1
|
Archambault JL, Hernandez TL, Nozik ES, Palmer C, Dillon M, Singh RJ, Galambos C, Delaney CA. Relationship between Urine Serotonin and Persistent Pulmonary Hypertension of the Newborn. J Pediatr 2024; 276:114290. [PMID: 39242076 DOI: 10.1016/j.jpeds.2024.114290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/22/2024] [Accepted: 08/30/2024] [Indexed: 09/09/2024]
Abstract
Urine serotonin (5-HT)/creatinine was lower at day of life 3 in newborns with pulmonary hypertension compared with controls, while the percent change in the 5-HT metabolite, 5-hydroxyindoleacetic acid (5-HIAA)/creatinine increased. We speculate that the changes in 5-HT and 5-HIAA reflect enhanced pulmonary 5-HT uptake and/or metabolism.
Collapse
Affiliation(s)
- Jamie L Archambault
- Section of Neonatology, Department of Pediatrics, University of Colorado, Aurora, CO
| | - Teri L Hernandez
- College of Nursing, University of Colorado, Aurora, CO; Section of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado, Aurora, CO
| | - Eva S Nozik
- Section of Pediatric Critical Care, Department of Pediatrics, University of Colorado, Aurora, CO
| | - Claire Palmer
- Section of Neonatology, Department of Pediatrics, University of Colorado, Aurora, CO
| | - Mairead Dillon
- Section of Neonatology, Department of Pediatrics, University of Colorado, Aurora, CO
| | - Ravinder J Singh
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN
| | - Csaba Galambos
- Section of Pathology, Department of Pediatrics, University of Colorado, Aurora, CO
| | - Cassidy A Delaney
- Section of Neonatology, Department of Pediatrics, University of Colorado, Aurora, CO.
| |
Collapse
|
2
|
Charoenpong P, Hall NM, Keller CM, Ram AK, Murnane KS, Goeders NE, Dhillon NK, Walter RE. Overview of Methamphetamine-Associated Pulmonary Arterial Hypertension. Chest 2024; 165:1518-1533. [PMID: 38211700 PMCID: PMC11177101 DOI: 10.1016/j.chest.2024.01.014] [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: 11/05/2023] [Revised: 12/26/2023] [Accepted: 01/06/2024] [Indexed: 01/13/2024] Open
Abstract
TOPIC IMPORTANCE The global surge in methamphetamine use is a critical public health concern, particularly due to its robust correlation with methamphetamine-associated pulmonary arterial hypertension (MA-PAH). This association raises urgent alarms about the potential escalation of MA-PAH incidence, posing a significant and imminent challenge to global public health. REVIEW FINDINGS This comprehensive review meticulously explores MA-PAH, offering insights into its epidemiology, pathophysiology, clinical presentation, diagnostic intricacies, and management strategies. The pathogenesis, yet to be fully described, involves complex molecular interactions, including alterations in serotonin signaling, reduced activity of carboxylesterase 1, oxidative stress, and dysregulation of pulmonary vasoconstrictors and vasodilators. These processes culminate in the structural remodeling of the pulmonary vasculature, resulting in pulmonary arterial hypertension. MA-PAH exhibits a more severe clinical profile in functional class and hemodynamics compared with idiopathic pulmonary arterial hypertension. Management involves a multifaceted approach, integrating pulmonary vasodilators, cessation of methamphetamine use, and implementing social and rehabilitation programs. These measures aim to enhance patient outcomes and detect potential relapses for timely intervention. SUMMARY This review consolidates our understanding of MA-PAH, pinpointing knowledge gaps for future studies. Addressing these gaps is crucial for advancing diagnostic accuracy, unraveling mechanisms, and optimizing treatment for MA-PAH, thereby addressing the evolving landscape of this complex health concern.
Collapse
Affiliation(s)
- Prangthip Charoenpong
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Louisiana Addiction Research Center, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA.
| | - Nicole M Hall
- Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Louisiana Addiction Research Center, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Department of Pharmacology, Toxicology & Neuroscience, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA
| | - Courtney M Keller
- Louisiana Addiction Research Center, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Department of Pharmacology, Toxicology & Neuroscience, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA
| | - Anil Kumar Ram
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Kevin S Murnane
- Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Louisiana Addiction Research Center, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Department of Pharmacology, Toxicology & Neuroscience, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Department of Psychiatry and Behavioral Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA
| | - Nicholas E Goeders
- Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Louisiana Addiction Research Center, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Department of Pharmacology, Toxicology & Neuroscience, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Department of Psychiatry and Behavioral Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA
| | - Navneet Kaur Dhillon
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Robert E Walter
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA; Louisiana Addiction Research Center, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA
| |
Collapse
|
3
|
Lamb FS, Choi H, Miller MR, Stark RJ. Vascular Inflammation and Smooth Muscle Contractility: The Role of Nox1-Derived Superoxide and LRRC8 Anion Channels. Hypertension 2024; 81:752-763. [PMID: 38174563 PMCID: PMC10954410 DOI: 10.1161/hypertensionaha.123.19434] [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] [Indexed: 01/05/2024]
Abstract
Vascular inflammation underlies the development of hypertension, and the mechanisms by which it increases blood pressure remain the topic of intense investigation. Proinflammatory factors including glucose, salt, vasoconstrictors, cytokines, wall stress, and growth factors enhance contractility and impair relaxation of vascular smooth muscle cells. These pathways share a dependence upon redox signaling, and excessive activation promotes oxidative stress that promotes vascular aging. Vascular smooth muscle cell phenotypic switching and migration into the intima contribute to atherosclerosis, while hypercontractility increases systemic vascular resistance and vasospasm that can trigger ischemia. Here, we review factors that drive the initiation and progression of this vasculopathy in vascular smooth muscle cells. Emphasis is placed on the contribution of reactive oxygen species generated by the Nox1 NADPH oxidase which produces extracellular superoxide (O2•-). The mechanisms of O2•- signaling remain poorly defined, but recent evidence demonstrates physical association of Nox1 with leucine-rich repeat containing 8 family volume-sensitive anion channels. These may provide a pathway for influx of O2•- to the cytoplasm, creating an oxidized cytoplasmic nanodomain where redox-based signals can affect both cytoskeletal structure and vasomotor function. Understanding the mechanistic links between inflammation, O2•- and vascular smooth muscle cell contractility may facilitate targeting of anti-inflammatory therapy in hypertension.
Collapse
Affiliation(s)
- Fred S Lamb
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Hyehun Choi
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Michael R Miller
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Ryan J Stark
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
4
|
Boucetta H, Zhang L, Sosnik A, He W. Pulmonary arterial hypertension nanotherapeutics: New pharmacological targets and drug delivery strategies. J Control Release 2024; 365:236-258. [PMID: 37972767 DOI: 10.1016/j.jconrel.2023.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/25/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a rare, serious, and incurable disease characterized by high lung pressure. PAH-approved drugs based on conventional pathways are still not exhibiting favorable therapeutic outcomes. Drawbacks like short half-lives, toxicity, and teratogenicity hamper effectiveness, clinical conventionality, and long-term safety. Hence, approaches like repurposing drugs targeting various and new pharmacological cascades and/or loaded in non-toxic/efficient nanocarrier systems are being investigated lately. This review summarizes the status of conventional, repurposed, either in vitro, in vivo, and/or in clinical trials of PAH treatment. In-depth description, discussion, and classification of the new pharmacological targets and nanomedicine strategies with a description of all the nanocarriers that showed promising efficiency in delivering drugs are discussed. Ultimately, an illustration of the different nucleic acids tailored and nanoencapsulated within different types of nanocarriers to restore the pathways affected by this disease is presented.
Collapse
Affiliation(s)
- Hamza Boucetta
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518107, China
| | - Lei Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Alejandro Sosnik
- Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion - Israel Institute of Technology, Technion City, Haifa 3200003, Israel.
| | - Wei He
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China.
| |
Collapse
|
5
|
Chen Y, Ma P, Bo L, Lv Y, Zhou W, Zhou R. Isorhamnetin alleviates symptoms and inhibits oxidative stress levels in rats with pulmonary arterial hypertension. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:1616-1623. [PMID: 39539446 PMCID: PMC11556765 DOI: 10.22038/ijbms.2024.75860.16421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 06/15/2024] [Indexed: 11/16/2024]
Abstract
Objectives Pulmonary arterial hypertension (PAH) is a malignant pulmonary vascular disease with high mortality. Isorhamnetin (ISO), one of the main natural flavonoids extracted from sea buckthorn, has pharmacological effects such as anti-inflammatory, anti-proliferative and antioxidant. This study aimed to investigate the protective effect of ISO on PAH and its relationship with the phosphorylation of the c-Src tyrosine kinase (p-c-src)/NOX1 signaling pathway. Materials and Methods Ninety-five rats were randomly divided into five groups. The normal group received only a subcutaneous injection of saline, while the other groups received a subcutaneous injection of monocrotaline(MCT) (60 mg/kg) to establish a PAH model. The treatment group received ISO (50, 100, 150 mg/kg/d) treatment for 21 days, and after 21 days, all rat lung tissues were separated. Results The results showed that ISO could significantly improve the hemodynamics of MCT-induced PAH rats, such as mean pulmonary artery pressure (mPAP) and right ventricular systolic pressure (RVSP), and had inhibitory effects on right ventricular hypertrophy in PAH rats, and on pulmonary vascular remodeling in PAH rats. In addition, ISO can reduce the content of 5-hydroxytryptamine (5-HT) in PAH rats, increase the expression of Nrf2 protein in the lung tissue of PAH rats, activate the antioxidant system, enhance the activity of SOD in lung tissue of PAH rats, and inhibit NOX1, 5-HTT, p-c-src and Proliferating Cell Nuclear Antigen(PCNA) protein expression, and decrease MDA content. Conclusion Our research confirmed the therapeutic effect of ISO on MCT-induced PAH rats, which may be related to regulating the p-c-src/NOX1 signaling pathway.
Collapse
Affiliation(s)
- Yefeng Chen
- School of Clinical Medicine, Ningxia Medical University,Yinchuan, China
- These authors contributed equally to this work
| | - Ping Ma
- General Hospital of Ningxia Medical University, Yinchuan, China
- These authors contributed equally to this work
| | - Lei Bo
- Department of Foreign Language Teaching, Ningxia Medical University, Yinchuan, China
| | - Yingjie Lv
- School of Public Health and Management, Ningxia Medical University, Yinchuan, China
| | - Wei Zhou
- People’s Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Ru Zhou
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China
- Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan 750004, China
- Ningxia Characteristic Traditional Chinese Medicine Modernization Engineering Technology Research Center, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
6
|
Archambault JL, Delaney CA. A Review of Serotonin in the Developing Lung and Neonatal Pulmonary Hypertension. Biomedicines 2023; 11:3049. [PMID: 38002049 PMCID: PMC10668978 DOI: 10.3390/biomedicines11113049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/03/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Serotonin (5-HT) is a bioamine that has been implicated in the pathogenesis of pulmonary hypertension (PH). The lung serves as an important site of 5-HT synthesis, uptake, and metabolism with signaling primarily regulated by tryptophan hydroxylase (TPH), the 5-HT transporter (SERT), and numerous unique 5-HT receptors. The 5-HT hypothesis of PH was first proposed in the 1960s and, since that time, preclinical and clinical studies have worked to elucidate the role of 5-HT in adult PH. Over the past several decades, accumulating evidence from both clinical and preclinical studies has suggested that the 5-HT signaling pathway may play an important role in neonatal cardiopulmonary transition and the development of PH in newborns. The expression of TPH, SERT, and the 5-HT receptors is developmentally regulated, with alterations resulting in pulmonary vasoconstriction and pulmonary vascular remodeling. However, much remains unknown about the role of 5-HT in the developing and newborn lung. The purpose of this review is to discuss the implications of 5-HT on fetal and neonatal pulmonary circulation and summarize the existing preclinical and clinical literature on 5-HT in neonatal PH.
