1
|
Jourd'heuil F, Mathai C, Cat Pham LG, Gilliard K, Balnis J, Overmyer KA, Coon JJ, Jaitovich A, Boivin B, Jourd'heuil D. Cytoglobin scavenges intracellular hydrogen peroxide and regulates redox signals in the vasculature. Redox Biol 2025; 83:103633. [PMID: 40252320 PMCID: PMC12023880 DOI: 10.1016/j.redox.2025.103633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/01/2025] [Accepted: 04/08/2025] [Indexed: 04/21/2025] Open
Abstract
The oxidant hydrogen peroxide (H2O2) serves as a signaling molecule that alters many aspects of cardiovascular functions and contributes to cardiovascular diseases. Recent studies suggest that cytoglobin - a member of the globin family - may promote electron transfer reactions with proposed functions in H2O2 decomposition. In the present study, we directly examined the ability of cytoglobin to decompose H2O2. Carotid arteries from cytoglobin knockout mice were more sensitive to glycolytic inhibition by H2O2 than arteries from wild type mice. In addition, the ectopic expression of cytoglobin in cultured cells limited the inhibitory effect of H2O2 on glycolysis and reversed the oxidative inactivation of the glycolytic enzyme GAPDH. Cytoglobin facilitated the reduction of the thiol-based H2O2 sensor Hyper7 after H2O2 challenge. The specific substitution of one of two cysteine residues on cytoglobin (C83) inhibited its antioxidant activity, as did the substitutions at the proximal and distal histidine residues. In vitro, direct measurements of H2O2 concentrations indicated that purified cytoglobin consumes H2O2 at rates comparable to that of peroxiredoxin 2 and that it competitively inhibits the hyperoxidation of peroxiredoxin 2. We propose that cytoglobin may serve as a regulator of intracellular redox signals initiated by H2O2.
Collapse
Affiliation(s)
- Frances Jourd'heuil
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Clinton Mathai
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Le Gia Cat Pham
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Kurrim Gilliard
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Joseph Balnis
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA; Division of Pulmonary and Critical Care Medicine, Albany Medical College, Albany, NY, USA
| | - Katherine A Overmyer
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, USA; Morgridge Institute for Research, Madison, WI, USA
| | - Joshua J Coon
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, USA; Morgridge Institute for Research, Madison, WI, USA; Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Ariel Jaitovich
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA; Division of Pulmonary and Critical Care Medicine, Albany Medical College, Albany, NY, USA
| | - Benoit Boivin
- Department of Nanoscale Science & Engineering, University at Albany, Albany, NY, USA
| | - David Jourd'heuil
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
2
|
Zhao Y, Li Z, Ma H, Pan Z, Cai B, Zhang C, Jiao J. METTL3-Mediated m 6A mRNA Modification Facilitates Neointimal Hyperplasia in Arteriovenous Fistula. Arterioscler Thromb Vasc Biol 2025. [PMID: 40401375 DOI: 10.1161/atvbaha.124.321014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/02/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Arteriovenous fistula (AVF) is the preferred vascular access for hemodialysis in patients with end-stage renal disease, yet its long-term patency is threatened by neointimal hyperplasia (NIH). N6-methyladenosine, a prevalent RNA modification catalyzed by METTL3 (methyltransferase-like 3), plays a regulatory role in cardiovascular remodeling. Our previous studies found that N6-methyladenosine methyltransferase METTL3 mediated cardiomyocyte proliferation and heart repair after myocardial ischemia. However, its impact on AVF-related NIH remains unclear. METHODS We examined m6A levels and METTL3 expression in human and murine AVF tissues. Using smooth muscle cell-specific METTL3 conditional knockout and METTL3-overexpressing (adeno-associated virus-METTL3) mouse models, we evaluated NIH formation. In vitro, we analyzed vascular smooth muscle cell proliferation, migration, phenotypic switching, and ferroptosis. m6A epitranscriptomic microarray and RNA stability assays were used to explore downstream targets and mechanisms. RESULTS METTL3 was significantly upregulated in AVF tissues and vascular smooth muscle cells undergoing dedifferentiation. METTL3 deletion attenuated, while overexpression exacerbated, NIH in vivo. METTL3 enhanced vascular smooth muscle cell proliferation, migration, and phenotypic switching, while suppressing ferroptosis. Mechanistically, METTL3 increased m6A modification of SLC7A11 (solute carrier family 7 member 11) mRNA, stabilized its transcript, and promoted translation via recruitment of the m6A reader YTHDF1 (YTH N6-methyladenosine RNA-binding protein 1). Silencing SLC7A11 or YTHDF1 abrogated METTL3-induced phenotypic changes and ferroptosis resistance. CONCLUSIONS The METTL3-YTHDF1-SLC7A11 axis facilitates AVF NIH by regulating vascular smooth muscle cell dedifferentiation and ferroptosis. These findings uncover a novel epitranscriptional mechanism and suggest a potential therapeutic target for AVF stenosis.
Collapse
Affiliation(s)
- Yao Zhao
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, China (Y.Z., Z.L., H.M., C.Z., J.J.)
| | - Zhaozheng Li
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, China (Y.Z., Z.L., H.M., C.Z., J.J.)
| | - Huimin Ma
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, China (Y.Z., Z.L., H.M., C.Z., J.J.)
| | - Zhenwei Pan
- Harbin Medical University, China. (Z.P., B.C.)
| | - Benzhi Cai
- Department of Pharmacy at The Second Affiliated Hospital, and Department of Pharmacology (The Key Laboratory of Cardiovascular Medicine Research, Ministry of Education) at College of Pharmacy, Department of Clinical Pharmacology (the Heilongjiang Key Laboratory of Drug Research) (B.C.)
- Harbin Medical University, China. (Z.P., B.C.)
| | - Chengwei Zhang
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, China (Y.Z., Z.L., H.M., C.Z., J.J.)
| | - Jundong Jiao
- Department of Nephrology, The Second Affiliated Hospital of Harbin Medical University, China (Y.Z., Z.L., H.M., C.Z., J.J.)
- Institute of Nephrology, Harbin Medical University, China (J.J.)
| |
Collapse
|
3
|
Mahgoup EM, Khaleel SA, El-Mahdy MA, Zweier JL. Electronic cigarette vape decreases nitric oxide bioavailability in vascular smooth muscle cells via increased cytoglobin-mediated metabolism. Free Radic Biol Med 2025; 228:339-349. [PMID: 39743029 PMCID: PMC11788057 DOI: 10.1016/j.freeradbiomed.2024.12.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/16/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
Cytoglobin (Cygb) regulates vascular tone by modulating nitric oxide (NO) metabolism in vascular smooth muscle cells (VSMCs). In the presence of its cytochrome B5a (B5)/B5 reductase-isoform-3 (B5R) reducing system, Cygb controls NO metabolism via oxygen-dependent NO dioxygenation. Electronic cigarette (EC) use has been shown to induce vascular dysfunction and decrease NO bioavailability; however, the role of Cygb-mediated NO metabolism in the pathophysiology of this process has not been previously investigated. Therefore, we utilized aortic VSMCs with EC vape extract (ECE) exposure to elucidate the effects of EC vape constituents on NO degradation and alterations in the process of Cygb-mediated NO metabolism. VSMCs were exposed to ECE, either nicotine-free (ECEV) or nicotine-containing (ECEN), for various durations. NO decay rates were measured along with cellular expression of Cygb and its B5/B5R reducing system. Exposure to ECEV led to a much higher rate of NO consumption by VSMCs, with an even larger effect following ECEN exposure. With 4 h of exposure, a modest increase in NO decay rate occurred that was followed by much higher increases with exposure times of 24-48 h. This effect was paralleled by upregulation of Cygb and B5/B5R expression. siRNA-mediated knock-down of Cygb expression largely reversed this ECE-induced increase in NO metabolism rate. Thus, ECE exposure led to increased Cygb-mediated NO metabolism in VSMCs with diminished NO bioavailability, which in turn can play a key role in EC-induced vascular dysfunction.
Collapse
Affiliation(s)
- Elsayed M Mahgoup
- Department of Internal Medicine, Division of Cardiovascular Medicine, and the EPR Center, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA; Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Sahar A Khaleel
- Department of Internal Medicine, Division of Cardiovascular Medicine, and the EPR Center, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA; Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Mohamed A El-Mahdy
- Department of Internal Medicine, Division of Cardiovascular Medicine, and the EPR Center, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Jay L Zweier
- Department of Internal Medicine, Division of Cardiovascular Medicine, and the EPR Center, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
4
|
Porto E, Loula P, Strand S, Hankeln T. Molecular analysis of the human cytoglobin mRNA isoforms. J Inorg Biochem 2024; 251:112422. [PMID: 38016326 DOI: 10.1016/j.jinorgbio.2023.112422] [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: 04/28/2023] [Revised: 09/26/2023] [Accepted: 10/29/2023] [Indexed: 11/30/2023]
Abstract
Multiple functions have been proposed for the ubiquitously expressed vertebrate globin cytoglobin (Cygb), including nitric oxide (NO) metabolism, lipid peroxidation/signalling, superoxide dismutase activity, reactive oxygen/nitrogen species (RONS) scavenging, regulation of blood pressure, antifibrosis, and both tumour suppressor and oncogenic effects. Since alternative splicing can expand the biological roles of a gene, we investigated whether this mechanism contributes to the functional diversity of Cygb. By mining of cDNA data and molecular analysis, we identified five alternative mRNA isoforms for the human CYGB gene (V-1 to V-5). Comprehensive RNA-seq analyses of public datasets from human tissues and cells confirmed that the canonical CYGB V-1 isoform is the primary CYGB transcript in the majority of analysed datasets. Interestingly, we revealed that isoform V-3 represented the predominant CYGB variant in hepatoblastoma (HB) cell lines and in the majority of analysed normal and HB liver tissues. CYGB V-3 mRNA is transcribed from an alternate upstream promoter and hypothetically encodes a N-terminally truncated CYGB protein, which is not recognized by some antibodies used in published studies. Little to no transcriptional evidence was found for the other CYGB isoforms. Comparative transcriptomics and flow cytometry on CYGB+/+ and gene-edited CYGB-/- HepG2 HB cells did not unveil a knockout phenotype and, thus, a potential function for CYGB V-3. Our study reveals that the CYGB gene is transcriptionally more complex than previously described as it expresses alternative mRNA isoforms of unknown function. Additional experimental data are needed to clarify the biological meaning of those alternative CYGB transcripts.
Collapse
Affiliation(s)
- Elena Porto
- Institute of Organismic and Molecular Evolution, Molecular Genetics & Genome Analysis Group, Johannes Gutenberg University Mainz, J. J. Becher-Weg 30A, D-55128 Mainz, Germany
| | - Paraskevi Loula
- Institute of Organismic and Molecular Evolution, Molecular Genetics & Genome Analysis Group, Johannes Gutenberg University Mainz, J. J. Becher-Weg 30A, D-55128 Mainz, Germany
| | - Susanne Strand
- Department of Internal Medicine I, Molecular Hepatology, University Medical Center, Johannes Gutenberg University Mainz, Obere Zahlbacher Strasse 63, 55131 Mainz, Germany
| | - Thomas Hankeln
- Institute of Organismic and Molecular Evolution, Molecular Genetics & Genome Analysis Group, Johannes Gutenberg University Mainz, J. J. Becher-Weg 30A, D-55128 Mainz, Germany.
| |
Collapse
|
5
|
Porto E, De Backer J, Thuy LTT, Kawada N, Hankeln T. Transcriptomics of a cytoglobin knockout mouse: Insights from hepatic stellate cells and brain. J Inorg Biochem 2024; 250:112405. [PMID: 37977965 DOI: 10.1016/j.jinorgbio.2023.112405] [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: 06/12/2023] [Revised: 10/10/2023] [Accepted: 10/16/2023] [Indexed: 11/19/2023]
Abstract
The vertebrate respiratory protein cytoglobin (Cygb) is thought to exert multiple cellular functions. Here we studied the phenotypic effects of a Cygb knockout (KO) in mouse on the transcriptome level. RNA sequencing (RNA-Seq) was performed for the first time on sites of major endogenous Cygb expression, i.e. quiescent and activated hepatic stellate cells (HSCs) and two brain regions, hippocampus and hypothalamus. The data recapitulated the up-regulation of Cygb during HSC activation and its expression in the brain. Differential gene expression analyses suggested a role of Cygb in the response to inflammation in HSCs and its involvement in retinoid metabolism, retinoid X receptor (RXR) activation-induced xenobiotics metabolism, and RXR activation-induced lipid metabolism and signaling in activated cells. Unexpectedly, only minor effects of the Cygb KO were detected in the transcriptional profiles in hippocampus and hypothalamus, precluding any enrichment analyses. Furthermore, the transcriptome data pointed at a previously undescribed potential of the Cygb- knockout allele to produce cis-acting effects, necessitating future verification studies.
Collapse
Affiliation(s)
- Elena Porto
- Institute of Organismic and Molecular Evolution, Molecular Genetics & Genome Analysis Group, Johannes Gutenberg University Mainz, J. J. Becher-Weg 30A, Mainz D-55128, Germany
| | - Joey De Backer
- Research Group PPES, Department of Biomedical Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, Antwerp 1610, Belgium
| | - Le Thi Thanh Thuy
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Japan
| | - Norifumi Kawada
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Japan
| | - Thomas Hankeln
- Institute of Organismic and Molecular Evolution, Molecular Genetics & Genome Analysis Group, Johannes Gutenberg University Mainz, J. J. Becher-Weg 30A, Mainz D-55128, Germany.
| |
Collapse
|
6
|
Mathai C, Jourd'heuil F, Pham LGC, Gilliard K, Balnis J, Jen A, Overmyer KA, Coon JJ, Jaitovich A, Boivin B, Jourd'heuil D. A role for cytoglobin in regulating intracellular hydrogen peroxide and redox signals in the vasculature. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.31.535146. [PMID: 37034694 PMCID: PMC10081330 DOI: 10.1101/2023.03.31.535146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
The oxidant hydrogen peroxide serves as a signaling molecule that alters many aspects of cardiovascular functions. Recent studies suggest that cytoglobin - a hemoglobin expressed in the vasculature - may promote electron transfer reactions with proposed functions in hydrogen peroxide decomposition. Here, we determined the extent to which cytoglobin regulates intracellular hydrogen peroxide and established mechanisms. We found that cytoglobin decreased the hyperoxidation of peroxiredoxins and maintained the activity of peroxiredoxin 2 following challenge with exogenous hydrogen peroxide. Cytoglobin promoted a reduced intracellular environment and facilitated the reduction of the thiol-based hydrogen peroxide sensor Hyper7 after bolus addition of hydrogen peroxide. Cytoglobin also limited the inhibitory effect of hydrogen peroxide on glycolysis and reversed the oxidative inactivation of the glycolytic enzyme GAPDH. Our results indicate that cytoglobin in cells exists primarily as oxyferrous cytoglobin (CygbFe 2+ -O 2 ) with its cysteine residues in the reduced form. We found that the specific substitution of one of two cysteine residues on cytoglobin (C83A) inhibited the reductive activity of cytoglobin on Hyper7 and GAPDH. Carotid arteries from cytoglobin knockout mice were more sensitive to glycolytic inhibition by hydrogen peroxide than arteries from wildtype mice. Together, these results support a role for cytoglobin in regulating intracellular redox signals associated with hydrogen peroxide through oxidation of its cysteine residues, independent of hydrogen peroxide reaction at its heme center.
Collapse
|
7
|
Mathai C, Jourd'heuil F, Pham LGC, Gilliard K, Howard D, Balnis J, Jaitovich A, Chittur SV, Rilley M, Peredo-Wende R, Ammoura I, Shin SJ, Barroso M, Barra J, Shishkova E, Coon JJ, Lopez-Soler RI, Jourd'heuil D. Regulation of DNA damage and transcriptional output in the vasculature through a cytoglobin-HMGB2 axis. Redox Biol 2023; 65:102838. [PMID: 37573836 PMCID: PMC10428073 DOI: 10.1016/j.redox.2023.102838] [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: 06/29/2023] [Revised: 07/24/2023] [Accepted: 08/02/2023] [Indexed: 08/15/2023] Open
Abstract
Identifying novel regulators of vascular smooth muscle cell function is necessary to further understand cardiovascular diseases. We previously identified cytoglobin, a hemoglobin homolog, with myogenic and cytoprotective roles in the vasculature. The specific mechanism of action of cytoglobin is unclear but does not seem to be related to oxygen transport or storage like hemoglobin. Herein, transcriptomic profiling of injured carotid arteries in cytoglobin global knockout mice revealed that cytoglobin deletion accelerated the loss of contractile genes and increased DNA damage. Overall, we show that cytoglobin is actively translocated into the nucleus of vascular smooth muscle cells through a redox signal driven by NOX4. We demonstrate that nuclear cytoglobin heterodimerizes with the non-histone chromatin structural protein HMGB2. Our results are consistent with a previously unknown function by which a non-erythrocytic hemoglobin inhibits DNA damage and regulates gene programs in the vasculature by modulating the genome-wide binding of HMGB2.
Collapse
Affiliation(s)
- Clinton Mathai
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Frances Jourd'heuil
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Le Gia Cat Pham
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Kurrim Gilliard
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Dennis Howard
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Joseph Balnis
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA; Division of Pulmonary and Critical Care Medicine, Albany Medical College, Albany, NY, USA
| | - Ariel Jaitovich
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA; Division of Pulmonary and Critical Care Medicine, Albany Medical College, Albany, NY, USA
| | - Sridar V Chittur
- Center for Functional Genomics, Cancer Research Center, University at Albany, New York, 12144, USA
| | - Mark Rilley
- Division of Rheumatology, Department of Medicine, Samuel Stratton VA Medical Center, Albany, NY, 12208, USA
| | - Ruben Peredo-Wende
- Division of Rheumatology, Department of Medicine, Samuel Stratton VA Medical Center, Albany, NY, 12208, USA
| | - Ibrahim Ammoura
- Department of Pathology and Medicine, Albany Medical Center, Albany, NY, 12208, USA
| | - Sandra J Shin
- Department of Pathology and Medicine, Albany Medical Center, Albany, NY, 12208, USA
| | - Margarida Barroso
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Jonathan Barra
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Evgenia Shishkova
- National Center for Quantitative Biology of Complex Systems, Madison, WI, 53706, USA; Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, 53506, USA
| | - Joshua J Coon
- Department of Pathology and Medicine, Albany Medical Center, Albany, NY, 12208, USA; Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, 53506, USA; Morgridge Institute for Research, Madison, WI, 53515, USA; Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53506, USA
| | - Reynold I Lopez-Soler
- Section of Renal Transplantation, Edward Hines VA Jr. Hospital, Hines, IL, 60141, USA; Department of Surgery, Division of Intra-Abdominal Transplantation, Stritch School of Medicine, Maywood, IL, 60153, USA
| | - David Jourd'heuil
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
8
|
Xu S, Chuang CY, Hawkins CL, Hägglund P, Davies MJ. Identification and quantification of protein nitration sites in human coronary artery smooth muscle cells in the absence and presence of peroxynitrous acid/peroxynitrite. Redox Biol 2023; 64:102799. [PMID: 37413764 PMCID: PMC10363479 DOI: 10.1016/j.redox.2023.102799] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/11/2023] [Accepted: 06/24/2023] [Indexed: 07/08/2023] Open
Abstract
Peroxynitrous acid/peroxynitrite (ONOOH/ONOO-) is a powerful oxidizing/nitrating system formed at sites of inflammation, which can modify biological targets, and particularly proteins. Here, we show that multiple proteins from primary human coronary artery smooth muscle cells are nitrated, with LC-MS peptide mass mapping providing data on the sites and extents of changes on cellular and extracellular matrix (ECM) proteins. Evidence is presented for selective and specific nitrations at Tyr and Trp on 11 cellular proteins (out of 3668, including 205 ECM species) in the absence of added reagent ONOOH/ONOO-, with this being consistent with low-level endogenous nitration. A number of these have key roles in cell signaling/sensing and protein turnover. With added ONOOH/ONOO-, more proteins were modified (84 total; with 129 nitrated Tyr and 23 nitrated Trp, with multiple modifications on some proteins), with this occurring at the same and additional sites to endogenous modification. With low concentrations of ONOOH/ONOO- (50 μM) nitration occurs on specific proteins at particular sites, and is not driven by protein or Tyr/Trp abundance, with modifications detected on some low abundance proteins. However, with higher ONOOH/ONOO- concentrations (500 μM), modification is primarily driven by protein abundance. ECM species are major targets and over-represented in the pool of modified proteins, with fibronectin and thrombospondin-1 being particularly heavily modified (12 sites in each case). Both endogenous and exogenous nitration of cell- and ECM-derived species may have significant effects on cell and protein function, and potentially be involved in the development and exacerbation of diseases such as atherosclerosis.
Collapse
Affiliation(s)
- Shuqi Xu
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark
| | - Christine Y Chuang
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark
| | - Clare L Hawkins
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark
| | - Per Hägglund
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark.
| | - Michael J Davies
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark.
| |
Collapse
|
9
|
Mathai C, Jourd'heuil F, Pham LGC, Gilliard K, Howard D, Balnis J, Jaitovich A, Chittur SV, Rilley M, Peredo-Wende R, Ammoura I, Shin SJ, Barroso M, Barra J, Shishkova E, Coon JJ, Lopez-Soler RI, Jourd'heuil D. Nuclear cytoglobin associates with HMGB2 and regulates DNA damage and genome-wide transcriptional output in the vasculature. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.10.540045. [PMID: 37214992 PMCID: PMC10197644 DOI: 10.1101/2023.05.10.540045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Identifying novel regulators of vascular smooth muscle cell function is necessary to further understand cardiovascular diseases. We previously identified cytoglobin, a hemoglobin homolog, with myogenic and cytoprotective roles in the vasculature. The specific mechanism of action of cytoglobin is unclear but does not seem to be related to oxygen transport or storage like hemoglobin. Herein, transcriptomic profiling of injured carotid arteries in cytoglobin global knockout mice revealed that cytoglobin deletion accelerated the loss of contractile genes and increased DNA damage. Overall, we show that cytoglobin is actively translocated into the nucleus of vascular smooth muscle cells through a redox signal driven by NOX4. We demonstrate that nuclear cytoglobin heterodimerizes with the non-histone chromatin structural protein HMGB2. Our results are consistent with a previously unknown function by which a non-erythrocytic hemoglobin inhibits DNA damage and regulates gene programs in the vasculature by modulating the genome-wide binding of HMGB2.
Collapse
|
10
|
Keller TCS, Lechauve C, Keller AS, Brooks S, Weiss MJ, Columbus L, Ackerman H, Cortese-Krott MM, Isakson BE. The role of globins in cardiovascular physiology. Physiol Rev 2022; 102:859-892. [PMID: 34486392 PMCID: PMC8799389 DOI: 10.1152/physrev.00037.2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 12/11/2022] Open
Abstract
Globin proteins exist in every cell type of the vasculature, from erythrocytes to endothelial cells, vascular smooth muscle cells, and peripheral nerve cells. Many globin subtypes are also expressed in muscle tissues (including cardiac and skeletal muscle), in other organ-specific cell types, and in cells of the central nervous system (CNS). The ability of each of these globins to interact with molecular oxygen (O2) and nitric oxide (NO) is preserved across these contexts. Endothelial α-globin is an example of extraerythrocytic globin expression. Other globins, including myoglobin, cytoglobin, and neuroglobin, are observed in other vascular tissues. Myoglobin is observed primarily in skeletal muscle and smooth muscle cells surrounding the aorta or other large arteries. Cytoglobin is found in vascular smooth muscle but can also be expressed in nonvascular cell types, especially in oxidative stress conditions after ischemic insult. Neuroglobin was first observed in neuronal cells, and its expression appears to be restricted mainly to the CNS and the peripheral nervous system. Brain and CNS neurons expressing neuroglobin are positioned close to many arteries within the brain parenchyma and can control smooth muscle contraction and thus tissue perfusion and vascular reactivity. Overall, reactions between NO and globin heme iron contribute to vascular homeostasis by regulating vasodilatory NO signals and scavenging reactive species in cells of the mammalian vascular system. Here, we discuss how globin proteins affect vascular physiology, with a focus on NO biology, and offer perspectives for future study of these functions.
Collapse
Affiliation(s)
- T C Stevenson Keller
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Christophe Lechauve
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Alexander S Keller
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Steven Brooks
- Physiology Unit, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, Maryland
| | - Mitchell J Weiss
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Linda Columbus
- Department of Chemistry, University of Virginia, Charlottesville, Virginia
| | - Hans Ackerman
- Physiology Unit, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, Maryland
| | - Miriam M Cortese-Krott
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology, Medical Faculty, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia
| |
Collapse
|
11
|
Mahgoup EM, Khaleel SA, El-Mahdy MA, Abd-Allah AR, Zweier JL. Role of cytoglobin in cigarette smoke constituent-induced loss of nitric oxide bioavailability in vascular smooth muscle cells. Nitric Oxide 2022; 119:9-18. [PMID: 34875385 PMCID: PMC8752519 DOI: 10.1016/j.niox.2021.12.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/22/2021] [Accepted: 12/03/2021] [Indexed: 02/03/2023]
Abstract
Cytoglobin (Cygb) has been identified as the major nitric oxide (NO) metabolizing protein in vascular smooth muscle cells (VSMCs) and is crucial for the regulation of vascular tone. In the presence of its requisite cytochrome B5a (B5)/B5 reductase-isoform-3 (B5R) reducing system, Cygb controls NO metabolism through the oxygen-dependent process of NO dioxygenation. Tobacco cigarette smoking (TCS) induces vascular dysfunction; however, the role of Cygb in the pathophysiology of TCS-induced cardiovascular disease has not been previously investigated. While TCS impairs NO biosynthesis, its effect on NO metabolism remains unclear. Therefore, we performed studies in aortic VSMCs with tobacco smoke extract (TSE) exposure to investigate the effects of cigarette smoke constituents on the rates of NO decay, with focus on the alterations that occur in the process of Cygb-mediated NO metabolism. TSE greatly enhanced the rates of NO metabolism by VSMCs. An initial increase in superoxide-mediated NO degradation was seen at 4 h of exposure. This was followed by much larger progressive increases at 24 and 48 h, accompanied by parallel increases in the expression of Cygb and B5/B5R. siRNA-mediated Cygb knockdown greatly decreased these TSE-induced elevations in NO decay rates. Therefore, upregulation of the levels of Cygb and its reducing system accounted for the large increase in NO metabolism rate seen after 24 h of TSE exposure. Thus, increased Cygb-mediated NO degradation would contribute to TCS-induced vascular dysfunction and partial inhibition of Cygb expression or its NO dioxygenase function could be a promising therapeutic target to prevent secondary cardiovascular disease.
Collapse
Affiliation(s)
- Elsayed M Mahgoup
- Department of Internal Medicine, Division of Cardiovascular Medicine, And the EPR Center, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA; Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Sahar A Khaleel
- Department of Internal Medicine, Division of Cardiovascular Medicine, And the EPR Center, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA; Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Mohamed A El-Mahdy
- Department of Internal Medicine, Division of Cardiovascular Medicine, And the EPR Center, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA; Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Adel R Abd-Allah
- Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Jay L Zweier
- Department of Internal Medicine, Division of Cardiovascular Medicine, And the EPR Center, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
12
|
Gardner PR. Ordered Motions in the Nitric-Oxide Dioxygenase Mechanism of Flavohemoglobin and Assorted Globins with Tightly Coupled Reductases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1414:45-96. [PMID: 36520413 DOI: 10.1007/5584_2022_751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Nitric-oxide dioxygenases (NODs) activate and combine O2 with NO to form nitrate. A variety of oxygen-binding hemoglobins with associated partner reductases or electron donors function as enzymatic NODs. Kinetic and structural investigations of the archetypal two-domain microbial flavohemoglobin-NOD have illuminated an allosteric mechanism that employs selective tunnels for O2 and NO, gates for NO and nitrate, transient O2 association with ferric heme, and an O2 and NO-triggered, ferric heme spin crossover-driven, motion-controlled, and dipole-regulated electron-transfer switch. The proposed mechanism facilitates radical-radical coupling of ferric-superoxide with NO to form nitrate while preventing suicidal ferrous-NO formation. Diverse globins display the structural and functional motifs necessary for a similar allosteric NOD mechanism. In silico docking simulations reveal monomeric erythrocyte hemoglobin alpha-chain and beta-chain intrinsically matched and tightly coupled with NADH-cytochrome b5 oxidoreductase and NADPH-cytochrome P450 oxidoreductase, respectively, forming membrane-bound flavohemoglobin-like mammalian NODs. The neuroprotective neuroglobin manifests a potential NOD role in a close-fitting ternary complex with membrane-bound NADH-cytochrome b5 oxidoreductase and cytochrome b5. Cytoglobin interfaces weakly with cytochrome b5 for O2 and NO-regulated electron-transfer and coupled NOD activity. The mechanistic model also provides insight into the evolution of O2 binding cooperativity in hemoglobin and a basis for the discovery of allosteric NOD inhibitors.
Collapse
|
13
|
Thorne LS, Rochford G, Williams TD, Southam AD, Rodriguez-Blanco G, Dunn WB, Hodges NJ. Cytoglobin protects cancer cells from apoptosis by regulation of mitochondrial cardiolipin. Sci Rep 2021; 11:985. [PMID: 33441751 PMCID: PMC7806642 DOI: 10.1038/s41598-020-79830-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 12/10/2020] [Indexed: 12/15/2022] Open
Abstract
Cytoglobin is important in the progression of oral squamous cell carcinoma but the molecular and cellular basis remain to be elucidated. In the current study, we develop a new cell model to study the function of cytoglobin in oral squamous carcinoma and response to cisplatin. Transcriptomic profiling showed cytoglobin mediated changes in expression of genes related to stress response, redox metabolism, mitochondrial function, cell adhesion, and fatty acid metabolism. Cellular and biochemical studies show that cytoglobin expression results in changes to phenotype associated with cancer progression including: increased cellular proliferation, motility and cell cycle progression. Cytoglobin also protects cells from cisplatin-induced apoptosis and oxidative stress with levels of the antioxidant glutathione increased and total and mitochondrial reactive oxygen species levels reduced. The mechanism of cisplatin resistance involved inhibition of caspase 9 activation and cytoglobin protected mitochondria from oxidative stress-induced fission. To understand the mechanism behind these phenotypic changes we employed lipidomic analysis and demonstrate that levels of the redox sensitive and apoptosis regulating cardiolipin are significantly up-regulated in cells expressing cytoglobin. In conclusion, our data shows that cytoglobin expression results in important phenotypic changes that could be exploited by cancer cells in vivo to facilitate disease progression.
Collapse
Affiliation(s)
- Lorna S Thorne
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Garret Rochford
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Timothy D Williams
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Andrew D Southam
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Phenome Centre Birmingham, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Giovanny Rodriguez-Blanco
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Phenome Centre Birmingham, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Warwick B Dunn
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Phenome Centre Birmingham, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Nikolas J Hodges
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
14
|
Ilangovan G, Khaleel SA, Kundu T, Hemann C, El-Mahdy MA, Zweier JL. Defining the reducing system of the NO dioxygenase cytoglobin in vascular smooth muscle cells and its critical role in regulating cellular NO decay. J Biol Chem 2021; 296:100196. [PMID: 33334890 PMCID: PMC7948950 DOI: 10.1074/jbc.ra120.016394] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/07/2020] [Accepted: 12/15/2020] [Indexed: 12/27/2022] Open
Abstract
In smooth muscle, cytoglobin (Cygb) functions as a potent nitric oxide (NO) dioxygenase and regulates NO metabolism and vascular tone. Major questions remain regarding which cellular reducing systems regulate Cygb-mediated NO metabolism. To better define the Cygb-mediated NO dioxygenation process in vascular smooth muscle cells (SMCs), and the requisite reducing systems that regulate cellular NO decay, we assessed the intracellular concentrations of Cygb and its putative reducing systems and examined their roles in the process of NO decay. Cygb and the reducing systems, cytochrome b5 (B5)/cytochrome b5 reductase (B5R) and cytochrome P450 reductase (CPR) were measured in aortic SMCs. Intracellular Cygb concentration was estimated as 3.5 μM, while B5R, B5, and CPR were 0.88, 0.38, and 0.15 μM, respectively. NO decay in SMCs was measured following bolus addition of NO to air-equilibrated cells. siRNA-mediated knockdown experiments indicated that ∼78% of NO metabolism in SMCs is Cygb-dependent. Of this, ∼87% was B5R- and B5-dependent. CPR knockdown resulted in a small decrease in the NO dioxygenation rate (VNO), while depletion of ascorbate had no effect. Kinetic analysis of VNO for the B5/B5R/Cygb system with variation of B5 or B5R concentrations from their SMC levels showed that VNO exhibits apparent Michaelis-Menten behavior for B5 and B5R. In contrast, linear variation was seen with change in Cygb concentration. Overall, B5/B5R was demonstrated to be the major reducing system supporting Cygb-mediated NO metabolism in SMCs with changes in cellular B5/B5R levels modulating the process of NO decay.
Collapse
Affiliation(s)
- Govindasamy Ilangovan
- Department of Internal Medicine, Division of Cardiovascular Medicine, and the EPR Center, Davis Heart & Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Sahar A Khaleel
- Department of Internal Medicine, Division of Cardiovascular Medicine, and the EPR Center, Davis Heart & Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, USA; Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Tapan Kundu
- Department of Internal Medicine, Division of Cardiovascular Medicine, and the EPR Center, Davis Heart & Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Craig Hemann
- Department of Internal Medicine, Division of Cardiovascular Medicine, and the EPR Center, Davis Heart & Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Mohamed A El-Mahdy
- Department of Internal Medicine, Division of Cardiovascular Medicine, and the EPR Center, Davis Heart & Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Jay L Zweier
- Department of Internal Medicine, Division of Cardiovascular Medicine, and the EPR Center, Davis Heart & Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, USA.
| |
Collapse
|
15
|
DeMartino AW, Amdahl MB, Bocian K, Rose JJ, Tejero J, Gladwin MT. Redox sensor properties of human cytoglobin allosterically regulate heme pocket reactivity. Free Radic Biol Med 2021; 162:423-434. [PMID: 33144263 PMCID: PMC7889637 DOI: 10.1016/j.freeradbiomed.2020.10.321] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 10/21/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022]
Abstract
Cytoglobin is a conserved hemoprotein ubiquitously expressed in mammalian tissues, which conducts electron transfer reactions with proposed signaling functions in nitric oxide (NO) and lipid metabolism. Cytoglobin has an E7 distal histidine (His81), which unlike related globins such as myoglobin and hemoglobin, is in equilibrium between a bound, hexacoordinate state and an unbound, pentacoordinate state. The His81 binding equilibrium appears to be allosterically modulated by the presence of an intramolecular disulfide between two cysteines (Cys38 and Cys83). The formation of this disulfide bridge regulates nitrite reductase activity and lipid binding. Herein, we attempt to clarify the effects of defined thiol oxidation states on small molecule binding of cytoglobin heme, using cyanide binding to probe the ferric state. Cyanide binding kinetics to wild-type cytoglobin reveal at least two kinetically distinct subpopulations, depending on thiol oxidation states. Experiments with covalent thiol modification by NEM, glutathione, and amino acid substitutions (C38S, C83S and H81A), indicate that subpopulations ranging from fully reduced thiols, single thiol oxidation, and intramolecular disulfide formation determine heme binding properties by modulating the histidine-heme affinity and ligand binding. The redox modulation of ligand binding is sensitive to physiological levels of hydrogen peroxide, with a functional midpoint redox potential for the native cytoglobin intramolecular disulfide bond of -189 ± 4 mV, a value within the boundaries of intracellular redox potentials. These results support the hypothesis that Cys38 and Cys83 on cytoglobin serve as sensitive redox sensors that modulate the cytoglobin distal heme pocket reactivity and ligand binding.
Collapse
Affiliation(s)
- Anthony W DeMartino
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Matthew B Amdahl
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kaitlin Bocian
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jason J Rose
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States; Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, United States; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15260, United States
| | - Jesús Tejero
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States; Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, United States; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15260, United States; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, United States.
| | - Mark T Gladwin
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States; Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, United States; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15260, United States.
| |
Collapse
|
16
|
Red Blood Cells and Hemoglobin in Human Atherosclerosis and Related Arterial Diseases. Int J Mol Sci 2020; 21:ijms21186756. [PMID: 32942605 PMCID: PMC7554753 DOI: 10.3390/ijms21186756] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 02/07/2023] Open
Abstract
As the main particulate component of the circulating blood, RBCs play major roles in physiological hemodynamics and impact all arterial wall pathologies. RBCs are the main determinant of blood viscosity, defining the frictional forces exerted by the blood on the arterial wall. This function is used in phylogeny and ontogeny of the cardiovascular (CV) system, allowing the acquisition of vasomotricity adapted to local metabolic demands, and systemic arterial pressure after birth. In pathology, RBCs collide with the arterial wall, inducing both local retention of their membranous lipids and local hemolysis, releasing heme-Fe++ with a high toxicity for arterial cells: endothelial and smooth muscle cells (SMCs) cardiomyocytes, neurons, etc. Specifically, overloading of cells by Fe++ promotes cell death. This local hemolysis is an event associated with early and advanced stages of human atherosclerosis. Similarly, the permanent renewal of mural RBC clotting is the major support of oxidation in abdominal aortic aneurysm. In parallel, calcifications promote intramural hemorrhages, and hemorrhages promote an osteoblastic phenotypic shift of arterial wall cells. Different plasma or tissue systems are able, at least in part, to limit this injury by acting at the different levels of this system.
Collapse
|
17
|
Amdahl MB, DeMartino AW, Gladwin MT. Inorganic nitrite bioactivation and role in physiological signaling and therapeutics. Biol Chem 2020; 401:201-211. [PMID: 31747370 DOI: 10.1515/hsz-2019-0349] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 11/02/2019] [Indexed: 01/23/2023]
Abstract
The bioactivation of inorganic nitrite refers to the conversion of otherwise 'inert' nitrite to the diatomic signaling molecule nitric oxide (NO), which plays important roles in human physiology and disease, notably in the regulation of vascular tone and blood flow. While the most well-known sources of NO are the nitric oxide synthase (NOS) enzymes, another source of NO is the nitrate-nitrite-NO pathway, whereby nitrite (obtained from reduction of dietary nitrate) is further reduced to form NO. The past few decades have seen extensive study of the mechanisms of NO generation through nitrate and nitrite bioactivation, as well as growing appreciation of the contribution of this pathway to NO signaling in vivo. This review, prepared for the volume 400 celebration issue of Biological Chemistry, summarizes some of the key reactions of the nitrate-nitrite-NO pathway such as reduction, disproportionation, dehydration, and oxidative denitrosylation, as well as current evidence for the contribution of the pathway to human cardiovascular physiology. Finally, ongoing efforts to develop novel medical therapies for multifarious conditions, especially those related to pathologic vasoconstriction and ischemia/reperfusion injury, are also explored.
Collapse
Affiliation(s)
- Matthew B Amdahl
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Anthony W DeMartino
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Mark T Gladwin
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA.,Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
18
|
Lessons from the post-genomic era: Globin diversity beyond oxygen binding and transport. Redox Biol 2020; 37:101687. [PMID: 32863222 PMCID: PMC7475203 DOI: 10.1016/j.redox.2020.101687] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/11/2020] [Accepted: 08/11/2020] [Indexed: 12/16/2022] Open
Abstract
Vertebrate hemoglobin (Hb) and myoglobin (Mb) were among the first proteins whose structures and sequences were determined over 50 years ago. In the subsequent pregenomic period, numerous related proteins came to light in plants, invertebrates and bacteria, that shared the myoglobin fold, a signature sequence motif characteristic of a 3-on-3 α-helical sandwich. Concomitantly, eukaryote and bacterial globins with a truncated 2-on-2 α-helical fold were discovered. Genomic information over the last 20 years has dramatically expanded the list of known globins, demonstrating their existence in a limited number of archaeal genomes, a majority of bacterial genomes and an overwhelming majority of eukaryote genomes. In vertebrates, 6 additional globin types were identified, namely neuroglobin (Ngb), cytoglobin (Cygb), globin E (GbE), globin X (GbX), globin Y (GbY) and androglobin (Adgb). Furthermore, functions beyond the familiar oxygen transport and storage have been discovered within the vertebrate globin family, including NO metabolism, peroxidase activity, scavenging of free radicals, and signaling functions. The extension of the knowledge on globin functions suggests that the original roles of bacterial globins must have been enzymatic, involved in defense against NO toxicity, and perhaps also as sensors of O2, regulating taxis away or towards high O2 concentrations. In this review, we aimed to discuss the evolution and remarkable functional diversity of vertebrate globins with particular focus on the variety of non-canonical expression sites of mammalian globins and their according impressive variability of atypical functions.
Collapse
|
19
|
Thuy LTT, Hai H, Kawada N. Role of cytoglobin, a novel radical scavenger, in stellate cell activation and hepatic fibrosis. Clin Mol Hepatol 2020; 26:280-293. [PMID: 32492766 PMCID: PMC7364355 DOI: 10.3350/cmh.2020.0037] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/09/2020] [Accepted: 03/13/2020] [Indexed: 12/17/2022] Open
Abstract
Cytoglobin (Cygb), a stellate cell-specific globin, has recently drawn attention due to its association with liver fibrosis. In the livers of both humans and rodents, Cygb is expressed only in stellate cells and can be utilized as a marker to distinguish stellate cells from hepatic fibroblast-derived myofibroblasts. Loss of Cygb accelerates liver fibrosis and cancer development in mouse models of chronic liver injury including diethylnitrosamine-induced hepatocellular carcinoma, bile duct ligation-induced cholestasis, thioacetamide-induced hepatic fibrosis, and choline-deficient L-amino acid-defined diet-induced non-alcoholic steatohepatitis. This review focuses on the history of research into the role of reactive oxygen species and nitrogen species in liver fibrosis and discusses the current perception of Cygb as a novel radical scavenger with an emphasis on its role in hepatic stellate cell activation and fibrosis.
Collapse
Affiliation(s)
- Le Thi Thanh Thuy
- Department of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Hoang Hai
- Department of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Norifumi Kawada
- Department of Hepatology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| |
Collapse
|
20
|
Abstract
Significance: Cytoglobin (Cygb) was discovered as a new addition to the globin superfamily and subsequently identified to have potent nitric oxide (NO) dioxygenase function. Cygb plays a critical role in the oxygen-dependent regulation of NO levels and vascular tone. Recent Advances: In recent years, the mechanism of the Cygb-mediated NO dioxygenation has been studied in isolated protein, smooth muscle cell, isolated blood vessel, and in vivo animal model systems. Studies in Cygb-/- mice have demonstrated that Cygb plays a critical role in regulating blood pressure and vascular tone. This review summarizes advances in the knowledge of NO dioxygenation/metabolism regulated by Cygb. Advances in measurement of NO diffusion dynamics across blood vessels and kinetic modeling of Cygb-mediated NO dioxygenation are summarized. The oxygen-dependent regulation of NO degradation by Cygb is also reviewed along with how Cygb paradoxically generates NO from nitrite under anaerobic conditions. The important role of Cygb in the regulation of vascular function and disease is reviewed. Critical Issues: Cygb is a more potent NO dioxygenase (NOD) than previously known globins with structural differences in heme coordination and environment, conferring it with a higher rate of reduction and more rapid process of NO dioxygenation with unique oxygen dependence. Various cellular reducing systems regenerate the catalytic oxyferrous Cygb species, supporting a high rate of NO dioxygenation. Future Directions: There remains a critical need to further characterize the factors and processes that modulate Cygb-mediated NOD function, and to develop pharmacological or other approaches to modulate Cygb function and expression.
Collapse
Affiliation(s)
- Jay L Zweier
- Division of Cardiovascular Medicine, Department of Internal Medicine, Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Govindasamy Ilangovan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, Ohio, USA
| |
Collapse
|
21
|
Mathai C, Jourd'heuil FL, Lopez-Soler RI, Jourd'heuil D. Emerging perspectives on cytoglobin, beyond NO dioxygenase and peroxidase. Redox Biol 2020; 32:101468. [PMID: 32087552 PMCID: PMC7033357 DOI: 10.1016/j.redox.2020.101468] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/05/2020] [Accepted: 02/13/2020] [Indexed: 12/18/2022] Open
Abstract
Cytoglobin is an evolutionary ancient hemoglobin with poor functional annotation. Rather than constrained to penta coordination, cytoglobin's heme iron may exist either as a penta or hexacoordinated arrangement when exposed to different intracellular environments. Two cysteine residues at the surface of the protein form an intramolecular disulfide bond that regulates iron coordination, ligand binding, and peroxidase activity. Overall, biochemical results do not support a role for cytoglobin as a direct antioxidant enzyme that scavenges hydrogen peroxide because the rate of the reaction of cytoglobin with hydrogen peroxide is several orders of magnitude slower than metal and thiol-based peroxidases. Thus, alternative substrates such as fatty acids have been suggested and regulation of nitric oxide bioavailability through nitric oxide dioxygenase and nitrite reductase activities has received experimental support. Cytoglobin is broadly expressed in connective, muscle, and nervous tissues. Rational for differential cellular distribution is poorly understood but inducibility in response to hypoxia is one of the most established features of cytoglobin expression with regulation through the transcription factor hypoxia-inducible factor (HIF). Phenotypic characterization of cytoglobin deletion in the mouse have indicated broad changes that include a heightened inflammatory response and fibrosis, increase tumor burden, cardiovascular dysfunction, and hallmarks of senescence. Some of these changes might be reversed upon inhibition of nitric oxide synthase. However, subcellular and molecular interactions have been seldom characterized. In addition, specific molecular mechanisms of action are still lacking. We speculate that cytoglobin functionality will extend beyond nitric oxide handling and will have to encompass indirect regulatory antioxidant and redox sensing functions.
Collapse
Affiliation(s)
- Clinton Mathai
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Frances L Jourd'heuil
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | | | - David Jourd'heuil
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
22
|
Lu HS, Schmidt AM, Hegele RA, Mackman N, Rader DJ, Weber C, Daugherty A. Reporting Sex and Sex Differences in Preclinical Studies. Arterioscler Thromb Vasc Biol 2019; 38:e171-e184. [PMID: 30354222 DOI: 10.1161/atvbaha.118.311717] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Hong S Lu
- From the Department of Physiology, Saha Cardiovascular Research Center, University of Kentucky, Lexington (H.S.L., A.D.)
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, New York University Langone Medical Center, New York, NY (A.M.S.)
| | - Robert A Hegele
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada (R.A.H.)
| | - Nigel Mackman
- Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Daniel J Rader
- Department of Medicine (D.J.R.), Perelman School of Medicine, University of Pennsylvania, Philadelphia.,Department of Genetics (D.J.R.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Christian Weber
- Department of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität, Munich, Germany (C.W.).,German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Alan Daugherty
- From the Department of Physiology, Saha Cardiovascular Research Center, University of Kentucky, Lexington (H.S.L., A.D.)
| |
Collapse
|
23
|
Amdahl MB, DeMartino AW, Tejero J, Gladwin MT. Cytoglobin at the Crossroads of Vascular Remodeling. Arterioscler Thromb Vasc Biol 2019; 37:1803-1805. [PMID: 28954806 DOI: 10.1161/atvbaha.117.310058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Matthew B Amdahl
- From the Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine (M.B.A., A.W.D., J.T., M.T.G.), Department of Bioengineering (M.B.A.), and Division of Pulmonary, Allergy, and Critical Care Medicine (A.W.D., J.T., M.T.G.), University of Pittsburgh, PA
| | - Anthony W DeMartino
- From the Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine (M.B.A., A.W.D., J.T., M.T.G.), Department of Bioengineering (M.B.A.), and Division of Pulmonary, Allergy, and Critical Care Medicine (A.W.D., J.T., M.T.G.), University of Pittsburgh, PA
| | - Jesús Tejero
- From the Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine (M.B.A., A.W.D., J.T., M.T.G.), Department of Bioengineering (M.B.A.), and Division of Pulmonary, Allergy, and Critical Care Medicine (A.W.D., J.T., M.T.G.), University of Pittsburgh, PA
| | - Mark T Gladwin
- From the Heart, Lung, Blood, and Vascular Medicine Institute, Department of Medicine (M.B.A., A.W.D., J.T., M.T.G.), Department of Bioengineering (M.B.A.), and Division of Pulmonary, Allergy, and Critical Care Medicine (A.W.D., J.T., M.T.G.), University of Pittsburgh, PA.
| |
Collapse
|
24
|
Wu W, Zhang W, Choi M, Zhao J, Gao P, Xue M, Singer HA, Jourd'heuil D, Long X. Vascular smooth muscle-MAPK14 is required for neointimal hyperplasia by suppressing VSMC differentiation and inducing proliferation and inflammation. Redox Biol 2019; 22:101137. [PMID: 30771750 PMCID: PMC6377391 DOI: 10.1016/j.redox.2019.101137] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 02/05/2019] [Indexed: 12/19/2022] Open
Abstract
Injury-induced stenosis is a serious vascular complication. We previously reported that p38α (MAPK14), a redox-regulated p38MAPK family member was a negative regulator of the VSMC contractile phenotype in vitro. Here we evaluated the function of VSMC-MAPK14 in vivo in injury-induced neointima hyperplasia and the underlying mechanism using an inducible SMC-MAPK14 knockout mouse line (iSMC-MAPK14-/-). We show that MAPK14 expression and activity were induced in VSMCs after carotid artery ligation injury in mice and ex vivo cultured human saphenous veins. While the vasculature from iSMC-MAPK14-/- mice was indistinguishable from wildtype littermate controls at baseline, these mice exhibited reduced neointima formation following carotid artery ligation injury. Concomitantly, there was an increased VSMC contractile protein expression in the injured vessels and a decrease in proliferating cells. Blockade of MAPK14 through a selective inhibitor suppressed, while activation of MAPK14 by forced expression of an upstream MAPK14 kinase promoted VSMC proliferation in cultured VSMCs. Genome wide RNA array combined with VSMC lineage tracing studies uncovered that vascular injury evoked robust inflammatory responses including the activation of proinflammatory gene expression and accumulation of CD45 positive inflammatory cells, which were attenuated in iSMC-MAPK14-/- mice. Using multiple pharmacological and molecular approaches to manipulate MAPK14 pathway, we further confirmed the critical role of MAPK14 in activating proinflammatory gene expression in cultured VSMCs, which occurs in a p65/NFkB-dependent pathway. Finally, we found that NOX4 contributes to MAPK14 suppression of the VSMC contractile phenotype. Our results revealed that VSMC-MAPK14 is required for injury-induced neointima formation, likely through suppressing VSMC differentiation and promoting VSMC proliferation and inflammation. Our study will provide mechanistic insights into therapeutic strategies for mitigation of vascular stenosis.
Collapse
Affiliation(s)
- Wen Wu
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Wei Zhang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Mihyun Choi
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Jinjing Zhao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Ping Gao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Min Xue
- Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, PR China
| | - Harold A Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - David Jourd'heuil
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States
| | - Xiaochun Long
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, United States.
| |
Collapse
|
25
|
Lilly B, Dammeyer K, Marosis S, McCallinhart PE, Trask AJ, Lowe M, Sawant D. Endothelial cell-induced cytoglobin expression in vascular smooth muscle cells contributes to modulation of nitric oxide. Vascul Pharmacol 2018; 110:7-15. [PMID: 29969687 PMCID: PMC6135703 DOI: 10.1016/j.vph.2018.06.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/14/2018] [Accepted: 06/23/2018] [Indexed: 12/20/2022]
Abstract
Cytoglobin is a widely expressed heme protein that binds oxygen, carbon monoxide and nitric oxide. Recent examination of cytoglobin in the vasculature indicates that it contributes to nitric oxide availability, which is central to normal blood vessel function through regulation of smooth muscle cell tone and physiological response. Given the potential implications of cytoglobin in vascular function, we examined how cytoglobin might be uniquely regulated in vascular smooth muscle cells. Our data demonstrate that endothelial cells can increase the expression of cytoglobin in vascular smooth muscle cells, and the induction of cytoglobin is cell contact-dependent. We show that Notch signaling is necessary for endothelial cell-induced cytoglobin expression and Notch2 and Notch3 are sufficient to drive its expression in aortic smooth muscle cells. We further reveal that in cytoglobin-depleted smooth muscle cells there is increased cellular nitric oxide. These data demonstrate that, in addition to being the main producer of vascular nitric oxide, endothelial cells facilitate the ability of smooth muscle cells to metabolize nitric oxide through upregulation of cytoglobin. Our results reveal a novel mechanism by which Notch signaling contributes to vascular function through regulation of a gene that controls nitric oxide levels.
Collapse
Affiliation(s)
- Brenda Lilly
- Center for Cardiovascular Research, The Ohio State University, Columbus, OH, USA; The Heart Center, Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA; Department of Pediatrics, The Ohio State University, Columbus, OH, USA; The Ohio State University, Columbus, OH, USA.
| | - Kristen Dammeyer
- Center for Cardiovascular Research, The Ohio State University, Columbus, OH, USA; The Ohio State University, Columbus, OH, USA
| | - Sam Marosis
- Center for Cardiovascular Research, The Ohio State University, Columbus, OH, USA; The Ohio State University, Columbus, OH, USA
| | - Patricia E McCallinhart
- Center for Cardiovascular Research, The Ohio State University, Columbus, OH, USA; The Heart Center, Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA
| | - Aaron J Trask
- Center for Cardiovascular Research, The Ohio State University, Columbus, OH, USA; The Heart Center, Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA; Department of Pediatrics, The Ohio State University, Columbus, OH, USA; The Ohio State University, Columbus, OH, USA
| | - Megan Lowe
- Center for Cardiovascular Research, The Ohio State University, Columbus, OH, USA; The Heart Center, Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA
| | - Dwitiya Sawant
- Center for Cardiovascular Research, The Ohio State University, Columbus, OH, USA; The Heart Center, Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
26
|
Ran MX, Li Y, Zhang Y, Liang K, Ren YN, Zhang M, Zhou GB, Zhou YM, Wu K, Wang CD, Huang Y, Luo B, Qazi IH, Zhang HM, Zeng CJ. Transcriptome Sequencing Reveals the Differentially Expressed lncRNAs and mRNAs Involved in Cryoinjuries in Frozen-Thawed Giant Panda ( Ailuropoda melanoleuca) Sperm. Int J Mol Sci 2018; 19:ijms19103066. [PMID: 30297640 PMCID: PMC6212861 DOI: 10.3390/ijms19103066] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/03/2018] [Accepted: 10/05/2018] [Indexed: 02/06/2023] Open
Abstract
Sperm cryopreservation and artificial insemination are important methods for giant panda breeding and preservation of extant genetic diversity. Lower conception rates limit the use of artificial insemination with frozen-thawed giant panda sperm, due to the lack of understanding of the cryodamaging or cryoinjuring mechanisms in cryopreservation. Long non-coding RNAs (lncRNAs) are involved in regulating spermatogenesis. However, their roles during cryopreservation remain largely unexplored. Therefore, this study aimed to identify differentially expressed lncRNAs and mRNAs associated with cryodamage or freeze tolerance in frozen-thawed sperm through high throughput sequencing. A total of 61.05 Gb clean reads and 22,774 lncRNA transcripts were obtained. From the sequencing results, 1477 significantly up-regulated and 1,396 significantly down-regulated lncRNA transcripts from fresh and frozen-thawed sperm of giant panda were identified. GO and KEGG showed that the significantly dysregulated lncRNAs and mRNAs were mainly involved in regulating responses to cold stress and apoptosis, such as the integral component of membrane, calcium transport, and various signaling pathways including PI3K-Akt, p53 and cAMP. Our work is the first systematic profiling of lncRNA and mRNA in fresh and frozen-thawed giant panda sperm, and provides valuableinsights into the potential mechanism of cryodamage in sperm.
Collapse
Affiliation(s)
- Ming-Xia Ran
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Yuan Li
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Yan Zhang
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Kai Liang
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Ying-Nan Ren
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Ming Zhang
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Guang-Bin Zhou
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| | - Ying-Min Zhou
- China Conservation and Research Center for the Giant Panda, Wolong 473000, China.
| | - Kai Wu
- China Conservation and Research Center for the Giant Panda, Wolong 473000, China.
| | - Cheng-Dong Wang
- China Conservation and Research Center for the Giant Panda, Wolong 473000, China.
| | - Yan Huang
- China Conservation and Research Center for the Giant Panda, Wolong 473000, China.
| | - Bo Luo
- China Conservation and Research Center for the Giant Panda, Wolong 473000, China.
| | - Izhar Hyder Qazi
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
- Department of Veterinary Anatomy & Histology, Faculty of Bio-Sciences, Shaheed Benazir Bhutto University of Veterinary and Animal Sciences, Sakrand 67210, Pakistan.
| | - He-Min Zhang
- China Conservation and Research Center for the Giant Panda, Wolong 473000, China.
| | - Chang-Jun Zeng
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China.
| |
Collapse
|
27
|
Peterson SM, Turner JE, Harrington A, Davis-Knowlton J, Lindner V, Gridley T, Vary CPH, Liaw L. Notch2 and Proteomic Signatures in Mouse Neointimal Lesion Formation. Arterioscler Thromb Vasc Biol 2018; 38:1576-1593. [PMID: 29853569 PMCID: PMC6023756 DOI: 10.1161/atvbaha.118.311092] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 05/16/2018] [Indexed: 12/29/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— Vascular remodeling is associated with complex molecular changes, including increased Notch2, which promotes quiescence in human smooth muscle cells. We used unbiased protein profiling to understand molecular signatures related to neointimal lesion formation in the presence or absence of Notch2 and to test the hypothesis that loss of Notch2 would increase neointimal lesion formation because of a hyperproliferative injury response. Approach and Results— Murine carotid arteries isolated at 6 or 14 days after ligation injury were analyzed by mass spectrometry using a data-independent acquisition strategy in comparison to uninjured or sham injured arteries. We used a tamoxifen-inducible, cell-specific Cre recombinase strain to delete the Notch2 gene in smooth muscle cells. Vessel morphometric analysis and immunohistochemical staining were used to characterize lesion formation, assess vascular smooth muscle cell proliferation, and validate proteomic findings. Loss of Notch2 in smooth muscle cells leads to protein profile changes in the vessel wall during remodeling but does not alter overall lesion morphology or cell proliferation. Loss of smooth muscle Notch2 also decreases the expression of enhancer of rudimentary homolog, plectin, and annexin A2 in vascular remodeling. Conclusions— We identified unique protein signatures that represent temporal changes in the vessel wall during neointimal lesion formation in the presence and absence of Notch2. Overall lesion formation was not affected with loss of smooth muscle Notch2, suggesting compensatory pathways. We also validated the regulation of known injury- or Notch-related targets identified in other vascular contexts, providing additional insight into conserved pathways involved in vascular remodeling.
Collapse
Affiliation(s)
- Sarah M Peterson
- From the Maine Medical Center Research Institute, Scarborough (S.M.P., J.E.T., A.H., J.D.-K., V.L., T.G., C.P.H.V., L.L.).,University of Maine Graduate School of Biomedical Science and Engineering, Orono (S.M.P., V.L., T.G., C.P.H.V., L.L.)
| | - Jacqueline E Turner
- From the Maine Medical Center Research Institute, Scarborough (S.M.P., J.E.T., A.H., J.D.-K., V.L., T.G., C.P.H.V., L.L.)
| | - Anne Harrington
- From the Maine Medical Center Research Institute, Scarborough (S.M.P., J.E.T., A.H., J.D.-K., V.L., T.G., C.P.H.V., L.L.)
| | - Jessica Davis-Knowlton
- From the Maine Medical Center Research Institute, Scarborough (S.M.P., J.E.T., A.H., J.D.-K., V.L., T.G., C.P.H.V., L.L.).,Tufts Sackler School of Graduate Biomedical Sciences, Boston, MA (J.D.-K., V.L., T.G., C.P.H.V., L.L.)
| | - Volkhard Lindner
- From the Maine Medical Center Research Institute, Scarborough (S.M.P., J.E.T., A.H., J.D.-K., V.L., T.G., C.P.H.V., L.L.).,University of Maine Graduate School of Biomedical Science and Engineering, Orono (S.M.P., V.L., T.G., C.P.H.V., L.L.).,Tufts Sackler School of Graduate Biomedical Sciences, Boston, MA (J.D.-K., V.L., T.G., C.P.H.V., L.L.)
| | - Thomas Gridley
- From the Maine Medical Center Research Institute, Scarborough (S.M.P., J.E.T., A.H., J.D.-K., V.L., T.G., C.P.H.V., L.L.).,University of Maine Graduate School of Biomedical Science and Engineering, Orono (S.M.P., V.L., T.G., C.P.H.V., L.L.).,Tufts Sackler School of Graduate Biomedical Sciences, Boston, MA (J.D.-K., V.L., T.G., C.P.H.V., L.L.)
| | - Calvin P H Vary
- From the Maine Medical Center Research Institute, Scarborough (S.M.P., J.E.T., A.H., J.D.-K., V.L., T.G., C.P.H.V., L.L.).,University of Maine Graduate School of Biomedical Science and Engineering, Orono (S.M.P., V.L., T.G., C.P.H.V., L.L.).,Tufts Sackler School of Graduate Biomedical Sciences, Boston, MA (J.D.-K., V.L., T.G., C.P.H.V., L.L.)
| | - Lucy Liaw
- From the Maine Medical Center Research Institute, Scarborough (S.M.P., J.E.T., A.H., J.D.-K., V.L., T.G., C.P.H.V., L.L.) .,University of Maine Graduate School of Biomedical Science and Engineering, Orono (S.M.P., V.L., T.G., C.P.H.V., L.L.).,Tufts Sackler School of Graduate Biomedical Sciences, Boston, MA (J.D.-K., V.L., T.G., C.P.H.V., L.L.)
| |
Collapse
|