Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.938
Filter
1.
Sci Transl Med ; 16(767): eadk5005, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39356748

ABSTRACT

Acute kidney injury (AKI) is a frequent and challenging clinical condition associated with high morbidity and mortality and represents a common complication in critically ill patients with COVID-19. In AKI, renal tubular epithelial cells (TECs) are a primary site of damage, and recovery from AKI depends on TEC plasticity. However, the molecular mechanisms underlying adaptation and maladaptation of TECs in AKI remain largely unclear. Here, our study of an autopsy cohort of patients with COVID-19 provided evidence that injury of TECs by myoglobin, released as a consequence of rhabdomyolysis, is a major pathophysiological mechanism for AKI in severe COVID-19. Analyses of human kidney biopsies, mouse models of myoglobinuric and gentamicin-induced AKI, and mouse kidney tubuloids showed that TEC injury resulted in activation of the glucocorticoid receptor by endogenous glucocorticoids, which aggravated tubular damage. The detrimental effect of endogenous glucocorticoids on injured TECs was exacerbated by the administration of a widely clinically used synthetic glucocorticoid, dexamethasone, as indicated by experiments in mouse models of myoglobinuric- and folic acid-induced AKI, human and mouse kidney tubuloids, and human kidney slice cultures. Mechanistically, studies in mouse models of AKI, mouse tubuloids, and human kidney slice cultures demonstrated that glucocorticoid receptor signaling in injured TECs orchestrated a maladaptive transcriptional program to hinder DNA repair, amplify injury-induced DNA double-strand break formation, and dampen mTOR activity and mitochondrial bioenergetics. This study identifies glucocorticoid receptor activation as a mechanism of epithelial maladaptation, which is functionally important for AKI.


Subject(s)
Acute Kidney Injury , COVID-19 , Epithelial Cells , Glucocorticoids , Receptors, Glucocorticoid , Animals , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Humans , Glucocorticoids/adverse effects , Glucocorticoids/pharmacology , COVID-19/complications , COVID-19/metabolism , Mice , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Epithelial Cells/pathology , Receptors, Glucocorticoid/metabolism , Disease Models, Animal , Male , Kidney Tubules/pathology , Kidney Tubules/metabolism , Kidney Tubules/drug effects , Myoglobin/metabolism , Dexamethasone/pharmacology , Dexamethasone/adverse effects , Stress, Physiological/drug effects , SARS-CoV-2 , Mice, Inbred C57BL , Female
2.
Inorg Chem ; 63(40): 18531-18535, 2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39311200

ABSTRACT

In this study, we constructed a metal-binding site close to the heme cofactor in myoglobin (Mb) by covalently attaching a nonnative metal-binding ligand of bipyridine to Cys46 through the F46C mutation in the heme distal site. The X-ray structure of the designed enzyme, termed F46C-mBpy Mb, was solved in the Cu(II)-bound form, which revealed the formation of a heterodinuclear center of Cu-His-H2O-heme. Cu(II)-F46C-mBpy Mb exhibits not only nitrite reductase reactivity but also cascade reaction activity involving both hydrolysis and oxidation. Furthermore, F46C-mBpy Mb displays Mn-peroxidase activity by the oxidation of Mn2+ to Mn3+ using H2O2 as an oxidant. This study shows that the construction of a nonnative metal-binding site close to the heme cofactor is a convenient approach to creating an artificial metalloenzyme with a heterodinuclear center that confers multiple functions.


Subject(s)
Heme , Myoglobin , Myoglobin/chemistry , Myoglobin/metabolism , Heme/chemistry , Heme/metabolism , Binding Sites , Models, Molecular , Copper/chemistry , Copper/metabolism , Oxidation-Reduction , Metalloproteins/chemistry , Metalloproteins/metabolism , Crystallography, X-Ray , Manganese/chemistry , Manganese/metabolism
3.
Anal Chem ; 96(39): 15765-15772, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39291743

