31 results on '"Kourosh Honarmand Ebrahimi"'
Search Results
2. The amyloid precursor protein (APP) does not have a ferroxidase site in its E2 domain.
- Author
-
Kourosh Honarmand Ebrahimi, Christian Dienemann, Sandra Hoefgen, Manuel E Than, Peter-Leon Hagedoorn, and Wilfred R Hagen
- Subjects
Medicine ,Science - Abstract
The ubiquitous 24-meric iron-storage protein ferritin and multicopper oxidases such as ceruloplasmin or hephaestin catalyze oxidation of Fe(II) to Fe(III), using molecular oxygen as oxidant. The ferroxidase activity of these proteins is essential for cellular iron homeostasis. It has been reported that the amyloid precursor protein (APP) also has ferroxidase activity. The activity is assigned to a ferroxidase site in the E2 domain of APP. A synthetic 22-residue peptide that carries the putative ferroxidase site of E2 domain (FD1 peptide) has been claimed to encompass the same activity. We previously tested the ferroxidase activity of the synthetic FD1 peptide but we did not observe any activity above the background oxidation of Fe(II) by molecular oxygen. Here we used isothermal titration calorimetry to study Zn(II) and Fe(II) binding to the natural E2 domain of APP, and we employed the transferrin assay and oxygen consumption measurements to test the ferroxidase activity of the E2 domain. We found that this domain neither in the presence nor in the absence of the E1 domain binds Fe(II) and it is not able to catalyze the oxidation of Fe(II). Binding of Cu(II) to the E2 domain did not induce ferroxidase activity contrary to the presence of redox active Cu(II) centers in ceruloplasmin or hephaestin. Thus, we conclude that E2 or E1 domains of APP do not have ferroxidase activity and that the potential involvement of APP as a ferroxidase in the pathology of Alzheimer's disease must be re-evaluated.
- Published
- 2013
- Full Text
- View/download PDF
3. A synthetic peptide with the putative iron binding motif of amyloid precursor protein (APP) does not catalytically oxidize iron.
- Author
-
Kourosh Honarmand Ebrahimi, Peter-Leon Hagedoorn, and Wilfred R Hagen
- Subjects
Medicine ,Science - Abstract
The β-amyloid precursor protein (APP), which is a key player in Alzheimer's disease, was recently reported to possess an Fe(II) binding site within its E2 domain which exhibits ferroxidase activity [Duce et al. 2010, Cell 142: 857]. The putative ligands of this site were compared to those in the ferroxidase site of ferritin. The activity was indirectly measured using transferrin, which scavenges the Fe(III) product of the reaction. A 22-residue synthetic peptide, named FD1, with the putative ferroxidase site of APP, and the E2 domain of APP were each reported to exhibit 40% of the ferroxidase activity of APP and of ceruloplasmin. It was also claimed that the ferroxidase activity of APP is inhibited by Zn(II) just as in ferritin. We measured the ferroxidase activity indirectly (i) by the incorporation of the Fe(III) product of the ferroxidase reaction into transferrin and directly (ii) by monitoring consumption of the substrate molecular oxygen. The results with the FD1 peptide were compared to the established ferroxidase activities of human H-chain ferritin and of ceruloplasmin. For FD1 we observed no activity above the background of non-enzymatic Fe(II) oxidation by molecular oxygen. Zn(II) binds to transferrin and diminishes its Fe(III) incorporation capacity and rate but it does not specifically bind to a putative ferroxidase site of FD1. Based on these results, and on comparison of the putative ligands of the ferroxidase site of APP with those of ferritin, we conclude that the previously reported results for ferroxidase activity of FD1 and - by implication - of APP should be re-evaluated.
- Published
- 2012
- Full Text
- View/download PDF
4. Bioinorganic Chemistry of Metals in Cell Function and Disease
- Author
-
Kourosh Honarmand Ebrahimi, Simone Ciofi Baffoni, and Peter-Leon Hagedoorn
- Published
- 2023
- Full Text
- View/download PDF
5. Iron–sulfur clusters as inhibitors and catalysts of viral replication
- Author
-
Kourosh Honarmand Ebrahimi, Simone Ciofi-Baffoni, Peter-Leon Hagedoorn, Yvain Nicolet, Nick E. Le Brun, Wilfred R. Hagen, Fraser A. Armstrong, Institute of Pharmaceutical Science, Kings' College London, Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine, Università degli Studi di Firenze = University of Florence (UniFI), Delft University of Technology (TU Delft), Institut de biologie structurale (IBS - UMR 5075), Centre National de la Recherche Scientifique (CNRS)-Institut de Recherche Interdisciplinaire de Grenoble (IRIG), Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Grenoble Alpes (UGA), Centre for Molecular and Structural Biochemistry, University of East Anglia [Norwich] (UEA), Inorganic Chemistry Laboratory [Oxford], and University of Oxford
- Subjects
Iron-Sulfur Proteins ,[SDV.BBM.BS]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Structural Biology [q-bio.BM] ,General Chemical Engineering ,Iron ,General Chemistry ,Virus Replication ,Catalysis ,Sulfur - Abstract
International audience; A virus hijacks host cellular machineries and metabolites in order to reproduce. In response, the innate immune system activates different processes to fight back. Although many aspects of these processes have been well investigated, the key roles played by iron-sulfur [FeS] clusters, which are among the oldest classes of bio-inorganic cofactors, have barely been considered. Here we discuss how several [FeS] cluster-containing proteins activate, support and modulate the innate immune response to restrict viral infections, and how some of these proteins simultaneously support the replication of viruses. We also propose models of function of some proteins in the innate immune response and argue that [FeS] clusters in many of these proteins act as biological 'fuses' to control the response. We hope this overview helps to inspire future research in the emerging field of bio-inorganic virology/immunology and that such studies may reveal new molecular insight into the links between viral infections and diseases like cancer and neurodegeneration.
- Published
- 2022
- Full Text
- View/download PDF
6. Hidden in Plain Sight: Natural Products of Commensal Microbiota as an Environmental Selection Pressure for the Rise of New Variants of SARS-CoV-2
- Author
-
Kourosh Honarmand Ebrahimi, Maria Andrea Mroginski, and Jovan Dragelj
- Subjects
Siderophore ,Ecological selection ,natural products ,NRPs ,Biology ,medicine.disease_cause ,Biochemistry ,Virus ,medicine ,Animals ,Humans ,Computer Simulation ,Protein Interaction Domains and Motifs ,Receptor ,Molecular Biology ,bile acids ,chemistry.chemical_classification ,Infectivity ,Genetics ,Mutation ,Biological Products ,Bacteria ,SARS-CoV-2 ,Communication ,Microbiota ,Organic Chemistry ,COVID-19 ,Genetic Variation ,commensal microbiota ,biology.organism_classification ,Communications ,chemistry ,Molecular Medicine ,Receptors, Virus ,Glycoprotein - Abstract
Since the emergence of SARS‐CoV‐2, little attention has been paid to the interplay between the interaction of virus and commensal microbiota. Here, we used molecular docking and dynamics simulations to study the interaction of some of the known metabolites and natural products (NPs) produced by commensal microbiota with the receptor binding domain (RBD) of the spike glycoprotein of SARS‐CoV‐2. The results predict that NPs of commensal microbiota such as bile acids and non‐ribosomal peptides (NRPs), of which some are siderophores, bind to the wild‐type RBD and interfere with its binding to the ACE2 receptor. N501Y mutation, which is present in many of the emerging variants of the virus, abolishes the predicted binding pocket of bile acids and NRPs. Based on these findings, available experimental data showing that bile acids reduce the binding affinity of wild‐type RBD to the ACE2 receptor, and the data suggesting that the respiratory tract microbiota affect viral infection we put forward the following proposal: mutations such as N501Y enable the RBD to bind to the ACE2 receptor more effectively in the presence of NPs produced by the respiratory tract bacteria thereby, increasing the infectivity rate of the virus. We hope our data stimulate future works to better understand the interactions of NPs produced by commensal microbiota with respiratory viruses like SARS‐CoV‐2., In silico studies suggest that NPs like bile acids and non‐ribosomal peptides (NRPs) produced by bacteria residing in the respiratory tract can interfere with the binding of the original strain of SARS‐CoV‐2 to the ACE2 receptor but not with the binding of the emerging variants having the N501Y mutation. Thus, it is hypothesized that NPs of respiratory tract bacteria are an environmental selection pressure for the rise of new variants of the virus with higher infectivity rate.
- Published
- 2021
7. A lipidomic view of SARS-CoV-2
- Author
-
Kourosh Honarmand Ebrahimi and James S. O. McCullagh
- Subjects
Immunology & Inflammation ,Coronavirus disease 2019 (COVID-19) ,HDL ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,viruses ,Biophysics ,Disease ,Biochemistry ,Viral infection ,lipids ,Immune system ,Economic cost ,Virology ,Commentaries ,Pandemic ,Medicine ,Homeostasis ,Humans ,Metabolomics ,Molecular Biology ,Pandemics ,business.industry ,SARS-CoV-2 ,COVID-19 ,Lipid metabolism ,Cell Biology ,Lipid Metabolism ,Metabolism ,Immunology ,Lipidomics ,business - Abstract
The global pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which started in late 2019, has caused huge social and economic losses. A growing number of investigators are focusing on understanding the interaction of SARS-CoV-2 with host cellular processes to find therapeutic approaches. New data suggest that lipid metabolism may play a significant role in regulating the response of immune cells like macrophages to viral infection, thereby affecting the outcome of the disease. Therefore, understanding the role of lipid metabolism could help develop new therapeutic approaches to mitigate the social and economic cost of coronavirus disease 2019 (COVID-19).
- Published
- 2021
8. Randomised Controlled Trial of Intravenous Nafamostat Mesylate in COVID Pneumonitis: Phase 1b/2a Experimental Study to Investigate Safety, Pharmacokinetics and Pharmacodynamics
- Author
-
Oliver Koch, Andrew Duncan, Jean Antonelli, Timothy S. Walsh, Annya Bruce, Kay Russell, Colin Ferrett, Colin Church, James S. O. McCullagh, Jürgen Schwarze, Cecilia Boz, Giulia Rinaldi, Sinéad Plant, Gareth Hardisty, Anne Moore, Ahsan R. Akram, Kourosh Honarmand Ebrahimi, Sarah McNamara, Feng Li, Kevin G. Blyth, K. Dhaliwal, David H. Dockrell, Asta Valanciute, Ross Mills, Bethany Mills, Claire L. Mackintosh, Daniel C. Anthony, Tom Quinn, Steve P. Rannard, John Norrie, Irene Young, Keith Finlayson, Kate Templeton, Alastair F. Nimmo, Richard A. O’Connor, Erin Gaughan, Emma Scholefield, James W. Dear, Richard Parker, Andrew Owen, Philip Emanuel, Manu Shankar-Hari, Grant C. Churchill, Adam Marshall, Nik Hirani, Islom B. Nazarov, Matthew Burgess, Emily Gwyer Findlay, and Sunny Jabball
- Subjects
Male ,Medicine (General) ,History ,Polymers and Plastics ,Kaplan-Meier Estimate ,Guanidines ,Industrial and Manufacturing Engineering ,law.invention ,Randomized controlled trial ,Informed consent ,law ,SARS-CoV-2/COVID-19 ,Nafamostat mesylate ,Medicine ,Aged, 80 and over ,education.field_of_study ,Anti-Inflammatory Agents, Non-Steroidal ,General Medicine ,Middle Aged ,Viral Load ,Nafamostat ,Thromboelastometry ,Total manuscript word count ,Treatment Outcome ,Anesthesia ,Infectious diseases ,Administration, Intravenous ,Female ,Half-Life ,Adult ,medicine.medical_specialty ,Population ,Drug Administration Schedule ,General Biochemistry, Genetics and Molecular Biology ,Immunophenotyping ,Respiratory medicine ,R5-920 ,Pharmacokinetics ,Internal medicine ,Humans ,Business and International Management ,education ,Aged ,Pneumonitis ,SARS-CoV-2 ,business.industry ,COVID-19 ,medicine.disease ,Benzamidines ,COVID-19 Drug Treatment ,Clinical trial ,Pharmacodynamics ,business ,Biomarkers - Abstract
Despite the success of vaccines and selected repurposed treatments, COVID-19 is likely to remain a global health problem and further chemotherapeutics are required. Many repurposed drugs have progressed rapidly to Phase 2 and 3 trials without characterisation of Pharmacokinetics (PK)/Pharmacodynamics (PD) including safety in COVID-19. One such drug is Nafamostat Mesylate (Nafamostat), a synthetic serine protease inhibitor with anticoagulant and anti-inflammatory properties. Preclinical data has demonstrated that it is has potent antiviral activity against SARS-CoV-2 by directly inhibiting the transmembrane protease serine 2 (TMPRSS2) dependent stage of host cell entry.MethodsWe present the findings of a phase Ib/II open label, platform randomised controlled trial (RCT), exploring the safety of intravenous Nafamostat in hospitalised patients with confirmed COVID-19 pneumonitis. Patients were assigned randomly to standard of care (SoC), Nafamostat or an alternative therapy. Secondary endpoints included clinical endpoints such as number of oxygen free days and clinical improvement/ deterioration, PK/PD, thromboelastometry, D Dimers, cytokines, immune cell flow cytometry and viral load.ResultsData is reported from 42 patients, 21 of which were randomly assigned to receive intravenous Nafamostat. The Nafamostat group developed significantly higher plasma creatinine levels, more adverse events and a lower number of oxygen free days. There were no other statistically significant differences in the primary or secondary endpoints between Nafamostat and SoC. PK data demonstrated that intravenous Nafamostat was rapidly broken down to inactive metabolites. We observed an antifibrinolytic profile, and no significant anticoagulant effects in thromboelastometry. Participants in the Nafamostat group had higher D Dimers compared to SoC. There were no differences in cytokine profile and immune cell phenotype and viral loads between the groups.ConclusionIn hospitalised patients with COVID-19, we did not observe evidence of anti-inflammatory, anticoagulant or antiviral activity with intravenous Nafamostat. Given the number of negative trials with repurposed drugs, our experimental medicine trial highlights the value of PK/PD studies prior to selecting drugs for efficacy trials. Given the mechanism of action, further evaluation of Nafamostat delivered via a different route may be warranted. This trial demonstrates the importance of experimental trials in new disease entities such as COVID-19 prior to selecting drugs for larger trials.
- Published
- 2021
- Full Text
- View/download PDF
9. Ferritin as a Platform for Creating Antiviral Mosaic Nanocages: Prospects for Treating COVID‐19
- Author
-
Kourosh Honarmand Ebrahimi
- Subjects
COVID-19 Vaccines ,Coronavirus disease 2019 (COVID-19) ,Disease ,010402 general chemistry ,01 natural sciences ,Antiviral Agents ,Biochemistry ,Virus ,Antibodies ,Mice ,Nanocages ,Viral entry ,Pandemic ,Animals ,Humans ,Molecular Biology ,Drug Carriers ,biology ,010405 organic chemistry ,SARS-CoV-2 ,Serine Endopeptidases ,Organic Chemistry ,COVID-19 ,Virus Internalization ,Virology ,0104 chemical sciences ,Nanostructures ,COVID-19 Drug Treatment ,Ferritin ,Ferritins ,biology.protein ,Molecular Medicine ,Angiotensin-Converting Enzyme 2 ,Antibody therapy - Abstract
Infectious diseases are a continues threat to human health and the economy worldwide. The latest example is the global pandemic of COVID-19 caused by SARS-CoV-2. Antibody therapy and vaccines are promising approaches to treat the disease; however, they have bottlenecks: they might have low efficacy or narrow breadth due to the continuous emergence of new strains of the virus or antibodies could cause antibody-dependent enhancement (ADE) of infection. To address these bottlenecks, I propose the use of 24-meric ferritin for the synthesis of mosaic nanocages to deliver a cocktail of antibodies or nanobodies alone or in combination with another therapeutic, like a nucleotide analogue, to mimic the viral entry process and deceive the virus, or to develop mosaic vaccines. I argue that available data showing the effectiveness of ferritin-antibody conjugates in targeting specific cells and ferritin-haemagglutinin nanocages in developing influenza vaccines strongly support my proposals.
- Published
- 2020
- Full Text
- View/download PDF
10. Interferon‐stimulated gene products as regulators of central carbon metabolism
- Author
-
Kourosh Honarmand Ebrahimi, James S. O. McCullagh, Javier Gilbert-Jaramillo, and William James
- Subjects
0301 basic medicine ,Oxidoreductases Acting on CH-CH Group Donors ,S-Adenosylmethionine ,Induced Pluripotent Stem Cells ,immunometabolism ,Virus Replication ,Antiviral Agents ,Models, Biological ,Biochemistry ,ISG ,03 medical and health sciences ,Viewpoint ,0302 clinical medicine ,Immune system ,Viral life cycle ,Humans ,viruses ,Molecular Biology ,Cells, Cultured ,Glyceraldehyde 3-phosphate dehydrogenase ,chemistry.chemical_classification ,Innate immune system ,biology ,SARS-CoV-2 ,GAPDH ,Macrophages ,Interferon-stimulated gene ,COVID-19 ,Proteins ,Cell Biology ,Carbon ,Immunity, Innate ,Cell biology ,Nitric oxide synthase ,HEK293 Cells ,030104 developmental biology ,Enzyme ,chemistry ,030220 oncology & carcinogenesis ,Viperin ,biology.protein ,Interferons ,Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating) ,viperin - Abstract
ISG products are believed to directly block viral replication. We propose an alternative mechanism based on emerging evidence. We discuss that the metabolic products of two ISG proteins, namely RSAD2 and NOS, inhibit activity of GAPDH to regulate central carbon metabolism and support a broad‐spectrum antiviral immune response via at least four mechanisms: eicosanoids storm, antigen cross‐presentation via MHC‐I, modulating NFAT and NF‐κB pathways, and S‐nitrosylation of viral proteins., In response to viral infections, the innate immune system rapidly activates expression of several interferon‐stimulated genes (ISGs), whose protein and metabolic products are believed to directly interfere with the viral life cycle. Here, we argue that biochemical reactions performed by two specific protein products of ISGs modulate central carbon metabolism to support a broad‐spectrum antiviral response. We demonstrate that the metabolites generated by metalloenzymes nitric oxide synthase and the radical S‐adenosylmethionine (SAM) enzyme RSAD2 inhibit the activity of the housekeeping and glycolytic enzyme glyceraldehyde 3‐phosphate dehydrogenase (GAPDH). We discuss that this inhibition is likely to stimulate a range of metabolic and signalling processes to support a broad‐spectrum immune response. Based on these analyses, we propose that inhibiting GAPDH in individuals with deteriorated cellular innate immune response like elderly might help in treating viral diseases such as COVID‐19.
- Published
- 2020
- Full Text
- View/download PDF
11. Differentiation of Human induced Pluripotent Stem Cells to Authentic Macrophages using Fully Defined, Serum Free, Open Source Media
- Author
-
Paul K. Reardon, Kourosh Honarmand Ebrahimi, William James, Alun Vaughan-Jackson, Cathy Browne, Sally A. Cowley, Szymon Stodolak, Javier Gilbert-Jaramillo, and Elisabete Pires
- Subjects
Chemically defined medium ,Open source ,Serum free ,Cellular differentiation ,Human Induced Pluripotent Stem Cells ,Biology ,Cell biology - Abstract
SummaryHuman iPSC and macrophages derived from them are increasingly popular tools for research into both infectious and degenerative diseases. However, as the field strives for greater modelling accuracy, it is becoming ever more challenging to justify the use of undefined and proprietary media for the culture of these cells. We describe here two fully defined, serum-free, open-source media for the culture of iPSC and differentiation of iPSC-derived macrophages. These media are equally capable of maintaining these cells compared to commercial alternatives. The macrophages differentiated in these defined media display improved terminally differentiated cell characteristics, reduced basal expression of induced anti-viral response genes, and improved polarisation capacity. We conclude that cells cultured in these media are an appropriate and malleable model for tissue resident macrophages, on which future differentiation techniques can be built.
- Published
- 2020
- Full Text
- View/download PDF
12. A Chemical-Physical View of the Interpretation of Wavefunction
- Author
-
Kourosh Honarmand Ebrahimi
- Abstract
Here, based on different experimental setups and thought experiments it is shown that some of the predictions of the nonlocal probabilistic interpretation of wavefunction are not observed and that this interpretation does not account for the real-world physical and chemical interactions. Considering these interactions, classical and quantum mechanics are unified and a testable, local, and probabilistic interpretation of wavefunction is proposed in explaining the outcome of a double-slit experiment. This analysis suggests that there is no spooky action at a distance.
- Published
- 2020
- Full Text
- View/download PDF
13. ddhCTP produced by the radical-SAM activity of RSAD2 (viperin) inhibits the NAD
- Author
-
Kourosh, Honarmand Ebrahimi, Jane, Vowles, Cathy, Browne, James, McCullagh, and William S, James
- Subjects
Models, Molecular ,Oxidoreductases Acting on CH-CH Group Donors ,Binding Sites ,L-Lactate Dehydrogenase ,Cytidine Triphosphate ,Macrophages ,Induced Pluripotent Stem Cells ,Proteins ,NAD ,Adenosine Diphosphate ,HEK293 Cells ,Malate Dehydrogenase ,Humans ,Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating) - Abstract
Radical S-adenosylmethionine (SAM) domain-containing protein 2 (RSAD2; viperin) is a key enzyme in innate immune responses that is highly expressed in response to viral infection and inflammatory stimuli in many cell types. Recently, it was found that RSAD2 catalyses transformation of cytidine triphosphate (CTP) to its analogue 3'-deoxy-3',4'-didehydro-CTP (ddhCTP). The cellular function of this metabolite is unknown. Here, we analysed the extra- and intracellular metabolite levels in human induced pluripotent stem cell (hiPSC)-derived macrophages using high-resolution LC-MS/MS. The results together with biochemical assays and molecular docking simulations revealed that ddhCTP inhibits the NAD
- Published
- 2020
14. Differentiation of human induced pluripotent stem cells to authentic macrophages using a defined, serum-free, open-source medium
- Author
-
Szymon Stodolak, Alun Vaughan-Jackson, Javier Gilbert-Jaramillo, Elisabete Pires, William James, Cathy Browne, Paul K. Reardon, Kourosh Honarmand Ebrahimi, and Sally A. Cowley
- Subjects
0301 basic medicine ,induced pluripotent stem cell ,transcriptional analysis ,Transcription, Genetic ,Cellular differentiation ,Induced Pluripotent Stem Cells ,HIV Infections ,macrophage ,Biology ,Biochemistry ,Article ,Culture Media, Serum-Free ,03 medical and health sciences ,0302 clinical medicine ,Serum free ,disease modeling ,Genetics ,Macrophage ,Homeostasis ,Humans ,human ,Transcriptional analysis ,Human Induced Pluripotent Stem Cells ,Induced pluripotent stem cell ,Gene ,Cell Shape ,Cells, Cultured ,Macrophages ,Correction ,Cell Differentiation ,differentiation ,Zika Virus ,Cell Biology ,Macrophage Activation ,culture ,Cell biology ,030104 developmental biology ,Open source ,Phenotype ,Transcriptome ,030217 neurology & neurosurgery ,Biomarkers ,Developmental Biology - Abstract
Summary Human induced pluripotent stem cells (iPSCs) and macrophages derived from them are increasingly popular tools for research into both infectious and degenerative diseases. However, as the field strives for greater modeling accuracy, it is becoming ever more challenging to justify the use of undefined and proprietary media for the culture of these cells. Here, we describe a defined, serum-free, open-source medium for the differentiation of iPSC-derived macrophages. This medium is equally capable of maintaining these cells compared with commercial alternatives. The macrophages differentiated in this medium display improved terminally differentiated cell characteristics, reduced basal expression of induced antiviral response genes, and improved polarization capacity. We conclude that cells cultured in this medium are an appropriate and malleable model for tissue-resident macrophages, on which future differentiation techniques can be built., Highlights • Transparency in composition of iPSC-derived macrophage culture medium • Transcriptomic analysis of iPSC-derived macrophages • Improved terminal differentiation of iPSC-derived macrophages, Vaughan-Jackson et al. sought to bring transparency and malleability to the composition of growth media for human iPSC-derived macrophages. They describe a new, simple, defined, and open-sourced medium which supports the growth of macrophages. Building upon previous methodologies for iPSC-derived macrophage differentiation, they improve terminal differentiation and provide fully competent authentic tissue-derived macrophages.
- Published
- 2021
- Full Text
- View/download PDF
15. Spectroscopic evidence for the presence of a high-valent Fe(IV) species in the ferroxidase reaction of an archaeal ferritin
- Author
-
Wilfred R. Hagen, Kourosh Honarmand Ebrahimi, Eckhard Bill, and Peter-Leon Hagedoorn
- Subjects
Models, Molecular ,Methane monooxygenase ,Archaeal Proteins ,Iron ,Tyrosine radical ,Inorganic chemistry ,Biophysics ,010402 general chemistry ,01 natural sciences ,Biochemistry ,Cofactor ,Spectroscopy, Mossbauer ,Structural Biology ,Catalytic Domain ,tyrosine radical ,Genetics ,Molecular Biology ,chemistry.chemical_classification ,biology ,010405 organic chemistry ,Chemistry ,ferritin ,Electron Spin Resonance Spectroscopy ,Ceruloplasmin ,Cell Biology ,Hypothesis ,0104 chemical sciences ,Pyrococcus furiosus ,Ferritin ,Fe(IV) ,peroxodiferric ,Crystallography ,Enzyme ,Catalytic cycle ,Spectrophotometry ,Ferritins ,Enzymology ,Biocatalysis ,biology.protein ,Tyrosine ,ferroxidase ,Oxidation-Reduction - Abstract
A high-valent Fe(IV) species is proposed to be generated from the decay of a peroxodiferric intermediate in the catalytic cycle at the di-iron cofactor center of dioxygen-activating enzymes such as methane monooxygenase. However, it is believed that this intermediate is not formed in the di-iron substrate site of ferritin, where oxidation of Fe(II) substrate to Fe(III) (the ferroxidase reaction) occurs also via a peroxodiferric intermediate. In opposition to this generally accepted view, here we present evidence for the occurrence of a high-valent Fe(IV) in the ferroxidase reaction of an archaeal ferritin, which is based on trapped intermediates obtained with the freeze-quench technique and combination of spectroscopic characterization. We hypothesize that a Fe(IV) intermediate catalyzes oxidation of excess Fe(II) nearby the ferroxidase center.
- Published
- 2017
- Full Text
- View/download PDF
16. Viperin, through its radical-SAM activity, depletes cellular nucleotide pools and interferes with mitochondrial metabolism to inhibit viral replication
- Author
-
Kourosh Honarmand Ebrahimi, William James, Fraser A. Armstrong, Duncan Howie, James S. O. McCullagh, and Jack S. Rowbotham
- Subjects
Oxidoreductases Acting on CH-CH Group Donors ,Cytidine triphosphate ,Cytidine Triphosphate ,Cell Respiration ,Biophysics ,Uridine Triphosphate ,Mitochondrion ,Virus Replication ,Biochemistry ,Antiviral Agents ,03 medical and health sciences ,chemistry.chemical_compound ,Inhibitory Concentration 50 ,Structural Biology ,Genetics ,Humans ,Nucleotide ,Molecular Biology ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Chemistry ,Nucleotides ,030302 biochemistry & molecular biology ,Proteins ,Cell Biology ,Cell biology ,Mitochondria ,Terminator (genetics) ,Viral replication ,Viperin ,Nucleoside ,Radical SAM - Abstract
Viperin (RSAD2) is an antiviral radical S-adenosylmethionine (SAM) enzyme highly expressed in different cell types upon viral infection. Recently, it has been reported that the radical-SAM activity of viperin transforms cytidine triphosphate (CTP) to its analogue 3'-deoxy-3',4'-didehydro-CTP (ddhCTP). Based on biochemical studies and cell biological experiments, it was concluded that ddhCTP and its nucleoside form ddhC do not affect the cellular concentration of nucleotide triphosphates and that ddhCTP acts as replication chain terminator. However, our re-evaluation of the reported data and new results indicate that ddhCTP is not an effective viral chain terminator but depletes cellular nucleotide pools and interferes with mitochondrial activity to inhibit viral replication. Our analysis is consistent with a unifying view of the antiviral and radical-SAM activities of viperin.
- Published
- 2020
17. The workings of ferritin: a crossroad of opinions
- Author
-
Wilfred R. Hagen, Kourosh Honarmand Ebrahimi, and Peter-Leon Hagedoorn
- Subjects
0301 basic medicine ,biology ,Chemistry ,Iron ,Metals and Alloys ,Biophysics ,Oxidation reduction ,010402 general chemistry ,01 natural sciences ,Biochemistry ,Cell Physiological Phenomena ,0104 chemical sciences ,Biomaterials ,Ferritin ,03 medical and health sciences ,030104 developmental biology ,Extant taxon ,Chemistry (miscellaneous) ,Computational chemistry ,Ferritins ,biology.protein ,Humans ,Oxidation-Reduction - Abstract
Biochemistry of the essential element iron is complicated by radical chemistry associated with Fe(II) ions and by the extremely low solubility of the Fe(III) ion in near-neutral water. To mitigate these problems cells from all domains of life synthesize the protein ferritin to take up and oxidize Fe(II) and to form a soluble storage of Fe(III) from which iron can be made available for physiology. A long history of studies on ferritin has not yet resulted in a generally accepted mechanism of action of this enzyme. In fact strong disagreement exists between extant ideas on several key steps in the workings of ferritin. The scope of this review is to explain the experimental background of these controversies and to indicate directions towards their possible resolution.
- Published
- 2017
- Full Text
- View/download PDF
18. Cover Feature: Mechanism of Diol Dehydration by a Promiscuous Radical‐SAM Enzyme Homologue of the Antiviral Enzyme Viperin (RSAD2) (ChemBioChem 11/2020)
- Author
-
Kourosh Honarmand Ebrahimi, Jack S. Rowbotham, James S. O. McCullagh, and William James
- Subjects
chemistry.chemical_classification ,Stereochemistry ,Organic Chemistry ,Diol ,medicine.disease ,Biochemistry ,chemistry.chemical_compound ,Enzyme ,chemistry ,Viperin ,medicine ,Molecular Medicine ,Dehydration ,Molecular Biology ,Radical SAM - Published
- 2020
- Full Text
- View/download PDF
19. The radical-SAM enzyme Viperin catalyzes reductive addition of a 5'-deoxyadenosyl radical to UDP-glucose in vitro
- Author
-
James S. O. McCullagh, James Cantley, James Wickens, Nicholas H. Rees, Stephen B. Carr, Fraser A. Armstrong, and Kourosh Honarmand Ebrahimi
- Subjects
0301 basic medicine ,Uridine Diphosphate Glucose ,S-Adenosylmethionine ,Free Radicals ,Stereochemistry ,Protein Conformation ,Biophysics ,Sordariales ,010402 general chemistry ,01 natural sciences ,Biochemistry ,Conserved sequence ,Fungal Proteins ,03 medical and health sciences ,Protein structure ,Structural Biology ,Genetics ,Amino Acid Sequence ,Molecular Biology ,Peptide sequence ,Conserved Sequence ,chemistry.chemical_classification ,Deoxyadenosines ,Cell Biology ,Nuclear magnetic resonance spectroscopy ,0104 chemical sciences ,Amino acid ,Molecular Docking Simulation ,030104 developmental biology ,chemistry ,Docking (molecular) ,Viperin ,Biocatalysis ,Radical SAM ,Oxidation-Reduction ,Hydrogen - Abstract
Viperin, a radical-SAM enzyme conserved from fungi to humans, can restrict replication of many viruses. Neither the molecular mechanism underlying the antiviral activity of Viperin, nor its exact physiological function, is understood: most importantly, no radical-SAM activity has been discovered for Viperin. Here, using electron paramagnetic resonance (EPR) spectroscopy, mass spectrometry, and NMR spectroscopy, we show that UDP-glucose is a substrate of a fungal Viperin (58% pairwise identity with human Viperin at the amino acid level) in vitro. Structural homology modelling and docking experiments reveal a highly conserved binding pocket in which the position of UDP-glucose is consistent with our experimental data regarding catalytic addition of a 5′-deoxyadenosyl radical and a hydrogen atom to UDP-glucose. This article is protected by copyright. All rights reserved.
- Published
- 2017
20. Fast and accurate enzyme activity measurements using a chip-based microfluidic calorimeter
- Author
-
Kourosh Honarmand Ebrahimi, Peter-Leon Hagedoorn, Morten M. C. H. van Schie, and Wilfred R. Hagen
- Subjects
0301 basic medicine ,Materials science ,Microfluidics ,Biophysics ,Nanotechnology ,Calorimetry ,Incomplete mixing ,Protein Engineering ,01 natural sciences ,Biochemistry ,Diffusion ,Phosphate ionization ,03 medical and health sciences ,Enzyme calorimetry ,Alkaline phosphatase ,Miniaturization ,Calibration ,Chemical calibration method ,Animals ,Molecular Biology ,010401 analytical chemistry ,Cell Biology ,Microfluidic Analytical Techniques ,Chip ,High-Throughput Screening Assays ,0104 chemical sciences ,Calorimeter ,030104 developmental biology ,Product inhibition ,Chip-based calorimetry ,Cattle - Abstract
Recent developments in microfluidic and nanofluidic technologies have resulted in development of new chip-based microfluidic calorimeters with potential use in different fields. One application would be the accurate high-throughput measurement of enzyme activity. Calorimetry is a generic way to measure activity of enzymes, but unlike conventional calorimeters, chip-based calorimeters can be easily automated and implemented in high-throughput screening platforms. However, application of chip-based microfluidic calorimeters to measure enzyme activity has been limited due to problems associated with miniaturization such as incomplete mixing and a decrease in volumetric heat generated. To address these problems we introduced a calibration method and devised a convenient protocol for using a chip-based microfluidic calorimeter. Using the new calibration method, the progress curve of alkaline phosphatase, which has product inhibition for phosphate, measured by the calorimeter was the same as that recorded by UV-visible spectroscopy. Our results may enable use of current chip-based microfluidic calorimeters in a simple manner as a tool for high-throughput screening of enzyme activity with potential applications in drug discovery and enzyme engineering.
- Published
- 2017
21. Unity in the Biochemistry of the Iron-Storage Proteins Ferritin and Bacterioferritin
- Author
-
Wilfred R. Hagen, Kourosh Honarmand Ebrahimi, and Peter-Leon Hagedoorn
- Subjects
Models, Molecular ,biology ,Biochemical Phenomena ,Chemistry ,General Chemistry ,Bacterioferritin ,Cytochrome b Group ,Iron storage ,Ferritin ,Bacterial Proteins ,Biochemistry ,Ferritins ,biology.protein - Published
- 2014
- Full Text
- View/download PDF
22. Mass Spectrometry Approach and ELISA Reveal the Effect of Codon Optimization on N-Linked Glycosylation of HIV-1 gp120
- Author
-
Kourosh Honarmand Ebrahimi, Graham M. West, and Ricardo Flefil
- Subjects
Proteomics ,Glycosylation ,Molecular Sequence Data ,Oligosaccharides ,envelope glycoprotein ,Enzyme-Linked Immunosorbent Assay ,HIV Envelope Protein gp120 ,gp-120 ,Biochemistry ,Article ,codon optimization ,chemistry.chemical_compound ,N-linked glycosylation ,Tandem Mass Spectrometry ,Humans ,Amino Acid Sequence ,Codon ,Gene ,Glycoproteins ,Genetics ,chemistry.chemical_classification ,Binding Sites ,biology ,General Chemistry ,Envelope glycoprotein GP120 ,Stop codon ,HEK293 Cells ,chemistry ,Codon usage bias ,Mutation ,HIV-1 ,biology.protein ,Electrophoresis, Polyacrylamide Gel ,Peptides ,Glycoprotein - Abstract
The genes encoding many viral proteins such as HIV-1 envelope glycoprotein gp120 have a tendency for codons that are poorly used by the human genome. Why these codons are frequently present in the HIV genome is not known. The presence of these codons limits expression of HIV-1 gp120 for biochemical studies. The poor codons are replaced by synonymous codons that are frequently present in the highly expressed human genes to overexpress this protein. Whether this codon optimization affects functional properties of gp120 such as its N-linked glycosylation is unknown. We applied a bottom-up mass-spectrometry-based workflow for the direct measurement of deglycosylated and unglycosylated peptides with putative N-linked glycosylation sites, that is, NxS/T motifs. Using this mass-spectrometry approach in combination with ELISA, it is found that codon optimization significantly reduces the frequency with which the dolichol pyrophosphate-linked oligosaccharide is added by the catalytic subunits of oligosaccharide transferase complex to the glycosylation sites. This reduction affects binding of glycan-dependent broadly neutralizing antibodies. These data are essential for biochemical studies of gp120 and successful development of a vaccine against HIV-1. Furthermore, they demonstrate a mass-spectrometry approach for studying the site-specific N-linked glycosylation efficiency of glycoproteins.
- Published
- 2014
- Full Text
- View/download PDF
23. Phosphate accelerates displacement of Fe(III) by Fe(II) in the ferroxidase center ofPyrococcus furiosusferritin
- Author
-
Peter-Leon Hagedoorn, Wilfred R. Hagen, and Kourosh Honarmand Ebrahimi
- Subjects
Models, Molecular ,Archaeal Proteins ,Iron ,Biophysics ,Phosphate ,Models, Biological ,Biochemistry ,Phosphates ,chemistry.chemical_compound ,Structural Biology ,Enzyme Stability ,Genetics ,Site-directed mutagenesis ,Molecular Biology ,Ferritin ,Binding Sites ,Bacteria ,biology ,Ceruloplasmin ,Cell Biology ,Displacement ,biology.organism_classification ,Archaea ,Pyrococcus furiosus ,Kinetics ,Crystallography ,Amino Acid Substitution ,chemistry ,Ferritins ,Ferroxidase center ,Mutagenesis, Site-Directed ,biology.protein ,Molecular mechanism ,Oxidation-Reduction - Abstract
The iron-storage protein, ferritin, is widely found in all Domains of life. A conserved diiron center in ferritin catalyzes oxidation of Fe(II) and regulates storage of the resultant Fe(III) oxidation product. When this center is filled with Fe(III), in bacterial or archaeal ferritin the presence of phosphate accelerates the rate of Fe(II) oxidation. The molecular mechanism underlying this stimulatory effect of phosphate is unknown. Using site directed mutagenesis of the residues in the diiron center of the archaeal ferritin from Pyrococcus furiosus we show that phosphate facilitates displacement of Fe(III) by Fe(II) from this site. Therefore, the rate of Fe(II) oxidation increases only when the ferroxidase center is filled with Fe(III).
- Published
- 2012
- Full Text
- View/download PDF
24. The catalytic center of ferritin regulates iron storage via Fe(II)-Fe(III) displacement
- Author
-
Peter-Leon Hagedoorn, Wilfred R. Hagen, Kourosh Honarmand Ebrahimi, and Eckhard Bill
- Subjects
Models, Molecular ,Binding Sites ,biology ,Chemistry ,Iron ,Ceruloplasmin ,Protein core ,Cell Biology ,Ferric Compounds ,Iron storage ,Catalysis ,Ferritin ,Crystallography ,Biochemistry ,Biocatalysis ,Ferritins ,biology.protein ,Ferrous Compounds ,Binding site ,Molecular Biology - Abstract
A conserved iron-binding site, the ferroxidase center, regulates the vital iron storage role of the ubiquitous protein ferritin in iron metabolism. It is commonly thought that two Fe(II) simultaneously bind the ferroxidase center and that the oxidized Fe(III)-O(H)-Fe(III) product spontaneously enters the cavity of ferritin as a unit. In contrast, in some bacterioferritins and in archaeal ferritins a persistent di-iron prosthetic group in this center is believed to mediate catalysis of core formation. Using a combination of binding experiments and isotopically labeled (57)Fe(II), we studied two systems in comparison: the ferritin from the hyperthermophilic archaeal anaerobe Pyrococcus furiosus (PfFtn) and the eukaryotic human H ferritin (HuHF). The results do not support either of the two paradigmatic models; instead they suggest a unifying mechanism in which the Fe(III)-O-Fe(III) unit resides in the ferroxidase center until it is sequentially displaced by Fe(II).
- Published
- 2012
- Full Text
- View/download PDF
25. Spectroscopic evidence for the role of a site of the di-iron catalytic center of ferritins in tuning the kinetics of Fe(II) oxidation
- Author
-
Kourosh Honarmand Ebrahimi, Peter-Leon Hagedoorn, Eckhard Bill, and Wilfred R. Hagen
- Subjects
Models, Molecular ,0301 basic medicine ,Kinetics ,Molecular Conformation ,010402 general chemistry ,01 natural sciences ,Catalysis ,03 medical and health sciences ,Nanocages ,Iron homeostasis ,Catalytic Domain ,Humans ,Protein Interaction Domains and Motifs ,Ferrous Compounds ,Molecular Biology ,Binding Sites ,biology ,Chemistry ,Spectrum Analysis ,Ceruloplasmin ,biology.organism_classification ,0104 chemical sciences ,Pyrococcus furiosus ,Ferritin ,Crystallography ,A-site ,030104 developmental biology ,Ferritins ,biology.protein ,Oxidation-Reduction ,Protein Binding ,Biotechnology - Abstract
Ferritin is a nanocage protein made of 24 subunits. Its major role is to manage intracellular concentrations of free Fe(ii) and Fe(iii) ions, which is pivotal for iron homeostasis across all domains of life. This function of the protein is regulated by a conserved di-iron catalytic center and has been the subject of extensive studies over the past 50 years. Yet, it has not been fully understood how Fe(ii) is oxidized in the di-iron catalytic center and it is not known why eukaryotic and microbial ferritins oxidize Fe(ii) with different kinetics. In an attempt to obtain a new insight into the mechanism of Fe(ii) oxidation and understand the origin of the observed differences in the catalysis of Fe(ii) oxidation among ferritins we studied and compared the mechanism of Fe(ii) oxidation in the eukaryotic human H-type ferritin (HuHF) and the archaeal ferritin from Pyrococcus furiosus (PfFtn). The results show that the spectroscopic characteristics of the intermediate of Fe(ii) oxidation and the Fe(iii)-products are the same in these two ferritins supporting the proposal of unity in the mechanism of Fe(ii) oxidation among eukaryotic and microbial ferritins. Moreover, we observed that a site in the di-iron catalytic center controls the distribution of Fe(ii) among subunits of HuHF and PfFtn differently. This observation explains the reported differences between HuHF and PfFtn in the kinetics of Fe(ii) oxidation and the amount of O2 consumed per Fe(ii) oxidized. These results provide a fresh understanding of the mechanism of Fe(ii) oxidation by ferritins.
- Published
- 2016
26. Accurate label-free reaction kinetics determination using initial rate heat measurements
- Author
-
Peter-Leon Hagedoorn, Wilfred R. Hagen, Denise Ilse Jacobs, and Kourosh Honarmand Ebrahimi
- Subjects
Hot Temperature ,Multidisciplinary ,Chromatography ,biology ,Chemistry ,Kinetics ,Substrate (chemistry) ,Isothermal titration calorimetry ,Calorimetry ,Models, Theoretical ,Article ,Enzyme assay ,Chemical kinetics ,Reaction rate ,Biochemistry ,biology.protein ,Enzyme kinetics - Abstract
Accurate label-free methods or assays to obtain the initial reaction rates have significant importance in fundamental studies of enzymes and in application-oriented high throughput screening of enzyme activity. Here we introduce a label-free approach for obtaining initial rates of enzyme activity from heat measurements, which we name initial rate calorimetry (IrCal). This approach is based on our new finding that the data recorded by isothermal titration calorimetry for the early stages of a reaction, which have been widely ignored, are correlated to the initial rates. Application of the IrCal approach to various enzymes led to accurate enzyme kinetics parameters as compared to spectroscopic methods and enabled enzyme kinetic studies with natural substrate, e.g. proteases with protein substrates. Because heat is a label-free property of almost all reactions, the IrCal approach holds promise in fundamental studies of various enzymes and in use of calorimetry for high throughput screening of enzyme activity.
- Published
- 2015
- Full Text
- View/download PDF
27. A conserved tyrosine in ferritin is a molecular capacitor
- Author
-
Peter-Leon Hagedoorn, Kourosh Honarmand Ebrahimi, and Wilfred R. Hagen
- Subjects
Pyrococcus ,biology ,Chemistry ,Organic Chemistry ,Kinetics ,biology.organism_classification ,Biochemistry ,Catalysis ,H-Ferritin ,Ferritin ,Residue (chemistry) ,Catalytic Domain ,Ferritins ,Pyrococcus furiosus ,biology.protein ,Molecular Medicine ,Tyrosine ,Ferrous Compounds ,Binding site ,Molecular Biology ,Oxidation-Reduction - Abstract
A highly conserved tyrosine residue of unknown function is present in the vicinity of the di-iron catalytic center of the ubiquitous iron-storage protein ferritin. The di-iron center with a gateway FeII/FeIII-binding site nearby provides the vital iron-storage mechanism of the protein. It is believed that, in eukaryotic ferritin, this center catalyzes simultaneous oxidation of two FeII ions, whereas in microbial ferritin it catalyzes simultaneous oxidation of three FeII ions. To understand the role of the conserved tyrosine, we studied the intermediates and products that are formed during catalysis of FeII oxidation in the di-iron catalytic centers of the hyperthermophilic archaeal Pyrococcus furiosus ferritin and of eukaryotic human H ferritin. Based on our spectroscopic studies and modeling, we propose a merger of the models for eukaryotic and bacterial ferritin into a common mechanism of FeII oxidation in which the conserved tyrosine acts as a single-electron molecular capacitor to facilitate oxidation of FeII.
- Published
- 2013
28. Erratum: Corrigendum: The catalytic center of ferritin regulates iron storage via Fe(II)-Fe(III) displacement
- Author
-
Kourosh Honarmand Ebrahimi, Eckhard Bill, Peter-Leon Hagedoorn, and Wilfred R. Hagen
- Subjects
Ferritin ,Crystallography ,biology ,Chemistry ,Inorganic chemistry ,biology.protein ,Displacement (orthopedic surgery) ,Center (algebra and category theory) ,Cell Biology ,Molecular Biology ,Iron storage ,Catalysis - Abstract
Corrigendum: The catalytic center of ferritin regulates iron storage via Fe( II )-Fe( III ) displacement
- Published
- 2013
- Full Text
- View/download PDF
29. Inhibition and stimulation of formation of the ferroxidase center and the iron core in Pyrococcus furiosus ferritin
- Author
-
Peter-Leon Hagedoorn, Kourosh Honarmand Ebrahimi, and Wilfred R. Hagen
- Subjects
Models, Molecular ,Time Factors ,Iron ,Protein subunit ,chemistry.chemical_element ,Zinc ,Calorimetry ,Biochemistry ,Catalysis ,Cofactor ,Phosphates ,Inorganic Chemistry ,iron oxidation ,Vanadate ,ferroxidase center ,Binding site ,phosphate ,Original Paper ,Binding Sites ,biology ,ferritin ,Isothermal titration calorimetry ,biology.organism_classification ,isothermal titration calorimetry ,Pyrococcus furiosus ,Ferritin ,Kinetics ,Crystallography ,chemistry ,Ferritins ,biology.protein ,Oxidation-Reduction - Abstract
Ferritin is a ubiquitous iron-storage protein that has 24 subunits. Each subunit of ferritins that exhibit high Fe(II) oxidation rates has a diiron binding site, the so-called ferroxidase center (FC). The role of the FC appears to be essential for the iron-oxidation catalysis of ferritins. Studies of the iron oxidation by mammalian, bacterial, and archaeal ferritin have indicated different mechanisms are operative for Fe(II) oxidation, and for inhibition of the Fe(II) oxidation by Zn(II). These differences are presumably related to the variations in the amino acid residues of the FC and/or transport channels. We have used a combination of UV-vis spectroscopy, fluorescence spectroscopy, and isothermal titration calorimetry to study the inhibiting action of Zn(II) ions on the iron-oxidation process by apoferritin and by ferritin aerobically preloaded with 48 Fe(II) per 24-meric protein, and to study a possible role of phosphate in initial iron mineralization by Pyrococcus furiosus ferritin (PfFtn). Although the empty FC can accommodate two zinc ions, binding of one zinc ion to the FC suffices to essentially abolish iron-oxidation activity. Zn(II) no longer binds to the FC nor does it inhibit iron core formation once the FC is filled with two Fe(III). Phosphate and vanadate facilitate iron oxidation only after formation of a stable FC, whereupon they become an integral part of the core. These results corroborate our previous proposal that the FC in PfFtn is a stable prosthetic group, and they suggest that its formation is essential for iron-oxidation catalysis by the protein.
- Full Text
- View/download PDF
30. A novel mechanism of iron-core formation by Pyrococcus furiosus archaeoferritin, a member of an uncharacterized branch of the ferritin-like superfamily
- Author
-
Peter Verhaert, Laura van der Weel, Wilfred R. Hagen, Peter-Leon Hagedoorn, and Kourosh Honarmand Ebrahimi
- Subjects
Ferritin-like superfamily ,Iron ,Molecular Sequence Data ,Storage ,Genome ,Polymerase Chain Reaction ,Biochemistry ,Ferroxidase ,Inorganic Chemistry ,Amino Acid Sequence ,Peptide sequence ,Alternative methods ,Original Paper ,biology ,Mass spectrometry ,Chemistry ,SUPERFAMILY ,Bacterioferritin ,biology.organism_classification ,Ferritin ,Pyrococcus furiosus ,Ferritins ,biology.protein ,Function (biology) - Abstract
Storage of iron in a nontoxic and bioavailable form is essential for many forms of life. Three subfamilies of the ferritin-like superfamily, namely, ferritin, bacterioferritin, and Dps (DNA-binding proteins from starved cells), are able to store iron. Although the function of these iron-storage proteins is constitutive to many organisms to sustain life, the genome of some organisms appears not to encode any of these proteins. In an attempt to identify new iron-storage systems, we have found and characterized a new member of the ferritin-like superfamily of proteins, which unlike the multimeric storage system of ferritin, bacterioferritin, and Dps is monomeric in the absence of iron. Monomers catalyze oxidation of Fe(II) and they store the Fe(III) product as they assemble to form structures comparable to those of 24-meric ferritin. We propose that this mechanism is an alternative method of iron storage by the ferritin-like superfamily of proteins in organisms that lack the regular preassociated 24-meric/12-meric ferritins. Electronic supplementary material The online version of this article (doi:10.1007/s00775-012-0913-0) contains supplementary material, which is available to authorized users.
- Full Text
- View/download PDF
31. Catalysis of iron core formation in Pyrococcus furiosus ferritin
- Author
-
Kourosh Honarmand Ebrahimi, Peter-Leon Hagedoorn, Wilfred R. Hagen, and Jaap A. Jongejan
- Subjects
Models, Molecular ,Archaeal Proteins ,Iron ,Kinetics ,chemistry.chemical_element ,Biochemistry ,Oxygen ,Ferroxidase ,Catalysis ,Inorganic Chemistry ,Electron transfer ,Oxidizing agent ,Animals ,Humans ,Original Paper ,Ferritin ,biology ,Chemistry ,Substrate (chemistry) ,biology.organism_classification ,UV–vis spectroscopy ,Pyrococcus furiosus ,Protein Subunits ,Crystallography ,Ferritins ,biology.protein ,Cattle ,Oxidation-Reduction - Abstract
The hollow sphere-shaped 24-meric ferritin can store large amounts of iron as a ferrihydrite-like mineral core. In all subunits of homomeric ferritins and in catalytically active subunits of heteromeric ferritins a diiron binding site is found that is commonly addressed as the ferroxidase center (FC). The FC is involved in the catalytic Fe(II) oxidation by the protein; however, structural differences among different ferritins may be linked to different mechanisms of iron oxidation. Non-heme ferritins are generally believed to operate by the so-called substrate FC model in which the FC cycles by filling with Fe(II), oxidizing the iron, and donating labile Fe(III)–O–Fe(III) units to the cavity. In contrast, the heme-containing bacterial ferritin from Escherichia coli has been proposed to carry a stable FC that indirectly catalyzes Fe(II) oxidation by electron transfer from a core that oxidizes Fe(II). Here, we put forth yet another mechanism for the non-heme archaeal 24-meric ferritin from Pyrococcus furiosus in which a stable iron-containing FC acts as a catalytic center for the oxidation of Fe(II), which is subsequently transferred to a core that is not involved in Fe(II)-oxidation catalysis. The proposal is based on optical spectroscopy and steady-state kinetic measurements of iron oxidation and dioxygen consumption by apoferritin and by ferritin preloaded with different amounts of iron. Oxidation of the first 48 Fe(II) added to apoferritin is spectrally and kinetically different from subsequent iron oxidation and this is interpreted to reflect FC building followed by FC-catalyzed core formation. Electronic supplementary material The online version of this article (doi:10.1007/s00775-009-0571-z) contains supplementary material, which is available to authorized users.
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.