-Hu HEP mice). The procedures include (1) maintenance and genotyping of the FRG mice, (2) intrasplenic injection of primary human hepatocytes (PHH), (3) 2-(2-nitro-4-fluoromethylbenzoyl)-1,3-cyclohexanedione (NTBC) drug reduction cycling to improve human hepatocyte repopulation, (4) human albumin detection, and (5) HBV infection and detection. The method is simple and allows for highly reproducible generation of FRG-Hu HEP mice for HBV infection and therapy investigations.


Asunto(s)
Modelos Animales de Enfermedad , Virus de la Hepatitis B , Hepatitis B , Hepatocitos , Hidrolasas , Hígado , Ratones Noqueados , Animales , Humanos , Ratones , Hidrolasas/genética , Hidrolasas/metabolismo , Hidrolasas/deficiencia , Hepatitis B/virología , Virus de la Hepatitis B/genética , Hígado/virología , Hígado/patología , Hepatocitos/virología , Hepatocitos/trasplante , Ratones Endogámicos NOD , Subunidad gamma Común de Receptores de Interleucina/genética , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Quimera , Ciclohexanonas , Nitrobenzoatos
18.
J Chem Theory Comput ; 20(14): 5807-5819, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38978395

RESUMEN

Because most proteins have buried active sites, protein tunnels or channels play a crucial role in the transport of small molecules into buried cavities for enzymatic catalysis. Tunnels can critically modulate the biological process of protein-ligand recognition. Various molecular dynamics methods have been developed for exploring and exploiting the protein-ligand conformational space to extract high-resolution details of the binding processes, a recent example being energetically unbiased high-throughput adaptive sampling simulations. The current study systematically contrasted the role of integrating prior knowledge while generating useful initial protein-ligand configurations, called seeds, for these simulations. Using a nontrivial system of a haloalkane dehalogenase mutant with multiple transport tunnels leading to a deeply buried active site, simulations were employed to derive kinetic models describing the process of association and dissociation of the substrate molecule. The most knowledge-based seed generation enabled high-throughput simulations that could more consistently capture the entire transport process, explore the complex network of transport tunnels, and predict equilibrium dissociation constants, koff/kon, on the same order of magnitude as experimental measurements. Overall, the infusion of more knowledge into the initial seeds of adaptive sampling simulations could render analyses of transport mechanisms in enzymes more consistent even for very complex biomolecular systems, thereby promoting drug development efforts and the rational design of enzymes with buried active sites.


Asunto(s)
Dominio Catalítico , Hidrolasas , Simulación de Dinámica Molecular , Ligandos , Hidrolasas/química , Hidrolasas/metabolismo , Cinética
19.
Front Cell Infect Microbiol ; 14: 1392249, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38915922

RESUMEN

In recent years, there has been increasing interest in studying gut microbiome-derived hydrolases in relation to oral drug metabolism, particularly focusing on natural product drugs. Despite the significance of natural product drugs in the field of oral medications, there is a lack of research on the regulatory interplay between gut microbiome-derived hydrolases and these drugs. This review delves into the interaction between intestinal microbiome-derived hydrolases and natural product drugs metabolism from three key perspectives. Firstly, it examines the impact of glycoside hydrolases, amide hydrolases, carboxylesterase, bile salt hydrolases, and epoxide hydrolase on the structure of natural products. Secondly, it explores how natural product drugs influence microbiome-derived hydrolases. Lastly, it analyzes the impact of interactions between hydrolases and natural products on disease development and the challenges in developing microbial-derived enzymes. The overarching goal of this review is to lay a solid theoretical foundation for the advancement of research and development in new natural product drugs and personalized treatment.


Asunto(s)
Productos Biológicos , Microbioma Gastrointestinal , Hidrolasas , Productos Biológicos/metabolismo , Productos Biológicos/farmacología , Humanos , Hidrolasas/metabolismo , Animales , Glicósido Hidrolasas/metabolismo , Bacterias/metabolismo , Bacterias/enzimología
20.
J Hazard Mater ; 474: 134838, 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-38850944

RESUMEN

Microplastics (MPs) pose an emerging threat to soil ecological function, yet effective solutions remain limited. This study introduces a novel approach using magnetic biochar immobilized PET hydrolase (MB-LCC-FDS) to degrade soil polyethylene terephthalate microplastics (PET-MPs). MB-LCC-FDS exhibited a 1.68-fold increase in relative activity in aquatic solutions and maintained 58.5 % residual activity after five consecutive cycles. Soil microcosm experiment amended with MB-LCC-FDS observed a 29.6 % weight loss of PET-MPs, converting PET into mono(2-hydroxyethyl) terephthalate (MHET). The generated MHET can subsequently be metabolized by soil microbiota to release terephthalic acid. The introduction of MB-LCC-FDS shifted the functional composition of soil microbiota, increasing the relative abundances of Microbacteriaceae and Skermanella while reducing Arthobacter and Vicinamibacteraceae. Metagenomic analysis revealed that MB-LCC-FDS enhanced nitrogen fixation, P-uptake and transport, and organic-P mineralization in PET-MPs contaminated soil, while weakening the denitrification and nitrification. Structural equation model indicated that changes in soil total carbon and Simpson index, induced by MB-LCC-FDS, were the driving factors for soil carbon and nitrogen transformation. Overall, this study highlights the synergistic role of magnetic biochar-immobilized PET hydrolase and soil microbiota in degrading soil PET-MPs, and enhances our understanding of the microbiome and functional gene responses to PET-MPs and MB-LCC-FDS in soil systems.


Asunto(s)
Carbón Orgánico , Hidrolasas , Fósforo , Tereftalatos Polietilenos , Microbiología del Suelo , Contaminantes del Suelo , Hidrolasas/metabolismo , Tereftalatos Polietilenos/química , Tereftalatos Polietilenos/metabolismo , Contaminantes del Suelo/metabolismo , Carbón Orgánico/química , Fósforo/metabolismo , Fósforo/química , Microplásticos/toxicidad , Biodegradación Ambiental , Enzimas Inmovilizadas/metabolismo , Enzimas Inmovilizadas/química , Nitrógeno/metabolismo , Ciclo del Nitrógeno , Microbiota/efectos de los fármacos , Bacterias/genética , Bacterias/metabolismo , Bacterias/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA