Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 140
Filter
1.
Int J Mol Sci ; 25(14)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39063122

ABSTRACT

Essential transition metals have key roles in oxygen transport, neurotransmitter synthesis, nucleic acid repair, cellular structure maintenance and stability, oxidative phosphorylation, and metabolism. The balance between metal deficiency and excess is typically ensured by several extracellular and intracellular mechanisms involved in uptake, distribution, and excretion. However, provoked by either intrinsic or extrinsic factors, excess iron, zinc, copper, or manganese can lead to cellular damage upon chronic or acute exposure, frequently attributed to oxidative stress. Intracellularly, mitochondria are the organelles that require the tightest control concerning reactive oxygen species production, which inevitably leaves them to be one of the most vulnerable targets of metal toxicity. Current therapies to counteract metal overload are focused on chelators, which often cause secondary effects decreasing patients' quality of life. New therapeutic options based on synthetic or natural antioxidants have proven positive effects against metal intoxication. In this review, we briefly address the cellular metabolism of transition metals, consequences of their overload, and current therapies, followed by their potential role in inducing oxidative stress and remedies thereof.


Subject(s)
Antioxidants , Oxidative Stress , Transition Elements , Humans , Antioxidants/therapeutic use , Antioxidants/metabolism , Oxidative Stress/drug effects , Transition Elements/metabolism , Animals , Mitochondria/metabolism , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Iron/metabolism , Metals/metabolism , Chelating Agents/therapeutic use , Chelating Agents/pharmacology
2.
J Biol Chem ; 300(8): 107589, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39032653

ABSTRACT

Transition metal ions are critically important across all kingdoms of life. The chemical properties of iron, copper, zinc, manganese, cobalt, and nickel make them very attractive for use as cofactors in metalloenzymes and/or metalloproteins. Their versatile chemistry in aqueous solution enables them to function both as electron donors and acceptors, and thus participate in both reduction and oxidation reactions respectively. Transition metal ions can also function as nonredox multidentate coordination sites that play essential roles in macromolecular structure and function. Malfunction in transition metal transport and homeostasis has been linked to a wide number of human diseases including cancer, diabetes, and neurodegenerative disorders. Transition metal transporters are central players in the physiology of transition metals whereby they move transition metals in and out of cellular compartments. In this review, we provide a comprehensive overview of in vitro reconstitution of the activity of integral membrane transition metal transporters and discuss strategies that have been successfully implemented to overcome the challenges. We also discuss recent advances in our understanding of transition metal transport mechanisms and the techniques that are currently used to decipher the molecular basis of transport activities of these proteins. Deep mechanistic insights into transition metal transport systems will be essential to understand their malfunction in human diseases and target them for potential therapeutic strategies.


Subject(s)
Transition Elements , Humans , Transition Elements/metabolism , Transition Elements/chemistry , Animals
3.
Sheng Li Xue Bao ; 76(3): 418-428, 2024 Jun 25.
Article in Chinese | MEDLINE | ID: mdl-38939936

ABSTRACT

Aging refers to a progressive decline in biological functions, leading to age-related diseases and mortality. The transition metals, including iron, copper, and manganese, play important roles in human physiological and pathological processes. Substantial research has demonstrated that senescent cells accumulate higher levels of transition metals, which in turn accelerates the process of cellular senescence and related diseases through mechanisms such as production of excessive reactive oxygen species (ROS), induction of oxidative stress, DNA damage, and mitochondrial dysfunction. This review article provides a comprehensive overview of the causes of transition metal accumulation in senescent cells, as well as the mechanisms by which it further promotes cellular senescence and related diseases. The aim is to provide insights into anti-aging and treatment of aging-related diseases caused by transition metal accumulation.