Collapse
Affiliation(s)
| | - Cassidy A. Delaney
- Section of Neonatology, Department of Pediatrics, University of Colorado, Aurora, CO 80045, USA;
| |
Collapse
|
7
|
Liu W, Liu C, Yu X, Zhai Y, He Q, Li J, Liu X, Ye X, Zhang Q, Wang R, Zhang Y, Ge P, Zhang D. Association between systemic immune-inflammatory markers and the risk of moyamoya disease: a case-control study. Ann Med 2023; 55:2269368. [PMID: 37865806 PMCID: PMC10591523 DOI: 10.1080/07853890.2023.2269368] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 10/06/2023] [Indexed: 10/23/2023] Open
Abstract
Background:Systemic immune-inflammatory markers such as neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), lymphocyte-to-monocyte ratio (LMR) and systemic immune-inflammatory index (SII) are associated with the prognosis of many cardiovascular and neoplastic diseases. Moyamoya disease (MMD) is associated with inflammation, but the relationship between systemic immune-inflammatory markers between MMD is unclear. The aim of our study was to analyse the association between systemic immune-inflammatory markers and the risk of MMD and its subtypes.Methods:We consecutively recruited 360 patients with MMD and 89 healthy control subjects in a case-control study to calculate and analyse the association of systemic immune-inflammatory markers with the risk of MMD and its subtypes.Results:The risk of MMD increased with higher levels of NLR (OR 1.237, 95% CI [1.008, 1.520], p = .042). When NLR and SII were assessed as quartile-spaced subgroups, the third quartile grouping of NLR and SII had a higher risk of MMD than the first quartile grouping (NLR: OR 3.206, 95% CI [1.271, 8.088], p = .014; SII: OR 3.074,95% CI [1.232,7.672], p = .016). When NLR was combined with SII, the highest subgroup had a higher risk of MMD than the lowest subgroup (OR2.643, 95% CI [1.340, 5.212], p = .005). The risk of subtypes also increased with higher levels of NLR and SII. The association between the levels of NLR and SII with the staging of the Suzuki stage follows an inverted U-shape. The highest levels of NLR and SII were found in patients with MMD at Suzuki stages 3-4.Conclusion:The risk of MMD increases with elevated systemic immune-inflammatory markers. This study analysed the association of systemic immune-inflammatory markers with the risk of developing MMD and its subtypes, and identified novel inflammatory markers for MMD.
Collapse
Affiliation(s)
- Wei Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Chenglong Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Xiaofan Yu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Yuanren Zhai
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Qiheng He
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Junsheng Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Xingju Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Xun Ye
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Qian Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Rong Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Yan Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Peicong Ge
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| | - Dong Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
- Beijing Translational Engineering Center for 3D Printer in Clinical Neuroscience, Beijing, China
| |
Collapse
|
8
|
Rybakova EY, Avdonin PP, Trufanov SK, Goncharov NV, Avdonin PV. Synergistic Interaction of 5-HT 1B and 5-HT 2B Receptors in Cytoplasmic Ca 2+ Regulation in Human Umbilical Vein Endothelial Cells: Possible Involvement in Pathologies. Int J Mol Sci 2023; 24:13833. [PMID: 37762136 PMCID: PMC10530667 DOI: 10.3390/ijms241813833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/28/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
The aim of this work was to explore the involvement of 5-HT1B and 5-HT2B receptors (5-HT1BR and 5-HT2BR) in the regulation of free cytoplasmic calcium concentration ([Ca2+]i) in human umbilical vein endothelial cells (HUVEC). We have shown by quantitative PCR analysis, that 5-HT1BR and 5-HT2BR mRNAs levels are almost equal in HUVEC. Immunofluorescent staining demonstrated, that 5-HT1BR and 5-HT2BR are expressed both in plasma membrane and inside the cells. Intracellular 5-HT1BR are localized mainly in the nuclear region, whereas 5-HT2BR receptors are almost evenly distributed in HUVEC. 5-HT, 5-HT1BR agonist CGS12066B, or 5-HT2BR agonist BW723C86 added to HUVEC caused a slight increase in [Ca2+]i, which was much lower than that of histamine, ATP, or SFLLRN, an agonist of protease-activated receptors (PAR1). However, activation of 5-HT1BR with CGS12066B followed by activation of 5-HT2BR with BW723C86 manifested a synergism of response, since several-fold higher rise in [Ca2+]i occurred. CGS12066B caused more than a 5-fold increase in [Ca2+]i rise in HUVEC in response to 5-HT. This 5-HT induced [Ca2+]i rise was abolished by 5-HT2BR antagonist RS127445, indicating that extracellular 5-HT acts through 5-HT2BR. Synergistic [Ca2+]i rise in response to activation of 5-HT1BR and 5-HT2BR persisted in a calcium-free medium. It was suppressed by the phospholipase C inhibitor U73122 and was not inhibited by the ryanodine and NAADP receptors antagonists dantrolene and NED-19. [Ca2+]i measurements in single cells demonstrated that activation of 5-HT2BR alone by BW723C86 caused single asynchronous [Ca2+]i oscillations in 19.8 ± 4.2% (n = 3) of HUVEC that occur with a long delay (66.1 ± 4.3 s, n = 71). On the contrary, histamine causes a simultaneous and almost immediate increase in [Ca2+]i in all the cells. Pre-activation of 5-HT1BR by CGS12066B led to a 3-4 fold increase in the number of HUVEC responding to BW723C86, to synchronization of their responses with a delay shortening, and to the bursts of [Ca2+]i oscillations in addition to single oscillations. In conclusion, to get a full rise of [Ca2+]i in HUVEC in response to 5-HT, simultaneous activation of 5-HT1BR and 5-HT2BR is required. 5-HT causes an increase in [Ca2+]i via 5-HT2BR while 5-HT1BR could be activated by the membrane-permeable agonist CGS12066B. We hypothesized that CGS12066B acts via intracellular 5-HT1BR inaccessible to extracellular 5-HT. Intracellular 5-HT1BR might be activated by 5-HT which could be accumulated in EC under certain pathological conditions.
Collapse
Affiliation(s)
- Elena Yu. Rybakova
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Moscow 119334, Russia; (E.Y.R.); (P.P.A.); (S.K.T.)
| | - Piotr P. Avdonin
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Moscow 119334, Russia; (E.Y.R.); (P.P.A.); (S.K.T.)
| | - Sergei K. Trufanov
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Moscow 119334, Russia; (E.Y.R.); (P.P.A.); (S.K.T.)
| | - Nikolay V. Goncharov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Saint Petersburg 194223, Russia;
| | - Pavel V. Avdonin
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, Moscow 119334, Russia; (E.Y.R.); (P.P.A.); (S.K.T.)
| |
Collapse
|
9
|
Yan Q, Liu S, Sun Y, Chen C, Yang S, Lin M, Long J, Yao J, Lin Y, Yi F, Meng L, Tan Y, Ai Q, Chen N, Yang Y. Targeting oxidative stress as a preventive and therapeutic approach for cardiovascular disease. J Transl Med 2023; 21:519. [PMID: 37533007 PMCID: PMC10394930 DOI: 10.1186/s12967-023-04361-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/16/2023] [Indexed: 08/04/2023] Open
Abstract
Cardiovascular diseases (CVDs) continue to exert a significant impact on global mortality rates, encompassing conditions like pulmonary arterial hypertension (PAH), atherosclerosis (AS), and myocardial infarction (MI). Oxidative stress (OS) plays a crucial role in the pathogenesis and advancement of CVDs, highlighting its significance as a contributing factor. Maintaining an equilibrium between reactive oxygen species (ROS) and antioxidant systems not only aids in mitigating oxidative stress but also confers protective benefits on cardiac health. Herbal monomers can inhibit OS in CVDs by activating multiple signaling pathways, such as increasing the activity of endogenous antioxidant systems and decreasing the level of ROS expression. Given the actions of herbal monomers to significantly protect the normal function of the heart and reduce the damage caused by OS to the organism. Hence, it is imperative to recognize the significance of herbal monomers as prospective therapeutic interventions for mitigating oxidative damage in CVDs. This paper aims to comprehensively review the origins and mechanisms underlying OS, elucidate the intricate association between CVDs and OS, and explore the therapeutic potential of antioxidant treatment utilizing herbal monomers. Furthermore, particular emphasis will be placed on examining the cardioprotective effects of herbal monomers by evaluating their impact on cardiac signaling pathways subsequent to treatment.
Collapse
Affiliation(s)
- Qian Yan
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Shasha Liu
- Department of Pharmacy, Changsha Hospital for Matemal&Child Health Care, Changsha, People's Republic of China
| | - Yang Sun
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Chen Chen
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Songwei Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Meiyu Lin
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Junpeng Long
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Jiao Yao
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Yuting Lin
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Fan Yi
- Key Laboratory of Cosmetic, China National Light Industry, Beijing Technology and Business University, Beijing, 100048, China
| | - Lei Meng
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Yong Tan
- Department of Nephrology, Xiangtan Central Hospital, Xiangtan, 411100, China
| | - Qidi Ai
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Naihong Chen
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Yantao Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
| |
Collapse
|
10
|
Morris HE, Neves KB, Nilsen M, Montezano AC, MacLean MR, Touyz RM. Notch3/Hes5 Induces Vascular Dysfunction in Hypoxia-Induced Pulmonary Hypertension Through ER Stress and Redox-Sensitive Pathways. Hypertension 2023; 80:1683-1696. [PMID: 37254738 PMCID: PMC10355806 DOI: 10.1161/hypertensionaha.122.20449] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 04/24/2023] [Indexed: 06/01/2023]
Abstract
BACKGROUND Notch3 (neurogenic locus notch homolog protein 3) is implicated in vascular diseases, including pulmonary hypertension (PH)/pulmonary arterial hypertension. However, molecular mechanisms remain elusive. We hypothesized increased Notch3 activation induces oxidative and endoplasmic reticulum (ER) stress and downstream redox signaling, associated with procontractile pulmonary artery state, pulmonary vascular dysfunction, and PH development. METHODS Studies were performed in TgNotch3R169C mice (harboring gain-of-function [GOF] Notch3 mutation) exposed to chronic hypoxia to induce PH, and examined by hemodynamics. Molecular and cellular studies were performed in pulmonary artery smooth muscle cells from pulmonary arterial hypertension patients and in mouse lung. Notch3-regulated genes/proteins, ER stress, ROCK (Rho-associated kinase) expression/activity, Ca2+ transients and generation of reactive oxygen species, and nitric oxide were measured. Pulmonary vascular reactivity was assessed in the presence of fasudil (ROCK inhibitor) and 4-phenylbutyric acid (ER stress inhibitor). RESULTS Hypoxia induced a more severe PH phenotype in TgNotch3R169C mice versus controls. TgNotch3R169C mice exhibited enhanced Notch3 activation and expression of Notch3 targets Hes Family BHLH Transcription Factor 5 (Hes5), with increased vascular contraction and impaired vasorelaxation that improved with fasudil/4-phenylbutyric acid. Notch3 mutation was associated with increased pulmonary vessel Ca2+ transients, ROCK activation, ER stress, and increased reactive oxygen species generation, with reduced NO generation and blunted sGC (soluble guanylyl cyclase)/cGMP signaling. These effects were ameliorated by N-acetylcysteine. pulmonary artery smooth muscle cells from patients with pulmonary arterial hypertension recapitulated Notch3/Hes5 signaling, ER stress and redox changes observed in PH mice. CONCLUSIONS Notch3 GOF amplifies vascular dysfunction in hypoxic PH. This involves oxidative and ER stress, and ROCK. We highlight a novel role for Notch3/Hes5-redox signaling and important interplay between ER and oxidative stress in PH.
Collapse
Affiliation(s)
- Hannah E Morris
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (H.E.M., K.B.N., A.C.M., R.M.T.)
| | - Karla B Neves
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (H.E.M., K.B.N., A.C.M., R.M.T.)
| | - Margaret Nilsen
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, United Kingdom (M.N., M.R.M.)
| | - Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (H.E.M., K.B.N., A.C.M., R.M.T.)
| | - Margaret R MacLean
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, United Kingdom (M.N., M.R.M.)
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (H.E.M., K.B.N., A.C.M., R.M.T.)
- Research Institute of McGill University Health Centre, McGill University, Canada (R.M.T.)
| |
Collapse
|
11
|
Cook CM, Craddock VD, Ram AK, Abraham AA, Dhillon NK. HIV and Drug Use: A Tale of Synergy in Pulmonary Vascular Disease Development. Compr Physiol 2023; 13:4659-4683. [PMID: 37358518 PMCID: PMC10693986 DOI: 10.1002/cphy.c210049] [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] [Indexed: 06/27/2023]
Abstract
Over the past two decades, with the advent and adoption of highly active anti-retroviral therapy, HIV-1 infection, a once fatal and acute illness, has transformed into a chronic disease with people living with HIV (PWH) experiencing increased rates of cardio-pulmonary vascular diseases including life-threatening pulmonary hypertension. Moreover, the chronic consequences of tobacco, alcohol, and drug use are increasingly seen in older PWH. Drug use, specifically, can have pathologic effects on the cardiovascular health of these individuals. The "double hit" of drug use and HIV may increase the risk of HIV-associated pulmonary arterial hypertension (HIV-PAH) and potentiate right heart failure in this population. This article explores the epidemiology and pathophysiology of PAH associated with HIV and recreational drug use and describes the proposed mechanisms by which HIV and drug use, together, can cause pulmonary vascular remodeling and cardiopulmonary hemodynamic compromise. In addition to detailing the proposed cellular and signaling pathways involved in the development of PAH, this article proposes areas ripe for future research, including the influence of gut dysbiosis and cellular senescence on the pathobiology of HIV-PAH. © 2023 American Physiological Society. Compr Physiol 13:4659-4683, 2023.
Collapse
Affiliation(s)
- Christine M Cook
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Vaughn D Craddock
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Anil K Ram
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Ashrita A Abraham
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Navneet K Dhillon
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
12
|
Zhu J, Yang L, Jia Y, Balistrieri A, Fraidenburg DR, Wang J, Tang H, Yuan JXJ. Pathogenic Mechanisms of Pulmonary Arterial Hypertension: Homeostasis Imbalance of Endothelium-Derived Relaxing and Contracting Factors. JACC. ASIA 2022; 2:787-802. [PMID: 36713766 PMCID: PMC9877237 DOI: 10.1016/j.jacasi.2022.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 08/29/2022] [Accepted: 09/14/2022] [Indexed: 12/23/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive and fatal disease. Sustained pulmonary vasoconstriction and concentric pulmonary vascular remodeling contribute to the elevated pulmonary vascular resistance and pulmonary artery pressure in PAH. Endothelial cells regulate vascular tension by producing endothelium-derived relaxing factors (EDRFs) and endothelium-derived contracting factors (EDCFs). Homeostasis of EDRF and EDCF production has been identified as a marker of the endothelium integrity. Impaired synthesis or release of EDRFs induces persistent vascular contraction and pulmonary artery remodeling, which subsequently leads to the development and progression of PAH. In this review, the authors summarize how EDRFs and EDCFs affect pulmonary vascular homeostasis, with special attention to the recently published novel mechanisms related to endothelial dysfunction in PAH and drugs associated with EDRFs and EDCFs.
Collapse
Key Words
- 5-HT, 5-hydroxytryptamine
- ACE, angiotensin-converting enzyme
- EC, endothelial cell
- EDCF, endothelium-derived contracting factor
- EDRF, endothelium-derived relaxing factor
- ET, endothelin
- PAH, pulmonary arterial hypertension
- PASMC, pulmonary artery smooth muscle cell
- PG, prostaglandin
- TPH, tryptophan hydroxylase
- TXA2, thromboxane A2
- cGMP, cyclic guanosine monophosphate
- endothelial dysfunction
- endothelium-derived relaxing factor
- pulmonary arterial hypertension
- vascular homeostasis
Collapse
Affiliation(s)
- Jinsheng Zhu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lei Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yangfan Jia
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Angela Balistrieri
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Dustin R. Fraidenburg
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Haiyang Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
13
|
Chen D, Zhang HF, Yuan TY, Sun SC, Wang RR, Wang SB, Fang LH, Lyu Y, Du GH. Puerarin-V prevents the progression of hypoxia- and monocrotaline-induced pulmonary hypertension in rodent models. Acta Pharmacol Sin 2022; 43:2325-2339. [PMID: 35190697 PMCID: PMC9433387 DOI: 10.1038/s41401-022-00865-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 01/09/2022] [Indexed: 02/06/2023] Open
Abstract
Pulmonary hypertension (PH) is a cardiopulmonary disease characterized by a progressive increase in pulmonary vascular resistance. One of the initial pathogenic factors of PH is pulmonary arterial remodeling under various stimuli. Current marketed drugs against PH mainly relieve symptoms without significant improvement in overall prognosis. Discovering and developing new therapeutic drugs that interfere with vascular remodeling is in urgent need. Puerarin is an isoflavone compound extracted from the root of Kudzu vine, which is widely used in the treatment of cardiovascular diseases. In the present study, we evaluated the efficacy of puerarin in the treatment of experimental PH. PH was induced in rats by a single injection of MCT (50 mg/kg, sc), and in mice by exposure to hypoxia (10% O2) for 14 days. After MCT injection the rats were administered puerarin (10, 30, 100 mg · kg-1 · d-1, i.g.) for 28 days, whereas hypoxia-treated mice were pre-administered puerarin (60 mg · kg-1 · d-1, i.g.) for 7 days. We showed that puerarin administration exerted significant protective effects in both experimental PH rodent models, evidenced by significantly reduced right ventricular systolic pressure (RVSP) and lung injury, improved pulmonary artery blood flow as well as pulmonary vasodilation and contraction function, inhibited inflammatory responses in lung tissues, improved resistance to apoptosis and abnormal proliferation in lung tissues, attenuated right ventricular injury and remodeling, and maintained normal function of the right ventricle. We revealed that MCT and hypoxia treatment significantly downregulated BMPR2/Smad signaling in the lung tissues and PPARγ/PI3K/Akt signaling in the lung tissues and right ventricles, which were restored by puerarin administration. In addition, we showed that a novel crystal type V (Puer-V) exerted better therapeutic effects than the crude form of puerarin (Puer). Furthermore, Puer-V was more efficient than bosentan (a positive control drug) in alleviating the abnormal structural changes and dysfunction of lung tissues and right ventricles. In conclusion, this study provides experimental evidence for developing Puer-V as a novel therapeutic drug to treat PH.
Collapse
Affiliation(s)
- Di Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Hui-Fang Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Tian-Yi Yuan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Shu-Chan Sun
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Ran-Ran Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Shou-Bao Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Lian-Hua Fang
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Yang Lyu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
- Beijing Key Laboratory of Polymorphic Drugs, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Guan-Hua Du
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
- Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
14
|
MacLean MR, Fanburg B, Hill N, Lazarus HM, Pack TF, Palacios M, Penumatsa KC, Wring SA. Serotonin and Pulmonary Hypertension; Sex and Drugs and ROCK and Rho. Compr Physiol 2022; 12:4103-4118. [PMID: 36036567 DOI: 10.1002/cphy.c220004] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Serotonin is often referred to as a "happy hormone" as it maintains good mood, well-being, and happiness. It is involved in communication between nerve cells and plays a role in sleeping and digestion. However, too much serotonin can have pathogenic effects and serotonin synthesis is elevated in pulmonary artery endothelial cells from patients with pulmonary arterial hypertension (PAH). PAH is characterized by elevated pulmonary pressures, right ventricular failure, inflammation, and pulmonary vascular remodeling; serotonin has been shown to be associated with these pathologies. The rate-limiting enzyme in the synthesis of serotonin in the periphery of the body is tryptophan hydroxylase 1 (TPH1). TPH1 expression and serotonin synthesis are elevated in pulmonary artery endothelial cells in patients with PAH. The serotonin synthesized in the pulmonary arterial endothelium can act on the adjacent pulmonary arterial smooth muscle cells (PASMCs), adventitial macrophages, and fibroblasts, in a paracrine fashion. In humans, serotonin enters PASMCs cells via the serotonin transporter (SERT) and it can cooperate with the 5-HT1B receptor on the plasma membrane; this activates both contractile and proliferative signaling pathways. The "serotonin hypothesis of pulmonary hypertension" arose when serotonin was associated with PAH induced by diet pills such as fenfluramine, aminorex, and chlorphentermine; these act as indirect serotonergic agonists causing the release of serotonin from platelets and cells through the SERT. Here the role of serotonin in PAH is reviewed. Targeting serotonin synthesis or signaling is a promising novel alternative approach which may lead to novel therapies for PAH. © 2022 American Physiological Society. Compr Physiol 12: 1-16, 2022.
Collapse
Affiliation(s)
- Margaret R MacLean
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland
| | - Barry Fanburg
- Pulmonary, Critical Care and Sleep Division, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | - Nicolas Hill
- Pulmonary, Critical Care and Sleep Division, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | | | | | | | - Krishna C Penumatsa
- Pulmonary, Critical Care and Sleep Division, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | | |
Collapse
|
15
|
Huang L, Zhang H, Liu Y, Long Y. The Role of Gut and Airway Microbiota in Pulmonary Arterial Hypertension. Front Microbiol 2022; 13:929752. [PMID: 35910623 PMCID: PMC9326471 DOI: 10.3389/fmicb.2022.929752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/17/2022] [Indexed: 11/13/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe clinical condition that is characterized pathologically by perivascular inflammation and pulmonary vascular remodeling that ultimately leads to right heart failure. However, current treatments focus on controlling vasoconstriction and have little effect on pulmonary vascular remodeling. Better therapies of PAH require a better understanding of its pathogenesis. With advances in sequencing technology, researchers have begun to focus on the role of the human microbiota in disease. Recent studies have shown that the gut and airway microbiota and their metabolites play an important role in the pathogenesis of PAH. In this review, we summarize the current literature on the relationship between the gut and airway microbiota and PAH. We further discuss the key crosstalk between the gut microbiota and the lung associated with PAH, and the potential link between the gut and airway microbiota in the pathogenesis of PAH. In addition, we discuss the potential of using the microbiota as a new target for PAH therapy.
Collapse
Affiliation(s)
- Linlin Huang
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
| | - Hongdie Zhang
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
| | - Yijun Liu
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
| | - Yang Long
- Experimental Medicine Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, China
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- *Correspondence: Yang Long
| |
Collapse
|
16
|
Target Nuclear Factor Erythroid 2-Related Factor 2 in Pulmonary Hypertension: Molecular Insight into Application. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7845503. [PMID: 35707273 PMCID: PMC9192195 DOI: 10.1155/2022/7845503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 04/07/2022] [Accepted: 04/08/2022] [Indexed: 11/17/2022]
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a key transcription factor involved in maintaining redox balance and activates the expression of downstream antioxidant enzymes. Nrf2 has received wide attention considering its crucial role in oxidative and electrophilic stress. Large amounts of studies have demonstrated the protective role of Nrf2 activation in various pulmonary hypertension (pH) models. Additionally, various kinds of natural phytochemicals acting as Nrf2 activators prevent the development of pH and provide a novel and promising therapeutic insight for the treatment of pH. In the current review, we give a brief introduction of Nrf2 and focus on the role and mechanism of Nrf2 in the pathophysiology of pH and then review the relevant research of Nrf2 agonists in pH in both experimental research and clinical trials.
Collapse
|
17
|
Cheron C, McBride SA, Antigny F, Girerd B, Chouchana M, Chaumais MC, Jaïs X, Bertoletti L, Sitbon O, Weatherald J, Humbert M, Montani D. Sex and gender in pulmonary arterial hypertension. Eur Respir Rev 2021; 30:30/162/200330. [PMID: 34750113 DOI: 10.1183/16000617.0330-2020] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 01/28/2021] [Indexed: 12/21/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare disease characterised by pulmonary vascular remodelling and elevated pulmonary pressure, which eventually leads to right heart failure and death. Registries worldwide have noted a female predominance of the disease, spurring particular interest in hormonal involvement in the disease pathobiology. Several experimental models have shown both protective and deleterious effects of oestrogens, suggesting that complex mechanisms participate in PAH pathogenesis. In fact, oestrogen metabolites as well as receptors and enzymes implicated in oestrogen signalling pathways and associated conditions such as BMPR2 mutation contribute to PAH penetrance more specifically in women. Conversely, females have better right ventricular function, translating to a better prognosis. Along with right ventricular adaptation, women tend to respond to PAH treatment differently from men. As some young women suffer from PAH, contraception is of particular importance, considering that pregnancy in patients with PAH is strongly discouraged due to high risk of death. When contraception measures fail, pregnant women need a multidisciplinary team-based approach. This article aims to review epidemiology, mechanisms underlying the higher female predominance, but better prognosis and the intricacies in management of women affected by PAH.
Collapse
Affiliation(s)
- Céline Cheron
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Susan Ainslie McBride
- Internal Medicine Residency Program, Dept of Medicine, University of Calgary, Calgary, Canada
| | - Fabrice Antigny
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Barbara Girerd
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Margot Chouchana
- Assistance Publique Hôpitaux de Paris, Service de Pharmacie Hôpital Bicêtre, Le Kremlin Bicêtre, France
| | - Marie-Camille Chaumais
- Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France.,Assistance Publique Hôpitaux de Paris, Service de Pharmacie Hôpital Bicêtre, Le Kremlin Bicêtre, France.,Université Paris-Saclay, Faculté de Pharmacie, Chatenay Malabry, France
| | - Xavier Jaïs
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Laurent Bertoletti
- Centre Hospitalier Universitaire de Saint-Etienne, Service de Médecine Vasculaire et Thérapeutique, Saint-Etienne, France.,INSERM U1059 et CIC1408, Université Jean-Monnet, Saint-Etienne, France
| | - Olivier Sitbon
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Jason Weatherald
- Division of Respirology, Dept of Medicine, University of Calgary, Calgary, Canada.,Libin Cardiovascular Institute, University of Calgary, Calgary, Canada
| | - Marc Humbert
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - David Montani
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France .,Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin-Bicêtre, France.,Inserm UMR_S 999, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| |
Collapse
|
18
|
Zhou SY, Tao L, Zhang Z, Zhang Z, An S. Mediators of neutrophil lymphocyte ratio in the relationship between ondansetron pre-treatment and the mortality of ICU patients on mechanical ventilation: causal mediation analysis from the MIMIC-IV database. Br J Clin Pharmacol 2021; 88:2747-2756. [PMID: 34964162 DOI: 10.1111/bcp.15204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 11/29/2021] [Accepted: 12/14/2021] [Indexed: 11/26/2022] Open
Abstract
AIMS The mortality of critically ill patients undergoing mechanical ventilation (MV) is high and few strategies are available. We explored the relationship between ondansetron pre-treatment, the neutrophil:lymphocyte ratio (NLR), and platelet:lymphocyte ratio (PLR), and mortality of ventilated patients in the intensive care unit. METHODS We developed a retrospective cohort study that involved patients undergoing MV in the Multiparameter Intelligent Monitoring in Intensive Care IV (MIMIC-IV) database. Causal mediation analysis was conducted to assess the relationship of ondansetron use and mortality and explore the potential causal pathway mediated by the NLR or PLR. The primary outcome was 28-day mortality. RESULTS A total of 17,927 eligible patients was obtained (5665 had taken ondansetron before MV initiation and 12,262 patients had not). The OR for 28-day mortality for ondansetron use uncorrelated with the mediator (NLR, PLR) was 0.72 (95%CI=0.64-0.81, P <0.001). Ondansetron was also associated with a reduction in 28-day mortality after controlling for the mediator of NLR (OR = 0.98, 95%CI = 0.97-0.99, P < 0.01). For the indirect effect, the NLR could explain 13.47% (95%CI = 8.59-20.54%, P < 0.01) of the impact of ondansetron use on 28-day mortality. The proportion mediated increased to 21.50% (95%CI = 12.36-47.44%, P < 0.01) for 90-day mortality. Adjusted mediation analysis revealed no suggestion of a causal mediation pathway for this effect by the PLR (P = 0.12). CONCLUSIONS NLR may play substantial roles in the relationship between ondansetron pre-treatment before initiation of mechanical ventilation and the reduction of death risk in ventilated patients.
Collapse
Affiliation(s)
- Shi Yu Zhou
- Department of Biostatistics, School of Public Health (Guangdong Provincial Key Laboratory of Tropical Disease Research), Southern Medical University, Guangzhou, Guangdong, China
| | - Lili Tao
- Department of Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zheng Zhang
- Department of Biostatistics, School of Public Health (Guangdong Provincial Key Laboratory of Tropical Disease Research), Southern Medical University, Guangzhou, Guangdong, China
| | - Zhenhui Zhang
- Department of Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shengli An
- Department of Biostatistics, School of Public Health (Guangdong Provincial Key Laboratory of Tropical Disease Research), Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
19
|
Alves-Lopes R, Neves KB, Strembitska A, Harvey AP, Harvey KY, Yusuf H, Haniford S, Hepburn RT, Dyet J, Beattie W, Haddow L, McAbney J, Graham D, Montezano AC. Osteoprotegerin regulates vascular function through syndecan-1 and NADPH oxidase-derived reactive oxygen species. Clin Sci (Lond) 2021; 135:2429-2444. [PMID: 34668009 DOI: 10.1042/cs20210643] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 01/27/2023]
Abstract
Osteogenic factors, such as osteoprotegerin (OPG), are protective against vascular calcification. However, OPG is also positively associated with cardiovascular damage, particularly in pulmonary hypertension, possibly through processes beyond effects on calcification. In the present study, we focused on calcification-independent vascular effects of OPG through activation of syndecan-1 and NADPH oxidases (Noxs) 1 and 4. Isolated resistance arteries from Wistar-Kyoto (WKY) rats, exposed to exogenous OPG, studied by myography exhibited endothelial and smooth muscle dysfunction. OPG decreased nitric oxide (NO) production, eNOS activation and increased reactive oxygen species (ROS) production in endothelial cells. In VSMCs, OPG increased ROS production, H2O2/peroxynitrite levels and activation of Rho kinase and myosin light chain. OPG vascular and redox effects were also inhibited by the syndecan-1 inhibitor synstatin (SSNT). Additionally, heparinase and chondroitinase abolished OPG effects on VSMCs-ROS production, confirming syndecan-1 as OPG molecular partner and suggesting that OPG binds to heparan/chondroitin sulphate chains of syndecan-1. OPG-induced ROS production was abrogated by NoxA1ds (Nox1 inhibitor) and GKT137831 (dual Nox1/Nox4 inhibitor). Tempol (SOD mimetic) inhibited vascular dysfunction induced by OPG. In addition, we studied arteries from Nox1 and Nox4 knockout (KO) mice. Nox1 and Nox4 KO abrogated OPG-induced vascular dysfunction. Vascular dysfunction elicited by OPG is mediated by a complex signalling cascade involving syndecan-1, Nox1 and Nox4. Our data identify novel molecular mechanisms beyond calcification for OPG, which may underlie vascular injurious effects of osteogenic factors in conditions such as hypertension and/or diabetes.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Hemodynamics/drug effects
- Male
- Mesenteric Arteries/drug effects
- Mesenteric Arteries/enzymology
- Mesenteric Arteries/physiopathology
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- NADPH Oxidase 1/genetics
- NADPH Oxidase 1/metabolism
- NADPH Oxidase 4/genetics
- NADPH Oxidase 4/metabolism
- NADPH Oxidases/genetics
- NADPH Oxidases/metabolism
- Osteoprotegerin/toxicity
- Oxidative Stress
- Rats, Inbred WKY
- Reactive Oxygen Species/metabolism
- Signal Transduction
- Syndecan-1/metabolism
- Mice
- Rats
Collapse
Affiliation(s)
- Rhéure Alves-Lopes
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, U.K
| | - Karla Bianca Neves
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, U.K
| | | | - Adam P Harvey
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, U.K
| | - Katie Y Harvey
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, U.K
| | - Hiba Yusuf
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, U.K
| | - Susan Haniford
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, U.K
| | - Ross T Hepburn
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, U.K
| | - Jennifer Dyet
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, U.K
| | - Wendy Beattie
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, U.K
| | - Laura Haddow
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, U.K
| | - John McAbney
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, U.K
| | - Delyth Graham
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, U.K
| | - Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, U.K
| |
Collapse
|
20
|
The occurrence and risk factors of bradycardia after the Maze procedure in patients with atrial fibrillation and tricuspid regurgitation. J Cardiothorac Surg 2021; 16:270. [PMID: 34565413 PMCID: PMC8474904 DOI: 10.1186/s13019-021-01653-1] [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: 08/05/2021] [Accepted: 09/17/2021] [Indexed: 11/30/2022] Open
Abstract
Objective To evaluate the occurrence and risk factors of bradycardia after the Maze procedure in patients with atrial fibrillation and tricuspid regurgitation. Methods All patients underwent mitral valve (MV) replacement and concomitant bi-atrial cut-and-sew Maze procedure along with other cardiac surgical procedures were recruited from the Department of Cardiovascular Surgery at the First Affiliated Hospital of Medical College of Xi'an Jiaotong University. According to the severity of tricuspid regurgitation, all patients were divided into mild tricuspid regurgitation group and moderate-to-severe tricuspid regurgitation group. The general clinical data, biochemical indexes, intraoperative and postoperative data were collected. The relationship between tricuspid regurgitation and sinus bradycardia after the Maze procedure was analyzed by multivariate logistic regression model. Results We enrolled 82 patients, including 24 males and 58 females. The patients had an average age of 56 ± 10 years old. There were 50 cases in mild tricuspid regurgitation group and 32 cases in moderate-to-severe tricuspid regurgitation group. Compared with the mild tricuspid regurgitation group, postoperative bradyarrhythmia (41% vs. 14%), pre-discharge bradyarrhythmia (63% vs. 14%), postoperative sinus bradycardia (34% vs. 10%) and pre-discharge sinus bradycardia (63% vs. 10%) in moderate-to-severe tricuspid regurgitation group were significantly increased (P < 0.01). In moderate-to-severe tricuspid regurgitation, the risk of sinus bradycardia increased after the Maze procedure (OR = 1.453, 95% CI 1.127–1.874), area under ROC curve was 0.81, the Jordan index was 0.665. Conclusion The severity of tricuspid regurgitation may be an important factor affecting sinus bradycardia after the Maze procedure. It can be considered as a factor to predict sinus bradycardia after the Maze procedure.
Collapse
|
21
|
Shimoda LA. Cellular Pathways Promoting Pulmonary Vascular Remodeling by Hypoxia. Physiology (Bethesda) 2021; 35:222-233. [PMID: 32490752 DOI: 10.1152/physiol.00039.2019] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Exposure to hypoxia increases pulmonary vascular resistance, leading to elevated pulmonary arterial pressure and, potentially, right heart failure. Vascular remodeling is an important contributor to the increased pulmonary vascular resistance. Hyperproliferation of smooth muscle, endothelial cells, and fibroblasts, and deposition of extracellular matrix lead to increased wall thickness, extension of muscle into normally non-muscular arterioles, and vascular stiffening. This review highlights intrinsic and extrinsic modulators contributing to the remodeling process.
Collapse
Affiliation(s)
- Larissa A Shimoda
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
22
|
Labazi H, Nilsen M, MacLean MR. Sex-dependent right ventricular hypertrophic gene changes after methamphetamine treatment in mice. Eur J Pharmacol 2021; 900:174066. [PMID: 33789156 PMCID: PMC8111419 DOI: 10.1016/j.ejphar.2021.174066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 03/17/2021] [Accepted: 03/22/2021] [Indexed: 11/19/2022]
Abstract
Methamphetamine (MA) abuse is associated with the development of pulmonary arterial hypertension (PAH) and subsequent right ventricular failure. A recent clinical study demonstrated that female sex is a major risk factor for MA-induced PAH. The mechanisms associated with increased prevalence and severity of MA-induced PAH in females are still unclear. We hypothesized that MA may promote changes in gene expression in the right ventricle contributing to the development and/or worsening of PAH in females. Male and female C57BL/6 mice were treated with either MA or vehicle. Right and left ventricular systolic pressures (RVSP and LVSP, respectively) were assessed and tissue samples were collected for gene expression and histology. LVSP and RVSP were not affected by MA in either males or females. Right ventricular hypertrophy was significantly increased by MA in females but it was not affected by MA in males. In the female mice, MA-induced right ventricular hypertrophy was associated with increased expression of brain natriuretic peptide gene and members of the TGF-β receptor signaling pathway such as TGF-β receptor-1, smad3 and smad7. In male mice, there were no changes in right ventricular gene expression. Our results suggest that MA caused right ventricular hypertrophy in female mice, but not in males and that this was associated with an increase in hypertrophic genes. The right ventricular hypertrophy was not dependent on increased RVSP suggesting a direct effect of MA on the right ventricle. If this translates to PAH patients, it might explain the poor outcome observed in MA-associated female PAH patients.
Collapse
Affiliation(s)
- Hicham Labazi
- Institute of Cardiovascular & Medical Sciences and College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK; Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK.
| | - Margaret Nilsen
- Institute of Cardiovascular & Medical Sciences and College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK; Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Margaret R MacLean
- Institute of Cardiovascular & Medical Sciences and College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK; Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
23
|
Song JL, Zheng SY, He RL, Gui LX, Lin MJ, Sham JSK. Serotonin and chronic hypoxic pulmonary hypertension activate a NADPH oxidase 4 and TRPM2 dependent pathway for pulmonary arterial smooth muscle cell proliferation and migration. Vascul Pharmacol 2021; 138:106860. [PMID: 33794383 DOI: 10.1016/j.vph.2021.106860] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 03/17/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023]
Abstract
5-Hydroxytryptamine (5-HT)-dependent signaling mediated through its transporters and receptors plays important roles in chronic hypoxic pulmonary hypertension (CHPH), which is associated with aberrant reactive oxygen species (ROS) production. NADPH oxidase 4 (NOX4) is one of the major sources of ROS in pulmonary vasculature, and has been implicated in the development of PH. NOX4 generates H2O2, which can activate the transient receptor potential melastatin 2 (TRPM2) channels, providing Ca2+ signals for cell proliferation and migration. However, the connection between 5-HT, NOX4, ROS and TRPM2 in the context of PH has not been established. Here we examined the level of 5-HT and expression of NOX4 and TRPM2, and their roles in pulmonary arterial smooth muscle cells (PASMCs) proliferation and migration. NOX4 and TRPM2 were upregulated in pulmonary arteries of CHPH rats, which were associated with elevated levels of 5-HT and ROS, and enhanced proliferation and migration in PASMCs. The increase in ROS, and the enhanced proliferation and migration of PASMCs from CHPH rats were mimicked by treating normoxic PASMCs with 5-HT. 5-HT; and CH-induced ROS production were reversed by catalase, the NOX1/NOX4 inhibitor GKT137831, and Nox4 siRNA. 5-HT and H2O2 elicited Ca2+ responses were significantly augmented in CHPH PASMCs; and the augmented Ca2+ responses were obliterated by the 2-Aminoethoxydiphenyl borate (2-APB) and Trpm2-specific siRNA. Moreover, 5-HT and CH-induced proliferation and migration were suppressed by Nox4 or Trpm2 siRNA; and simultaneous transfection of both siRNA did not cause further inhibition. These results suggest that the 5-HT and CH-induced PASMC proliferation and migration were mediated, at least in part, by TRPM2 via activation of NOX4-dependent ROS production; and revealed a novel NOX4-ROS-TRPM2 signaling pathway for the pathogenesis of CHPH.
Collapse
MESH Headings
- Animals
- Calcium Signaling
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Chronic Disease
- Disease Models, Animal
- Hypoxia/complications
- Male
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- NADPH Oxidase 4/genetics
- NADPH Oxidase 4/metabolism
- Pulmonary Arterial Hypertension/enzymology
- Pulmonary Arterial Hypertension/etiology
- Pulmonary Arterial Hypertension/pathology
- Pulmonary Arterial Hypertension/physiopathology
- Pulmonary Artery/drug effects
- Pulmonary Artery/enzymology
- Pulmonary Artery/pathology
- Pulmonary Artery/physiopathology
- Rats, Sprague-Dawley
- Reactive Oxygen Species/metabolism
- Serotonin/metabolism
- Serotonin/pharmacology
- TRPM Cation Channels/genetics
- TRPM Cation Channels/metabolism
- Vascular Remodeling/drug effects
- Rats
Collapse
Affiliation(s)
- Jia-Lin Song
- Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, PR China
| | - Si-Yi Zheng
- Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, PR China
| | - Rui-Lan He
- Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, PR China
| | - Long-Xin Gui
- Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, PR China
| | - Mo-Jun Lin
- Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, PR China.
| | - James S K Sham
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
24
|
Liu Y, Nie X, Zhu J, Wang T, Li Y, Wang Q, Sun Z. NDUFA4L2 in smooth muscle promotes vascular remodeling in hypoxic pulmonary arterial hypertension. J Cell Mol Med 2021; 25:1221-1237. [PMID: 33340241 PMCID: PMC7812284 DOI: 10.1111/jcmm.16193] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 11/10/2020] [Accepted: 11/21/2020] [Indexed: 12/12/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by a progressive increase in pulmonary vascular resistance and obliterative pulmonary vascular remodelling (PVR). The imbalance between the proliferation and apoptosis of pulmonary artery smooth muscle cells (PASMCs) is an important cause of PVR leading to PAH. Mitochondria play a key role in the production of hypoxia-induced pulmonary hypertension (HPH). However, there are still many issues worth studying in depth. In this study, we demonstrated that NADH dehydrogenase (ubiquinone) 1 alpha subcomplex 4 like 2 (NDUFA4L2) was a proliferation factor and increased in vivo and in vitro through various molecular biology experiments. HIF-1α was an upstream target of NDUFA4L2. The plasma levels of 4-hydroxynonene (4-HNE) were increased both in PAH patients and hypoxic PAH model rats. Knockdown of NDUFA4L2 decreased the levels of malondialdehyde (MDA) and 4-HNE in human PASMCs in hypoxia. Elevated MDA and 4-HNE levels might be associated with excessive ROS generation and increased expression of 5-lipoxygenase (5-LO) in hypoxia, but this effect was blocked by siNDUFA4L2. Further research found that p38-5-LO was a downstream signalling pathway of PASMCs proliferation induced by NDUFA4L2. Up-regulated NDUFA4L2 plays a critical role in the development of HPH, which mediates ROS production and proliferation of PASMCs, suggesting NDUFA4L2 as a potential new therapeutic target for PAH.
Collapse
MESH Headings
- Aldehydes/metabolism
- Animals
- Arachidonate 5-Lipoxygenase/metabolism
- Cell Hypoxia
- Cell Proliferation
- Disease Models, Animal
- Electron Transport Complex I/genetics
- Electron Transport Complex I/metabolism
- Endothelial Cells/metabolism
- Gene Expression Regulation
- Gene Silencing
- Humans
- Hypoxia/complications
- Hypoxia/physiopathology
- Male
- Malondialdehyde/metabolism
- Models, Biological
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Oxidation-Reduction
- Oxygen Consumption
- Pulmonary Arterial Hypertension/complications
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/pathology
- Pulmonary Arterial Hypertension/physiopathology
- Pulmonary Artery/pathology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Wistar
- Reactive Oxygen Species/metabolism
- Vascular Remodeling/genetics
- p38 Mitogen-Activated Protein Kinases/metabolism
- Rats
Collapse
Affiliation(s)
- Yun Liu
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, China
- Department of Pharmacy, The Affiliated Lianyungang Hospital of Xuzhou Medical University/The First People's Hospital of Lianyungang, Lianyungang, China
| | - Xiaowei Nie
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Shenzhen, China
- Lung Transplant Group, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Jinquan Zhu
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Tianyan Wang
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Yanli Li
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Qian Wang
- Department of Anesthesiology, Children's Hospital of Soochow University, Suzhou, China
| | - Zengxian Sun
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, China
- Department of Pharmacy, The Affiliated Lianyungang Hospital of Xuzhou Medical University/The First People's Hospital of Lianyungang, Lianyungang, China
| |
Collapse
|
25
|
景 灵, 赵 华, 刘 观. [Comparison of two techniques for lateral collateral ligament complex repair in treatment of terrible triad of elbow]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2020; 34:1550-1554. [PMID: 33319534 PMCID: PMC8171572 DOI: 10.7507/1002-1892.201908111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 09/21/2020] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To compare the effectiveness of transosseous tunnel fixation and drilling fixation for repair of lateral collateral ligament complex (LCLC) in treatment of terrible triad of elbow (TTE). METHODS A clinical data of 50 patients with TTE between June 2012 and January 2018 were retrospectively analyzed. The LCLC was repaired with transosseous tunnel fixation in 22 patients (transosseous tunnel fixation group) and with drilling fixation in 28 patients (drilling fixation group). There was no significant difference between the two groups ( P>0.05) in gender, age, fracture side, time from injury to admission, coronoid process fracture classification, radial head fracture classification, and TTE classification. The operation time, intraoperative blood loss, fracture healing time, and complications of the two groups were recorded. At last follow-up, the Mayo elbow performance system (MEPS) score, range of motion of elbow joint, and Broberg-Morrey classification were recorded. RESULTS The operation of two groups were successfully completed. There was no significant difference in the operation time and intraoperative blood loss between the two group ( P>0.05). The follow-up time was (24.43±6.84) months in the transosseous tunnel fixation group and (21.55±6.16) months in the drilling fixation group, and the difference was not significant ( t=1.534, P=0.132). X-ray films showed that the coronoid process and radial head fractures in the two groups healed, and there was no significant difference in the healing time ( P>0.05). At last follow-up, there was no significant difference in the flexion-extension activity, rotation activity, MEPS score, and Broberg-Morrey grading ( P>0.05). During the follow-up, there was no re-dislocation or instability of the elbow joint. The incidence of complication was 28.57% (8/28) in the transosseous tunnel fixation group and 27.27% (6/22) in the drilling fixation group, showing no significant difference ( χ 2=2.403, P=0.121). CONCLUSION Both transosseous tunnel fixation and drilling fixation can achieve good results in repair of LCLC for TTE.
Collapse
Affiliation(s)
- 灵勇 景
- 宁波大学医学院(浙江宁波 315211)Medicine School of Ningbo University, Ningbo Zhejiang, 315211, P.R.China
| | - 华国 赵
- 宁波大学医学院(浙江宁波 315211)Medicine School of Ningbo University, Ningbo Zhejiang, 315211, P.R.China
| | - 观燚 刘
- 宁波大学医学院(浙江宁波 315211)Medicine School of Ningbo University, Ningbo Zhejiang, 315211, P.R.China
| |
Collapse
|
26
|
Yan S, Resta TC, Jernigan NL. Vasoconstrictor Mechanisms in Chronic Hypoxia-Induced Pulmonary Hypertension: Role of Oxidant Signaling. Antioxidants (Basel) 2020; 9:E999. [PMID: 33076504 PMCID: PMC7602539 DOI: 10.3390/antiox9100999] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/06/2020] [Accepted: 10/06/2020] [Indexed: 02/06/2023] Open
Abstract
Elevated resistance of pulmonary circulation after chronic hypoxia exposure leads to pulmonary hypertension. Contributing to this pathological process is enhanced pulmonary vasoconstriction through both calcium-dependent and calcium sensitization mechanisms. Reactive oxygen species (ROS), as a result of increased enzymatic production and/or decreased scavenging, participate in augmentation of pulmonary arterial constriction by potentiating calcium influx as well as activation of myofilament sensitization, therefore mediating the development of pulmonary hypertension. Here, we review the effects of chronic hypoxia on sources of ROS within the pulmonary vasculature including NADPH oxidases, mitochondria, uncoupled endothelial nitric oxide synthase, xanthine oxidase, monoamine oxidases and dysfunctional superoxide dismutases. We also summarize the ROS-induced functional alterations of various Ca2+ and K+ channels involved in regulating Ca2+ influx, and of Rho kinase that is responsible for myofilament Ca2+ sensitivity. A variety of antioxidants have been shown to have beneficial therapeutic effects in animal models of pulmonary hypertension, supporting the role of ROS in the development of pulmonary hypertension. A better understanding of the mechanisms by which ROS enhance vasoconstriction will be useful in evaluating the efficacy of antioxidants for the treatment of pulmonary hypertension.
Collapse
Affiliation(s)
| | | | - Nikki L. Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; (S.Y.); (T.C.R.)
| |
Collapse
|
27
|
Docherty CK, Denver N, Fisher S, Nilsen M, Hillyard D, Openshaw RL, Labazi H, MacLean MR. Direct Delivery of MicroRNA96 to the Lungs Reduces Progression of Sugen/Hypoxia-Induced Pulmonary Hypertension in the Rat. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 22:396-405. [PMID: 33230444 PMCID: PMC7533346 DOI: 10.1016/j.omtn.2020.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/03/2020] [Indexed: 12/21/2022]
Abstract
The 5HT1B receptor (5HT1BR) contributes to the pathogenic effects of serotonin in pulmonary arterial hypertension. Here, we determine the effect of a microRNA96 (miR96) mimic delivered directly to the lungs on development of severe pulmonary hypertension in rats. Female rats were dosed with sugen (30 mg/kg) and subjected to 3 weeks of hypobaric hypoxia. In normoxia, rats were dosed with either a 5HT1BR antagonist SB216641 (7.5 mg/kg/day for 3 weeks), miR96, or scramble sequence (50 μg per rat), delivered by intratracheal (i.t) administration, once a week for 3 weeks. Cardiac hemodynamics were determined, pulmonary vascular remodeling was assessed, and gene expression was assessed by qRT-PCR, and in situ hybridization and protein expression were assessed by western blot and ELISA. miR96 expression was increased in pulmonary arteries and associated with a downregulation of the 5HT1BR protein in the lung. miR96 reduced progression of right ventricular systolic pressure, pulmonary arterial remodeling, right ventricular hypertrophy, and the occurrence of occlusive pulmonary lesions. Importantly, miR96 had no off-target effects and did not affect fibrotic markers of liver and kidney function. In conclusion, direct delivery of miR96 to the lungs was effective, reducing progression of sugen/hypoxia-induced pulmonary hypertension with no measured off-target effects. miR96 may be a novel therapy for pulmonary arterial hypertension, acting through downregulation of 5HT1BR.
Collapse
Affiliation(s)
- Craig K Docherty
- Strathclyde Institute of Pharmacy and Biological Sciences, University of Strathclyde, Glasgow G4 0RE, Scotland.,Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, Scotland
| | - Nina Denver
- Strathclyde Institute of Pharmacy and Biological Sciences, University of Strathclyde, Glasgow G4 0RE, Scotland
| | - Simon Fisher
- Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, Scotland
| | - Margaret Nilsen
- Strathclyde Institute of Pharmacy and Biological Sciences, University of Strathclyde, Glasgow G4 0RE, Scotland.,Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, Scotland
| | - Dianne Hillyard
- Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, Scotland
| | - Rebecca L Openshaw
- Strathclyde Institute of Pharmacy and Biological Sciences, University of Strathclyde, Glasgow G4 0RE, Scotland
| | - Hicham Labazi
- Strathclyde Institute of Pharmacy and Biological Sciences, University of Strathclyde, Glasgow G4 0RE, Scotland
| | - Margaret R MacLean
- Strathclyde Institute of Pharmacy and Biological Sciences, University of Strathclyde, Glasgow G4 0RE, Scotland.,Institute of Cardiovascular and Medical Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, Scotland
| |
Collapse
|
28
|
Sun Y, Lu Y, Saredy J, Wang X, Drummer Iv C, Shao Y, Saaoud F, Xu K, Liu M, Yang WY, Jiang X, Wang H, Yang X. ROS systems are a new integrated network for sensing homeostasis and alarming stresses in organelle metabolic processes. Redox Biol 2020; 37:101696. [PMID: 32950427 PMCID: PMC7767745 DOI: 10.1016/j.redox.2020.101696] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/17/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS) are critical for the progression of cardiovascular diseases, inflammations and tumors. However, the mechanisms of how ROS sense metabolic stress, regulate metabolic pathways and initiate proliferation, inflammation and cell death responses remain poorly characterized. In this analytic review, we concluded that: 1) Based on different features and functions, eleven types of ROS can be classified into seven functional groups: metabolic stress-sensing, chemical connecting, organelle communication, stress branch-out, inflammasome-activating, dual functions and triple functions ROS. 2) Among the ROS generation systems, mitochondria consume the most amount of oxygen; and nine types of ROS are generated; thus, mitochondrial ROS systems serve as the central hub for connecting ROS with inflammasome activation, trained immunity and immunometabolic pathways. 3) Increased nuclear ROS production significantly promotes cell death in comparison to that in other organelles. Nuclear ROS systems serve as a convergent hub and decision-makers to connect unbearable and alarming metabolic stresses to inflammation and cell death. 4) Balanced ROS levels indicate physiological homeostasis of various metabolic processes in subcellular organelles and cytosol, while imbalanced ROS levels present alarms for pathological organelle stresses in metabolic processes. Based on these analyses, we propose a working model that ROS systems are a new integrated network for sensing homeostasis and alarming stress in metabolic processes in various subcellular organelles. Our model provides novel insights on the roles of the ROS systems in bridging metabolic stress to inflammation, cell death and tumorigenesis; and provide novel therapeutic targets for treating those diseases. (Word count: 246).
Collapse
Affiliation(s)
- Yu Sun
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - Yifan Lu
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - Jason Saredy
- Metabolic Disease Research and Cardiovascular Research and Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Xianwei Wang
- Metabolic Disease Research and Cardiovascular Research and Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Charles Drummer Iv
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - Ying Shao
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - Fatma Saaoud
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - Keman Xu
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - Ming Liu
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA
| | - William Y Yang
- Metabolic Disease Research and Cardiovascular Research and Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Xiaohua Jiang
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA; Metabolic Disease Research and Cardiovascular Research and Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Hong Wang
- Metabolic Disease Research and Cardiovascular Research and Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Xiaofeng Yang
- Centers for Cardiovascular Research and Inflammation, Translational and Clinical Lung Research, USA; Metabolic Disease Research and Cardiovascular Research and Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
29
|
Shi J, Yang Y, Cheng A, Xu G, He F. Metabolism of vascular smooth muscle cells in vascular diseases. Am J Physiol Heart Circ Physiol 2020; 319:H613-H631. [PMID: 32762559 DOI: 10.1152/ajpheart.00220.2020] [Citation(s) in RCA: 166] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Vascular smooth muscle cells (VSMCs) are the fundamental component of the medial layer of arteries and are essential for arterial physiology and pathology. It is becoming increasingly clear that VSMCs can alter their metabolism to fulfill the bioenergetic and biosynthetic requirements. During vascular injury, VSMCs switch from a quiescent "contractile" phenotype to a highly migratory and proliferative "synthetic" phenotype. Recent studies have found that the phenotype switching of VSMCs is driven by a metabolic switch. Metabolic pathways, including aerobic glycolysis, fatty acid oxidation, and amino acid metabolism, have distinct, indispensable roles in normal and dysfunctional vasculature. VSMCs metabolism is also related to the metabolism of endothelial cells. In the present review, we present a brief overview of VSMCs metabolism and how it regulates the progression of several vascular diseases, including atherosclerosis, systemic hypertension, diabetes, pulmonary hypertension, vascular calcification, and aneurysms, and the effect of the risk factors for vascular disease (aging, cigarette smoking, and excessive alcohol drinking) on VSMC metabolism to clarify the role of VSMCs metabolism in the key pathological process.
Collapse
Affiliation(s)
- Jia Shi
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Yang
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anying Cheng
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Xu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan He
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
30
|
Spaczyńska M, Rocha SF, Oliver E. Pharmacology of Pulmonary Arterial Hypertension: An Overview of Current and Emerging Therapies. ACS Pharmacol Transl Sci 2020; 3:598-612. [PMID: 32832865 DOI: 10.1021/acsptsci.0c00048] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Indexed: 12/21/2022]
Abstract
Pulmonary arterial hypertension is a rare and devastating disease characterized by an abnormal chronic increase in pulmonary arterial pressure above 20 mmHg at rest, with a poor prognosis if not treated. Currently, there is not a single fully effective therapy, even though a dozen of drugs have been developed in the last decades. Pulmonary arterial hypertension is a multifactorial disease, meaning that several molecular mechanisms are implicated in its pathology. The main molecular pathways regulating the pulmonary vasomotor tone-endothelin, nitric oxide, and prostacyclin-are the most biologically and therapeutically explored to date. However, drugs targeting these pathways have already found their limitations. In the last years, translational research and clinical trials have made a strong effort in suggesting and testing novel therapeutic strategies for this disease. These approaches involve targeting the main molecular pathways with novel drugs, drug repurposing for novel targets, and also using combinatorial therapies. In this review, we summarize current strategies and drugs targeting the endothelin, nitric oxide, and prostacyclin pathways, as well as, the emerging new drugs proposed to cope with vascular remodelling, metabolic switch, perivascular inflammation, epigenetic modifications, estrogen deregulation, serotonin, and other neurohumoral mechanisms characteristic of this disease. Nowadays, pulmonary arterial hypertension remains an incurable disease; however, the incoming new knowledge makes us believe that new promising therapies are coming to the clinical arena soon.
Collapse
Affiliation(s)
- Monika Spaczyńska
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, 28029, Spain
| | - Susana F Rocha
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, 28029, Spain
| | - Eduardo Oliver
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, 28029, Spain.,Centro de Investigaciones Biomédicas en Red Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, 28029, Spain
| |
Collapse
|
31
|
Mair KM, Gaw R, MacLean MR. Obesity, estrogens and adipose tissue dysfunction - implications for pulmonary arterial hypertension. Pulm Circ 2020; 10:2045894020952019. [PMID: 32999709 PMCID: PMC7506791 DOI: 10.1177/2045894020952023] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 08/02/2020] [Indexed: 12/12/2022] Open
Abstract
Obesity is a prevalent global public health issue characterized by excess body fat. Adipose tissue is now recognized as an important endocrine organ releasing an abundance of bioactive adipokines including, but not limited to, leptin, adiponectin and resistin. Obesity is a common comorbidity amongst pulmonary arterial hypertension patients, with 30% to 40% reported as obese, independent of other comorbidities associated with pulmonary arterial hypertension (e.g. obstructive sleep apnoea). An 'obesity paradox' has been observed, where obesity has been associated with subclinical right ventricular dysfunction but paradoxically may confer a protective effect on right ventricular function once pulmonary hypertension develops. Obesity and pulmonary arterial hypertension share multiple pathophysiological mechanisms including inflammation, oxidative stress, elevated leptin (proinflammatory) and reduced adiponectin (anti-inflammatory). The female prevalence of pulmonary arterial hypertension has instigated the hypothesis that estrogens may play a causative role in its development. Adipose tissue, a major site for storage and metabolism of sex steroids, is the primary source of estrogens and circulating estrogens levels which are elevated in postmenopausal women and men with pulmonary arterial hypertension. This review discusses the functions of adipose tissue in both health and obesity and the links between obesity and pulmonary arterial hypertension. Shared pathophysiological mechanisms and the contribution of specific fat depots, metabolic and sex-dependent differences are discussed.
Collapse
Affiliation(s)
- Kirsty M. Mair
- Strathclyde Institute of Pharmacy and Biomedical
Sciences (SIPBS), University of Strathclyde, Glasgow, UK
| | - Rosemary Gaw
- Strathclyde Institute of Pharmacy and Biomedical
Sciences (SIPBS), University of Strathclyde, Glasgow, UK
| | - Margaret R. MacLean
- Strathclyde Institute of Pharmacy and Biomedical
Sciences (SIPBS), University of Strathclyde, Glasgow, UK
| |
Collapse
|
32
|
Alcántara-Vázquez O, Villamil-Hernández MT, Sánchez-López A, Pertz HH, Villalón CM, Centurión D. Blocking properties of terguride at the 5-HT 2 receptor subtypes mediating cardiovascular responses in the rat. Can J Physiol Pharmacol 2020; 98:511-521. [PMID: 32268074 DOI: 10.1139/cjpp-2019-0591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In vitro studies have suggested that terguride blocks the contractile and relaxant responses produced by 5-hydroxytryptamine (5-HT) via 5-HT2A/2B receptors. This study has now investigated terguride's blocking properties on central/peripheral 5-HT2 receptors in anaesthetized or pithed rats. Male Wistar anaesthetized/pithed rats were cannulated for recording blood pressure and heart rate and for i.v. administration of several compounds. In both groups of rats, i.v. bolus injections of 5-HT or (±)-DOI (a 5-HT2 receptor agonist; 1-1000 μg/kg) produced dose-dependent increases in diastolic blood pressure and heart rate. These responses were dose-dependently antagonized by terguride (10-3000 μg/kg). In anaesthetized rats, i.v. bolus injections of BW723C86 (a 5-HT2B receptor agonist; 1-1000 μg/kg) produced dose-dependent increases in diastolic blood pressure and not dose-dependent increases in heart rate, while in pithed rats, these responses were attenuated. The vasopressor responses elicited by BW723C86 in anaesthetized rats were dose-dependently blocked by terguride (10-300 μg/kg), whereas its the tachycardic responses were dose-independently blocked. These results, taken together, suggest that terguride behaved as an antagonist at the 5-HT2 receptors located in the central nervous system and (or) the systemic vasculature. This is the first evidence demonstrating that terguride can block central/peripheral 5-HT2 receptors mediating cardiovascular responses in anaesthetized or pithed rats.
Collapse
Affiliation(s)
- Oscar Alcántara-Vázquez
- Centro Interdisciplinario de Ciencias de la Salud, Unidad Milpa Alta, IPN, Ex-Hacienda del Mayorazgo, Km. 39.5 Carretera Xochimilco-Oaxtepec, C.P 12000, México City, México
| | - Ma Trinidad Villamil-Hernández
- Centro Interdisciplinario de Ciencias de la Salud, Unidad Milpa Alta, IPN, Ex-Hacienda del Mayorazgo, Km. 39.5 Carretera Xochimilco-Oaxtepec, C.P 12000, México City, México
| | - Araceli Sánchez-López
- Departamento de Farmacobiología, Cinvestav-Coapa, Czda. de los Tenorios 235, Col. Granjas-Coapa, Deleg. Tlalpan, C.P.14330, Mexico City, Mexico
| | - Heinz H Pertz
- Institut für Pharmazie, Freie Universität Berlin, Königin-Luise-Str. 2, 14195 Berlin (Dahlem), Germany
| | - Carlos M Villalón
- Departamento de Farmacobiología, Cinvestav-Coapa, Czda. de los Tenorios 235, Col. Granjas-Coapa, Deleg. Tlalpan, C.P.14330, Mexico City, Mexico
| | - David Centurión
- Departamento de Farmacobiología, Cinvestav-Coapa, Czda. de los Tenorios 235, Col. Granjas-Coapa, Deleg. Tlalpan, C.P.14330, Mexico City, Mexico
| |
Collapse
|
33
|
Gomez AP, Moreno MJ, Hernández A. Adventitial growth and lung connective tissue growth factor expression in pulmonary arterioles due to hypobaric hypoxia in broilers. Poult Sci 2020; 99:1832-1837. [PMID: 32241463 PMCID: PMC7587700 DOI: 10.3382/ps/pez157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 03/11/2019] [Indexed: 11/25/2022] Open
Abstract
Forty broilers maintained under natural hypobaric hypoxia (2,638 m above sea level) and 20 maintained under relative normoxia (460 m above sea level) were selected as pulmonary hypertensive (PHB) and nonpulmonary hypertensive (NPHB), to estimate the degree of the adventitial vascular thickness in lung arterioles and connective tissue growth factor (CTGF) expression in lung. In each group, the adventitial thickness (%AT) of 20 arterioles with 100 to 250 μm of external diameter was measured in lung samples of 24 and 42-day-old broilers. Also, mRNA extraction and real-time reverse transcription-PCR analysis were used to measure lung CTGF expression. The %AT was higher in PHB at 42 D as compared to NPHB at both ages and PHB at 24 D; however, the same differences were not evidenced at 24 D. In the 2 ages evaluated, differences were observed in the %AT between broilers under hypobaric hypoxia (PHB and NPHB) and under relative normoxia (P < 0.01). In broilers subjected to relative normoxia, no significant differences were found at any of the 2 ages. The expression levels of CTGF mRNA were higher in PHB compared to NPHB at the 2 ages. The %AT was higher in PHB with high levels of expression of CTGF mRNA than those NPHB with low expression of CTGF mRNA. This study showed that adventitial thickening is part of the pulmonary hypertension (PH) physiopathology in broilers exposed to hypobaric hypoxia, in which CTGF appears to be a fibrosis enhancer. Although present data suggest that adventitial engrossment could be a time-dependent process, individual susceptibility and the variable time-course of PH pathophysiology have to be considered.
Collapse
Affiliation(s)
- A P Gomez
- Facultad de Medicina Veterinaria y de Zootecnia, Universidad Nacional de Colombia, Bogotá DC 111311, Colombia
| | - M J Moreno
- Facultad de Medicina Veterinaria y de Zootecnia, Universidad Nacional de Colombia, Bogotá DC 111311, Colombia
| | - A Hernández
- Facultad de Medicina Veterinaria y de Zootecnia, Universidad Nacional de Colombia, Bogotá DC 111311, Colombia.
| |
Collapse
|
34
|
Impact of Nutrition on Pulmonary Arterial Hypertension. Nutrients 2020; 12:nu12010169. [PMID: 31936113 PMCID: PMC7019983 DOI: 10.3390/nu12010169] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/26/2019] [Accepted: 01/03/2020] [Indexed: 02/07/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by sustained vasoconstriction, vascular remodeling, inflammation, and in situ thrombosis. Although there have been important advances in the knowledge of the pathophysiology of PAH, it remains a debilitating, limiting, and rapidly progressive disease. Vitamin D and iron deficiency are worldwide health problems of pandemic proportions. Notably, these nutritional alterations are largely more prevalent in PAH patients than in the general population and there are several pieces of evidence suggesting that they may trigger or aggravate disease progression. There are also several case reports associating scurvy, due to severe vitamin C deficiency, with PAH. Flavonoids such as quercetin, isoflavonoids such as genistein, and other dietary polyphenols including resveratrol slow the progression of the disease in animal models of PAH. Finally, the role of the gut microbiota and its interplay with the diet, host immune system, and energy metabolism is emerging in multiple cardiovascular diseases. The alteration of the gut microbiota has also been reported in animal models of PAH. It is thus possible that in the near future interventions targeting the nutritional status and the gut dysbiosis will improve the outcome of these patients.
Collapse
|
35
|
|
36
|
Knock GA. NADPH oxidase in the vasculature: Expression, regulation and signalling pathways; role in normal cardiovascular physiology and its dysregulation in hypertension. Free Radic Biol Med 2019; 145:385-427. [PMID: 31585207 DOI: 10.1016/j.freeradbiomed.2019.09.029] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/29/2019] [Accepted: 09/23/2019] [Indexed: 02/06/2023]
Abstract
The last 20-25 years have seen an explosion of interest in the role of NADPH oxidase (NOX) in cardiovascular function and disease. In vascular smooth muscle and endothelium, NOX generates reactive oxygen species (ROS) that act as second messengers, contributing to the control of normal vascular function. NOX activity is altered in response to a variety of stimuli, including G-protein coupled receptor agonists, growth-factors, perfusion pressure, flow and hypoxia. NOX-derived ROS are involved in smooth muscle constriction, endothelium-dependent relaxation and smooth muscle growth, proliferation and migration, thus contributing to the fine-tuning of blood flow, arterial wall thickness and vascular resistance. Through reversible oxidative modification of target proteins, ROS regulate the activity of protein tyrosine phosphatases, kinases, G proteins, ion channels, cytoskeletal proteins and transcription factors. There is now considerable, but somewhat contradictory evidence that NOX contributes to the pathogenesis of hypertension through oxidative stress. Specific NOX isoforms have been implicated in endothelial dysfunction, hyper-contractility and vascular remodelling in various animal models of hypertension, pulmonary hypertension and pulmonary arterial hypertension, but also have potential protective effects, particularly NOX4. This review explores the multiplicity of NOX function in the healthy vasculature and the evidence for and against targeting NOX for antihypertensive therapy.
Collapse
Affiliation(s)
- Greg A Knock
- Dpt. of Inflammation Biology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, UK.
| |
Collapse
|
37
|
MESH Headings
- Animals
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aorta, Abdominal/physiopathology
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- Gene Expression Regulation
- Humans
- MicroRNAs/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Phenotype
- Signal Transduction
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transcription, Genetic
- Vascular Diseases/genetics
- Vascular Diseases/pathology
- Vascular Diseases/physiopathology
- Vascular Remodeling/physiology
Collapse
Affiliation(s)
- Ning Shi
- Department of Surgery, University of Missouri, Columbia, MO
- Department of Physiology & pharmacology, The University of Georgia, Athens, GA
| | - Xiaohan Mei
- Department of Physiology & pharmacology, The University of Georgia, Athens, GA
| | - Shi-You Chen
- Department of Surgery, University of Missouri, Columbia, MO
- Department of Physiology & pharmacology, The University of Georgia, Athens, GA
- Correspondence to: Shi-You Chen, PhD, Department of Surgery, University of Missouri, 1 Hospital Drive, Columbia, MO 65212, , Tel: (573) 882-3137, Fax: (573)884-4585
| |
Collapse
|
38
|
Lu HS, Schmidt AM, Hegele RA, Mackman N, Rader DJ, Weber C, Daugherty A. Reporting Sex and Sex Differences in Preclinical Studies. Arterioscler Thromb Vasc Biol 2019; 38:e171-e184. [PMID: 30354222 DOI: 10.1161/atvbaha.118.311717] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Hong S Lu
- From the Department of Physiology, Saha Cardiovascular Research Center, University of Kentucky, Lexington (H.S.L., A.D.)
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, New York University Langone Medical Center, New York, NY (A.M.S.)
| | - Robert A Hegele
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada (R.A.H.)
| | - Nigel Mackman
- Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Daniel J Rader
- Department of Medicine (D.J.R.), Perelman School of Medicine, University of Pennsylvania, Philadelphia.,Department of Genetics (D.J.R.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Christian Weber
- Department of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität, Munich, Germany (C.W.).,German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Alan Daugherty
- From the Department of Physiology, Saha Cardiovascular Research Center, University of Kentucky, Lexington (H.S.L., A.D.)
| |
Collapse
|
39
|
Daneva Z, Laubach VE, Sonkusare SK. Novel Regulators and Targets of Redox Signaling in Pulmonary Vasculature. CURRENT OPINION IN PHYSIOLOGY 2019; 9:87-93. [PMID: 31406951 DOI: 10.1016/j.cophys.2019.04.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Dysregulated redox signaling in pulmonary vasculature is central to the development of pulmonary arterial hypertension (PAH) and lung injury. Modulators of reactive oxygen species (ROS) production and downstream signaling targets are critical for mediating the physiological or pathological effects of ROS. Understanding the complex interactions between the modulators and signaling targets of ROS is essential for developing novel strategies to prevent or attenuate lung pathologies. In this review, we discuss recent studies on the modulators and targets of ROS in pulmonary endothelial and smooth muscle cells, their cellular effects, and the disease conditions associated with dysregulated redox signaling.
Collapse
Affiliation(s)
- Zdravka Daneva
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, Charlottesville, VA, 22908, USA
| | - Victor E Laubach
- Department of Surgery, University of Virginia-School of Medicine, Charlottesville, VA, 22908, USA
| | - Swapnil K Sonkusare
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, Charlottesville, VA, 22908, USA.,Department of Molecular Physiology and Biological Physics, University of Virginia-School of Medicine, Charlottesville, VA, 22908, USA
| |
Collapse
|
40
|
Cheng J, Wen J, Wang N, Wang C, Xu Q, Yang Y. Ion Channels and Vascular Diseases. Arterioscler Thromb Vasc Biol 2019; 39:e146-e156. [DOI: 10.1161/atvbaha.119.312004] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Jun Cheng
- From the Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China (J.C., J.W., N.W., Q.X., Y.Y.)
| | - Jing Wen
- From the Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China (J.C., J.W., N.W., Q.X., Y.Y.)
| | - Na Wang
- From the Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China (J.C., J.W., N.W., Q.X., Y.Y.)
| | - Claire Wang
- Gonville and Caius College, University of Cambridge, United Kingdom (C.W.)
| | - Qingbo Xu
- From the Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China (J.C., J.W., N.W., Q.X., Y.Y.)
- School of Cardiovascular Medicine and Sciences, King’s College London BHF Centre, London, United Kingdom (Q.X.)
| | - Yan Yang
- From the Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China (J.C., J.W., N.W., Q.X., Y.Y.)
| |
Collapse
|
41
|
Docherty CK, Harvey KY, Mair KM, Griffin S, Denver N, MacLean MR. The Role of Sex in the Pathophysiology of Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1065:511-528. [PMID: 30051404 DOI: 10.1007/978-3-319-77932-4_31] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease characterised by increased pulmonary vascular resistance and pulmonary artery remodelling as result of increased vascular tone and vascular cell proliferation, respectively. Eventually, this leads to right heart failure. Heritable PAH is caused by a mutation in the bone morphogenetic protein receptor-II (BMPR-II). Female susceptibility to PAH has been known for some time, and most recent figures show a female-to-male ratio of 4:1. Variations in the female sex hormone estrogen and estrogen metabolism modify FPAH risk, and penetrance of the disease in BMPR-II mutation carriers is increased in females. Several lines of evidence point towards estrogen being pathogenic in the pulmonary circulation, and thus increasing the risk of females developing PAH. Recent studies have also suggested that estrogen metabolism may be crucial in the development and progression of PAH with studies indicating that downstream metabolites such as 16α-hydroxyestrone are upregulated in several forms of experimental pulmonary hypertension (PH) and can cause pulmonary artery smooth muscle cell proliferation and subsequent vascular remodelling. Conversely, other estrogen metabolites such as 2-methoxyestradiol have been shown to be protective in the context of PAH. Estrogen may also upregulate the signalling pathways of other key mediators of PAH such as serotonin.
Collapse
Affiliation(s)
- Craig K Docherty
- Research Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Katie Yates Harvey
- Research Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Kirsty M Mair
- Research Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Sinead Griffin
- Research Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Nina Denver
- Research Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Margaret R MacLean
- Research Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
42
|
Huetsch JC, Suresh K, Shimoda LA. Regulation of Smooth Muscle Cell Proliferation by NADPH Oxidases in Pulmonary Hypertension. Antioxidants (Basel) 2019; 8:antiox8030056. [PMID: 30841544 PMCID: PMC6466559 DOI: 10.3390/antiox8030056] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/25/2019] [Accepted: 03/01/2019] [Indexed: 02/07/2023] Open
Abstract
Hyperproliferation of pulmonary arterial smooth muscle cells is a key component of vascular remodeling in the setting of pulmonary hypertension (PH). Numerous studies have explored factors governing the changes in smooth muscle cell phenotype that lead to the increased wall thickness, and have identified various potential candidates. A role for reactive oxygen species (ROS) has been well documented in PH. ROS can be generated from a variety of sources, including mitochondria, uncoupled nitric oxide synthase, xanthine oxidase, and reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. In this article, we will review recent data supporting a role for ROS generated from NADPH oxidases in promoting pulmonary arterial smooth muscle cell proliferation during PH.
Collapse
Affiliation(s)
- John C Huetsch
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21224, USA.
| | - Karthik Suresh
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21224, USA.
| | - Larissa A Shimoda
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21224, USA.
| |
Collapse
|
43
|
Majesky MW. Vascular Development. Arterioscler Thromb Vasc Biol 2019; 38:e17-e24. [PMID: 29467221 DOI: 10.1161/atvbaha.118.310223] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 01/22/2018] [Indexed: 12/13/2022]
Abstract
The vascular system forms as a branching network of endothelial cells that acquire identity as arterial, venous, hemogenic, or lymphatic. Endothelial specification depends on gene targets transcribed by Ets domain-containing factors, including Ets variant gene 2 (Etv2), together with the activity of chromatin-remodeling complexes containing Brahma-related gene-1 (Brg1). Once specified and assembled into vessels, mechanisms regulating lumen diameter and axial growth ensure that the structure of the branching vascular network matches the need for perfusion of target tissues. In addition, blood vessels provide important morphogenic cues that guide or direct the development of organs forming around them. As the embryo grows and lumen diameters increase, smooth muscle cells wrap around the nascent vessel walls to provide mechanical strength and vasomotor control of the circulation. Increasing mechanical stretch and wall strain promote smooth muscle cell differentiation via coupling of actin cytoskeletal remodeling to myocardin and serum response factor-dependent transcription. Remodeling of artery walls by developmental signaling pathways reappears in postnatal blood vessels during physiological and pathological adaptation to vessel wall injury, inflammation, or chronic hypoxia. Recent reports providing insights into major steps in vascular development are reviewed here with a particular emphasis on studies that have been recently published in Arteriosclerosis, Thrombosis, and Vascular Biology.
Collapse
Affiliation(s)
- Mark W Majesky
- From the Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, WA; and Departments of Pediatrics and Pathology, University of Washington, Seattle.
| |
Collapse
|
44
|
Abstract
SIGNIFICANCE G protein-coupled receptors (GPCR) are the largest group of cell surface receptors, which link cells to their environment. Reactive oxygen species (ROS) can act as important cellular signaling molecules. The family of NADPH oxidases generates ROS in response to activated cell surface receptors. Recent Advances: Various signaling pathways linking GPCRs and activation of NADPH oxidases have been characterized. CRITICAL ISSUES Still, a more detailed analysis of G proteins involved in the GPCR-mediated activation of NADPH oxidases is needed. In addition, a more precise discrimination of NADPH oxidase activation due to either upregulation of subunit expression or post-translational subunit modifications is needed. Also, the role of noncanonical modulators of NADPH oxidase activation in the response to GPCRs awaits further analyses. FUTURE DIRECTIONS As GPCRs are one of the most popular classes of investigational drug targets, further detailing of G protein-coupled mechanisms in the activation mechanism of NADPH oxidases as well as better understanding of the link between newly identified NADPH oxidase interaction partners and GPCR signaling will provide new opportunities for improved efficiency and decreased off target effects of therapies targeting GPCRs.
Collapse
Affiliation(s)
- Andreas Petry
- 1 Experimental and Molecular Pediatric Cardiology, German Heart Center Munich , TU Munich, Munich, Germany
| | - Agnes Görlach
- 1 Experimental and Molecular Pediatric Cardiology, German Heart Center Munich , TU Munich, Munich, Germany .,2 DZHK (German Centre for Cardiovascular Research) , Partner Site Munich, Munich Heart Alliance, Munich, Germany
| |
Collapse
|
45
|
Strassheim D, Karoor V, Stenmark K, Verin A, Gerasimovskaya E. A current view of G protein-coupled receptor - mediated signaling in pulmonary hypertension: finding opportunities for therapeutic intervention. ACTA ACUST UNITED AC 2018; 2. [PMID: 31380505 PMCID: PMC6677404 DOI: 10.20517/2574-1209.2018.44] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pathological vascular remodeling is observed in various cardiovascular diseases including pulmonary hypertension (PH), a disease of unknown etiology that has been characterized by pulmonary artery vasoconstriction, right ventricular hypertrophy, vascular inflammation, and abnormal angiogenesis in pulmonary circulation. G protein-coupled receptors (GPCRs) are the largest family in the genome and widely expressed in cardiovascular system. They regulate all aspects of PH pathophysiology and represent therapeutic targets. We overview GPCRs function in vasoconstriction, vasodilation, vascular inflammation-driven remodeling and describe signaling cross talk between GPCR, inflammatory cytokines, and growth factors. Overall, the goal of this review is to emphasize the importance of GPCRs as critical signal transducers and targets for drug development in PH.
Collapse
Affiliation(s)
- Derek Strassheim
- Departments of Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| | - Vijaya Karoor
- Departments of Medicine, University of Colorado Denver, Aurora, CO 80045, USA.,Cardiovascular and Pulmonary Research laboratories, University of Colorado Denver, Aurora, CO 80045, USA
| | - Kurt Stenmark
- Cardiovascular and Pulmonary Research laboratories, University of Colorado Denver, Aurora, CO 80045, USA.,Department of Pediatrics, Pulmonary and Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| | - Alexander Verin
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
| | - Evgenia Gerasimovskaya
- Cardiovascular and Pulmonary Research laboratories, University of Colorado Denver, Aurora, CO 80045, USA.,Department of Pediatrics, Pulmonary and Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| |
Collapse
|
46
|
Aaronson PI. Actin polymerization contributes to ROS- and Rho-dependent Ca 2+ sensitization in pulmonary arteries from chronic hypoxic rats. Am J Physiol Heart Circ Physiol 2018; 315:H314-H317. [DOI: 10.1152/ajpheart.00135.2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Philip I. Aaronson
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
47
|
Iyinikkel J, Murray F. GPCRs in pulmonary arterial hypertension: tipping the balance. Br J Pharmacol 2018; 175:3063-3079. [PMID: 29468655 PMCID: PMC6031878 DOI: 10.1111/bph.14172] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 01/30/2018] [Accepted: 02/01/2018] [Indexed: 02/06/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive, fatal disease characterised by increased pulmonary vascular resistance and excessive proliferation of pulmonary artery smooth muscle cells (PASMC). GPCRs, which are attractive pharmacological targets, are important regulators of pulmonary vascular tone and PASMC phenotype. PAH is associated with the altered expression and function of a number of GPCRs in the pulmonary circulation, which leads to the vasoconstriction and proliferation of PASMC and thereby contributes to the imbalance of pulmonary vascular tone associated with PAH; drugs targeting GPCRs are currently used clinically to treat PAH and extensive preclinical work supports the utility of a number of additional GPCRs. Here we review how GPCR expression and function changes with PAH and discuss why GPCRs continue to be relevant drug targets for the disease.
Collapse
Affiliation(s)
- Jean Iyinikkel
- College of Life Sciences and Medicine, School of Medicine, Medical Sciences and NutritionUniversity of AberdeenAberdeenUK
| | - Fiona Murray
- College of Life Sciences and Medicine, School of Medicine, Medical Sciences and NutritionUniversity of AberdeenAberdeenUK
| |
Collapse
|
48
|
MacLean MMR. The serotonin hypothesis in pulmonary hypertension revisited: targets for novel therapies (2017 Grover Conference Series). Pulm Circ 2018; 8:2045894018759125. [PMID: 29468941 PMCID: PMC5826007 DOI: 10.1177/2045894018759125] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Increased synthesis of serotonin and/or activity of serotonin in pulmonary arteries has been implicated in the pathobiology of pulmonary arterial hypertension (PAH). The incidence of PAH associated with diet pills such as aminorex, fenfluramine, and chlorphentermine initially led to the “serotonin hypothesis of pulmonary hypertension.” Over the last couple of decades there has been an accumulation of convincing evidence that targeting serotonin synthesis or signaling is a novel and promising approach to the development of novel therapies for PAH. Pulmonary endothelial serotonin synthesis via tryptophan hydroxlase 1 (TPH1) is increased in patients with PAH and serotonin can act in a paracrine fashion on underlying pulmonary arterial smooth muscle cells (PASMCs), In humans, serotonin can enter PASMCs via the serotonin transporter (SERT) or activate the 5-HT1B receptor; 5-HT1B activation and SERT activity cooperate to induce PASMC contraction and proliferation via activation of downstream proliferative and contractile signaling pathways. Here we will review the current status of the serotonin hypothesis and discuss potential and novel therapeutic targets.
Collapse
Affiliation(s)
- Margaret Mandy R MacLean
- Research Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
49
|
Castillo JG, Naib T, Zacks JS, Adams DH. Echocardiography in functional midgut neuroendocrine tumors: When and how often. Rev Endocr Metab Disord 2017; 18:411-421. [PMID: 29080935 DOI: 10.1007/s11154-017-9434-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The management of patients with midgut neuroendocrine tumors (MNET) is rapidly evolving. Current preoperative detection rates of primary tumor sites are higher than ever and progression-free survival in patients with already advanced disease is expanding due to the implementation of novel efficacious treatment strategies. This survival benefit may potentially translate into a need for a multidisciplinary approach to an even more heterogenous variety of clinical conditions, among these, carcinoid syndrome (CS) and carcinoid heart disease (CHD). The latter often triggers substantial morbidity and mortality, hence a systematic screening, an accurate diagnosis, as well as effective interventions are critically important. The rarity of the disease has result in a relative lack of statistically powerful evidence, which in turn may have rendered significant variability between practices. In this regard, despite recent guidelines, the optimal follow-up of patients with CHD remain debatable to some authors, perhaps due to the preponderance of certain schools throughout the manuscript. Herein, we present a concise and practical guidance document on clinical screening and echocardiographic surveillance of patients with CHD based on a comprehensive review of the literature, and complemented by our experience at the Center for Carcinoid and Neuroendocrine Tumors at The Mount Sinai Hospital.
Collapse
Affiliation(s)
- Javier G Castillo
- Department of Cardiovascular Surgery, Mount Sinai Health System, The Mount Sinai Hospital, 1190 Fifth Avenue, GP2 West, New York, NY, 10029-6574, USA.
- Center for Carcinoid and Neuroendocrine Tumors, Mount Sinai Health System, The Mount Sinai Hospital, New York, NY, USA.
| | - Tara Naib
- Department of Clinical Cardiology, Mount Sinai Health System, The Mount Sinai Hospital, New York, NY, USA
| | - Jerome S Zacks
- Center for Carcinoid and Neuroendocrine Tumors, Mount Sinai Health System, The Mount Sinai Hospital, New York, NY, USA
- Department of Clinical Cardiology, Mount Sinai Health System, The Mount Sinai Hospital, New York, NY, USA
| | - David H Adams
- Department of Cardiovascular Surgery, Mount Sinai Health System, The Mount Sinai Hospital, 1190 Fifth Avenue, GP2 West, New York, NY, 10029-6574, USA
| |
Collapse
|
50
|
Arora TK, Arora AK, Sachdeva MK, Rajput SK, Sharma AK. Pulmonary hypertension: Molecular aspects of current therapeutic intervention and future direction. J Cell Physiol 2017; 233:3794-3804. [DOI: 10.1002/jcp.26191] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 09/08/2017] [Indexed: 12/28/2022]
Affiliation(s)
| | - Amit K. Arora
- Cardiovascular DivisionSir Ganga ram HospitalNew DelhiIndia
| | | | - Satyendra K. Rajput
- Department of Cardiovascular PharmacologyAmity UniversityNoidaUttar PradeshIndia
| | - Arun K. Sharma
- Department of Cardiovascular PharmacologyAmity UniversityNoidaUttar PradeshIndia
| |
Collapse
|