Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Anal Chem ; 92(14): 9429-9440, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32490668

ABSTRACT

Many research institutions, clinical diagnostic laboratories, and blood banks are desperately searching for a possibility to identify and quantify heme in different physiological and pathological settings as well as various research applications. The reasons for this are the toxicity of the heme and the fact that it acts as a hemolytic and pro-inflammatory molecule. Heme only exerts these severe and undesired effects when it is not incorporated in hemoproteins. Upon release from the hemoproteins, it enters a biologically available state (labile heme), in which it is loosely associated with proteins, lipids, nucleic acids, or other molecules. While the current methods and procedures for quantitative determination of heme have been used for many years in different settings, their value is limited by the challenging chemical properties of heme. A major cause of inadequate quantification is the separation of labile and permanently bound heme and its high aggregation potential. Thus, none of the current methods are utilized as a generally applicable, standardized approach. The aim of this Feature is to describe and summarize the most common and frequently used chemical, analytical, and biochemical methods for the quantitative determination of heme. Based on this overview, the most promising approaches for future solutions to heme quantification are highlighted.


Subject(s)
Chromatography/methods , Electrophoresis, Capillary/methods , Enzyme Assays/methods , Heme/chemistry , Hemeproteins/chemistry , Humans , Molecular Structure , Spectrum Analysis
2.
BMC Bioinformatics ; 21(1): 124, 2020 Mar 27.
Article in English | MEDLINE | ID: mdl-32216745

ABSTRACT

BACKGROUND: The notion of heme as a regulator of many physiological processes via transient binding to proteins is one that is recently being acknowledged. The broad spectrum of the effects of heme makes it important to identify further heme-regulated proteins to understand physiological and pathological processes. Moreover, several proteins were shown to be functionally regulated by interaction with heme, yet, for some of them the heme-binding site(s) remain unknown. The presented application HeMoQuest enables identification and qualitative evaluation of such heme-binding motifs from protein sequences. RESULTS: We present HeMoQuest, an online interface (http://bit.ly/hemoquest) to algorithms that provide the user with two distinct qualitative benefits. First, our implementation rapidly detects transient heme binding to nonapeptide motifs from protein sequences provided as input. Additionally, the potential of each predicted motif to bind heme is qualitatively gauged by assigning binding affinities predicted by an ensemble learning implementation, trained on experimentally determined binding affinity data. Extensive testing of our implementation on both existing and new manually curated datasets reveal that our method produces an unprecedented level of accuracy (92%) in identifying those residues assigned "heme binding" in all of the datasets used. Next, the machine learning implementation for the prediction and qualitative assignment of binding affinities to the predicted motifs achieved 71% accuracy on our data. CONCLUSIONS: Heme plays a crucial role as a regulatory molecule exerting functional consequences via transient binding to surfaces of target proteins. HeMoQuest is designed to address this imperative need for a computational approach that enables rapid detection of heme-binding motifs from protein datasets. While most existing implementations attempt to predict sites of permanent heme binding, this application is to the best of our knowledge, the first of its kind to address the significance of predicting transient heme binding to proteins.


Subject(s)
Amino Acid Motifs , Heme/metabolism , Software , Algorithms , Binding Sites , Internet , Machine Learning , Protein Binding , Sequence Analysis, Protein
3.
Sci Rep ; 9(1): 16893, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31729440

ABSTRACT

Cytokines of the interleukin (IL)-1 family regulate immune and inflammatory responses. The recently discovered IL-36 family members are involved in psoriasis, rheumatoid arthritis, and pulmonary diseases. Here, we show that IL-36α interacts with heme thereby contributing to its regulation. Based on in-depth spectroscopic analyses, we describe two heme-binding sites in IL-36α that associate with heme in a pentacoordinated fashion. Solution NMR analysis reveals structural features of IL-36α and its complex with heme. Structural investigation of a truncated IL-36α supports the notion that the N-terminus is necessary for association with its cognate receptor. Consistent with our structural studies, IL-36-mediated signal transduction was negatively regulated by heme in synovial fibroblast-like synoviocytes from rheumatoid arthritis patients. Taken together, our results provide a structural framework for heme-binding proteins and add IL-1 cytokines to the group of potentially heme-regulated proteins.


Subject(s)
Heme/metabolism , Interleukin-1/metabolism , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cells, Cultured , Cytokines/agonists , Cytokines/chemistry , Cytokines/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Inflammation Mediators/agonists , Inflammation Mediators/chemistry , Inflammation Mediators/metabolism , Interleukin-1/agonists , Interleukin-1/chemistry , Models, Molecular , Molecular Dynamics Simulation , Protein Binding , Protein Conformation , Psoriasis/metabolism , Psoriasis/pathology , Structure-Activity Relationship , Synovial Membrane/metabolism , Synovial Membrane/pathology
4.
J Magn Reson ; 308: 106561, 2019 11.
Article in English | MEDLINE | ID: mdl-31345774

ABSTRACT

The N-terminal segment of human cystathionine-ß-synthase (CBS(1-40)) constitutes an intrinsically disordered protein stretch that transiently interacts with heme. We illustrate that the HCBCACON experimental protocol provides an efficient alternative approach for probing transient interactions of intrinsically disordered proteins with heme in situations where the applicability of the conventional [1H, 15N]-HSQC experiment may be limited. This experiment starting with the excitation of protein side chain protons delivers information about the proline residues and thereby makes it possible to use these residues in interaction mapping experiments. Employing this approach in conjunction with site-specific mutation we show that transient heme binding is mediated by the Cys15-Pro16 motif of CBS(1-40).


Subject(s)
Cystathionine beta-Synthase/chemistry , Heme/chemistry , Intrinsically Disordered Proteins/chemistry , Magnetic Resonance Spectroscopy/methods , Peptides/chemistry , Humans , Recombinant Fusion Proteins/chemistry
5.
Biomol NMR Assign ; 13(1): 155-161, 2019 04.
Article in English | MEDLINE | ID: mdl-30758717

ABSTRACT

Interleukins are cytokines performing central tasks in the human immune system. Interleukin-36ß (IL-36ß) is a member of the interleukin-1 superfamily as are its homologues IL-36α and IL-36γ. All of them interact with a common receptor composed of IL-36R and IL-1R/acP. IL-36 cytokines can activate IL-36R to proliferation of CD4 + lymphocytes or stimulate M2 macrophages as potently as IL-1ß. Within our efforts to study the structure-function relationship of the three interleukins IL-36α, IL-36ß and IL-36γ by heteronuclear multidimensional NMR, we here report the 1H, 13C, and 15N resonance assignments for the backbone and side chain nuclei of cytokine interleukin-36ß isoform-2.


Subject(s)
Interleukin-1/chemistry , Nuclear Magnetic Resonance, Biomolecular , Amino Acid Sequence , Carbon Isotopes , Deuterium Exchange Measurement , Humans , Hydrogen Bonding , Nitrogen Radioisotopes , Protein Isoforms/chemistry , Protons , Temperature
6.
Biosci Rep ; 39(1)2019 01 31.
Article in English | MEDLINE | ID: mdl-30622148

ABSTRACT

Deviant levels of available heme and related molecules can result from pathological situations such as impaired heme biosynthesis or increased hemolysis as a consequence of vascular trauma or bacterial infections. Heme-related biological processes are affected by these situations, and it is essential to fully understand the underlying mechanisms. While heme has long been known as an important prosthetic group of various proteins, its function as a regulatory and signaling molecule is poorly understood. Diseases such as porphyria are caused by impaired heme metabolism, and heme itself might be used as a drug in order to downregulate its own biosynthesis. In addition, heme-driven side effects and symptoms emerging from heme-related pathological conditions are not fully comprehended and thus impede adequate medical treatment. Several heme-regulated proteins have been identified in the past decades, however, the molecular basis of transient heme-protein interactions remains to be explored. Herein, we summarize the results of an in-depth analysis of heme binding to proteins, which revealed specific binding modes and affinities depending on the amino acid sequence. Evaluating the binding behavior of a plethora of heme-peptide complexes resulted in the implementation of a prediction tool (SeqD-HBM) for heme-binding motifs, which eventually led and will perspectively lead to the identification and verification of so far unknown heme-regulated proteins. This systematic approach resulted in a broader picture of the alternative functions of heme as a regulator of proteins. However, knowledge on heme regulation of proteins is still a bottomless barrel that leaves much scope for future research and development.


Subject(s)
Heme/genetics , Hemeproteins/genetics , Multiprotein Complexes/genetics , Peptides/genetics , Amino Acid Sequence , Databases, Genetic , Heme/metabolism , Hemeproteins/metabolism , Humans , Multiprotein Complexes/chemistry , Peptides/chemistry , Protein Binding/genetics
7.
Biochim Biophys Acta Gen Subj ; 1862(9): 1964-1972, 2018 09.
Article in English | MEDLINE | ID: mdl-29908817

ABSTRACT

BACKGROUND: Tight regulation of heme homeostasis is a critical mechanism in pathogenic bacteria since heme functions as iron source and prosthetic group, but is also toxic at elevated concentrations. Hemolysin-activating lysine-acyltransferase (HlyC) from Escherichia coli is crucial for maturation of hemolysin A, which lyses several mammalian cells including erythrocytes liberating large amounts of heme for bacterial uptake. A possible impact and functional consequences of the released heme on events employing bacterial HlyC have remained unexplored. METHODS: Heme binding to HlyC was investigated using UV/vis and SPR spectroscopy. Functional impact of heme association was examined using an in vitro hemolysis assay. The interaction was further studied by homology modeling, molecular docking and dynamics simulations. RESULTS: We identified HlyC as potential heme-binding protein possessing heme-regulatory motifs. Using wild-type protein and a double alanine mutant we demonstrated that heme binds to HlyC via histidine 151 (H151). We could show further that heme inhibits the enzymatic activity of wild-type HlyC. Computational studies illustrated potential interaction sites in addition to H151 confirming the results from spectroscopy indicating more than one heme-binding site. CONCLUSIONS: Taken together, our results reveal novel insights into heme-protein interactions and regulation of a component of the heme uptake system in one of the major causative agents of urinary tract infections in humans. GENERAL SIGNIFICANCE: This study points to a possible novel mechanism of regulation as present in many uropathogenic E. coli strains at an early stage of heme iron acquisition from erythrocytes for subsequent internalization by the bacterial heme-uptake machinery.


Subject(s)
Acyltransferases/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Heme/metabolism , Hemolysin Proteins/metabolism , Lysine Acetyltransferases/metabolism , Acyltransferases/chemistry , Animals , Binding Sites , Erythrocytes/metabolism , Escherichia coli Proteins/chemistry , Heme/chemistry , Hemolysin Proteins/chemistry , Hemolysis , Lysine Acetyltransferases/chemistry , Sheep
8.
Sci Rep ; 8(1): 2474, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29410458

ABSTRACT

Cystathionine-ß-synthase (CBS) belongs to a large family of pyridoxal 5'-phosphate (PLP)-dependent enzymes, responsible for the sulfur metabolism. The heme-dependent protein CBS is part of regulatory pathways also involving the gasotransmitter hydrogen sulfide. Malfunction of CBS can lead to pathologic conditions like cancer, cardiovascular and neurodegenerative disorders. Truncation of residues 1-40, absent in X-ray structures of CBS, reduces but does not abolish the activity of the enzyme. Here we report the NMR resonance assignment and heme interaction studies for the N-terminal peptide stretch of CBS. We present NMR-spectral evidence that residues 1-40 constitute an intrinsically disordered region in CBS and interact with heme via a cysteine-proline based motif.


Subject(s)
Cystathionine beta-Synthase/chemistry , Heme/chemistry , Intrinsically Disordered Proteins/chemistry , Peptides/chemistry , Pyridoxal Phosphate/chemistry , Recombinant Fusion Proteins/chemistry , Amino Acid Sequence , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Cloning, Molecular , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Heme/metabolism , Humans , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/metabolism , Kinetics , Nuclear Magnetic Resonance, Biomolecular , Peptides/genetics , Peptides/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Pyridoxal Phosphate/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity , Thermodynamics
9.
Chembiochem ; 18(16): 1561-1564, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28547798

ABSTRACT

How free is heme? A fluorescent sensor has been used by Niles and co-workers to detect heme in P. falciparum. ECFP and EYFP were attached to its termini to give a FRET signal that was quenched by heme binding. The sensor was used to investigate alterations in the labile heme pool during the parasite's life cycle as well as under the influence of the drug chloroquine.


Subject(s)
Antigens, Protozoan/chemistry , Biosensing Techniques , Heme/analysis , Hemeproteins/chemistry , Luminescent Proteins/chemistry , Protozoan Proteins/chemistry , Biosensing Techniques/methods , Hemeproteins/genetics , Luminescent Proteins/genetics , Peptide Fragments/chemistry , Plasmodium falciparum/metabolism
10.
J Med Chem ; 60(1): 373-385, 2017 01 12.
Article in English | MEDLINE | ID: mdl-27976903

ABSTRACT

Labile heme has been suggested to have an impact in several severe diseases. In the context of Alzheimer's disease (AD), however, decreased levels of free heme have been reported. Therefore, we were looking for an assay system that can be used for heme concentration determination. From a biochemical point of view the peroxidase activity of the Aß-heme complex seemed quite attractive to pursue this goal. As a consequence, a peptide that is able to increase the readout even in the case of a low heme concentration is favorable. The examination of Aß- and non-Aß-derived peptides in complex with heme revealed that the peroxidase-like activity significantly depends on the peptide sequence and length. A 23mer His-based peptide derived from human fatty acyl-CoA reductase 1 in complex with heme exhibited a significantly higher peroxidase activity than Aß(40)-heme. Structural modeling of both complexes demonstrated that heme binding via a histidine can be supported by hydrogen bond interactions of a basic residue near the propionate carboxyl function of protoporphyrin IX. Furthermore, the interplay of Aß-heme and the lipoprotein LDL as a potential physiological effector of Aß was examined.


Subject(s)
Amyloid beta-Peptides/physiology , Heme/metabolism , Histidine/metabolism , Peroxidases/metabolism , Amino Acid Sequence , Amyloid beta-Peptides/chemistry , Binding Sites , Catalysis , Lipoproteins, LDL/metabolism , Models, Molecular , Peroxidases/chemistry , Spectrometry, Mass, Electrospray Ionization
11.
Biomol NMR Assign ; 10(2): 329-33, 2016 10.
Article in English | MEDLINE | ID: mdl-27351892

ABSTRACT

Interleukin-36α (IL-36α) is a recently characterised member of the interleukin-1 superfamily. It is involved in the pathogenesis of inflammatory arthritis in one third of psoriasis patients. By binding of IL-36α to its receptor IL-36R via the NF-κB pathway other cytokines involved in inflammatory and apoptotic cascade are activated. The efficacy of complex formation is controlled by N-terminal processing. To obtain a more detailed view on the structure function relationship we performed a heteronuclear multidimensional NMR investigation and here report the (1)H, (13)C, and (15)N resonance assignments for the backbone and side chain nuclei of the pro-inflammatory cytokine interleukin-36α.


Subject(s)
Interleukin-1/chemistry , Interleukin-1/metabolism , Nuclear Magnetic Resonance, Biomolecular , Inflammation/metabolism
12.
ACS Chem Biol ; 8(8): 1785-93, 2013 Aug 16.
Article in English | MEDLINE | ID: mdl-23730736

ABSTRACT

Regulatory heme binds to specific motifs in proteins and controls a variety of biochemical processes. Several of these proteins were recently shown to form complexes with ferric and/or ferrous heme via a cysteine residue as axial ligand. The objective of this study was to examine the heme-binding properties of a series of cysteine-containing peptides with focus on CP motif sequences. The peptides displayed different binding behavior upon Fe(III) heme application with characteristic wavelength shifts of the Soret band to 370 nm or 420-430 nm and in some cases to both wavelengths. Whereas for most of the peptides containing a cysteine only a shift to 420-430 nm was observed, CP-containing peptides exhibited a preference for a shift to 370 nm. Detailed structural investigation using Raman and NMR spectroscopy on selected representatives revealed different binding modes with respect to iron ion coordination, which reflected the results of the UV-vis studies. A predicted short sequence stretch derived from dipeptidyl peptidase 8 was additionally examined with respect to CP motif binding to heme on the peptide as well as on the protein level. The heme association was confirmed with the first solution structure of a CP-peptide-heme complex and, moreover, an inhibitory effect of Fe(III) heme on the enzyme's activity. The relevance of both the use of model compounds to elucidate the molecular mechanism underlying regulatory heme binding and its potential for the investigation of regulatory heme control is discussed.


Subject(s)
Cysteine/chemistry , Ferric Compounds/chemistry , Heme/chemistry , Proteins/chemistry , Amino Acid Sequence , Magnetic Resonance Spectroscopy , Molecular Structure , Peptides/chemistry , Peptides/genetics , Protein Binding , Protein Structure, Tertiary , Spectrum Analysis, Raman
SELECTION OF CITATIONS
SEARCH DETAIL
...