Subject(s)
Aging , Cellular Senescence , DNA Damage , Oxidative Stress , Reactive Oxygen Species , Cellular Senescence/physiology , Humans , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Aging/physiology , Aging/metabolism , Animals , Transition Elements/metabolism , Iron/metabolism , Mitochondria/metabolism , Mitochondria/physiology , Copper/metabolism , Manganese/metabolism
4.
Environ Res ; 258: 119460, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38906451

ABSTRACT

To investigate the inhibitory effects of various transition metal ions on nitrogen removal and their underlying mechanisms, the single and combined effects of Cu2+ Ni2+ and Zn2+ on Heterotrophic nitrification-aerobic denitrification (HN-AD) bacteria Acinetobacter sp. TAC-1 were studied in a batch experiment system. The results revealed that increasing concentrations of Cu2+ and Ni2+ had a detrimental effect on the removal of ammonium nitrogen (NH4+-N) and total nitrogen (TN). Specifically, Cu2+ concentration of 10 mg/L, the TN degradation rate was 55.09%, compared to 77.60% in the control group. Cu2+ exhibited a pronounced inhibitory effect. In contrast, Zn2+ showed no apparent inhibitory effect on NH4+-N removal and even enhanced TN removal at lower concentrations. However, when the mixed ion concentration of Zn2++Ni2+ exceeded 5 mg/L, the removal rates of NH4+-N and TN were significantly reduced. Moreover, transition metal ions did not significantly impact the removal rates of chemical oxygen demand (COD). The inhibition model fitting results indicated that the inhibition sequence was Cu2+ > Zn2+ > Ni2+. Transcriptome analysis demonstrated that metal ions influence TAC-1 activity by modulating the expression of pivotal genes, including zinc ABC transporter substrate binding protein (znuA), ribosomal protein (rpsM), and chromosome replication initiation protein (dnaA) and DNA replication of TAC-1 under metal ion stress, leading to disruptions in transcription, translation, and cell membrane structure. Finally, a conceptual model was proposed by us to summarize the inhibition mechanism and possible response strategies of TAC-1 bacteria under metal ion stress, and to address the lack of understanding regarding the influence mechanism of TAC-1 on nitrogen removal in wastewater co-polluted by metal and ammonia nitrogen. The results provided practical guidance for the management of transition metal and ammonia nitrogen co-polluted water bodies, as well as the removal of high nitrogen.


Subject(s)
Denitrification , Nitrification , Acinetobacter/metabolism , Acinetobacter/genetics , Heterotrophic Processes , Aerobiosis , Transition Elements/metabolism , Nitrogen/metabolism , Water Pollutants, Chemical/metabolism
5.
Molecules ; 28(19)2023 Oct 01.
Article in English | MEDLINE | ID: mdl-37836742

ABSTRACT

Inflammatory mediators constitute a recently coined term in the field of metal-based complexes with antiplatelet activities. Our strategy targets Platelet-Activating Factor (PAF) and its receptor, which is the most potent lipid mediator of inflammation. Thus, the antiplatelet (anti-PAF) potency of any substance could be exerted by inhibiting the PAF-induced aggregation in washed rabbit platelets (WRPs), which internationally is a well-accepted methodology. Herein, a series of mononuclear (mer-[Cr(pqx)Cl3(H2O]) (1), [Co(pqx)Cl2(DMF)] (2) (DMF = N,N'-dimethyl formamide), [Cu(pqx)Cl2(DMSO)] (3) (DMSO = dimethyl sulfoxide), [Zn(pqx)Cl2] (4)) and dinuclear complexes ([Mn(pqx)(H2O)2Cl2]2 (5), [Fe(pqx)Cl2]2 (6) and [Ni(pqx)Cl2]2 (7)) incorporating the 2-(2'-pyridyl)quinoxaline ligand (pqx), were biologically evaluated as inhibitors of the PAF- and thrombin-induced aggregation in washed rabbit platelets (WRPs). The molecular structure of the five-co-ordinate analog (3) has been elucidated by single-crystal X-ray diffraction revealing a trigonal bipyramidal geometry. All complexes are potent inhibitors of the PAF-induced aggregation in WRPs in the micromolar range. Complex (6) displayed a remarkable in vitro dual inhibition against PAF and thrombin, with IC50 values of 1.79 µM and 0.46 µM, respectively. Within the series, complex (5) was less effective (IC50 = 39 µM) while complex (1) was almost 12-fold more potent against PAF, as opposed to thrombin-induced aggregation. The biological behavior of complexes 1, 6 and 7 on PAF's basic metabolic enzymatic pathways reveals that they affect key biosynthetic and catabolic enzymes of PAF underlying the anti-inflammatory properties of the relevant complexes. The in vitro cytotoxic activities of all complexes in HEK293T (human embryonic kidney cells) and HeLa cells (cervical cancer cells) are described via the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay. The results reveal that complex 3 is the most potent within the series.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Transition Elements , Animals , Humans , Rabbits , Platelet Aggregation , Platelet Activating Factor/pharmacology , Platelet Activating Factor/metabolism , Blood Platelets/metabolism , Thrombin/metabolism , Coordination Complexes/pharmacology , Coordination Complexes/metabolism , Ligands , Inflammation Mediators/metabolism , Dimethyl Sulfoxide/pharmacology , Quinoxalines/pharmacology , HEK293 Cells , HeLa Cells , Antineoplastic Agents/pharmacology , Transition Elements/metabolism
6.
J Inorg Biochem ; 227: 111687, 2022 02.
Article in English | MEDLINE | ID: mdl-34953313

ABSTRACT

Sulfide and transition metals often came together in Biology. The variety of possible structural combinations enabled living organisms to evolve an array of highly versatile metal-sulfide centers to fulfill different physiological roles. The ubiquitous iron­sulfur centers, with their structural, redox, and functional diversity, are certainly the best-known partners, but other metal-sulfide centers, involving copper, nickel, molybdenum or tungsten, are equally crucial for Life. This review provides a concise overview of the exclusive sulfide properties as a metal ligand, with emphasis on the structural aspects and biosynthesis. Sulfide as catalyst and as a substrate is discussed. Different enzymes are considered, including xanthine oxidase, formate dehydrogenases, nitrogenases and carbon monoxide dehydrogenases. The sulfide effect on the activity and function of iron­sulfur, heme and zinc proteins is also addressed.


Subject(s)
Iron-Sulfur Proteins , Metalloproteins , Sulfides , Transition Elements , Heme/chemistry , Heme/metabolism , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/metabolism , Metalloproteins/chemistry , Metalloproteins/metabolism , Sulfides/chemistry , Sulfides/metabolism , Transition Elements/chemistry , Transition Elements/metabolism
7.
J Am Chem Soc ; 143(43): 18073-18090, 2021 11 03.
Article in English | MEDLINE | ID: mdl-34699194

ABSTRACT

Human calprotectin (CP, S100A8/S100A9 oligomer, MRP8/MRP14 oligomer) is an abundant innate immune protein that contributes to the host metal-withholding response. Its ability to sequester transition metal nutrients from microbial pathogens depends on a complex interplay of Ca(II) binding and self-association, which converts the αß heterodimeric apo protein into a Ca(II)-bound (αß)2 heterotetramer that displays enhanced transition metal affinities, antimicrobial activity, and protease stability. A paucity of structural data on the αß heterodimer has hampered molecular understanding of how Ca(II) binding enables CP to exert its metal-sequestering innate immune function. We report solution NMR data that reveal how Ca(II) binding affects the structure and dynamics of the CP αß heterodimer. These studies provide a structural model in which the apo αß heterodimer undergoes conformational exchange and switches between two states, a tetramerization-incompetent or "inactive" state and a tetramerization-competent or "active" state. Ca(II) binding to the EF-hands of the αß heterodimer causes the active state to predominate, resulting in self-association and formation of the (αß)2 heterotetramer. Moreover, Ca(II) binding causes local and allosteric ordering of the His3Asp and His6 metal-binding sites. Ca(II) binding to the noncanonical EF-hand of S100A9 positions (A9)D30 and organizes the His3Asp site. Remarkably, Ca(II) binding causes allosteric effects in the C-terminal region of helix αIV of S100A9, which stabilize the α-helicity at positions H91 and H95 and thereby organize the functionally versatile His6 site. Collectively, this study illuminates the molecular basis for how CP responds to high extracellular Ca(II) concentrations, which enables its metal-sequestering host-defense function.


Subject(s)
Calcium/metabolism , Leukocyte L1 Antigen Complex/metabolism , Protein Multimerization/drug effects , Transition Elements/metabolism , Calgranulin A/genetics , Calgranulin A/metabolism , Calgranulin B/genetics , Calgranulin B/metabolism , Histidine/chemistry , Humans , Leukocyte L1 Antigen Complex/genetics , Metals, Heavy/metabolism , Mutation , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Conformation, alpha-Helical/drug effects , Protein Multimerization/genetics
8.
Inorg Chem ; 60(20): 15096-15100, 2021 Oct 18.
Article in English | MEDLINE | ID: mdl-34529407

ABSTRACT

We report on the synthesis of the tetrasubstituted sandwich-type Keggin silicotungstates as the pure Na salts Na14[(A-α-SiW10O37)2{Co4(OH)2(H2O)2}]·37H2O (Na{SiW10Co2}2) and Na14[(A-α-SiW10O37)2{Ni4(OH)2(H2O)2}]·77.5H2O (Na{SiW10Ni2}2), which were prepared by applying a new synthesis protocol and characterized thoroughly in the solid state by single-crystal and powder X-ray diffraction, IR spectroscopy, thermogravimetric analysis, and elemental analysis. Proteinase K was applied as a model protein and the polyoxotungstate (POT)-protein interactions of Na{SiW10Co2}2 and Na{SiW10Ni2}2 were studied side by side with the literature-known K5Na3[A-α-SiW9O34(OH)3{Co4(OAc)3}]·28.5H2O ({SiW9Co4}) featuring the same number of transition metals. Testing the solution behavior of applied POTs under the crystallization conditions (sodium acetate buffer, pH 5.5) by time-dependent UV/vis spectroscopy and electrospray ionization mass spectrometry speciation studies revealed an initial dissociation of the sandwich POTs to the disubstituted Keggin anions HxNa5-x[SiW10Co2O38]3- and HxNa5-x[SiW10Ni2O38]3- ({SiW10M2}, M = CoII and NiII) followed by partial rearrangement to the monosubstituted compounds (α-{SiW11Co} and α-{SiW11Ni}) after 1 week of aging. The protein crystal structure analysis revealed monosubstituted α-Keggin POTs in two conserved binding positions for all three investigated compounds, with one of these positions featuring a covalent attachment of the POT anion to an aspartate carboxylate. Despite the presence of both mono- and disubstituted anions in a crystallization mixture, proteinase K selectively binds to monosubstituted anions because of their preferred charge density for POT-protein interaction.


Subject(s)
Cobalt/chemistry , Coordination Complexes/chemistry , Endopeptidase K/chemistry , Silicon/chemistry , Transition Elements/chemistry , Tungsten Compounds/chemistry , Cobalt/metabolism , Coordination Complexes/metabolism , Crystallography, X-Ray , Endopeptidase K/metabolism , Hypocreales/enzymology , Models, Molecular , Molecular Structure , Silicon/metabolism , Transition Elements/metabolism , Tungsten Compounds/metabolism
9.
Chem Soc Rev ; 50(8): 4804-4811, 2021 Apr 26.
Article in English | MEDLINE | ID: mdl-33657202

ABSTRACT

Oxygen evolution and reduction reactions are fundamental processes in biological energy conversion schemes, which represent an attractive method for artificial energy conversion for a world still largely depending on fossil fuels. A range of metalloenzymes achieve these challenging tasks in biology by activating water and dioxygen using cheap and abundant transition metals, such as iron, copper, and manganese. High-valent metal-oxo/oxyl, metal-superoxo, and/or metal-(hydro)peroxo species are common reactive intermediates that are found in the O-O bond formation and activation reactions. The transient nature of the metal-oxygen intermediates has, however, prevented their isolation and characterization in most cases. As a consequence, unambiguous mechanistic assignments in the O-O bond formation and cleavage processes in biological and chemical entries remain elusive, especially for the intermediates and mechanisms involved in the O-O bond formation reactions. This viewpoint article aims at summarizing the information obtained to date in enzymatic and biomimetic systems that fuels the debate regarding the nature of the active oxidants and the mechanistic uncertainties associated with the transition metal-mediated O-O bond formation and cleavage reactions.


Subject(s)
Oxygen/metabolism , Transition Elements/metabolism , Oxygen/chemistry , Transition Elements/chemistry
10.
J Cell Physiol ; 236(10): 7144-7158, 2021 10.
Article in English | MEDLINE | ID: mdl-33694161

ABSTRACT

Transition metals refer to the elements in the d and ds blocks of the periodic table. Since the success of cisplatin and auranofin, transition metal-based compounds have become a prospective source for drug development, particularly in cancer treatment. In recent years, extensive studies have shown that numerous transition metal-based compounds could modulate autophagy, promising a new therapeutic strategy for metal-related diseases and the design of metal-based agents. Copper, zinc, and manganese, which are common components in physiological pathways, play important roles in the progression of cancer, neurodegenerative diseases, and cardiovascular diseases. Furthermore, enrichment of copper, zinc, or manganese can regulate autophagy. Thus, we summarized the current advances in elucidating the mechanisms of some metals/metal-based compounds and their functions in autophagy regulation, which is conducive to explore the intricate roles of autophagy and exploit novel therapeutic drugs for human diseases.


Subject(s)
Autophagy/drug effects , Cardiovascular Diseases/drug therapy , Coordination Complexes/therapeutic use , Metals/therapeutic use , Neoplasms/drug therapy , Neurodegenerative Diseases/drug therapy , Transition Elements/therapeutic use , Animals , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Coordination Complexes/metabolism , Humans , Metals/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Transition Elements/metabolism
11.
J Trace Elem Med Biol ; 60: 126495, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32179426

ABSTRACT

Addiction is a pressing social problem worldwide and opioid dependence can be considered the strongest and most difficult addiction to treat. Mesolimbic and mesocortical dopaminergic pathways play an important role in modulation of cognitive processes and decision making and, therefore, changes in dopamine metabolism are considered the central basis for the development of dependence. Disturbances caused by excesses or deficiency of certain elements have a significant impact on the functioning of the central nervous system (CNS) both in physiological conditions and in pathology and can affect the cerebral reward system and therefore, may modulate processes associated with the development of addiction. In this paper we review the mechanisms of interactions between morphine and zinc, manganese, chromium, cadmium, lead, fluoride, their impact on neural pathways associated with addiction, and on antinociception and morphine tolerance and dependence.


Subject(s)
Morphine Dependence/metabolism , Morphine/metabolism , Transition Elements/metabolism , Animals , Humans , Morphine/chemistry , Neural Pathways/metabolism , Transition Elements/chemistry
12.
Curr Opin Microbiol ; 55: 17-25, 2020 06.
Article in English | MEDLINE | ID: mdl-32062305

ABSTRACT

Transition metals from manganese to zinc function as catalytic and structural cofactors for an amazing diversity of proteins and enzymes, and thus are essential for all forms of life. During infection, inflammatory host proteins limit the accessibility of multiple transition metals to invading pathogens in a process termed nutritional immunity. In order to respond to host-mediated metal starvation, bacteria employ both protein and RNA-based mechanisms to sense prevailing transition metal concentrations that collectively regulate systems-level strategies to maintain cellular metallostasis. In this review, we discuss a number of recent advances in our understanding of how bacteria orchestrate the adaptive response to host-mediated multi-metal restriction, highlighting crosstalk among these regulatory systems.


Subject(s)
Bacteria/metabolism , Host-Pathogen Interactions , Leukocyte L1 Antigen Complex/physiology , Metals/immunology , Metals/metabolism , Bacterial Physiological Phenomena , Humans , Immunity , Transition Elements/metabolism
13.
Metallomics ; 12(1): 8-11, 2020 01 29.
Article in English | MEDLINE | ID: mdl-31913381

ABSTRACT

After exposure to micron-sized TiO2 particles, anatase and/or rutile, Rhodococcus ruber GIN-1 accumulates an increased concentration (2.2 ± 0.2 mg kg-1) of mobilized Ti into its biomass with concomitant decreases in cellular biometals Fe, Zn, and possibly Mn, while levels of Cu and Al are unaffected.


Subject(s)
Rhodococcus/drug effects , Rhodococcus/metabolism , Titanium/pharmacology , Transition Elements/metabolism , Aluminum/metabolism , Biomass , Copper/metabolism , Iron/metabolism , Manganese/metabolism , Zinc/metabolism
14.
J Phys Chem B ; 124(1): 101-109, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31829598

ABSTRACT

We have studied the effects of different 3d orbitals in divalent transition-metal ions [G2+ = Mn2+ (d5), Fe2+ (d6), Co2+ (d7), Ni2+ (d8), Cu2+ (d9), or Zn2+ (d10)] on the conformations of leucine encephalin (LE) and methionine encephalin (ME) in the gas phase using hydrogen/deuterium exchange mass spectrometry (HDX-MS) and theoretical calculations at the molecular level. The HDX-MS reveals a 1:1 stoichiometric monovalent complex of [LE/ME + G - H]+ and observed that the different HDX reactivities follow the trend Fe2+ < Co2+ < Ni2+ < Mn2+ < Cu2+ ≈ Zn2+ and that [ME + Mn/Cu/Zn - H]+ > [LE + Mn/Cu/Zn - H]+, while [LE + Fe/Co/Ni - H]+ > [ME + Fe/Co/Ni - H]+. We cross-correlated the collision-induced dissociation energies of the complexes with the HDX results and found that the more stable the complex, the harder it is for it to undergo HDX. Furthermore, we used theoretical calculations to optimize the favorable conformations of the complexes and found the same interaction structure of G2+ coordination with the five carbonyl oxygens of LE/ME that have different bond lengths. Finally, we calculated the proton affinity (PA) values of the optimized complexes in order to interpret the HDX observations that the higher the PA values, the more difficult it is for the complex to undergo HDX. Overall, both the experiments and the theoretical calculations show that the six metal ions have different effects on the LE/ME conformation, with the low-energy stability of the G2+ 3d orbitals corresponding to more dramatic effects on the LE/ME conformation. In addition, the hardness of the ionic acid corresponding to the fully filled Mn2+ and half-filled Zn2+ orbitals also contributes strongly to the coordination effect; the conformation effect of Fe2+/Co2+/Ni2+ on LE is greater than that on ME, whereas the conformation effect of Mn2+/Cu2+/Zn2+ on ME is greater than that on LE.


Subject(s)
Enkephalins/chemistry , Mass Spectrometry/methods , Transition Elements/chemistry , Amino Acid Sequence , Deuterium Exchange Measurement , Enkephalins/metabolism , Ions/chemistry , Quantum Theory , Transition Elements/metabolism
15.
Acta Biochim Pol ; 66(4): 459-462, 2019 Dec 11.
Article in English | MEDLINE | ID: mdl-31826048

ABSTRACT

One of the antioxidant roles of melanin is binding redox-active transition metal ions. The aim of this study was to examine the redox reactions accompanying iron binding by melanin. Two kinds of synthetic eumelanin were mixed with iron (II) and iron (III) in the presence and absence of citrate and ADP in the aerobic and anaerobic system. The iron binding was examined by electron paramagnetic resonance (EPR) spectroscopy and thiocyanate assay. Obtained results indicate that although melanin reduces iron (III) that is unbound to this polymer, binding of iron (II) is accompanied by its oxidation by melanin.


Subject(s)
Iron/metabolism , Melanins/metabolism , Adenosine Diphosphate/pharmacology , Citric Acid/pharmacology , Ions , Oxidation-Reduction , Protein Binding , Transition Elements/metabolism
16.
Molecules ; 24(17)2019 Aug 31.
Article in English | MEDLINE | ID: mdl-31480486

ABSTRACT

Elucidation of the structure and function of biomolecules provides us knowledge that can be transferred into the generation of new materials and eventually applications in e.g., catalysis or bioassays. The main problems, however, concern the complexity of the natural systems and their limited availability, which necessitates utilization of simple biomimetic analogues that are, to a certain degree, similar in terms of structure and thus behaviour. We have, therefore, devised a small library of six tridentate N-heterocyclic coordinating agents (L1-L6), which, upon complexation, form two groups of artificial, monometallic non-heme iron species. Utilization of iron(III) chloride leads to the formation of the 1:1 (Fe:Ln) 'open' complexes, whereas iron(II) trifluoromethanosulfonate allows for the synthesis of 1:2 (M:Ln) 'closed' systems. The structural differences between the individual complexes are a result of the information encoded within the metallic centre and the chosen counterion, whereas the organic scaffold influences the observed properties. Indeed, the number and nature of the external hydrogen bond donors coming from the presence of (benz)imidazole moieties in the ligand framework are responsible for the observed biological behaviour in terms of mimicking phenoxazinone synthase activity and interaction with DNA.


Subject(s)
Benzimidazoles/chemistry , Biomimetic Materials/chemistry , DNA/metabolism , Iron/chemistry , Oxidoreductases/metabolism , Schiff Bases/chemistry , Aminophenols/metabolism , Animals , Binding, Competitive , Catalysis , Cattle , Fluorescence , Imidazoles , Kinetics , Ligands , Oxazines , Oxidation-Reduction , Schiff Bases/chemical synthesis , Transition Elements/metabolism
17.
Chem Commun (Camb) ; 55(62): 9148-9151, 2019 Jul 30.
Article in English | MEDLINE | ID: mdl-31304493

ABSTRACT

Using superoxide reductase as a model system, a computational approach reveals how histidine tautomerism tunes the redox properties of metalloenzymes to enable their catalytic function. Inspired by these experimentally inaccessible insights, non-canonical histidine congeners are introduced as new versatile tools for the rational engineering of biological transition metal sites.


Subject(s)
Oxidoreductases/chemistry , Oxidoreductases/metabolism , Transition Elements/metabolism , Biocatalysis , Models, Molecular , Molecular Structure , Oxidation-Reduction , Transition Elements/chemistry
18.
Biometals ; 32(3): 369-384, 2019 06.
Article in English | MEDLINE | ID: mdl-30788645

ABSTRACT

Host-associated microbial communities provide critical functions for their hosts. Transition metals are essential for both the mammalian host and the majority of commensal bacteria. As such, access to transition metals is an important component of host-microbe interactions in the gastrointestinal tract. In mammals, transition metal ions are often sequestered by metal binding proteins to limit microbial access under homeostatic conditions. In response to invading pathogens, the mammalian host further decreases availability of these micronutrients by regulating their trafficking or releasing high-affinity metal chelating proteins, a process termed nutritional immunity. Bacterial pathogens have evolved several mechanisms to subvert nutritional immunity. Here, we provide an overview on how metal ion availability shapes host-microbe interactions in the gut with a particular focus on intestinal inflammatory diseases.


Subject(s)
Host Microbial Interactions , Intestines/microbiology , Transition Elements/metabolism , Animals , Humans
19.
J Biomol Struct Dyn ; 37(17): 4590-4600, 2019 10.
Article in English | MEDLINE | ID: mdl-30526382

ABSTRACT

We report molecular dynamics simulations of three possible adducts of Fe(II) to the N-terminal 1-16 fragments of the amyloid-ß peptide, along with analogous simulations of Cu(II) and Zn(II) adducts. We find that multiple simulations from different starting points reach pseudo-equilibration within 100-300 ns, leading to over 900 ns of equilibrated trajectory data for each system. The specifics of the coordination modes for Fe(II) have only a weak effect on peptide secondary and tertiary structures, and we therefore compare one of these with analogous models of Cu(II) and Zn(II) complexes. All share broadly similar structural features, with mixture of coil, turn and bend in the N-terminal region and helical structure for residues 11-16. Within this overall pattern, subtle effects due to changes in metal are evident: Fe(II) complexes are more compact and are more likely to occupy bridge and ribbon regions of Ramachandran maps, while Cu(II) coordination leads to greater occupancy of the poly-proline region. Analysis of representative clusters in terms of molecular mechanics energy and atoms-in-molecules properties indicates similarity of four-coordinate Cu and Zn complexes, compared to five-coordinate Fe complex that exhibits lower stability and weaker metal-ligand bonding. Communicated by Ramaswamy H. Sarma.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Metals/metabolism , Molecular Dynamics Simulation , Transition Elements/metabolism , Cluster Analysis , Hydrogen Bonding , Protein Binding , Protein Structure, Secondary
20.
J Inorg Biochem ; 191: 69-76, 2019 02.
Article in English | MEDLINE | ID: mdl-30468944

ABSTRACT

Islet Amyloid Polypeptide (IAPP), also known as amylin, is a 37-amino-acid peptide hormone that is secreted by pancreatic islet ß-cells. Amylin is complementary to insulin in regulating and maintaining blood glucose levels in the human body. The misfolding and aggregation of amylin is primarily associated with type 2 diabetes mellitus, which is classified as an amyloid disease. Recently, the interactions between amylin and specific metal ions, e.g., copper(II), zinc(II), and iron(II), were found to impact its performance and aggregation processes. Therefore, the focus in this review will be on how the chemistry and structural properties of amylin are affected by these interactions. In addition, the impact of amylin and other amyloidogenic peptides interacting with metal ions on the cell membranes is discussed. In particular, recent studies on the interactions of amylin with copper, zinc, iron, nickel, gold, ruthenium, and vanadium are discussed.


Subject(s)
Islet Amyloid Polypeptide/metabolism , Transition Elements/metabolism , Animals , Cell Membrane/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL