1
|
Aktar MS, Madhuresh NKD, Ghiladi RA, Franzen S. The role of proton-coupled electron transfer from protein to heme in dehaloperoxidase. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2025; 1873:141053. [PMID: 39424090 DOI: 10.1016/j.bbapap.2024.141053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/01/2024] [Accepted: 10/06/2024] [Indexed: 10/21/2024]
Abstract
At least two of the six methionine (Met) residues in dehaloperoxidase (DHP) are shown to act as electron donors in both autoreduction and protein-heme crosslinking. Autoreduction observed in the two isozymes, DHP-A and DHP-B, is explained by the high heme reduction potential and an endogenous source of electrons from methionine (Met) or cysteine (Cys). This study provides evidence of a connection to protein-heme crosslinking that occurs when DHP is activated by H2O2 in competition with substrate oxidation and autoreduction. The autoreduction yields of DHP-A and DHP-B are comparable and both are inversely proportional to DHP concentration. Both isoenzymes show an anti-cooperative effect on autoreduction kinetics associated with protein dimerization. Despite the presence of five tyrosine (Tyr) amino acids in DHP-A and four Tyr in DHP-B, the mass spectral evidence does not support a Tyr-heme or interprotein Tyr-Tyr crosslinking event as observed in some mammalian myoglobins. LC-MS and tandem MS/MS studies revealed three amino acids that were involved in the heme-protein crosslink, Cys73, Met63 and Met64. Cys73 facilitates dimer formation in DHP-A which also appears to slow the rate of autoreduction, but is not involved in covalent protein-heme crosslinking. Based on mutational studies, Met63 and 64 are involved in both covalent heme crosslinking and autoreduction. Proton-coupled electron transfer and crosslinking by Met to the heme may serve to regulate DHP function and protect it from uncontrolled oxidative damage.
Collapse
Affiliation(s)
- Mst Sharmin Aktar
- Department of Chemistry, North Carolina State University, Raleigh, NC 27695, United States of America
| | | | - Reza A Ghiladi
- Department of Chemistry, North Carolina State University, Raleigh, NC 27695, United States of America
| | - Stefan Franzen
- Department of Chemistry, North Carolina State University, Raleigh, NC 27695, United States of America.
| |
Collapse
|
2
|
Zhao J, Chen Y, Alford H, Franzen S. The mechanism of autoreduction in Dehaloperoxidase-A. Biochem Biophys Res Commun 2024; 745:151217. [PMID: 39729674 DOI: 10.1016/j.bbrc.2024.151217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 12/29/2024]
Abstract
Hemoglobin and myoglobin are known to undergo autoxidation, in which the oxyferrous form of the heme is oxidized to the ferric state by O2. Dehaloperoxidase-A (DHP-A), a multifunctional catalytic hemoglobin from Amphitrite ornata is an exception and is observed to undergo the reverse process, during which the ferric heme is spontaneously reduced to the oxyferrous form under aerobic conditions. The high reduction potential of DHP (+202 mV at pH 7.0) partially explains this unusual behavior, but the endogenous source of reducing equivalents has remained obscure. Cysteine, methionine, tyrosine, and tryptophan are the principal endogenous reducing agents in proteins that may explain the observed autoreduction in DHP-A. In fact, DHP-A has six methionines, which may be of particular importance for the observed autoreduction. To investigate the role of the sulfur-containing residues, we created seven mutants (C73S, C73 S/M49C, S78C, M63L, M64L, M63 L/M64L, and H55V) by site-directed mutagenesis and conducted a series of CO-driven autoreduction kinetic measurements. Mutational analysis suggests a role for the pair of methionines M63 and M64 increaing the autoreduction rate. Adding surface cysteines has little effect, but the C73S mutation that eliminates the only native surface cysteine accelerates the autoreduction process. The kinetics had a sigmoidal form which was found to be a result of anti-cooperative behavior. This observation suggests that DHP-A's monomer-dimer equilibrium in solution may play a role in regulating the autoreduction process.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Chemistry, North Carolina State University, Raleigh, NC, 27695, USA
| | - Yinglu Chen
- Department of Chemistry, North Carolina State University, Raleigh, NC, 27695, USA
| | - Hunter Alford
- Department of Chemistry, North Carolina State University, Raleigh, NC, 27695, USA
| | - Stefan Franzen
- Department of Chemistry, North Carolina State University, Raleigh, NC, 27695, USA.
| |
Collapse
|
3
|
Feng Y, Kong L, Zheng R, Wu X, Zhou J, Xu X, Liu S. Adjusted bacterial cooperation in anammox community to adapt to high ammonium in wastewater treatment plant. WATER RESEARCH X 2024; 25:100258. [PMID: 39381622 PMCID: PMC11460484 DOI: 10.1016/j.wroa.2024.100258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/21/2024] [Accepted: 09/15/2024] [Indexed: 10/10/2024]
Abstract
Bacterial cooperation is very important for anammox bacteria which perform low-carbon and energy-efficient nitrogen removal, yet its variation to adapt to high NH4 +-N concentration in actual wastewater treatment plants (WWTPs) remains unclear. Here, we found wide and varied cross-feedings of anammox bacteria and symbiotic bacteria in the two series connected full-scale reactors with different NH4 +-N concentrations (297.95 ± 54.84 and 76.03 ± 34.01 mg/L) treating sludge digester liquor. The uptake of vitamin B6 as highly effective antioxidants secreted by the symbiotic bacteria was beneficial for anammox bacteria to resist the high NH4 +-N concentration and varied dissolved oxygen (DO). When NH4 +-N concentration in influent (1785.46 ± 228.5 mg/L) increased, anammox bacteria tended to reduce the amino acids supply to symbiotic bacteria to save metabolic costs. A total of 26.1% bacterial generalists switched to specialists to increase the stability and functional heterogeneity of the microbial community at high NH4 +-N conditions. V/A-type ATPase for anammox bacteria to adapt to the change of NH4 +-N was highly important to strive against cellular alkalization caused by free ammonia. This study expands the understanding of the adjusted bacterial cooperation within anammox consortia at high NH4 +-N conditions, providing new insights into bacterial adaptation to adverse environments from a sociomicrobiology perspective.
Collapse
Affiliation(s)
- Yiming Feng
- College of Environmental Sciences and Engineering, Peking University, Beijing, 100871, China
- Key Laboratory of Water and Sediment Sciences, Ministry of Education of China, Beijing, 100871, China
| | - Lingrui Kong
- College of Environmental Sciences and Engineering, Peking University, Beijing, 100871, China
- Key Laboratory of Water and Sediment Sciences, Ministry of Education of China, Beijing, 100871, China
| | - Ru Zheng
- College of Environmental Sciences and Engineering, Peking University, Beijing, 100871, China
- Key Laboratory of Water and Sediment Sciences, Ministry of Education of China, Beijing, 100871, China
| | - Xiaogang Wu
- College of Environmental Sciences and Engineering, Peking University, Beijing, 100871, China
- Key Laboratory of Water and Sediment Sciences, Ministry of Education of China, Beijing, 100871, China
| | - Jianhang Zhou
- College of Environmental Sciences and Engineering, Peking University, Beijing, 100871, China
- Key Laboratory of Water and Sediment Sciences, Ministry of Education of China, Beijing, 100871, China
| | - Xiaochen Xu
- Key Laboratory of Industrial Ecology and Environmental Engineering (MOE), School of Environment Sciences and Technology, Dalian University of Technology, Linggong Road 2, Dalian, 116024, China
| | - Sitong Liu
- College of Environmental Sciences and Engineering, Peking University, Beijing, 100871, China
- Key Laboratory of Water and Sediment Sciences, Ministry of Education of China, Beijing, 100871, China
| |
Collapse
|
4
|
Tan R, Hoare M, Bellomio P, Broas S, Camacho K, Swovick K, Welle KA, Hryhorenko JR, Ghaemmaghami S. Formylation facilitates the reduction of oxidized initiator methionines. Proc Natl Acad Sci U S A 2024; 121:e2403880121. [PMID: 39499632 PMCID: PMC11572973 DOI: 10.1073/pnas.2403880121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 10/02/2024] [Indexed: 11/07/2024] Open
Abstract
Within a cell, protein-bound methionines can be chemically or enzymatically oxidized, and subsequently reduced by methionine sulfoxide reductases (Msrs). Methionine oxidation can result in structural damage or be the basis of functional regulation of enzymes. In addition to participating in redox reactions, methionines play an important role as the initiator residue of translated proteins where they are commonly modified at their α-amine group by formylation or acetylation. Here, we investigated how formylation and acetylation of initiator methionines impact their propensity for oxidation and reduction. We show that in vitro, N-terminal methionine residues are particularly prone to chemical oxidation and that their modification by formylation or acetylation greatly enhances their subsequent enzymatic reduction by MsrA and MsrB. Concordantly, in vivo ablation of methionyl-tRNA formyltransferase (MTF) in Escherichia coli increases the prevalence of oxidized methionines within synthesized proteins. We show that oxidation of formylated initiator methionines is detrimental in part because it obstructs their ensuing deformylation by peptide deformylase (PDF) and hydrolysis by methionyl aminopeptidase (MAP). Thus, by facilitating their reduction, formylation mitigates the misprocessing of oxidized initiator methionines.
Collapse
Affiliation(s)
- Ruiyue Tan
- Department of Biology, University of Rochester, Rochester, NY14627
| | - Margaret Hoare
- Department of Biology, University of Rochester, Rochester, NY14627
| | - Philip Bellomio
- Department of Biology, University of Rochester, Rochester, NY14627
| | - Sarah Broas
- Department of Biology, University of Rochester, Rochester, NY14627
| | | | - Kyle Swovick
- Mass Spectrometry Resource Laboratory, University of Rochester Medical Center, Rochester, NY14627
| | - Kevin A. Welle
- Mass Spectrometry Resource Laboratory, University of Rochester Medical Center, Rochester, NY14627
| | - Jennifer R. Hryhorenko
- Mass Spectrometry Resource Laboratory, University of Rochester Medical Center, Rochester, NY14627
| | - Sina Ghaemmaghami
- Department of Biology, University of Rochester, Rochester, NY14627
- Mass Spectrometry Resource Laboratory, University of Rochester Medical Center, Rochester, NY14627
| |
Collapse
|
5
|
Interlandi G. Exploring ligands that target von Willebrand factor selectively under oxidizing conditions through docking and molecular dynamics simulations. Proteins 2024; 92:1261-1275. [PMID: 38829206 PMCID: PMC11471382 DOI: 10.1002/prot.26706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/25/2024] [Accepted: 05/13/2024] [Indexed: 06/05/2024]
Abstract
The blood protein von Willebrand factor (VWF) is a large multimeric protein that, when activated, binds to blood platelets, tethering them to the site of vascular injury and initiating blood coagulation. This process is critical for the normal hemostatic response, but especially under inflammatory conditions, it is thought to be a major player in pathological thrombus formation. For this reason, VWF has been the target for the development of anti-thrombotic therapeutics. However, it is challenging to prevent pathological thrombus formation while still allowing normal physiological blood coagulation, as currently available anti-thrombotic therapeutics are known to cause unwanted bleeding, in particular intracranial hemorrhage. This work explores the possibility of inhibiting VWF selectively under the inflammatory conditions present during pathological thrombus formation. In particular, the A2 domain of VWF is known to inhibit the neighboring A1 domain from binding to the platelet surface receptor GpIbα, and this auto-inhibitory mechanism has been shown to be removed by oxidizing agents released during inflammation. Hence, finding drug molecules that bind at the interface between A1 and A2 only under oxidizing conditions could restore such an auto-inhibitory mechanism. Here, by using a combination of computational docking, molecular dynamics simulations, and free energy perturbation calculations, a ligand from the ZINC15 database was identified that binds at the A1A2 interface, with the interaction being stronger under oxidizing conditions. The results provide a framework for the discovery of drug molecules that bind to a protein selectively in the presence of inflammatory conditions.
Collapse
Affiliation(s)
- Gianluca Interlandi
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| |
Collapse
|
6
|
Lee SH, Cho S, Lee JY, Kim JY, Kim S, Jeong M, Hong JY, Kim GY, Lee SW, Kim E, Kim J, Kim JW, Hwa J, Kim WH. Methionine sulfoxide reductase B2 protects against cardiac complications in diabetes mellitus. Diabetol Metab Syndr 2024; 16:149. [PMID: 38970135 PMCID: PMC11225187 DOI: 10.1186/s13098-024-01390-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/25/2024] [Indexed: 07/07/2024] Open
Abstract
Diabetes mellitus (DM) is a progressive, chronic metabolic disorder characterized by high oxidative stress, which can lead to cardiac damage. Methionine sulfoxylation (MetO) of proteins by excessive reactive oxygen species (ROS) can impair the basic functionality of essential cellular proteins, contributing to heart failure. Methionine sulfoxide reductase B2 (MsrB2) can reverse oxidation induced MetO in mitochondrial proteins, so we investigated its role in diabetic cardiomyopathy. We observed that DM-induced heart damage in diabetic mice model is characterized by increased ROS, increased protein MetO with mitochondria structural pathology, and cardiac fibrosis. In addition, MsrB2 was significantly increased in mouse DM cardiomyocytes, supporting the induction of a protective process. Further, MsrB2 directly induces Parkin and LC3 activation (mitophagy markers) in cardiomyocytes. In MsrB2, knockout mice displayed abnormal electrophysiological function, as determined by ECG analysis. Histological analysis confirmed increased cardiac fibrosis and disrupted cardiac tissue in MsrB2 knockout DM mice. We then corroborated our findings in human DM heart samples. Our study demonstrates that increased MsrB2 expression in the heart protects against diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Seung Hee Lee
- Division of Cardiovascular Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Republic of Korea.
- Division of Endocrine and Kidney Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Republic of Korea.
| | - Suyeon Cho
- Division of Cardiovascular Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Jong Youl Lee
- Division of Cardiovascular Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Ji Yeon Kim
- Division of Cardiovascular Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Suji Kim
- Division of Cardiovascular Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Myoungho Jeong
- Division of Cardiovascular Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Jung Yeon Hong
- Division of Cardiovascular Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Geun-Young Kim
- Division of Cardiovascular Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Seung Woo Lee
- Division of Cardiovascular Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Eunmi Kim
- Division of Cardiovascular Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Jihwa Kim
- Division of Cardiovascular Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Jee Woong Kim
- Division of Research Support, Department of Research Planning and Coordination, Korea National Institute of Health, Cheongju, Republic of Korea
| | - John Hwa
- Yale Cardiovascular Research Center, New Haven, USA.
| | - Won-Ho Kim
- Division of Cardiovascular Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Republic of Korea.
| |
Collapse
|
7
|
Yurina LV, Vasilyeva AD, Gavrilina ES, Ivanov VS, Obydennyi SI, Chabin IA, Indeykina MI, Kononikhin AS, Nikolaev EN, Rosenfeld MA. A role of methionines in the functioning of oxidatively modified fibrinogen. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2024; 1872:141013. [PMID: 38582358 DOI: 10.1016/j.bbapap.2024.141013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/05/2024] [Accepted: 04/02/2024] [Indexed: 04/08/2024]
Abstract
Posttranslational modifications in fibrinogen resulting from induced oxidation or oxidative stress in the organism can have deleterious influence on optimal functioning of fibrinogen, causing a disturbance in assembly and properties of fibrin. The protective mechanism supporting the ability of fibrinogen to function in ROS-generating environment remains completely unexplored. The effects of very low and moderately low HOCl/-OCl concentrations on fibrinogen oxidative modifications, the fibrin network structure as well as the kinetics of both fibrinogen-to-fibrin conversion and fibrin hydrolysis have been explored in the current study. As opposed to 25 Μm, HOCl/-OCl, 10 μM HOCl/-OCl did not affect the functional activity of fibrinogen. It is shown for the first time that a number of Met residues, AαMet476, AαMet517, AαMet584, BβMet367, γMet264, and γMet94, identified in 10 μM HOCl/-OCl fibrinogen by the HPLC-MS/MS method, operate as ROS scavengers, performing an important antioxidant function. In turn, this indicates that the fibrinogen structure is adapted to the detrimental action of ROS. The results obtained in our study provide evidence for a protective mechanism responsible for maintaining the structure and functioning of fibrinogen molecules in the bloodstream under conditions of mild and moderate oxidative stress.
Collapse
Affiliation(s)
- L V Yurina
- N.M. Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Russia.
| | - A D Vasilyeva
- N.M. Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Russia
| | - E S Gavrilina
- N.M. Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Russia
| | - V S Ivanov
- N.M. Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Russia
| | - S I Obydennyi
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology of Ministry of Healthcare of the Russian Federation, Russia; Centre for Theoretical Problems of Physicochemical Pharmacology, Russia
| | - I A Chabin
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology of Ministry of Healthcare of the Russian Federation, Russia; Sechenov First Moscow State Medical University (Sechenov University), Russia
| | - M I Indeykina
- N.M. Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Russia; Center for Molecular and Cellular Biology, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - A S Kononikhin
- Center for Molecular and Cellular Biology, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - E N Nikolaev
- Center for Molecular and Cellular Biology, Skolkovo Institute of Science and Technology, Moscow, Russia.
| | - M A Rosenfeld
- N.M. Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Russia
| |
Collapse
|
8
|
Wu Y, Ma Y, Zhong W, Shen H, Ye J, Du S, Li P. Alleviation of endothelial dysfunction of Pheretima guillemi (Michaelsen)-derived protein DPf3 in ponatinib-induced thrombotic zebrafish and mechanisms explored through ox-LDL-induced HUVECs and TMT-based proteomics. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117669. [PMID: 38159828 DOI: 10.1016/j.jep.2023.117669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/20/2023] [Accepted: 12/25/2023] [Indexed: 01/03/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Thrombus generation is one of the leading causes of death in human, and vascular endothelial dysfunction is a major contributor to thrombosis. Pheretima guillemi (Michaelsen), a traditional medicinal animal known as "Dilong", has been utilized to cure thrombotic disorders for many years. DPf3, a group of functional proteins extracted from P. guillemi, has been characterized and identified to possess antithrombotic bioactivity via in vitro and ex vivo experiments. AIM OF THE STUDY This study is aimed to investigate the vascular-protection activity and related mechanism of antithrombotic protein DPf3 purified from Pheretima guillelmi systematically. MATERIALS AND METHODS The antithrombotic activity and vascular endothelium protection effect of DPf3 was explored in vivo using ponatinib-induced vascular endothelial injury zebrafish thrombus model. Then, (hi) ox-LDL-induced HUVECs was applied to investigate the protection mechanism of DPf3 against the injury of vascular endothelium. In addition, TMT-based proteomics analysis was used to study the biomarkers, biological processes and signal pathways involved in the antithrombotic and vascular protective effects of DPf3 holistically. RESULTS DPf3 exerted robust in vivo antithrombosis and vascular endothelial protection ability. DPf3 was identified to prevent HUVECs from damage by reducing ROS production, and to reduce monocyte adhesion by decreasing the protein content of adhesion factor VCAM 1. DPf3 was also observed to weaken the migration ability of injured cells and inhibit abnormal angiogenesis. The mechanism of DPf3's antithrombotic and vascular protective activity was mainly related to the regulation of lipid metabolism, energy metabolism, complement and coagulation system, ECM receptor interaction, MAPK signal pathway, etc. CONCLUSIONS: This study demonstrates that DPf3 has strong antithrombotic and endothelial protective effects. The endothelial protective ability and related mechanisms of DPf3 provide a scientific reference for the traditional use of earthworms in the treatment of thrombosis.
Collapse
Affiliation(s)
- Yali Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China; Henan Province Engineering Laboratory for Clinical Evaluation Technology of Chinese Medicine, Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450000, China.
| | - Yunnan Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Wanling Zhong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Huijuan Shen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Jinhong Ye
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Shouying Du
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Pengyue Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
9
|
Interlandi G. Exploring ligands that target von Willebrand factor selectively under oxidizing conditions through docking and molecular dynamics simulations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.22.586354. [PMID: 38585752 PMCID: PMC10996496 DOI: 10.1101/2024.03.22.586354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
The blood protein von Willebrand factor (VWF) is a large multimeric protein that, when activated, binds to blood platelets tethering them to the site of vascular injury initiating blood coagulation. This process is critical for the normal haemostatic response, but especially under inflammatory conditions it is thought to be a major player in pathological thrombus formation. For this reason, VWF has been the target for the development of anti-thrombotic therapeutics. However, it is challenging to prevent pathological thrombus formation while still allowing normal physiological blood coagulation as currently available anti-thrombotic therapeutics are known to cause unwanted bleeding in particular intracranial haemorrhage. This work explores the possibility of inhibiting VWF selectively under the inflammatory conditions present during pathological thrombus formation. In particular, the A2 domain of VWF is known to inhibit the neighboring A1 domain from binding to the platelet surface receptor GpIbα and this auto-inhibitory mechanism has been shown to be removed by oxidizing agents released during inflammation. Hence, finding drug molecules that bind at the interface between A1 and A2 only under oxidizing conditions could restore such auto-inhibitory mechanism. Here, by using a combination of computational docking, molecular dynamics simulations and free energy perturbation calculations, a ligand from the ZINC15 database was identified that binds at the A1A2 interface with the interaction being stronger under oxidizing conditions. The results provide a framework for the discovery of drug molecules that bind to a protein selectively in inflammatory conditions.
Collapse
|
10
|
Wang AC, Qi XM, Li QF, Feng YJ, Zhang YL, Wei HZ, Li JS, Qiao YB, Li QS. Methionine redox regulation of actin-interacting proteins primarily governs antioxidative signaling and response to the salvianolic acid B treatment in EA.hy926 cells. Toxicol Appl Pharmacol 2024; 483:116835. [PMID: 38272317 DOI: 10.1016/j.taap.2024.116835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 01/27/2024]
Abstract
Actin-interacting proteins are important molecules for filament assembly and cytoskeletal signaling within vascular endothelium. Disruption in their interactions causes endothelial pathogenesis through redox imbalance. Actin filament redox regulation remains largely unexplored, in the context of pharmacological treatment. This work focused on the peptidyl methionine (M) redox regulation of actin-interacting proteins, aiming at elucidating its role on governing antioxidative signaling and response. Endothelial EA.hy926 cells were subjected to treatment with salvianolic acid B (Sal B) and tert-butyl-hydroperoxide (tBHP) stimulation. Mass spectrometry was employed to characterize redox status of proteins, including actin, myosin-9, kelch-like erythroid-derived cap-n-collar homology-associated protein 1 (Keap1), plastin-3, prelamin-A/C and vimentin. The protein redox landscape revealed distinct stoichiometric ratios or reaction site transitions mediated by M sulfoxide reductase and reactive oxygen species. In comparison with effects of tBHP stimulation, Sal B treatment prevented oxidation at actin M325, myosin-9 M1489/1565, Keap1 M120, plastin-3 M592, prelamin-A/C M187/371/540 and vimentin M344. For Keap1, reaction site was transitioned within its scaffolding region to the actin ring. These protein M oxidation regulations contributed to the Sal B cytoprotective effects on actin filament. Additionally, regarding the Keap1 homo-dimerization region, Sal B preventive roles against M120 oxidation acted as a primary signal driver to activate nuclear factor erythroid 2-related factor 2 (Nrf2). Transcriptional splicing of non-POU domain-containing octamer-binding protein was validated during the Sal B-mediated overexpression of NAD(P)H dehydrogenase [quinone] 1. This molecular redox regulation of actin-interacting proteins provided valuable insights into the phenolic structures of Sal B analogs, showing potential antioxidative effects on vascular endothelium.
Collapse
Affiliation(s)
- Ai-Cheng Wang
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China
| | - Xiao-Ming Qi
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China.
| | - Qing-Fang Li
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China
| | - Yi-Jia Feng
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China
| | - Yuan-Lin Zhang
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China.
| | - Hui-Zhi Wei
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China; School of Public Health Science & Pharmaceutical Science, Shanxi Medical University, Taiyuan 030001, China.
| | - Jin-Shan Li
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China
| | - Yuan-Biao Qiao
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China.
| | - Qing-Shan Li
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China; School of Public Health Science & Pharmaceutical Science, Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
11
|
Cui FP, Miao Y, Liu AX, Deng YL, Liu C, Zhang M, Zeng JY, Li YF, Liu HY, Liu CJ, Zeng Q. Associations of exposure to disinfection by-products with blood coagulation parameters among women: Results from the Tongji reproductive and environmental (TREE) study. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 269:115741. [PMID: 38029584 DOI: 10.1016/j.ecoenv.2023.115741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/15/2023] [Accepted: 11/23/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND Experimental studies have shown that disinfection byproducts (DBPs) induce coagulotoxicity, but human evidence is scarce. OBJECTIVE This study aimed to explore the relationships of DBP exposures with blood coagulation parameters. METHODS Among 858 women from the Tongji Reproductive and Environmental (TREE) study, urinary dichloroacetic acid (DCAA) and trichloroacetic acid (TCAA) were detected as internal biomarkers of DBP exposures. We measured activated partial thromboplastin time (APTT), fibrinogen (Fbg), international normalized ratio (INR), prothrombin time (PT), and thrombin time (TT) as blood coagulation parameters. Multivariable linear regression models were utilized to estimate the relationships between urinary DCAA and TCAA and blood coagulation parameters. The effect modifications by demographic and lifestyle characteristics were further explored. RESULTS Elevated tertiles of urinary DCAA concentrations were associated with increased PT and INR (11.29%, 95% CI: 1.66%, 20.92% and 0.99%, 95% CI: 0.08%, 1.90% for the third vs. first tertile, respectively; both P for trends < 0.05). Stratification analysis showed that the positive associations were only observed among younger (< 30 years), leaner (body mass index < 24.0 kg/m2), and non-passive smoking women. Moreover, elevated tertiles of urinary TCAA concentrations in positive associations with PT and INR were observed among younger women (17.89%, 95% CI: 2.50%, 33.29% and 1.82%, 95% CI: 0.34%, 3.30% for the third vs. first tertile, respectively; both P for trends < 0.05) but not among older women (both P for interactions < 0.05). CONCLUSION Higher levels of urinary DCAA and TCAA are associated with prolonged clotting time among women.
Collapse
Affiliation(s)
- Fei-Peng Cui
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yu Miao
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - A-Xue Liu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yan-Ling Deng
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Chong Liu
- Department of Environmental Health, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Zhang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Jia-Yue Zeng
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yu-Feng Li
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Hai-Yi Liu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Chang-Jiang Liu
- NHC Key Laboratory of Birth Defects and Reproductive Health, Chongqing Population and Family Planning Science and Technology Research Institute, Chongqing, PR China.
| | - Qiang Zeng
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China.
| |
Collapse
|
12
|
Li J, He Q, Liu C, Zeng C, Tao C, Zhai Y, Liu W, Zhang Q, Wang R, Zhang Y, Ge P, Zhang D, Zhao J. Integrated analysis of the association between methionine cycle and risk of moyamoya disease. CNS Neurosci Ther 2023; 29:3212-3227. [PMID: 37183324 PMCID: PMC10580345 DOI: 10.1111/cns.14254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/10/2023] [Accepted: 04/24/2023] [Indexed: 05/16/2023] Open
Abstract
OBJECTIVE The role of methionine (Met) cycle in the pathogenesis and progression of cardiovascular and cerebrovascular diseases has been established, but its association with moyamoya disease (MMD) has rarely been studied. This study aimed to analyze the levels of Met cycle-related metabolites and constructed a risk model to explore its association with the risk of MMD. METHODS In this prospective study, a total of 302 adult MMD patients and 88 age-matched healthy individuals were consecutively recruited. The serum levels of Met cycle-related metabolites were quantified by liquid chromatography-mass spectrometry (LC-MS). Participants were randomly divided into training set and testing set at a ratio of 1:1. The training set was used to construct the risk score model by LASSO regression. The association between Met cycle-related risk score and the risk of MMD was analyzed using logistic regression and assessed by ROC curves. The testing set was used for validation. RESULTS The levels of methionine sulfoxide and homocysteine were significantly increased, while the levels of betaine and choline were significantly decreased in MMD and its subtypes compared to healthy controls (p < 0.05 for all). The training set was used to construct the risk model and the risk score of each participant has been calculated. After adjusting for potential confounders, the risk score was independently associated with the risk of MMD and its subtypes (p < 0.05 for all). We then divided the participants into low-risk and high-risk groups, the high-risk score was significantly associated with the risk of MMD and its subtypes (p < 0.05 for all). The risk scores were further assessed as tertiles, the highest tertile was significantly associated with a higher risk of MMD and its subtypes compared to the lowest (p < 0.05 for all). The results were validated in the testing set. CONCLUSION This study has constructed and validated a risk model based on Met cycle-related metabolites, which was independently associated with the risk of MMD and its subtypes. The findings provided a new perspective on the risk evaluation and prevention of MMD.
Collapse
Affiliation(s)
- Junsheng Li
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Qiheng He
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Chenglong Liu
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Chaofan Zeng
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Chuming Tao
- Department of NeurosurgeryThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yuanren Zhai
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Wei Liu
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Qian Zhang
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Rong Wang
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Yan Zhang
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Peicong Ge
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Dong Zhang
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Jizong Zhao
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| |
Collapse
|
13
|
Yang M, Smith BC. Cysteine and methionine oxidation in thrombotic disorders. Curr Opin Chem Biol 2023; 76:102350. [PMID: 37331217 PMCID: PMC10527720 DOI: 10.1016/j.cbpa.2023.102350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 06/20/2023]
Abstract
Thrombosis is the leading cause of death in many diseased conditions. Oxidative stress is characteristic of these conditions. Yet, the mechanisms through which oxidants become prothrombotic are unclear. Recent evidence suggests protein cysteine and methionine oxidation as prothrombotic regulators. These oxidative post-translational modifications occur on proteins that participate in the thrombotic process, including Src family kinases, protein disulfide isomerase, β2 glycoprotein I, von Willebrand factor, and fibrinogen. New chemical tools to identify oxidized cysteine and methionine proteins in thrombosis and hemostasis, including carbon nucleophiles for cysteine sulfenylation and oxaziridines for methionine, are critical to understanding why clots occur during oxidative stress. These mechanisms will identify alternative or novel therapeutic approaches to treat thrombotic disorders in diseased conditions.
Collapse
Affiliation(s)
- Moua Yang
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02115, USA.
| | - Brian C Smith
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
14
|
Rosenfeld MA, Yurina LV, Vasilyeva AD. Antioxidant role of methionine-containing intra- and extracellular proteins. Biophys Rev 2023; 15:367-383. [PMID: 37396452 PMCID: PMC10310685 DOI: 10.1007/s12551-023-01056-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/24/2023] [Indexed: 07/04/2023] Open
Abstract
Significant evidence suggests that reversible oxidation of methionine residues provides a mechanism capable of scavenging reactive species, thus creating a cycle with catalytic efficiency to counteract or mitigate deleterious effects of ROS on other functionally important amino acid residues. Because of the absence of MSRs in the blood plasma, oxidation of methionines in extracellular proteins is effectively irreversible and, therefore, the ability of methionines to serve as interceptors of oxidant molecules without impairment of the structure and function of plasma proteins is still debatable. This review presents data on the oxidative modification of both intracellular and extracellular proteins that differ drastically in their spatial structures and functions indicating that the proteins contain antioxidant methionines/the oxidation of which does not affect (or has a minor effect) on their functional properties. The functional consequences of methionine oxidation in proteins have been mainly identified from studies in vitro and, to a very limited extent, in vivo. Hence, much of the functioning of plasma proteins constantly subjected to oxidative stress remains unclear and requires further research to understand the evolutionary role of methionine oxidation in proteins for the maintenance of homeostasis and risk factors affecting the development of ROS-related pathologies. Data presented in this review contribute to increased evidence of antioxidant role of surface-exposed methionines and can be useful for understanding a possible mechanism that supports or impairs structure-function relationships of proteins subjected to oxidative stress.
Collapse
Affiliation(s)
- Mark A. Rosenfeld
- N. M. Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119334 Russia
| | - Lyubov V. Yurina
- N. M. Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119334 Russia
| | - Alexandra D. Vasilyeva
- N. M. Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119334 Russia
| |
Collapse
|
15
|
Babu S, Claville MO, Fronczek FR, Uppu RM. l-Me-thion-yl-l-tyrosine monohydrate. IUCRDATA 2023; 8:x230551. [PMID: 37936870 PMCID: PMC10626622 DOI: 10.1107/s2414314623005515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 06/22/2023] [Indexed: 11/09/2023] Open
Abstract
The study of the oxidation of various proteins necessitates scrutiny of the amino acid sequence. Since me-thio-nine (Met) and tyrosine (Tyr) are easily oxidized, peptides that contain these amino acids are frequently studied using a variety of oxidation methods, including, but not limited to, pulse radiolysis, electrochemical oxidation, and laser flash photolysis. To date, the oxidation of the Met-Tyr dipeptide is not fully understood. Several investigators have proposed a mechanism of intra-molecular electron transfer between the sulfide radical of Met and the Tyr residue. Our elucidation of the structure and absolute configuration of l-Met-l-Tyr monohydrate, C14H20N2O4S·H2O (systematic name: (2S)-2-{[(2S)-2-amino-4-methyl-sulfanyl-butano-yl]amino}-3-(4-hy-droxy-phen-yl)propanoic acid monohydrate) is presented herein and provides information about the zwitterionic nature of the dipeptide. We suspect that the zwitterionic state of the dipeptide and its inter-action within the solvent medium may play a major role in the oxidation of the dipeptide. In the crystal, all the potential donor atoms inter-act via strong N-H⋯O, C-H⋯O, O-H⋯S, and O-H⋯O hydrogen bonds.
Collapse
Affiliation(s)
- Sainath Babu
- Department of Biological Science, Hampton University, Hampton, VA 23668, USA
| | | | - Frank R. Fronczek
- Department of Chemistry, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Rao M. Uppu
- Department of Environmental Toxicology, Southern University and A&M College, Baton Rouge, LA 70813, USA
| |
Collapse
|
16
|
Lu Z, Feng L, Jiang W, Wu P, Liu Y, Jiang J, Kuang S, Tang L, Li S, Zhong C, Zhou X. Mannan oligosaccharides alleviate oxidative injury in the head kidney and spleen in grass carp (Ctenopharyngodon idella) via the Nrf2 signaling pathway after Aeromonas hydrophila infection. J Anim Sci Biotechnol 2023; 14:58. [PMID: 37060042 PMCID: PMC10105433 DOI: 10.1186/s40104-023-00844-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 01/31/2023] [Indexed: 04/16/2023] Open
Abstract
BACKGROUND Mannan oligosaccharides (MOS) are recommended as aquaculture additives owing to their excellent antioxidant properties. In the present study, we examined the effects of dietary MOS on the head kidney and spleen of grass carp (Ctenopharyngodon idella) with Aeromonas hydrophila infection. METHODS A total of 540 grass carp were used for the study. They were administered six gradient dosages of the MOS diet (0, 200, 400, 600, 800, and 1,000 mg/kg) for 60 d. Subsequently, we performed a 14-day Aeromonas hydrophila challenge experiment. The antioxidant capacity of the head kidney and spleen were examined using spectrophotometry, DNA fragmentation, qRT-PCR, and Western blotting. RESULTS After infection with Aeromonas hydrophila, 400-600 mg/kg MOS supplementation decreased the levels of reactive oxygen species, protein carbonyl, and malonaldehyde and increased the levels of anti-superoxide anion, anti-hydroxyl radical, and glutathione in the head kidney and spleen of grass carp. The activities of copper-zinc superoxide dismutase, manganese superoxide dismutase, catalase, glutathione S-transferase, glutathione reductase, and glutathione peroxidase were also enhanced by supplementation with 400-600 mg/kg MOS. Furthermore, the expression of most antioxidant enzymes and their corresponding genes increased significantly with supplementation of 200-800 mg/kg MOS. mRNA and protein levels of nuclear factor erythroid 2-related factor 2 also increased following supplementation with 400-600 mg/kg MOS. In addition, supplementation with 400-600 mg/kg MOS reduced excessive apoptosis by inhibiting the death receptor pathway and mitochondrial pathway processes. CONCLUSIONS Based on the quadratic regression analysis of the above biomarkers (reactive oxygen species, malondialdehyde, and protein carbonyl) of oxidative damage in the head kidney and spleen of on-growing grass carp, the recommended MOS supplementation is 575.21, 557.58, 531.86, 597.35, 570.16, and 553.80 mg/kg, respectively. Collectively, MOS supplementation could alleviate oxidative injury in the head kidney and spleen of grass carp infected with Aeromonas hydrophila.
Collapse
Affiliation(s)
- Zhiyuan Lu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu, 611130, China
| | - Weidan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu, 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu, 611130, China
| | - Jun Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu, 611130, China
| | - Shengyao Kuang
- Sichuan Animal Science Academy, Sichuan Animtech Feed Co. Ltd, Chengdu, 610066, China
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu, 610066, China
| | - Ling Tang
- Sichuan Animal Science Academy, Sichuan Animtech Feed Co. Ltd, Chengdu, 610066, China
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu, 610066, China
| | - Shuwei Li
- Sichuan Animal Science Academy, Sichuan Animtech Feed Co. Ltd, Chengdu, 610066, China
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu, 610066, China
| | - Chengbo Zhong
- Sichuan Animal Science Academy, Sichuan Animtech Feed Co. Ltd, Chengdu, 610066, China
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu, 610066, China
| | - Xiaoqiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China.
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China.
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
17
|
Qin X, Zhu L, Zhong Y, Wang Y, Wu G, Qiu J, Wang G, Qu K, Zhang K, Wu W. Spontaneously Right-Side-Out-Orientated Coupling-Driven ROS-Sensitive Nanoparticles on Cell Membrane Inner Leaflet for Efficient Renovation in Vascular Endothelial Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205093. [PMID: 36703487 PMCID: PMC9951580 DOI: 10.1002/advs.202205093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 12/19/2022] [Indexed: 06/18/2023]
Abstract
Biomimetic cell membrane camouflaged technology has drawn extensive attention as a feasible and efficient way to realize the biological functions of nanoparticles from the parent cells. As the burgeoning nanotherapeutic, the right-side-out orientation self-assembly and pathological dependent "on-demand" cargo release of cell membrane camouflaged nanocarriers remarkably limit further development for practical applications. In the present study, a spontaneously right-side-out-orientated coupling-driven ROS-sensitive nanotherapeutic has been constructed for target endothelial cells (ECs) repair through the synergistic effects of spontaneously right-side-out-orientated camouflaging. This condition results from the specific affinity between the intracellular domain of key transmembrane receptors band 3 on cell membrane inner leaflet and the corresponding P4.2 peptide-modified nanoparticles without the additional coextrusion. The "on-demand" cargo release results from the pathological ROS-cleavable prodrug. Particularly, the red blood cell camouflaged nanotherapeutics (RBC-LVTNPs) can enhance target drug delivery through low oscillatory shear stress (LSS) blood flow in the injured ECs lesion. Both in vitro and in vivo results collectively confirm that RBC-LVTNPs can restore the damaged ECs and function with the recovered vascular permeability and low inflammation microenvironment. The findings provide a powerful and universal approach for developing the biomimetic cell membrane camouflaged nanotechnology.
Collapse
Affiliation(s)
- Xian Qin
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400030China
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic DiseasesChongqing University Three Gorges HospitalChongqing404000China
| | - Li Zhu
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400030China
| | - Yuan Zhong
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400030China
| | - Yi Wang
- College of Basic Medical SciencesChongqing Medical UniversityChongqing400016China
| | - Guicheng Wu
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic DiseasesChongqing University Three Gorges HospitalChongqing404000China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400030China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400030China
- JinFeng LaboratoryChongqing401329China
| | - Kai Qu
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400030China
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic DiseasesChongqing University Three Gorges HospitalChongqing404000China
| | - Kun Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400030China
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic DiseasesChongqing University Three Gorges HospitalChongqing404000China
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400030China
- JinFeng LaboratoryChongqing401329China
| |
Collapse
|
18
|
Li J, Ge P, He Q, Liu C, Zeng C, Tao C, Zhai Y, Wang J, Zhang Q, Wang R, Zhang Y, Zhang D, Zhao J. Association between methionine sulfoxide and risk of moyamoya disease. Front Neurosci 2023; 17:1158111. [PMID: 37123363 PMCID: PMC10130537 DOI: 10.3389/fnins.2023.1158111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/16/2023] [Indexed: 05/02/2023] Open
Abstract
Objective Methionine sulfoxide (MetO) has been identified as a risk factor for vascular diseases and was considered as an important indicator of oxidative stress. However, the effects of MetO and its association with moyamoya disease (MMD) remained unclear. Therefore, we performed this study to evaluate the association between serum MetO levels and the risk of MMD and its subtypes. Methods We eventually included consecutive 353 MMD patients and 88 healthy controls (HCs) with complete data from September 2020 to December 2021 in our analyzes. Serum levels of MetO were quantified using liquid chromatography-mass spectrometry (LC-MS) analysis. We evaluated the role of MetO in MMD using logistic regression models and confirmed by receiver-operating characteristic (ROC) curves and area under curve (AUC) values. Results We found that the levels of MetO were significantly higher in MMD and its subtypes than in HCs (p < 0.001 for all). After adjusting for traditional risk factors, serum MetO levels were significantly associated with the risk of MMD and its subtypes (p < 0.001 for all). We further divided the MetO levels into low and high groups, and the high MetO level was significantly associated with the risk of MMD and its subtypes (p < 0.05 for all). When MetO levels were assessed as quartiles, we found that the third (Q3) and fourth (Q4) MetO quartiles had a significantly increased risk of MMD compared with the lowest quartile (Q3, OR: 2.323, 95%CI: 1.088-4.959, p = 0.029; Q4, OR: 5.559, 95%CI: 2.088-14.805, p = 0.001). Conclusion In this study, we found that a high level of serum MetO was associated with an increased risk of MMD and its subtypes. Our study raised a novel perspective on the pathogenesis of MMD and suggested potential therapeutic targets.
Collapse
Affiliation(s)
- Junsheng Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Peicong Ge
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Qiheng He
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Chenglong Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Chaofan Zeng
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Chuming Tao
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuanren Zhai
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Jia Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Qian Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Rong Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Yan Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Dong Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
- *Correspondence: Dong Zhang,
| | - Jizong Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
- Savaid Medical School, University of the Chinese Academy of Sciences, Beijing, China
- Jizong Zhao,
| |
Collapse
|
19
|
Liang Z, Chen Y, Gu R, Guo Q, Nie X. Asiaticoside Prevents Oxidative Stress and Apoptosis in Endothelial Cells by Activating ROS-dependent p53/Bcl-2/Caspase-3 Signaling Pathway. Curr Mol Med 2023; 23:1116-1129. [PMID: 36284389 DOI: 10.2174/1566524023666221024120825] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/06/2022] [Accepted: 09/06/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Asiaticoside (AC) is a triterpenoid saponin found in Centella asiatica (L.) urban extract that has a wide range of pharmacological properties. Our previous study demonstrated that AC could promote angiogenesis in diabetic wounds, but the specific mechanisms remain unknown. OBJECTIVE This study aimed to examine the effectiveness and mechanism of AC on human umbilical vein endothelial cells (HUVECs) exposed to tert-butyl hydroperoxide (t-BHP) toxicity. METHODS Senescence was confirmed using senescence-associated betagalactosidase (SA-β-gal) activity and expression of the cell cycle phase markers p16 and p21. The levels of SOD, NO, MDA, GSH-Px, and ROS were tested. Furthermore, several cell death-related genes and proteins (p53, Bax, Bcl-2 and Caspase-3) were assessed with RT-qPCR and Western blotting. RESULTS AC significantly reduced SA-β-gal activity, with both the suppression of cellcycle inhibitors p16 and p21. We also found that the induced oxidative stress and apoptosis caused by t-BHP treatment resulted in the decrease of antioxidant enzymes activities, the surge of ROS and MDA, the up-regulation of p53, Bax and caspase-3, and the decrease of SOD, NO, GSH-Px and Bcl-2. These biochemical changes were all reversed by treatment with varying doses of AC. CONCLUSION AC alleviates t-BHP-induced oxidative injury and apoptosis in HUVECs through the ROS-dependent p53/Bcl-2/Caspase-3 signaling pathway. It may be a potential antioxidant applied in metabolic disorders and pharmaceutical products.
Collapse
Affiliation(s)
- Zhenwen Liang
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
- Key Laboratory of Basic Pharmacology of the Ministry of Education, Zunyi Medical University, Zunyi 563003, China
- College of Pharmacy, Zunyi Medical University, Zunyi 563003, China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563003, China
| | - Yu Chen
- Key Laboratory of Basic Pharmacology of the Ministry of Education, Zunyi Medical University, Zunyi 563003, China
- College of Pharmacy, Zunyi Medical University, Zunyi 563003, China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563003, China
| | - Rifang Gu
- College of Pharmacy, Zunyi Medical University, Zunyi 563003, China
| | - Qi Guo
- College of Rehabilitation Sciences, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Xuqiang Nie
- Key Laboratory of Basic Pharmacology of the Ministry of Education, Zunyi Medical University, Zunyi 563003, China
- College of Pharmacy, Zunyi Medical University, Zunyi 563003, China
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563003, China
| |
Collapse
|
20
|
Chen Y, Fu W, Zheng Y, Yang J, Liu Y, Qi Z, Wu M, Fan Z, Yin K, Chen Y, Gao W, Ding Z, Dong J, Li Q, Zhang S, Hu L. Galectin 3 enhances platelet aggregation and thrombosis via Dectin-1 activation: a translational study. Eur Heart J 2022; 43:3556-3574. [PMID: 35165707 PMCID: PMC9989600 DOI: 10.1093/eurheartj/ehac034] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 11/25/2021] [Accepted: 01/18/2022] [Indexed: 01/25/2023] Open
Abstract
AIMS Galectin-3, a β-galactoside-binding lectin, is abnormally increased in cardiovascular disease. Plasma Galectin-3 receives a Class II recommendation for heart failure management and has been extensively studied for multiple cellular functions. The direct effects of Galectin-3 on platelet activation remain unclear. This study explores the direct effects of Galectin-3 on platelet activation and thrombosis. METHODS AND RESULTS A strong positive correlation between plasma Galectin-3 concentration and platelet aggregation or whole blood thrombus formation was observed in patients with coronary artery disease (CAD). Multiple platelet function studies demonstrated that Galectin-3 directly potentiated platelet activation and in vivo thrombosis. Mechanistic studies using the Dectin-1 inhibitor, laminarin, and Dectin-1-/- mice revealed that Galectin-3 bound to and activated Dectin-1, a receptor not previously reported in platelets, to phosphorylate spleen tyrosine kinase and thus increased Ca2+ influx, protein kinase C activation, and reactive oxygen species production to regulate platelet hyperreactivity. TD139, a Galectin-3 inhibitor in a Phase II clinical trial, concentration dependently suppressed Galectin-3-potentiated platelet activation and inhibited occlusive thrombosis without exacerbating haemorrhage in ApoE-/- mice, which spontaneously developed increased plasma Galectin-3 levels. TD139 also suppressed microvascular thrombosis to protect the heart from myocardial ischaemia-reperfusion injury in ApoE-/- mice. CONCLUSION Galectin-3 is a novel positive regulator of platelet hyperreactivity and thrombus formation in CAD. As TD139 has potent antithrombotic effects without bleeding risk, Galectin-3 inhibitors may have therapeutic advantages as potential antiplatelet drugs for patients with high plasma Galectin-3 levels.
Collapse
Affiliation(s)
- Yufei Chen
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.,Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Wanrong Fu
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yunbo Zheng
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Yang
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yangyang Liu
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhiyong Qi
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Meiling Wu
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Zhichao Fan
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT, USA
| | - Kanhua Yin
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yunfeng Chen
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Wen Gao
- Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhongren Ding
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianzeng Dong
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qi Li
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Si Zhang
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugates Research, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Liang Hu
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.,Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
21
|
Plasma Oxylipins and Their Precursors Are Strongly Associated with COVID-19 Severity and with Immune Response Markers. Metabolites 2022; 12:metabo12070619. [PMID: 35888743 PMCID: PMC9319897 DOI: 10.3390/metabo12070619] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 12/13/2022] Open
Abstract
COVID-19 is characterised by a dysregulated immune response, that involves signalling lipids acting as mediators of the inflammatory process along the innate and adaptive phases. To promote understanding of the disease biochemistry and provide targets for intervention, we applied a range of LC-MS platforms to analyse over 100 plasma samples from patients with varying COVID-19 severity and with detailed clinical information on inflammatory responses (>30 immune markers). The second publication in a series reports the results of quantitative LC-MS/MS profiling of 63 small lipids including oxylipins, free fatty acids, and endocannabinoids. Compared to samples taken from ward patients, intensive care unit (ICU) patients had 2−4-fold lower levels of arachidonic acid (AA) and its cyclooxygenase-derived prostanoids, as well as lipoxygenase derivatives, exhibiting negative correlations with inflammation markers. The same derivatives showed 2−5-fold increases in recovering ward patients, in paired comparison to early hospitalisation. In contrast, ICU patients showed elevated levels of oxylipins derived from poly-unsaturated fatty acids (PUFA) by non-enzymatic peroxidation or activity of soluble epoxide hydrolase (sEH), and these oxylipins positively correlated with markers of macrophage activation. The deficiency in AA enzymatic products and the lack of elevated intermediates of pro-resolving mediating lipids may result from the preference of alternative metabolic conversions rather than diminished stores of PUFA precursors. Supporting this, ICU patients showed 2-to-11-fold higher levels of linoleic acid (LA) and the corresponding fatty acyl glycerols of AA and LA, all strongly correlated with multiple markers of excessive immune response. Our results suggest that the altered oxylipin metabolism disrupts the expected shift from innate immune response to resolution of inflammation.
Collapse
|
22
|
Karu N, Kindt A, van Gammeren AJ, Ermens AAM, Harms AC, Portengen L, Vermeulen RCH, Dik WA, Langerak AW, van der Velden VHJ, Hankemeier T. Severe COVID-19 Is Characterised by Perturbations in Plasma Amines Correlated with Immune Response Markers, and Linked to Inflammation and Oxidative Stress. Metabolites 2022; 12:618. [PMID: 35888742 PMCID: PMC9321395 DOI: 10.3390/metabo12070618] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/25/2022] [Accepted: 06/26/2022] [Indexed: 02/01/2023] Open
Abstract
The COVID-19 pandemic raised a need to characterise the biochemical response to SARS-CoV-2 infection and find biological markers to identify therapeutic targets. In support of these aims, we applied a range of LC-MS platforms to analyse over 100 plasma samples from patients with varying COVID-19 severity and with detailed clinical information on inflammatory responses (>30 immune markers). The first publication in a series reports the results of quantitative LC-MS/MS profiling of 56 amino acids and derivatives. A comparison between samples taken from ICU and ward patients revealed a notable increase in ten post-translationally modified amino acids that correlated with markers indicative of an excessive immune response: TNF-alpha, neutrophils, markers for macrophage, and leukocyte activation. Severe patients also had increased kynurenine, positively correlated with CRP and cytokines that induce its production. ICU and ward patients with high IL-6 showed decreased levels of 22 immune-supporting and anti-oxidative amino acids and derivatives (e.g., glutathione, GABA). These negatively correlated with CRP and IL-6 and positively correlated with markers indicative of adaptive immune activation. Including corresponding alterations in convalescing ward patients, the overall metabolic picture of severe COVID-19 reflected enhanced metabolic demands to maintain cell proliferation and redox balance, alongside increased inflammation and oxidative stress.
Collapse
Affiliation(s)
- Naama Karu
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands; (A.K.); (A.C.H.)
| | - Alida Kindt
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands; (A.K.); (A.C.H.)
| | - Adriaan J. van Gammeren
- Department of Clinical Chemistry and Hematology, Amphia Hospital, 4818 CK Breda, The Netherlands; (A.J.v.G.); (A.A.M.E.)
| | - Anton A. M. Ermens
- Department of Clinical Chemistry and Hematology, Amphia Hospital, 4818 CK Breda, The Netherlands; (A.J.v.G.); (A.A.M.E.)
| | - Amy C. Harms
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands; (A.K.); (A.C.H.)
| | - Lutzen Portengen
- Department of Population Health Sciences, Institute for Risk Assessment Sciences, University Utrecht, 3584 CK Utrecht, The Netherlands; (L.P.); (R.C.H.V.)
| | - Roel C. H. Vermeulen
- Department of Population Health Sciences, Institute for Risk Assessment Sciences, University Utrecht, 3584 CK Utrecht, The Netherlands; (L.P.); (R.C.H.V.)
| | - Willem A. Dik
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (W.A.D.); (A.W.L.); (V.H.J.v.d.V.)
| | - Anton W. Langerak
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (W.A.D.); (A.W.L.); (V.H.J.v.d.V.)
| | - Vincent H. J. van der Velden
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (W.A.D.); (A.W.L.); (V.H.J.v.d.V.)
| | - Thomas Hankemeier
- Metabolomics and Analytics Centre, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands; (A.K.); (A.C.H.)
| |
Collapse
|
23
|
Nascimento RO, Prado FM, Massafera MP, Di Mascio P, Ronsein GE. Dehydromethionine is a common product of methionine oxidation by singlet molecular oxygen and hypohalous acids. Free Radic Biol Med 2022; 187:17-28. [PMID: 35580773 DOI: 10.1016/j.freeradbiomed.2022.05.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/29/2022] [Accepted: 05/05/2022] [Indexed: 10/18/2022]
Abstract
Methionine is one of the main targets for biological oxidants. Its reaction with the majority of oxidants generates only methionine sulfoxide. However, when N-terminal methionine reacts with hypohalous acids (HOCl and HOBr) or singlet molecular oxygen (1O2), it can also generate a cyclic product called dehydromethionine (DHM). Previously, DHM was suggested as a biomarker of oxidative stress induced by hypohalous acids. However, DHM can also be generated by 1O2 -oxidation of methionine, and the contribution of this pathway of DHM formation in a context of a site-specific redox imbalance in an organism is unknown. In this work, a through comparison of the reactions of hypohalous acids and 1O2 with methionine, either free or inserted in peptides and proteins was undertaken. In addition, we performed methionine photooxidation in heavy water (H218O) to determine the influence of the pH in the mechanism of DHM formation. We showed that for free methionine, or methionine-containing peptides, the yields of DHM formation in the reactions with 1O2 were close to those achieved by HOBr oxidation, but much higher than the yields obtained with HOCl as the oxidant. This was true for all pH tested (5, 7.4, and 9). Interestingly, for the protein ubiquitin, DHM yields after reaction with 1O2 were higher than those obtained with both hypohalous acids. Our results indicate that 1O2 may also be an important source of DHM in biological systems.
Collapse
Affiliation(s)
| | - Fernanda Manso Prado
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Mariana Pereira Massafera
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Paolo Di Mascio
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, 05508-000, Brazil.
| | - Graziella Eliza Ronsein
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, 05508-000, Brazil.
| |
Collapse
|
24
|
Gu SX, Dayal S. Redox Mechanisms of Platelet Activation in Aging. Antioxidants (Basel) 2022; 11:995. [PMID: 35624860 PMCID: PMC9137594 DOI: 10.3390/antiox11050995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/14/2022] [Accepted: 05/17/2022] [Indexed: 02/01/2023] Open
Abstract
Aging is intrinsically linked with physiologic decline and is a major risk factor for a broad range of diseases. The deleterious effects of advancing age on the vascular system are evidenced by the high incidence and prevalence of cardiovascular disease in the elderly. Reactive oxygen species are critical mediators of normal vascular physiology and have been shown to gradually increase in the vasculature with age. There is a growing appreciation for the complexity of oxidant and antioxidant systems at the cellular and molecular levels, and accumulating evidence indicates a causal association between oxidative stress and age-related vascular disease. Herein, we review the current understanding of mechanistic links between oxidative stress and thrombotic vascular disease and the changes that occur with aging. While several vascular cells are key contributors, we focus on oxidative changes that occur in platelets and their mediation in disease progression. Additionally, we discuss the impact of comorbid conditions (i.e., diabetes, atherosclerosis, obesity, cancer, etc.) that have been associated with platelet redox dysregulation and vascular disease pathogenesis. As we continue to unravel the fundamental redox mechanisms of the vascular system, we will be able to develop more targeted therapeutic strategies for the prevention and management of age-associated vascular disease.
Collapse
Affiliation(s)
- Sean X. Gu
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT 06511, USA;
| | - Sanjana Dayal
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Iowa City VA Healthcare System, Iowa City, IA 52246, USA
| |
Collapse
|
25
|
Li HY, Sun H, Zhang AH, He LW, Qiu S, Xue JR, Wu F, Wang XJ. Therapeutic Effect and Mechanism of Si-Miao-Yong-An-Tang on Thromboangiitis Obliterans Based on the Urine Metabolomics Approach. Front Pharmacol 2022; 13:827733. [PMID: 35273504 PMCID: PMC8902467 DOI: 10.3389/fphar.2022.827733] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/12/2022] [Indexed: 11/13/2022] Open
Abstract
Si-Miao-Yong-An-Tang (SMYAT) is a classic prescription for the treatment of thromboangiitis obliterans (TAO). However, the effect and mechanism are still unclear. This experiment aims to evaluate the therapeutic effect and mechanism of SMYAT on sodium laurate solution induced thromboangiitis obliterans model rats using urine metabolomics. The therapeutic effect of SMYAT was evaluated by histopathology, hemorheology and other indexes. The urine metabolomic method, principal component analysis (PCA) and orthogonal partial least squares discriminant analysis (OPLS-DA) were used for clustering group and discriminant analysis to screen urine differential metabolic biomarkers, and explore new insight into pathophysiological mechanisms of SMYAT in the treatment of TAO. SMYAT has significant antithrombotic and anti-inflammatory effects, according to the results of urine metabolomic analysis, and regulate the metabolic profile of TAO rats, and its return profile is close to the state of control group. Through metabolomics technology, a total of 35 urine biomarkers of TAO model were characterized. Among them, SMYAT treatment can regulate 22 core biomarkers, such as normetanephrine and 4-pyridoxic acid. It is found that the therapeutic effect of SMYAT is closely related to the tyrosine metabolism, vitamin B6 metabolism and cysteine and methionine metabolism. It preliminarily explored the therapeutic mechanism of SMYAT, and provided a scientific basis for the application of SMYAT.
Collapse
Affiliation(s)
- Hui-Yu Li
- National Engineering Laboratory for the Development of Southwestern Endangered Medicinal Materials, Guangxi Botanical Garden of Medicinal Plant, Nanning, China
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Functional Metabolomics Laboratory, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Hui Sun
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Functional Metabolomics Laboratory, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ai-Hua Zhang
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Functional Metabolomics Laboratory, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Lu-Wen He
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Functional Metabolomics Laboratory, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shi Qiu
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Functional Metabolomics Laboratory, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jun-Ru Xue
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Functional Metabolomics Laboratory, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Fangfang Wu
- National Engineering Laboratory for the Development of Southwestern Endangered Medicinal Materials, Guangxi Botanical Garden of Medicinal Plant, Nanning, China
| | - Xi-Jun Wang
- National Engineering Laboratory for the Development of Southwestern Endangered Medicinal Materials, Guangxi Botanical Garden of Medicinal Plant, Nanning, China
- National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Functional Metabolomics Laboratory, Heilongjiang University of Chinese Medicine, Harbin, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao SAR, China
| |
Collapse
|
26
|
Microbiome and metabolome features of the cardiometabolic disease spectrum. Nat Med 2022; 28:303-314. [PMID: 35177860 PMCID: PMC8863577 DOI: 10.1038/s41591-022-01688-4] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 01/07/2022] [Indexed: 02/08/2023]
Abstract
Previous microbiome and metabolome analyses exploring non-communicable diseases have paid scant attention to major confounders of study outcomes, such as common, pre-morbid and co-morbid conditions, or polypharmacy. Here, in the context of ischemic heart disease (IHD), we used a study design that recapitulates disease initiation, escalation and response to treatment over time, mirroring a longitudinal study that would otherwise be difficult to perform given the protracted nature of IHD pathogenesis. We recruited 1,241 middle-aged Europeans, including healthy individuals, individuals with dysmetabolic morbidities (obesity and type 2 diabetes) but lacking overt IHD diagnosis and individuals with IHD at three distinct clinical stages—acute coronary syndrome, chronic IHD and IHD with heart failure—and characterized their phenome, gut metagenome and serum and urine metabolome. We found that about 75% of microbiome and metabolome features that distinguish individuals with IHD from healthy individuals after adjustment for effects of medication and lifestyle are present in individuals exhibiting dysmetabolism, suggesting that major alterations of the gut microbiome and metabolome might begin long before clinical onset of IHD. We further categorized microbiome and metabolome signatures related to prodromal dysmetabolism, specific to IHD in general or to each of its three subtypes or related to escalation or de-escalation of IHD. Discriminant analysis based on specific IHD microbiome and metabolome features could better differentiate individuals with IHD from healthy individuals or metabolically matched individuals as compared to the conventional risk markers, pointing to a pathophysiological relevance of these features. By studying individuals along a spectrum of cardiometabolic disease and adjusting for effects of lifestyle and medication, this investigation identifies alterations of the metabolome and microbiome from dysmetabolic conditions, such as obesity and type 2 diabetes, to ischemic heart disease.
Collapse
|
27
|
Wu Y, Wu ZM, Zhang SS, Liu LY, Sun F, Jiao WH, Wang SP, Lin HW. Axinellasins A-D, Immunosuppressive Cycloheptapeptide Diastereomers, Discovered via a Precursor Ion Scanning-Supercritical Fluid Chromatography Strategy from the Marine Sponge Axinella species. Org Lett 2022; 24:934-938. [PMID: 35044186 DOI: 10.1021/acs.orglett.1c04309] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The precursor ion scanning-supercritical fluid chromatography (PI-SFC) method was applied to explore new methionine sulfoxide-containing cycloheptapeptides, axinellasins A-D (1-4), from the marine sponge Axinella sp. Their structures, including absolute configurations, were elucidated by detailed spectroscopic analyses and X-ray crystallography. The total synthesis of 4 was completed via an Fmoc solid/solution-phase synthesis. Compounds 1-4 exhibited immunosuppressive effects via inhibition of T and B cell proliferation, and 1 and 4 showed better inhibitory activities than their corresponding diastereomers.
Collapse
Affiliation(s)
- Ying Wu
- Research Center for Marine Drugs, State Key Laboratory of Oncogene and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Zong-Mei Wu
- Research Center for Marine Drugs, State Key Laboratory of Oncogene and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Shuai-Shuai Zhang
- Research Center for Marine Drugs, State Key Laboratory of Oncogene and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Li-Yun Liu
- Research Center for Marine Drugs, State Key Laboratory of Oncogene and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Fan Sun
- Research Center for Marine Drugs, State Key Laboratory of Oncogene and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Wei-Hua Jiao
- Research Center for Marine Drugs, State Key Laboratory of Oncogene and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Shu-Ping Wang
- Research Center for Marine Drugs, State Key Laboratory of Oncogene and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Hou-Wen Lin
- Research Center for Marine Drugs, State Key Laboratory of Oncogene and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
28
|
Gadgaard S, van der Velden WJC, Schiellerup SP, Hunt JE, Gabe MBN, Windeløv JA, Boer GA, Kissow H, Ørskov C, Holst JJ, Hartmann B, Rosenkilde MM. Novel agonist- and antagonist-based radioligands for the GLP-2 receptor - useful tools for studies of basic GLP-2R pharmacology. Br J Pharmacol 2021; 179:1998-2015. [PMID: 34855984 PMCID: PMC9303331 DOI: 10.1111/bph.15766] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/22/2021] [Accepted: 11/10/2021] [Indexed: 11/27/2022] Open
Abstract
Background Glucagon‐like peptide‐2 (GLP‐2) is a pro‐glucagon‐derived hormone secreted from intestinal enteroendocrine L cells with actions on gut and bones. GLP‐2(1–33) is cleaved by DPP‐4, forming GLP‐2(3–33), having low intrinsic activity and competitive antagonism properties at GLP‐2 receptors. We created radioligands based on these two molecules. Experimental approach The methionine in position 10 of GLP‐2(1–33) and GLP‐2(3–33) was substituted with tyrosine (M10Y) enabling oxidative iodination, creating [125I]‐hGLP‐2(1–33,M10Y) and [125I]‐hGLP‐2(3–33,M10Y). Both were characterized by competition binding, on‐and‐off‐rate determination and receptor activation. Receptor expression was determined by target‐tissue autoradiography and immunohistochemistry. Key results Both M10Y‐substituted peptides induced cAMP production via the GLP‐2 receptor comparable to the wildtype peptides. GLP‐2(3–33,M10Y) maintained the antagonistic properties of GLP‐2(3–33). However, hGLP‐2(1–33,M10Y) had lower arrestin recruitment than hGLP‐2(1–33). High affinities for the hGLP‐2 receptor were observed using [125I]‐hGLP‐2(1–33,M10Y) and [125I]‐hGLP‐2(3–33,M10Y) with KD values of 59.3 and 40.6 nM. The latter (with antagonistic properties) had higher Bmax and faster on and off rates compared to the former (full agonist). Both bound the hGLP‐1 receptor with low affinity (Ki of 130 and 330 nM, respectively). Autoradiography in wildtype mice revealed strong labelling of subepithelial myofibroblasts, confirmed by immunohistochemistry using a GLP‐2 receptor specific antibody that in turn was confirmed in GLP‐2 receptor knock‐out mice. Conclusion and implications Two new radioligands with different binding kinetics, one a full agonist and the other a weak partial agonist with antagonistic properties were developed and subepithelial myofibroblasts identified as a major site for GLP‐2 receptor expression.
Collapse
Affiliation(s)
- Sarina Gadgaard
- Laboratory of Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Bainan Biotech, Copenhagen, Denmark
| | - Wijnand J C van der Velden
- Laboratory of Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Present address: Department of Computational & Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, California, USA
| | - Sine P Schiellerup
- Laboratory of Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jenna Elizabeth Hunt
- Endocrinology and Metabolism, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Maria B N Gabe
- Laboratory of Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Johanne Agerlin Windeløv
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Geke Aline Boer
- Endocrinology and Metabolism, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Hannelouise Kissow
- Endocrinology and Metabolism, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Cathrine Ørskov
- Endocrinology and Metabolism, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Jens J Holst
- Endocrinology and Metabolism, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Bolette Hartmann
- Endocrinology and Metabolism, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Mette M Rosenkilde
- Laboratory of Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
29
|
Macrophage membrane camouflaged reactive oxygen species responsive nanomedicine for efficiently inhibiting the vascular intimal hyperplasia. J Nanobiotechnology 2021; 19:374. [PMID: 34789284 PMCID: PMC8600790 DOI: 10.1186/s12951-021-01119-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/02/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Intimal hyperplasia caused by vascular injury is an important pathological process of many vascular diseases, especially occlusive vascular disease. In recent years, Nano-drug delivery system has attracted a wide attention as a novel treatment strategy, but there are still some challenges such as high clearance rate and insufficient targeting. RESULTS In this study, we report a biomimetic ROS-responsive MM@PCM/RAP nanoparticle coated with macrophage membrane. The macrophage membrane with the innate "homing" capacity can superiorly regulate the recruitment of MM@PCM/RAP to inflammatory lesion to enhance target efficacy, and can also disguise MM@PCM/RAP nanoparticle as the autologous cell to avoid clearance by the immune system. In addition, MM@PCM/RAP can effectively improve the solubility of rapamycin and respond to the high concentration level of ROS accumulated in pathological lesion for controlling local cargo release, thereby increasing drug availability and reducing toxic side effects. CONCLUSIONS Our findings validate that the rational design, biomimetic nanoparticles MM@PCM/RAP, can effectively inhibit the pathological process of intimal injury with excellent biocompatibility.
Collapse
|
30
|
Metabolomic Profile in Venous Thromboembolism (VTE). Metabolites 2021; 11:metabo11080495. [PMID: 34436436 PMCID: PMC8400436 DOI: 10.3390/metabo11080495] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 01/19/2023] Open
Abstract
Venous thromboembolism (VTE) is a condition comprising deep venous thrombosis (DVT) and pulmonary embolism (PE). The prevalence of this disease is constantly increasing and it is also a chief reason for morbidity. Therefore, the primary prevention of VTE remains a highly important public health issue. At present, its diagnosis generally relies on subjective clinical examination and ultrasound imaging. D-dimer is also used as a biomarker, but it is considered to be poorly specific and only moderately sensitive. There are also no reliable methods that could accurately guide the type of treatment and potentially identify patients who may benefit from more aggressive therapies without the risk of bleeding. The application of metabolomics profiling in the area of vascular diseases may become a turning point in early diagnosis and patient management. Among the most described metabolites possibly related to VTE are carnitine species, glucose, phenylalanine, 3-hydroxybutarate, lactic acid, tryptophan and some monounsaturated and polyunsaturated fatty acids. The cell response to acute PE was suggested to involve the uncoupling between glycolysis and oxidative phosphorylation. Despite technological advancement in the identification of metabolites and their alteration in thrombosis, we still do not understand the mechanisms and pathways responsible for the occurrence of observed alterations.
Collapse
|
31
|
Kehm R, Baldensperger T, Raupbach J, Höhn A. Protein oxidation - Formation mechanisms, detection and relevance as biomarkers in human diseases. Redox Biol 2021; 42:101901. [PMID: 33744200 PMCID: PMC8113053 DOI: 10.1016/j.redox.2021.101901] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/06/2021] [Accepted: 02/12/2021] [Indexed: 12/16/2022] Open
Abstract
Generation of reactive oxygen species and related oxidants is an inevitable consequence of life. Proteins are major targets for oxidation reactions, because of their rapid reaction rates with oxidants and their high abundance in cells, extracellular tissues, and body fluids. Additionally, oxidative stress is able to degrade lipids and carbohydrates to highly reactive intermediates, which eventually attack proteins at various functional sites. Consequently, a wide variety of distinct posttranslational protein modifications is formed by protein oxidation, glycoxidation, and lipoxidation. Reversible modifications are relevant in physiological processes and constitute signaling mechanisms ("redox signaling"), while non-reversible modifications may contribute to pathological situations and several diseases. A rising number of publications provide evidence for their involvement in the onset and progression of diseases as well as aging processes. Certain protein oxidation products are chemically stable and formed in large quantity, which makes them promising candidates to become biomarkers of oxidative damage. Moreover, progress in the development of detection and quantification methods facilitates analysis time and effort and contributes to their future applicability in clinical routine. The present review outlines the most important classes and selected examples of oxidative protein modifications, elucidates the chemistry beyond their formation and discusses available methods for detection and analysis. Furthermore, the relevance and potential of protein modifications as biomarkers in the context of disease and aging is summarized.
Collapse
Affiliation(s)
- Richard Kehm
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Germany.
| | - Tim Baldensperger
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Germany.
| | - Jana Raupbach
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Germany.
| | - Annika Höhn
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764, Muenchen-Neuherberg, Germany.
| |
Collapse
|
32
|
Dillinger JG, Laine M, Bouajila S, Paganelli F, Henry P, Bonello L. Antithrombotic strategies in elderly patients with acute coronary syndrome. Arch Cardiovasc Dis 2021; 114:232-245. [PMID: 33632631 DOI: 10.1016/j.acvd.2020.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 12/21/2020] [Accepted: 12/21/2020] [Indexed: 12/22/2022]
Abstract
Elderly patients represent a growing proportion of the acute coronary syndrome population in Western countries. However, their frequent atypical symptoms at presentation often lead to delays in management and to misdiagnosis. Furthermore, their prognosis is poorer than that of younger patients because of physiological changes in platelet function, haemostasis and fibrinolysis, but also a higher proportion of comorbidities and frailty, both of which increase the risk of recurrent thrombotic and bleeding events. This complex situation, with ischaemic and haemorrhagic risk factors often being intertwined, may lead to confusion about the required treatment strategy, sometimes resulting in inadequate management or even to therapeutic nihilism. It is therefore critical to provide a comprehensive overview of our understanding of the pathophysiological processes underlying acute coronary syndrome in elderly patients, and to summarise the results from the latest clinical trials to help decision making for these high-risk patients.
Collapse
Affiliation(s)
- Jean-Guillaume Dillinger
- Department of cardiology, Hôpital Lariboisière, AP-HP, Inserm U-942, Université de Paris, 2, rue Ambroise-Paré, 75010 Paris, France.
| | - Marc Laine
- Mediterranean Association for research and studies in cardiology (MARS cardio), Centre for cardiovascular and nutrition research, AP-HM, Aix-Marseille University, INSERM 1263, INRA 1260, 13015 Marseille, France; Cardiology department, Hôpital Nord, 13015 Marseille, France; Mediterranean Association for research and studies in cardiology (MARS cardio), 13015 Marseille, France
| | - Sara Bouajila
- Department of cardiology, Hôpital Lariboisière, AP-HP, Inserm U-942, Université de Paris, 2, rue Ambroise-Paré, 75010 Paris, France
| | - Franck Paganelli
- Mediterranean Association for research and studies in cardiology (MARS cardio), Centre for cardiovascular and nutrition research, AP-HM, Aix-Marseille University, INSERM 1263, INRA 1260, 13015 Marseille, France; Cardiology department, Hôpital Nord, 13015 Marseille, France
| | - Patrick Henry
- Department of cardiology, Hôpital Lariboisière, AP-HP, Inserm U-942, Université de Paris, 2, rue Ambroise-Paré, 75010 Paris, France
| | - Laurent Bonello
- Mediterranean Association for research and studies in cardiology (MARS cardio), Centre for cardiovascular and nutrition research, AP-HM, Aix-Marseille University, INSERM 1263, INRA 1260, 13015 Marseille, France; Cardiology department, Hôpital Nord, 13015 Marseille, France; Mediterranean Association for research and studies in cardiology (MARS cardio), 13015 Marseille, France
| |
Collapse
|
33
|
Tsai R, Interlandi G. Oxidation shuts down an auto-inhibitory mechanism of von Willebrand factor. Proteins 2021; 89:731-741. [PMID: 33550613 DOI: 10.1002/prot.26055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 12/24/2020] [Accepted: 01/31/2021] [Indexed: 01/13/2023]
Abstract
The blood protein von Willebrand factor (VWF) is a key link between inflammation and pathological thrombus formation. In particular, oxidation of methionine residues in specific domains of VWF due to the release of oxidants in inflammatory conditions has been linked to an increased platelet-binding activity. However, the atomistic details of how methionine oxidation activates VWF have not been elucidated to date. Yet understanding the activation mechanism of VWF under oxidizing conditions can lead to the development of novel therapeutics that target VWF selectively under inflammatory conditions in order to reduce its thrombotic activity while maintaining its haemostatic function. In this manuscript, we used a combination of a dynamic flow assay and molecular dynamics (MD) simulations to investigate how methionine oxidation removes an auto-inhibitory mechanism of VWF. Results from the dynamic flow assay revealed that oxidation does not directly activate the A1 domain, which is the domain in VWF that contains the binding site to the platelet surface receptor glycoprotein Ibα (GpIbα), but rather removes the inhibitory function of the neighboring A2 and A3 domains. Furthermore, the MD simulations combined with free energy perturbation calculations suggested that methionine oxidation may destabilize the binding interface between the A1 and A2 domains leading to unmasking of the GpIbα-binding site in the A1 domain.
Collapse
Affiliation(s)
- Rachel Tsai
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Gianluca Interlandi
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| |
Collapse
|
34
|
Zhang C, Recacha R, Ruddock LW, Moilanen A. Efficient soluble production of folded cat allergen Fel d 1 in Escherichia coli. Protein Expr Purif 2020; 180:105809. [PMID: 33338588 DOI: 10.1016/j.pep.2020.105809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 10/22/2022]
Abstract
The major cat allergen Fel d 1 is one of the most common and potent causes of animal related allergy. Medical treatment of cat allergy has relied on immunotherapy carried out with cat dander extract. This approach has been problematic, mainly due to inconsistent levels of the major allergen in the produced extracts. Recombinant DNA technology has been proposed as an alternative method to produce more consistent pharmaceuticals for immunotherapy and diagnostics of allergy. Current approaches to produce recombinant Fel d 1 (recFel d 1) in the cytoplasm of Escherichia coli have however resulted in protein folding deficiencies and insoluble inclusion body formation, requiring elaborate in vitro processing to acquire folded material. In this study, we introduce an efficient method for cytoplasmic production of recFel d 1 that utilizes eukaryotic folding factors to aid recFel d 1 to fold and be produced in the soluble fraction of E. coli. The solubly expressed recFel d 1 is shown by biophysical in vitro experiments to contain structural disulfides, is extremely stable, and has a sensitivity for methionine sulfoxidation. The latter is discussed in the context of functional relevance.
Collapse
Affiliation(s)
- Chi Zhang
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie 7, 90220, Oulu, Finland.
| | - Rosario Recacha
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie 7, 90220, Oulu, Finland.
| | - Lloyd W Ruddock
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie 7, 90220, Oulu, Finland.
| | - Antti Moilanen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie 7, 90220, Oulu, Finland.
| |
Collapse
|
35
|
Ligthart-Melis GC, Engelen MPKJ, Simbo SY, Ten Have GAM, Thaden JJ, Cynober L, Deutz NEP. Metabolic Consequences of Supplemented Methionine in a Clinical Context. J Nutr 2020; 150:2538S-2547S. [PMID: 33000166 DOI: 10.1093/jn/nxaa254] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 04/01/2020] [Accepted: 07/31/2020] [Indexed: 12/27/2022] Open
Abstract
The central position of methionine (Met) in protein metabolism indicates the importance of this essential amino acid for growth and maintenance of lean body mass. Therefore, Met might be a tempting candidate for supplementation. However, because Met is also the precursor of homocysteine (Hcy), a deficient intake of B vitamins or excessive intake of Met may result in hyperhomocysteinemia (HHcy), which is a risk factor for cardiovascular disease. This review discusses the evidence generated in preclinical and clinical studies on the importance and potentially harmful effects of Met supplementation and elaborates on potential clinical applications of supplemental Met with reference to clinical studies performed over the past 20 y. Recently acquired knowledge about the NOAEL (no observed adverse effect level) of 46.3 mg · kg-1 · d-1 and the LOAEL (lowest observed adverse effect level) of 91 mg · kg-1 · d-1 of supplemented Met will guide the design of future studies to further establish the role of Met as a potential (safe) candidate for nutritional supplementation in clinical applications.
Collapse
Affiliation(s)
- Gerdien C Ligthart-Melis
- Center for Translational Research in Aging & Longevity, Department of Health & Kinesiology, Texas A&M University, College Station, Texas, USA
| | - Mariëlle P K J Engelen
- Center for Translational Research in Aging & Longevity, Department of Health & Kinesiology, Texas A&M University, College Station, Texas, USA
| | - Sunday Y Simbo
- Center for Translational Research in Aging & Longevity, Department of Health & Kinesiology, Texas A&M University, College Station, Texas, USA
| | - Gabrie A M Ten Have
- Center for Translational Research in Aging & Longevity, Department of Health & Kinesiology, Texas A&M University, College Station, Texas, USA
| | - John J Thaden
- Center for Translational Research in Aging & Longevity, Department of Health & Kinesiology, Texas A&M University, College Station, Texas, USA
| | - Luc Cynober
- Department of Clinical Chemistry, Hôpital Cochin, Hôpitaux Universitaires Paris Centre, Paris, France
| | - Nicolaas E P Deutz
- Center for Translational Research in Aging & Longevity, Department of Health & Kinesiology, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
36
|
Abstract
![]()
The
methionine–iodine reaction was reinvestigated spectrophotometrically
in detail monitoring the absorbance belonging to the isosbestic point
of iodine at 468 nm, at T = 25.0 ± 0.1 °C,
and at 0.5 M ionic strength in buffered acidic medium. The stoichiometric
ratio of the reactants was determined to be 1:1 producing methionine
sulfoxide as the lone sulfur-containing product. The direct reaction
between methionine and iodine was found to be relatively rapid in
the absence of initially added iodide ion, and it can conveniently
be followed by the stopped-flow technique. Reduction of iodine eventually
leads to the formation of iodide ion that inhibits the reaction making
the whole system autoinhibitory with respect to the halide ion. We
have also shown that this inhibitory effect appears quite prominently,
and addition of iodide ion in the millimole concentration range may
result in a rate law where the formal kinetic order of this species
becomes −2. In contrast to this, hydrogen ion has just a mildly
inhibitory effect giving rise to the fact that iodine is the kinetically
active species in the system but not hypoiodous acid. The surprisingly
complex kinetics of this simple reaction may readily be interpreted
via the initiating rapidly established iodonium-transfer process between
the reactants followed by the subsequent hydrolytic decomposition
of the short-lived iodinated methionine. A seven-step kinetic model
to be able to describe the most important characteristics of the measured
kinetic curves is established and discussed in detail.
Collapse
Affiliation(s)
- Li Xu
- Department of Chemical Engineering and Technology, School of Chemistry, Biology and Material of Science, East China University of Technology, Nanchang 330013, Jiangxi Province People's Republic of China
| | - György Csekő
- Department of Inorganic Chemistry, Faculty of Sciences, University of Pécs, Ifjúság u. 6, Pécs, Hungary, H-7624
| | - Attila K Horváth
- Department of Inorganic Chemistry, Faculty of Sciences, University of Pécs, Ifjúság u. 6, Pécs, Hungary, H-7624
| |
Collapse
|
37
|
Bogdanović Pristov J, Opačić M, Bajčetić M, Mandić V, Maglić D, Miković Ž, Spasojević I. Oxidative status of maternal blood in pregnancies burdened by inherited thrombophilias. PLoS One 2020; 15:e0234253. [PMID: 32555583 PMCID: PMC7299371 DOI: 10.1371/journal.pone.0234253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 05/21/2020] [Indexed: 11/18/2022] Open
Abstract
Oxidative status of maternal blood represents an important parameter of pregnancy that is involved in both, regulation of physiological processes and (if significantly altered) development of different pregnancy complications. Inherited thrombophilias represent genetic disorders that increase the risk of thromboembolism in pregnancy. Little is known about the impact of thrombophilia on the oxidative status of maternal blood. In this study, we analyzed oxidative status of blood of 56 women with pregnancies burdened by inherited thrombophilias. The status was established at three different trimesters using biochemical assays and electrochemical measurements, and it was compared to 10 age- and trimester-matching controls. Activities of superoxide dismutase, catalase, and glutathione reductase in the 1st and the 2nd trimester of thrombophilic pregnancy were lower than controls. Also, there was less oxidation in the plasma, according to higher concentration of reduced thiols and lower oxidation-reduction potential. Therefore, it appears that thrombophilic mothers do not experience oxidative stress in the circulation in the first two trimesters. However, the rise in GPx, GR and SOD activities in the 3rd trimester of thrombophilic pregnancy implies that the risk of oxidative stress is increased during the late pregnancy. These results are important for developing antioxidative treatment that could tackle thrombophilia-related pregnancy complications.
Collapse
Affiliation(s)
- Jelena Bogdanović Pristov
- Life Sciences Department, Institute for Multidisciplinary Research, University of Belgrade, Belgrade, Serbia
| | - Miloš Opačić
- Life Sciences Department, Institute for Multidisciplinary Research, University of Belgrade, Belgrade, Serbia
| | - Milica Bajčetić
- Department of Pharmacology, Clinical Pharmacology and Toxicology, School of Medicine, University of Belgrade, Belgrade, Serbia.,Clinical Pharmacology Unit, University Children's Hospital, Belgrade, Serbia
| | - Vesna Mandić
- Department for High-risk Pregnancies, University Clinic for Gynecology and Obstetrics "Narodni front", Belgrade, Serbia.,Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Dragana Maglić
- Department for High-risk Pregnancies, University Clinic for Gynecology and Obstetrics "Narodni front", Belgrade, Serbia.,Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Željko Miković
- Department for High-risk Pregnancies, University Clinic for Gynecology and Obstetrics "Narodni front", Belgrade, Serbia.,Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Ivan Spasojević
- Life Sciences Department, Institute for Multidisciplinary Research, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
38
|
Holte KB, Svanteson M, Hanssen KF, Sveen KA, Seljeflot I, Solheim S, Sell DR, Monnier VM, Berg TJ. Collagen methionine sulfoxide and glucuronidine/LW-1 are markers of coronary artery disease in long-term survivors with type 1 diabetes. The Dialong study. PLoS One 2020; 15:e0233174. [PMID: 32401813 PMCID: PMC7219747 DOI: 10.1371/journal.pone.0233174] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 04/29/2020] [Indexed: 01/07/2023] Open
Abstract
Objectives Type 1 diabetes is a risk factor for coronary heart disease. The underlying mechanism behind the accelerated atherosclerosis formation is not fully understood but may be related to the formation of oxidation products and advanced glycation end-products (AGEs). We aimed to examine the associations between the collagen oxidation product methionine sulfoxide; the collagen AGEs methylglyoxal hydroimidazolone (MG-H1), glucosepane, pentosidine, glucuronidine/LW-1; and serum receptors for AGE (RAGE) with measures of coronary artery disease in patients with long-term type 1 diabetes. Methods In this cross-sectional study, 99 participants with type 1 diabetes of ≥ 45-year duration and 63 controls without diabetes had either established coronary heart disease (CHD) or underwent Computed Tomography Coronary Angiography (CTCA) measuring total, calcified and soft/mixed plaque volume. Skin collagen methionine sulfoxide and AGEs were measured by liquid chromatography-mass spectrometry and serum sRAGE/esRAGE by ELISA. Results In the diabetes group, low levels of methionine sulfoxide (adjusted for age, sex and mean HbA1c) were associated with normal coronary arteries, OR 0.48 (95% CI 0.27–0.88). Glucuronidine/LW-1 was associated with established CHD, OR 2.0 (1.16–3.49). MG-H1 and glucuronidine/LW-1 correlated with calcified plaque volume (r = 0.23–0.28, p<0.05), while pentosidine correlated with soft/mixed plaque volume (r = 0.29, p = 0.008), also in the adjusted analysis. Conclusions Low levels of collagen-bound methionine sulfoxide were associated with normal coronary arteries while glucuronidine/LW-1 was positively associated with established CHD in long-term type 1 diabetes, suggesting a role for metabolic and oxidative stress in the formation of atherosclerosis in diabetes.
Collapse
Affiliation(s)
- Kristine B. Holte
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- * E-mail:
| | - Mona Svanteson
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Kristian F. Hanssen
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- The Norwegian Diabetics’ Center, Oslo, Norway
| | - Kari Anne Sveen
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Ingebjørg Seljeflot
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital, Oslo, Norway
| | - Svein Solheim
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital, Oslo, Norway
| | - David R. Sell
- Department of Pathology and Biochemistry, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Vincent M. Monnier
- Department of Pathology and Biochemistry, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Tore Julsrud Berg
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
39
|
Ma N, Yang Y, Liu X, Li S, Qin Z, Li J. Plasma metabonomics and proteomics studies on the anti-thrombosis mechanism of aspirin eugenol ester in rat tail thrombosis model. J Proteomics 2019; 215:103631. [PMID: 31891783 DOI: 10.1016/j.jprot.2019.103631] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/10/2019] [Accepted: 12/27/2019] [Indexed: 01/09/2023]
Abstract
Aspirin eugenol eater (AEE), a new drug compound, was synthesized through the combination of aspirin and eugenol. Antithrombotic effects of AEE have been confirmed in carrageenan-induced rat tail thrombosis model. However, its mechanism is unclear. With the application of integrated approach combining proteomics and metabolomics, the profilings of protein and metabolite in plasma were examined in thrombosis rat pretreated with AEE, aspirin and eugenol, respectively. A clear separation of the plasma metabolic profiles from different groups was found in score plots. 15 metabolites related with the metabolism of fatty acid, energy and amino acid were found. A total of 144, 38, 41 and 54 differentially abundant proteins (DAPs) were identified in control, AEE, aspirin and eugenol group, respectively. Proteomic results showed that aspirin modulated 7 proteins in amino acid metabolism and 4 proteins in complement system; eugenol regulated the 8 proteins related with coagulation cascades and fibrinogen; AEE improved 3 proteins in TCA cycle and 3 in lipid metabolism. Integrated analysis suggested that AEE improved fatty acid, energy and lipid metabolism to against thrombosis. Results of this study indicated AEE had different action mechanism on thrombosis from aspirin and eugenol, and contribute to understanding the mechanisms of AEE on thrombosis. SIGNIFICANCE: Thrombosis is a threat to human health, and there is an urgent need for new drug. In this study, compared with the model group, plasma metabolic profiles in AEE-treated rats were clearly separated; 15 metabolites and 38 proteins were picked out. These metabolites and proteins may assist in understanding the action mechanism of AEE on thrombosis. The results of plasma metabonomics and proteomics also revealed the different action mechanism among AEE, aspirin and eugenol on thrombosis. This study established the foundation to further evaluate the druggability of AEE on thrombosis treatment.
Collapse
Affiliation(s)
- Ning Ma
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China; College of Veterinary Medicine, Agricultural University of Hebei, Baoding, Hebei 071000, PR China
| | - Yajun Yang
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China
| | - Xiwang Liu
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China
| | - Shihong Li
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China
| | - Zhe Qin
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China
| | - Jianyong Li
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development, Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Science of Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China.
| |
Collapse
|
40
|
Trostchansky A, Moore-Carrasco R, Fuentes E. Oxidative pathways of arachidonic acid as targets for regulation of platelet activation. Prostaglandins Other Lipid Mediat 2019; 145:106382. [PMID: 31634570 DOI: 10.1016/j.prostaglandins.2019.106382] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 08/12/2019] [Accepted: 09/03/2019] [Indexed: 12/17/2022]
Abstract
Platelet activation plays an important role in acute and chronic cardiovascular disease states. Multiple pathways contribute to platelet activation including those dependent upon arachidonic acid. Arachidonic acid is released from the platelet membrane by phospholipase A2 action and is then metabolized in the cytosol by specific arachidonic acid oxidation enzymes including prostaglandin H synthase, 12-lipoxygenase, and cytochrome P450 to produce pro- and anti-inflammatory eicosanoids. This review aims to analyze the role of arachidonic acid oxidation on platelet activation, the enzymes that use it as a substrate associated as novel therapeutics target for antiplatelet drugs.
Collapse
Affiliation(s)
- Andres Trostchansky
- Departamento de Bioquimica and Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la República, Montevideo, Uruguay.
| | - Rodrigo Moore-Carrasco
- Departamento de Bioquímica Clínica e Inmunohematología, Facultad de Ciencias de la Salud, Programa de Investigación Asociativa en Cáncer Gástrico (PIA-CG), Universidad de Talca, Chile
| | - Eduardo Fuentes
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Center on Aging, Universidad de Talca, Talca, Chile.
| |
Collapse
|
41
|
Lee SH, Lee S, Du J, Jain K, Ding M, Kadado AJ, Atteya G, Jaji Z, Tyagi T, Kim W, Herzog RI, Patel A, Ionescu CN, Martin KA, Hwa J. Mitochondrial MsrB2 serves as a switch and transducer for mitophagy. EMBO Mol Med 2019; 11:e10409. [PMID: 31282614 PMCID: PMC6685081 DOI: 10.15252/emmm.201910409] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 06/07/2019] [Accepted: 06/13/2019] [Indexed: 01/01/2023] Open
Abstract
Mitophagy can selectively remove damaged toxic mitochondria, protecting a cell from apoptosis. The molecular spatial-temporal mechanisms governing autophagosomal selection of reactive oxygen species (ROS)-damaged mitochondria, particularly in a platelet (no genomic DNA for transcriptional regulation), remain unclear. We now report that the mitochondrial matrix protein MsrB2 plays an important role in switching on mitophagy by reducing Parkin methionine oxidation (MetO), and transducing mitophagy through ubiquitination by Parkin and interacting with LC3. This biochemical signaling only occurs at damaged mitochondria where MsrB2 is released from the mitochondrial matrix. MsrB2 platelet-specific knockout and in vivo peptide inhibition of the MsrB2/LC3 interaction lead to reduced mitophagy and increased platelet apoptosis. Pathophysiological importance is highlighted in human subjects, where increased MsrB2 expression in diabetes mellitus leads to increased platelet mitophagy, and in platelets from Parkinson's disease patients, where reduced MsrB2 expression is associated with reduced mitophagy. Moreover, Parkin mutations at Met192 are associated with Parkinson's disease, highlighting the structural sensitivity at the Met192 position. Release of the enzyme MsrB2 from damaged mitochondria, initiating autophagosome formation, represents a novel regulatory mechanism for oxidative stress-induced mitophagy.
Collapse
Affiliation(s)
- Seung Hee Lee
- Yale Cardiovascular Research CenterSection of Cardiovascular MedicineDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
- Division of Cardiovascular DiseasesCenter for Biomedical SciencesNational Institute of HealthCheongjuChungbukKorea
| | - Suho Lee
- Departments of Neurology and NeurobiologyCellular Neuroscience, Neurodegeneration and Repair ProgramYale University School of MedicineNew HavenCTUSA
| | - Jing Du
- Yale Cardiovascular Research CenterSection of Cardiovascular MedicineDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
| | - Kanika Jain
- Yale Cardiovascular Research CenterSection of Cardiovascular MedicineDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
| | - Min Ding
- Yale Cardiovascular Research CenterSection of Cardiovascular MedicineDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
| | - Anis J Kadado
- Yale Cardiovascular Research CenterSection of Cardiovascular MedicineDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
| | - Gourg Atteya
- Yale Cardiovascular Research CenterSection of Cardiovascular MedicineDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
| | - Zainab Jaji
- Yale Cardiovascular Research CenterSection of Cardiovascular MedicineDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
| | - Tarun Tyagi
- Yale Cardiovascular Research CenterSection of Cardiovascular MedicineDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
| | - Won‐ho Kim
- Division of Cardiovascular DiseasesCenter for Biomedical SciencesNational Institute of HealthCheongjuChungbukKorea
| | - Raimund I Herzog
- Section of EndocrinologyDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
| | - Amar Patel
- Division of Movement DisordersDepartments of Neurology and NeurobiologyYale University School of MedicineNew HavenCTUSA
| | - Costin N Ionescu
- Yale Cardiovascular MedicineDepartment of Internal MedicineYale‐New Haven HospitalNew HavenCTUSA
| | - Kathleen A Martin
- Yale Cardiovascular Research CenterSection of Cardiovascular MedicineDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
| | - John Hwa
- Yale Cardiovascular Research CenterSection of Cardiovascular MedicineDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
| |
Collapse
|
42
|
Touyz RM, Alves-Lopes R, Rios FJ, Camargo LL, Anagnostopoulou A, Arner A, Montezano AC. Vascular smooth muscle contraction in hypertension. Cardiovasc Res 2019; 114:529-539. [PMID: 29394331 PMCID: PMC5852517 DOI: 10.1093/cvr/cvy023] [Citation(s) in RCA: 391] [Impact Index Per Article: 65.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 01/30/2018] [Indexed: 12/19/2022] Open
Abstract
Hypertension is a major risk factor for many common chronic diseases, such as heart failure, myocardial infarction, stroke, vascular dementia, and chronic kidney disease. Pathophysiological mechanisms contributing to the development of hypertension include increased vascular resistance, determined in large part by reduced vascular diameter due to increased vascular contraction and arterial remodelling. These processes are regulated by complex-interacting systems such as the renin-angiotensin-aldosterone system, sympathetic nervous system, immune activation, and oxidative stress, which influence vascular smooth muscle function. Vascular smooth muscle cells are highly plastic and in pathological conditions undergo phenotypic changes from a contractile to a proliferative state. Vascular smooth muscle contraction is triggered by an increase in intracellular free calcium concentration ([Ca2+]i), promoting actin–myosin cross-bridge formation. Growing evidence indicates that contraction is also regulated by calcium-independent mechanisms involving RhoA-Rho kinase, protein Kinase C and mitogen-activated protein kinase signalling, reactive oxygen species, and reorganization of the actin cytoskeleton. Activation of immune/inflammatory pathways and non-coding RNAs are also emerging as important regulators of vascular function. Vascular smooth muscle cell [Ca2+]i not only determines the contractile state but also influences activity of many calcium-dependent transcription factors and proteins thereby impacting the cellular phenotype and function. Perturbations in vascular smooth muscle cell signalling and altered function influence vascular reactivity and tone, important determinants of vascular resistance and blood pressure. Here, we discuss mechanisms regulating vascular reactivity and contraction in physiological and pathophysiological conditions and highlight some new advances in the field, focusing specifically on hypertension.
Collapse
Affiliation(s)
- Rhian M Touyz
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Rheure Alves-Lopes
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Francisco J Rios
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Livia L Camargo
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Aikaterini Anagnostopoulou
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Anders Arner
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Augusto C Montezano
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| |
Collapse
|
43
|
Chen L, He FJ, Dong Y, Huang Y, Harshfield GA, Zhu H. Sodium Reduction, Metabolomic Profiling, and Cardiovascular Disease Risk in Untreated Black Hypertensives. Hypertension 2019; 74:194-200. [PMID: 31079530 PMCID: PMC9116731 DOI: 10.1161/hypertensionaha.119.12880] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Dietary sodium restriction has multiple beneficial effects on cardiovascular health. The underlying mechanisms are not fully understood, and the roles of metabolomics have been rarely studied. We aimed to test the hypothesis that the reduction in dietary sodium intake would induce changes in metabolomic profiling among black hypertensives, and the changes would be associated with reduced blood pressure (BP) and improved skin capillary density. A total of 64 untreated black hypertensives were included from a randomized, double-blind, placebo-controlled cross-over trial of sodium reduction. The participants were given either 9 slow sodium tablets (10 mmol sodium per tablet) or placebo tablets daily for 6 weeks, they then crossed over to receive the other tablets for another 6 weeks, while on reduced sodium diet aiming at achieving daily sodium intake around 2.0 g. Untargeted metabolomic profiling was performed in paired serum samples, which were collected at the end of each period, so were BP and capillary density. Mixed-effects models were used. There were 34 metabolites identified with raw P's<0.05. Among those, 2 metabolites including β-hydroxyisovalerate and methionine sulfone were significantly increased with sodium reduction (false discovery rate =0.006 and 0.099, respectively). Increased β-hydroxyisovalerate was associated with reduced office systolic BP and ambulatory daytime systolic BP, whereas increased methionine sulfone was associated with reduced 24-hour diastolic BP, ambulatory nighttime diastolic BP, and increased skin capillary density. Our results suggest that dietary sodium reduction increases the circulating levels of β-hydroxyisovalerate and methionine sulfone. Further studies are warranted. Clinical Trial Registration- URL: http://www.clinicaltrials.gov . Unique identifier: NCT00152074.
Collapse
Affiliation(s)
- Li Chen
- From the Georgia Prevention Institute, Department of Population Health Sciences, Medical College of Georgia, Augusta University (L.C., Y.D., Y.H., G.A.H., H.Z.)
| | - Feng J He
- Center for Environmental and Preventive Medicine, Wolfson Institute of Preventive Medicine, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (F.J.H.)
| | - Yanbin Dong
- From the Georgia Prevention Institute, Department of Population Health Sciences, Medical College of Georgia, Augusta University (L.C., Y.D., Y.H., G.A.H., H.Z.)
| | - Ying Huang
- From the Georgia Prevention Institute, Department of Population Health Sciences, Medical College of Georgia, Augusta University (L.C., Y.D., Y.H., G.A.H., H.Z.)
| | - Gregory A Harshfield
- From the Georgia Prevention Institute, Department of Population Health Sciences, Medical College of Georgia, Augusta University (L.C., Y.D., Y.H., G.A.H., H.Z.)
| | - Haidong Zhu
- From the Georgia Prevention Institute, Department of Population Health Sciences, Medical College of Georgia, Augusta University (L.C., Y.D., Y.H., G.A.H., H.Z.)
| |
Collapse
|
44
|
Sartore G, Chilelli NC, Seraglia R, Ragazzi E, Marin R, Roverso M, Cosma C, Vaccaro O, Burlina S, Lapolla A. Long-term effect of pioglitazone vs glimepiride on lipoprotein oxidation in patients with type 2 diabetes: a prospective randomized study. Acta Diabetol 2019; 56:505-513. [PMID: 30740640 DOI: 10.1007/s00592-018-01278-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 12/14/2018] [Indexed: 01/07/2023]
Abstract
AIMS Type 2 diabetes (DM2) is associated to oxidative modifications of high-density lipoproteins (HDL), which can interfere with their function. Pioglitazone has proved effective in raising HDL cholesterol (HDL-C) and lowering small dense low-density lipoprotein (LDL), but no clinical studies have examined its effect on lipoprotein oxidation in patients with DM2. METHODS We assessed the effect of pioglitazone vs glimepiride after 1 year on HDL oxidation, expressed as relative abundance of peptides containing Met112O in ApoA-I (oxApoA-I) estimated by mass spectrometry (MALDI/TOF/TOF), in 95 patients with DM2. The oxLDL and AGE were quantified by ELISA. RESULTS Patients receiving pioglitazone showed a significant increase in the concentration of ApoA-I (Δ = 7.2 ± 14.8 mg/dL, p < 0.02) and a reduction in oxApoA-I (Δ = - 1.0 ± 2.6%, p < 0.02); this reduction was not significantly different from glimepiride. oxLDL showed a slight, but not significant increase in both treatment groups. Regression analysis showed a correlation between ΔoxApoA-I and ΔAGE (r = 0.30; p = 0.007) in all patients, while both of these parameters were unrelated to changes in HbA1c, HDL-C, duration of illness, or use of statins. CONCLUSIONS Long-term treatment with pioglitazone was effective in reducing the oxidation of HDL, but not LDL in patients with DM2, while glimepiride didn't. This finding seems to be associated to the change of glyco-oxidation status, not to any improvement in glycemic control or lipid profile. TRIAL REGISTRATION NCT00700856, ClinicalTrials.gov Registered June 18, 2008.
Collapse
Affiliation(s)
- Giovanni Sartore
- Diabetology and Dietetics, Department of Medicine (DIMED), University of Padova, via Giustiniani, 2, 35100, Padua, Italy
| | - Nino Cristiano Chilelli
- Diabetology and Dietetics, Department of Medicine (DIMED), University of Padova, via Giustiniani, 2, 35100, Padua, Italy.
| | - Roberta Seraglia
- National Research Council-Institute for Energy and Interphases, Padua, Italy
| | - Eugenio Ragazzi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padua, Italy
| | - Raffaella Marin
- Lipid Laboratory, Department of Medicine (DIMED), University of Padova, Padua, Italy
| | - Marco Roverso
- National Research Council-Institute for Energy and Interphases, Padua, Italy
| | - Chiara Cosma
- Department of Laboratory Medicine, University of Padova, Padua, Italy
| | - Olga Vaccaro
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Silvia Burlina
- Diabetology and Dietetics, Department of Medicine (DIMED), University of Padova, via Giustiniani, 2, 35100, Padua, Italy
| | - Annunziata Lapolla
- Diabetology and Dietetics, Department of Medicine (DIMED), University of Padova, via Giustiniani, 2, 35100, Padua, Italy
| |
Collapse
|
45
|
Soares MSP, da Silveira de Mattos B, Ávila AA, Spohr L, Pedra NS, Teixeira FC, Bona NP, Oliveira PS, Stefanello FM, Spanevello RM. High levels of methionine and methionine sulfoxide: Impact on adenine nucleotide hydrolysis and redox status in platelets and serum of young rats. J Cell Biochem 2019; 120:2289-2303. [PMID: 30216509 DOI: 10.1002/jcb.27554] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 08/02/2018] [Indexed: 01/24/2023]
Abstract
We investigated acute and chronic effects administration of methionine (Met) and/or methionine sulfoxide (MetO) on ectonucleotidases and oxidative stress in platelets and serum of young rats. Wistar rats were divided into four groups: control, Met, MetO, and Met + MetO. In acute treatment, the animals received a single subcutaneous injection of amino acid(s) and were euthanized after 1 and 3 hours. In chronic protocol, Met and/or MetO were administered twice a day with an 8-hour interval from the 6th to the 28th day of life. Nucleoside triphosphate phosphohydrolase and 5'-nucleotidase activities were reduced in platelets and serum by Met, MetO, and Met + MetO after 3 hours and 21 days. Adenosine deaminase activity reduced in platelets at 3 hours after MetO and Met + MetO administration and increased after 21 days in animals treated with Met + MetO. Superoxide dismutase and catalase activities decreased in platelets in MetO and Met + MetO groups after 3 hours, while reactive oxygen species (ROS) levels increased in same groups. Catalase activity in platelets decreased in all experimental groups after chronic treatment. Met, MetO, and Met + MetO administration increased plasmatic ROS levels in acute and chronic protocols; glutathione S-transferase activity increased by MetO and Met + MetO administration at 3 hours, and ascorbic acid decreased in all experimental groups in acute and chronic protocols. Thiobarbituric acid reactive substances increased, superoxide dismutase and catalase activities reduced in the Met and/or MetO groups at 3 hours and in chronic treatment. Our data demonstrated that Met and/or MetO induced changes in adenine nucleotide hydrolysis and redox status of platelets and serum, which can be associated with platelet dysfunction in hypermethioninemia.
Collapse
Affiliation(s)
- Mayara Sandrielly Pereira Soares
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Bruna da Silveira de Mattos
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Anita Almeida Ávila
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Luiza Spohr
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Nathalia Stark Pedra
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Fernanda Cardoso Teixeira
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Natália Pontes Bona
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Pathise Souto Oliveira
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Francieli Moro Stefanello
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Roselia Maria Spanevello
- Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| |
Collapse
|
46
|
D'Aronco S, Crotti S, Agostini M, Traldi P, Chilelli NC, Lapolla A. The role of mass spectrometry in studies of glycation processes and diabetes management. MASS SPECTROMETRY REVIEWS 2019; 38:112-146. [PMID: 30423209 DOI: 10.1002/mas.21576] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 07/03/2018] [Indexed: 06/09/2023]
Abstract
In the last decade, mass spectrometry has been widely employed in the study of diabetes. This was mainly due to the development of new, highly sensitive, and specific methods representing powerful tools to go deep into the biochemical and pathogenetic processes typical of the disease. The aim of this review is to give a panorama of the scientifically valid results obtained in this contest. The recent studies on glycation processes, in particular those devoted to the mechanism of production and to the reactivity of advanced glycation end products (AGEs, AGE peptides, glyoxal, methylglyoxal, dicarbonyl compounds) allowed to obtain a different view on short and long term complications of diabetes. These results have been employed in the research of effective markers and mass spectrometry represented a precious tool allowing the monitoring of diabetic nephropathy, cardiovascular complications, and gestational diabetes. The same approaches have been employed to monitor the non-insulinic diabetes pharmacological treatments, as well as in the discovery and characterization of antidiabetic agents from natural products. © 2018 Wiley Periodicals, Inc. Mass Spec Rev 38:112-146, 2019.
Collapse
Affiliation(s)
- Sara D'Aronco
- Department of Surgical, Oncological and Gastroenterological Sciences, University of Padova, Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Sara Crotti
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Marco Agostini
- Department of Surgical, Oncological and Gastroenterological Sciences, University of Padova, Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Pietro Traldi
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | | | | |
Collapse
|
47
|
Pennington SM, Klutho PR, Xie L, Broadhurst K, Koval OM, McCormick ML, Spitz DR, Grumbach IM. Defective protein repair under methionine sulfoxide A deletion drives autophagy and ARE-dependent gene transcription. Redox Biol 2018; 16:401-413. [PMID: 29649787 PMCID: PMC5953240 DOI: 10.1016/j.redox.2018.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 03/21/2018] [Accepted: 04/01/2018] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVE Reduction of oxidized methionines is emerging as a major protein repair pathway. The lack of methionine sulfoxide reductase A (MsrA) exacerbates cardiovascular disease phenotypes driven by increased oxidative stress. However, the role of MsrA on maintaining cellular homeostasis in the absence of excessive oxidative stress is less well understood. METHODS AND RESULTS Constitutive genetic deletion of MsrA increased formation of p62-containing protein aggregates, activated autophagy, and decreased a marker of apoptosis in vascular smooth muscle cells (VSMC). The association of Keap1 with p62 was augmented in MsrA-/- VSMC. Keap1 targets the transcription factor Nrf2, which regulates antioxidant genes, for proteasomal degradation. However, in MsrA-/- VSMC, the association of Nrf2 with Keap1 was diminished. Whereas Nrf2 mRNA levels were not decreased in MsrA-/- VSMC, we detected decreased ubiquitination of Nrf2 and a corresponding increase in total Nrf2 protein in the absence of biochemical markers of oxidative stress. Moreover, nuclear-localized Nrf2 was increased under MsrA deficiency, resulting in upregulation of Nrf2-dependent transcriptional activity. Consequently, transcription, protein levels and enzymatic activity of glutamate-cysteine ligase and glutathione reductase were greatly augmented in MsrA-/- VSMC. SUMMARY Our findings demonstrate that reversal of methionine oxidation is required for maintenance of cellular homeostasis in the absence of increased oxidative stress. These data provide the first link between autophagy and activation of Nrf2 in the setting of MsrA deletion.
Collapse
Affiliation(s)
- Steven M Pennington
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Paula R Klutho
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Litao Xie
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Kim Broadhurst
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Olha M Koval
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Michael L McCormick
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA
| | - Douglas R Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA
| | - Isabella M Grumbach
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA; Veterans Affairs Healthcare System, Iowa City, IA 52246, USA.
| |
Collapse
|
48
|
Rosenfeld MA, Vasilyeva AD, Yurina LV, Bychkova AV. Oxidation of proteins: is it a programmed process? Free Radic Res 2017; 52:14-38. [DOI: 10.1080/10715762.2017.1402305] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Mark A. Rosenfeld
- N. M. Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, Russia
| | - Alexandra D. Vasilyeva
- N. M. Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, Russia
| | - Lyubov V. Yurina
- N. M. Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, Russia
| | - Anna V. Bychkova
- N. M. Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
49
|
Abstract
Protein function can be regulated via post-translational modifications by numerous enzymatic and non-enzymatic mechanisms, including oxidation of cysteine and methionine residues. Redox-dependent regulatory mechanisms have been identified for nearly every cellular process, but the major paradigm has been that cellular components are oxidized (damaged) by reactive oxygen species (ROS) in a relatively unspecific way, and then reduced (repaired) by designated reductases. While this scheme may work with cysteine, it cannot be ascribed to other residues, such as methionine, whose reaction with ROS is too slow to be biologically relevant. However, methionine is clearly oxidized in vivo and enzymes for its stereoselective reduction are present in all three domains of life. Here, we revisit the chemistry and biology of methionine oxidation, with emphasis on its generation by enzymes from the monooxygenase family. Particular attention is placed on MICALs, a recently discovered family of proteins that harbor an unusual flavin-monooxygenase domain with an NADPH-dependent methionine sulfoxidase activity. Based on structural and kinetic information we provide a rational framework to explain MICAL mechanism, inhibition, and regulation. Methionine residues that are targeted by MICALs are reduced back by methionine sulfoxide reductases, suggesting that reversible methionine oxidation may be a general mechanism analogous to the regulation by phosphorylation by kinases/phosphatases. The identification of new enzymes that catalyze the oxidation of methionine will open a new area of research at the forefront of redox signaling.
Collapse
Affiliation(s)
- Bruno Manta
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Vadim N Gladyshev
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
50
|
Whole-exome sequencing in evaluation of patients with venous thromboembolism. Blood Adv 2017; 1:1224-1237. [PMID: 29296762 DOI: 10.1182/bloodadvances.2017005249] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 05/26/2017] [Indexed: 01/05/2023] Open
Abstract
Genetics play a significant role in venous thromboembolism (VTE), yet current clinical laboratory-based testing identifies a known heritable thrombophilia (factor V Leiden, prothrombin gene mutation G20210A, or a deficiency of protein C, protein S, or antithrombin) in only a minority of VTE patients. We hypothesized that a substantial number of VTE patients could have lesser-known thrombophilia mutations. To test this hypothesis, we performed whole-exome sequencing (WES) in 64 patients with VTE, focusing our analysis on a novel 55-gene extended thrombophilia panel that we compiled. Our extended thrombophilia panel identified a probable disease-causing genetic variant or variant of unknown significance in 39 of 64 study patients (60.9%), compared with 6 of 237 control patients without VTE (2.5%) (P < .0001). Clinical laboratory-based thrombophilia testing identified a heritable thrombophilia in only 14 of 54 study patients (25.9%). The majority of WES variants were either associated with thrombosis based on prior reports in the literature or predicted to affect protein structure based on protein modeling performed as part of this study. Variants were found in major thrombophilia genes, various SERPIN genes, and highly conserved areas of other genes with established or potential roles in coagulation or fibrinolysis. Ten patients (15.6%) had >1 variant. Sanger sequencing performed in family members of 4 study patients with and without VTE showed generally concordant results with thrombotic history. WES and extended thrombophilia testing are promising tools for improving our understanding of VTE pathogenesis and identifying inherited thrombophilias.
Collapse
|