Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 180
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
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.
Nature ; 593(7859): 418-423, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33727703

RESUMEN

The COVID-19 pandemic is the third outbreak this century of a zoonotic disease caused by a coronavirus, following the emergence of severe acute respiratory syndrome (SARS) in 20031 and Middle East respiratory syndrome (MERS) in 20122. Treatment options for coronaviruses are limited. Here we show that clofazimine-an anti-leprosy drug with a favourable safety profile3-possesses inhibitory activity against several coronaviruses, and can antagonize the replication of SARS-CoV-2 and MERS-CoV in a range of in vitro systems. We found that this molecule, which has been approved by the US Food and Drug Administration, inhibits cell fusion mediated by the viral spike glycoprotein, as well as activity of the viral helicase. Prophylactic or therapeutic administration of clofazimine in a hamster model of SARS-CoV-2 pathogenesis led to reduced viral loads in the lung and viral shedding in faeces, and also alleviated the inflammation associated with viral infection. Combinations of clofazimine and remdesivir exhibited antiviral synergy in vitro and in vivo, and restricted viral shedding from the upper respiratory tract. Clofazimine, which is orally bioavailable and comparatively cheap to manufacture, is an attractive clinical candidate for the treatment of outpatients and-when combined with remdesivir-in therapy for hospitalized patients with COVID-19, particularly in contexts in which costs are an important factor or specialized medical facilities are limited. Our data provide evidence that clofazimine may have a role in the control of the current pandemic of COVID-19 and-possibly more importantly-in dealing with coronavirus diseases that may emerge in the future.


Asunto(s)
Antivirales/farmacología , Clofazimina/farmacología , Coronavirus/clasificación , Coronavirus/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Adenosina Monofosfato/uso terapéutico , Alanina/análogos & derivados , Alanina/farmacología , Alanina/uso terapéutico , Animales , Antiinflamatorios/farmacocinética , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Antivirales/farmacocinética , Antivirales/uso terapéutico , Disponibilidad Biológica , Fusión Celular , Línea Celular , Clofazimina/farmacocinética , Clofazimina/uso terapéutico , Coronavirus/crecimiento & desarrollo , Coronavirus/patogenicidad , Cricetinae , ADN Helicasas/antagonistas & inhibidores , Sinergismo Farmacológico , Femenino , Humanos , Estadios del Ciclo de Vida/efectos de los fármacos , Masculino , Mesocricetus , Profilaxis Pre-Exposición , SARS-CoV-2/crecimiento & desarrollo , Especificidad de la Especie , Glicoproteína de la Espiga del Coronavirus/antagonistas & inhibidores , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética
3.
Proc Natl Acad Sci U S A ; 121(4): e2311630121, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38232278

RESUMEN

Copper is an essential trace element for the human body, and its requirement for optimistic immune functions has been recognized for decades. How copper is involved in the innate immune pathway, however, remains to be clarified. Here, we report that copper serves as a signal molecule to regulate the kinase activity of alpha-kinase 1 (ALPK1), a cytosolic pattern-recognition receptor (PRR), and therefore promotes host cell defense against bacterial infection. We show that in response to infection, host cells actively accumulate copper in the cytosol, and the accumulated cytosolic copper enhances host cell defense against evading pathogens, including intracellular and, unexpectedly, extracellular bacteria. Subsequently, we demonstrate that copper activates the innate immune pathway of host cells in an ALPK1-dependent manner. Further mechanistic studies reveal that copper binds to ALPK1 directly and is essential for the kinase activity of this cytosolic PRR. Moreover, the binding of copper to ALPK1 enhances the sensitivity of ALPK1 to the bacterial metabolite ADP-heptose and eventually prompts host cells to elicit an enhanced immune response during bacterial infection. Finally, using a zebrafish in vivo model, we show that a copper-treated host shows an increased production of proinflammatory cytokines, enhanced recruitment of phagosome cells, and promoted bacterial clearance. Our findings uncover a previously unrecognized role of copper in the modulation of host innate immune response against bacterial pathogens and advance our knowledge on the cross talk between cytosolic copper homeostasis and immune system.


Asunto(s)
Infecciones Bacterianas , Cobre , Animales , Humanos , Pez Cebra , Inmunidad Innata , Citocinas , Receptores de Reconocimiento de Patrones
4.
Proc Natl Acad Sci U S A ; 119(11): e2119417119, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35263219

RESUMEN

Colistin is considered the last-line antimicrobial for the treatment of multidrug-resistant gram-negative bacterial infections. The emergence and spread of superbugs carrying the mobile colistin resistance gene (mcr) have become the most serious and urgent threat to healthcare. Here, we discover that silver (Ag+), including silver nanoparticles, could restore colistin efficacy against mcr-positive bacteria. We show that Ag+ inhibits the activity of the MCR-1 enzyme via substitution of Zn2+ in the active site. Unexpectedly, a tetra-silver center was found in the active-site pocket of MCR-1 as revealed by the X-ray structure of the Ag-bound MCR-1, resulting in the prevention of substrate binding. Moreover, Ag+effectively slows down the development of higher-level resistance and reduces mutation frequency. Importantly, the combined use of Ag+ at a low concentration with colistin could relieve dermonecrotic lesions and reduce the bacterial load of mice infected with mcr-1­carrying pathogens. This study depicts a mechanism of Ag+ inhibition of MCR enzymes and demonstrates the potentials of Ag+ as broad-spectrum inhibitors for the treatment of mcr-positive bacterial infection in combination with colistin.


Asunto(s)
Antibacterianos , Colistina , Farmacorresistencia Bacteriana Múltiple , Proteínas de Escherichia coli , Escherichia coli , Plata , Antibacterianos/farmacología , Colistina/farmacología , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Farmacorresistencia Bacteriana Múltiple/genética , Escherichia coli/efectos de los fármacos , Escherichia coli/enzimología , Escherichia coli/genética , Proteínas de Escherichia coli/antagonistas & inhibidores , Proteínas de Escherichia coli/genética , Pruebas de Sensibilidad Microbiana , Plásmidos/genética , Plata/farmacología
5.
Clin Immunol ; 263: 110205, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38575044

RESUMEN

Increasing clinical data show that the imbalance of host metallome is closely associated with different kinds of disease, however, the intrinsic mechanisms of action of metals in immunity and pathogenesis of disease remain largely undefined. There is lack of multiplexed profiling system to integrate the metalloproteome-immunoproteome information at systemic level for exploring the roles of metals in immunity and disease pathogenesis. In this study, we build up a metal-coding assisted multiplexed proteome assay platform for serum metalloproteomic and immunoproteomic profiling. By taking COVID-19 as a showcase, we unbiasedly uncovered the most evident modulation of iron-related proteins, i.e., Ft and Tf, in serum of severe COVID-19 patients, and the value of Ft/Tf could work as a robust biomarker for COVID-19 severity stratification, which overtakes the well-established clinical risk factors (cytokines). We further uncovered a tight association of transferrin with inflammation mediator IL-10 in COVID-19 patients, which was proved to be mainly governed by the monocyte/macrophage of liver, shedding light on new pathophysiological and immune regulatory mechanisms of COVID-19 disease. We finally validated the beneficial effects of iron chelators as anti-viral agents in SARS-CoV-2-infected K18-hACE2 mice through modulation of iron dyshomeostasis and alleviating inflammation response. Our findings highlight the critical role of liver-mediated iron dysregulation in COVID-19 disease severity, providing solid evidence on the involvement of iron-related proteins in COVID-19 pathophysiology and immunity.


Asunto(s)
COVID-19 , Hierro , Proteoma , SARS-CoV-2 , COVID-19/inmunología , Humanos , Animales , SARS-CoV-2/inmunología , Ratones , Hierro/metabolismo , Proteómica/métodos , Transferrina/metabolismo , Metaloproteínas/inmunología , Metaloproteínas/metabolismo , Masculino , Femenino , Biomarcadores/sangre , Biomarcadores/metabolismo , Quelantes del Hierro/uso terapéutico , Quelantes del Hierro/farmacología , Interleucina-10/inmunología , Interleucina-10/metabolismo , Persona de Mediana Edad
6.
J Biol Chem ; 298(3): 101587, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35032550

RESUMEN

Catabolite control protein A (CcpA) of the human pathogen Staphylococcus aureus is an essential DNA regulator for carbon catabolite repression and virulence, which facilitates bacterial survival and adaptation to a changing environment. Here, we report that copper (II) signaling mediates the DNA-binding capability of CcpA in vitro and in vivo. Copper (II) catalyzes the oxidation of two cysteine residues (Cys216 and Cys242) in CcpA to form intermolecular disulfide bonds between two CcpA dimers, which results in the formation and dissociation of a CcpA tetramer of CcpA from its cognate DNA promoter. We further demonstrate that the two cysteine residues on CcpA are important for S. aureus to resist host innate immunity, indicating that S. aureus CcpA senses the redox-active copper (II) ions as a natural signal to cope with environmental stress. Together, these findings reveal a novel regulatory mechanism for CcpA activity through copper (II)-mediated oxidation.


Asunto(s)
Proteínas Bacterianas , Cobre , ADN Bacteriano , Proteínas Represoras , Staphylococcus aureus , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Cationes Bivalentes , Cobre/química , Cobre/metabolismo , Cisteína/química , Cisteína/metabolismo , ADN Bacteriano/metabolismo , Oxidación-Reducción , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Staphylococcus aureus/metabolismo
7.
J Biol Inorg Chem ; 28(2): 225-234, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36662362

RESUMEN

The emergence and rapid spread of the mobile colistin resistance gene mcr-1 among bacterial species and hosts significantly challenge the efficacy of "last-line" antibiotic colistin. Previously, we reported silver nitrate and auranofin serve as colistin adjuvants for combating mcr-1-positive bacteria. Herein, we uncovered more gold-based drugs and nanoparticles, and found that they exhibited varying degree of synergisms with colistin on killing mcr-1-positive bacteria. However, pre-activation of the drugs by either glutathione or N-acetyl cysteine, thus releasing and accumulating gold ions, is perquisite for their abilities to substitute zinc cofactor from MCR-1 enzyme. X-ray crystallography and biophysical studies further supported the proposed mechanism. This study not only provides basis for combining gold-based drugs and colistin for combating mcr-1-positive bacterial infections, but also undoubtedly opens a new horizon for metabolism details of gold-based drugs in overcoming antimicrobial resistance.


Asunto(s)
Colistina , Proteínas de Escherichia coli , Colistina/farmacología , Antibacterianos/farmacología , Bacterias , Oro/farmacología , Farmacorresistencia Bacteriana/genética , Plásmidos , Proteínas de Escherichia coli/química , Pruebas de Sensibilidad Microbiana
8.
PLoS Biol ; 17(6): e3000292, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31181061

RESUMEN

Despite the broad-spectrum antimicrobial activities of silver, its internal usage is restricted, owing to the toxicity. Strategies to enhance its efficacy are highly desirable but rely heavily on the understanding of its molecular mechanism of action. However, up to now, no direct silver-targeting proteins have been mined at a proteome-wide scale, which hinders systemic studies on the biological pathways interrupted by silver. Herein, we build up a unique system, namely liquid chromatography gel electrophoresis inductively coupled plasma mass spectrometry (LC-GE-ICP-MS), allowing 34 proteins directly bound by silver ions to be identified in Escherichia coli. By using integrated omic approaches, including metalloproteomics, metabolomics, bioinformatics, and systemic biology, we delineated the first dynamic antimicrobial actions of silver (Ag+) in E. coli, i.e., it primarily damages multiple enzymes in glycolysis and tricarboxylic acid (TCA) cycle, leading to the stalling of the oxidative branch of the TCA cycle and an adaptive metabolic divergence to the reductive glyoxylate pathway. It then further damages the adaptive glyoxylate pathway and suppresses the cellular oxidative stress responses, causing systemic damages and death of the bacterium. To harness these novel findings, we coadministrated metabolites involved in the Krebs cycles with Ag+ and found that they can significantly potentiate the efficacy of silver both in vitro and in an animal model. Our study reveals the comprehensive and dynamic mechanisms of Ag+ toxicity in E. coli cells and offers a novel and general approach for deciphering molecular mechanisms of metallodrugs in various pathogens and cells to facilitate the development of new therapeutics.


Asunto(s)
Biología Computacional/métodos , Escherichia coli/metabolismo , Plata/metabolismo , Plata/uso terapéutico , Antibacterianos/farmacología , Antiinfecciosos , Bacterias , Cromatografía Liquida/métodos , Proteínas de Escherichia coli/metabolismo , Espectrometría de Masas/métodos , Metabolómica , Proteómica
9.
Chem Soc Rev ; 50(21): 12037-12069, 2021 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-34533144

RESUMEN

Bismuth as a relatively non-toxic and inexpensive metal with exceptional properties has numerous biomedical applications. Bismuth-based compounds are used extensively as medicines for the treatment of gastrointestinal disorders including dyspepsia, gastric ulcers and H. pylori infections. Recently, its medicinal application was further extended to potential treatments of viral infection, multidrug resistant microbial infections, cancer and also imaging, drug delivery and biosensing. In this review we have highlighted the unique chemistry and biological chemistry of bismuth-209 as a prelude to sections covering the unique antibacterial activity of bismuth including a description of research undertaken to date to elucidate key molecular mechanisms of action against H. pylori, the development of novel compounds to treat infection from microbes beyond H. pylori and the significant role bismuth compounds can play as resistance breakers. Furthermore we have provided an account of the potential therapeutic application of bismuth-213 in targeted alpha therapy as well as a summary of the biomedical applications of bismuth-based nanoparticles and composites. Ultimately this review aims to provide the state of the art, highlight the untapped biomedical potential of bismuth and encourage original contributions to this exciting and important field.


Asunto(s)
Helicobacter pylori , Nanopartículas , Compuestos Organometálicos , Preparaciones Farmacéuticas , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Bismuto , Química Farmacéutica
10.
PLoS Biol ; 16(1): e2003887, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29320492

RESUMEN

Urease as a potential target of antimicrobial drugs has received considerable attention given its versatile roles in microbial infection. Development of effective urease inhibitors, however, is a significant challenge due to the deeply buried active site and highly specific substrate of a bacterial urease. Conventionally, urease inhibitors are designed by either targeting the active site or mimicking substrate of urease, which is not efficient. Up to now, only one effective inhibitor-acetohydroxamic acid (AHA)-is clinically available, but it has adverse side effects. Herein, we demonstrate that a clinically used drug, colloidal bismuth subcitrate, utilizes an unusual way to inhibit urease activity, i.e., disruption of urease maturation process via functional perturbation of a metallochaperone, UreG. Similar phenomena were also observed in various pathogenic bacteria, suggesting that UreG may serve as a general target for design of new types of urease inhibitors. Using Helicobacter pylori UreG as a showcase, by virtual screening combined with experimental validation, we show that two compounds targeting UreG also efficiently inhibited urease activity with inhibitory concentration (IC)50 values of micromolar level, resulting in attenuated virulence of the pathogen. We further demonstrate the efficacy of the compounds in a mammalian cell infection model. This study opens up a new opportunity for the design of more effective urease inhibitors and clearly indicates that metallochaperones involved in the maturation of important microbial metalloenzymes serve as new targets for devising a new type of antimicrobial drugs.


Asunto(s)
Proteínas Bacterianas/efectos de los fármacos , Proteínas Portadoras/efectos de los fármacos , Compuestos Organometálicos/farmacología , Ureasa/antagonistas & inhibidores , Antiinfecciosos/farmacología , Proteínas Bacterianas/fisiología , Proteínas Portadoras/fisiología , Dominio Catalítico , Helicobacter pylori/metabolismo , Metalochaperonas/farmacología , Proteínas de Unión a Fosfato , Ureasa/fisiología , Virulencia
11.
J Biol Chem ; 294(20): 7990-8000, 2019 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-30936204

RESUMEN

Knowledge of the molecular mechanisms of specific bacterial virulence factors can significantly contribute to antibacterial drug discovery. Helicobacter pylori is a Gram-negative microaerophilic bacterium that infects almost half of the world's population, leading to gastric disorders and even gastric cancer. H. pylori expresses a series of virulence factors in the host, among which high-temperature requirement A (HpHtrA) is a newly identified serine protease secreted by H. pylori. HpHtrA cleaves the extracellular domain of the epithelial cell surface adhesion protein E-cadherin and disrupts gastric epithelial cell junctions, allowing H. pylori to access the intercellular space. Here we report the first crystal structure of HpHtrA at 3.0 Å resolution. The structure revealed a new type of HtrA protease trimer stabilized by unique N-terminal domain swapping distinct from other known HtrA homologs. We further observed that truncation of the N terminus completely abrogates HpHtrA trimer formation as well as protease activity. In the presence of unfolded substrate, HpHtrA assembled into cage-like 12-mers or 24-mers. Combining crystallographic, biochemical, and mutagenic data, we propose a mechanistic model of how HpHtrA recognizes and cleaves the well-folded E-cadherin substrate. Our study provides a fundamental basis for the development of anti-H. pylori agents by using a previously uncharacterized HtrA protease as a target.


Asunto(s)
Proteínas Bacterianas/química , Helicobacter pylori/enzimología , Modelos Biológicos , Serina Endopeptidasas/química , Factores de Virulencia/química , Antígenos CD/química , Antígenos CD/genética , Antígenos CD/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Cadherinas/química , Cadherinas/genética , Cadherinas/metabolismo , Cristalografía por Rayos X , Helicobacter pylori/genética , Helicobacter pylori/patogenicidad , Humanos , Dominios Proteicos , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Especificidad por Sustrato , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
12.
J Biol Chem ; 294(45): 16978-16991, 2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31586033

RESUMEN

Transition metals serve as an important class of micronutrients that are indispensable for bacterial physiology but are cytotoxic when they are in excess. Bacteria have developed exquisite homeostatic systems to control the uptake, storage, and efflux of each of biological metals and maintain a thermodynamically balanced metal quota. However, whether the pathways that control the homeostasis of different biological metals cross-talk and render cross-resistance or sensitivity in the host-pathogen interface remains largely unknown. Here, we report that zinc (Zn) excess perturbs iron (Fe) and copper (Cu) homeostasis in Escherichia coli, resulting in increased Fe and decreased Cu levels in the cell. Gene expression analysis revealed that Zn excess transiently up-regulates Fe-uptake genes and down-regulates Fe-storage genes and thereby increases the cellular Fe quota. In vitro and in vivo protein-DNA binding assays revealed that the elevated intracellular Fe poisons the primary Cu detoxification transcription regulator CueR, resulting in dysregulation of its target genes copA and cueO and activation of the secondary Cu detoxification system CusSR-cusCFBA Supplementation with the Fe chelator 2,2'-dipyridyl (DIP) or with the reducing agent GSH abolished the induction of cusCFBA during Zn excess. Consistent with the importance of this metal homeostatic network in cell physiology, combined metal treatment, including simultaneously overloading cells with both Zn (0.25 mm) and Cu (0.25 mm) and sequestering Fe with DIP (50 µm), substantially inhibited E. coli growth. These results advance our understanding of bacterial metallobiology and may inform the development of metal-based antimicrobial regimens to manage infectious diseases.


Asunto(s)
Cobre/farmacología , Escherichia coli/efectos de los fármacos , Escherichia coli/metabolismo , Hierro/metabolismo , Zinc/farmacología , Transporte Biológico/efectos de los fármacos , Escherichia coli/citología , Homeostasis/efectos de los fármacos , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Estrés Oxidativo/efectos de los fármacos
13.
Bioinformatics ; 35(19): 3812-3814, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30825371

RESUMEN

SUMMARY: We present MetaMarker, a pipeline for discovering metagenomic biomarkers from whole-metagenome sequencing samples. Different from existing methods, MetaMarker is based on a de novo approach that does not require mapping raw reads to a reference database. We applied MetaMarker on whole-metagenome sequencing of colorectal cancer (CRC) stool samples from France to discover CRC specific metagenomic biomarkers. We showed robustness of the discovered biomarkers by validating in independent samples from Hong Kong, Austria, Germany and Denmark. We further demonstrated these biomarkers could be used to build a machine learning classifier for CRC prediction. AVAILABILITY AND IMPLEMENTATION: MetaMarker is freely available at https://bitbucket.org/mkoohim/metamarker under GPLv3 license. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Metagenoma , Biomarcadores de Tumor , Neoplasias Colorrectales , Bases de Datos Factuales , Humanos , Metagenómica , Programas Informáticos
14.
Acc Chem Res ; 52(1): 216-227, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30596427

RESUMEN

Metallodrugs have been widely used as diagnostic and therapeutic agents. Understanding their mechanisms of action may lead to advances in rational drug design. However, to achieve this, diversified approaches are required because of the complexity of metal-biomolecule interactions. Bismuth drugs in combination with antibiotics as a quadruple therapy show excellent success rates in the eradication of Helicobacter pylori, even for antibiotic-resistant strains, and in fact, they have been used in the clinic for decades for the treatment of infection. Understanding the mechanism of action of bismuth drugs may extend their medicinal application beyond the treatment of H. pylori infection. This Account describes several general strategies for mechanistic studies of metallodrugs, including system pharmacology and metalloproteomics approaches. The application of these approaches is exemplified using bismuth drugs. Through a system pharmacology approach, we showed that glutathione- and multidrug-resistance-associated protein 1-mediated self-propelled disposal of bismuth in human cells might explain the selective toxicity of bismuth drugs to H. pylori but not the human host. The development of metalloproteomics has enabled extensive studies of the putative protein targets of metallodrugs with a dynamic range of affinity. Continuous-flow GE-ICP-MS allows simultaneous monitoring of metals and their associated proteins with relatively high affinity on a proteome-wide scale. The fluorescence approach relies on unique M n+-NTA-based fluorescence probes and is particularly applicable for mining those proteins that bind to metals/metallodrugs weakly or transiently. Integration of these methods with quantitative proteomics makes it possible to maximum coverage of bismuth-associated proteins, and the sustained efficacy of bismuth drugs lies in their ability to disrupt multiple biological pathways through binding and functional perturbation of key enzymes. The knowledge acquired by mechanistic studies of bismuth drugs led to the discovery of UreG as a new target for the development of urease inhibitors. The ability of Bi(III) to inhibit metallo-ß-lactamase (MBL) activity through displacement of the Zn(II) cofactor renders bismuth drugs new potential as broad-spectrum inhibitors of MBLs. Therefore, bismuth drugs could be repurposed together with clinically used antibiotics as a cotherapy to cope with the current antimicrobial resistance crisis. We anticipate that the methodologies described in this Account are generally applicable for understanding the (patho)physiological roles of metals/metallodrugs. Our mechanism-guided discovery of new druggable targets as well as new medicinal applications of bismuth drugs will inspire researchers in relevant fields to engage in the rational design of drugs and reuse of existing drugs, eventually leading to the development of new effective therapeutics.


Asunto(s)
Antibacterianos/farmacología , Bismuto/química , Helicobacter pylori/efectos de los fármacos , Compuestos Organometálicos/farmacología , Animales , Antibacterianos/química , Línea Celular , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Ratones , Compuestos Organometálicos/química , Farmacología/métodos , Proteómica/métodos , Ureasa/antagonistas & inhibidores
15.
Inorg Chem ; 58(20): 13673-13685, 2019 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-31298530

RESUMEN

The clinical application and development of potent metal-based drugs relies heavily on a full understanding of their underlying molecular mechanisms. The emerging research field of metalloproteomics for unveiling the correlation between metals and biological proteomes has received growing attention and has been successfully implemented in medicinal inorganic chemistry. In this article, we introduce recent progress in the mechanistic studies of representative metal-based drugs by using state-of-the-art metalloproteomic approaches, with a focus on how the application of analytical chemistry, biocompatible chemical probes, and integrative omics help to advance the knowledge of metallodrug targeting profiles within a systemic perspective.


Asunto(s)
Complejos de Coordinación/metabolismo , Metaloproteínas/metabolismo , Metales/metabolismo , Preparaciones Farmacéuticas/metabolismo , Proteómica , Complejos de Coordinación/química , Metaloproteínas/química , Metales/química , Estructura Molecular , Preparaciones Farmacéuticas/química
16.
Hum Hered ; 83(2): 79-91, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30347404

RESUMEN

AIMS: We propose a novel machine learning approach to expand the knowledge about drug-target interactions. Our method may help to develop effective, less harmful treatment strategies and to enable the detection of novel indications for existing drugs. METHODS: We developed a novel machine learning strategy to predict drug-target interactions based on drug side effects and traits from genome-wide association studies. We integrated data from the databases SIDER and GWASdb and utilized them in a unique way by a neural network approach. RESULTS: We validate our method using drug-target interactions from the STITCH database. In addition, we compare the chemical similarity of the predicted target to known targets of the drug under consideration and present literature-based evidence for predicted interactions. We find drug combination warnings for drugs we predict to target the same protein, hinting to synergistic effects aggravating harmful events. This substantiates the translational value of our approach, because we are able to detect drugs that should be taken together with care due to common mechanisms of action. CONCLUSION: Taken together, we conclude that our approach is able to generate a novel and clinically applicable insight into the molecular determinants of drug action.


Asunto(s)
Interacciones Farmacológicas , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Estudio de Asociación del Genoma Completo , Aprendizaje Automático , Humanos , Redes Neurales de la Computación
17.
Anal Chem ; 90(17): 10465-10471, 2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-30080396

RESUMEN

Arsenic has long been used as therapeutic agents. Understanding the mechanism of action of arsenic-based drugs enables more effective arsenic drugs to be developed. Cell cycle has been known to play a critical role for cell division and growth. Herein, we establish a methodology to evaluate the uptake of two arsenic-based drugs (ATO and ZIO-101) across the cell cycle in single leukemia cells, i.e., NB4 and HL60, using a double thymidine block combined with time-resolved inductively coupled plasma mass spectrometry (ICPMS). We show that cells absorb maximal amounts of both ATO and ZIO-101 in G2/M phase and minimum in S phase, and such variation is less apparent for ZIO-101 than ATO (NB4-G2/M:S = 2.5:1 for ATO and 1.6:1 for ZIO-101). We subsequently demonstrate that AQP9, an ATO transporter, is highly expressed in the G1 phase (50.2-46.9%) and minimum value was observed in the S phase (27.6-24.6%); whereas xCT, a ZIO-101 transporter, is maximally expressed in the G2/M phase (74.8-76.1%) and minimally expressed in the G1 phases (55.4-59.8%). Different expression levels of AQP9 and xCT are only partially accountable for the observed differences in arsenic uptake across cell cycle, indicative of the presence of other importers for both ATO and ZIO-101. Furthermore, we show that the cytotoxicity of ATO and ZIO-101 on NB4 cells is also cell cycle dependent, with the highest cytotoxicity at S + G2/M phase and the lowest at G1 + S phase. Our studies provide the first evidence on cell cycle dependent uptake and cytotoxicity of arsenic-based drugs at single cell levels, may have general implications for precise evaluation of other anticancer drugs by considering cell cycle phase.


Asunto(s)
Antineoplásicos/farmacocinética , Arsénico/farmacocinética , Ciclo Celular , Leucemia/tratamiento farmacológico , Leucemia/patología , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Arsénico/farmacología , Arsénico/uso terapéutico , Ciclo Celular/efectos de los fármacos , Citometría de Flujo , Humanos , Espectrometría de Masas
18.
J Biol Inorg Chem ; 23(3): 471-480, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29623422

RESUMEN

The presence of ionic titanium in the serum of patients with titanium implants is currently unexplained. This is presumed due to corrosion, and yet the serum titanium concentration measured in patients is far greater than that predicted by its solubility. The binding of titanium ion as Ti(IV) to human transferrin (hTF) in serum indicates that Ti(IV) ions interact with human physiology. This is an intriguing finding since there is currently no known role for titanium ions in human physiology. Thus, understanding the factors that determine in vivo titanium ion release is relevant to further understanding this metal's interactions with human biochemistry. The present study sought to determine the extent of titanium ion release of into human serum in vitro, and the role of citrate, lactate and hTF in this process. It was found that, when surgical devices of commercially pure titanium were placed into human serum, citrate and lactate concentrations were the prime determinants of titanium release. Crystallography revealed Ti(IV) bound to hTF in the presence of citrate alone, signalling that citrate can act as an independent ligand for Ti(IV) binding to hTF. Based on these findings, a two-stage process of titanium ion release into human serum that is dependent upon both citrate and hTF is proposed to explain the ongoing presence of titanium ion in human subjects with implanted titanium devices.


Asunto(s)
Ácido Cítrico/sangre , Ácido Láctico/sangre , Prótesis e Implantes , Suero , Titanio/farmacocinética , Transferrina/metabolismo , Corrosión , Cristalografía , Humanos , Microscopía Electrónica de Rastreo , Titanio/sangre
19.
Proc Natl Acad Sci U S A ; 112(11): 3211-6, 2015 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-25737551

RESUMEN

Glutathione and multidrug resistance protein (MRP) play an important role on the metabolism of a variety of drugs. Bismuth drugs have been used to treat gastrointestinal disorder and Helicobacter pylori infection for decades without exerting acute toxicity. They were found to interact with a wide variety of biomolecules, but the major metabolic pathway remains unknown. For the first time (to our knowledge), we systematically and quantitatively studied the metabolism of bismuth in human cells. Our data demonstrated that over 90% of bismuth was passively absorbed, conjugated to glutathione, and transported into vesicles by MRP transporter. Mathematical modeling of the system reveals an interesting phenomenon. Passively absorbed bismuth consumes intracellular glutathione, which therefore activates de novo biosynthesis of glutathione. Reciprocally, sequestration by glutathione facilitates the passive uptake of bismuth and thus completes a self-sustaining positive feedback circle. This mechanism robustly removes bismuth from both intra- and extracellular space, protecting critical systems of human body from acute toxicity. It elucidates the selectivity of bismuth drugs between human and pathogens that lack of glutathione, such as Helicobacter pylori, opening new horizons for further drug development.


Asunto(s)
Bismuto/metabolismo , Glutatión/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Bismuto/farmacología , Compartimento Celular/efectos de los fármacos , Línea Celular , Coloides/metabolismo , Coloides/farmacología , Escherichia coli/metabolismo , Humanos , Inactivación Metabólica/efectos de los fármacos , Transporte Iónico/efectos de los fármacos , Modelos Biológicos , Compuestos Organometálicos/metabolismo , Compuestos Organometálicos/farmacología , Proteómica , Estadística como Asunto , Factores de Tiempo
20.
Proc Natl Acad Sci U S A ; 112(10): 2948-53, 2015 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-25713372

RESUMEN

Small molecule-based fluorescent probes have been used for real-time visualization of live cells and tracking of various cellular events with minimal perturbation on the cells being investigated. Given the wide utility of the (histidine)6-Ni(2+)-nitrilotriacetate (Ni-NTA) system in protein purification, there is significant interest in fluorescent Ni(2+)-NTA-based probes. Unfortunately, previous Ni-NTA-based probes suffer from poor membrane permeability and cannot label intracellular proteins. Here, we report the design and synthesis of, to our knowledge, the first membrane-permeable fluorescent probe Ni-NTA-AC via conjugation of NTA with fluorophore and arylazide followed by coordination with Ni(2+) ions. The probe, driven by Ni(2+)-NTA, binds specifically to His-tags genetically fused to proteins and subsequently forms a covalent bond upon photoactivation of the arylazide, leading to a 13-fold fluorescence enhancement. The arylazide is indispensable not only for fluorescence enhancement, but also for strengthening the binding between the probe and proteins. Significantly, the Ni-NTA-AC probe can rapidly enter different types of cells, even plant tissues, to target His-tagged proteins. Using this probe, we visualized the subcellular localization of a DNA repair protein, Xeroderma pigmentosum group A (XPA122), which is known to be mainly enriched in the nucleus. We also demonstrated that the probe can image a genetically engineered His-tagged protein in plant tissues. This study thus offers a new opportunity for in situ visualization of large libraries of His-tagged proteins in various prokaryotic and eukaryotic cells.


Asunto(s)
Histidina/metabolismo , Proteínas/metabolismo , Colorantes Fluorescentes , Células HeLa , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA