1
|
Singerer I, Tempel L, Gruen K, Heiß J, Gutte C, Matasci M, Schrepper A, Bauer R, Berndt A, Jung C, Schulze PC, Neri D, Franz M. Extra domain A-containing fibronectin in pulmonary hypertension and treatment effects of a function-blocking antibody. Cardiovasc Res 2024; 120:1485-1497. [PMID: 39023231 DOI: 10.1093/cvr/cvae146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 04/07/2024] [Accepted: 06/01/2024] [Indexed: 07/20/2024] Open
Abstract
AIMS Pulmonary vascular and right ventricular (RV) remodelling processes are important for development and progression of pulmonary hypertension (PH). The current study analysed the functional role of the extra domain A-containing fibronectin (ED-A+ Fn) for the development of PH by comparing ED-A+ Fn knockout (KO) and wild-type (WT) mice as well as the effects of an antibody-based therapeutic approach in a model of monocrotaline (MCT)-induced PH, which will be validated in a model of Sugen 5416/hypoxia-induced PH. METHODS AND RESULTS PH was induced using MCT (PH mice). Sixty-nine mice were divided into the following groups: sham-treated controls (WT: n = 7; KO: n = 7), PH mice without specific treatment (WT: n = 12; KO: n = 10), PH mice treated with a dual endothelin receptor antagonist (macitentan; WT: n = 6; KO: n = 11), WT PH mice treated with the F8 antibody, specifically recognizing ED-A+ Fn, (n = 8), and WT PH mice treated with an antibody of irrelevant antigen specificity (KSF, n = 8). Compared to controls, WT_PH mice showed a significant elevation of the RV systolic pressure (P = 0.04) and RV functional impairment including increased basal RV (P = 0.016) diameter or tricuspid annular plane systolic excursion (P = 0.008). In contrast, KO PH did not show such effects compared to controls (P = n.s.). In WT_PH mice treated with F8, haemodynamic and echocardiographic parameters were significantly improved compared to untreated WT_PH mice or those treated with the KSF antibody (P < 0.05). On the microscopic level, KO_PH mice showed significantly less tissue damage compared to the WT_PH mice (P = 0.008). Furthermore, lung tissue damage could significantly be reduced after F8 treatment (P = 0.04). Additionally, these findings could be verified in the Sugen 5416/hypoxia mouse model, in which F8 significantly improved echocardiographic, haemodynamic, and histologic parameters. CONCLUSION ED-A+ Fn is of crucial importance for PH pathogenesis representing a promising therapeutic target in PH. We here show a novel therapeutic approach using antibody-mediated functional blockade of ED-A+ Fn capable of attenuating and partially reversing PH-associated tissue remodelling.
Collapse
MESH Headings
- Animals
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/immunology
- Fibronectins/metabolism
- Fibronectins/genetics
- Mice, Knockout
- Disease Models, Animal
- Monocrotaline
- Ventricular Function, Right/drug effects
- Ventricular Remodeling/drug effects
- Mice, Inbred C57BL
- Pyrimidines/pharmacology
- Pulmonary Artery/physiopathology
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Male
- Endothelin Receptor Antagonists/pharmacology
- Vascular Remodeling/drug effects
- Antibodies, Blocking/pharmacology
- Antibodies, Monoclonal/pharmacology
- Hypertrophy, Right Ventricular/metabolism
- Hypertrophy, Right Ventricular/physiopathology
- Hypertrophy, Right Ventricular/pathology
- Sulfonamides/pharmacology
Collapse
Affiliation(s)
- Isabell Singerer
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
- Department of Cardiology, Angiology and Intensive Care Medicine, Cardiovascular Center Rotenburg, Klinikum Hersfeld-Rotenburg, Heinz-Meise-Str. 100, 36199 Rotenburg an der Fulda, Germany
| | - Laura Tempel
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Katja Gruen
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Judith Heiß
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Clara Gutte
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
| | | | - Andrea Schrepper
- Department of Cardiothoracic Surgery, University Hospital Jena, Jena, Germany
| | - Reinhard Bauer
- Center for Molecular Biomedicine, Institute of Molecular Cell Biology, University Hospital Jena, Jena, Germany
| | - Alexander Berndt
- Section Pathology, Institute of Legal Medicine, University Hospital Jena, Jena, Germany
| | - Christian Jung
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - P Christian Schulze
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
| | | | - Marcus Franz
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
- Department of Cardiology, Angiology and Intensive Care Medicine, Cardiovascular Center Rotenburg, Klinikum Hersfeld-Rotenburg, Heinz-Meise-Str. 100, 36199 Rotenburg an der Fulda, Germany
| |
Collapse
|
2
|
Steppan J, Wang H, Nandakumar K, Gadkari M, Poe A, Pak L, Brady T, Berkowitz DE, Shimoda LA, Santhanam L. LOXL2 inhibition ameliorates pulmonary artery remodeling in pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2024; 327:L423-L438. [PMID: 39010824 PMCID: PMC11482525 DOI: 10.1152/ajplung.00327.2023] [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: 10/30/2023] [Revised: 06/16/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024] Open
Abstract
Conduit pulmonary arterial stiffening and the resultant increase in pulmonary vascular impedance have emerged as an important underlying driver of pulmonary arterial hypertension (PAH). Given that matrix deposition is central to vascular remodeling, we evaluated the role of the collagen cross-linking enzyme lysyl oxidase like 2 (LOXL2) in this study. Human pulmonary artery smooth muscle cells (PASMCs) subjected to hypoxia showed increased LOXL2 secretion. LOXL2 activity and expression were markedly higher in primary PASMCs isolated from the pulmonary arteries of the rat Sugen 5416 + hypoxia (SuHx) model of severe pulmonary hypertension (PH). Similarly, LOXL2 protein and mRNA levels were increased in the pulmonary arteries (PA) and lungs of rats with PH (SuHx and monocrotaline (MCT) models). Pulmonary arteries (PAs) isolated from the rats with PH exhibited hypercontractility to phenylephrine and attenuated vasorelaxation elicited by acetylcholine, indicating severe endothelial dysfunction. Tensile testing revealed a significant increase in PA stiffness in PH. Treatment with PAT-1251, a novel small-molecule LOXL2 inhibitor, improved active and passive properties of the PA ex vivo. There was an improvement in right heart function as measured by right ventricular pressure volume loops in vivo with PAT-1251. Importantly, PAT-1251 treatment ameliorated PH, resulting in improved pulmonary artery pressures, right ventricular remodeling, and survival. Hypoxia-induced LOXL2 activation is a causal mechanism in pulmonary artery stiffening in PH and pulmonary artery mechanical and functional decline. LOXL2 inhibition with PAT-1251 could be a promising approach to improve pulmonary artery pressures, right ventricular elastance, cardiac relaxation, and survival in PAH.NEW & NOTEWORTHY Pulmonary arterial stiffening contributes to the progression of PAH and the deterioration of right heart function. This study shows that LOXL2 is upregulated in rat models of PH. LOXL2 inhibition halts pulmonary vascular remodeling and improves PA contractility, endothelial function, and PA pressure, resulting in prolonged survival. Thus, LOXL2 is an important mediator of PA remodeling and stiffening in PH and a promising target to improve PA pressures and survival in PH.
Collapse
Affiliation(s)
- Jochen Steppan
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Huilei Wang
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Kavitha Nandakumar
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Mahin Gadkari
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore Maryland, United States
| | - Alan Poe
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Lydia Pak
- Department of Molecular and Cellular Biology, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, Maryland, United States
| | - Travis Brady
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Dan E Berkowitz
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Larissa A Shimoda
- Division of Pulmonary and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Lakshmi Santhanam
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore Maryland, United States
| |
Collapse
|
3
|
Wang X, Cui L, Wang Y, Zeng Z, Wang H, Tian L, Guo J, Chen Y. Mechanistic investigation of wogonin in delaying the progression of endothelial mesenchymal transition by targeting the TGF-β1 pathway in pulmonary hypertension. Eur J Pharmacol 2024; 978:176786. [PMID: 38942264 DOI: 10.1016/j.ejphar.2024.176786] [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: 02/20/2024] [Revised: 06/15/2024] [Accepted: 06/26/2024] [Indexed: 06/30/2024]
Abstract
Pulmonary hypertension (PH) is characterized by pulmonary vascular remodeling, which endothelial-to-mesenchymal transition (EndMT) being its main progressive phase. Wogonin, a flavonoid extracted from the root of Scutellaria baicalensis Georgi, hinders the abnormal proliferation of cells and has been employed in the treatment of several cardiopulmonary diseases. This study was designed to investigate how wogonin affected EndMT during PH. Monocrotaline (MCT) was used to induce PH in rats. Binding capacity of TGF-β1 receptor to wogonin detected by molecular docking and molecular dynamics. EndMT model was established in pulmonary microvascular endothelial cells (PMVECs) by transforming growth factor beta-1 (TGF-β1). The result demonstrated that wogonin (20 mg/kg/day) attenuated right ventricular systolic pressure (RVSP), right ventricular hypertrophy and pulmonary vascular thickness in PH rats. EndMT in the pulmonary vascular was inhibited after wogonin treatment as evidenced by the restored expression of CD31 and decreased expression of α-SMA. Wogonin has strong affinity for both TGFBRI and TGFBRII, and has a better binding stability for TGFBRI. In TGF-β1-treated PMVECs, wogonin (0.3, 1, and 3 μM) exhibited significant inhibitory effects on this transformation process via down-regulating the expression of p-Smad2 and Snail, while up-regulating the expression of p-Smad1/5. Additionally, results of Western blot and fluorescence shown that the expression of α-SMA were decrease with increasing level of CD31 in PMVECs. In conclusion, our research showed that wogonin suppressed EndMT via the TGF-β1/Smad pathway which may lead to its alleviated effect on PH. Wogonin may be a promising drug against PH.
Collapse
Affiliation(s)
- Xinyue Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Lidan Cui
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yichen Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zuomei Zeng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Hongjuan Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Leiyu Tian
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jian Guo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Yucai Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
4
|
Krstic AM, Jones TLM, Power AS, Ward ML. The Monocrotaline Rat Model of Right Heart Disease Induced by Pulmonary Artery Hypertension. Biomedicines 2024; 12:1944. [PMID: 39335458 PMCID: PMC11428269 DOI: 10.3390/biomedicines12091944] [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: 07/24/2024] [Revised: 08/16/2024] [Accepted: 08/17/2024] [Indexed: 09/30/2024] Open
Abstract
Pulmonary artery hypertension (PAH) is characterised by increased pulmonary vascular resistance (PVR) resulting in elevated pressure in the pulmonary artery supplying the pulmonary circulation. Disease of the right ventricle (RV) often manifests as a result of PAH placing excessive pressure on the right side of the heart. Although a relatively rare disease in humans, the impact of sustained PAH is severe, with poor outcomes even in treated individuals. As PAH develops, the blood flow is restricted through the pulmonary arteries and the right ventricle hypertrophies due to the increased strain of pumping blood through the pulmonary circulation. With time, RV hypertrophy progresses to right heart failure, impacting the supply of blood to the left ventricle and systemic circulation. Although right heart failure can currently be treated, it cannot be cured. There is therefore a need for more research into the physiological changes that cause the heart to fail under pressure overload. This review aims to evaluate the monocrotaline (MCT) rat model of PAH as a means of studying the cellular mechanisms associated with the development of RV hypertrophy and right heart failure.
Collapse
Affiliation(s)
- Anna Maria Krstic
- Department of Physiology, University of Auckland, Auckland 1142, New Zealand
| | - Timothy L M Jones
- Department of Physiology, University of Auckland, Auckland 1142, New Zealand
- Division of Cardiology, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA
| | - Amelia S Power
- Department of Physiology, University of Auckland, Auckland 1142, New Zealand
| | - Marie-Louise Ward
- Department of Physiology, University of Auckland, Auckland 1142, New Zealand
| |
Collapse
|
5
|
Hołda MK, Raźny U, Sordyl M, Góralska J, Kapusta M, Słowińska-Solnica K, Wojtysiak D, Lis G, Solnica B, Kopeć G, Hołda J. Autophagy and ubiquitin-dependent proteolysis processes in left ventricular mass loss in pulmonary arterial hypertension. Sci Rep 2024; 14:15133. [PMID: 38956194 PMCID: PMC11220073 DOI: 10.1038/s41598-024-64950-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/14/2024] [Indexed: 07/04/2024] Open
Abstract
The goal of this study was to evaluate the intensity of autophagy and ubiquitin-dependent proteolysis processes occurring in myocardium of left ventricle (LV) in subsequent stages of pulmonary arterial hypertension (PAH) to determine mechanisms responsible for LV mass loss in a monocrotaline-induced PAH rat model. LV myocardium samples collected from 32 Wistar rats were analyzed in an early PAH group (n = 8), controls time-paired (n = 8), an end-stage PAH group (n = 8), and their controls (n = 8). Samples were subjected to histological analyses with immunofluorescence staining, autophagy assessment by western blotting, and evaluation of ubiquitin-dependent proteolysis in the LV by immunoprecipitation of ubiquitinated proteins. Echocardiographic, hemodynamic, and heart morphometric parameters were assessed regularly throughout the experiment. Considerable morphological and hemodynamic remodeling of the LV was observed over the course of PAH. The end-stage PAH was associated with significantly impaired LV systolic function and a decrease in LV mass. The LC3B-II expression in the LV was significantly higher in the end-stage PAH group compared to the early PAH group (p = 0.040). The measured LC3B-II/LC3B-I ratios in the end-stage PAH group were significantly elevated compared to the controls (p = 0.039). Immunofluorescence staining showed a significant increase in the abundance of LC3 puncta in the end-stage PAH group compared to the matched controls. There were no statistically significant differences in the levels of expression of all ubiquitinated proteins when comparing both PAH groups and matched controls. Autophagy may be considered as the mechanism behind the LV mass loss at the end stage of PAH.
Collapse
Affiliation(s)
- Mateusz K Hołda
- HEART - Heart Embryology and Anatomy Research Team, Department of Anatomy, Jagiellonian University Medical College, Kopernika 12, 31-034, Kraków, Poland.
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, UK.
| | - Urszula Raźny
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Kraków, Poland
| | - Maria Sordyl
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Kraków, Poland
| | - Joanna Góralska
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Kraków, Poland
| | - Maria Kapusta
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Kraków, Poland
| | | | - Dorota Wojtysiak
- Department of Animal Genetics, Breeding and Ethology, University of Agriculture in Cracow, Kraków, Poland
| | - Grzegorz Lis
- Department of Histology, Jagiellonian University Medical College, Kraków, Poland
| | - Bogdan Solnica
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Kraków, Poland
| | - Grzegorz Kopeć
- Department of Cardiac and Vascular Diseases, Jagiellonian University Medical College, Kraków, Poland
| | - Jakub Hołda
- HEART - Heart Embryology and Anatomy Research Team, Department of Anatomy, Jagiellonian University Medical College, Kopernika 12, 31-034, Kraków, Poland
| |
Collapse
|
6
|
Leite LB, Soares LL, Portes AMO, Soares TI, da Silva BAF, Dias TR, Costa SFF, Guimarães-Ervilha LO, Assis MQ, Lavorato VN, da Silva AN, Machado-Neves M, Reis ECC, Natali AJ. Combined physical training protects the left ventricle from structural and functional damages in experimental pulmonary arterial hypertension. Clin Hypertens 2024; 30:12. [PMID: 38689333 PMCID: PMC11061945 DOI: 10.1186/s40885-024-00270-z] [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: 12/05/2023] [Accepted: 04/05/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Under the adverse remodeling of the right ventricle and interventricular septum in pulmonary arterial hypertension (PAH) the left ventricle (LV) dynamics is impaired. Despite the benefits of combined aerobic and resistance physical trainings to individuals with PAH, its impact on the LV is not fully understood. OBJECTIVE To test whether moderate-intensity combined physical training performed during the development of PAH induced by MCT in rats is beneficial to the LV's structure and function. METHODS Male Wistar rats were divided into two groups: Sedentary Hypertensive Survival (SHS, n = 7); and Exercise Hypertensive Survival (EHS, n = 7) to test survival. To investigate the effects of combined physical training, another group of rats were divided into three groups: Sedentary Control (SC, n = 7); Sedentary Hypertensive (SH, n = 7); and Exercise Hypertensive (EH, n = 7). PAH was induced through an intraperitoneal injection of MCT (60 mg/kg). Echocardiographic evaluations were conducted on the 22nd day after MCT administration. Animals in the EHS and EH groups participated in a combined physical training program, alternating aerobic (treadmill running: 50 min, 60% maximum running speed) and resistance (ladder climbing: 15 climbs with 1 min interval, 60% maximum carrying load) exercises, one session/day, 5 days/week for approximately 4 weeks. RESULTS The physical training increased survival and tolerance to aerobic (i.e., maximum running speed) and resistance (i.e., maximum carrying load) exertions and prevented reductions in ejection fraction and fractional shortening. In addition, the physical training mitigated oxidative stress (i.e., CAT, SOD and MDA) and inhibited adverse LV remodeling (i.e., Collagen, extracellular matrix, and cell dimensions). Moreover, the physical training preserved the amplitude and velocity of contraction and hindered the reductions in the amplitude and velocity of the intracellular Ca2+ transient in LV single myocytes. CONCLUSION Moderate-intensity combined physical training performed during the development of MCT-induced PAH in rats protects their LV from damages to its structure and function and hence increases their tolerance to physical exertion and prolongs their survival.
Collapse
Affiliation(s)
- Luciano Bernardes Leite
- Department of Physical Education, Laboratory of Exercise Biology, Federal University of Viçosa, Viçosa, Brazil.
| | - Leôncio Lopes Soares
- Department of Physical Education, Laboratory of Exercise Biology, Federal University of Viçosa, Viçosa, Brazil
| | | | - Thayana Inácia Soares
- Department of Physical Education, Laboratory of Exercise Biology, Federal University of Viçosa, Viçosa, Brazil
| | | | - Taís Rodrigues Dias
- Department of Physical Education, Laboratory of Exercise Biology, Federal University of Viçosa, Viçosa, Brazil
| | | | - Luiz Otávio Guimarães-Ervilha
- Department of General Biology, Laboratory of Structural Biology, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | - Mirian Quintão Assis
- Department of General Biology, Laboratory of Structural Biology, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | - Victor Neiva Lavorato
- Department of Physical Education, Governador Ozanam Coelho University Center, Ubá, Minas Gerais, Brazil
| | | | - Mariana Machado-Neves
- Department of General Biology, Laboratory of Structural Biology, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | | | - Antônio José Natali
- Department of Physical Education, Laboratory of Exercise Biology, Federal University of Viçosa, Viçosa, Brazil
| |
Collapse
|
7
|
Murugesan P, Zhang Y, Huang Y, Chenggong Zong N, Youn JY, Chen W, Wang C, Loscalzo J, Cai H. Reversal of Pulmonary Hypertension in a Human-Like Model: Therapeutic Targeting of Endothelial DHFR. Circ Res 2024; 134:351-370. [PMID: 38299369 PMCID: PMC10880947 DOI: 10.1161/circresaha.123.323090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 01/06/2024] [Accepted: 01/15/2024] [Indexed: 02/02/2024]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a progressive disorder characterized by remodeling of the pulmonary vasculature and elevated mean pulmonary arterial pressure, resulting in right heart failure. METHODS Here, we show that direct targeting of the endothelium to uncouple eNOS (endothelial nitric oxide synthase) with DAHP (2,4-diamino 6-hydroxypyrimidine; an inhibitor of GTP cyclohydrolase 1, the rate-limiting synthetic enzyme for the critical eNOS cofactor tetrahydrobiopterin) induces human-like, time-dependent progression of PH phenotypes in mice. RESULTS Critical phenotypic features include progressive elevation in mean pulmonary arterial pressure, right ventricular systolic blood pressure, and right ventricle (RV)/left ventricle plus septum (LV+S) weight ratio; extensive vascular remodeling of pulmonary arterioles with increased medial thickness/perivascular collagen deposition and increased expression of PCNA (proliferative cell nuclear antigen) and alpha-actin; markedly increased total and mitochondrial superoxide production, substantially reduced tetrahydrobiopterin and nitric oxide bioavailabilities; and formation of an array of human-like vascular lesions. Intriguingly, novel in-house generated endothelial-specific dihydrofolate reductase (DHFR) transgenic mice (tg-EC-DHFR) were completely protected from the pathophysiological and molecular features of PH upon DAHP treatment or hypoxia exposure. Furthermore, DHFR overexpression with a pCMV-DHFR plasmid transfection in mice after initiation of DAHP treatment completely reversed PH phenotypes. DHFR knockout mice spontaneously developed PH at baseline and had no additional deterioration in response to hypoxia, indicating an intrinsic role of DHFR deficiency in causing PH. RNA-sequencing experiments indicated great similarity in gene regulation profiles between the DAHP model and human patients with PH. CONCLUSIONS Taken together, these results establish a novel human-like murine model of PH that has long been lacking in the field, which can be broadly used for future mechanistic and translational studies. These data also indicate that targeting endothelial DHFR deficiency represents a novel and robust therapeutic strategy for the treatment of PH.
Collapse
Affiliation(s)
- Priya Murugesan
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (P.M., Y.Z., Y.H., N.C.Z., J.Y.Y., H.C.)
| | - Yixuan Zhang
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (P.M., Y.Z., Y.H., N.C.Z., J.Y.Y., H.C.)
| | - Yuanli Huang
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (P.M., Y.Z., Y.H., N.C.Z., J.Y.Y., H.C.)
| | - Nobel Chenggong Zong
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (P.M., Y.Z., Y.H., N.C.Z., J.Y.Y., H.C.)
| | - Ji Youn Youn
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (P.M., Y.Z., Y.H., N.C.Z., J.Y.Y., H.C.)
| | - Wenhui Chen
- Peking Union Medical College and Chinese Academy of Medical Sciences, Department of Respiratory Medicine, China-Japan Friendship Hospital, Beijing (W.C., C.W.)
| | - Chen Wang
- Peking Union Medical College and Chinese Academy of Medical Sciences, Department of Respiratory Medicine, China-Japan Friendship Hospital, Beijing (W.C., C.W.)
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA (J.L.)
| | - Hua Cai
- Division of Molecular Medicine, Department of Anesthesiology, Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (P.M., Y.Z., Y.H., N.C.Z., J.Y.Y., H.C.)
| |
Collapse
|
8
|
Shima N, Yamamura A, Fujiwara M, Amano T, Matsumoto K, Sekine T, Okano H, Kondo R, Suzuki Y, Yamamura H. Up-regulated expression of two-pore domain K + channels, KCNK1 and KCNK2, is involved in the proliferation and migration of pulmonary arterial smooth muscle cells in pulmonary arterial hypertension. Front Cardiovasc Med 2024; 11:1343804. [PMID: 38410243 PMCID: PMC10894933 DOI: 10.3389/fcvm.2024.1343804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/29/2024] [Indexed: 02/28/2024] Open
Abstract
Background Pulmonary arterial hypertension (PAH) is a severe and rare disease in the cardiopulmonary system. Its pathogenesis involves vascular remodeling of the pulmonary artery, which results in progressive increases in pulmonary arterial pressure. Chronically increased pulmonary arterial pressure causes right ventricular hypertrophy and subsequent right heart failure. Pulmonary vascular remodeling is attributed to the excessive proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs), which are induced by enhanced Ca2+ signaling following the up-/down-regulation of ion channel expression. Objectives In the present study, the functional expression of two-pore domain potassium KCNK channels was investigated in PASMCs from idiopathic PAH (IPAH) patients and experimental pulmonary hypertensive (PH) animals. Results In IPAH-PASMCs, the expression of KCNK1/TWIK1 and KCNK2/TREK1 channels was up-regulated, whereas that of KCNK3/TASK1 and KCNK6/TWIK2 channels was down-regulated. The similar up-regulated expression of KCNK1 and KCNK2 channels was observed in the pulmonary arterial smooth muscles of monocrotaline-induced PH rats, Sugen 5416/hypoxia-induced PH rats, and hypoxia-induced PH mice. The facilitated proliferation of IPAH-PASMCs was suppressed by the KCNK channel blockers, quinine and tetrapentylammonium. The migration of IPAH-PASMCs was also suppressed by these channel blockers. Furthermore, increases in the proliferation and migration were inhibited by the siRNA knockdown of KCNK1 or KCNK2 channels. The siRNA knockdown also caused membrane depolarization and subsequent decrease in cytosolic [Ca2+]. The phosphorylated level of c-Jun N-terminal kinase (JNK) was elevated in IPAH-PASMCs compared to normal-PASMCs. The increased phosphorylation was significantly reduced by the siRNA knockdown of KCNK1 or KCNK2 channels. Conclusion Collectively, these findings indicate that the up-regulated expression of KCNK1 and KCNK2 channels facilitates the proliferation and migration of PASMCs via enhanced Ca2+ signaling and JNK signaling pathway, which is associated with vascular remodeling in PAH.
Collapse
Affiliation(s)
- Natsumi Shima
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Aya Yamamura
- Department of Physiology, Aichi Medical University, Nagakute, Japan
| | - Moe Fujiwara
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Taiki Amano
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Kazuyuki Matsumoto
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Taiga Sekine
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Haruka Okano
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Rubii Kondo
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Yoshiaki Suzuki
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Hisao Yamamura
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| |
Collapse
|
9
|
van Heerden JK, Louw EH, Thienemann F, Engel ME, Allwood BW. The prevalence of pulmonary hypertension in post-tuberculosis and active tuberculosis populations: a systematic review and meta-analysis. Eur Respir Rev 2024; 33:230154. [PMID: 38232991 DOI: 10.1183/16000617.0154-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 11/23/2023] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND The prevalence of tuberculosis (TB)-associated pulmonary hypertension (PH) has not previously been quantified, resulting in an underappreciated burden of disease. We aimed to estimate the prevalence of PH in post-TB and active TB populations. METHODS In this systematic review and meta-analysis, we searched PubMed/Medline, Cochrane Library, EBSCOhost, Scopus, African Journals Online and Google Scholar, with no language restriction, for available literature published after 1950. Eligible studies described adult participants (≥16 years), with documented evidence of active or prior TB, diagnosed with PH. Study quality was assessed using a risk of bias tool specifically developed for prevalence studies. Aggregate prevalence estimates with 95% confidence intervals were synthesised using a random-effects meta-analysis model, incorporating the Freeman-Tukey transformation. Subgroup analysis was conducted to ascertain prevalence estimates in specific patient populations. RESULTS We identified 1452 unique records, of which 34 met our inclusion criteria. 23 studies, with an acceptable risk of bias and where PH was diagnosed at right heart catheterisation or echocardiography, were included in the meta-analysis. In post-TB studies (14/23), the prevalence of PH was 67.0% (95% CI 50.8-81.4) in patients with chronic respiratory failure, 42.4% (95% CI 31.3-54.0) in hospitalised or symptomatic patients and 6.3% (95% CI 2.3-11.8) in nonhealthcare-seeking outpatients (I2=96%). There was a lower estimated prevalence of PH in studies of populations with active TB (9.4%, 95% CI 6.3-13.0), I2=84%). CONCLUSION Our results highlight the significant burden of PH in post-TB and active TB populations. We emphasise the need for increased recognition of TB-associated PH and additional high-quality prevalence data.
Collapse
Affiliation(s)
- Jennifer K van Heerden
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Elizabeth H Louw
- Division of Pulmonology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University and Tygerberg Hospital, Cape Town, South Africa
| | - Friedrich Thienemann
- General Medicine and Global Health, Department of Medicine, and Cape Heart Institute, Faculty of Health Sciences, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa
- Department of Internal Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Mark E Engel
- Department of Medicine, and Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- South African Medical Research Council, Cape Town, South Africa
| | - Brian W Allwood
- Division of Pulmonology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University and Tygerberg Hospital, Cape Town, South Africa
| |
Collapse
|
10
|
Zhao Q, Liu Z, Song P, Yuan Z, Zou MH. Mitochondria-derived Vesicle Packaging as a Novel Therapeutic Mechanism in Pulmonary Hypertension. Am J Respir Cell Mol Biol 2024; 70:39-49. [PMID: 37713305 PMCID: PMC10768832 DOI: 10.1165/rcmb.2023-0010oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 09/14/2023] [Indexed: 09/17/2023] Open
Abstract
Increasing evidence suggests that mitochondrial dysfunction in pulmonary endothelial cells (ECs) plays a causative role in the initiation and progression of pulmonary hypertension (PH); how mitochondria become dysfunctional in PH remains elusive. Mitochondria-derived vesicles (MDVs) are small subcellular vesicles that excise from mitochondria. Whether MDV deregulation causes mitochondrial dysfunction in PH is unknown. The aim of this study was to determine MDV regulation in ECs and to elucidate how MDV deregulation in ECs leads to PH. MDV formation and mitochondrial morphology/dynamics were examined in ECs of EC-specific liver kinase B1 (LKB1) knockout mice (LKB1ec-/-), in monocrotaline-induced PH rats, and in lungs of patients with PH. Pulmonary ECs of patients with PH and hypoxia-treated pulmonary ECs exhibited increased mitochondrial fragmentation and disorganized mitochondrial ultrastructure characterized by electron lucent-swelling matrix compartments and concentric layering of the cristae network, together with defective MDV shedding. MDVs actively regulated mitochondrial membrane dynamics and mitochondrial ultrastructure via removing mitofission-related cargoes. The shedding of MDVs from parental mitochondria required LKB1-mediated mitochondrial recruitment of Rab9 GTPase. LKB1ec-/- mice spontaneously developed PH with decreased mitochondrial pools of Rab9 GTPase, defective MDV shedding, and disequilibrium of the mitochondrial fusion-fission cycle in pulmonary ECs. Aerosol intratracheal delivery of adeno-associated virus LKB1 reversed PH, together with improved MDV shedding and mitochondrial function in rats in vivo. We conclude that LKB1 regulates MDV shedding and mitochondrial dynamics in pulmonary ECs by enhancing mitochondrial recruitment of Rab9 GTPase. Defects of LKB1-mediated MDV shedding from parental mitochondria instigate EC dysfunction and PH.
Collapse
Affiliation(s)
- Qiang Zhao
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia; and
- Department of Cardiology, The First Affiliated Hospital of Xian Jiaotong University, Xi’an, Shaanxi, China
| | - Zhixue Liu
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia; and
| | - Ping Song
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia; and
| | - Zuyi Yuan
- Department of Cardiology, The First Affiliated Hospital of Xian Jiaotong University, Xi’an, Shaanxi, China
| | - Ming-Hui Zou
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia; and
| |
Collapse
|
11
|
Zeng C, Liu J, Zheng X, Hu X, He Y. Prostaglandin and prostaglandin receptors: present and future promising therapeutic targets for pulmonary arterial hypertension. Respir Res 2023; 24:263. [PMID: 37915044 PMCID: PMC10619262 DOI: 10.1186/s12931-023-02559-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 10/09/2023] [Indexed: 11/03/2023] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH), Group 1 pulmonary hypertension (PH), is a type of pulmonary vascular disease characterized by abnormal contraction and remodeling of the pulmonary arterioles, manifested by pulmonary vascular resistance (PVR) and increased pulmonary arterial pressure, eventually leading to right heart failure or even death. The mechanisms involved in this process include inflammation, vascular matrix remodeling, endothelial cell apoptosis and proliferation, vasoconstriction, vascular smooth muscle cell proliferation and hypertrophy. In this study, we review the mechanisms of action of prostaglandins and their receptors in PAH. MAIN BODY PAH-targeted therapies, such as endothelin receptor antagonists, phosphodiesterase type 5 inhibitors, activators of soluble guanylate cyclase, prostacyclin, and prostacyclin analogs, improve PVR, mean pulmonary arterial pressure, and the six-minute walk distance, cardiac output and exercise capacity and are licensed for patients with PAH; however, they have not been shown to reduce mortality. Current treatments for PAH primarily focus on inhibiting excessive pulmonary vasoconstriction, however, vascular remodeling is recalcitrant to currently available therapies. Lung transplantation remains the definitive treatment for patients with PAH. Therefore, it is imperative to identify novel targets for improving pulmonary vascular remodeling in PAH. Studies have confirmed that prostaglandins and their receptors play important roles in the occurrence and development of PAH through vasoconstriction, vascular smooth muscle cell proliferation and migration, inflammation, and extracellular matrix remodeling. CONCLUSION Prostacyclin and related drugs have been used in the clinical treatment of PAH. Other prostaglandins also have the potential to treat PAH. This review provides ideas for the treatment of PAH and the discovery of new drug targets.
Collapse
Affiliation(s)
- Cheng Zeng
- Department of Cardiology, The Second Xiangya Hospital of Central South University, No.139, Middle Ren-min Road, Changsha, 410011, Hunan Province, People's Republic of China
| | - Jing Liu
- Department of Cardiology, The Second Xiangya Hospital of Central South University, No.139, Middle Ren-min Road, Changsha, 410011, Hunan Province, People's Republic of China
| | - Xialei Zheng
- Department of Cardiology, The Second Xiangya Hospital of Central South University, No.139, Middle Ren-min Road, Changsha, 410011, Hunan Province, People's Republic of China
| | - Xinqun Hu
- Department of Cardiology, The Second Xiangya Hospital of Central South University, No.139, Middle Ren-min Road, Changsha, 410011, Hunan Province, People's Republic of China.
| | - Yuhu He
- Department of Cardiology, The Second Xiangya Hospital of Central South University, No.139, Middle Ren-min Road, Changsha, 410011, Hunan Province, People's Republic of China.
| |
Collapse
|
12
|
Steppan J, Wang H, Nandakumar K, Poe A, Pak L, Brady T, Gadkari M, Berkowitz DE, Shimoda LA, Santhanam L. LOXL2 inhibition ameliorates pulmonary artery remodeling in pulmonary hypertension. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563874. [PMID: 37961202 PMCID: PMC10634806 DOI: 10.1101/2023.10.24.563874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Background Conduit pulmonary arterial stiffening and the resultant increase in pulmonary vascular impedance has emerged as an important underlying driver of pulmonary arterial hypertension (PAH). Given that matrix deposition is central to vascular remodeling, we evaluated the role of the collagen crosslinking enzyme lysyl oxidase like 2 (LOXL2) in this study. Methods and Results Human pulmonary artery smooth muscle cells (PASMCs) subjected to hypoxia showed increased LOXL2 secretion. LOXL2 activity and expression were markedly higher in primary PASMCs isolated from pulmonary arteries of the rat Sugen 5416 + hypoxia (SuHx) model of severe PH. Similarly, LOXL2 protein and mRNA levels were increased in pulmonary arteries (PA) and lungs of rats with PH (SuHx and monocrotaline (MCT) models). Pulmonary arteries (PAs) isolated from rats with PH exhibited hypercontractility to phenylephrine and attenuated vasorelaxation elicited by acetylcholine, indicating severe endothelial dysfunction. Tensile testing revealed a a significant increase in PA stiffness in PH. Treatment with PAT-1251, a novel small-molecule LOXL2 inhibitor, improved active and passive properties of the PA ex vivo. There was an improvement in right heart function as measured by right ventricular pressure volume loops in-vivo with PAT-1251. Importantly PAT-1251 treatment ameliorated PH, resulting in improved pulmonary artery pressures, right ventricular remodeling, and survival. Conclusion Hypoxia induced LOXL2 activation is a causal mechanism in pulmonary artery stiffening in PH, as well as pulmonary artery mechanical and functional decline. LOXL2 inhibition with PAT-1251 is a promising approach to improve pulmonary artery pressures, right ventricular elastance, cardiac relaxation, and survival in PAH. New & Noteworthy Pulmonary arterial stiffening contributes to the progression of PAH and the deterioration of right heart function. This study shows that LOXL2 is upregulated in rat models of PH. LOXL2 inhibition halts pulmonary vascular remodeling and improves PA contractility, endothelial function and improves PA pressure, resulting in prolonged survival. Thus, LOXL2 is an important mediator of PA remodeling and stiffening in PH and a promising target to improve PA pressures and survival in PH.
Collapse
|
13
|
Sarkar T, Isbatan A, Moinuddin SM, Chen J, Ahsan F. Catheterization of Pulmonary and Carotid Arteries for Concurrent Measurement of Mean Pulmonary and Systemic Arterial Pressure in Rat Models of Pulmonary Arterial Hypertension. Bio Protoc 2023; 13:e4737. [PMID: 37645695 PMCID: PMC10461069 DOI: 10.21769/bioprotoc.4737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/19/2023] [Accepted: 05/14/2023] [Indexed: 08/31/2023] Open
Abstract
Pulmonary hypertension (PH) is a group of pulmonary vascular disorders in which mean pulmonary arterial pressure (mPAP) becomes abnormally high because of various pathological conditions, including remodeling of the pulmonary arteries, lung and heart disorders, or congenital conditions. Various animal models, including mouse and rat models, have been used to recapitulate elevated mPAP observed in PH patients. However, the measurement and recording of mPAP and mean systemic arterial pressure (mSAP) in small animals require microsurgical procedures and a sophisticated data acquisition system. In this paper, we describe the surgical procedures for right heart catheterizations (RHC) to measure mPAP in rats. We also explain the catheterization of the carotid artery for simultaneous measurement of mPAP and mSAP using the PowerLab Data Acquisition system. We enumerate the surgical steps involved in exposing the jugular vein and the carotid artery for catheterizing these two blood vessels. We list the tools used for microsurgery in rats, describe the methods for preparing catheters, and illustrate the process for inserting the catheters in the pulmonary and carotid arteries. Finally, we delineate the steps involved in the calibration and setup of the PowerLab system for recording both mPAP and mSAP. This is the first protocol wherein we meticulously explain the surgical procedures for RHC in rats and the recording of mPAP and mSAP. We believe this protocol will be essential for PH research. Investigators with little training in animal handling can reproduce this microsurgical procedure for RHC in rats and measure mPAP and mSAP in rat models of PH. Further, this protocol is likely to help master RHC in rats that are performed for other conditions, such as heart failure, congenital heart disease, heart valve disorders, and heart transplantation.
Collapse
Affiliation(s)
- Tanoy Sarkar
- Department of Pharmaceutical and Biomedical Sciences, California Northstate University College of Pharmacy, Elk Grove, USA
| | - Ayman Isbatan
- Cardiovascular Research Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Sakib M. Moinuddin
- Department of Pharmaceutical and Biomedical Sciences, California Northstate University College of Pharmacy, Elk Grove, USA
| | - Jiwang Chen
- Cardiovascular Research Center, University of Illinois at Chicago, Chicago, IL, USA
- Department of Medicine, Section of Pulmonary, Critical Care Medicine, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, USA
| | - Fakhrul Ahsan
- Department of Pharmaceutical and Biomedical Sciences, California Northstate University College of Pharmacy, Elk Grove, USA
| |
Collapse
|
14
|
Jacobs S, Payne C, Shaboodien S, Kgatla T, Pretorius A, Jumaar C, Sanni O, Butrous G, Maarman G. Gut microbiota crosstalk mechanisms are key in pulmonary hypertension: The involvement of melatonin is instrumental too. Pulm Circ 2023; 13:e12277. [PMID: 37583483 PMCID: PMC10423855 DOI: 10.1002/pul2.12277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 08/02/2023] [Accepted: 08/02/2023] [Indexed: 08/17/2023] Open
Abstract
The microbiota refers to a plethora of microorganisms with a gene pool of approximately three million, which inhabits the human gastrointestinal tract or gut. The latter, not only promotes the transport of nutrients, ions, and fluids from the lumen to the internal environment but is linked with the development of diseases including coronary artery disease, heart failure, and lung diseases. The exact mechanism of how the microbiota achieves crosstalk between itself and distant organs/tissues is not clear, but factors released to other organs may play a role, like inflammatory and genetic factors, and now we highlight melatonin as a novel mediator of the gut-lung crosstalk. Melatonin is present in high concentrations in the gut and the lung and has recently been linked to the pathogenesis of pulmonary hypertension (PH). In this comprehensive review of the literature, we suggest that melatonin is an important link between the gut microbiota and the development of PH (where suppressed melatonin-crosstalk between the gut and lungs could promote the development of PH). More studies are needed to investigate the link between the gut microbiota, melatonin and PH. Studies could also investigate whether microbiota genes play a role in the epigenetic aspects of PH. This is relevant because, for example, dysbiosis (caused by epigenetic factors) could reduce melatonin signaling between the gut and lungs, reduce subcellular melatonin concentrations in the gut/lungs, or reduce melatonin serum levels secondary to epigenetic factors. This area of research is largely unexplored and further studies are warranted.
Collapse
Affiliation(s)
- Steve Jacobs
- CARMA: Centre for Cardio‐Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine & Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Carmen Payne
- CARMA: Centre for Cardio‐Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine & Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Sara Shaboodien
- CARMA: Centre for Cardio‐Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine & Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Thato Kgatla
- CARMA: Centre for Cardio‐Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine & Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Amy Pretorius
- CARMA: Centre for Cardio‐Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine & Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Chrisstoffel Jumaar
- CARMA: Centre for Cardio‐Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine & Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Olakunle Sanni
- CARMA: Centre for Cardio‐Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine & Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Ghazwan Butrous
- School of Pharmacy, Imperial College of LondonUniversity of KentCanterburyUK
| | - Gerald Maarman
- CARMA: Centre for Cardio‐Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine & Health SciencesStellenbosch UniversityCape TownSouth Africa
| |
Collapse
|
15
|
Zhang C, Sun Y, Guo Y, Xu J, Zhao H. JMJD1C promotes smooth muscle cell proliferation by activating glycolysis in pulmonary arterial hypertension. Cell Death Discov 2023; 9:98. [PMID: 36934091 PMCID: PMC10024756 DOI: 10.1038/s41420-023-01390-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 02/23/2023] [Accepted: 02/28/2023] [Indexed: 03/20/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a chronic disorder characterized by hyperproliferation of pulmonary arterial smooth muscle cells (PASMCs). JMJD1C, a member of the Jumonji domain containing C (JMJC) histone demethylase family, contributes to cardiovascular dysfunction. However, the role of JMJD1C in PAH remains unknown. Mice were exposed to hypoxia to mimic several features associated with PAH clinically. We found that JMJD1C was highly expressed in the lungs of mice after hypoxia exposure. JMJD1C knockdown ameliorated hypoxia-induced right ventricular remodeling and thickening of the pulmonary arterial wall. PASMC hyperproliferation and resistance to apoptosis in mice exposed to hypoxia were suppressed by JMJD1C inhibition. We demonstrated that JMJD1C silencing reduced glycolytic enzymes (HK2, PGK1 and LDHA) and lactate overaccumulation in the lungs of mice exposed to hypoxia. In vitro, hypoxia-induced hyperproliferation and activated glycolytic processes in mouse PASMCs were impaired by JMJD1C knockdown. In addition, the activation of STAT3 signaling by hypoxia was suppressed by JMJD1C silencing both in vivo and in vitro. The overexpression of STAT3 reversed the inhibitory effect of JMJD1C depletion on proliferation and glycolysis in PASMCs under hypoxia. Thus, JMJD1C induces glycolytic processes by activating STAT3 signaling to promote PASMC proliferation and pulmonary vascular remodeling, suggesting the potential role of JMJD1C in regulating the metabolic program and vascular remodeling in PAH.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yue Sun
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yingying Guo
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jingjing Xu
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Haiyan Zhao
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
16
|
Upton PD, Dunmore BJ, Li W, Morrell NW. An emerging class of new therapeutics targeting TGF, Activin, and BMP ligands in pulmonary arterial hypertension. Dev Dyn 2023; 252:327-342. [PMID: 35434863 PMCID: PMC10952790 DOI: 10.1002/dvdy.478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/21/2022] [Accepted: 04/07/2022] [Indexed: 11/10/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is an often fatal condition, the primary pathology of which involves loss of pulmonary vascular perfusion due to progressive aberrant vessel remodeling. The reduced capacity of the pulmonary circulation places increasing strain on the right ventricle of the heart, leading to death by heart failure. Currently, licensed therapies are primarily vasodilators, which have increased the median post-diagnosis life expectancy from 2.8 to 7 years. Although this represents a substantial improvement, the search continues for transformative therapeutics that reverse established disease. The genetics of human PAH heavily implicates reduced endothelial bone morphogenetic protein (BMP) signaling as a causal role for the disease pathobiology. Recent approaches have focused on directly enhancing BMP signaling or removing the inhibitory influence of pathways that repress BMP signaling. In this critical commentary, we review the evidence underpinning the development of two approaches: BMP-based agonists and inhibition of activin/GDF signaling. We also address the key considerations and questions that remain regarding these approaches.
Collapse
Affiliation(s)
- Paul D. Upton
- Department of MedicineUniversity of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth HospitalsCambridgeUK
| | - Benjamin J. Dunmore
- Department of MedicineUniversity of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth HospitalsCambridgeUK
| | - Wei Li
- Department of MedicineUniversity of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth HospitalsCambridgeUK
| | - Nicholas W. Morrell
- Department of MedicineUniversity of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth HospitalsCambridgeUK
| |
Collapse
|
17
|
Heiss J, Grün K, Tempel L, Matasci M, Schrepper A, Schwarzer M, Bauer R, Förster M, Berndt A, Jung C, Schulze PC, Neri D, Franz M. Targeted Interleukin-9 delivery in pulmonary hypertension: Comparison of immunocytokine formats and effector cell study. Eur J Clin Invest 2023; 53:e13907. [PMID: 36377348 DOI: 10.1111/eci.13907] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/19/2022] [Accepted: 10/27/2022] [Indexed: 11/16/2022]
Abstract
AIMS Pulmonary hypertension (PH) is accompanied by pulmonary vascular remodelling. By targeted delivery of Interleukin-9 (IL9) via the immunocytokine F8IL9, beneficial effects could be demonstrated in a mouse model of PH. This study aimed to compare two immunocytokine formats (single-chain Fv and full IgG) and to identify potential target cells of IL9. METHODS The Monocrotaline mouse model of PH (PH, n = 12) was chosen to evaluate the treatment effects of F8IL9F8 (n = 12) and F8IgGIL9 (n = 6) compared with sham-induced animals (control, n = 10), the dual endothelin receptor antagonist Macitentan (MAC, n = 12) or IL9-based immunocytokines with irrelevant antigen specificity (KSFIL9KSF, n = 12; KSFIgGIL9 n = 6). Besides comparative validation of treatment effects, the study was focused on the detection and quantification of mast cells (MCs) and regulatory T cells (Tregs). RESULTS There was a significantly elevated systolic right ventricular pressure (104 ± 36 vs. 45 ± 17 mmHg) and an impairment of right ventricular echocardiographic parameters (RVbasal: 2.52 ± 0.25 vs. 1.94 ± 0.13 mm) in untreated PH compared with controls (p < 0.05). Only the groups treated with F8IL9, irrespective of the format, showed consistent beneficial effects (p < 0.05). Moreover, F8IL9F8 but not F8IgGIL9 treatment significantly reduced lung tissue damage compared with untreated PH mice (p < 0.05). There was a significant increase in Tregs in F8IL9-treated compared with control animals, the untreated PH and the MAC group (p < 0.05). CONCLUSIONS Beneficial treatment effects of targeted IL9 delivery in a preclinical model of PH could be convincingly validated. IL9-mediated recruitment of Tregs into lung tissue might play a crucial role in the induction of anti-inflammatory and anti-proliferative mechanisms potentially contributing to a novel disease-modifying concept.
Collapse
Affiliation(s)
- Judith Heiss
- Department of Internal Medicine I, University Hospital Jena, Jena, Germany.,Else Kröner Graduate School for Medical Students "JSAM", Jena University Hospital, Jena, Germany
| | - Katja Grün
- Department of Internal Medicine I, University Hospital Jena, Jena, Germany
| | - Laura Tempel
- Department of Internal Medicine I, University Hospital Jena, Jena, Germany
| | | | - Andrea Schrepper
- Department of Cardiothoracic Surgery, University Hospital Jena, Jena, Germany
| | - Michael Schwarzer
- Department of Cardiothoracic Surgery, University Hospital Jena, Jena, Germany
| | - Reinhard Bauer
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine, University Hospital Jena, Jena, Germany
| | - Martin Förster
- Department of Internal Medicine I, University Hospital Jena, Jena, Germany
| | - Alexander Berndt
- Section Pathology, Institute of Legal Medicine, University Hospital Jena, Jena, Germany
| | - Christian Jung
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | | | | | - Marcus Franz
- Department of Internal Medicine I, University Hospital Jena, Jena, Germany
| |
Collapse
|
18
|
Spang MT, Middleton R, Diaz M, Hunter J, Mesfin J, Banka A, Sullivan H, Wang R, Lazerson TS, Bhatia S, Corbitt J, D'Elia G, Sandoval-Gomez G, Kandell R, Vratsanos MA, Gnanasekaran K, Kato T, Igata S, Luo C, Osborn KG, Gianneschi NC, Eniola-Adefeso O, Cabrales P, Kwon EJ, Contijoch F, Reeves RR, DeMaria AN, Christman KL. Intravascularly infused extracellular matrix as a biomaterial for targeting and treating inflamed tissues. Nat Biomed Eng 2023; 7:94-109. [PMID: 36581694 PMCID: PMC10166066 DOI: 10.1038/s41551-022-00964-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 10/18/2022] [Indexed: 12/31/2022]
Abstract
Decellularized extracellular matrix in the form of patches and locally injected hydrogels has long been used as therapies in animal models of disease. Here we report the safety and feasibility of an intravascularly infused extracellular matrix as a biomaterial for the repair of tissue in animal models of acute myocardial infarction, traumatic brain injury and pulmonary arterial hypertension. The biomaterial consists of decellularized, enzymatically digested and fractionated ventricular myocardium, localizes to injured tissues by binding to leaky microvasculature, and is largely degraded in about 3 d. In rats and pigs with induced acute myocardial infarction followed by intracoronary infusion of the biomaterial, we observed substantially reduced left ventricular volumes and improved wall-motion scores, as well as differential expression of genes associated with tissue repair and inflammation. Delivering pro-healing extracellular matrix by intravascular infusion post injury may provide translational advantages for the healing of inflamed tissues 'from the inside out'.
Collapse
Affiliation(s)
- Martin T Spang
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Ryan Middleton
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Miranda Diaz
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Jervaughn Hunter
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Joshua Mesfin
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Alison Banka
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Holly Sullivan
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Raymond Wang
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Tori S Lazerson
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Saumya Bhatia
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - James Corbitt
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Gavin D'Elia
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Gerardo Sandoval-Gomez
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Rebecca Kandell
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Maria A Vratsanos
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
| | - Karthikeyan Gnanasekaran
- Department of Chemistry, International Institute for Nanotechnology, Chemistry of Life Processes Institute, Simpson Querrey Institute, Northwestern University, Evanston, IL, USA
| | - Takayuki Kato
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Sachiyo Igata
- Division of Cardiovascular Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Colin Luo
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Kent G Osborn
- Animal Care Program, University of California San Diego, La Jolla, CA, USA
| | - Nathan C Gianneschi
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
- Department of Chemistry, International Institute for Nanotechnology, Chemistry of Life Processes Institute, Simpson Querrey Institute, Northwestern University, Evanston, IL, USA
- Department of Biomedical Engineering and Department of Pharmacology, Northwestern University, Evanston, IL, USA
| | - Omolola Eniola-Adefeso
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Pedro Cabrales
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Ester J Kwon
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Francisco Contijoch
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Department of Radiology, University of California San Diego, La Jolla, CA, USA
| | - Ryan R Reeves
- Division of Cardiovascular Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Anthony N DeMaria
- Division of Cardiovascular Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Karen L Christman
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA.
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA.
| |
Collapse
|
19
|
Yang JZ, Mokhlesi B, Mesarwi OA. Obstructive sleep apnea and pulmonary hypertension: the pendulum swings again. J Clin Sleep Med 2023; 19:209-211. [PMID: 36533401 PMCID: PMC9892742 DOI: 10.5664/jcsm.10454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Affiliation(s)
- Jenny Z. Yang
- Division of Pulmonary, Critical Care, and Sleep Medicine and Physiology, University of California, San Diego, La Jolla, California
| | - Babak Mokhlesi
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
| | - Omar A. Mesarwi
- Division of Pulmonary, Critical Care, and Sleep Medicine and Physiology, University of California, San Diego, La Jolla, California
| |
Collapse
|
20
|
Wang YX, Reyes-García J, Di Mise A, Zheng YM. Role of ryanodine receptor 2 and FK506-binding protein 12.6 dissociation in pulmonary hypertension. J Gen Physiol 2023; 155:213798. [PMID: 36625865 PMCID: PMC9836826 DOI: 10.1085/jgp.202213100] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 07/29/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Pulmonary hypertension (PH) is a devastating disease characterized by a progressive increase in pulmonary arterial pressure leading to right ventricular failure and death. A major cellular response in this disease is the contraction of smooth muscle cells (SMCs) of the pulmonary vasculature. Cell contraction is determined by the increase in intracellular Ca2+ concentration ([Ca2+]i), which is generated and regulated by various ion channels. Several studies by us and others have shown that ryanodine receptor 2 (RyR2), a Ca2+-releasing channel in the sarcoplasmic reticulum (SR), is an essential ion channel for the control of [Ca2+]i in pulmonary artery SMCs (PASMCs), thereby mediating the sustained vasoconstriction seen in PH. FK506-binding protein 12.6 (FKBP12.6) strongly associates with RyR2 to stabilize its functional activity. FKBP12.6 can be dissociated from RyR2 by a hypoxic stimulus to increase channel function and Ca2+ release, leading to pulmonary vasoconstriction and PH. More specifically, dissociation of the RyR2-FKBP12.6 complex is a consequence of increased mitochondrial ROS generation mediated by the Rieske iron-sulfur protein (RISP) at the mitochondrial complex III after hypoxia. Overall, RyR2/FKBP12.6 dissociation and the corresponding signaling pathway may be an important factor in the development of PH. Novel drugs and biologics targeting RyR2, FKBP12.6, and related molecules may become unique effective therapeutics for PH.
Collapse
Affiliation(s)
- Yong-Xiao Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA,Correspondence to Yong-Xiao Wang:
| | - Jorge Reyes-García
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA,Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México,Ciudad de México, México
| | - Annarita Di Mise
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA,Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Yun-Min Zheng
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA,Yun-Min Zheng:
| |
Collapse
|
21
|
Alleviating experimental pulmonary hypertension via co-delivering FoxO1 stimulus and apoptosis activator to hyperproliferating pulmonary arteries. Acta Pharm Sin B 2022. [DOI: 10.1016/j.apsb.2022.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
22
|
Cai G, Zhang Y, Wang X, Li S, Cheng Y, Huang X. Comparison of hypoxia-induced pulmonary hypertension rat models caused by different hypoxia protocols. Exp Lung Res 2022:1-11. [DOI: 10.1080/01902148.2022.2148016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Gexiang Cai
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang, China
| | - Yaxin Zhang
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang, China
| | - Xinghong Wang
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang, China
| | - Shini Li
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang, China
| | - Yushan Cheng
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang, China
| | - Xiaoying Huang
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Zhejiang, China
| |
Collapse
|
23
|
Wu J, Liu T, Shi S, Fan Z, Hiram R, Xiong F, Cui B, Su X, Chang R, Zhang W, Yan M, Tang Y, Huang H, Wu G, Huang C. Dapagliflozin reduces the vulnerability of rats with pulmonary arterial hypertension-induced right heart failure to ventricular arrhythmia by restoring calcium handling. Cardiovasc Diabetol 2022; 21:197. [PMID: 36171554 PMCID: PMC9516842 DOI: 10.1186/s12933-022-01614-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 09/01/2022] [Indexed: 12/20/2022] Open
Abstract
Background Malignant ventricular arrhythmia (VA) is a major contributor to sudden cardiac death (SCD) in patients with pulmonary arterial hypertension (PAH)-induced right heart failure (RHF). Recently, dapagliflozin (DAPA), a sodium/glucose cotransporter-2 inhibitor (SGLT2i), has been found to exhibit cardioprotective effects in patients with left ventricular systolic dysfunction. In this study, we examined the effects of DAPA on VA vulnerability in a rat model of PAH-induced RHF. Methods Rats randomly received monocrotaline (MCT, 60 mg/kg) or vehicle via a single intraperitoneal injection. A day later, MCT-injected rats were randomly treated with placebo, low-dose DAPA (1 mg/kg/day), or high-dose (3 mg/kg/day) DAPA orally for 35 days. Echocardiographic analysis, haemodynamic experiments, and histological assessments were subsequently performed to confirm the presence of PAH-induced RHF. Right ventricle (RV) expression of calcium (Ca2+) handling proteins were detected via Western blotting. RV expression of connexin 43 (Cx43) was determined via immunohistochemical staining. An optical mapping study was performed to assess the electrophysiological characteristics in isolated hearts. Cellular Ca2+ imaging from RV cardiomyocytes (RVCMs) was recorded using Fura-2 AM or Fluo-4 AM. Results High-dose DAPA treatment attenuated RV structural remodelling, improved RV function, alleviated Cx43 remodelling, increased the conduction velocity, restored the expression of key Ca2+ handling proteins, increased the threshold for Ca2+ and action potential duration (APD) alternans, decreased susceptibility to spatially discordant APD alternans and spontaneous Ca2+ events, promoted cellular Ca2+ handling, and reduced VA vulnerability in PAH-induced RHF rats. Low-dose DAPA treatment also showed antiarrhythmic effects in hearts with PAH-induced RHF, although with a lower level of efficacy. Conclusion DAPA administration reduced VA vulnerability in rats with PAH-induced RHF by improving RVCM Ca2+ handling. Supplementary Information The online version contains supplementary material available at 10.1186/s12933-022-01614-5.
Collapse
Affiliation(s)
- Jinchun Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Department of Cardiology, Qinghai Provincial People's Hospital, No.2 Gong He Road, Xining, 810007, People's Republic of China
| | - Tao Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China. .,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China. .,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.
| | - Shaobo Shi
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China
| | - Zhixing Fan
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China
| | - Roddy Hiram
- Department of Medicine, Faculty of Medicine, Montreal Heart Institute (MHI), Université de Montréal, Montreal, QC, Canada
| | - Feng Xiong
- Department of Medicine, Faculty of Medicine, Montreal Heart Institute (MHI), Université de Montréal, Montreal, QC, Canada
| | - Bo Cui
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China
| | - Xiaoling Su
- Department of Cardiology, Qinghai Provincial People's Hospital, No.2 Gong He Road, Xining, 810007, People's Republic of China
| | - Rong Chang
- Department of Cardiology, Shenzhen Longhua District Central Hospital, The Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, No. 187 Guanlan Road, Longhua District, Shenzhen, 518109, China
| | - Wei Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China
| | - Min Yan
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China
| | - Yanhong Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China
| | - He Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China
| | - Gang Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China. .,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China. .,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuhan, 430060, People's Republic of China. .,Cardiovascular Research Institute, Wuhan University, 238 Jiefang Road, Wuhan, 430060, People's Republic of China. .,Hubei Key Laboratory of Cardiology, 238 Jiefang Road, Wuhan, 430060, People's Republic of China.
| |
Collapse
|
24
|
Sarkar T, Nguyen T, Moinuddin SM, Stenmark KR, Nozik ES, Saha D, Ahsan F. A Protocol for Fabrication and on-Chip Cell Culture to Recreate PAH-Afflicted Pulmonary Artery on a Microfluidic Device. MICROMACHINES 2022; 13:mi13091483. [PMID: 36144106 PMCID: PMC9504537 DOI: 10.3390/mi13091483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 05/08/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a rare pulmonary vascular disease that affects people of all ethnic origins and age groups including newborns. In PAH, pulmonary arteries and arterioles undergo a series of pathological changes including remodeling of the entire pulmonary vasculatures and extracellular matrices, mis-localized growth of pulmonary arterial cells, and development of glomeruloid-like lesions called plexiform lesions. Traditionally, various animal and cellular models have been used to understand PAH pathophysiology, investigate sex-disparity in PAH and monitor therapeutic efficacy of PAH medications. However, traditional models can only partially capture various pathological features of PAH, and they are not adaptable to combinatorial study design for deciphering intricately intertwined complex cellular processes implicated in PAH pathogenesis. While many microfluidic chip-based models are currently available for major diseases, no such disease-on-a-device model is available for PAH, an under investigated disease. In the absence of any chip-based models of PAH, we recently proposed a five-channel polydimethylsiloxane (PDMS)-based microfluidic device that can emulate major pathological features of PAH. However, our proposed model can make a bigger impact on the PAH field only when the larger scientific community engaged in PAH research can fabricate the device and develop the model in their laboratory settings. With this goal in mind, in this study, we have described the detailed methodologies for fabrication and development of the PAH chip model including a thorough explanation of scientific principles for various steps for chip fabrication, a detailed list of reagents, tools and equipment along with their source and catalogue numbers, description of laboratory setup, and cautionary notes. Finally, we explained the methodologies for on-chip cell seeding and application of this model for studying PAH pathophysiology. We believe investigators with little or no training in microfluidic chip fabrication can fabricate this eminently novel PAH-on-a-chip model. As such, this study will have a far-reaching impact on understanding PAH pathophysiology, unravelling the biological mystery associated with sexual dimorphism in PAH, and developing PAH therapy based on patient sex and age.
Collapse
Affiliation(s)
- Tanoy Sarkar
- Department of Pharmaceutical and Biomedical Sciences, California Northstate University College of Pharmacy, Elk Grove, CA 95757, USA
| | - Trieu Nguyen
- Department of Pharmaceutical and Biomedical Sciences, California Northstate University College of Pharmacy, Elk Grove, CA 95757, USA
| | - Sakib M. Moinuddin
- Department of Pharmaceutical and Biomedical Sciences, California Northstate University College of Pharmacy, Elk Grove, CA 95757, USA
| | - Kurt R. Stenmark
- Department of Pediatrics and Medicine, Cardiovascular Pulmonary Research Laboratories, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Eva S. Nozik
- Department of Pediatrics and Medicine, Cardiovascular Pulmonary Research Laboratories, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Dipongkor Saha
- Department of Pharmaceutical and Biomedical Sciences, California Northstate University College of Pharmacy, Elk Grove, CA 95757, USA
- Correspondence: (D.S.); (F.A.)
| | - Fakhrul Ahsan
- Department of Pharmaceutical and Biomedical Sciences, California Northstate University College of Pharmacy, Elk Grove, CA 95757, USA
- MedLuidics, Elk Grove, CA 95757, USA
- Correspondence: (D.S.); (F.A.)
| |
Collapse
|
25
|
Abstract
Pulmonary hypertension (PH) because of chronic lung disease is categorized as Group 3 PH in the most recent classification system. Prevalence of these diseases is increasing over time, creating a growing need for effective therapeutic options. Recent approval of the first pulmonary arterial hypertension therapy for the treatment of Group 3 PH related to interstitial lung disease represents an encouraging advancement. This review focuses on molecular mechanisms contributing to pulmonary vasculopathy in chronic hypoxia, the pathology and epidemiology of Group 3 PH, the right ventricular dysfunction observed in this population and clinical trial data that inform the use of pulmonary vasodilators in Group 3 PH.
Collapse
Affiliation(s)
- Navneet Singh
- Division of Pulmonary, Critical Care and Sleep Medicine (N.S., C.E.V.), Brown University, Providence, RI
| | - Peter Dorfmüller
- Department of Pathology, Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig University, Germany (P.D.).,German Center for Lung Research (DZL), Giessen, Germany (P.D.)
| | - Oksana A Shlobin
- Advanced Lung Disease and Transplant Program, Inova Fairfax Hospital, Falls Church, VA (O.A.S.)
| | - Corey E Ventetuolo
- Division of Pulmonary, Critical Care and Sleep Medicine (N.S., C.E.V.), Brown University, Providence, RI.,Department of Health Services, Policy and Practice (C.E.V.), Brown University, Providence, RI
| |
Collapse
|
26
|
Wu XH, Ma JL, Ding D, Ma YJ, Wei YP, Jing ZC. Experimental animal models of pulmonary hypertension: Development and challenges. Animal Model Exp Med 2022; 5:207-216. [PMID: 35333455 PMCID: PMC9240731 DOI: 10.1002/ame2.12220] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/11/2022] [Accepted: 02/20/2022] [Indexed: 12/16/2022] Open
Abstract
Pulmonary hypertension (PH) is clinically divided into 5 major types, characterized by elevation in pulmonary arterial pressure (PAP) and pulmonary vascular resistance (PVR), finally leading to right heart failure and death. The pathogenesis of this arteriopathy remains unclear, leaving it impossible to target pulmonary vascular remodeling and reverse the deterioration of right ventricular (RV) function. Different animal models have been designed to reflect the complex mechanistic origins and pathology of PH, roughly divided into 4 categories according to the modeling methods: non‐invasive models in vivo, invasive models in vivo, gene editing models, and multi‐means joint modeling. Though each model shares some molecular and pathological changes with different classes of human PH, in most cases the molecular etiology of human PH is poorly known. The appropriate use of classic and novel PH animal models is essential for the hunt of molecular targets to reverse severe phenotypes.
Collapse
Affiliation(s)
- Xiao-Han Wu
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie-Ling Ma
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dong Ding
- Medical Science Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue-Jiao Ma
- Medical Science Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yun-Peng Wei
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhi-Cheng Jing
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
27
|
Maarman GJ. Reviewing the suitability of mitochondrial transplantation as therapeutic approach for pulmonary hypertension in the era of personalised medicine. Am J Physiol Lung Cell Mol Physiol 2022; 322:L641-L646. [PMID: 35318860 DOI: 10.1152/ajplung.00484.2021] [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] Open
Abstract
Pulmonary hypertension (PH) is a fatal disease, defined as a mean pulmonary artery pressure ≥ 25 mm Hg. It is caused, in part, by mitochondrial dysfunction. Among the various biological therapies proposed to rescue mitochondrial dysfunction, evidence going back as far as 2009, suggests that mitochondrial transplantation is an alternative. Although scant, recent PH findings and other literature supports a role for mitochondrial transplantation as a therapeutic approach in the context of PH. In experimental models of PH, it confers beneficial effects that include reduced pulmonary vasoconstriction, reduced pulmonary vascular remodelling, and improved right ventricular function. It also reduces the proliferation of pulmonary artery smooth muscle cells. However, first, we must understand that more research is needed before mitochondrial transplantation can be considered an effective therapy in the clinical setting, as many of the mechanisms or potential long-term risks are still unknown. Second, the current challenges of mitochondrial transplantation are surmountable and should not deter researchers from further investigating its effectiveness and trying to overcome these challenges in creative ways.
Collapse
Affiliation(s)
- Gerald J Maarman
- CARMA: Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Department of Biomedical Sciences, Stellenbosch University, Tygerberg, South Africa
| |
Collapse
|
28
|
Habeeb E, Aldosari S, Saghir SA, Cheema M, Momenah T, Husain K, Omidi Y, Rizvi SA, Akram M, Ansari RA. Role of Environmental Toxicants in the Development of Hypertensive and Cardiovascular Diseases. Toxicol Rep 2022; 9:521-533. [PMID: 35371924 PMCID: PMC8971584 DOI: 10.1016/j.toxrep.2022.03.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/17/2022] [Indexed: 12/12/2022] Open
Abstract
The incidence of hypertension with diabetes mellitus (DM) as a co-morbid condition is on the rise worldwide. In 2000, an estimated 972 million adults had hypertension, which is predicted to grow to 1.56 billion by 2025. Hypertension often leads to diabetes mellitus that strongly puts the patients at an increased risk of cardiovascular, kidney, and/or atherosclerotic diseases. Hypertension has been identified as a major risk factor for the development of diabetes; patients with hypertension are at two-to-three-fold higher risk of developing diabetes than patients with normal blood pressure (BP). Causes for the increase in hypertension and diabetes are not well understood, environmental factors (e.g., exposure to environmental toxicants like heavy metals, organic solvents, pesticides, alcohol, and urban lifestyle) have been postulated as one of the reasons contributing to hypertension and cardiovascular diseases (CVD). The mechanism of action(s) of these toxicants in developing hypertension and CVDs is not well defined. Research studies have linked hypertension with the chronic consumption of alcohol and exposure to metals like lead, mercury, and arsenic have also been linked to hypertension and CVD. Workers chronically exposed to styrene have a higher incidence of CVD. Recent studies have demonstrated that exposure to particulate matter (PM) in diesel exhaust and urban air contributes to increased CVD and mortality. In this review, we have imparted the role of environmental toxicants such as heavy metals, organic pollutants, PM, alcohol, and some drugs in hypertension and CVD along with possible mechanisms and limitations in extrapolating animal data to humans. Rising incidence of hypertension may be linked to chronic exposure with environmental toxicants. Urban lifestyle and alcohol intake may be responsible for increased incidence of hypertension among urbanites. Exposure with organic solvent, heavy metals and pesticides could also be contributing to the rise in blood pressure.
Collapse
Affiliation(s)
- Ehsan Habeeb
- Department of Pharmaceutical Sciences, College of Pharmacy, Health Professions Division, Nova Southeastern University, 3200S University Drive, Fort Lauderdale, FL 33200, USA
| | - Saad Aldosari
- Department of Pharmaceutical Sciences, College of Pharmacy, Health Professions Division, Nova Southeastern University, 3200S University Drive, Fort Lauderdale, FL 33200, USA
| | - Shakil A. Saghir
- The Scotts Company LLC, Marysville, OH 43041, USA
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | - Mariam Cheema
- Department of Pharmaceutical Sciences, College of Pharmacy, Health Professions Division, Nova Southeastern University, 3200S University Drive, Fort Lauderdale, FL 33200, USA
| | - Tahani Momenah
- Department of Pharmaceutical Sciences, College of Pharmacy, Health Professions Division, Nova Southeastern University, 3200S University Drive, Fort Lauderdale, FL 33200, USA
| | - Kazim Husain
- Department of Gastrointestinal Oncology (FOB-2), Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Health Professions Division, Nova Southeastern University, 3200S University Drive, Fort Lauderdale, FL 33200, USA
| | - Syed A.A. Rizvi
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton University, VA 23668, USA
| | - Muhammad Akram
- Department of Eastern Medicine and Surgery, Government College University Faisalabad, Faisalabad, Pakistan
| | - Rais A. Ansari
- Department of Pharmaceutical Sciences, College of Pharmacy, Health Professions Division, Nova Southeastern University, 3200S University Drive, Fort Lauderdale, FL 33200, USA
- Corresponding author.
| |
Collapse
|
29
|
Jasińska-Stroschein M. A review of genetically-driven rodent models of pulmonary hypertension. Vascul Pharmacol 2022; 144:106970. [PMID: 35150934 DOI: 10.1016/j.vph.2022.106970] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 01/19/2022] [Accepted: 02/05/2022] [Indexed: 11/19/2022]
Abstract
An increasing number of models used to examine the role of particular signaling pathways in vasculature and the development of pulmonary hypertension (PH) are based on animals with different genetic modifications. The present study explores the severity of PH-related lesions that can be provided by a genetic particular model in accordance to the most common non-genetic PH inducers such as chronic exposure to hypoxia or single injection of monocrotaline. A review of 516 interventions on a variety of animal models was performed. It examined the advantages of various genetically-driven procedures intended to develop spontaneous PH, and the effects of combining such procedures with common PH models or other stimuli ('second-hit') with the aim of exacerbating pulmonary artery remodeling, right ventricle hypertrophy and hemodynamics or animal mortality. A wide range of genetically-modified rodents are used for pre-clinical studies on PH, with different response to the genetic modification as compared to the most common non-genetic stimuli. Nevertheless, they could highlight the mechanisms and pathways that contribute to the expression of pathophysiological features of the disease, and they could be helpful in the identification of additional targets for new drugs.
Collapse
|
30
|
Nakano M, Koga M, Hashimoto T, Matsushita N, Masukawa D, Mizuno Y, Uchimura H, Niikura R, Miyazaki T, Nakamura F, Zou S, Shimizu T, Saito M, Tamura K, Goto T, Goshima Y. Right ventricular overloading is attenuated in monocrotaline-induced pulmonary hypertension model rats with a disrupted Gpr143 gene, the gene that encodes the 3,4-l-dihydroxyphenyalanine (l-DOPA) receptor. J Pharmacol Sci 2022; 148:214-220. [PMID: 35063136 DOI: 10.1016/j.jphs.2021.11.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/18/2021] [Accepted: 11/29/2021] [Indexed: 12/17/2022] Open
Abstract
Pulmonary hypertension (PH) is a severe and progressive disease that causes elevated right ventricular systolic pressure, right ventricular hypertrophy and ultimately right heart failure. However, the underlying pathophysiologic mechanisms are poorly understood. We previously showed that 3,4-l-dihydroxylphenyalanine (DOPA) sensitizes vasomotor response to sympathetic tone via coupling between the adrenergic receptor alpha1 (ADRA1) and a G protein-coupled receptor 143 (GPR143), a DOPA receptor. We investigated whether DOPA similarly enhances ADRA1-mediated contraction in pulmonary arteries isolated from rats, and whether GPR143 is involved in the PH pathogenesis. Pretreating the isolated pulmonary arteries with DOPA 1 μM enhanced vasoconstriction in response to phenylephrine, an ADRA1 agonist, but not to U-46619, a thromboxane A2 agonist or endothelin-1. We generated Gpr143 gene-deficient (Gpr143-/y) rats, and confirmed that DOPA did not augment phenylephrine-induced contractile response in Gpr143-/y rat pulmonary arteries. We utilized a rat model of monocrotaline (MCT)-induced PH. In the MCT model, the right ventricular systolic pressure was attenuated in the Gpr143-/y rats than in WT rats. Phenylephrine-induced cell migration and proliferation were also suppressed in Gpr143-/y pulmonary artery smooth muscle cells than in WT cells. Our result suggests that GPR143 is involved in the PH pathogenesis in the rat models of PH.
Collapse
MESH Headings
- 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology
- Animals
- Disease Models, Animal
- Heart Failure/etiology
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/genetics
- Hypertrophy, Right Ventricular/etiology
- In Vitro Techniques
- Male
- Monocrotaline/adverse effects
- Pulmonary Artery/physiology
- Rats, Sprague-Dawley
- Receptors, Adrenergic, alpha-1/physiology
- Receptors, G-Protein-Coupled/physiology
- Receptors, Neurotransmitter/genetics
- Systole
- Vasoconstriction/drug effects
- Vasoconstriction/genetics
- Ventricular Dysfunction, Right/etiology
- Ventricular Function, Right/genetics
- Rats
Collapse
Affiliation(s)
- Masayuki Nakano
- Department of Molecular Pharmacology and Neurobiology, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan; Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan
| | - Motokazu Koga
- Department of Molecular Pharmacology and Neurobiology, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan; Department of Anesthesiology, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan; Department of Anesthesiology, Kanagawa Cancer Center, Yokohama, 241-8515, Japan
| | - Tatsuo Hashimoto
- Department of Molecular Pharmacology and Neurobiology, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan; Department of Internal Medicine, Graduate School of Dentistry, Kanagawa Dental University, Yokosuka 238-8680, Japan, Yokosuka, 238-8570, Japan
| | - Natsuki Matsushita
- Division of Laboratory Animal Research, Aichi Medical University, Nagakute, 480-1195, Aichi, Japan
| | - Daiki Masukawa
- Department of Molecular Pharmacology and Neurobiology, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan
| | - Yusuke Mizuno
- Department of Anesthesiology, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan
| | - Hiraku Uchimura
- Department of Molecular Pharmacology and Neurobiology, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan; Department of Psychiatry, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan
| | - Ryo Niikura
- Department of Anesthesiology, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan
| | - Tomoyuki Miyazaki
- Department of Anesthesiology, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan; Department of Physiology, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan
| | - Fumio Nakamura
- Department of Molecular Pharmacology and Neurobiology, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan; Department of Biochemistry, School of Medicine, Tokyo Women's Medical University, Tokyo, 162- 8666, Japan
| | - Suo Zou
- Department of Pharmacology, Kochi Medical School, Kochi University, Nankoku, Kochi, 783-8505, Japan
| | - Takahiro Shimizu
- Department of Pharmacology, Kochi Medical School, Kochi University, Nankoku, Kochi, 783-8505, Japan
| | - Motoaki Saito
- Department of Pharmacology, Kochi Medical School, Kochi University, Nankoku, Kochi, 783-8505, Japan
| | - Kouichi Tamura
- Department of Medical Science and Cardiorenal Medicine, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan
| | - Takahisa Goto
- Department of Anesthesiology, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan
| | - Yoshio Goshima
- Department of Molecular Pharmacology and Neurobiology, Graduate School of Medicine, Yokohama City University, Yokohama, 236-0004, Japan.
| |
Collapse
|
31
|
Calvier L, Herz J, Hansmann G. Interplay of Low-Density Lipoprotein Receptors, LRPs, and Lipoproteins in Pulmonary Hypertension. JACC Basic Transl Sci 2022; 7:164-180. [PMID: 35257044 PMCID: PMC8897182 DOI: 10.1016/j.jacbts.2021.09.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 12/21/2022]
Abstract
The low-density lipoprotein receptor (LDLR) gene family includes LDLR, very LDLR, and LDL receptor-related proteins (LRPs) such as LRP1, LRP1b (aka LRP-DIT), LRP2 (aka megalin), LRP4, and LRP5/6, and LRP8 (aka ApoER2). LDLR family members constitute a class of closely related multifunctional, transmembrane receptors, with diverse functions, from embryonic development to cancer, lipid metabolism, and cardiovascular homeostasis. While LDLR family members have been studied extensively in the systemic circulation in the context of atherosclerosis, their roles in pulmonary arterial hypertension (PAH) are understudied and largely unknown. Endothelial dysfunction, tissue infiltration of monocytes, and proliferation of pulmonary artery smooth muscle cells are hallmarks of PAH, leading to vascular remodeling, obliteration, increased pulmonary vascular resistance, heart failure, and death. LDLR family members are entangled with the aforementioned detrimental processes by controlling many pathways that are dysregulated in PAH; these include lipid metabolism and oxidation, but also platelet-derived growth factor, transforming growth factor β1, Wnt, apolipoprotein E, bone morpohogenetic proteins, and peroxisome proliferator-activated receptor gamma. In this paper, we discuss the current knowledge on LDLR family members in PAH. We also review mechanisms and drugs discovered in biological contexts and diseases other than PAH that are likely very relevant in the hypertensive pulmonary vasculature and the future care of patients with PAH or other chronic, progressive, debilitating cardiovascular diseases.
Collapse
Key Words
- ApoE, apolipoprotein E
- Apoer2
- BMP
- BMPR, bone morphogenetic protein receptor
- BMPR2
- COPD, chronic obstructive pulmonary disease
- CTGF, connective tissue growth factor
- HDL, high-density lipoprotein
- KO, knockout
- LDL receptor related protein
- LDL, low-density lipoprotein
- LDLR
- LDLR, low-density lipoprotein receptor
- LRP
- LRP, low-density lipoprotein receptor–related protein
- LRP1
- LRP1B
- LRP2
- LRP4
- LRP5
- LRP6
- LRP8
- MEgf7
- Mesd, mesoderm development
- PAH
- PAH, pulmonary arterial hypertension
- PASMC, pulmonary artery smooth muscle cell
- PDGF
- PDGFR-β, platelet-derived growth factor receptor-β
- PH, pulmonary hypertension
- PPARγ
- PPARγ, peroxisome proliferator-activated receptor gamma
- PVD
- RV, right ventricle/ventricular
- RVHF
- RVSP, right ventricular systolic pressure
- TGF-β1
- TGF-β1, transforming growth factor β1
- TGFBR, transforming growth factor β1 receptor
- TNF, tumor necrosis factor receptor
- VLDLR
- VLDLR, very low density lipoprotein receptor
- VSMC, vascular smooth muscle cell
- Wnt
- apolipoprotein E receptor 2
- endothelial cell
- gp330
- low-density lipoprotein receptor
- mRNA, messenger RNA
- megalin
- monocyte
- multiple epidermal growth factor-like domains 7
- pulmonary arterial hypertension
- pulmonary vascular disease
- right ventricle heart failure
- smooth muscle cell
- very low density lipoprotein receptor
- β-catenin
Collapse
Affiliation(s)
- Laurent Calvier
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Georg Hansmann
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany
- Pulmonary Vascular Research Center, Hannover Medical School, Hannover, Germany
| |
Collapse
|
32
|
Myocardial Afterload Is a Key Biomechanical Regulator of Atrioventricular Myocyte Differentiation in Zebrafish. J Cardiovasc Dev Dis 2022; 9:jcdd9010022. [PMID: 35050232 PMCID: PMC8779957 DOI: 10.3390/jcdd9010022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 02/05/2023] Open
Abstract
Heart valve development is governed by both genetic and biomechanical inputs. Prior work has demonstrated that oscillating shear stress associated with blood flow is required for normal atrioventricular (AV) valve development. Cardiac afterload is defined as the pressure the ventricle must overcome in order to pump blood throughout the circulatory system. In human patients, conditions of high afterload can cause valve pathology. Whether high afterload adversely affects embryonic valve development remains poorly understood. Here we describe a zebrafish model exhibiting increased myocardial afterload, caused by vasopressin, a vasoconstrictive drug. We show that the application of vasopressin reliably produces an increase in afterload without directly acting on cardiac tissue in zebrafish embryos. We have found that increased afterload alters the rate of growth of the cardiac chambers and causes remodeling of cardiomyocytes. Consistent with pathology seen in patients with clinically high afterload, we see defects in both the form and the function of the valve leaflets. Our results suggest that valve defects are due to changes in atrioventricular myocyte signaling, rather than pressure directly acting on the endothelial valve leaflet cells. Cardiac afterload should therefore be considered a biomechanical factor that particularly impacts embryonic valve development.
Collapse
|
33
|
Boulate D, Loisel F, Coblence M, Provost B, Todesco A, Decante B, Beurnier A, Herve P, Perros F, Humbert M, Fadel E, Mercier O, Chemla D. Pulsatile pulmonary artery pressure in a large animal model of chronic thromboembolic pulmonary hypertension: Similarities and differences with human data. Pulm Circ 2022; 12:e12017. [PMID: 35506099 PMCID: PMC9052967 DOI: 10.1002/pul2.12017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 10/27/2021] [Accepted: 11/20/2021] [Indexed: 11/20/2022] Open
Abstract
A striking feature of the human pulmonary circulation is that mean (mPAP) and systolic (sPAP) pulmonary artery pressures (PAPs) are strongly related and, thus, are essentially redundant. According to the empirical formula documented under normotensive and hypertensive conditions (mPAP = 0.61 sPAP + 2 mmHg), sPAP matches ~160%mPAP on average. This attests to the high pulsatility of PAP, as also witnessed by the near equality of PA pulse pressure and mPAP. Our prospective study tested if pressure redundancy and high pulsatility also apply in a piglet model of chronic thromboembolic pulmonary hypertension (CTEPH). At baseline (Week‐0, W0), Sham (n = 8) and CTEPH (n = 27) had similar mPAP and stroke volume. At W6, mPAP increased in CTEPH only, with a two‐ to three‐fold increase in PA stiffness and total pulmonary resistance. Seven CTEPH piglets were also studied at W16 at baseline, after volume loading, and after acute pulmonary embolism associated with dobutamine infusion. There was a strong linear relationship between sPAP and mPAP (1) at W0 and W6 (n = 70 data points, r² = 0.95); (2) in the subgroup studied at W16 (n = 21, r² = 0.97); and (3) when all data were pooled (n = 91, r² = 0.97, sPAP range 9–112 mmHg). The PA pulsatility was lower than that expected based on observations in humans: sPAP matched ~120%mPAP only and PA pulse pressure was markedly lower than mPAP. In conclusion, the redundancy between mPAP and sPAP seems a characteristic of the pulmonary circulation independent of the species. However, it is suggested that the sPAP thresholds used to define PH in animals are species‐ and/or model‐dependent and thus must be validated.
Collapse
Affiliation(s)
- David Boulate
- Hôpital Marie Lannelongue INSERM UMR_S 999 Le Plessis Robinson France
| | - Fanny Loisel
- Hôpital Marie Lannelongue INSERM UMR_S 999 Le Plessis Robinson France
| | - Mathieu Coblence
- Hôpital Marie Lannelongue INSERM UMR_S 999 Le Plessis Robinson France
| | - Bastien Provost
- Hôpital Marie Lannelongue INSERM UMR_S 999 Le Plessis Robinson France
| | - Alban Todesco
- Hôpital Marie Lannelongue INSERM UMR_S 999 Le Plessis Robinson France
| | - Benoit Decante
- Hôpital Marie Lannelongue INSERM UMR_S 999 Le Plessis Robinson France
| | - Antoine Beurnier
- Hôpital Marie Lannelongue INSERM UMR_S 999 Le Plessis Robinson France
| | - Philippe Herve
- Hôpital Marie Lannelongue INSERM UMR_S 999 Le Plessis Robinson France
| | - Frédéric Perros
- Hôpital Marie Lannelongue INSERM UMR_S 999 Le Plessis Robinson France
| | - Marc Humbert
- Hôpital Marie Lannelongue INSERM UMR_S 999 Le Plessis Robinson France
- Service de Pneumologie, Hôpital Bicêtre DMU‐THORINO, AP‐HP Le Kremlin‐Bicêtre France
| | - Elie Fadel
- Hôpital Marie Lannelongue INSERM UMR_S 999 Le Plessis Robinson France
- Pôle Thoracique, Vasculaire et Transplantations Hôpital Marie Lannelongue Le Plessis Robinson France
| | - Olaf Mercier
- Hôpital Marie Lannelongue INSERM UMR_S 999 Le Plessis Robinson France
- Pôle Thoracique, Vasculaire et Transplantations Hôpital Marie Lannelongue Le Plessis Robinson France
| | - Denis Chemla
- Hôpital Marie Lannelongue INSERM UMR_S 999 Le Plessis Robinson France
- Service d'Explorations Fonctionnelles Multidisciplinaires Bi‐site, Hôpitaux Antoine Béclère–Kremlin Bicêtre, Faculté de médecine‐Université Paris Saclay DMU‐CORREVE, AP‐HP Le Kremlin‐Bicêtre France
| |
Collapse
|
34
|
Earl CC, Damen FW, Yin M, Aasa KL, Burris SK, Goergen CJ. Strain Estimation of the Murine Right Ventricle Using High-Frequency Speckle-Tracking Ultrasound. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:3291-3300. [PMID: 34373135 PMCID: PMC8488001 DOI: 10.1016/j.ultrasmedbio.2021.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 06/28/2021] [Accepted: 07/03/2021] [Indexed: 05/25/2023]
Abstract
Right ventricular (RV) strain measurements from ultrasound via speckle-tracking techniques are being used more frequently as a non-invasive diagnostic tool for a variety of cardiopulmonary pathologies. However, despite the clinical utility of ultrasound RV strain measurements, quantification of RV strain in rodents remains difficult owing to unique image artifacts and non-standardized methodologies. We demonstrate here a simple approach for measuring RV strain in both mice and rats using high-frequency ultrasound and automated speckle tracking. Our results show estimated peak RV free-wall longitudinal strain values (mean ± standard error of the mean) in mice (n = 15) and rats (n = 5) of, respectively, -10.38% ± 0.4% and -4.85% ± 0.42%. We further estimated the 2-D Green-Lagrange strain within the RV free wall, with longitudinal components estimated at -5.7% ± 0.48% in mice and -2.1% ± 0.28% in rats. These methods and data may provide a foundation for future work aimed at evaluating murine RV strain levels in different disease models.
Collapse
Affiliation(s)
- Conner C Earl
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Frederick W Damen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| | - Melissa Yin
- Fujifilm VisualSonics Inc., Toronto, Ontario, Canada
| | | | | | - Craig J Goergen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA.
| |
Collapse
|
35
|
Wu C, Ha Y, Zou Y, Liao X, Zhang S, Zhang X, Li R, Xing J, Jie W, Guo J, Li J, Shen Z. Pathologic role of peptidyl-prolyl isomerase Pin1 in pulmonary artery remodeling. Am J Transl Res 2021; 13:11162-11177. [PMID: 34786049 PMCID: PMC8581927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 08/06/2021] [Indexed: 06/13/2023]
Abstract
Peptidyl-prolyl isomerase Pin1 is crucial for cell proliferation, but its role in pulmonary artery remodeling (PAR) is unclear. In the present study, we aimed to evaluate the expression and contribution of Pin1 in PAR. Treatment with Pin1 inhibitor Juglone or Pin1-specific siRNAs ameliorated the expression of Pin1 and proliferating cell nuclear antigen (PCNA) in human pulmonary artery smooth muscle cells (PASMCs) in vitro, and Juglone treatment arrested the cell cycle at the G1 phase. Treatment with transforming growth factor β1 (TGF-β1) also enhanced Pin1 expression and PASMC proliferation. Immunohistochemical staining revealed that Pin1 and PCNA expression levels were increased and positively correlated with each other in PAR samples from humans and monocrotaline-treated Sprague-Dawley rats; these proteins were mainly localized in arteries undergoing remodeling, as well as inflammatory cells, and hyperplastic bronchial epithelial cells. Intraperitoneal injection of Juglone also led to morphologic and hemodynamic changes in PAR rats. Additionally, PAR rats displayed higher serum and lung TGF-β1 levels compared with controls, while administration of Juglone to PAR rats suppressed serum and lung TGF-β1 levels. The findings in this study suggest that TGF-β1 and Pin1 constitute a positive feedback loop, which plays an important role in the pathophysiology of PAR.
Collapse
Affiliation(s)
- Caixia Wu
- Department of Pathology & Pathophysiology, School of Basic Medicine Sciences, Guangdong Medical UniversityZhanjiang 524023, Guangdong, China
| | - Yanping Ha
- Department of Pathology & Pathophysiology, School of Basic Medicine Sciences, Guangdong Medical UniversityZhanjiang 524023, Guangdong, China
| | - Yuan Zou
- Department of Pathology & Pathophysiology, School of Basic Medicine Sciences, Guangdong Medical UniversityZhanjiang 524023, Guangdong, China
| | - Xiaomin Liao
- Department of Pathology & Pathophysiology, School of Basic Medicine Sciences, Guangdong Medical UniversityZhanjiang 524023, Guangdong, China
| | - Shuya Zhang
- Key Laboratory for Tropical Cardiovascular Diseases Research of Hainan Province, The First Affiliated Hospital of Hainan Medical UniversityHaikou 571199, Hainan, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical UniveraityHaikou 571199, Hainan, China
| | - Xiaodian Zhang
- Key Laboratory for Tropical Cardiovascular Diseases Research of Hainan Province, The First Affiliated Hospital of Hainan Medical UniversityHaikou 571199, Hainan, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical UniveraityHaikou 571199, Hainan, China
| | - Rujia Li
- Department of Pathology & Pathophysiology, School of Basic Medicine Sciences, Guangdong Medical UniversityZhanjiang 524023, Guangdong, China
| | - Jingci Xing
- Department of Pathology & Pathophysiology, School of Basic Medicine Sciences, Guangdong Medical UniversityZhanjiang 524023, Guangdong, China
| | - Wei Jie
- Department of Pathology & Pathophysiology, School of Basic Medicine Sciences, Guangdong Medical UniversityZhanjiang 524023, Guangdong, China
- Key Laboratory for Tropical Cardiovascular Diseases Research of Hainan Province, The First Affiliated Hospital of Hainan Medical UniversityHaikou 571199, Hainan, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical UniveraityHaikou 571199, Hainan, China
| | - Junli Guo
- Key Laboratory for Tropical Cardiovascular Diseases Research of Hainan Province, The First Affiliated Hospital of Hainan Medical UniversityHaikou 571199, Hainan, China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical UniveraityHaikou 571199, Hainan, China
| | - Jingquan Li
- Department of Oncology, The First Affiliated Hospital of Hainan Medical UniversityHaikou 570102, Hainan, China
| | - Zhihua Shen
- Department of Pathology & Pathophysiology, School of Basic Medicine Sciences, Guangdong Medical UniversityZhanjiang 524023, Guangdong, China
| |
Collapse
|
36
|
Mamazhakypov A, Hein L, Lother A. Mineralocorticoid receptors in pulmonary hypertension and right heart failure: From molecular biology to therapeutic targeting. Pharmacol Ther 2021; 231:107987. [PMID: 34480966 DOI: 10.1016/j.pharmthera.2021.107987] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/21/2021] [Accepted: 08/31/2021] [Indexed: 12/20/2022]
Abstract
Pulmonary hypertension (PH) is a devastating condition characterized by pulmonary vascular remodelling, leading to progressive increase in pulmonary artery pressure and subsequent right ventricular failure. Aldosterone and the mineralocorticoid receptor (MR), a nuclear transcription factor, are key drivers of cardiovascular disease and MR antagonists are well-established in heart failure. Now, a growing body of evidence points at a detrimental role of MR in PH. Pharmacological MR blockade attenuated PH and prevented RV failure in experimental models. Mouse models with cell selective MR deletion suggest that this effect is mediated by MR in endothelial cells. While the evidence from experimental studies appears convincing, the available clinical data on MR antagonist use in patients with PH is more controversial. Integrated analysis of clinical data together with MR-dependent molecular alterations may provide insights why some patients respond to MRA treatment while others do not. Potential ways to identify MRA 'responders' include the analysis of underlying PH causes, stage of disease, or sex, as well as new biomarkers.
Collapse
Affiliation(s)
- Argen Mamazhakypov
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Germany
| | - Lutz Hein
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Germany
| | - Achim Lother
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Germany; Heart Center Freiburg University, Department of Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Germany.
| |
Collapse
|
37
|
Kawade A, Yamamura A, Fujiwara M, Kobayashi S, Mori S, Horii C, Hiraku A, Suzumura S, Tsukamoto K, Ohara N, Kondo R, Suzuki Y, Yamamura H. Comparative analysis of age in monocrotaline-induced pulmonary hypertensive rats. J Pharmacol Sci 2021; 147:81-85. [PMID: 34294376 DOI: 10.1016/j.jphs.2021.05.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/13/2021] [Accepted: 05/24/2021] [Indexed: 12/23/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare, progressive, and fatal cardiovascular/lung disease. The incidence rate is affected by age. Monocrotaline (MCT, 60 mg/kg)-treated rats are widely used as an experimental PAH model. Here, we found that young rats died at a mean of 23.4 days after MCT injection, whereas adult rats survived for over 42 days. However, young (7-week-old) and adult (20-week-old) MCT-treated rats developed PAH, and had upregulated Ca2+-sensing receptor and transient receptor potential canonical subfamily 6 channel expression in pulmonary arteries. The present study provides novel information for elucidating the mechanism underlying the age difference in PAH patients.
Collapse
Affiliation(s)
- Akiko Kawade
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabedori Mizuhoku, Nagoya, 467-8603, Japan
| | - Aya Yamamura
- Department of Physiology, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan; Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori Moriyamaku, Nagoya, 463-8521, Japan
| | - Moe Fujiwara
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabedori Mizuhoku, Nagoya, 467-8603, Japan
| | - Saki Kobayashi
- Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori Moriyamaku, Nagoya, 463-8521, Japan
| | - Shiho Mori
- Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori Moriyamaku, Nagoya, 463-8521, Japan
| | - Chihiro Horii
- Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori Moriyamaku, Nagoya, 463-8521, Japan
| | - Akari Hiraku
- Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori Moriyamaku, Nagoya, 463-8521, Japan
| | - Sayo Suzumura
- Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori Moriyamaku, Nagoya, 463-8521, Japan
| | - Kikuo Tsukamoto
- Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori Moriyamaku, Nagoya, 463-8521, Japan
| | - Naoki Ohara
- Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori Moriyamaku, Nagoya, 463-8521, Japan
| | - Rubii Kondo
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabedori Mizuhoku, Nagoya, 467-8603, Japan
| | - Yoshiaki Suzuki
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabedori Mizuhoku, Nagoya, 467-8603, Japan
| | - Hisao Yamamura
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabedori Mizuhoku, Nagoya, 467-8603, Japan.
| |
Collapse
|
38
|
Diaba-Nuhoho P, Cour M, Hadebe N, Marais D, Lecour S, Blackhurst D. Chronic and moderate consumption of reduced-alcohol wine confers cardiac benefits in a rat model of pulmonary arterial hypertension. BMC Res Notes 2021; 14:324. [PMID: 34425891 PMCID: PMC8381534 DOI: 10.1186/s13104-021-05738-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/12/2021] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES In pulmonary arterial hypertension (PAH), right ventricular (RV) dysfunction develops via mechanisms involving oxidative stress. Moderate and chronic red wine (RW) consumption reduces oxidative stress and confers cardioprotection but its effect on PAH is unknown. We evaluated whether moderate and chronic consumption of reduced-alcohol RW (RARW) confers cardioprotection in a monocrotaline (MCT)-induced PAH rat model. RESULTS Rats were randomly grouped: control; MCT; RARW; MCT + RARW. Wine was diluted to mimic moderate intake for humans, and consumed from 7 days before, until 28 days after MCT-injection. Echocardiography measured pulmonary artery acceleration time (PAAT) and RV thickness. Conjugated dienes (CD), and thiobarbituric acid reactive substances (TBARS) concentrations were assessed. MCT induced RV thickness and decreased PAAT compared to controls [1.22 ± 0.09 mm vs 0.46 ± 0.02 mm and 14 ± 1 vs 23 ± 2 m/s, respectively (p < 0.001)]. Chronic RARW consumption limited MCT-induced RV hypertrophy and increased PAAT. CD and TBARS increased in MCT-treated animals compared to controls (672 ± 43 nmol/L vs 453 ± 35 nmol/L; p < 0.01 and 13 ± 2 µmol/L vs 4 ± 0.3 µmol/L; p < 0.01). RARW reduced MCT-induced CD (472 ± 27 nmol/L vs 672 ± 43 nmol/L; p < 0.01). CONCLUSION Chronic and moderate intake of RARW ameliorates MCT-induced PAH in rats, which may be partly attributable to reduction of lipid peroxidation.
Collapse
Affiliation(s)
- Patrick Diaba-Nuhoho
- Division of Chemical Pathology, Department of Pathology, University of Cape Town, Cape Town, South Africa.
- Division of Vascular Endothelium and Microcirculation, Department of Medicine III, University of Technology Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
| | - Martin Cour
- Cardioprotection Group, Hatter Cardiovascular Institute and Lionel Opie Preclinical Imaging Core Facility, University of Cape Town, Cape Town, South Africa
| | - Nkanyiso Hadebe
- Cardioprotection Group, Hatter Cardiovascular Institute and Lionel Opie Preclinical Imaging Core Facility, University of Cape Town, Cape Town, South Africa
- Department of Anaesthesia, Groote Schuur Hospital and University of Cape Town, Cape Town, South Africa
| | - David Marais
- Division of Chemical Pathology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Sandrine Lecour
- Cardioprotection Group, Hatter Cardiovascular Institute and Lionel Opie Preclinical Imaging Core Facility, University of Cape Town, Cape Town, South Africa
| | - Dee Blackhurst
- Division of Chemical Pathology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
39
|
Mukherjee D, Konduri GG. Pediatric Pulmonary Hypertension: Definitions, Mechanisms, Diagnosis, and Treatment. Compr Physiol 2021; 11:2135-2190. [PMID: 34190343 PMCID: PMC8289457 DOI: 10.1002/cphy.c200023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pediatric pulmonary hypertension (PPH) is a multifactorial disease with diverse etiologies and presenting features. Pulmonary hypertension (PH), defined as elevated pulmonary artery pressure, is the presenting feature for several pulmonary vascular diseases. It is often a hidden component of other lung diseases, such as cystic fibrosis and bronchopulmonary dysplasia. Alterations in lung development and genetic conditions are an important contributor to pediatric pulmonary hypertensive disease, which is a distinct entity from adult PH. Many of the causes of pediatric PH have prenatal onset with altered lung development due to maternal and fetal conditions. Since lung growth is altered in several conditions that lead to PPH, therapy for PPH includes both pulmonary vasodilators and strategies to restore lung growth. These strategies include optimal alveolar recruitment, maintaining physiologic blood gas tension, nutritional support, and addressing contributing factors, such as airway disease and gastroesophageal reflux. The outcome for infants and children with PH is highly variable and largely dependent on the underlying cause. The best outcomes are for neonates with persistent pulmonary hypertension (PPHN) and reversible lung diseases, while some genetic conditions such as alveolar capillary dysplasia are lethal. © 2021 American Physiological Society. Compr Physiol 11:2135-2190, 2021.
Collapse
Affiliation(s)
- Devashis Mukherjee
- Division of Neonatology, Department of Pediatrics, Medical College of Wisconsin, Children’s Research Institute, Children’s Wisconsin, Milwaukee, Wisconsin, 53226 USA
| | - Girija G. Konduri
- Division of Neonatology, Department of Pediatrics, Medical College of Wisconsin, Children’s Research Institute, Children’s Wisconsin, Milwaukee, Wisconsin, 53226 USA
| |
Collapse
|
40
|
Ouyang S, Chen W, Gaofeng Z, Changcheng L, Guoping T, Minyan Z, Yang L, Min Y, Luo J. Cyanidin‑3‑O‑β‑glucoside protects against pulmonary artery hypertension induced by monocrotaline via the TGF‑β1/p38 MAPK/CREB signaling pathway. Mol Med Rep 2021; 23:338. [PMID: 33760143 PMCID: PMC7974420 DOI: 10.3892/mmr.2021.11977] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 04/09/2020] [Indexed: 12/22/2022] Open
Abstract
Pulmonary artery hypertension (PAH) is a disease with high morbidity and mortality. Cyanidin‑3‑O‑β‑glucoside (Cy‑3‑g), a classical anthocyanin, has a variety of biological effects. The present study evaluated whether Cy‑3‑g attenuated PAH, and explored the potential mechanism of action. Rats were injected with monocrotaline (MCT; 60 mg per kg of body weight) and then treated with Cy‑3‑g (200 or 400 mg per kg of body weight) for 4 weeks. Protein expression was determined in vitro in transforming growth factor‑β1 (TGF‑β1)‑mediated human pulmonary arterial smooth muscle cells (SMCs). The results indicated that Cy‑3‑g significantly inhibited the mean pulmonary artery pressure, right ventricular systolic pressure and right ventricular hypertrophy index, as well as vascular remodeling induced by MCT in PAH rats. Further experiments showed that Cy‑3‑g suppressed the expression of pro‑-inflammatory factors and enhanced the levels of anti‑inflammatory factors. Cy‑3‑g blocked oxidative stress and improved vascular endothelial injury. Cy‑3‑g also reduced the proliferation of SMCs. Furthermore, the MCT‑ and TGF‑β1‑induced increase in TGF‑β1, phosphorylated (p)‑p38 mitogen‑activated protein kinase (MAPK) and p‑cAMP‑response element binding protein (CREB) expression was blocked by Cy‑3‑g treatment in vivo and in vitro. These results indicated that Cy‑3‑g could prevent vascular remodeling in PAH via inhibition of the TGF‑β1/p38 MAPK/CREB axis.
Collapse
Affiliation(s)
- Shao Ouyang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Wei Chen
- Department of Respiratory Medicine, The Second Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Zeng Gaofeng
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Lei Changcheng
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Tian Guoping
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Zhu Minyan
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Liu Yang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yang Min
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Jiahao Luo
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
41
|
Fan Y, Hao Y, Gao D, Li G, Zhang Z. Phenotype and function of macrophage polarization in monocrotaline-induced pulmonary arterial hypertension rat model. Physiol Res 2021; 70:213-226. [PMID: 33676385 PMCID: PMC8820576 DOI: 10.33549/physiolres.934456] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 01/26/2021] [Indexed: 12/31/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) isa fatal disease characterized by vascular remodeling and chronic inflammation. Macrophages are the key orchestrators of inflammatory and repair responses, and have been demonstrated to be vital in the pathogenesis of PAH. However, specific phenotype of macrophage polarization (M1 & M2 macrophage) in the development of PAH and the underlying mechanisms how they work are still largely unclear. A rat model of monocrotaline (MCT) induced PAH was used. Hemodynamic analysis and histopathological experiments were conducted at day 3, 7, 14, 21 and 28, respectively. In PAH rat lung tissue, confocal microscopic images showed that CD68+NOS2+ M1-like macrophages were remarkably infiltrated on early stage, but dramatically decreased in mid-late stage. Meanwhile, CD68+CD206+ M2-like macrophages in lung tissue accumulated gradually since day 7 to day 28, and the relative ratio of M2/M1 macrophage increased over time. Results detected by western blot and immunohistochemistry were consistent. Further vitro functional studies revealed the possible mechanism involved in this pathophysiological process. By using Transwell co-culture system, it was found that M1 macrophages inducedendothelial cellapoptosis, while M2 macrophages significantly promoted proliferation of both endothelial cell and smooth muscle cell.These data preliminarily demonstrated a temporal dynamic change of macrophage M1/M2 polarization status in the development of experimental PAH. M1 macrophages participated in the initial stage of inflammation by accelerating apoptosis of endothelial cell, while M2 macrophages predominated in the reparative stage of inflammation and the followed stage of aberrant tissue remodeling.
Collapse
Affiliation(s)
- Yong Fan
- Department of Rheumatology and Clinical Immunology, Peking University First Hospital, Beijing, China.
| | | | | | | | | |
Collapse
|
42
|
Goncharova NS, Andreeva EM, Vakhrushev AD, Leandro HIC, Murashova LA, Voronin SE, Korobchenko LE, Mitrofanova LB, Skorik YA, Galagudza MM, Moiseeva OM, Mikhaylov EN. Modeling of Acute Pulmonary Arterial Hypertension in Pigs Using a Stable Thromboxane A 2 Analogue (U46619): Dose Adjustment and Assessment of Hemodynamic Reactions. Bull Exp Biol Med 2021; 170:729-733. [PMID: 33893968 DOI: 10.1007/s10517-021-05142-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Indexed: 11/29/2022]
Abstract
U46619, a synthetic analogue of thromboxane A2 was used for modeling acute stable and reversible pulmonary arterial hypertension. Administration of U46619 in high doses led to vascular collapse and inhibition of cardiac function. The doses of U46619 were empirically selected that allow attaining the target level of pulmonary hypertension without systemic hemodynamic disturbances. The possibility of attaining the target level of pulmonary hypertension and reversibility of changes after termination of U46619 infusion make this model attractive for evaluation of the efficiency of different therapeutic methods of treatment of pulmonary hypertension in large animals.
Collapse
Affiliation(s)
- N S Goncharova
- Non-Coronary Heart Diseases Research Department, St. Petersburg, Russia.
| | - E M Andreeva
- Non-Coronary Heart Diseases Research Department, St. Petersburg, Russia
| | - A D Vakhrushev
- Neuromodulation Research Laboratory, St. Petersburg, Russia
| | | | - L A Murashova
- Center for Preclinical and Translational Research, St. Petersburg, Russia
| | - S E Voronin
- Center for Preclinical and Translational Research, St. Petersburg, Russia
| | | | - L B Mitrofanova
- Research Laboratory of Pathomorphology, V. A. Almazov National Medical Research Centre, Ministry of Health the Russian Federation, St. Petersburg, Russia
| | - Y A Skorik
- Center for Preclinical and Translational Research, St. Petersburg, Russia
| | - M M Galagudza
- Center for Preclinical and Translational Research, St. Petersburg, Russia
| | - O M Moiseeva
- Non-Coronary Heart Diseases Research Department, St. Petersburg, Russia
| | - E N Mikhaylov
- Neuromodulation Research Laboratory, St. Petersburg, Russia.,Arrhythmia Research Department, St. Petersburg, Russia
| |
Collapse
|
43
|
Proteomic analysis of hypoxia and non-hypoxia secretome mesenchymal stem-like cells from human breastmilk. Saudi J Biol Sci 2021; 28:4399-4407. [PMID: 34354424 PMCID: PMC8324926 DOI: 10.1016/j.sjbs.2021.04.034] [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: 09/07/2020] [Revised: 04/08/2021] [Accepted: 04/11/2021] [Indexed: 11/18/2022] Open
Abstract
Introduction Breastmilk contains proteins and cells which have stem cell properties. The human breastmilk stem cell mimick mesenchymal stem cells and expresses pluripotency genes. The protein level of breastmilk is high in colostrum and gradually subsides in the first year of lactation. The mesenchymal stem cells from breastmilk can be an alternative source of stem cells that can potentially affect cardiovascular therapy. This study aimed to identify the proteomic analysis of secretome mesenchymal stem-like cells under hypoxia compared to non-hypoxia from human breastmilk stem cells. Material and methods The human breastmilk was collected from six healthy breastfeeding women and transported to the laboratory under aseptic conditions. The breastmilk cells were isolated then cultured. After 72 h, the human breastmilk stem cells reached confluence then cleaned up and isolated in serum-free media (spheroid) to allow serial passaging every 48 h. The acquisition stem cell was made with flow cytometry. The cells were divided into hBSC secretomes under hypoxia (A) and non-hypoxia (B) and analyzed for LC-MS to identify the peptide structure. Results The human breastmilk cells contained several mesenchymal stem-like cells in density 2.4 × 106 cell/mL for hypoxia and 2 × 106 cell/mL for non-hypoxia conditions. The human breastmilk stem cell surface markers derived from the third cell passage process were 93.77% for CD44, 98.69% for CD73, 88.45% for CD90, and 96.30% for CD105. The protein level of secretome mesenchymal stem -like cells under hypoxia was measured at 5.56 μg/mL and 4.28 μg/mL for non-hypoxia. The liquid chromatography-mass spectrometry analysis identified 130 and 59 peptides from hypoxia and non-hypoxia of the human breastmilk stem cell secretome sequentially. Some important proteomics structures were found in the hypoxic human breastmilk stem cell secretome, such as transforming growth factor-β, VE-cadherin, and caspase. Conclusion The human breastmilk cells contain mesenchymal stem-like cells and a high concentration of CD44, CD73, CD90, and CD105 as surface markers at third passage culture. The hypoxic hBSC secretome produces a higher protein level compare to non-hypoxia. The transforming growth factor -β was found in the hypoxic hBSC secretome as a modulator of VEGF-mediated angiogenesis.
Collapse
Key Words
- AFP, Alpha-Fetoprotein
- ATP, Adenosine Triphosphate
- BD, Becton Dickinson
- BMPR-II, Bone morphogenetic protein type II
- BSA, Bovine Serum Albumin
- EHD3, EH Domain-containing Protein 3
- FACS, Fluorescence-Activated Cell Sorting
- FBS, Fetal Bovine Serum
- HIF-1α, Hypoxia Inducible Factor-1α
- Hypoxia
- IGF1, Insulin-like Growth Factor 1
- LALBA, α-Lactalbumin
- LC-MS
- LC-MS, Liquid Chromatography-Mass Spectrometry
- LF, Lactoferrin
- MAPK, Mitogen-Activated Protein Kinase
- MPS, Multi Proliferative Supplement
- MPZL1, Myelin Protein Zero-like Protein 1
- MSC, Mesenchymal Stem Cell
- Mesenchymal stem-like cell
- PBS, Phosphate-buffered Saline
- SDS, Sodium Dodecyl Sulfate
- SMA, Smooth Muscle Actin
- SMAD, Signals Mothers Against the Decapentaplegic
- Secretome
- TGF-β, Transforming Growth Factor-Beta
- VEGF, Vascular Endothelial Growth Factor
- cDNA, complementary Deoxyribonucleic Acid
- hBSC
- hBSC, Human Breastmilk Stem Cell
- mRNA, messenger Ribonucleic Acid
Collapse
|
44
|
Gouyou B, Grün K, Kerschenmeyer A, Villa A, Matasci M, Schrepper A, Pfeil A, Bäz L, Jung C, Schulze PC, Neri D, Franz M. Therapeutic Evaluation of Antibody-Based Targeted Delivery of Interleukin 9 in Experimental Pulmonary Hypertension. Int J Mol Sci 2021; 22:ijms22073460. [PMID: 33801620 PMCID: PMC8037792 DOI: 10.3390/ijms22073460] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 12/22/2022] Open
Abstract
Background and Aims: Pulmonary hypertension (PH) is a heterogeneous disorder associated with poor prognosis. For the majority of patients, only limited therapeutic options are available. Thus, there is great interest to develop novel treatment strategies focusing on pulmonary vascular and right ventricular remodeling. Interleukin 9 (IL9) is a pleiotropic cytokine with pro- and anti-inflammatory functions. The aim of this study was to evaluate the therapeutic activity of F8IL9F8 consisting of IL9 fused to the F8 antibody, specific to the alternatively-spliced EDA domain of fibronectin, which is abundantly expressed in pulmonary vasculature and right ventricular myocardium in PH. Methods: The efficacy of F8IL9F8 in attenuating PH progression in the monocrotaline mouse model was evaluated in comparison to an endothelin receptor antagonist (ERA) or an IL9 based immunocytokine with irrelevant antibody specificity (KSFIL9KSF). Treatment effects were assessed by right heart catheterization, echocardiography as well as histological and immunohistochemical tissue analyses. Results: Compared to controls, systolic right ventricular pressure (RVPsys) was significantly elevated and a variety of right ventricular echocardiographic parameters were significantly impaired in all MCT-induced PH groups except for the F8IL9F8 group. Both, F8IL9F8 and ERA treatments lead to a significant reduction in RVPsys and an improvement of echocardiographic parameters when compared to the MCT group not observable for the KSFIL9KSF group. Only F8IL9F8 significantly reduced lung tissue damage and displayed a significant decrease of leukocyte and macrophage accumulation in the lungs and right ventricles. Conclusions: Our study provides first pre-clinical evidence for the use of F8IL9F8 as a new therapeutic agent for PH in terms of a disease-modifying concept addressing cardiovascular remodeling.
Collapse
Affiliation(s)
- Baptiste Gouyou
- Philochem AG, CH-8112 Otelfingen, Switzerland; (B.G.); (A.K.); (A.V.); (M.M.); (D.N.)
| | - Katja Grün
- Department of Internal Medicine I, Univerisity Hospital Jena, 07747 Jena, Germany; (K.G.); (L.B.); (P.C.S.)
| | - Anne Kerschenmeyer
- Philochem AG, CH-8112 Otelfingen, Switzerland; (B.G.); (A.K.); (A.V.); (M.M.); (D.N.)
| | - Alessandra Villa
- Philochem AG, CH-8112 Otelfingen, Switzerland; (B.G.); (A.K.); (A.V.); (M.M.); (D.N.)
| | - Mattia Matasci
- Philochem AG, CH-8112 Otelfingen, Switzerland; (B.G.); (A.K.); (A.V.); (M.M.); (D.N.)
| | - Andrea Schrepper
- Department of Cardiothoracic Surgery, Univerisity Hospital Jena, 07747 Jena, Germany;
| | - Alexander Pfeil
- Department of Internal Medicine III, Univerisity Hospital Jena, 07747 Jena, Germany;
| | - Laura Bäz
- Department of Internal Medicine I, Univerisity Hospital Jena, 07747 Jena, Germany; (K.G.); (L.B.); (P.C.S.)
| | - Christian Jung
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| | - P. Christian Schulze
- Department of Internal Medicine I, Univerisity Hospital Jena, 07747 Jena, Germany; (K.G.); (L.B.); (P.C.S.)
| | - Dario Neri
- Philochem AG, CH-8112 Otelfingen, Switzerland; (B.G.); (A.K.); (A.V.); (M.M.); (D.N.)
| | - Marcus Franz
- Department of Internal Medicine I, Univerisity Hospital Jena, 07747 Jena, Germany; (K.G.); (L.B.); (P.C.S.)
- Correspondence: ; Tel.: +49-3641-9324127
| |
Collapse
|
45
|
Mondéjar-Parreño G, Cogolludo A, Perez-Vizcaino F. Potassium (K +) channels in the pulmonary vasculature: Implications in pulmonary hypertension Physiological, pathophysiological and pharmacological regulation. Pharmacol Ther 2021; 225:107835. [PMID: 33744261 DOI: 10.1016/j.pharmthera.2021.107835] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/26/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023]
Abstract
The large K+ channel functional diversity in the pulmonary vasculature results from the multitude of genes expressed encoding K+ channels, alternative RNA splicing, the post-transcriptional modifications, the presence of homomeric or heteromeric assemblies of the pore-forming α-subunits and the existence of accessory β-subunits modulating the functional properties of the channel. K+ channels can also be regulated at multiple levels by different factors controlling channel activity, trafficking, recycling and degradation. The activity of these channels is the primary determinant of membrane potential (Em) in pulmonary artery smooth muscle cells (PASMC), providing an essential regulatory mechanism to dilate or contract pulmonary arteries (PA). K+ channels are also expressed in pulmonary artery endothelial cells (PAEC) where they control resting Em, Ca2+ entry and the production of different vasoactive factors. The activity of K+ channels is also important in regulating the population and phenotype of PASMC in the pulmonary vasculature, since they are involved in cell apoptosis, survival and proliferation. Notably, K+ channels play a major role in the development of pulmonary hypertension (PH). Impaired K+ channel activity in PH results from: 1) loss of function mutations, 2) downregulation of its expression, which involves transcription factors and microRNAs, or 3) decreased channel current as a result of increased vasoactive factors (e.g., hypoxia, 5-HT, endothelin-1 or thromboxane), exposure to drugs with channel-blocking properties, or by a reduction in factors that positively regulate K+ channel activity (e.g., NO and prostacyclin). Restoring K+ channel expression, its intracellular trafficking and the channel activity is an attractive therapeutic strategy in PH.
Collapse
Affiliation(s)
- Gema Mondéjar-Parreño
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain; Ciber Enfermedades Respiratorias (CIBERES), Spain
| | - Angel Cogolludo
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain; Ciber Enfermedades Respiratorias (CIBERES), Spain
| | - Francisco Perez-Vizcaino
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain; Ciber Enfermedades Respiratorias (CIBERES), Spain.
| |
Collapse
|
46
|
Dignam JP, Scott TE, Kemp-Harper BK, Hobbs AJ. Animal models of pulmonary hypertension: Getting to the heart of the problem. Br J Pharmacol 2021; 179:811-837. [PMID: 33724447 DOI: 10.1111/bph.15444] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/04/2021] [Accepted: 03/06/2021] [Indexed: 12/12/2022] Open
Abstract
Despite recent therapeutic advances, pulmonary hypertension (PH) remains a fatal disease due to the development of right ventricular (RV) failure. At present, no treatments targeted at the right ventricle are available, and RV function is not widely considered in the preclinical assessment of new therapeutics. Several small animal models are used in the study of PH, including the classic models of exposure to either hypoxia or monocrotaline, newer combinational and genetic models, and pulmonary artery banding, a surgical model of pure RV pressure overload. These models reproduce selected features of the structural remodelling and functional decline seen in patients and have provided valuable insight into the pathophysiology of RV failure. However, significant reversal of remodelling and improvement in RV function remains a therapeutic obstacle. Emerging animal models will provide a deeper understanding of the mechanisms governing the transition from adaptive remodelling to a failing right ventricle, aiding the hunt for druggable molecular targets.
Collapse
Affiliation(s)
- Joshua P Dignam
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Tara E Scott
- Department of Pharmacology, Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University Clayton Campus, Clayton, Victoria, Australia.,Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University Parkville Campus, Parkville, Victoria, Australia
| | - Barbara K Kemp-Harper
- Department of Pharmacology, Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University Clayton Campus, Clayton, Victoria, Australia
| | - Adrian J Hobbs
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
47
|
Inagaki T, Pearson JT, Tsuchimochi H, Schwenke DO, Saito S, Higuchi T, Masaki T, Umetani K, Shirai M, Nakaoka Y. Evaluation of right coronary vascular dysfunction in severe pulmonary hypertensive rats using synchrotron radiation microangiography. Am J Physiol Heart Circ Physiol 2021; 320:H1021-H1036. [PMID: 33481696 DOI: 10.1152/ajpheart.00327.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 01/06/2021] [Accepted: 01/06/2021] [Indexed: 11/22/2022]
Abstract
Pulmonary hypertension (PH) causes cardiac hypertrophy in the right ventricle (RV) and eventually leads to RV failure due to persistently elevated ventricular afterload. We hypothesized that the mechanical stress on the RV associated with increased afterload impairs vasodilator function of the right coronary artery (RCA) in PH. Coronary vascular response was assessed using microangiography with synchrotron radiation (SR) in two well-established PH rat models, monocrotaline injection or the combined exposure to chronic hypoxia and vascular endothelial growth factor receptor blockade with Su5416 (SuHx model). In the SuHx model, the effect of the treatment with the nonselective endothelin-1 receptor antagonist (ERA), macitentan, was also examined. Myocardial viability was determined in SuHx model rats, using 18F-FDG Positron emission tomography (PET) and magnetic resonance imaging (MRI). Endothelium-dependent and endothelium-independent vasodilator responses were significantly attenuated in the medium and small arteries of severe PH rats. ERA treatment significantly improved RCA vascular function compared with the untreated group. ERA treatment improved both the decrease in ejection fraction and the increased glucose uptake, and reduced RV remodeling. In addition, the upregulation of inflammatory genes in the RV was almost suppressed by ERA treatment. We found impairment of vasodilator responses in the RCA of severe PH rat models. Endothelin-1 activation in the RCA plays a major role in impaired vascular function in PH rats and is partially restored by ERA treatment. Treatment of PH with ERA may improve RV function in part by indirectly attenuating right heart afterload and in part by associated improvements in right coronary endothelial function.NEW & NOTEWORTHY We demonstrated for the first time the impairment of vascular responses in the right coronary artery (RCA) of the dysfunctional right heart in pulmonary hypertensive rats in vivo. Treatment with an endothelin-1 receptor antagonist ameliorated vascular dysfunction in the RCA, enabled tissue remodeling of the right heart, and improved cardiac function. Our results suggest that impaired RCA function might also contribute to the early progression to heart failure in patients with severe pulmonary arterial hypertension (PAH). The endothelium of the coronary vasculature might be considered as a potential target in treatments to prevent heart failure in severe patients with PAH.
Collapse
MESH Headings
- Animals
- Antihypertensive Agents/pharmacology
- Coronary Angiography
- Coronary Vessels/diagnostic imaging
- Coronary Vessels/drug effects
- Coronary Vessels/metabolism
- Coronary Vessels/physiopathology
- Disease Models, Animal
- Endothelin Receptor Antagonists/pharmacology
- Endothelin-1/genetics
- Endothelin-1/metabolism
- Hypertrophy, Right Ventricular/diagnostic imaging
- Hypertrophy, Right Ventricular/drug therapy
- Hypertrophy, Right Ventricular/metabolism
- Hypertrophy, Right Ventricular/physiopathology
- Hypoxia/complications
- Indoles
- Monocrotaline
- Predictive Value of Tests
- Pulmonary Arterial Hypertension/diagnostic imaging
- Pulmonary Arterial Hypertension/drug therapy
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/physiopathology
- Pyrimidines/pharmacology
- Pyrroles
- Rats, Sprague-Dawley
- Severity of Illness Index
- Sulfonamides/pharmacology
- Synchrotrons
- Vasodilation/drug effects
- Ventricular Dysfunction, Right/diagnostic imaging
- Ventricular Dysfunction, Right/drug therapy
- Ventricular Dysfunction, Right/metabolism
- Ventricular Dysfunction, Right/physiopathology
- Ventricular Function, Right
- Ventricular Remodeling
- Rats
Collapse
Affiliation(s)
- Tadakatsu Inagaki
- Department of Vascular Physiology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - James T Pearson
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
- Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Australia
| | - Hirotsugu Tsuchimochi
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Daryl O Schwenke
- Department of Physiology Heart-Otago, University of Otago, Dunedin, New Zealand
| | - Shigeyoshi Saito
- Department of Bio_Medical Imaging, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Takahiro Higuchi
- Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Takeshi Masaki
- Department of Vascular Physiology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Keiji Umetani
- Japan Synchrotron Radiation Research Institute, Harima, Japan
| | - Mikiyasu Shirai
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
- Department of Advanced Medical Research for Pulmonary Hypertension, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Yoshikazu Nakaoka
- Department of Vascular Physiology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
48
|
Liu H, Wang N, Li J, Wang W, Han W, Li Q. AAV1-Mediated shRNA Knockdown of SASH1 in Rat Bronchus Attenuates Hypoxia-Induced Pulmonary Artery Remodeling. Hum Gene Ther 2021; 32:796-805. [PMID: 33297837 DOI: 10.1089/hum.2020.242] [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: 11/13/2022] Open
Abstract
Pulmonary hypertension (PH) is a proliferative disease characterized by pulmonary arterial remodeling (PAR). SAM and SH3 domain containing 1 (SASH1) is a novel tumor suppressor gene whose biological function in PH is unclear. In this study, a hypoxia-induced pulmonary hypertension (HPH) rat model was constructed to explore the role of SASH1 in PAR. Histopathological changes in the lung tissue and hemodynamic alteration were detected in SASH1-knockdown rats through adeno-associated virus type-1 (AAV1) infection. In vitro, primary human pulmonary arterial smooth muscle cells (HPASMCs) were transfected with SASH1siRNA to investigate the effects of SASH1 on hypoxia-induced proliferation and migration. The molecular mechanisms associated with SASH1 were explored through knockdown and overexpression approaches. We found that SASH1 expression was significantly increased in rat pulmonary arteries and HPASMCs after hypoxia exposure. In vivo, silencing the SASH1 gene expression improved HPH in rats. The SASH1 downregulation inhibited proliferation and migration of hypoxia-induced HPASMCs. The protein expression of phospho-AKT (known as protein kinase B), proliferating cell nuclear antigen, and matrix metalloproteinase 9 (MMP9) in HPASMCs were increased after SASH1 overexpression, whereas these effects were inhibited by SASH1 knockdown. In conclusion, SASH1 downregulation improved hypoxia-induced PAR and PH. SASH1 may be a novel target for PH gene therapy in the era of precision medicine.
Collapse
Affiliation(s)
- Hong Liu
- Departments of Respiratory and Critical Care Medicine and.,Departments of Respiratory Disease Key Laboratory of Qingdao, Qingdao Municipal Hospital, Qingdao, China
| | - Ning Wang
- Departments of Respiratory and Critical Care Medicine and
| | - Jun Li
- Departments of Respiratory and Critical Care Medicine and
| | - Wenting Wang
- Departments of Oncology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | - Wei Han
- Departments of Respiratory and Critical Care Medicine and.,Departments of Respiratory Disease Key Laboratory of Qingdao, Qingdao Municipal Hospital, Qingdao, China
| | - Qinghai Li
- Departments of Respiratory and Critical Care Medicine and.,Departments of Respiratory Disease Key Laboratory of Qingdao, Qingdao Municipal Hospital, Qingdao, China
| |
Collapse
|
49
|
Park JF, Clark VR, Banerjee S, Hong J, Razee A, Williams T, Fishbein G, Saddic L, Umar S. Transcriptomic Analysis of Right Ventricular Remodeling in Two Rat Models of Pulmonary Hypertension: Identification and Validation of Epithelial-to-Mesenchymal Transition in Human Right Ventricular Failure. Circ Heart Fail 2021; 14:e007058. [PMID: 33541093 DOI: 10.1161/circheartfailure.120.007058] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Right ventricular (RV) dysfunction is a significant prognostic determinant of morbidity and mortality in pulmonary arterial hypertension (PAH). Despite the importance of RV function in PAH, the underlying molecular mechanisms of RV dysfunction secondary to PAH remain unclear. We aim to identify and compare molecular determinants of RV failure using RNA sequencing of RV tissue from 2 clinically relevant animal models of PAH. METHODS We performed RNA sequencing on RV from rats treated with monocrotaline or Sugen with hypoxia/normoxia. PAH and RV failure were confirmed by catheterization and echocardiography. We validated the RV transcriptome results using quantitative real-time polymerase chain reaction, immunofluorescence, and Western blot. Immunohistochemistry and immunofluorescence were performed on human RV tissue from control (n=3) and PAH-induced RV failure patients (n=5). RESULTS We identified similar transcriptomic profiles of RV from monocrotaline- and Sugen with hypoxia-induced RV failure. Pathway analysis showed genes enriched in epithelial-to-mesenchymal transition, inflammation, and metabolism. Histological staining of human RV tissue from patients with RV failure secondary to PAH revealed significant RV fibrosis and endothelial-to-mesenchymal transition, as well as elevated cellular communication network factor 2 (top gene implicated in epithelial-to-mesenchymal transition/endothelial-to-mesenchymal transition) expression in perivascular areas compared with normal RV. CONCLUSIONS Transcriptomic signature of RV failure in monocrotaline and Sugen with hypoxia models showed similar gene expressions and biological pathways. We provide translational relevance of this transcriptomic signature using RV from patients with PAH to demonstrate evidence of epithelial-to-mesenchymal transition/endothelial-to-mesenchymal transition and protein expression of cellular communication network factor 2 (CTGF [connective tissue growth factor]). Targeting specific molecular mechanisms responsible for RV failure in monocrotaline and Sugen with hypoxia models may identify novel therapeutic strategies for PAH-associated RV failure.
Collapse
Affiliation(s)
- John F Park
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine (J.F.P., V.R.C., S.B., J.H., A.R., T.W., L.S., S.U.), David Geffen School of Medicine, UCLA, Los Angeles, CA
| | - Varina R Clark
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine (J.F.P., V.R.C., S.B., J.H., A.R., T.W., L.S., S.U.), David Geffen School of Medicine, UCLA, Los Angeles, CA
| | - Somanshu Banerjee
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine (J.F.P., V.R.C., S.B., J.H., A.R., T.W., L.S., S.U.), David Geffen School of Medicine, UCLA, Los Angeles, CA
| | - Jason Hong
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine (J.F.P., V.R.C., S.B., J.H., A.R., T.W., L.S., S.U.), David Geffen School of Medicine, UCLA, Los Angeles, CA
- Division of Pulmonary Critical Care Medicine, Department of Medicine, UCLA, Los Angeles, CA (J.H.)
| | - Asif Razee
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine (J.F.P., V.R.C., S.B., J.H., A.R., T.W., L.S., S.U.), David Geffen School of Medicine, UCLA, Los Angeles, CA
| | - Tiffany Williams
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine (J.F.P., V.R.C., S.B., J.H., A.R., T.W., L.S., S.U.), David Geffen School of Medicine, UCLA, Los Angeles, CA
| | - Gregory Fishbein
- Department of Pathology (G.F.), David Geffen School of Medicine, UCLA, Los Angeles, CA
| | - Lou Saddic
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine (J.F.P., V.R.C., S.B., J.H., A.R., T.W., L.S., S.U.), David Geffen School of Medicine, UCLA, Los Angeles, CA
| | - Soban Umar
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine (J.F.P., V.R.C., S.B., J.H., A.R., T.W., L.S., S.U.), David Geffen School of Medicine, UCLA, Los Angeles, CA
| |
Collapse
|
50
|
Boengler K, Rohrbach S, Weissmann N, Schulz R. Importance of Cx43 for Right Ventricular Function. Int J Mol Sci 2021; 22:ijms22030987. [PMID: 33498172 PMCID: PMC7863922 DOI: 10.3390/ijms22030987] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/13/2021] [Accepted: 01/18/2021] [Indexed: 11/16/2022] Open
Abstract
In the heart, connexins form gap junctions, hemichannels, and are also present within mitochondria, with connexin 43 (Cx43) being the most prominent connexin in the ventricles. Whereas the role of Cx43 is well established for the healthy and diseased left ventricle, less is known about the importance of Cx43 for the development of right ventricular (RV) dysfunction. The present article focusses on the importance of Cx43 for the developing heart. Furthermore, we discuss the expression and localization of Cx43 in the diseased RV, i.e., in the tetralogy of Fallot and in pulmonary hypertension, in which the RV is affected, and RV hypertrophy and failure occur. We will also introduce other Cx molecules that are expressed in RV and surrounding tissues and have been reported to be involved in RV pathophysiology. Finally, we highlight therapeutic strategies aiming to improve RV function in pulmonary hypertension that are associated with alterations of Cx43 expression and function.
Collapse
|