1
|
Costa ADSD, Vadym K, Park K. Engineered endothelium model enables recapitulation of vascular function and early atherosclerosis development. Biomaterials 2025; 314:122889. [PMID: 39423515 DOI: 10.1016/j.biomaterials.2024.122889] [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: 04/30/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
Human health relies heavily on the vascular endothelium. Here, we propose a novel engineered endothelium model (EEM), which recapitulated both normal vascular function and pathology. An artificial basement membrane (aBM), where porous polyvinyl alcohol hydrogel was securely integrated with human fibroblast-derived, decellularized extracellular matrix on both sides was fabricated first and followed by endothelial cells (ECs) and pericytes (PCs) adhesion, respectively. Our EEM formed robust adherens junction (VE-cad) and built an impermeable barrier with time, along with the nitric oxide (NO) secretion. In our EEM, ECs and PCs interacted each other via aBM and led to hemoglobin alpha 1 (Hb-α1) development, which was involved in NO control and was strongly interconnected with VE-cad as well. A resilient property of EEM under inflammatory milieu was also confirmed by VE-cad and barrier recovery with time. In particular interest, foam cells formation, a hallmark of atherosclerotic initiation was successfully recapitulated in our EEM, where a series of sequential events were confirmed: human monocytes adhesion, transendothelial migration, and oxidized low-density lipoprotein uptake by macrophages. Collectively, our EEM is excellent in recapitulating not only normal endothelium but early pathologic one, thereby enabling EEM to be a physiologically relevant model for vascular study and disease modeling.
Collapse
Affiliation(s)
| | - Kopych Vadym
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| | - Kwideok Park
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792, Republic of Korea.
| |
Collapse
|
2
|
Rodriguez-Muñoz A, Motahari-Rad H, Martin-Chaves L, Benitez-Porres J, Rodriguez-Capitan J, Gonzalez-Jimenez A, Insenser M, Tinahones FJ, Murri M. A Systematic Review of Proteomics in Obesity: Unpacking the Molecular Puzzle. Curr Obes Rep 2024; 13:403-438. [PMID: 38703299 PMCID: PMC11306592 DOI: 10.1007/s13679-024-00561-4] [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] [Accepted: 03/14/2024] [Indexed: 05/06/2024]
Abstract
PURPOSE OF REVIEW The present study aims to review the existing literature to identify pathophysiological proteins in obesity by conducting a systematic review of proteomics studies. Proteomics may reveal the mechanisms of obesity development and clarify the links between obesity and related diseases, improving our comprehension of obesity and its clinical implications. RECENT FINDINGS Most of the molecular events implicated in obesity development remain incomplete. Proteomics stands as a powerful tool for elucidating the intricate interactions among proteins in the context of obesity. This methodology has the potential to identify proteins involved in pathological processes and to evaluate changes in protein abundance during obesity development, contributing to the identification of early disease predisposition, monitoring the effectiveness of interventions and improving disease management overall. Despite many non-targeted proteomic studies exploring obesity, a comprehensive and up-to-date systematic review of the molecular events implicated in obesity development is lacking. The lack of such a review presents a significant challenge for researchers trying to interpret the existing literature. This systematic review was conducted following the PRISMA guidelines and included sixteen human proteomic studies, each of which delineated proteins exhibiting significant alterations in obesity. A total of 41 proteins were reported to be altered in obesity by at least two or more studies. These proteins were involved in metabolic pathways, oxidative stress responses, inflammatory processes, protein folding, coagulation, as well as structure/cytoskeleton. Many of the identified proteomic biomarkers of obesity have also been reported to be dysregulated in obesity-related disease. Among them, seven proteins, which belong to metabolic pathways (aldehyde dehydrogenase and apolipoprotein A1), the chaperone family (albumin, heat shock protein beta 1, protein disulfide-isomerase A3) and oxidative stress and inflammation proteins (catalase and complement C3), could potentially serve as biomarkers for the progression of obesity and the development of comorbidities, contributing to personalized medicine in the field of obesity. Our systematic review in proteomics represents a substantial step forward in unravelling the complexities of protein alterations associated with obesity. It provides valuable insights into the pathophysiological mechanisms underlying obesity, thereby opening avenues for the discovery of potential biomarkers and the development of personalized medicine in obesity.
Collapse
Affiliation(s)
- Alba Rodriguez-Muñoz
- Endocrinology and Nutrition UGC, Hospital Universitario Virgen de La Victoria, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Clínico Virgen de La Victoria, Málaga, Spain
- CIBER Fisiopatología de La Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Málaga, Spain
| | - Hanieh Motahari-Rad
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Laura Martin-Chaves
- Heart Area, Hospital Universitario Virgen de La Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Malaga, Spain
- Department of Dermatology and Medicine, Faculty of Medicine, University of Malaga, Malaga, Spain
| | - Javier Benitez-Porres
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Clínico Virgen de La Victoria, Málaga, Spain
- Department of Human Physiology, Physical Education and Sport, Faculty of Medicine, University of Malaga, Malaga, Spain
| | - Jorge Rodriguez-Capitan
- Heart Area, Hospital Universitario Virgen de La Victoria, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Malaga, Spain
- Biomedical Research Network Center for Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | | | - Maria Insenser
- Diabetes, Obesity and Human Reproduction Research Group, Department of Endocrinology & Nutrition, Hospital Universitario Ramón y Cajal & Universidad de Alcalá & Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) & Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain.
| | - Francisco J Tinahones
- Endocrinology and Nutrition UGC, Hospital Universitario Virgen de La Victoria, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Clínico Virgen de La Victoria, Málaga, Spain
- CIBER Fisiopatología de La Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Málaga, Spain
- Department of Dermatology and Medicine, Faculty of Medicine, University of Malaga, Malaga, Spain
| | - Mora Murri
- Endocrinology and Nutrition UGC, Hospital Universitario Virgen de La Victoria, Málaga, Spain.
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Clínico Virgen de La Victoria, Málaga, Spain.
- CIBER Fisiopatología de La Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Málaga, Spain.
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
3
|
Romero G, Martin B, Gabris B, Salama G. Relaxin suppresses atrial fibrillation, reverses fibrosis and reduces inflammation in aged hearts. Biochem Pharmacol 2024; 227:116407. [PMID: 38969298 DOI: 10.1016/j.bcp.2024.116407] [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: 03/19/2024] [Revised: 06/18/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Healthy aging results in cardiac structural and electrical remodeling that increase susceptibility to cardiovascular diseases. Relaxin has shown broad cardioprotective effects including anti-fibrotic, anti-arrhythmic and anti-inflammatory outcomes in multiple models. This paper focuses on the cardioprotective effects of Relaxin in a rat model of aging. Sustained atrial or ventricular fibrillation are readily induced in the hearts of aged but not young control animals. Treatment with Relaxin suppressed this arrhythmogenic response by increasing conduction velocity, decreasing fibrosis and promoting substantial cardiac remodeling. Relaxin treatment resulted in a significant increase in the levels of: Nav1.5, Cx43, βcatenin and Wnt1 in rat hearts. In isolated cardiomyocytes, Relaxin increased Nav1.5 expression. These effects were mimicked by CHIR 99021, a pharmacological activator of canonical Wnt signaling, but blocked by the canonical Wnt inhibitor Dickkopf1. Relaxin prevented TGF-β-dependent differentiation of cardiac fibroblasts into myofibroblasts while increasing the expression of Wnt1; the effects of Relaxin on cardiac fibroblast differentiation were blocked by Dickkopf1. RNASeq studies demonstrated reduced expression of pro-inflammatory cytokines and an increase in the expression of α- and β-globin in Relaxin-treated aged males. Relaxin reduces arrhythmogenicity in the hearts of aged rats by reduction of fibrosis and increased conduction velocity. These changes are accompanied by substantial remodeling of the cardiac tissue and appear to be mediated by increased canonical Wnt signaling. Relaxin also exerts significant anti-inflammatory and anti-oxidant effects in the hearts of aged rodents. The mechanisms by which Relaxin increases the expression of Wnt ligands, promotes Wnt signaling and reprograms gene expression remain to be determined.
Collapse
Affiliation(s)
- Guillermo Romero
- Departments of Pharmacology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Brian Martin
- Departments of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Beth Gabris
- Departments of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Guy Salama
- Departments of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
4
|
Lim L. Modifying Alzheimer's disease pathophysiology with photobiomodulation: model, evidence, and future with EEG-guided intervention. Front Neurol 2024; 15:1407785. [PMID: 39246604 PMCID: PMC11377238 DOI: 10.3389/fneur.2024.1407785] [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: 03/27/2024] [Accepted: 08/01/2024] [Indexed: 09/10/2024] Open
Abstract
This manuscript outlines a model of Alzheimer's Disease (AD) pathophysiology in progressive layers, from its genesis to the development of biomarkers and then to symptom expression. Genetic predispositions are the major factor that leads to mitochondrial dysfunction and subsequent amyloid and tau protein accumulation, which have been identified as hallmarks of AD. Extending beyond these accumulations, we explore a broader spectrum of pathophysiological aspects, including the blood-brain barrier, blood flow, vascular health, gut-brain microbiodata, glymphatic flow, metabolic syndrome, energy deficit, oxidative stress, calcium overload, inflammation, neuronal and synaptic loss, brain matter atrophy, and reduced growth factors. Photobiomodulation (PBM), which delivers near-infrared light to selected brain regions using portable devices, is introduced as a therapeutic approach. PBM has the potential to address each of these pathophysiological aspects, with data provided by various studies. They provide mechanistic support for largely small published clinical studies that demonstrate improvements in memory and cognition. They inform of PBM's potential to treat AD pending validation by large randomized controlled studies. The presentation of brain network and waveform changes on electroencephalography (EEG) provide the opportunity to use these data as a guide for the application of various PBM parameters to improve outcomes. These parameters include wavelength, power density, treatment duration, LED positioning, and pulse frequency. Pulsing at specific frequencies has been found to influence the expression of waveforms and modifications of brain networks. The expression stems from the modulation of cellular and protein structures as revealed in recent studies. These findings provide an EEG-based guide for the use of artificial intelligence to personalize AD treatment through EEG data feedback.
Collapse
Affiliation(s)
- Lew Lim
- Vielight Inc., Toronto, ON, Canada
| |
Collapse
|
5
|
Abbineni PS, Baid S, Weiss MJ. A moonlighting job for α-globin in blood vessels. Blood 2024; 144:834-844. [PMID: 38848504 DOI: 10.1182/blood.2023022192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/08/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
ABSTRACT Red blood cells express high levels of hemoglobin A tetramer (α2β2) to facilitate oxygen transport. Hemoglobin subunits and related proteins are also expressed at lower levels in other tissues across the animal kingdom. Physiological functions for most nonerythroid globins likely derive from their ability to catalyze reduction-oxidation (redox) reactions via electron transfer through heme-associated iron. An interesting example is illustrated by the recent discovery that α-globin without β-globin is expressed in some arteriolar endothelial cells (ECs). α-globin binds EC nitric oxide (NO) synthase (eNOS) and degrades its enzymatic product NO, a potent vasodilator. Thus, depletion of α-globin in ECs or inhibition of its association with eNOS causes arteriolar relaxation and lowering of blood pressure in mice. Some of these findings have been replicated in isolated human blood vessels, and genetic studies are tractable in populations in which α-thalassemia alleles are prevalent. Two small studies identified associations between loss of α-globin genes in humans and NO-regulated vascular responses elicited by local hypoxia-induced blood flow or thermal stimulation. In a few larger population-based studies, no associations were detected between loss of α-globin genes and blood pressure, ischemic stroke, or pulmonary hypertension. In contrast, a significant positive association between α-globin gene copy number and kidney disease was detected in an African American cohort. Further studies are required to define comprehensively the expression of α-globin in different vascular beds and ascertain their overall impact on normal and pathological vascular physiology.
Collapse
Affiliation(s)
- Prabhodh S Abbineni
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL
| | - Srishti Baid
- Life Sciences Institute, University of Michigan, Ann Arbor, MI
| | - Mitchell J Weiss
- Department of Hematology, St Jude Children's Research Hospital, Memphis, TN
| |
Collapse
|
6
|
Ruoss S, Nasamran CA, Ball ST, Chen JL, Halter KN, Bruno KA, Whisenant TC, Parekh JN, Dorn SN, Esparza MC, Bremner SN, Fisch KM, Engler AJ, Ward SR. Comparative single-cell transcriptional and proteomic atlas of clinical-grade injectable mesenchymal source tissues. SCIENCE ADVANCES 2024; 10:eadn2831. [PMID: 38996032 PMCID: PMC11244553 DOI: 10.1126/sciadv.adn2831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 06/06/2024] [Indexed: 07/14/2024]
Abstract
Bone marrow aspirate concentrate (BMAC) and adipose-derived stromal vascular fraction (ADSVF) are the most marketed stem cell therapies to treat a variety of conditions in the general population and elite athletes. Both tissues have been used interchangeably clinically even though their detailed composition, heterogeneity, and mechanisms of action have neither been rigorously inventoried nor compared. This lack of information has prevented investigations into ideal dosages and has facilitated anecdata and misinformation. Here, we analyzed single-cell transcriptomes, proteomes, and flow cytometry profiles from paired clinical-grade BMAC and ADSVF. This comparative transcriptional atlas challenges the prevalent notion that there is one therapeutic cell type present in both tissues. We also provide data of surface markers that may enable isolation and investigation of cell (sub)populations. Furthermore, the proteome atlas highlights intertissue and interpatient heterogeneity of injected proteins with potentially regenerative or immunomodulatory capacities. An interactive webtool is available online.
Collapse
Affiliation(s)
- Severin Ruoss
- Department of Orthopaedic Surgery, UC San Diego, La Jolla, CA, USA
| | - Chanond A. Nasamran
- Center for Computational Biology and Bioinformatics, UC San Diego, La Jolla, CA, USA
| | - Scott T. Ball
- Department of Orthopaedic Surgery, UC San Diego, La Jolla, CA, USA
| | - Jeffrey L. Chen
- Department of Anesthesiology, Center for Pain, UC San Diego, La Jolla, CA, USA
| | - Kenneth N. Halter
- Department of Anesthesiology, Center for Pain, UC San Diego, La Jolla, CA, USA
| | - Kelly A. Bruno
- Department of Anesthesiology, Center for Pain, UC San Diego, La Jolla, CA, USA
| | - Thomas C. Whisenant
- Center for Computational Biology and Bioinformatics, UC San Diego, La Jolla, CA, USA
| | - Jesal N. Parekh
- Department of Orthopaedic Surgery, UC San Diego, La Jolla, CA, USA
| | - Shanelle N. Dorn
- Department of Orthopaedic Surgery, UC San Diego, La Jolla, CA, USA
| | - Mary C. Esparza
- Department of Orthopaedic Surgery, UC San Diego, La Jolla, CA, USA
| | | | - Kathleen M. Fisch
- Center for Computational Biology and Bioinformatics, UC San Diego, La Jolla, CA, USA
- Department of Obstetrics, Gynecology & Reproductive Sciences, UC San Diego, La Jolla, CA, USA
| | - Adam J. Engler
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Samuel R. Ward
- Department of Orthopaedic Surgery, UC San Diego, La Jolla, CA, USA
- Chien-Lay Department of Bioengineering, UC San Diego, La Jolla, CA, USA
- Department of Radiology, UC San Diego, La Jolla, CA, USA
| |
Collapse
|
7
|
Suh SB, Suh JY, Cho SB. Analyzing secretory proteins in human dermal fibroblast-conditioned medium for angiogenesis: A bioinformatic approach. Skin Res Technol 2024; 30:e13568. [PMID: 38200622 PMCID: PMC10781896 DOI: 10.1111/srt.13568] [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: 12/07/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND The conditioned medium from human dermal fibroblasts (dermal fibroblast-conditioned medium; DFCM) contains a diverse array of secretory proteins, including growth factors and wound repair-promoting proteins. Angiogenesis, a crucial process that facilitates the infiltration of inflammatory cells during wound repair, is induced by a hypoxic environment and inflammatory cytokines. METHODS In this study, we conducted a comprehensive bioinformatic analysis of 337 proteins identified through proteomics analysis of DFCM. We specifically focused on 64 DFCM proteins with potential involvement in angiogenesis. These proteins were further classified based on their characteristics, and we conducted a detailed analysis of their protein-protein interactions. RESULTS Gene Ontology protein classification categorized these 64 DFCM proteins into various classes, including metabolite interconversion enzymes (N = 11), protein modifying enzymes (N = 10), protein-binding activity modulators (N = 9), cell adhesion molecules (N = 6), extracellular matrix proteins (N = 6), transfer/carrier proteins (N = 3), calcium-binding proteins (N = 2), chaperones (N = 2), cytoskeletal proteins (N = 2), RNA metabolism proteins (N = 1), intercellular signal molecules (N = 1), transporters (N = 1), scaffold/adaptor proteins (N = 1), and unclassified proteins (N = 9). Furthermore, our protein-protein interaction network analysis of DFCM proteins revealed two distinct networks: one with medium confidence level interaction scores, consisting of 60 proteins with significant connections, and another at a high confidence level, comprising 52 proteins with significant interactions. CONCLUSIONS Our bioinformatic analysis highlights the presence of a multitude of secretory proteins in DFCM that form significant protein-protein interaction networks crucial for regulating angiogenesis. These findings underscore the critical roles played by DFCM proteins in various stages of angiogenesis during the wound repair process.
Collapse
Affiliation(s)
| | | | - Sung Bin Cho
- Yonsei Seran Dermatology and Laser ClinicSeoulSouth Korea
| |
Collapse
|
8
|
Thomas G, Banton KL, Garrett R, Palacio CH, Acuna D, Madayag R, Bar-Or D. Hypoxia Dysregulates the Transcription of Myoendothelial Junction Proteins Involved with Nitric Oxide Production in Brain Endothelial Cells. Biomedicines 2023; 12:75. [PMID: 38255181 PMCID: PMC10813549 DOI: 10.3390/biomedicines12010075] [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: 12/04/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
Myoendothelial junctions (MEJs) are structures that allow chemical signals to be transmitted between endothelial cells (ECs) and vascular smooth muscle cells, which control vascular tone. MEJs contain hemoglobin alpha (Hbα) and endothelial nitric oxide synthase (eNOS) complexes that appear to control the production and scavenging of nitric oxide (NO) along with the activity of cytochrome b5 reductase 3 (CYB5R3). The aim of this study was to examine how hypoxia affected the regulation of proteins involved in the production of NO in brain ECs. In brief, human brain microvascular endothelial cells (HBMEC) were exposed to cobalt chloride (CoCl2), a hypoxia mimetic, and a transcriptional analysis was performed using primers for eNOS, CYB5R3, and Hbα2 with ΔΔCt relative gene expression normalized to GAPDH. NO production was also measured after treatment using 4,5-diaminofluorescein diacetate (DAF-DA), a fluorescent NO indicator. When HBMEC were exposed to CoCl2 for 48 h, eNOS and CYB5R3 messenger RNA significantly decreased (up to -17.8 ± 4.30-fold and -10.4 ± 2.8, respectively) while Hbα2 increased to detectable levels. Furthermore, CoCl2 treatment caused a redistribution of peripheral membrane-generated NO production to a perinuclear region. To the best of our knowledge, this is the first time this axis has been studied in brain ECs and these findings imply that hypoxia may cause dysregulation of proteins that regulate NO production in brain MEJs.
Collapse
Affiliation(s)
- Gregory Thomas
- Trauma and Stroke Research Lab, 601 East Hampden Ave, Englewood, CO 80113, USA
| | - Kaysie L. Banton
- Trauma and Surgery Services, Swedish Medical Center, 501 East Hampden Ave, Englewood, CO 80113, USA
| | - Raymond Garrett
- Trauma and Stroke Research Lab, 601 East Hampden Ave, Englewood, CO 80113, USA
| | - Carlos H. Palacio
- Trauma and Surgery Services, South Texas Health System-McAllen, 301 West Expressway 83, McAllen, TX 78503, USA
| | - David Acuna
- Trauma and Surgery Services, Wesley Medical Center, 550 North Hillside St, Wichita, KS 67214, USA
| | - Robert Madayag
- Trauma and Surgery Services, Lutheran Medical Center, 8300 W. 38th Ave, Wheat Ridge, CO 80033, USA
| | - David Bar-Or
- Trauma and Stroke Research Lab, 601 East Hampden Ave, Englewood, CO 80113, USA
- Trauma and Surgery Services, Swedish Medical Center, 501 East Hampden Ave, Englewood, CO 80113, USA
- Trauma and Surgery Services, South Texas Health System-McAllen, 301 West Expressway 83, McAllen, TX 78503, USA
- Trauma and Surgery Services, Wesley Medical Center, 550 North Hillside St, Wichita, KS 67214, USA
- Department of Molecular Biology, Rocky Vista University, 8401 S Chambers Rd, Parker, CO 80134, USA
| |
Collapse
|
9
|
Robertson JO, Bazeley P, Erzurum SC, Asosingh K. Single-cell transcriptomic profiling of microvascular endothelial cell heterogeneity in congenital diaphragmatic hernia. Sci Rep 2023; 13:9851. [PMID: 37330615 PMCID: PMC10276841 DOI: 10.1038/s41598-023-37050-y] [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: 03/10/2022] [Accepted: 06/15/2023] [Indexed: 06/19/2023] Open
Abstract
Congenital diaphragmatic hernia (CDH) is a neonatal anomaly that includes pulmonary hypoplasia and hypertension. We hypothesized that microvascular endothelial cell (EC) heterogeneity is different in CDH lungs and related to lung underdevelopment and remodeling. To test this, we evaluated rat fetuses at E21.5 in a nitrofen model of CDH to compare lung transcriptomes among healthy controls (2HC), nitrofen-exposed controls (NC) and nitrofen-exposed subjects with CDH. Single-cell RNA sequencing with unbiased clustering revealed 3 distinct microvascular EC clusters: a general population (mvEC), a proliferative population and a population high in hemoglobin. Only the CDH mvEC cluster had a distinct inflammatory transcriptomic signature as compared to the 2HC and NC endothelial cells, e.g. greater activation and adhesion of inflammatory cells and production of reactive oxygen species. Furthermore, CDH mvECs had downregulated Ca4, Apln and Ednrb gene expression. Those genes are markers for ECs important to lung development, gas exchange and alveolar repair (mvCa4+). mvCa4+ ECs were reduced in CDH (2HC [22.6%], NC [13.1%] and CDH [5.3%], p < 0.0001). Overall, these findings identify transcriptionally distinct microvascular endothelial cell clusters in CDH, including the distinctly inflammatory mvEC cluster and the depleted group of mvCa4+ ECs, which together may contribute to pathogenesis.
Collapse
Affiliation(s)
- Jason O Robertson
- Department of Pediatric Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic Children's, 9500 Euclid Avenue/A10, Cleveland, OH, 44195, USA.
| | - Peter Bazeley
- Department of Quantitative Health Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, 44195, USA
| | - Serpil C Erzurum
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, 44195, USA
| | - Kewal Asosingh
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, 44195, USA
| |
Collapse
|
10
|
Sumi MP, Tupta B, Roychowdhury S, Comhair S, Asosingh K, Stuehr DJ, Erzurum SC, Ghosh A. Hemoglobin resident in the lung epithelium is protective for smooth muscle soluble guanylate cyclase function. Redox Biol 2023; 63:102717. [PMID: 37120930 PMCID: PMC10172757 DOI: 10.1016/j.redox.2023.102717] [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: 03/22/2023] [Revised: 04/17/2023] [Accepted: 04/24/2023] [Indexed: 05/02/2023] Open
Abstract
Hemoglobin (Hb) present in the lung epithelium is of unknown significance. However Hb being an nitric oxide (NO) scavenger can bind to NO and reduce its deleterious effects. Hence we postulated an NO scavenging role for this lung Hb. Doing transwell co-culture with bronchial epithelial cells, A549/16-HBE (apical) and human airway smooth muscle cells (HASMCs as basal), we found that Hb can protect the smooth muscle soluble guanylyl cyclase (sGC) from excess NO. Inducing the apical A549/16-HBE cells with cytokines to trigger iNOS expression and NO generation caused a time dependent increase in SNO-sGC and this was accompanied with a concomitant drop in sGC-α1β1 heterodimerization. Silencing Hbαβ in the apical cells further increased the SNO on sGC with a faster drop in the sGC heterodimer and these effects were additive along with further silencing of thioredoxin 1 (Trx1). Since heme of Hb is critical for NO scavenging we determined the Hb heme in a mouse model of allergic asthma (OVA) and found that Hb in the inflammed OVA lungs was low in heme or heme-free relative to those of naïve lungs. Further we established a direct correlation between the status of the sGC heterodimer and the Hb heme from lung samples of human asthma, iPAH, COPD and cystic fibrosis. These findings present a new mechanism of protection of lung sGC by the epithelial Hb, and suggests that this protection maybe lost in asthma or COPD where lung Hb is unable to scavenge the NO due to it being heme-deprived.
Collapse
Affiliation(s)
- Mamta P Sumi
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Blair Tupta
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Sanjoy Roychowdhury
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Suzy Comhair
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Kewal Asosingh
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Dennis J Stuehr
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Serpil C Erzurum
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Arnab Ghosh
- Department of Inflammation and Immunity, Lerner Research Institute, The Cleveland Clinic, Cleveland, OH, 44195, USA.
| |
Collapse
|
11
|
Xu A, Han F, Zhang Y, Zhou T, Gao T. Comparative Transcriptomic Analyses Revealed the Effects of Poly (I:C) on the Liver and Spleen of Argyrosomus japonicus. Int J Mol Sci 2022; 23:ijms23179801. [PMID: 36077207 PMCID: PMC9455969 DOI: 10.3390/ijms23179801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Poly (I:C) can work as an immunostimulant and a viral vaccine; however, its functional mechanism in aquatic animals needs to be further investigated. In this study, comparative transcriptomic analyses were performed to investigate the effects of poly (I:C) on Argyrosomus japonicus at 12 h and 48 h postinjection. A total of 194 and 294 differentially expressed genes were obtained in the liver and spleen, respectively. At 12 h, poly (I:C) injection could significantly influence the function of the metabolism-related pathways and immune-related pathways in the liver through the upregulation of the genes GST, LPIN, FOXO1, CYP24A1, ECM1, and SGK1, and the downregulation of the genes IL-1β, CXC19, TNFAIP3, and IRF1. At 48 h, poly (I:C) could enhance the liver energy metabolism by upregulating the genes TXNRD and ECM1, while it also induced some injury in the cells with the downregulation of the genes HBA and CYP24A1. In the spleen, poly (I:C) could regulate the fish immunity and inflammatory response by upregulating the genes DDIT4, C3, EFNA, and MNK, and by downregulating the genes ABCA1, SORT1, TNF, TLR2, IL8, and MHCII at 12 h, and at 48 h, the poly (I:C) had a similar influence as that in the liver. Intersection analyses demonstrated that CYP24A1 and ECM1 were the main functional genes that contributed to the health of the liver. Ten and four genes participated in maintaining the health of the two tissues after 12 h and 48 h, respectively. In summary, our results provided a new insight into ploy (I:C) application in A. japonicus, and it also helped us to better understand the fish response mechanism to the viral vaccine injection.
Collapse
Affiliation(s)
- Anle Xu
- Fisheries College, Zhejiang Ocean University, Zhoushan 316022, China
| | - Fei Han
- Fisheries College, Ocean University of China, Qingdao 266003, China
| | - Yuan Zhang
- Fisheries College, Ocean University of China, Qingdao 266003, China
| | - Tao Zhou
- College of Ocean and Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Tianxiang Gao
- Fisheries College, Zhejiang Ocean University, Zhoushan 316022, China
- Correspondence: ; Tel.: +86-1-35-8707-2063
| |
Collapse
|
12
|
King DR, Sedovy MW, Eaton X, Dunaway LS, Good ME, Isakson BE, Johnstone SR. Cell-To-Cell Communication in the Resistance Vasculature. Compr Physiol 2022; 12:3833-3867. [PMID: 35959755 DOI: 10.1002/cphy.c210040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The arterial vasculature can be divided into large conduit arteries, intermediate contractile arteries, resistance arteries, arterioles, and capillaries. Resistance arteries and arterioles primarily function to control systemic blood pressure. The resistance arteries are composed of a layer of endothelial cells oriented parallel to the direction of blood flow, which are separated by a matrix layer termed the internal elastic lamina from several layers of smooth muscle cells oriented perpendicular to the direction of blood flow. Cells within the vessel walls communicate in a homocellular and heterocellular fashion to govern luminal diameter, arterial resistance, and blood pressure. At rest, potassium currents govern the basal state of endothelial and smooth muscle cells. Multiple stimuli can elicit rises in intracellular calcium levels in either endothelial cells or smooth muscle cells, sourced from intracellular stores such as the endoplasmic reticulum or the extracellular space. In general, activation of endothelial cells results in the production of a vasodilatory signal, usually in the form of nitric oxide or endothelial-derived hyperpolarization. Conversely, activation of smooth muscle cells results in a vasoconstriction response through smooth muscle cell contraction. © 2022 American Physiological Society. Compr Physiol 12: 1-35, 2022.
Collapse
Affiliation(s)
- D Ryan King
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, Virginia Tech, Roanoke, Virginia, USA
| | - Meghan W Sedovy
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, Virginia Tech, Roanoke, Virginia, USA.,Translational Biology, Medicine, and Health Graduate Program, Virginia Tech, Blacksburg, Virginia, USA
| | - Xinyan Eaton
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, Virginia Tech, Roanoke, Virginia, USA
| | - Luke S Dunaway
- Robert M. Berne Cardiovascular Research Centre, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Miranda E Good
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Centre, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Scott R Johnstone
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, Virginia Tech, Roanoke, Virginia, USA.,Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
13
|
Novotny T, Eckhardt A, Doubkova M, Knitlova J, Vondrasek D, Vanaskova E, Ostadal M, Uhlik J, Bacakova L, Musilkova J. The possible role of hypoxia in the affected tissue of relapsed clubfoot. Sci Rep 2022; 12:4462. [PMID: 35292718 PMCID: PMC8924187 DOI: 10.1038/s41598-022-08519-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 03/07/2022] [Indexed: 02/07/2023] Open
Abstract
Our aim was to study the expression of hypoxia-related proteins as a possible regulatory pathway in the contracted side tissue of relapsed clubfoot. We compared the expression of hypoxia-related proteins in the tissue of the contracted (medial) side of relapsed clubfoot, and in the tissue of the non-contracted (lateral) side of relapsed clubfoot. Tissue samples from ten patients were analyzed by immunohistochemistry and image analysis, Real-time PCR and Mass Spectrometry to evaluate the differences in protein composition and gene expression. We found a significant increase in the levels of smooth muscle actin, transforming growth factor-beta, hypoxia-inducible factor 1 alpha, lysyl oxidase, lysyl oxidase-like 2, tenascin C, matrix metalloproteinase-2, matrix metalloproteinase-9, fibronectin, collagen types III and VI, hemoglobin subunit alpha and hemoglobin subunit beta, and an overexpression of ACTA2, FN1, TGFB1, HIF1A and MMP2 genes in the contracted medial side tissue of clubfoot. In the affected tissue, we have identified an increase in the level of hypoxia-related proteins, together with an overexpression of corresponding genes. Our results suggest that the hypoxia-associated pathway is potentially a factor contributing to the etiology of clubfoot relapses, as it stimulates both angioproliferation and fibroproliferation, which are considered to be key factors in the progression and development of relapses.
Collapse
Affiliation(s)
- Tomas Novotny
- Department of Orthopaedics, University J.E. Purkinje and Masaryk Hospital, Usti nad Labem, Czech Republic.,Department of Histology and Embryology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Adam Eckhardt
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| | - Martina Doubkova
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic. .,Second Faculty of Medicine, Charles University, Prague, Czech Republic.
| | - Jarmila Knitlova
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - David Vondrasek
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.,Faculty of Physical Education and Sport, Charles University, Prague, Czech Republic
| | - Eliska Vanaskova
- Department of Orthopaedics, University J.E. Purkinje and Masaryk Hospital, Usti nad Labem, Czech Republic
| | - Martin Ostadal
- Department of Orthopaedics, University Hospital Bulovka, Charles University, Prague, Czech Republic
| | - Jiri Uhlik
- Department of Histology and Embryology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Lucie Bacakova
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jana Musilkova
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
14
|
Akyüz Ünsal Aİ, Key Ö, Güler D, Kurt Omurlu İ, Anık A, Demirci B, Dündar S. Can Complete Blood Count Parameters Predict Retinopathy of Prematurity? Turk J Ophthalmol 2021; 50:87-93. [PMID: 32367699 PMCID: PMC7204894 DOI: 10.4274/tjo.galenos.2019.45313] [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] [Indexed: 12/20/2022] Open
Abstract
Objectives To predict the risk of retinopathy of prematurity (ROP) development according to routine complete blood count (CBC) parameters. Materials and Methods The medical records and CBC results of 150 premature neonates were retrospectively evaluated. As ROP develops 1 month after birth, first month CBC profiles of neonates without ROP (non-ROP), with ROP (ROP group), and those with Type 1, Type 2, and Stage 1+2 ROP were compared. Besides known statistical methods like Student's t-test, logistic regression and classification & regression tree (C&RT) analysis were also done to identify a reliable quantitative predictive parameter. Results Mean gestational age and birth weight of the ROP group (n=99) and non-ROP (n=43) group were 29.39±3.43 and 32.05±2.20 weeks and 1382.44±545.30 and 1691.51±360.84 grams, respectively (p<0.001, p<0.001). Average hemoglobin (Hb) (p<0.001), hematocrit (HCT) (p<0.001), erythrocyte (p=0.005), mean corpuscular hemoglobin (MCH) (p=0.020), and MCH concentration (p=0.019) values of the ROP group were lower than those of the non-ROP group. Leukocyte was higher in the ROP group (p=0.018). Hb [odds ratio (OR)=0.668, 95% confidence interval (CI)=0.555-0.804, p<0.001], red cell distribution width (RDW) (OR=1.282, 95% CI=1.012-1.624, p=0.040), leukocyte (OR=1.157, 95% CI=1.053-1.271, p=0.002), and platelet (OR=0.997, 95% CI: 0.994-0.999, p=0.036) values differed significantly between the two groups. Platelet, MCV, and MCH parameters were found to be lower in the Type 1 ROP group compared to the Stage 1+2 ROP group (p<0.005). MCH was the most prominent predictor (cut-off: 34.43 pg) according to the results of C&RT analysis. Conclusion As Hb plays an important role in oxygen transport, low levels of Hb and especially MCH may cause increased vascular endothelial growth factor secretion from the hypoxic retina, thereby causing ROP. Therefore, the results of this study are encouraging regarding the use of the abovementioned CBC parameters as a simple screening test to predict ROP.
Collapse
Affiliation(s)
- Ayşe İpek Akyüz Ünsal
- Aydın Adnan Menderes University Faculty of Medicine, Department of Ophthalmology, Aydın, Turkey
| | - Özge Key
- Aydın Adnan Menderes University Faculty of Medicine, Department of Ophthalmology, Aydın, Turkey
| | - Duygu Güler
- Aydın Adnan Menderes University Faculty of Medicine, Department of Ophthalmology, Aydın, Turkey
| | - İmran Kurt Omurlu
- Aydın Adnan Menderes University Faculty of Medicine, Department of Biostatistics, Aydın, Turkey
| | - Ayşe Anık
- Aydın Adnan Menderes University Faculty of Medicine, Department of Pediatrics Neonatology Division, Aydın, Turkey
| | - Buket Demirci
- Aydın Adnan Menderes University Faculty of Medicine, Department of Medical Pharmacology, Aydın, Turkey
| | - Sema Dündar
- Aydın Adnan Menderes University Faculty of Medicine, Department of Ophthalmology, Aydın, Turkey
| |
Collapse
|
15
|
Mohr AE, Reiss RA, Beaudet M, Sena J, Naik JS, Walker BR, Sweazea KL. Short-term high fat diet alters genes associated with metabolic and vascular dysfunction during adolescence in rats: a pilot study. PeerJ 2021; 9:e11714. [PMID: 34285833 PMCID: PMC8274493 DOI: 10.7717/peerj.11714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 06/11/2021] [Indexed: 11/20/2022] Open
Abstract
Background Diet-induced metabolic dysfunction precedes multiple disease states including diabetes, heart disease, and vascular dysfunction. The critical role of the vasculature in disease progression is established, yet the details of how gene expression changes in early cardiovascular disease remain an enigma. The objective of the current pilot project was to evaluate whether a quantitative assessment of gene expression within the aorta of six-week old healthy male Sprague-Dawley rats compared to those exhibiting symptoms of metabolic dysfunction could reveal potential mediators of vascular dysfunction. Methods RNA was extracted from the aorta of eight rats from a larger experiment; four animals fed a high-fat diet (HFD) known to induce symptoms of metabolic dysfunction (hypertension, increased adiposity, fasting hyperglycemia) and four age-matched healthy animals fed a standard chow diet (CHOW). The bioinformatic workflow included Gene Ontology (GO) biological process enrichment and network analyses. Results The resulting network contained genes relevant to physiological processes including fat and protein metabolism, oxygen transport, hormone regulation, vascular regulation, thermoregulation, and circadian rhythm. The majority of differentially regulated genes were downregulated, including several associated with circadian clock function. In contrast, leptin and 3-hydroxy-3-methylglutaryl-CoA synthase 2 (Hmgcs2) were notably upregulated. Leptin is involved in several major energy balance signaling pathways and Hmgcs2 is a mitochondrial enzyme that catalyzes the first reaction of ketogenesis. Conclusion Together, these data describe changes in gene expression within the aortic wall of HFD rats with early metabolic dysfunction and highlight potential pathways and signaling intermediates that may impact the development of early vascular dysfunction.
Collapse
Affiliation(s)
- Alex E Mohr
- College of Health Solutions, Arizona State University, Phoenix, AZ, United States
| | - Rebecca A Reiss
- Biology Department, New Mexico Institute of Mining and Technology, Socorro, NM, United States
| | - Monique Beaudet
- Biology Department, New Mexico Institute of Mining and Technology, Socorro, NM, United States
| | - Johnny Sena
- National Center for Genome Resources, Santa Fe, NM, USA
| | - Jay S Naik
- The Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States
| | - Benjimen R Walker
- The Department of Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States
| | - Karen L Sweazea
- College of Health Solutions & School of Life Sciences, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
16
|
Denton CC, Shah P, Suriany S, Liu H, Thuptimdang W, Sunwoo J, Chalacheva P, Veluswamy S, Kato R, Wood JC, Detterich JA, Khoo MCK, Coates TD. Loss of alpha-globin genes in human subjects is associated with improved nitric oxide-mediated vascular perfusion. Am J Hematol 2021; 96:277-281. [PMID: 33247606 PMCID: PMC10653668 DOI: 10.1002/ajh.26058] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 11/23/2020] [Indexed: 01/19/2023]
Abstract
Alpha thalassemia is a hemoglobinopathy due to decreased production of the α-globin protein from loss of up to four α-globin genes, with one or two missing in the trait phenotype. Individuals with sickle cell disease who co-inherit the loss of one or two α-globin genes have been known to have reduced risk of morbid outcomes, but the underlying mechanism is unknown. While α-globin gene deletions affect sickle red cell deformability, the α-globin genes and protein are also present in the endothelial wall of human arterioles and participate in nitric oxide scavenging during vasoconstriction. Decreased production of α-globin due to α-thalassemia trait may thereby limit nitric oxide scavenging and promote vasodilation. To evaluate this potential mechanism, we performed flow-mediated dilation and microvascular post-occlusive reactive hyperemia in 27 human subjects (15 missing one or two α-globin genes and 12 healthy controls). Flow-mediated dilation was significantly higher in subjects with α-trait after controlling for age (P = .0357), but microvascular perfusion was not different between groups. As none of the subjects had anemia or hemolysis, the improvement in vascular function could be attributed to the difference in α-globin gene status. This may explain the beneficial effect of α-globin gene loss in sickle cell disease and suggests that α-globin gene status may play a role in other vascular diseases.
Collapse
Affiliation(s)
- Christopher C. Denton
- Division of Hematology/Oncology, Department of Pediatrics, Childrenʼs Hospital Los Angeles, Los Angeles, California
| | - Payal Shah
- Division of General Pediatrics, Childrenʼs Hospital Los Angeles, Los Angeles, California
| | - Silvie Suriany
- Division of Cardiology, Department of Pediatrics, Children’s Hospital Los Angeles, Los Angeles, California
| | - Honglei Liu
- Division of Cardiology, Department of Pediatrics, Children’s Hospital Los Angeles, Los Angeles, California
| | - Wanwara Thuptimdang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California
| | - John Sunwoo
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California
| | - Patjanaporn Chalacheva
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California
| | - Saranya Veluswamy
- Division of Hematology/Oncology, Department of Pediatrics, Childrenʼs Hospital Los Angeles, Los Angeles, California
| | - Roberta Kato
- Division of Pulmonology, Department of Pediatrics, Children’s Hospital Los Angeles, Los Angeles, California
| | - John C. Wood
- Division of Cardiology, Department of Pediatrics, Children’s Hospital Los Angeles, Los Angeles, California
| | - Jon A. Detterich
- Division of Cardiology, Department of Pediatrics, Children’s Hospital Los Angeles, Los Angeles, California
| | - Michael C. K. Khoo
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California
| | - Thomas D. Coates
- Division of Hematology/Oncology, Department of Pediatrics, Childrenʼs Hospital Los Angeles, Los Angeles, California
| |
Collapse
|
17
|
Wolpe AG, Ruddiman CA, Hall PJ, Isakson BE. Polarized Proteins in Endothelium and Their Contribution to Function. J Vasc Res 2021; 58:65-91. [PMID: 33503620 DOI: 10.1159/000512618] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 10/27/2020] [Indexed: 12/11/2022] Open
Abstract
Protein localization in endothelial cells is tightly regulated to create distinct signaling domains within their tight spatial restrictions including luminal membranes, abluminal membranes, and interendothelial junctions, as well as caveolae and calcium signaling domains. Protein localization in endothelial cells is also determined in part by the vascular bed, with differences between arteries and veins and between large and small arteries. Specific protein polarity and localization is essential for endothelial cells in responding to various extracellular stimuli. In this review, we examine protein localization in the endothelium of resistance arteries, with occasional references to other vessels for contrast, and how that polarization contributes to endothelial function and ultimately whole organism physiology. We highlight the protein localization on the luminal surface, discussing important physiological receptors and the glycocalyx. The protein polarization to the abluminal membrane is especially unique in small resistance arteries with the presence of the myoendothelial junction, a signaling microdomain that regulates vasodilation, feedback to smooth muscle cells, and ultimately total peripheral resistance. We also discuss the interendothelial junction, where tight junctions, adherens junctions, and gap junctions all convene and regulate endothelial function. Finally, we address planar cell polarity, or axial polarity, and how this is regulated by mechanosensory signals like blood flow.
Collapse
Affiliation(s)
- Abigail G Wolpe
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Claire A Ruddiman
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Phillip J Hall
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA, .,Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia, USA,
| |
Collapse
|
18
|
Skeffington KL, Bond AR, Bigotti MG, AbdulGhani S, Iacobazzi D, Kang SL, Heesom KJ, Wilson MC, Stoica S, Martin R, Caputo M, Suleiman MS, Ghorbel MT. Changes in inflammation and oxidative stress signalling pathways in coarcted aorta triggered by bicuspid aortic valve and growth in young children. Exp Ther Med 2020; 20:48. [PMID: 32973936 PMCID: PMC7506967 DOI: 10.3892/etm.2020.9171] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/24/2020] [Indexed: 12/11/2022] Open
Abstract
Neonates with coarctation of the aorta (CoA) combined with a bicuspid aortic valve (BAV) show significant structural differences compared to neonatal CoA patients with a normal tricuspid aortic valve (TAV). These effects are likely to change over time in response to growth. This study investigated proteomic differences between coarcted aortic tissue of BAV and TAV patients in children older than one month. Aortic tissue just proximal to the coarctation site was collected from 10 children (BAV; n=6, 1.9±1.7 years, TAV; n=4, 1.7±1.5 years, (mean ± SEM, P=0.92.) Tissue were snap frozen, proteins extracted, and the extracts used for proteomic and phosphoproteomic analysis using Tandem Mass Tag (TMT) analysis. A total of 1811 protein and 76 phosphoprotein accession numbers were detected, of which 40 proteins and 6 phosphoproteins were significantly differentially expressed between BAV and TAV patients. Several canonical pathways involved in inflammation demonstrated enriched protein expression, including acute phase response signalling, EIF2 signalling and macrophage production of IL12 and reactive oxygen species. Acute phase response signalling also demonstrated enriched phosphoprotein expression, as did Th17 activation. Other pathways with significantly enriched protein expression include degradation of superoxide radicals and several pathways involved in apoptosis. This work suggests that BAV CoA patients older than one month have an altered proteome consistent with changes in inflammation, apoptosis and oxidative stress compared to TAV CoA patients of the same age. There is no evidence of structural differences, suggesting the pathology associated with BAV evolves with age in paediatric CoA patients.
Collapse
Affiliation(s)
- Katie L Skeffington
- Bristol Heart Institute, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Andrew R Bond
- Bristol Heart Institute, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - M Giulia Bigotti
- Bristol Heart Institute, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Safa AbdulGhani
- Bristol Heart Institute, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK.,Department of Congenital Heart Disease, Bristol Children's Hospital, Bristol BS2 8JB, UK
| | - Dominga Iacobazzi
- Bristol Heart Institute, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Sok-Leng Kang
- Department of Physiology, Faculty of Medicine, Al-Quds University, P.O Box 89, Abu Dis, Palestine
| | - Kate J Heesom
- Proteomics Facility, University of Bristol, Bristol BS8 1RJ, UK
| | | | - Serban Stoica
- Department of Physiology, Faculty of Medicine, Al-Quds University, P.O Box 89, Abu Dis, Palestine
| | - Robin Martin
- Department of Physiology, Faculty of Medicine, Al-Quds University, P.O Box 89, Abu Dis, Palestine
| | - Massimo Caputo
- Bristol Heart Institute, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK.,Department of Physiology, Faculty of Medicine, Al-Quds University, P.O Box 89, Abu Dis, Palestine
| | - M Saadeh Suleiman
- Bristol Heart Institute, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Mohamed T Ghorbel
- Bristol Heart Institute, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| |
Collapse
|
19
|
Abstract
Of the 21 members of the connexin family, 4 (Cx37, Cx40, Cx43, and Cx45) are expressed in the endothelium and/or smooth muscle of intact blood vessels to a variable and dynamically regulated degree. Full-length connexins oligomerize and form channel structures connecting the cytosol of adjacent cells (gap junctions) or the cytosol with the extracellular space (hemichannels). The different connexins vary mainly with regard to length and sequence of their cytosolic COOH-terminal tails. These COOH-terminal parts, which in the case of Cx43 are also translated as independent short isoforms, are involved in various cellular signaling cascades and regulate cell functions. This review focuses on channel-dependent and -independent effects of connexins in vascular cells. Channels play an essential role in coordinating and synchronizing endothelial and smooth muscle activity and in their interplay, in the control of vasomotor actions of blood vessels including endothelial cell reactivity to agonist stimulation, nitric oxide-dependent dilation, and endothelial-derived hyperpolarizing factor-type responses. Further channel-dependent and -independent roles of connexins in blood vessel function range from basic processes of vascular remodeling and angiogenesis to vascular permeability and interactions with leukocytes with the vessel wall. Together, these connexin functions constitute an often underestimated basis for the enormous plasticity of vascular morphology and function enabling the required dynamic adaptation of the vascular system to varying tissue demands.
Collapse
Affiliation(s)
- Ulrich Pohl
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Planegg-Martinsried, Germany; Biomedical Centre, Cardiovascular Physiology, LMU Munich, Planegg-Martinsried, Germany; German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany; and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
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
|
Garcia V, Park EJ, Siragusa M, Frohlich F, Mahfuzul Haque M, Pascale JV, Heberlein KR, Isakson BE, Stuehr DJ, Sessa WC. Unbiased proteomics identifies plasminogen activator inhibitor-1 as a negative regulator of endothelial nitric oxide synthase. Proc Natl Acad Sci U S A 2020; 117:9497-9507. [PMID: 32300005 PMCID: PMC7196906 DOI: 10.1073/pnas.1918761117] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) produced by endothelial nitric oxide synthase (eNOS) is a critical mediator of vascular function. eNOS is tightly regulated at various levels, including transcription, co- and posttranslational modifications, and by various protein-protein interactions. Using stable isotope labeling with amino acids in cell culture (SILAC) and mass spectrometry (MS), we identified several eNOS interactors, including the protein plasminogen activator inhibitor-1 (PAI-1). In cultured human umbilical vein endothelial cells (HUVECs), PAI-1 and eNOS colocalize and proximity ligation assays demonstrate a protein-protein interaction between PAI-1 and eNOS. Knockdown of PAI-1 or eNOS eliminates the proximity ligation assay (PLA) signal in endothelial cells. Overexpression of eNOS and HA-tagged PAI-1 in COS7 cells confirmed the colocalization observations in HUVECs. Furthermore, the source of intracellular PAI-1 interacting with eNOS was shown to be endocytosis derived. The interaction between PAI-1 and eNOS is a direct interaction as supported in experiments with purified proteins. Moreover, PAI-1 directly inhibits eNOS activity, reducing NO synthesis, and the knockdown or antagonism of PAI-1 increases NO bioavailability. Taken together, these findings place PAI-1 as a negative regulator of eNOS and disruptions in eNOS-PAI-1 binding promote increases in NO production and enhance vasodilation in vivo.
Collapse
Affiliation(s)
- Victor Garcia
- Vascular Biology and Therapeutics Program, Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520
| | - Eon Joo Park
- Vascular Biology and Therapeutics Program, Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520
| | - Mauro Siragusa
- Institute for Vascular Signaling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Florian Frohlich
- Vascular Biology and Therapeutics Program, Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520
- Department of Biology/Chemistry, Molecular Membrane Biology Section, University of Osnabrück, 49076 Osnabrück, Germany
| | - Mohammad Mahfuzul Haque
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Jonathan V Pascale
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595
| | - Katherine R Heberlein
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Dennis J Stuehr
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - William C Sessa
- Vascular Biology and Therapeutics Program, Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520;
| |
Collapse
|
22
|
Corliss BA, Delalio LJ, Stevenson Keller TC, Keller AS, Keller DA, Corliss BH, Beers JM, Peirce SM, Isakson BE. Vascular Expression of Hemoglobin Alpha in Antarctic Icefish Supports Iron Limitation as Novel Evolutionary Driver. Front Physiol 2019; 10:1389. [PMID: 31780954 PMCID: PMC6861181 DOI: 10.3389/fphys.2019.01389] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 10/24/2019] [Indexed: 12/16/2022] Open
Abstract
Frigid temperatures of the Southern Ocean are known to be an evolutionary driver in Antarctic fish. For example, many fish have reduced red blood cell (RBC) concentration to minimize vascular resistance. Via the oxygen-carrying protein hemoglobin, RBCs contain the vast majority of the body's iron, which is known to be a limiting nutrient in marine ecosystems. Since lower RBC levels also lead to reduced iron requirements, we hypothesize that low iron availability was an additional evolutionary driver of Antarctic fish speciation. Antarctic Icefish of the family Channichthyidae are known to have an extreme alteration of iron metabolism due to loss of RBCs and two iron-binding proteins, hemoglobin and myoglobin. Loss of hemoglobin is considered a maladaptive trait allowed by relaxation of predator selection since extreme adaptations are required to compensate for the loss of oxygen-carrying capacity. However, iron dependency minimization may have driven hemoglobin loss instead of a random evolutionary event. Given the variety of functions that hemoglobin serves in the endothelium, we suspected the protein corresponding to the 3' truncated Hbα fragment (Hbα-3'f) that was not genetically excluded by icefish may still be expressed as a protein. Using whole mount confocal microscopy, we show that Hbα-3'f is expressed in the vascular endothelium of icefish retina, suggesting this Hbα fragment may still serve an important role in the endothelium. These observations support a novel hypothesis that iron minimization could have influenced icefish speciation with the loss of the iron-binding portion of Hbα in Hbα-3'f, as well as hemoglobin β and myoglobin.
Collapse
Affiliation(s)
- Bruce A Corliss
- Biomedical Engineering Department, University of Virginia, Charlottesville, VA, United States
| | - Leon J Delalio
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - T C Stevenson Keller
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States.,Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Alexander S Keller
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | | | - Bruce H Corliss
- Graduate School of Oceanography, University of Rhode Island, Kingston, RI, United States
| | - Jody M Beers
- Department of Biology, College of Charleston, Charleston, SC, United States
| | - Shayn M Peirce
- Biomedical Engineering Department, University of Virginia, Charlottesville, VA, United States
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States.,Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
23
|
Pogoda K, Kameritsch P, Mannell H, Pohl U. Connexins in the control of vasomotor function. Acta Physiol (Oxf) 2019; 225:e13108. [PMID: 29858558 DOI: 10.1111/apha.13108] [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: 09/12/2017] [Revised: 05/25/2018] [Accepted: 05/28/2018] [Indexed: 12/13/2022]
Abstract
Vascular endothelial cells, as well as smooth muscle cells, show heterogeneity with regard to their receptor expression and reactivity. For the vascular wall to act as a functional unit, the various cells' responses require integration. Such an integration is not only required for a homogeneous response of the vascular wall, but also for the vasomotor behaviour of consecutive segments of the microvascular arteriolar tree. As flow resistances of individual sections are connected in series, sections require synchronization and coordination to allow effective changes of conductivity and blood flow. A prerequisite for the local coordination of individual vascular cells and different sections of an arteriolar tree is intercellular communication. Connexins are involved in a dual manner in this coordination. (i) By forming gap junctions between cells, they allow an intercellular exchange of signalling molecules and electrical currents. In particular, the spread of electrical currents allows for coordination of cell responses over longer distances. (ii) Connexins are able to interact with other proteins to form signalling complexes. In this way, they can modulate and integrate individual cells' responses also in a channel-independent manner. This review outlines mechanisms allowing the vascular connexins to exert their coordinating function and to regulate the vasomotor reactions of blood vessels both locally, and in vascular networks. Wherever possible, we focus on the vasomotor behaviour of small vessels and arterioles which are the main vessels determining vascular resistance, blood pressure and local blood flow.
Collapse
Affiliation(s)
- K. Pogoda
- Walter-Brendel-Centre of Experimental Medicine; University Hospital; LMU Munich; Munich Germany
- Biomedical Center; Cardiovascular Physiology; LMU Munich; Munich Germany
- DZHK (German Center for Cardiovascular Research); Partner Site Munich Heart Alliance; Munich Germany
| | - P. Kameritsch
- Walter-Brendel-Centre of Experimental Medicine; University Hospital; LMU Munich; Munich Germany
- Biomedical Center; Cardiovascular Physiology; LMU Munich; Munich Germany
- DZHK (German Center for Cardiovascular Research); Partner Site Munich Heart Alliance; Munich Germany
| | - H. Mannell
- Walter-Brendel-Centre of Experimental Medicine; University Hospital; LMU Munich; Munich Germany
- Biomedical Center; Cardiovascular Physiology; LMU Munich; Munich Germany
| | - U. Pohl
- Walter-Brendel-Centre of Experimental Medicine; University Hospital; LMU Munich; Munich Germany
- Biomedical Center; Cardiovascular Physiology; LMU Munich; Munich Germany
- DZHK (German Center for Cardiovascular Research); Partner Site Munich Heart Alliance; Munich Germany
- Munich Cluster for Systems Neurology (SyNergy); Munich Germany
| |
Collapse
|
24
|
Knight AK, Dunlop AL, Kilaru V, Cobb D, Corwin EJ, Conneely KN, Smith AK. Characterization of gene expression changes over healthy term pregnancies. PLoS One 2018; 13:e0204228. [PMID: 30303981 PMCID: PMC6179206 DOI: 10.1371/journal.pone.0204228] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 08/14/2018] [Indexed: 12/20/2022] Open
Abstract
During pregnancy, women experience numerous physiological changes but, to date, there is limited published data that characterize accompanying changes in gene expression over pregnancy. This study sought to characterize the complexity of the transcriptome over the course of pregnancy among women with healthy pregnancies. Subjects provided a venous blood sample during early (6-15 weeks) and late (22-33 weeks) pregnancy, which was used to isolate peripheral blood mononuclear cells prior to RNA extraction. Gene expression was examined for 63 women with uncomplicated, term deliveries. We evaluated the association between weeks gestation at sample collection and expression of each transcript. Of the 16,311 transcripts evaluated, 439 changed over pregnancy after a Bonferroni correction to account for multiple comparisons. Genes whose expression increased over pregnancy were associated with oxygen transport, the immune system, and host response to bacteria. Characterization of changes in the transcriptome over the course of healthy term pregnancies may enable the identification of genes whose expression predicts complications or adverse outcomes of pregnancy.
Collapse
Affiliation(s)
- Anna K. Knight
- Genetics and Molecular Biology Program, Emory University, Atlanta, GA, United States of America
| | - Anne L. Dunlop
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, United States of America
| | - Varun Kilaru
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Dawayland Cobb
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Elizabeth J. Corwin
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, United States of America
| | - Karen N. Conneely
- Genetics and Molecular Biology Program, Emory University, Atlanta, GA, United States of America
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Alicia K. Smith
- Genetics and Molecular Biology Program, Emory University, Atlanta, GA, United States of America
- Department of Gynecology and Obstetrics, Emory University School of Medicine, Atlanta, GA, United States of America
- Department of Psychiatry, Emory University School of Medicine, Atlanta, GA, United States of America
- * E-mail:
| |
Collapse
|
25
|
Lechauve C, Butcher JT, Freiwan A, Biwer LA, Keith JM, Good ME, Ackerman H, Tillman HS, Kiger L, Isakson BE, Weiss MJ. Endothelial cell α-globin and its molecular chaperone α-hemoglobin-stabilizing protein regulate arteriolar contractility. J Clin Invest 2018; 128:5073-5082. [PMID: 30295646 DOI: 10.1172/jci99933] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 08/21/2018] [Indexed: 12/18/2022] Open
Abstract
Arteriolar endothelial cell-expressed (EC-expressed) α-globin binds endothelial NOS (eNOS) and degrades its enzymatic product, NO, via dioxygenation, thereby lessening the vasodilatory effects of NO on nearby vascular smooth muscle. Although this reaction potentially affects vascular physiology, the mechanisms that regulate α-globin expression and dioxygenase activity in ECs are unknown. Without β-globin, α-globin is unstable and cytotoxic, particularly in its oxidized form, which is generated by dioxygenation and recycled via endogenous reductases. We show that the molecular chaperone α-hemoglobin-stabilizing protein (AHSP) promotes arteriolar α-globin expression in vivo and facilitates its reduction by eNOS. In Ahsp-/- mice, EC α-globin was decreased by 70%. Ahsp-/- and Hba1-/- mice exhibited similar evidence of increased vascular NO signaling, including arteriolar dilation, blunted α1-adrenergic vasoconstriction, and reduced blood pressure. Purified α-globin bound eNOS or AHSP, but not both together. In ECs in culture, eNOS or AHSP enhanced α-globin expression posttranscriptionally. However, only AHSP prevented oxidized α-globin precipitation in solution. Finally, eNOS reduced AHSP-bound α-globin approximately 6-fold faster than did the major erythrocyte hemoglobin reductases (cytochrome B5 reductase plus cytochrome B5). Our data support a model whereby redox-sensitive shuttling of EC α-globin between AHSP and eNOS regulates EC NO degradation and vascular tone.
Collapse
Affiliation(s)
- Christophe Lechauve
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Joshua T Butcher
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Abdullah Freiwan
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Lauren A Biwer
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Julia M Keith
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Miranda E Good
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Hans Ackerman
- Laboratory of Malaria and Vector Research, National Institutes of Allergy and Infectious Diseases, Rockville, Maryland, USA
| | - Heather S Tillman
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Mitchell J Weiss
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
26
|
Alvarez RA, Miller MP, Hahn SA, Galley JC, Bauer E, Bachman T, Hu J, Sembrat J, Goncharov D, Mora AL, Rojas M, Goncharova E, Straub AC. Targeting Pulmonary Endothelial Hemoglobin α Improves Nitric Oxide Signaling and Reverses Pulmonary Artery Endothelial Dysfunction. Am J Respir Cell Mol Biol 2017; 57:733-744. [PMID: 28800253 PMCID: PMC5765416 DOI: 10.1165/rcmb.2016-0418oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 07/12/2017] [Indexed: 12/13/2022] Open
Abstract
Pulmonary hypertension is characterized by pulmonary endothelial dysfunction. Previous work showed that systemic artery endothelial cells (ECs) express hemoglobin (Hb) α to control nitric oxide (NO) diffusion, but the role of this system in pulmonary circulation has not been evaluated. We hypothesized that up-regulation of Hb α in pulmonary ECs contributes to NO depletion and pulmonary vascular dysfunction in pulmonary hypertension. Primary distal pulmonary arterial vascular smooth muscle cells, lung tissue sections from unused donor (control) and idiopathic pulmonary artery (PA) hypertension lungs, and rat and mouse models of SU5416/hypoxia-induced pulmonary hypertension (PH) were used. Immunohistochemical, immunocytochemical, and immunoblot analyses and transfection, infection, DNA synthesis, apoptosis, migration, cell count, and protein activity assays were performed in this study. Cocultures of human pulmonary microvascular ECs and distal pulmonary arterial vascular smooth muscle cells, lung tissue from control and pulmonary hypertensive lungs, and a mouse model of chronic hypoxia-induced PH were used. Immunohistochemical, immunoblot analyses, spectrophotometry, and blood vessel myography experiments were performed in this study. We find increased expression of Hb α in pulmonary endothelium from humans and mice with PH compared with controls. In addition, we show up-regulation of Hb α in human pulmonary ECs cocultured with PA smooth muscle cells in hypoxia. We treated pulmonary ECs with a Hb α mimetic peptide that disrupts the association of Hb α with endothelial NO synthase, and found that cells treated with the peptide exhibited increased NO signaling compared with a scrambled peptide. Myography experiments using pulmonary arteries from hypoxic mice show that the Hb α mimetic peptide enhanced vasodilation in response to acetylcholine. Our findings reveal that endothelial Hb α functions as an endogenous scavenger of NO in the pulmonary endothelium. Targeting this pathway may offer a novel therapeutic target to increase endogenous levels of NO in PH.
Collapse
MESH Headings
- Animals
- Biomimetic Materials/pharmacology
- Coculture Techniques
- Disease Models, Animal
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Female
- Hemoglobin A/biosynthesis
- Humans
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/physiopathology
- Male
- Mice
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Nitric Oxide/metabolism
- Nitric Oxide Synthase Type III/genetics
- Nitric Oxide Synthase Type III/metabolism
- Peptides/pharmacology
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/physiopathology
- Rats
- Up-Regulation/drug effects
- Vasodilation/drug effects
Collapse
Affiliation(s)
- Roger A. Alvarez
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Miller School of Medicine, University of Miami, Miami, Florida; and
| | | | | | - Joseph C. Galley
- Heart, Lung, Blood, and Vascular Medicine Institute
- Department of Pharmacology and Chemical Biology
| | | | - Timothy Bachman
- Heart, Lung, Blood, and Vascular Medicine Institute
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jian Hu
- Heart, Lung, Blood, and Vascular Medicine Institute
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - John Sembrat
- Heart, Lung, Blood, and Vascular Medicine Institute
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Dmitry Goncharov
- Heart, Lung, Blood, and Vascular Medicine Institute
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ana L. Mora
- Heart, Lung, Blood, and Vascular Medicine Institute
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Mauricio Rojas
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Elena Goncharova
- Heart, Lung, Blood, and Vascular Medicine Institute
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Adam C. Straub
- Heart, Lung, Blood, and Vascular Medicine Institute
- Department of Pharmacology and Chemical Biology
| |
Collapse
|
27
|
沈 安, 胡 水, 黄 敬, 郑 德, 胡 兆. [Screening and verification of plasma biomarkers for stable angina pectoris: a differential proteomic analysis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2017; 37:1370-1374. [PMID: 29070468 PMCID: PMC6743951 DOI: 10.3969/j.issn.1673-4254.2017.10.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Indexed: 06/07/2023]
Abstract
OBJECTIVE To compare and analyze the differentially expressed plasma proteome between patients with stable angina pectoris (SAP) and healthy donors to identify the biomarkers for early diagnosis of SAP. METHODS Plasma samples from 60 patients with SAP and 60 healthy controls were collected. Twenty samples (100 mL each) randomly selected from each group were pooled and after removing high-abundance proteins from the pooled plasma, two-dimensional gel electrophoresis (2DE) was performed to isolate the total proteins. The protein spots with more than 2 fold changes were selected after 2D analysis using software, and the differentially expressed proteins were identified by MALDI TOF/TOF mass spectrometer. ELISA was performed to detect hemoglobin subunit delta (HBD) levels in 40 randomly selected samples from each group for verification of the results of 2DE. RESULTS A total of 7 differentially expressed proteins were found in plasma samples from patients with SAP, including 3 up regulated proteins (serum albumin, hemoglobin subunit alpha and hemoglobin subunit delta,) and 4 down?regulated ones (apolipoprotein L1, apolipoprotein C3, apolipoprotein E and complement C4B). ELISA results showed that HBD level was increased in SAP plasma, which was consistent with the results of 2DE. CONCLUSION Patients with SAP have different plasma protein profiles from those of healthy controls, and HBD may serve as a potential specific biomarker for early diagnosis of SAP.
Collapse
Affiliation(s)
- 安娜 沈
- 南方医科大学第三附属医院 心血管内科, 广东 广州 510630Department of Cardiovascular Medicine, Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - 水旺 胡
- 南方医科大学病理生理学教研室, 广东 广州 510515Department of Clinical Laboratory, Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - 敬 黄
- 南方医科大学第三附属医院 检验科, 广东 广州 510630Department of Pathophysiology, Southern Medical University, Guangzhou 510515, China
| | - 德仲 郑
- 南方医科大学第三附属医院 心血管内科, 广东 广州 510630Department of Cardiovascular Medicine, Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - 兆霆 胡
- 南方医科大学第三附属医院 心血管内科, 广东 广州 510630Department of Cardiovascular Medicine, Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| |
Collapse
|
28
|
沈 安, 胡 水, 黄 敬, 郑 德, 胡 兆. [Screening and verification of plasma biomarkers for stable angina pectoris: a differential proteomic analysis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2017; 37:1370-1374. [PMID: 29070468 PMCID: PMC6743951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Indexed: 07/30/2024]
Abstract
OBJECTIVE To compare and analyze the differentially expressed plasma proteome between patients with stable angina pectoris (SAP) and healthy donors to identify the biomarkers for early diagnosis of SAP. METHODS Plasma samples from 60 patients with SAP and 60 healthy controls were collected. Twenty samples (100 mL each) randomly selected from each group were pooled and after removing high-abundance proteins from the pooled plasma, two-dimensional gel electrophoresis (2DE) was performed to isolate the total proteins. The protein spots with more than 2 fold changes were selected after 2D analysis using software, and the differentially expressed proteins were identified by MALDI TOF/TOF mass spectrometer. ELISA was performed to detect hemoglobin subunit delta (HBD) levels in 40 randomly selected samples from each group for verification of the results of 2DE. RESULTS A total of 7 differentially expressed proteins were found in plasma samples from patients with SAP, including 3 up regulated proteins (serum albumin, hemoglobin subunit alpha and hemoglobin subunit delta,) and 4 down?regulated ones (apolipoprotein L1, apolipoprotein C3, apolipoprotein E and complement C4B). ELISA results showed that HBD level was increased in SAP plasma, which was consistent with the results of 2DE. CONCLUSION Patients with SAP have different plasma protein profiles from those of healthy controls, and HBD may serve as a potential specific biomarker for early diagnosis of SAP.
Collapse
Affiliation(s)
- 安娜 沈
- 南方医科大学第三附属医院 心血管内科, 广东 广州 510630Department of Cardiovascular Medicine, Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - 水旺 胡
- 南方医科大学病理生理学教研室, 广东 广州 510515Department of Clinical Laboratory, Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - 敬 黄
- 南方医科大学第三附属医院 检验科, 广东 广州 510630Department of Pathophysiology, Southern Medical University, Guangzhou 510515, China
| | - 德仲 郑
- 南方医科大学第三附属医院 心血管内科, 广东 广州 510630Department of Cardiovascular Medicine, Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - 兆霆 胡
- 南方医科大学第三附属医院 心血管内科, 广东 广州 510630Department of Cardiovascular Medicine, Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| |
Collapse
|
29
|
Wang R, Pan Q, Kuebler WM, Li JKJ, Pries AR, Ning G. Modeling of pulsatile flow-dependent nitric oxide regulation in a realistic microvascular network. Microvasc Res 2017; 113:40-49. [PMID: 28478072 DOI: 10.1016/j.mvr.2017.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 04/14/2017] [Accepted: 05/02/2017] [Indexed: 11/29/2022]
Abstract
Hemodynamic pulsatility has been reported to regulate microcirculatory function. To quantitatively assess the impact of flow pulsatility on the microvasculature, a mathematical model was first developed to simulate the regulation of NO production by pulsatile flow in the microcirculation. Shear stress and pressure pulsatility were selected as regulators of endothelial NO production and NO-dependent vessel dilation as feedback to control microvascular hemodynamics. The model was then applied to a real microvascular network of the rat mesentery consisting of 546 microvessels. As compared to steady flow conditions, pulsatile flow increased the average NO concentration in arterioles from 256.8±93.1nM to 274.8±101.1nM (P<0.001), with a corresponding increase in vessel dilation by approximately 7% from 27.5±10.6% to 29.4±11.4% (P<0.001). In contrast, NO concentration and vessel size showed a far lesser increase (about 1.7%) in venules under pulsatile flow as compared to steady flow conditions. Network perfusion and flow heterogeneity were improved under pulsatile flow conditions, and vasodilation within the network was more sensitive to heart rate changes than pulse pressure amplitude. The proposed model simulates the role of flow pulsatility in the regulation of a complex microvascular network in terms of NO concentration and hemodynamics under varied physiological conditions.
Collapse
Affiliation(s)
- Ruofan Wang
- Department of Biomedical Engineering, MOE Key Laboratory of Biomedical Engineering, Zhejiang University, 38 Zheda Road, Hangzhou 310027, China
| | - Qing Pan
- College of Information Engineering, Zhejiang University of Technology, 288 Liuhe Road, Hangzhou 310023, China
| | - Wolfgang M Kuebler
- Keenan Research Centre for Biomedical Science of St. Michael's, University of Toronto, 30 Bond Street, Toronto M5B 1W8, Canada; Department of Physiology and Center for Cardiovascular Research, Charité Universitätsmediz in Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - John K-J Li
- Cardiovascular Research, Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, USA
| | - Axel R Pries
- Department of Physiology and Center for Cardiovascular Research, Charité Universitätsmediz in Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Gangmin Ning
- Department of Biomedical Engineering, MOE Key Laboratory of Biomedical Engineering, Zhejiang University, 38 Zheda Road, Hangzhou 310027, China.
| |
Collapse
|
30
|
Etyang AO, Khayeka-Wandabwa C, Kapesa S, Muthumbi E, Odipo E, Wamukoya M, Ngomi N, Haregu T, Kyobutungi C, Tendwa M, Makale J, Macharia A, Cruickshank JK, Smeeth L, Scott JAG, Williams TN. Blood Pressure and Arterial Stiffness in Kenyan Adolescents With α +Thalassemia. J Am Heart Assoc 2017; 6:JAHA.117.005613. [PMID: 28381468 PMCID: PMC5533038 DOI: 10.1161/jaha.117.005613] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Background Recent studies have discovered that α‐globin is expressed in blood vessel walls where it plays a role in regulating vascular tone. We tested the hypothesis that blood pressure (BP) might differ between normal individuals and those with α+thalassemia, in whom the production of α‐globin is reduced. Methods and Results The study was conducted in Nairobi, Kenya, among 938 adolescents aged 11 to 17 years. Twenty‐four‐hour ambulatory BP monitoring and arterial stiffness measurements were performed using an arteriograph device. We genotyped for α+thalassemia by polymerase chain reaction. Complete data for analysis were available for 623 subjects; 223 (36%) were heterozygous (−α/αα) and 47 (8%) were homozygous (−α/−α) for α+thalassemia whereas the remaining 353 (55%) were normal (αα/αα). Mean 24‐hour systolic BP ±SD was 118±12 mm Hg in αα/αα, 117±11 mm Hg in −α/αα, and 118±11 mm Hg in −α/−α subjects, respectively. Mean 24‐hour diastolic BP ±SD in these groups was 64±8, 63±7, and 65±8 mm Hg, respectively. Mean pulse wave velocity (PWV)±SD was 7±0.8, 7±0.8, and 7±0.7 ms−1, respectively. No differences were observed in PWV and any of the 24‐hour ambulatory BP monitoring‐derived measures between those with and without α+thalassemia. Conclusions These data suggest that the presence of α+thalassemia does not affect BP and/or arterial stiffness in Kenyan adolescents.
Collapse
Affiliation(s)
- Anthony O Etyang
- KEMRI-Wellcome Trust Research Program, Kilifi, Kenya .,London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | | | | | | - Emily Odipo
- KEMRI-Wellcome Trust Research Program, Kilifi, Kenya
| | | | - Nicholas Ngomi
- African Population and Health Research Centre, Nairobi, Kenya
| | - Tilahun Haregu
- African Population and Health Research Centre, Nairobi, Kenya
| | | | | | | | - Alex Macharia
- KEMRI-Wellcome Trust Research Program, Kilifi, Kenya
| | | | - Liam Smeeth
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - J Anthony G Scott
- KEMRI-Wellcome Trust Research Program, Kilifi, Kenya.,London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Thomas N Williams
- KEMRI-Wellcome Trust Research Program, Kilifi, Kenya.,Imperial College, London, United Kingdom
| |
Collapse
|
31
|
Kollau A, Russwurm M, Neubauer A, Rechberger G, Schmidt K, Koesling D, Fassett J, Schrammel A, Mayer B. Scavenging of nitric oxide by hemoglobin in the tunica media of porcine coronary arteries. Nitric Oxide 2016; 54:8-14. [PMID: 26805578 PMCID: PMC5933522 DOI: 10.1016/j.niox.2016.01.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 01/19/2016] [Accepted: 01/20/2016] [Indexed: 11/23/2022]
Abstract
Scavenging of nitric oxide (NO) often interferes with studies on NO signaling in cell-free preparations. We observed that formation of cGMP by NO-stimulated purified soluble guanylate cyclase (sGC) was virtually abolished in the presence of cytosolic preparations of porcine coronary arteries, with the scavenging activity localized in the tunica media (smooth muscle layer). Electrochemical measurement of NO release from a donor compound and light absorbance spectroscopy showed that cytosolic preparations contained a reduced heme protein that scavenged NO. This protein, which reacted with anti-human hemoglobin antibodies, was efficiently removed from the preparations by haptoglobin affinity chromatography. The cleared cytosols showed only minor scavenging of NO according to electrochemical measurements and did not decrease cGMP formation by NO-stimulated sGC. In contrast, the column flow-through caused a nearly 2-fold increase of maximal sGC activity (from 33.1 ± 1.6 to 54.9 ± 2.2 μmol × min(-1) × mg(-1)). The proteins retained on the affinity column were identified as hemoglobin α and β subunits. The results indicate that hemoglobin, presumably derived from vasa vasorum erythrocytes, is present and scavenges NO in preparations of porcine coronary artery smooth muscle. Selective removal of hemoglobin-mediated scavenging unmasked stimulation of maximal NO-stimulated sGC activity by a soluble factor expressed in vascular tissue.
Collapse
Affiliation(s)
- Alexander Kollau
- Institute of Pharmaceutical Sciences, Department of Pharmacology and Toxicology, University of Graz, Austria
| | - Michael Russwurm
- Department of Pharmacology and Toxicology, Ruhr University Bochum, Germany
| | - Andrea Neubauer
- Institute of Pharmaceutical Sciences, Department of Pharmacology and Toxicology, University of Graz, Austria
| | - Gerald Rechberger
- Institute of Molecular Biosciences, NAWI Graz, University of Graz, Austria; Omics-Center, BioTechMed-Graz, Austria
| | - Kurt Schmidt
- Institute of Pharmaceutical Sciences, Department of Pharmacology and Toxicology, University of Graz, Austria
| | - Doris Koesling
- Department of Pharmacology and Toxicology, Ruhr University Bochum, Germany
| | - John Fassett
- Institute of Pharmaceutical Sciences, Department of Pharmacology and Toxicology, University of Graz, Austria
| | - Astrid Schrammel
- Institute of Pharmaceutical Sciences, Department of Pharmacology and Toxicology, University of Graz, Austria
| | - Bernd Mayer
- Institute of Pharmaceutical Sciences, Department of Pharmacology and Toxicology, University of Graz, Austria.
| |
Collapse
|
32
|
Shu X, Keller TCS, Begandt D, Butcher JT, Biwer L, Keller AS, Columbus L, Isakson BE. Endothelial nitric oxide synthase in the microcirculation. Cell Mol Life Sci 2015; 72:4561-75. [PMID: 26390975 PMCID: PMC4628887 DOI: 10.1007/s00018-015-2021-0] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 07/21/2015] [Accepted: 08/11/2015] [Indexed: 02/07/2023]
Abstract
Endothelial nitric oxide synthase (eNOS, NOS3) is responsible for producing nitric oxide (NO)--a key molecule that can directly (or indirectly) act as a vasodilator and anti-inflammatory mediator. In this review, we examine the structural effects of regulation of the eNOS enzyme, including post-translational modifications and subcellular localization. After production, NO diffuses to surrounding cells with a variety of effects. We focus on the physiological role of NO and NO-derived molecules, including microvascular effects on vessel tone and immune response. Regulation of eNOS and NO action is complicated; we address endogenous and exogenous mechanisms of NO regulation with a discussion of pharmacological agents used in clinical and laboratory settings and a proposed role for eNOS in circulating red blood cells.
Collapse
Affiliation(s)
- Xiaohong Shu
- College of Pharmacy, Dalian Medical University, Dalian, 116044, China
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, P.O. Box 801394, Charlottesville, VA, 22908, USA
| | - T C Stevenson Keller
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, P.O. Box 801394, Charlottesville, VA, 22908, USA
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, USA
| | - Daniela Begandt
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, P.O. Box 801394, Charlottesville, VA, 22908, USA
| | - Joshua T Butcher
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, P.O. Box 801394, Charlottesville, VA, 22908, USA
| | - Lauren Biwer
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, P.O. Box 801394, Charlottesville, VA, 22908, USA
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, USA
| | - Alexander S Keller
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, P.O. Box 801394, Charlottesville, VA, 22908, USA
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, USA
| | - Linda Columbus
- Department of Chemistry, University of Virginia, Charlottesville, USA
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, P.O. Box 801394, Charlottesville, VA, 22908, USA.
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, USA.
| |
Collapse
|
33
|
Wang Z, Teoh SH, Hong M, Luo F, Teo EY, Chan JKY, Thian ES. Dual-Microstructured Porous, Anisotropic Film for Biomimicking of Endothelial Basement Membrane. ACS APPLIED MATERIALS & INTERFACES 2015; 7:13445-13456. [PMID: 26030777 DOI: 10.1021/acsami.5b02464] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Human endothelial basement membrane (BM) plays a pivotal role in vascular development and homeostasis. Here, a bioresponsive film with dual-microstructured geometries was engineered to mimic the structural roles of the endothelial BM in developing vessels, for vascular tissue engineering (TE) application. Flexible poly(ε-caprolactone) (PCL) thin film was fabricated with microscale anisotropic ridges/grooves and through-holes using a combination of uniaxial thermal stretching and direct laser perforation, respectively. Through optimizing the interhole distance, human mesenchymal stem cells (MSCs) cultured on the PCL film's ridges/grooves obtained an intact cell alignment efficiency. With prolonged culturing for 8 days, these cells formed aligned cell multilayers as found in native tunica media. By coculturing human umbilical vein endothelial cells (HUVECs) on the opposite side of the film, HUVECs were observed to build up transmural interdigitation cell-cell contact with MSCs via the through-holes, leading to a rapid endothelialization on the PCL film surface. Furthermore, vascular tissue construction based on the PCL film showed enhanced bioactivity with an elevated total nitric oxide level as compared to single MSCs or HUVECs culturing and indirect MSCs/HUVECs coculturing systems. These results suggested that the dual-microstructured porous and anisotropic film could simulate the structural roles of endothelial BM for vascular reconstruction, with aligned stromal cell multilayers, rapid endothelialization, and direct cell-cell interaction between the engineered stromal and endothelial components. This study has implications of recapitulating endothelial BM architecture for the de novo design of vascular TE scaffolds.
Collapse
Affiliation(s)
- Zuyong Wang
- †Department of Mechanical Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Swee Hin Teoh
- ‡School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Singapore
| | - Minghui Hong
- §Department of Electrical and Computer Engineering, National University of Singapore, 2 Engineering Drive 3, Singapore 117576, Singapore
| | - Fangfang Luo
- §Department of Electrical and Computer Engineering, National University of Singapore, 2 Engineering Drive 3, Singapore 117576, Singapore
| | - Erin Yiling Teo
- ⊥Department of Reproductive Medicine, KK Women's and Children's Hospital, 100 Buikit Timah Road, Singapore 229899, Singapore
| | - Jerry Kok Yen Chan
- ⊥Department of Reproductive Medicine, KK Women's and Children's Hospital, 100 Buikit Timah Road, Singapore 229899, Singapore
- ∥Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore
- ⊗Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, 8 College Road, Singapore 169857, Singapore
| | - Eng San Thian
- †Department of Mechanical Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| |
Collapse
|
34
|
Mutchler SM, Straub AC. Compartmentalized nitric oxide signaling in the resistance vasculature. Nitric Oxide 2015; 49:8-15. [PMID: 26028569 DOI: 10.1016/j.niox.2015.05.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 05/15/2015] [Accepted: 05/20/2015] [Indexed: 01/23/2023]
Abstract
Nitric oxide (NO) was first described as a bioactive molecule through its ability to stimulate soluble guanylate cyclase, but the revelation that NO was the endothelium derived relaxation factor drove the field to its modern state. The wealth of research conducted over the past 30 years has provided us with a picture of how diverse NO signaling can be within the vascular wall, going beyond simple vasodilation to include such roles as signaling through protein S-nitrosation. This expanded view of NO's actions requires highly regulated and compartmentalized production. Importantly, resistance arteries house multiple proteins involved in the production and transduction of NO allowing for efficient movement of the molecule to regulate vascular tone and reactivity. In this review, we focus on the many mechanisms regulating NO production and signaling action in the vascular wall, with a focus on the control of endothelial nitric oxide synthase (eNOS), the enzyme responsible for synthesizing most of the NO within these confines. We also explore how cross talk between the endothelium and smooth muscle in the microcirculation can modulate NO signaling, illustrating that this one small molecule has the capability to produce a plethora of responses.
Collapse
Affiliation(s)
- Stephanie M Mutchler
- Heart, Lung, Blood and Vascular Medicine Institute, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15216, USA
| | - Adam C Straub
- Heart, Lung, Blood and Vascular Medicine Institute, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15216, USA.
| |
Collapse
|