ABSTRACT

I. BACKGROUND: Human induced pluripotent stem cell (hiPSC) derived cardiomyocytes (CMs) have been utilized in drug toxicity evaluation, drug discovery, and treating heart failure patients, showing substantial effects. Ensuring the quality, purity, and maturation of hiPSC-CMs during large-scale production is crucial. There is a growing demand for a novel method to characterize cell molecular profiles without labels and without causing damage. II. METHODS: In this study, we employed label-free Raman microscopy to evaluate hiPSC-derived CMs. The study involved the characterization of cell molecular profiles without labels and without causing damage. The correlation between Raman spectroscopy of specific components, such as cytochrome c and myoglobin, and CM purity and maturation following hiPSC differentiation was investigated. Additionally, the validation of this correlation was performed by assessing mixtures of commercially available CMs (iCell cardiomyocytes2) and fibroblasts at various ratios as well as hiPSC-derived CMs with different efficiencies. Furthermore, CMs were matured using rapid pacing of traveling waves, and the Raman profiles of matured CMs were compared to those of immature ones. III. RESULTS: Raman spectroscopy indicated that the cytochrome c and myoglobin showed correlation with the purity and maturation of CMs following differentiation of hiPSCs. This correlation was validated through experiments involving different CM-fibroblast mixtures and hiPSC-derived CMs with varying efficiencies. Moreover, matured CMs exhibited markedly different Raman profiles compared to immature ones, indicating the potential of Raman imaging as a tool for assessing CM maturation. IV. CONCLUSIONS: We discovered that Raman spectroscopy of certain components, such as cytochrome c and myoglobin, correlates with the CM purity and maturation following hiPSC differentiation. The findings of this study highlight the potential of label-free Raman microscopy as a nondestructive, high-content, and time-efficient method for quality control of hiPSC-derived CMs. This approach could significantly contribute to ensuring the quality and maturity of hiPSC-CMs for various applications in drug discovery and regenerative medicine.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells , Myocytes, Cardiac , Spectrum Analysis, Raman , Humans , Spectrum Analysis, Raman/methods , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myoglobin/analysis , Myoglobin/metabolism , Cytochromes c/metabolism , Cytochromes c/analysis , Cells, Cultured
4.
Bioorg Med Chem ; 112: 117900, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39217687

ABSTRACT

The incorporation of non-canonical amino acids (ncAAs) into the metal coordination environments of proteins has endowed metalloproteins with enhanced properties and novel activities, particularly in hemoproteins. In this work, we disclose a scalable synthetic strategy that enables the production of myoglobin (Mb) variants with non-canonical heme ligands, i.e., HoCys and f4Tyr. The ncAA-containing Mb* variants (with H64V/V68A mutations) were obtained through two consecutive native chemical ligations and a subsequent desulfurization step, with overall isolated yield up to 28.6 % in over 10-milligram scales. After refolding and heme b cofactor reconstitution, the synthetic Mb* variants showed typical electronic absorption bands. When subjected to the catalysis of the cyclopropanation of styrene, both synthetic variants, however, were not as competent as the His-ligated Mb*. We envisioned that the synthetic method reported herein would be useful for incorporating a variety of ncAAs with diverse structures and properties into Mb for varied purposes.


Subject(s)
Heme , Myoglobin , Myoglobin/chemistry , Myoglobin/metabolism , Ligands , Heme/chemistry , Heme/metabolism , Molecular Structure , Amino Acids/chemistry , Amino Acids/metabolism
5.
J Inorg Biochem ; 260: 112688, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39111220

ABSTRACT

New-to-Nature biocatalysis has emerged as a promising tool in organic synthesis thanks to progress in protein engineering. Notably, hemeproteins have been evolved into robust catalysts for carbene and nitrene transfers and related sigmatropic rearrangements. In this work, we report the first example of a [2,3]-sigmatropic Sommelet-Hauser rearrangement initiated by a carbene transfer of the sperm whale myoglobin mutant L29S,H64V,V68F that was previously reported to catalyze the mechanistically similar [2,3]-sigmatropic Doyle-Kirmse rearrangement. This repurposed heme enzyme catalyzes the Sommelet-Hauser rearrangement between ethyl diazoacetate and benzyl thioethers bearing strong electron-withdrawing substituents with good yields and enantiomeric excess. Optimized catalytic conditions in the absence of any reductant led to an increased asymmetric induction with up to 59% enantiomeric excess. This myoglobin mutant is therefore one of the few catalysts for the asymmetric Sommelet-Hauser rearrangement. This work broadens the scope of abiological reactions catalyzed by iron-carbene transferases with a new example of asymmetric sigmatropic rearrangement.


Subject(s)
Myoglobin , Myoglobin/chemistry , Myoglobin/genetics , Myoglobin/metabolism , Methane/analogs & derivatives , Methane/chemistry , Methane/metabolism , Biocatalysis , Transferases/metabolism , Transferases/genetics , Transferases/chemistry , Animals , Sperm Whale , Protein Engineering/methods
6.
Nat Commun ; 15(1): 5714, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38977701

ABSTRACT

Genetic code expansion has emerged as a powerful tool for precisely introducing unnatural chemical structures into proteins to improve their catalytic functions. Given the high catalytic propensity of histidine in the enzyme pocket, increasing the chemical diversity of catalytic histidine could result in new characteristics of biocatalysts. Herein, we report the genetically encoded Nδ-Vinyl Histidine (δVin-H) and achieve the wild-type-like incorporation efficiency by the evolution of pyrrolysyl tRNA synthetase. As histidine usually acts as the nucleophile or the metal ligand in the catalytic center, we replace these two types of catalytic histidine to δVin-H to improve the performance of the histidine-involved catalytic center. Additionally, we further demonstrate the improvements of the hydrolysis activity of a previously reported organocatalytic esterase (the OE1.3 variant) in the acidic condition and myoglobin (Mb) catalyzed carbene transfer reactions under the aerobic condition. As histidine is one of the most frequently used residues in the enzyme catalytic center, the derivatization of the catalytic histidine by δVin-H holds a great potential to promote the performance of biocatalysts.


Subject(s)
Catalytic Domain , Histidine , Histidine/metabolism , Histidine/chemistry , Histidine/genetics , Myoglobin/genetics , Myoglobin/chemistry , Myoglobin/metabolism , Biocatalysis , Catalysis , Amino Acyl-tRNA Synthetases/genetics , Amino Acyl-tRNA Synthetases/metabolism , Amino Acyl-tRNA Synthetases/chemistry , Esterases/genetics , Esterases/metabolism , Esterases/chemistry , Hydrolysis , Escherichia coli/genetics , Escherichia coli/metabolism
7.
Sci Rep ; 14(1): 16337, 2024 07 16.
Article in English | MEDLINE | ID: mdl-39014025

ABSTRACT

It has been suggested that sodium-glucose cotransporter 2 (SGLT2) inhibitors have cardioprotective effects during myocardial ischemia/reperfusion (I/R) independent of glucose-lowering action. However, the effects of SGLT2 inhibitors on structural damage to cardiomyocytes in the ischemic region during I/R remain unknown. We applied a microdialysis technique to the heart of anesthetized rats and investigated the effects of an SGLT2 inhibitor, dapagliflozin, on myocardial interstitial myoglobin levels in the ischemic region during coronary occlusion followed by reperfusion. Dapagliflozin was administered systemically (40 µg/body iv) or locally via a dialysis probe (100 µM and 1 mM) 30 min before coronary occlusion. In the vehicle group, coronary occlusion increased the dialysate myoglobin concentration in the ischemic region. Reperfusion further increased the dialysate myoglobin concentration. Intravenous administration of dapagliflozin reduced dialysate myoglobin concentration during ischemia and at 0-15 min after reperfusion, but local administration (100 µM and 1 mM) did not. Therefore, acute systemic administration of dapagliflozin prior to ischemia has cardioprotective effects on structural damage during I/R.


Subject(s)
Benzhydryl Compounds , Glucosides , Myocardial Reperfusion Injury , Myocytes, Cardiac , Myoglobin , Animals , Benzhydryl Compounds/pharmacology , Myoglobin/metabolism , Glucosides/pharmacology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/drug therapy , Rats , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Male , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Microdialysis
8.
Vitam Horm ; 125: 31-46, 2024.
Article in English | MEDLINE | ID: mdl-38997168

ABSTRACT

Post-translational modification of proteins by Maillard reaction, known as glycation, is thought to be the root cause of different complications, particularly in diabetes mellitus and age-related disorders. Methylglyoxal (MG), a reactive α-oxoaldehyde, increases in diabetic condition and reacts with the proteins to form advanced glycation end products (AGEs) following a Maillard-like reaction. In a time-dependent reaction study of MG with the heme protein myoglobin (Mb), MG was found to induce significant structural alterations of the heme protein, such as heme loss, changes in tryptophan fluorescence, and decrease of α-helicity with increased ß-sheet content. These changes were found to occur gradually with increasing period of incubation. Incubation of Mb with MG induced the formation of several AGE adducts, including, carboxyethyllysine at Lys-16, carboxymethyllysine at Lys-87, carboxyethyllysine or pyrraline-carboxymethyllysine at Lys-133, carboxyethyllysine at Lys-42 and hydroimidazolone or argpyrimidine at Arg-31 and Arg-139. MG induced amyloid-like aggregation of Mb was detected at a longer period of incubation. MG-derived AGEs, therefore, appear to have an important role as the precursors of protein aggregation, which, in turn, may be associated with pathophysiological complications.


Subject(s)
Glycation End Products, Advanced , Myoglobin , Protein Aggregates , Pyruvaldehyde , Animals , Humans , Glycation End Products, Advanced/metabolism , Glycosylation , Maillard Reaction , Myoglobin/metabolism , Myoglobin/chemistry , Protein Processing, Post-Translational , Pyruvaldehyde/metabolism
9.
Chembiochem ; 25(17): e202400329, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-38926093

ABSTRACT

Photodynamic therapy (PDT) is a noninvasive approach to cancer treatment, wherein cell death is initiated by singlet oxygen (1O2) production via energy transfer from excited photosensitizers to ground-state O2. Effective clinical photosensitizers necessitate water solubility for in vivo administration. Hydrophobic dyes, such as phthalocyanines, cannot be used directly as photosensitizers. Herein, we synthesized a myoglobin-(human serum albumin) fusion protein reconstituted with zinc-phthalocyanine (ZnPc), termed ZnPcMb-HSA. The photophysical properties of ZnPcMb-HSA closely resemble those of ZnPc-substituted Mb. Notably, ZnPc dissociates from ZnPcMb-HSA and selectively accumulates within cancer cells, while the protein components remain extracellular. Treatment of four distinct cell lines with ZnPcMb-HSA, followed by red-light irradiation, effectively induced apoptosis. The half-maximal inhibitory concentrations (IC50) against these cancer cell lines ranged between 0.1-0.5 µM. Reconstituted Mb-HSA emerges as a promising carrier for transporting various water-insoluble porphyrinoid photosensitizer to target cancer cells in PDT applications.


Subject(s)
Indoles , Isoindoles , Myoglobin , Organometallic Compounds , Photochemotherapy , Photosensitizing Agents , Zinc Compounds , Humans , Indoles/chemistry , Indoles/pharmacology , Myoglobin/chemistry , Myoglobin/metabolism , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemical synthesis , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Organometallic Compounds/chemical synthesis , Zinc Compounds/chemistry , Cell Line, Tumor , Serum Albumin, Human/chemistry , Serum Albumin, Human/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/metabolism , Apoptosis/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Neoplasms/drug therapy , Neoplasms/pathology
10.
Toxicon ; 247: 107835, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-38942240

ABSTRACT

Serine peptidases and metallopeptidases are the primary toxins found in Bothrops snakes venoms, which act on proteins in the tissues of victims or prey, and release of peptides formed through proteolytic activity. Various studies have indicated that these peptides, released by the proteolytic activity of heterologous enzymes, generate molecules with unidentified functions, referred to as cryptids. To address this, we purified serine peptidases from Bothrops jararaca venom using molecular exclusion chromatography and then incubated them with the endogenous substrate myoglobin. As a control, we also incubated the substrate with trypsin. The resulting proteolytic fragments were analyzed, separated, and collected via HPLC. These fractions were then tested on cell cultures, the active fractions were sequenced (ALELFR and TGHPETLEK) and synthesized. After confirming their activity, the peptides underwent sequencing and synthesis for additional cell tests, including the increase of cell viability, cycle phases, proliferation, signaling, growth kinetics, angiogenesis, and migration. The results revealed that the synthesized peptides exhibited cellular repair properties, suggesting a potential role in tissue repair in the range of 0.05-5 µ M. Additionally, the effects of fragments resulting from myoglobin degradation isolated (ALELFR and TGHPETLEK) revealed a regenerative action on tissue.


Subject(s)
Bothrops , Crotalid Venoms , Myoglobin , Serine Proteases , Animals , Crotalid Venoms/chemistry , Serine Proteases/metabolism , Serine Proteases/chemistry , Myoglobin/metabolism , Peptides/pharmacology , Peptides/chemistry , Humans , Cell Survival/drug effects , Bothrops jararaca
11.
Angew Chem Int Ed Engl ; 63(36): e202403485, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-38780472

ABSTRACT

Design of metal cofactor ligands is essential for controlling the reactivity of metalloenzymes. We investigated a carbene transfer reaction catalyzed by myoglobins containing iron porphyrin cofactors with one and two trifluoromethyl groups at peripheral sites (FePorCF3 and FePor(CF3)2, respectively), native heme and iron porphycene (FePc). These four myoglobins show a wide range of Fe(II)/Fe(III) redox potentials in the protein of +147 mV, +87 mV, +42 mV and -198 mV vs. NHE, respectively. Myoglobin reconstituted with FePor(CF3)2 has a more positive potential, which enhances the reactivity of a carbene intermediate with alkenes, and demonstrates superior cyclopropanation of inert alkenes, such as aliphatic and internal alkenes. In contrast, engineered myoglobin reconstituted with FePc has a more negative redox potential, which accelerates the formation of the intermediate, but has low reactivity for inert alkenes. Mechanistic studies indicate that myoglobin with FePor(CF3)2 generates an undetectable active intermediate with a radical character. In contrast, this reaction catalyzed by myoglobin with FePc includes a detectable iron-carbene species with electrophilic character. This finding highlights the importance of redox-focused design of the iron porphyrinoid cofactor in hemoproteins to tune the reactivity of the carbene transfer reaction.


Subject(s)
Myoglobin , Oxidation-Reduction , Myoglobin/chemistry , Myoglobin/metabolism , Protein Engineering , Cyclopropanes/chemistry , Metalloporphyrins/chemistry , Methane/chemistry , Methane/analogs & derivatives
12.
Physiol Rev ; 104(4): 1611-1642, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38696337

ABSTRACT

A canonical view of the primary physiological function of myoglobin (Mb) is that it is an oxygen (O2) storage protein supporting mitochondrial oxidative phosphorylation, especially as the tissue O2 partial pressure (Po2) drops and Mb off-loads O2. Besides O2 storage/transport, recent findings support functions for Mb in lipid trafficking and sequestration, interacting with cellular glycolytic metabolites such as lactate (LAC) and pyruvate (PYR), and "ectopic" expression in some types of cancer cells and in brown adipose tissue (BAT). Data from Mb knockout (Mb-/-) mice and biochemical models suggest additional metabolic roles for Mb, especially regulation of nitric oxide (NO) pools, modulation of BAT bioenergetics, thermogenesis, and lipid storage phenotypes. From these and other findings in the literature over many decades, Mb's function is not confined to delivering O2 in support of oxidative phosphorylation but may serve as an O2 sensor that modulates intracellular Po2- and NO-responsive molecular signaling pathways. This paradigm reflects a fundamental change in how oxidative metabolism and cell regulation are viewed in Mb-expressing cells such as skeletal muscle, heart, brown adipocytes, and select cancer cells. Here, we review historic and emerging views related to the physiological roles for Mb and present working models illustrating the possible importance of interactions between Mb, gases, and small-molecule metabolites in regulation of cell signaling and bioenergetics.


Subject(s)
Energy Metabolism , Myoglobin , Oxygen , Animals , Myoglobin/metabolism , Humans , Oxygen/metabolism , Energy Metabolism/physiology , Adipose Tissue, Brown/metabolism , Oxidative Phosphorylation , Thermogenesis/physiology
13.
Int J Biol Macromol ; 271(Pt 1): 132402, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38754662

ABSTRACT

In this paper, the effects of chitosan film containing star anise essential oil nanofiltration (CFSAO) and superchilled (SC) temperature on the changes of physicochemical and microbiological indexes of rabbit meat patties within 15 days of storage were studied. The total aerobic bacteria counts, malondialdehyde content, protein carbonyl content, total sulfhydryl content, and metmyoglobin content continued to grow throughout the entire experimental period, and the maximum absorption peak at the soret region of myoglobin gradually decreased. Along with the storage time extended, the brightness and redness of rabbit meat significantly decreased (P < 0.05), while the yellowness significantly increased (P < 0.05). The results of storage experiments showed that chitosan composite films and SC temperature had good inhibition on lipid oxidation, myoglobin oxidation and degradation, sulfhydryl content reduction, and microbial growth of rabbit meat after 15 days of storage, and could slow down the change of rabbit meat color.


Subject(s)
Chitosan , Food Packaging , Food Storage , Meat , Oils, Volatile , Animals , Chitosan/chemistry , Chitosan/pharmacology , Rabbits , Food Packaging/methods , Oils, Volatile/chemistry , Oils, Volatile/pharmacology , Meat/microbiology , Food Storage/methods , Myoglobin/metabolism , Myoglobin/chemistry , Food Preservation/methods , Food Quality
14.
Int J Mol Sci ; 25(9)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38731803

ABSTRACT

This study explores the effects of normobaric hypoxia and intermittent hypoxic training (IHT) on the physiological condition of the cardiac muscle in swimmers. Hypoxia has been reported to elicit both beneficial and adverse changes in the cardiovascular system, but its impact on the myocardium during acute exercise and altitude/hypoxic training remains less understood. We aimed to determine how a single bout of intense interval exercise and a four-week period of high-intensity endurance training under normobaric hypoxia affect cardiac marker activity in swimmers. Sixteen young male swimmers were divided into two groups: one undergoing training in hypoxia and the other in normoxia. Cardiac markers, including troponin I and T (cTnI and cTnT), heart-type fatty acid-binding protein (H-FABP), creatine kinase-MB isoenzyme (CK-MB), and myoglobin (Mb), were analyzed to assess the myocardium's response. We found no significant differences in the physiological response of the cardiac muscle to intense physical exertion between hypoxia and normoxia. Four weeks of IHT did not alter the resting levels of cTnT, cTnI, and H-FABP, but it resulted in a noteworthy decrease in the resting concentration of CK-MB, suggesting enhanced cardiac muscle adaptation to exercise. In contrast, a reduction in resting Mb levels was observed in the control group training in normoxia. These findings suggest that IHT at moderate altitudes does not adversely affect cardiac muscle condition and may support cardiac muscle adaptation, affirming the safety and efficacy of IHT as a training method for athletes.


Subject(s)
Athletes , Biomarkers , Hypoxia , Humans , Male , Hypoxia/metabolism , Pilot Projects , Swimming/physiology , Young Adult , Myocardium/metabolism , Myoglobin/metabolism , Troponin I/metabolism , Fatty Acid Binding Protein 3/metabolism , Adolescent , Fatty Acid-Binding Proteins/metabolism , Physical Endurance/physiology , Creatine Kinase, MB Form/blood , Creatine Kinase, MB Form/metabolism , Adaptation, Physiological , Altitude
15.
J Inorg Biochem ; 257: 112595, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38759262

ABSTRACT

Globins, such as myoglobin (Mb) and neuroglobin (Ngb), are ideal protein scaffolds for the design of functional metalloenzymes. To date, numerous approaches have been developed for enzyme design. This review presents a summary of the progress made in the design of functional metalloenzymes based on Mb and Ngb, with a focus on the exploitation of covalent interactions, including coordination bonds and covalent modifications. These include the construction of a metal-binding site, the incorporation of a non-native metal cofactor, the formation of Cys/Tyr-heme covalent links, and the design of disulfide bonds, as well as other Cys-covalent modifications. As exemplified by recent studies from our group and others, the designed metalloenzymes have potential applications in biocatalysis and bioconversions. Furthermore, we discuss the current trends in the design of functional metalloenzymes and highlight the importance of covalent interactions in the design of functional metalloenzymes.


Subject(s)
Globins , Myoglobin , Nerve Tissue Proteins , Neuroglobin , Neuroglobin/metabolism , Neuroglobin/chemistry , Myoglobin/chemistry , Myoglobin/metabolism , Globins/chemistry , Globins/metabolism , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/chemistry , Humans , Animals , Heme/chemistry , Heme/metabolism , Binding Sites , Metalloproteins/chemistry , Metalloproteins/metabolism , Protein Engineering/methods
16.
Mol Nutr Food Res ; 68(9): e2300889, 2024 May.
Article in English | MEDLINE | ID: mdl-38676468

ABSTRACT

SCOPE: Epidemiological studies have linked excessive red and processed meat intake to gut disorders. Under laboratory conditions, high heme content is considered the primary health risk factor for red meat. However, heme in meat is present in myoglobin, which is an indigestible protein, suggesting the different functions between myoglobin and heme. This study aims to explore how dietary myoglobin and heme affect gut health and microbiota differently. METHODS AND RESULTS: Histological and biochemical assessments as well as 16S rRNA sequencing are performed. Moderate myoglobin intake (equivalent to the recommended intake of 150 g meat per day for human) has beneficial effects on the duodenal barrier. However, a too high myoglobin diet (equivalent to intake of 3000 g meat per day for human) triggers duodenum injury and alters the microbial community. The hemin diet destroys intestinal tissue and ileal microbiota more significantly. The in vitro experiments further confirm that free heme exhibits high toxicity to beneficial gut bacteria while myoglobin promotes the growth and metabolism of Limosilactobacillus reuteri. CONCLUSION: Moderate intake of myoglobin or hemin is beneficial to intestinal health and microbiota, but too high amounts lead to tissue inflammation and injury in the small intestine by reshaping ileal microbiota.


Subject(s)
Gastrointestinal Microbiome , Hemin , Inflammation , Myoglobin , Gastrointestinal Microbiome/drug effects , Animals , Myoglobin/metabolism , Hemin/pharmacology , Male , Diet/methods , Intestine, Small/drug effects , Intestine, Small/metabolism , Limosilactobacillus reuteri , Duodenum/metabolism , RNA, Ribosomal, 16S/genetics , Heme
17.
Basic Res Cardiol ; 119(3): 435-451, 2024 06.
Article in English | MEDLINE | ID: mdl-38499702

ABSTRACT

Myocardial infarction (MI) is a serious acute cardiovascular syndrome that causes myocardial injury due to blood flow obstruction to a specific myocardial area. Under ischemic-reperfusion settings, a burst of reactive oxygen species is generated, leading to redox imbalance that could be attributed to several molecules, including myoglobin. Myoglobin is dynamic and exhibits various oxidation-reduction states that have been an early subject of attention in the food industry, specifically for meat consumers. However, rarely if ever have the myoglobin optical properties been used to measure the severity of MI. In the current study, we develop a novel imaging pipeline that integrates tissue clearing, confocal and light sheet fluorescence microscopy, combined with imaging analysis, and processing tools to investigate and characterize the oxidation-reduction states of myoglobin in the ischemic area of the cleared myocardium post-MI. Using spectral imaging, we have characterized the endogenous fluorescence of the myocardium and demonstrated that it is partly composed by fluorescence of myoglobin. Under ischemia-reperfusion experimental settings, we report that the infarcted myocardium spectral signature is similar to that of oxidized myoglobin signal that peaks 3 h post-reperfusion and decreases with cardioprotection. The infarct size assessed by oxidation-reduction imaging at 3 h post-reperfusion was correlated to the one estimated with late gadolinium enhancement MRI at 24 h post-reperfusion. In conclusion, this original work suggests that the redox state of myoglobin can be used as a promising imaging biomarker for characterizing and estimating the size of the MI during early phases of reperfusion.


Subject(s)
Myocardial Infarction , Myocardial Reperfusion Injury , Myocardium , Myoglobin , Oxidation-Reduction , Animals , Disease Models, Animal , Microscopy, Confocal , Microscopy, Fluorescence , Myocardial Infarction/metabolism , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/diagnostic imaging , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Myocardium/pathology , Myoglobin/metabolism
18.
Acta Physiol (Oxf) ; 240(5): e14139, 2024 05.
Article in English | MEDLINE | ID: mdl-38509816

ABSTRACT

AIM: Endurance exercise training is known to increase mitochondrial respiration in skeletal muscle. However, the molecular mechanisms behind this are not fully understood. Myoglobin (Mb) is a member of the globin family, which is highly expressed in skeletal and cardiac muscles. We recently found that Mb localizes inside mitochondria in skeletal muscle and interacts with cytochrome c oxidase subunit IV (COXIV), a subunit of mitochondrial complex IV, which regulates respiration by augmenting complex IV activity. In the present study, we investigated the effect of endurance training on Mb-COXIV interaction within mitochondria in rat skeletal muscle. METHODS: Eight-week-old male Wistar rats were subjected to 6-week treadmill running training. Forty-eight hours after the last training session, the plantaris muscle was removed under anesthesia and used for biochemical analysis. RESULTS: The endurance training increased mitochondrial content in the skeletal muscle. It also augmented complex IV-dependent oxygen consumption and complex IV activity in isolated mitochondria from skeletal muscle. Furthermore, endurance training increased Mb expression at the whole muscle level. Importantly, mitochondrial Mb content and Mb-COXIV binding were increased by endurance training. CONCLUSION: These findings suggest that an increase in mitochondrial Mb and the concomitant enhancement of Mb interaction with COXIV may contribute to the endurance training-induced upregulation of mitochondrial respiration by augmenting complex IV activity.


Subject(s)
Electron Transport Complex IV , Muscle, Skeletal , Myoglobin , Physical Conditioning, Animal , Rats, Wistar , Animals , Male , Muscle, Skeletal/metabolism , Electron Transport Complex IV/metabolism , Rats , Physical Conditioning, Animal/physiology , Myoglobin/metabolism , Endurance Training , Mitochondria, Muscle/metabolism , Oxygen Consumption/physiology , Physical Endurance/physiology
19.
Structure ; 32(6): 838-848.e3, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38508191

ABSTRACT

Protein missense mutations and resulting protein stability changes are important causes for many human genetic diseases. However, the accurate prediction of stability changes due to mutations remains a challenging problem. To address this problem, we have developed an unbiased effective model: PMSPcnn that is based on a convolutional neural network. We have included an anti-symmetry property to build a balanced training dataset, which improves the prediction, in particular for stabilizing mutations. Persistent homology, which is an effective approach for characterizing protein structures, is used to obtain topological features. Additionally, a regression stratification cross-validation scheme has been proposed to improve the prediction for mutations with extreme ΔΔG. For three test datasets: Ssym, p53, and myoglobin, PMSPcnn achieves a better performance than currently existing predictors. PMSPcnn also outperforms currently available methods for membrane proteins. Overall, PMSPcnn is a promising method for the prediction of protein stability changes caused by single point mutations.


Subject(s)
Neural Networks, Computer , Point Mutation , Protein Stability , Humans , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism , Myoglobin/chemistry , Myoglobin/genetics , Myoglobin/metabolism , Databases, Protein , Mutation, Missense , Models, Molecular , DNA Glycosylases
20.
Chemistry ; 30(28): e202400268, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38472116

ABSTRACT

Modern approaches in metallodrug research focus on compounds that bind protein targets rather than DNA. However, the identification of protein targets and binding sites is challenging. Using intact mass spectrometry and proteomics, we investigated the binding of the antimetastatic agent RAPTA-C to the model proteins ubiquitin, cytochrome c, lysozyme, and myoglobin. Binding to cytochrome c and lysozyme was negligible. However, ubiquitin bound up to three Ru moieties, two of which were localized at Met1 and His68 as [Ru(cym)], and [Ru(cym)] or [Ru(cym)(PTA)] adducts, respectively. Myoglobin bound up to four [Ru(cym)(PTA)] moieties and five sites were identified at His24, His36, His64, His81/82 and His113. Collision-induced unfolding (CIU) studies via ion-mobility mass spectrometry allowed measuring protein folding as a function of collisional activation. CIU of protein-RAPTA-C adducts showed binding of [Ru(cym)] to Met1 caused a significant compaction of ubiquitin, likely from N-terminal S-Ru-N chelation, while binding of [Ru(cym)(PTA)] to His residues of ubiquitin or myoglobin induced a smaller effect. Interestingly, the folded state of ubiquitin formed by His functionalization was more stable than Met1 metalation. The data suggests that selective metalation of amino acids at different positions on the protein impacts the conformation and potentially the biological activity of anticancer compounds.


Subject(s)
Cytochromes c , Muramidase , Myoglobin , Protein Folding , Ubiquitin , Ubiquitin/chemistry , Ubiquitin/metabolism , Myoglobin/chemistry , Myoglobin/metabolism , Binding Sites , Cytochromes c/chemistry , Cytochromes c/metabolism , Muramidase/chemistry , Muramidase/metabolism , Protein Binding , Ruthenium/chemistry , Coordination Complexes/chemistry , Coordination Complexes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL