1
|
Dominoni M, Melito C, Schirinzi S, Ghio S, Scelsi L, Greco A, Turco A, Broglia F, Fuardo M, Delmonte MP, Perotti F, Gardella B, Spinillo A. When pulmonary arterial hypertension and pregnancy meet: a multidisciplinary clinical experts review. Arch Gynecol Obstet 2024; 310:2839-2852. [PMID: 39570387 DOI: 10.1007/s00404-024-07827-1] [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: 07/08/2024] [Accepted: 11/03/2024] [Indexed: 11/22/2024]
Abstract
Pulmonary hypertension (PH) is a rare condition characterized by elevated pulmonary arterial pressure and pulmonary vascular resistance, potentially leading to right ventricular failure. Pulmonary arterial hypertension (PAH) is the most common type of PH in women of childbearing age and, as per the modified World Health Organization (mWHO) classification of maternal cardiovascular risk, it falls into mWHO class IV and pregnancy is contraindicated. These patients face an exceptionally high risk of maternal mortality and morbidity, with estimated maternal cardiac event rates ranging from 40 to 100% during pregnancy, because physiological changes happening in pregnancy exacerbate the disorder. Despite these recommendations, there is a growing incidence of pregnancy among women with PAH. Early referral to specialized centers, personalized therapies and expert multidisciplinary care involving pulmonary hypertension specialists, obstetricians, critical care specialists, anesthesiologists, and neonatologists are crucial steps to ensure positive outcomes for both mother and fetus. This review aims to examine the current understanding of pregnancy in patients with PAH, drawing on the experience of our center in the multidisciplinary management of pregnant women with this condition. In particular, we want to focus the attention of clinicians on the following aspects: early referral of pregnant patients to specialized centers, detailed counseling on the implications of pregnancy, initiation of therapy in treatment-naive patients and potential adjustment of therapy in non-naive patients, periodic risk assessment, evaluation of the appropriate timing of delivery, multidisciplinary management of the most critical periods, which are delivery and the post-partum phase.
Collapse
Affiliation(s)
- Mattia Dominoni
- Department of Clinical, Surgical, Diagnostic and Paediatric Sciences, University of Pavia, Corso Strada Nuova, 65, 27100, Pavia, Italy.
- Department of Obstetrics and Gynecology, IRCCS Foundation Policlinico San Matteo, Viale Camillo Golgi, 19, 27100, Pavia, Italy.
| | - Chiara Melito
- Department of Clinical, Surgical, Diagnostic and Paediatric Sciences, University of Pavia, Corso Strada Nuova, 65, 27100, Pavia, Italy
- Department of Obstetrics and Gynecology, IRCCS Foundation Policlinico San Matteo, Viale Camillo Golgi, 19, 27100, Pavia, Italy
| | - Sandra Schirinzi
- Division of Cardiology, Fondazione IRCCS Policlinico San Matteo, 27100, Pavia, Italy
| | - Stefano Ghio
- Division of Cardiology, Fondazione IRCCS Policlinico San Matteo, 27100, Pavia, Italy
| | - Laura Scelsi
- Division of Cardiology, Fondazione IRCCS Policlinico San Matteo, 27100, Pavia, Italy
| | - Alessandra Greco
- Division of Cardiology, Fondazione IRCCS Policlinico San Matteo, 27100, Pavia, Italy
| | - Annalisa Turco
- Division of Cardiology, Fondazione IRCCS Policlinico San Matteo, 27100, Pavia, Italy
| | - Federica Broglia
- Department of Anesthesia and Intensive Care 1, Unit of Obstetric Anesthesia, I Fondazione IRCCS Policlinico San Matteo, 27100, Pavia, Italy
| | - Marinella Fuardo
- Department of Anesthesia and Intensive Care 1, Unit of Obstetric Anesthesia, I Fondazione IRCCS Policlinico San Matteo, 27100, Pavia, Italy
| | - Maria Paola Delmonte
- Department of Anesthesia and Intensive Care 1, Unit of Obstetric Anesthesia, I Fondazione IRCCS Policlinico San Matteo, 27100, Pavia, Italy
| | - Francesca Perotti
- Department of Clinical, Surgical, Diagnostic and Paediatric Sciences, University of Pavia, Corso Strada Nuova, 65, 27100, Pavia, Italy
- Department of Obstetrics and Gynecology, IRCCS Foundation Policlinico San Matteo, Viale Camillo Golgi, 19, 27100, Pavia, Italy
| | - Barbara Gardella
- Department of Clinical, Surgical, Diagnostic and Paediatric Sciences, University of Pavia, Corso Strada Nuova, 65, 27100, Pavia, Italy
- Department of Obstetrics and Gynecology, IRCCS Foundation Policlinico San Matteo, Viale Camillo Golgi, 19, 27100, Pavia, Italy
| | - Arsenio Spinillo
- Department of Clinical, Surgical, Diagnostic and Paediatric Sciences, University of Pavia, Corso Strada Nuova, 65, 27100, Pavia, Italy
- Department of Obstetrics and Gynecology, IRCCS Foundation Policlinico San Matteo, Viale Camillo Golgi, 19, 27100, Pavia, Italy
| |
Collapse
|
2
|
Coulis A, Levanon S, Randhawa G, Brailovsky Y, Raza F, Oliveros E. Cardiopulmonary exercise testing in pulmonary arterial hypertension and chronic thromboembolic pulmonary hypertension. Front Sports Act Living 2024; 6:1477562. [PMID: 39635043 PMCID: PMC11614647 DOI: 10.3389/fspor.2024.1477562] [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: 08/08/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
Cardiopulmonary exercise testing allows for a comprehensive assessment of the mechanism of exercise limitation in pulmonary arterial hypertension and chronic thromboembolic pulmonary hypertension. Competitive pathophysiologic mechanisms may affect the clinical interpretation of cardiopulmonary disease as they relate to dyspnea, leg fatigue, and exercise intolerance.
Collapse
Affiliation(s)
- Alexis Coulis
- Temple Heart and Vascular Institute, Temple University Hospital, Philadelphia, PA, United States
| | - Shir Levanon
- Temple Heart and Vascular Institute, Temple University Hospital, Philadelphia, PA, United States
| | - Gurshaun Randhawa
- Temple Heart and Vascular Institute, Temple University Hospital, Philadelphia, PA, United States
| | - Yevgeniy Brailovsky
- Jefferson Heart Institute, Thomas Jefferson University, Philadelphia, PA, United States
| | - Farhan Raza
- Department of Medicine-Cardiovascular Division, University of Wisconsin Hospital, Madison, WI, United States
| | - Estefania Oliveros
- Temple Heart and Vascular Institute, Temple University Hospital, Philadelphia, PA, United States
| |
Collapse
|
3
|
Freire TC, Ferreira MS, De Angelis K, Paula-Ribeiro M. Respiratory, cardiovascular and musculoskeletal mechanisms involved in the pathophysiology of pulmonary hypertension: An updated systematic review of preclinical and clinical studies. Heart Lung 2024; 68:81-91. [PMID: 38941771 DOI: 10.1016/j.hrtlng.2024.06.001] [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: 01/16/2024] [Revised: 04/22/2024] [Accepted: 06/04/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND Progressive exercise intolerance is a hallmark of pulmonary hypertension (pH), severely impacting patients' independence and quality of life (QoL). Accumulating evidence over the last decade shows that combined abnormalities in peripheral reflexes and target organs contribute to disease progression and exercise intolerance. OBJECTIVE The aim of this study was to review the literature of the last decade on the contribution of the cardiovascular, respiratory, and musculoskeletal systems to pathophysiology and exercise intolerance in pH. METHODS A systematic literature search was conducted using specific terms in PubMed, SciELO, and the Cochrane Library databases for original pre-clinical or clinical studies published between 2013 and 2023. Studies followed randomized controlled/non-randomized controlled and pre-post designs. RESULTS The systematic review identified 25 articles reporting functional or structural changes in the respiratory, cardiovascular, and musculoskeletal systems in pH. Moreover, altered biomarkers in these systems, lower cardiac baroreflex, and heightened peripheral chemoreflex activity seemed to contribute to functional changes associated with poor prognosis and exercise intolerance in pH. Potential therapeutic strategies acutely explored involved manipulating the baroreflex and peripheral chemoreflex, improving cardiovascular autonomic control via cardiac vagal control, and targeting specific pathways such as GPER1, GDF-15, miR-126, and the JMJD1C gene. CONCLUSION Information published in the last 10 years advances the notion that pH pathophysiology involves functional and structural changes in the respiratory, cardiovascular, and musculoskeletal systems and their integration with peripheral reflexes. These findings suggest potential therapeutic targets, yet unexplored in clinical trials, that could assist in improving exercise tolerance and QoL in patients with pH.
Collapse
Affiliation(s)
- Thaís C Freire
- Translational Physiology Laboratory, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil; Department of Physiology, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Marília S Ferreira
- Translational Physiology Laboratory, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil; Department of Physiology, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Kátia De Angelis
- Translational Physiology Laboratory, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil; Department of Physiology, Federal University of Sao Paulo, Sao Paulo, Brazil
| | | |
Collapse
|
4
|
Signoretti C, Matsumura S, Fatehi S, D'Silva M, Mathew R, Cendali F, D'Alessandro A, Alam SMS, Garcia V, Miano JM, Gupte SA. G6pdN126D Variant Increases the Risk of Developing VEGFR (Vascular Endothelial Growth Factor Receptor) Blocker-Induced Pulmonary Vascular Disease. J Am Heart Assoc 2024; 13:e035174. [PMID: 39291493 DOI: 10.1161/jaha.123.035174] [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: 03/24/2024] [Accepted: 08/09/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND G6PD (glucose-6-phosphate-dehydrogenase) is a key enzyme in the glycolytic pathway and has been implicated in the pathogenesis of cancer and pulmonary hypertension-associated vascular remodeling. Here, we investigated the role of an X-linked G6pd mutation (N126D polymorphism), which is known to increase the risk of cardiovascular disease in individuals from sub-Saharan Africa and many others with African ancestry, in the pathogenesis of pulmonary hypertension induced by a vascular endothelial cell growth factor receptor blocker used for treating cancer. METHODS AND RESULTS CRISPR-Cas9 genome editing was used to generate the G6pd variant (N126D; G6pdN126D) in rats. A single dose of the vascular endothelial cell growth factor receptor blocker sugen-5416 (SU; 20 mg/kg in DMSO), which is currently in a Phase 2/3 clinical trial for cancer treatment, was subcutaneously injected into G6pdN126D rats and their wild-type littermates. After 8 weeks of normoxic conditions, right ventricular pressure and hypertrophy, pulmonary artery remodeling, the metabolic profile, and cytokine expression were assessed. Right ventricular pressure and pulmonary arterial wall thickness were increased in G6PDN126D+SU/normoxic rats. Simultaneously, levels of oxidized glutathione, inositol triphosphate, and intracellular Ca2+ were increased in the lungs of G6PDN126D+SU/normoxic rats, whereas nitric oxide was decreased. Also increased in G6PDN126D+SU/normoxic rats were pulmonary levels of plasminogen activator inhibitor-1, thrombin-antithrombin complex, and expression of proinflammatory cytokines CCL3 (chemokine [C-C motif] ligand), CCL5, and CCL7. CONCLUSIONS Our results suggest G6PDN126D increases inositol triphosphate-Ca2+ signaling, inflammation, thrombosis, and hypertrophic pulmonary artery remodeling in SU-treated rats. This suggests an increased risk of vascular endothelial cell growth factor receptor blocker-induced pulmonary hypertension in those carrying this G6PD variant.
Collapse
MESH Headings
- Animals
- Glucosephosphate Dehydrogenase/genetics
- Glucosephosphate Dehydrogenase/metabolism
- Receptors, Vascular Endothelial Growth Factor/genetics
- Rats
- Male
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/physiopathology
- Hypertension, Pulmonary/chemically induced
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/physiopathology
- Disease Models, Animal
- Vascular Remodeling/drug effects
- Rats, Sprague-Dawley
- Indoles/pharmacology
- Hypertrophy, Right Ventricular/genetics
- Hypertrophy, Right Ventricular/metabolism
- Hypertrophy, Right Ventricular/physiopathology
- Pyrroles
Collapse
Affiliation(s)
| | - Shun Matsumura
- Department of Pharmacology New York Medical College Valhalla NY USA
| | - Samuel Fatehi
- Department of Pharmacology New York Medical College Valhalla NY USA
| | - Melinee D'Silva
- Department of Pharmacology New York Medical College Valhalla NY USA
| | - Rajamma Mathew
- Department of Medicine, Division of Pediatric Cardiology, Physiology New York Medical College Valhalla NY USA
| | - Francesca Cendali
- Department of Biochemistry and Molecular Genetics University of Colorado Anschutz Medical Campus Aurora CO USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics University of Colorado Anschutz Medical Campus Aurora CO USA
| | - S M Shafiqul Alam
- Department of Pathology, Microbiology, and Immunology (PMI) New York Medical College Valhalla NY USA
| | - Victor Garcia
- Department of Pharmacology New York Medical College Valhalla NY USA
| | - Joseph M Miano
- Department of Medicine Vascular Biology Center, Medical College of Georgia at Augusta University Augusta GA USA
| | - Sachin A Gupte
- Department of Pharmacology New York Medical College Valhalla NY USA
| |
Collapse
|
5
|
Zhang S, Wang J, Wen J, Xin Q, Wang J, Ju Z, Luan Y. MSC-derived exosomes attenuates pulmonary hypertension via inhibiting pulmonary vascular remodeling. Exp Cell Res 2024; 442:114256. [PMID: 39299482 DOI: 10.1016/j.yexcr.2024.114256] [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: 07/18/2024] [Revised: 09/05/2024] [Accepted: 09/14/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a serious cardiopulmonary disease with significant morbidity and mortality. Vascular obstruction leads to a continuous increase in pulmonary vascular resistance, vascular remodeling, and right ventricular hypertrophy and failure, which are the main pathological features of PH. Currently, the treatments for PH are very limited, so new methods are urgently needed. Msenchymal stem cells-derived exosomes have been shown to have significant therapeutic effects in PH, however, the mechanism still very blurry. Here, we investigated the possible mechanism by which umbilical cord mesenchymal stem cell-derived exosomes (hUC-MSC-EXO) inhibited monocrotaline (MCT)-induced pulmonary vascular remodeling in a rat model of PH by regulating the NF-κB/BMP signaling pathway. Our data revealed that hUC-MSC-EXO could significantly attenuate MCT-induced PH and right ventricular hypertrophy. Moreover, the protein expression level of BMPR2, BMP-4, BMP-9 and ID1 was significantly increased, but NF-κB p65, p-NF-κB-p65 and BMP antagonists Gremlin-1 was increased in vitro and vivo. Collectively, this study revealed that the mechanism of hUC-MSC-EXO attenuates pulmonary hypertension may be related to inhibition of NF-κB signaling to further activation of BMP signaling. The present study provided a promising therapeutic strategy for PH vascular remodeling.
Collapse
Affiliation(s)
- Shanshan Zhang
- Department of Emergency, The Second Hospital of Shandong University, PR China
| | - Junfu Wang
- College of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250000, PR China
| | - Jiang Wen
- Institute of Medical Sciences, The Second Hospital of Shandong University, No. 247, Beiyuan Dajie, Jinan, 250000, PR China
| | - Qian Xin
- Institute of Medical Sciences, The Second Hospital of Shandong University, No. 247, Beiyuan Dajie, Jinan, 250000, PR China
| | - Jue Wang
- Institute of Medical Sciences, The Second Hospital of Shandong University, No. 247, Beiyuan Dajie, Jinan, 250000, PR China
| | - Zhiye Ju
- Department of Ultrasound, Shandong Provincial Public Health Clinical Center, No. 46, Lishan Road, Jinan, 250000, PR China.
| | - Yun Luan
- Institute of Medical Sciences, The Second Hospital of Shandong University, No. 247, Beiyuan Dajie, Jinan, 250000, PR China.
| |
Collapse
|
6
|
Wang S, Awad KS, Chen LY, Siddique MAH, Ferreyra GA, Wang CL, Joseph T, Yu ZX, Takeda K, Demirkale CY, Zhao YY, Elinoff JM, Danner RL. Endothelial PHD2 deficiency induces apoptosis resistance and inflammation via AKT activation and AIP1 loss independent of HIF2α. Am J Physiol Lung Cell Mol Physiol 2024; 327:L503-L519. [PMID: 39159362 PMCID: PMC11482463 DOI: 10.1152/ajplung.00077.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/16/2024] [Accepted: 08/09/2024] [Indexed: 08/21/2024] Open
Abstract
In hypoxic and pseudohypoxic rodent models of pulmonary hypertension (PH), hypoxia-inducible factor (HIF) inhibition attenuates disease initiation. However, HIF activation alone, due to genetic alterations or use of inhibitors of prolyl hydroxylase domain (PHD) enzymes, has not been definitively shown to cause PH in humans, indicating the involvement of other mechanisms. Given the association between endothelial cell dysfunction and PH, the effects of pseudohypoxia and its underlying pathways were investigated in primary human lung endothelial cells. PHD2 silencing or inhibition, while activating HIF2α, induced apoptosis-resistance and IFN/STAT activation in endothelial cells, independent of HIF signaling. Mechanistically, PHD2 deficiency activated AKT and ERK, inhibited JNK, and reduced AIP1 (ASK1-interacting protein 1), all independent of HIF2α. Like PHD2, AIP1 silencing affected these same kinase pathways and produced a similar dysfunctional endothelial cell phenotype, which was partially reversed by AKT inhibition. Consistent with these in vitro findings, AIP1 protein levels in lung endothelial cells were decreased in Tie2-Cre/Phd2 knockout mice compared with wild-type controls. Lung vascular endothelial cells from patients with pulmonary arterial hypertension (PAH) showed IFN/STAT activation. Lung tissue from both SU5416/hypoxia PAH rats and patients with PAH all showed AKT activation and dysregulated AIP1 expression. In conclusion, PHD2 deficiency in lung vascular endothelial cells drives an apoptosis-resistant and inflammatory phenotype, mediated by AKT activation and AIP1 loss independent of HIF signaling. Targeting these pathways, including PHD2, AKT, and AIP1, holds the potential for developing new treatments for endothelial dysfunction in PH.NEW & NOTEWORTHY HIF activation alone does not conclusively lead to human PH, suggesting that HIF-independent signaling may also contribute to hypoxia-induced PH. This study demonstrated that PHD2 silencing-induced pseudohypoxia in human lung endothelial cells suppresses apoptosis and activates STAT, effects that persist despite HIF2α inhibition or knockdown and are attributed to AKT and ERK activation, JNK inhibition, and AIP1 loss. These findings align with observations in lung endothelial cells and tissues from PAH rodent models and patients.
Collapse
Affiliation(s)
- Shuibang Wang
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, United States
| | - Keytam S Awad
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, United States
| | - Li-Yuan Chen
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, United States
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Mohammad A H Siddique
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, United States
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Gabriela A Ferreyra
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, United States
| | - Caroline L Wang
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, United States
| | - Thea Joseph
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, United States
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Zu-Xi Yu
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Kazuyo Takeda
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Cumhur Y Demirkale
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, United States
| | - You-Yang Zhao
- Section for Injury Repair and Regeneration, Stanley Manne Children Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, United States
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States
| | - Jason M Elinoff
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, United States
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Robert L Danner
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
7
|
Krause PN, McGeorge G, McPeek JL, Khalid S, Nelin LD, Liu Y, Chen B. Pde3a and Pde3b regulation of murine pulmonary artery smooth muscle cell growth and metabolism. Physiol Rep 2024; 12:e70089. [PMID: 39435735 PMCID: PMC11494452 DOI: 10.14814/phy2.70089] [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: 09/05/2024] [Revised: 10/05/2024] [Accepted: 10/05/2024] [Indexed: 10/23/2024] Open
Abstract
A role for metabolically active adipose tissue in pulmonary hypertension (PH) pathogenesis is emerging. Alterations in cellular metabolism in metabolic syndrome are triggers of PH-related vascular dysfunction. Metabolic reprogramming in proliferative pulmonary vascular cells causes a metabolic switch from oxidative phosphorylation to glycolysis. PDE3A and PDE3B subtypes in the regulation of metabolism in pulmonary artery smooth muscle cells (PASMC) are poorly understood. We previously found that PDE3A modulates the cellular energy sensor, AMPK, in human PASMC. We demonstrate that global Pde3a knockout mice have right ventricular (RV) hypertrophy, elevated RV systolic pressures, and metabolic dysfunction with elevated serum free fatty acids (FFA). Therefore, we sought to delineate Pde3a/Pde3b regulation of metabolic pathways in PASMC. We found that PASMC Pde3a deficiency, and to a lesser extent Pde3b deficiency, downregulates AMPK, CREB and PPARγ, and upregulates pyruvate kinase dehydrogenase expression, suggesting decreased oxidative phosphorylation. Interestingly, siRNA Pde3a knockdown in adipocytes led to elevated FFA secretion. Furthermore, PASMC exposed to siPDE3A-transfected adipocyte media led to decreased α-SMA, AMPK and CREB phosphorylation, and greater viable cell numbers compared to controls under the same conditions. These data demonstrate that deficiencies of Pde3a and Pde3b alter pathways that affect cell growth and metabolism in PASMC.
Collapse
MESH Headings
- Animals
- Male
- Mice
- AMP-Activated Protein Kinases/metabolism
- AMP-Activated Protein Kinases/genetics
- Cell Proliferation
- Cells, Cultured
- Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism
- Cyclic Nucleotide Phosphodiesterases, Type 3/genetics
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/cytology
- Myocytes, Smooth Muscle/metabolism
- PPAR gamma/metabolism
- PPAR gamma/genetics
- Pulmonary Artery/metabolism
- Pulmonary Artery/cytology
Collapse
Affiliation(s)
- Paulina N. Krause
- Center for Perinatal ResearchAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
| | - Gabrielle McGeorge
- Center for Perinatal ResearchAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
| | - Jennifer L. McPeek
- Center for Perinatal ResearchAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
| | - Sidra Khalid
- Center for Perinatal ResearchAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
| | - Leif D. Nelin
- Center for Perinatal ResearchAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
- Department of PediatricsThe Ohio State University College of MedicineColumbusOhioUSA
| | - Yusen Liu
- Center for Perinatal ResearchAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
- Department of PediatricsThe Ohio State University College of MedicineColumbusOhioUSA
| | - Bernadette Chen
- Center for Perinatal ResearchAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
- Department of PediatricsThe Ohio State University College of MedicineColumbusOhioUSA
| |
Collapse
|
8
|
Ishii S, Hatano M, Minatsuki S, Hirose K, Saito A, Yagi H, Shimbo M, Soma K, Konoeda C, Sato M, Nakajima J, Komuro I. Comprehensive Risk Assessment in Patients With Pulmonary Arterial Hypertension Referred for Lung Transplantation. Circ J 2024; 88:1610-1617. [PMID: 38403681 DOI: 10.1253/circj.cj-23-0790] [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] [Indexed: 02/27/2024]
Abstract
BACKGROUND Whether comprehensive risk assessment predicts post-referral outcome in patients with pulmonary arterial hypertension (PAH) referred for lung transplantation (LT) in Japan is unknown. METHODS AND RESULTS We retrospectively analyzed 52 PAH patients referred for LT. Risk status at referral was assessed using 3- and 4-strata models from the 2022 European Society of Cardiology and European Respiratory Society guidelines. The 3-strata model intermediate-risk group was further divided into 2 groups based on the median proportion of low-risk variables (modified risk assessment [MRA]). The primary outcome was post-referral mortality. During follow-up, 9 patients died and 13 patients underwent LT. There was no survival difference among 3-strata model groups. The 4-strata model classified 33, 16, and 3 patients as low intermediate, high intermediate, and high risk, respectively. The 4-strata model identified high-risk patients with a 1-year survival rate of 33%, but did not discriminate survival between the intermediate-risk groups. The MRA classified 15, 28, 8, and 1 patients as low, low intermediate, high intermediate, and high risk, respectively. High intermediate- or high-risk patients had worse survival (P<0.001), with 1- and 3-year survival rates of 64% and 34%, respectively. MRA high intermediate- or high-risk classification was associated with mortality (hazard ratio 12.780; 95% confidence interval 2.583-63.221; P=0.002). CONCLUSIONS Patients classified as high intermediate or high risk by the MRA after treatment should be referred for LT.
Collapse
Affiliation(s)
- Satoshi Ishii
- Department of Cardiovascular Medicine, The University of Tokyo Hospital
| | - Masaru Hatano
- Department of Cardiovascular Medicine, The University of Tokyo Hospital
- Department of Advanced Medical Center for Heart Failure, The University of Tokyo Hospital
| | - Shun Minatsuki
- Department of Cardiovascular Medicine, The University of Tokyo Hospital
| | - Kazutoshi Hirose
- Department of Cardiovascular Medicine, The University of Tokyo Hospital
| | - Akihito Saito
- Department of Cardiovascular Medicine, The University of Tokyo Hospital
| | - Hiroki Yagi
- Department of Cardiovascular Medicine, The University of Tokyo Hospital
| | - Mai Shimbo
- Department of Cardiovascular Medicine, The University of Tokyo Hospital
- Department of Computational Diagnostic Radiology and Preventive Medicine, The University of Tokyo Hospital
| | - Katsura Soma
- Department of Cardiovascular Medicine, The University of Tokyo Hospital
| | - Chihiro Konoeda
- Department of Thoracic Surgery, The University of Tokyo Hospital
| | - Masaaki Sato
- Department of Thoracic Surgery, The University of Tokyo Hospital
| | - Jun Nakajima
- Department of Thoracic Surgery, The University of Tokyo Hospital
| | - Issei Komuro
- Department of Cardiovascular Medicine, The University of Tokyo Hospital
- Department of Frontier Cardiovascular Science, The University of Tokyo Hospital
- International University of Health and Welfare
| |
Collapse
|
9
|
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
|
10
|
Yuan Q, Xiao LW, Zhang Y, Li L, Xia T, Xu Q, Xing SG, Wang LS. Inverted U-Shaped relationship Between Systemic Immune-Inflammation Index and Pulmonary Function: A Large Population-Based Study in US Adults. Int J Chron Obstruct Pulmon Dis 2024; 19:1971-1987. [PMID: 39247667 PMCID: PMC11379031 DOI: 10.2147/copd.s471068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 08/13/2024] [Indexed: 09/10/2024] Open
Abstract
Background Systemic immune-inflammation index (SII) is a novel comprehensive inflammatory marker. Inflammation is associated with impaired lung function. We aimed to explore the possible relationship between SII and lung function to examine the potential of SII in predicting lung function decline. Methods A cross-sectional survey was conducted using the data of the NHANES from 2007 to 2012. Multiple linear regression models were used to analyze the linear relationship between SII and pulmonary functions. Sensitivity analyses, subgroup analyses, and interaction tests were used to examine the robustness of this relationship across populations. Fitted smooth curves and threshold effect analysis were used to describe the nonlinear relationships. Results A total of 10,125 patients were included in this study. After adjusting for all covariates, multiple linear regression model analysis showed that high Log2-SII level was significantly associated with decreased FVC(β, -23.4061; 95% CI, -42.2805- -4.5317), FEV1(β, -46.7730; 95% CI, -63.3371- -30.2089), FEV1%(β, -0.7923; 95% CI, -1.1635- -0.4211), FEV1/FVC(β, -0.6366; 95% CI, -0.8328- -0.4404) and PEF(β, -121.4468; 95% CI,-164.1939- -78.6998). The negative correlation between Log2-SII and pulmonary function indexes remained stable in trend test and stratified analysis. Inverted U-shaped relationships between Log2-SII and FVC, FEV1, FEV1%, and PEF were observed, while a negative linear correlation existed between FEV1/FVC and Log2-SII. The cutoff values of the nonlinear relationship between Log2-SII and FVC, FEV1, FEV1%, PEF were 8.3736, 8.0688, 8.3745, and 8.5255, respectively. When SII exceeded the critical value, the lung function decreased significantly. Conclusion This study found a close correlation between SII and pulmonary function indicators. This study investigated the SII threshold when lung functions began to decline in the overall population. SII may become a promising serological indicator for predicting lung function decline. However, prospective studies were needed further to establish the causal relationship between these two factors.
Collapse
Affiliation(s)
- Qian Yuan
- Department of Thoracic Surgery, Nan Jing Gaochun PEople's Hospital (The Gaochun Affiliated Hospital of Jiang Su University), Nanjing, Jiangsu, 210000, People's Republic of China
| | - Long-Wu Xiao
- Department of Thoracic Surgery, Nan Jing Gaochun PEople's Hospital (The Gaochun Affiliated Hospital of Jiang Su University), Nanjing, Jiangsu, 210000, People's Republic of China
| | - Yao Zhang
- Department of Thoracic Surgery, Nan Jing Gaochun PEople's Hospital (The Gaochun Affiliated Hospital of Jiang Su University), Nanjing, Jiangsu, 210000, People's Republic of China
| | - Long Li
- Department of Thoracic Surgery, Nan Jing Gaochun PEople's Hospital (The Gaochun Affiliated Hospital of Jiang Su University), Nanjing, Jiangsu, 210000, People's Republic of China
| | - Teng Xia
- Department of Thoracic Surgery, Nan Jing Gaochun PEople's Hospital (The Gaochun Affiliated Hospital of Jiang Su University), Nanjing, Jiangsu, 210000, People's Republic of China
| | - Qing Xu
- Department of Thoracic Surgery, Nan Jing Gaochun PEople's Hospital (The Gaochun Affiliated Hospital of Jiang Su University), Nanjing, Jiangsu, 210000, People's Republic of China
| | - Shi-Gui Xing
- Department of Thoracic Surgery, Nan Jing Gaochun PEople's Hospital (The Gaochun Affiliated Hospital of Jiang Su University), Nanjing, Jiangsu, 210000, People's Republic of China
| | - Liu-Shun Wang
- Department of Thoracic Surgery, Nan Jing Gaochun PEople's Hospital (The Gaochun Affiliated Hospital of Jiang Su University), Nanjing, Jiangsu, 210000, People's Republic of China
| |
Collapse
|
11
|
Papadimitriou TI, Lemmers JMJ, van Caam APM, Vos JL, Vitters EL, Stinissen L, van Leuven SI, Koenders MI, van der Kraan PM, Koenen HJPM, Smeets RL, Nijveldt R, Vonk MC, Thurlings RM. Systemic sclerosis-associated pulmonary arterial hypertension is characterized by a distinct peripheral T helper cell profile. Rheumatology (Oxford) 2024; 63:2525-2534. [PMID: 38552313 PMCID: PMC11371376 DOI: 10.1093/rheumatology/keae190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 03/17/2024] [Indexed: 09/05/2024] Open
Abstract
OBJECTIVES Systemic sclerosis (SSc) is characterized by multiple clinical manifestations. Vasculopathy is a main disease hallmark and ranges in severity from an exacerbated Raynaud phenomenon to pulmonary arterial hypertension (PAH). The potential involvement of the immune system in SSc-associated vascular abnormalities is not clear. Here, we set out to study SSc-related immune parameters and determine whether and which peripheral T cell subsets associate with vascular severity in SSc patients. METHODS Peripheral blood and clinical data were collected from 30 SSc patients, 5 patients with idiopathic PAH and 15 age and sex-matched healthy donors (HD). In this cross-sectional cohort, SSc patients with PAH (n = 15) were matched for their age, sex and medication with SSc patients with no signs of PAH (n = 15). Lymphocyte subsets were quantified by multi-colour flow cytometry. RESULTS SSc patients exhibited elevated percentages of T peripheral helper cells (Tph), CD4+GZMB+ T cells and decreased levels of Th1 cells compared with HD. Increased presence of both CD4+ and CD8+ exhausted-like (CD28-) T cells, characterized by raised cytokine and cytotoxic signature, was also observed in SSc compared with HD blood. Furthermore, IL-4 expressing CD4+CD8+ T cells were significantly increased in SSc peripheral blood. Interestingly, the presence of PAH in SSc was accompanied by a distinct T helper profile, characterized by raised percentages of Th17 and Tph cells. CONCLUSION SSc patients with severe vasculopathy (presence of PAH) exhibited a distinct T cell profile, suggesting a potential role of auto-immune inflammation in SSc vascular complications.
Collapse
Affiliation(s)
- Theodoros Ioannis Papadimitriou
- Department of Rheumatology, Radboudumc, Nijmegen, The Netherlands
- Department of Laboratory Medicine – Medical Immunology, Radboudumc, Nijmegen, The Netherlands
| | | | | | | | - Elly L Vitters
- Department of Rheumatology, Radboudumc, Nijmegen, The Netherlands
| | - Lizan Stinissen
- Department of Rheumatology, Radboudumc, Nijmegen, The Netherlands
| | | | | | | | - Hans J P M Koenen
- Department of Laboratory Medicine – Medical Immunology, Radboudumc, Nijmegen, The Netherlands
| | - Ruben L Smeets
- Department of Laboratory Medicine – Medical Immunology, Radboudumc, Nijmegen, The Netherlands
- Radboudumc Laboratory for Diagnostics, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Robin Nijveldt
- Department of Cardiology, Radboudumc, Nijmegen, The Netherlands
| | - Madelon C Vonk
- Department of Rheumatology, Radboudumc, Nijmegen, The Netherlands
| | | |
Collapse
|
12
|
Mahajan A, Kumar A, Chen L, Dhillon NK. LncRNA-536 and RNA Binding Protein RBM25 Interactions in Pulmonary Arterial Hypertension. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.27.610011. [PMID: 39253448 PMCID: PMC11383286 DOI: 10.1101/2024.08.27.610011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
OBJECTIVE Hyperproliferation of pulmonary artery smooth muscle cells (PASMCs) is one of the essential features of the maladaptive inward remodeling of the pulmonary arteries in pulmonary arterial hypertension (PAH). In this study, we define the mechanistic association between long-noncoding RNA: ENST00000495536 (Lnc-536) and anti-proliferative HOXB13 in mediating smooth muscle hyperplasia. METHODS Antisense oligonucleotide-based GapmeRs or plasmid overexpressing lnc-536 were used to evaluate the role of lnc-536 in mediating hyperproliferation of PDGF-treated or idiopathic PAH (IPAH) PASMCs. Further, we pulled down lnc536 to identify the proteins directly interacting with lnc536. The in-vivo role of lnc-536 was determined in Sugen-hypoxia and HIV-transgenic pulmonary hypertensive rats. RESULTS Increased levels of lnc-536 in PDGF-treated or IPAH PASMCs promote hyperproliferative phenotype by downregulating the HOXB13 expression. Knockdown of lnc-536 in-vivo prevented increased RVSP, Fulton Index, and pulmonary vascular remodeling in Sugen-Hypoxia rats. The lncRNA-536 pull-down assay demonstrated the interactions of RNA binding protein: RBM25 with SFPQ, a transcriptional regulator that has a binding motif on HOXB13 exon Further, The RNA-IP experiment using the SFPQ antibody showed direct interaction of RBM25 with SFPQ and knockdown of RBM25 resulted in increased interactions of SFPQ and HOXB13 mRNA while attenuating PASMC proliferation. Finally, we examined the role of lnc-536 and HOXB13 axis in the PASMCs exposed to the dual hit of HIV and a stimulant: cocaine as well. CONCLUSION lnc-536 acts as a decoy for RBM25, which in turn sequesters SFPQ, leading to the decrease in HOXB13 expression and hyperproliferation of smooth muscle cells associated with PAH development.
Collapse
Affiliation(s)
- Aatish Mahajan
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Ashok Kumar
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Ling Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Navneet K Dhillon
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS
| |
Collapse
|
13
|
Xiao R, Liu J, Shi L, Zhang T, Liu J, Qiu S, Ruiz M, Dupuis J, Zhu L, Wang L, Wang Z, Hu Q. Au-modified ceria nanozyme prevents and treats hypoxia-induced pulmonary hypertension with greatly improved enzymatic activity and safety. J Nanobiotechnology 2024; 22:492. [PMID: 39160624 PMCID: PMC11331617 DOI: 10.1186/s12951-024-02738-4] [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: 11/23/2023] [Accepted: 07/24/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND Despite recent advances the prognosis of pulmonary hypertension remains poor and warrants novel therapeutic options. Extensive studies, including ours, have revealed that hypoxia-induced pulmonary hypertension is associated with high oxidative stress. Cerium oxide nanozyme or nanoparticles (CeNPs) have displayed catalytic activity mimicking both catalase and superoxide dismutase functions and have been widely used as an anti-oxidative stress approach. However, whether CeNPs can attenuate hypoxia-induced pulmonary vascular oxidative stress and pulmonary hypertension is unknown. RESULTS In this study, we designed a new ceria nanozyme or nanoparticle (AuCeNPs) exhibiting enhanced enzyme activity. The AuCeNPs significantly blunted the increase of reactive oxygen species and intracellular calcium concentration while limiting proliferation of pulmonary artery smooth muscle cells and pulmonary vasoconstriction in a model of hypoxia-induced pulmonary hypertension. In addition, the inhalation of nebulized AuCeNPs, but not CeNPs, not only prevented but also blunted hypoxia-induced pulmonary hypertension in rats. The benefits of AuCeNPs were associated with limited increase of intracellular calcium concentration as well as enhancement of extracellular calcium-sensing receptor (CaSR) activity and expression in rat pulmonary artery smooth muscle cells. Nebulised AuCeNPs showed a favorable safety profile, systemic arterial pressure, liver and kidney function, plasma Ca2+ level, and blood biochemical parameters were not affected. CONCLUSION We conclude that AuCeNPs is an improved reactive oxygen species scavenger that effectively prevents and treats hypoxia-induced pulmonary hypertension.
Collapse
Affiliation(s)
- Rui Xiao
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), 13 Hangkong Road, Wuhan, 430030, China
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, HUST, Wuhan, China
| | - Jia Liu
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, HUST, Wuhan, China
| | - Lin Shi
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, HUST, Wuhan, China
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, HUST, Wuhan, China
| | - Ting Zhang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), 13 Hangkong Road, Wuhan, 430030, China
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, HUST, Wuhan, China
| | - Jie Liu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), 13 Hangkong Road, Wuhan, 430030, China
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, HUST, Wuhan, China
| | - Shuyi Qiu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), 13 Hangkong Road, Wuhan, 430030, China
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, HUST, Wuhan, China
| | - Matthieu Ruiz
- Department of Nutrition, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Montréal, Québec, Canada
| | - Jocelyn Dupuis
- Montreal Heart Institute, Montréal, Québec, Canada
- Department of medicine, Université de Montréal, Montréal, Québec, Canada
| | - Liping Zhu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), 13 Hangkong Road, Wuhan, 430030, China.
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, HUST, Wuhan, China.
| | - Lin Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, HUST, Wuhan, China.
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, HUST, Wuhan, China.
| | - Zheng Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, HUST, Wuhan, China.
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, HUST, Wuhan, China.
| | - Qinghua Hu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology (HUST), 13 Hangkong Road, Wuhan, 430030, China.
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, HUST, Wuhan, China.
| |
Collapse
|
14
|
Correale M, Tricarico L, Chirivì F, Bevere EML, Ruggeri D, Migliozzi C, Rossi L, Vitullo A, Granatiero M, Granato M, Villani D, Giannetti L, Iacoviello M, Brunetti ND. Endothelial Function Correlates With Pulmonary Pressures in Subjects With Clinically Suspected Pulmonary Hypertension. Am J Cardiol 2024; 225:61-66. [PMID: 38897266 DOI: 10.1016/j.amjcard.2024.06.019] [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: 01/08/2024] [Accepted: 06/15/2024] [Indexed: 06/21/2024]
Abstract
Impaired pulmonary circulation hemodynamics are characteristic of pulmonary hypertension (PH). We therefore sought to evaluate possible correlations between endothelial function noninvasively assessed by brachial artery flow-mediated dilation (FMD) and hemodynamic parameters at right-sided cardiac catheterization in patients with clinically suspected PH. Consecutive outpatients with suspected PH were enrolled in the study. In all patients, endothelial function was assessed by FMD and hemodynamic parameters (pulmonary artery pressure [PAP]); pulmonary vascular resistances [PVR]) were derived by right-sided cardiac catheterization. For this study, 95 consecutive patients with suspected PH were enrolled (mean age 63 ± 13 years, 58% male) and included in the analysis. FMD values were significantly correlated with systolic (s)PAP levels (r = -0.29, p = 0.016); correlation with PVR was of borderline significance (r = -0.21, p = 0.78). After multivariable regression analysis including age, gender, tricuspid annular plane systolic excursion and peak tricuspid regurgitation velocity (peak TRV), and FMD, the latter remained significantly correlated with systolic pulmonary artery pressure (sPAP) values (B = -47, p = 0.02). After classifying patients according to median levels of peak TRV and FMD into 3 groups (neither, either, or both impaired), progressively increased levels of sPAP, mean PAP, and PVR were found (p for trend <0.001 in all cases). FMD values were inversely related to sPAP levels in a small population of patients with clinically suspected PH. In combination with peak TRV levels, FMD values noninvasively assessed were predictive of increased sPAP, mean PAP, and PVR.
Collapse
Affiliation(s)
- Michele Correale
- Cardiothoracic Department, Ospedali Riuniti University Hospital, Foggia, Italy
| | - Lucia Tricarico
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Francesco Chirivì
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | | | - Debora Ruggeri
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Celeste Migliozzi
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Luciano Rossi
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Antonio Vitullo
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Michele Granatiero
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Mattia Granato
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Deborah Villani
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Laura Giannetti
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Massimo Iacoviello
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | | |
Collapse
|
15
|
Bordan Z, Batori RK, Haigh S, Li X, Meadows ML, Brown ZL, West MA, Dong K, Han W, Su Y, Ma Q, Huo Y, Zhou J, Abdelbary M, Sullivan JC, Weintraub NL, Stepp DW, Chen F, Barman SA, Fulton DJR. PDZ-Binding Kinase, a Novel Regulator of Vascular Remodeling in Pulmonary Arterial Hypertension. Circulation 2024; 150:393-410. [PMID: 38682326 DOI: 10.1161/circulationaha.123.067095] [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: 09/15/2023] [Accepted: 03/04/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is high blood pressure in the lungs that originates from structural changes in small resistance arteries. A defining feature of PAH is the inappropriate remodeling of pulmonary arteries (PA) leading to right ventricle failure and death. Although treatment of PAH has improved, the long-term prognosis for patients remains poor, and more effective targets are needed. METHODS Gene expression was analyzed by microarray, RNA sequencing, quantitative polymerase chain reaction, Western blotting, and immunostaining of lung and isolated PA in multiple mouse and rat models of pulmonary hypertension (PH) and human PAH. PH was assessed by digital ultrasound, hemodynamic measurements, and morphometry. RESULTS Microarray analysis of the transcriptome of hypertensive rat PA identified a novel candidate, PBK (PDZ-binding kinase), that was upregulated in multiple models and species including humans. PBK is a serine/threonine kinase with important roles in cell proliferation that is minimally expressed in normal tissues but significantly increased in highly proliferative tissues. PBK was robustly upregulated in the medial layer of PA, where it overlaps with markers of smooth muscle cells. Gain-of-function approaches show that active forms of PBK increase PA smooth muscle cell proliferation, whereas silencing PBK, dominant negative PBK, and pharmacological inhibitors of PBK all reduce proliferation. Pharmacological inhibitors of PBK were effective in PH reversal strategies in both mouse and rat models, providing translational significance. In a complementary genetic approach, PBK was knocked out in rats using CRISPR/Cas9 editing, and loss of PBK prevented the development of PH. We found that PBK bound to PRC1 (protein regulator of cytokinesis 1) in PA smooth muscle cells and that multiple genes involved in cytokinesis were upregulated in experimental models of PH and human PAH. Active PBK increased PRC1 phosphorylation and supported cytokinesis in PA smooth muscle cells, whereas silencing or dominant negative PBK reduced cytokinesis and the number of cells in the G2/M phase of the cell cycle. CONCLUSIONS PBK is a newly described target for PAH that is upregulated in proliferating PA smooth muscle cells, where it contributes to proliferation through changes in cytokinesis and cell cycle dynamics to promote medial thickening, fibrosis, increased PA resistance, elevated right ventricular systolic pressure, right ventricular remodeling, and PH.
Collapse
Affiliation(s)
- Zsuzsanna Bordan
- Vascular Biology Center (Z.B., R.K.B., S.H., Z.L.B., M.A.W., Q.M., Y.H., N.L.W., D.W.S., D.J.R.F.), Medical College of Georgia, Augusta University
| | - Robert K Batori
- Vascular Biology Center (Z.B., R.K.B., S.H., Z.L.B., M.A.W., Q.M., Y.H., N.L.W., D.W.S., D.J.R.F.), Medical College of Georgia, Augusta University
| | - Stephen Haigh
- Vascular Biology Center (Z.B., R.K.B., S.H., Z.L.B., M.A.W., Q.M., Y.H., N.L.W., D.W.S., D.J.R.F.), Medical College of Georgia, Augusta University
| | - Xueyi Li
- Departments of Ophthalmology and Medicine, Stanford University School of Medicine, Palo Alto, CA (X.L.)
| | - Mary Louise Meadows
- Department of Pharmacology and Toxicology (M.L.M., W.H., Y.S., J.Z., S.A.B., D.J.R.F.), Medical College of Georgia, Augusta University
| | - Zach L Brown
- Vascular Biology Center (Z.B., R.K.B., S.H., Z.L.B., M.A.W., Q.M., Y.H., N.L.W., D.W.S., D.J.R.F.), Medical College of Georgia, Augusta University
| | - Madison A West
- Vascular Biology Center (Z.B., R.K.B., S.H., Z.L.B., M.A.W., Q.M., Y.H., N.L.W., D.W.S., D.J.R.F.), Medical College of Georgia, Augusta University
| | - Kunzhe Dong
- Vascular Biology Center (Z.B., R.K.B., S.H., Z.L.B., M.A.W., Q.M., Y.H., N.L.W., D.W.S., D.J.R.F.), Medical College of Georgia, Augusta University
| | - Weihong Han
- Department of Pharmacology and Toxicology (M.L.M., W.H., Y.S., J.Z., S.A.B., D.J.R.F.), Medical College of Georgia, Augusta University
| | - Yunchao Su
- Department of Pharmacology and Toxicology (M.L.M., W.H., Y.S., J.Z., S.A.B., D.J.R.F.), Medical College of Georgia, Augusta University
| | - Qian Ma
- Vascular Biology Center (Z.B., R.K.B., S.H., Z.L.B., M.A.W., Q.M., Y.H., N.L.W., D.W.S., D.J.R.F.), Medical College of Georgia, Augusta University
| | - Yuqing Huo
- Vascular Biology Center (Z.B., R.K.B., S.H., Z.L.B., M.A.W., Q.M., Y.H., N.L.W., D.W.S., D.J.R.F.), Medical College of Georgia, Augusta University
| | - Jiliang Zhou
- Department of Pharmacology and Toxicology (M.L.M., W.H., Y.S., J.Z., S.A.B., D.J.R.F.), Medical College of Georgia, Augusta University
| | - Mahmoud Abdelbary
- School of Medicine, Oregon Health & Science University, Portland (M.A.)
| | - Jennifer C Sullivan
- Immunology Center of Georgia (K.D.), Department of Physiology (J.C.S.), Medical College of Georgia, Augusta University
| | - Neal L Weintraub
- Vascular Biology Center (Z.B., R.K.B., S.H., Z.L.B., M.A.W., Q.M., Y.H., N.L.W., D.W.S., D.J.R.F.), Medical College of Georgia, Augusta University
| | - David W Stepp
- Vascular Biology Center (Z.B., R.K.B., S.H., Z.L.B., M.A.W., Q.M., Y.H., N.L.W., D.W.S., D.J.R.F.), Medical College of Georgia, Augusta University
| | - Feng Chen
- Department of Forensic Medicine, Nanjing Medical University, China (F.C.)
| | - Scott A Barman
- Department of Pharmacology and Toxicology (M.L.M., W.H., Y.S., J.Z., S.A.B., D.J.R.F.), Medical College of Georgia, Augusta University
| | - David J R Fulton
- Vascular Biology Center (Z.B., R.K.B., S.H., Z.L.B., M.A.W., Q.M., Y.H., N.L.W., D.W.S., D.J.R.F.), Medical College of Georgia, Augusta University
- Department of Pharmacology and Toxicology (M.L.M., W.H., Y.S., J.Z., S.A.B., D.J.R.F.), Medical College of Georgia, Augusta University
| |
Collapse
|
16
|
Jacobs S, Payne C, Shaboodien S, Kgatla T, Pretorius A, Jumaar C, Maarman G, Sanni O. Pulmonary hypertension and the potential of 'drug' repurposing: A case for African medicinal plants. Afr J Thorac Crit Care Med 2024; 30:e1352. [PMID: 39171151 PMCID: PMC11334905 DOI: 10.7196/ajtccm.2024.v30i2.1352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 03/25/2024] [Indexed: 08/23/2024] Open
Abstract
Abstract Pulmonary hypertension (PH) is a haemodynamic disorder in which elevated blood pressure in the pulmonary circulation is caused by abnormal vascular tone. Despite advances in treatment, PH mortality remains high, and drug repurposing has been proposed as a mitigating approach. This article reviews the studies that have investigated drug repurposing as a viable option for PH. We provide an overview of PH and highlight pharmaceutical drugs with repurposing potential, based on limited evidence of their mechanisms of action. Moreover, studies have demonstrated the benefits of medicinal plants in PH, most of which are of Indian or Asian origin. Africa is a rich source of many medicinal plants that have been scientifically proven to counteract myriad pathologies. When perusing these studies, one will notice that some African medicinal plants can counteract the molecular pathways (e.g. proliferation, vasoconstriction, inflammation, oxidative stress and mitochondrial dysfunction) that are also involved in the pathogenesis of PH. We review the actions of these plants with actions applicable to PH and highlight that they could be repurposed as adjunct PH therapies. However, these plants have either never been tested in PH, or there is little evidence of their actions against PH. We therefore encourage caution, as more research is needed to study these plants further in experimental models of PH while acknowledging that the outcomes of such proof of-concept studies may not always yield promising findings. Regardless, this article aims to stimulate future research that could make timely contributions to the field. Study synopsis What the study adds. Pulmonary hypertension (PH) remains a fatal disease, and 80% of the patients live in developing countries where resources are scarce and specialised therapies are often unavailable. Drug repurposing is a viable option to try to improve treatment outcomes.Implications of the findings. We propose that another form of 'drug' repurposing is the use of medicinal plants, many of which have demonstrated benefits against pathological processes that are also key in PH, e.g. apoptosis, tumour-like growth of cells, proliferation, oxidative stress and mitochondrial dysfunction.
Collapse
Affiliation(s)
- S Jacobs
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - C Payne
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - S Shaboodien
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - T Kgatla
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - A Pretorius
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - C Jumaar
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - G Maarman
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - O Sanni
- Centre for Cardiometabolic Research in Africa (CARMA), Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
17
|
Kiskin FN, Yang Y, Yang H, Zhang JZ. Cracking the code of the cardiovascular enigma: hPSC-derived endothelial cells unveil the secrets of endothelial dysfunction. J Mol Cell Cardiol 2024; 192:65-78. [PMID: 38761989 DOI: 10.1016/j.yjmcc.2024.05.005] [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: 03/13/2024] [Revised: 05/08/2024] [Accepted: 05/10/2024] [Indexed: 05/20/2024]
Abstract
Endothelial dysfunction is a central contributor to the development of most cardiovascular diseases and is characterised by the reduced synthesis or bioavailability of the vasodilator nitric oxide together with other abnormalities such as inflammation, senescence, and oxidative stress. The use of patient-specific and genome-edited human pluripotent stem cell-derived endothelial cells (hPSC-ECs) has shed novel insights into the role of endothelial dysfunction in cardiovascular diseases with strong genetic components such as genetic cardiomyopathies and pulmonary arterial hypertension. However, their utility in studying complex multifactorial diseases such as atherosclerosis, metabolic syndrome and heart failure poses notable challenges. In this review, we provide an overview of the different methods used to generate and characterise hPSC-ECs before comprehensively assessing their effectiveness in cardiovascular disease modelling and high-throughput drug screening. Furthermore, we explore current obstacles that will need to be overcome to unleash the full potential of hPSC-ECs in facilitating patient-specific precision medicine. Addressing these challenges holds great promise in advancing our understanding of intricate cardiovascular diseases and in tailoring personalised therapeutic strategies.
Collapse
Affiliation(s)
- Fedir N Kiskin
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Yuan Yang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Hao Yang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Joe Z Zhang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| |
Collapse
|
18
|
Schulze KM, Horn AG, Muller-Delp JM, White ZJ, Hall SE, Medarev SL, Weber RE, Poole DC, Musch TI, Behnke BJ. Pulmonary hypertension impairs vasomotor function in rat diaphragm arterioles. Microvasc Res 2024; 154:104686. [PMID: 38614154 PMCID: PMC11198381 DOI: 10.1016/j.mvr.2024.104686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/02/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
Pulmonary hypertension (PH) is a chronic, progressive condition in which respiratory muscle dysfunction is a primary contributor to exercise intolerance and dyspnea in patients. Contractile function, blood flow distribution, and the hyperemic response are altered in the diaphragm with PH, and we sought to determine whether this may be attributed, in part, to impaired vasoreactivity of the resistance vasculature. We hypothesized that there would be blunted endothelium-dependent vasodilation and impaired myogenic responsiveness in arterioles from the diaphragm of PH rats. Female Sprague-Dawley rats were randomized into healthy control (HC, n = 9) and monocrotaline-induced PH rats (MCT, n = 9). Endothelium-dependent and -independent vasodilation and myogenic responses were assessed in first-order arterioles (1As) from the medial costal diaphragm in vitro. There was a significant reduction in endothelium-dependent (via acetylcholine; HC, 78 ± 15% vs. MCT, 47 ± 17%; P < 0.05) and -independent (via sodium nitroprusside; HC, 89 ± 10% vs. MCT, 66 ± 10%; P < 0.05) vasodilation in 1As from MCT rats. MCT-induced PH also diminished myogenic constriction (P < 0.05) but did not alter passive pressure responses. The diaphragmatic weakness, impaired hyperemia, and blood flow redistribution associated with PH may be due, in part, to diaphragm vascular dysfunction and thus compromised oxygen delivery which occurs through both endothelium-dependent and -independent mechanisms.
Collapse
Affiliation(s)
- Kiana M Schulze
- Department of Kinesiology, Kansas State University, Manhattan, KS 66506, United States of America.
| | - Andrew G Horn
- Department of Kinesiology, Kansas State University, Manhattan, KS 66506, United States of America
| | - Judy M Muller-Delp
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, United States of America
| | - Zachary J White
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS 66506, United States of America
| | - Stephanie E Hall
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS 66506, United States of America
| | - Steven L Medarev
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, United States of America
| | - Ramona E Weber
- Department of Kinesiology, Kansas State University, Manhattan, KS 66506, United States of America
| | - David C Poole
- Department of Kinesiology, Kansas State University, Manhattan, KS 66506, United States of America; Department of Anatomy and Physiology, Kansas State University, Manhattan, KS 66506, United States of America
| | - Timothy I Musch
- Department of Kinesiology, Kansas State University, Manhattan, KS 66506, United States of America; Department of Anatomy and Physiology, Kansas State University, Manhattan, KS 66506, United States of America
| | - Bradley J Behnke
- Department of Kinesiology, Kansas State University, Manhattan, KS 66506, United States of America
| |
Collapse
|
19
|
Donovan MK, Abdel-Rahman AA. Ethanol-induced lung and cardiac right ventricular inflammation and remodeling underlie progression to pulmonary arterial hypertension. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:1250-1260. [PMID: 38710650 PMCID: PMC11236493 DOI: 10.1111/acer.15341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 04/05/2024] [Accepted: 04/16/2024] [Indexed: 05/08/2024]
Abstract
BACKGROUND Current research on ethanol-induced cardiovascular anomalies has focused on left ventricular (LV) function and blood pressure. To extend this area of research, we sought to determine whether ethanol-induced alterations in the structure and function of the right cardiac ventricle (RV) and pulmonary artery (PA) lead to pulmonary arterial hypertension (PAH). METHODS Two groups of male Sprague-Dawley rats received a balanced liquid diet containing 5% ethanol (w/v) or a pair-fed isocaloric liquid diet for 8 weeks. Weekly echocardiography was conducted to evaluate cardiopulmonary function, and lung and RV tissues were collected for ex vivo histological and molecular studies. RESULTS The ethanol-treated rats exhibited: (1) Elevated mean pulmonary arterial pressure and decreased pulmonary artery acceleration time/ejection time; (2) Pulmonary vascular remodeling comprising intrapulmonary artery medial layer thickening; and (3) RV hypertrophy along with increased RV/LV + septum, RV diameter, RV cardiomyocyte cross-sectional area, and LV mass/body weight ratio. These responses were associated with increased lung and RV pro-inflammatory markers, endothelin-1 (ET-1), TNF-α, and IL-6 levels and higher ET-1, ET-1 type A/B receptor ratio, and downregulation of the cytoprotective protein, bone morphogenetic protein receptor 2 (BMPR2), in the lungs. CONCLUSION These findings show that moderate ethanol-induced cardiopulmonary changes underlie progression to PAH via an upregulated proinflammatory ET1-TNFα-IL6 pathway and suppression of the anti-inflammatory BMPR2.
Collapse
Affiliation(s)
- Mary Katherine Donovan
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - Abdel A Abdel-Rahman
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| |
Collapse
|
20
|
Heise EL, Salman J, Webs KS, Höffler K, Brandenberger C, Böthig D, Mühlfeld C, Haverich A. Hypoxic perfusion of pulmonary arterial vasa vasorum increases pulmonary arterial pressure. Am J Physiol Lung Cell Mol Physiol 2024; 327:L79-L85. [PMID: 38651234 DOI: 10.1152/ajplung.00346.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/22/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024] Open
Abstract
The pathophysiology of pulmonary hypertension (PH) is not fully understood. Here, we tested the hypothesis that hypoxic perfusion of the vasa vasorum of the pulmonary arterial (PA) wall causes PH. Young adult pig lungs were explanted and placed into a modified ex vivo lung perfusion unit (organ care system, OCS) allowing the separate adjustment of parameters for mechanical ventilation, as well as PA perfusion and bronchial arterial (BA) perfusion. The PA vasa vasorum are branches of the BA. The lungs were used either as the control group (n = 3) or the intervention group (n = 8). The protocol for the intervention group was as follows: normoxic ventilation and perfusion (steady state), hypoxic BA perfusion, steady state, and hypoxic BA perfusion. During hypoxic BA perfusion, ventilation and PA perfusion maintained normal. Control lungs were kept under steady-state conditions for 105 min. During the experiments, PA pressure (PAP) and blood gas analysis were frequently monitored. Hypoxic perfusion of the BA resulted in an increase in systolic and mean PAP, a reaction that was reversible upon normoxic BA perfusion. The PAP increase was reproducible during the second hypoxic BA perfusion. Under control conditions, the PAP stayed constant until about 80 min of the experiment. In conclusion, the results of the current study prove that hypoxic perfusion of the vasa vasorum of the PA directly increases PAP in an ex situ lung perfusion setup, suggesting that PA vasa vasorum function and wall ischemia may contribute to the development of PH.NEW & NOTEWORTHY Hypoxic perfusion of the vasa vasorum of the pulmonary artery directly increased pulmonary arterial pressure in an ex vivo lung perfusion setup. This suggests that the function of pulmonary arterial vasa vasorum and wall ischemia may contribute to the development of pulmonary hypertension.
Collapse
Affiliation(s)
- Emma L Heise
- Hannover Medical School, Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover, Germany
| | - Jawad Salman
- Hannover Medical School, Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Karolin S Webs
- Hannover Medical School, Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Klaus Höffler
- Hannover Medical School, Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover, Germany
| | - Christina Brandenberger
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
- Hannover Medical School, Institute of Functional and Applied Anatomy, Hannover, Germany
- Institute of Functional Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Dietmar Böthig
- Hannover Medical School, Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover, Germany
| | - Christian Mühlfeld
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
- Hannover Medical School, Institute of Functional and Applied Anatomy, Hannover, Germany
| | - Axel Haverich
- Hannover Medical School, Department of Cardiothoracic, Transplant and Vascular Surgery, Hannover, Germany
| |
Collapse
|
21
|
Yang Z, Li F, Thandavarayan RA, Natarajan K, Martin DR, Li Z, Guha A. Early detection of pulmonary arterial hypertension through [ 18F] positron emission tomography imaging with a vascular endothelial receptor small molecule. Pulm Circ 2024; 14:e12393. [PMID: 39072304 PMCID: PMC11273098 DOI: 10.1002/pul2.12393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 05/10/2024] [Accepted: 05/20/2024] [Indexed: 07/30/2024] Open
Abstract
The objective of this study is to provide a positron emission tomography (PET) imaging modality targeting vascular endothelial growth factor receptors (VEGFR) for the early noninvasive detection and assessment of pulmonary arterial hypertension (PAH) severity. To validate the effectiveness of the [18F]VEGFR PET tracer, we utilized a monocrotaline (MCT)-induced PAH rat model. Molecular optical imaging, using a Cy5.5-conjugated VEGFR targeting agent, was employed to demonstrate the uptake of the agent at pulmonary arterioles, correlating with the onset and progression of PAH. Histological examinations of the MCT-PAH rat lung revealed a significant correlation between VEGFR2 expression and the pathogenesis of PAH. Molecular optical imaging demonstrated heightened uptake of the Cy5.5-conjugated VEGFR targeting agent at pulmonary arterioles, corresponding with the onset and progression of PAH. [18F]VEGFR PET showed increased lung uptake detectable in early-stage PAH before increase in pulmonary artery pressures, and this uptake correlated with increased PAH severity. Moreover, when compared to [18F]FDG PET, [18F]VEGFR PET exhibited markedly lower background cardiac signal, enhancing imaging sensitivity for lung abnormalities. Our study provides a compelling evidence for the potential utility of the innovative [18F]VEGFR PET tracer, in non-invasively detecting early signs of PAH, and monitoring its progression. The observed correlations between VEGFR2 expression, molecular optical imaging results, and [18F]VEGFR PET findings support the use of this tracer for early detection, and assessment of PAH severity. The lower background cardiac signal observed with [18F]VEGFR PET further enhances its imaging sensitivity, emphasizing its potential clinical significance.
Collapse
Affiliation(s)
- Zhen Yang
- Department of RadiologyHouston Methodist Academic InstituteHoustonTexasUSA
| | - Feng Li
- Department of RadiologyHouston Methodist Academic InstituteHoustonTexasUSA
| | | | - Kartiga Natarajan
- Department of Cardiovascular SciencesHouston Methodist Research InstituteHoustonTexasUSA
| | - Diego R. Martin
- Department of RadiologyHouston Methodist Academic InstituteHoustonTexasUSA
- Department of RadiologyWeil Cornell MedicineNew YorkNew YorkUSA
| | - Zheng Li
- Department of RadiologyHouston Methodist Academic InstituteHoustonTexasUSA
- Center for Scientific ReviewNIHBethesdaMarylandUSA
| | - Ashrith Guha
- Department of CardiologyHouston Methodist HospitalHoustonTexasUSA
| |
Collapse
|
22
|
Dabral S, Noh M, Werner F, Krebes L, Völker K, Maier C, Aleksic I, Novoyatleva T, Hadzic S, Schermuly RT, Perez VADJ, Kuhn M. C-type natriuretic peptide/cGMP/FoxO3 signaling attenuates hyperproliferation of pericytes from patients with pulmonary arterial hypertension. Commun Biol 2024; 7:693. [PMID: 38844781 PMCID: PMC11156916 DOI: 10.1038/s42003-024-06375-3] [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: 07/27/2023] [Accepted: 05/23/2024] [Indexed: 06/09/2024] Open
Abstract
Pericyte dysfunction, with excessive migration, hyperproliferation, and differentiation into smooth muscle-like cells contributes to vascular remodeling in Pulmonary Arterial Hypertension (PAH). Augmented expression and action of growth factors trigger these pathological changes. Endogenous factors opposing such alterations are barely known. Here, we examine whether and how the endothelial hormone C-type natriuretic peptide (CNP), signaling through the cyclic guanosine monophosphate (cGMP) -producing guanylyl cyclase B (GC-B) receptor, attenuates the pericyte dysfunction observed in PAH. The results demonstrate that CNP/GC-B/cGMP signaling is preserved in lung pericytes from patients with PAH and prevents their growth factor-induced proliferation, migration, and transdifferentiation. The anti-proliferative effect of CNP is mediated by cGMP-dependent protein kinase I and inhibition of the Phosphoinositide 3-kinase (PI3K)/AKT pathway, ultimately leading to the nuclear stabilization and activation of the Forkhead Box O 3 (FoxO3) transcription factor. Augmentation of the CNP/GC-B/cGMP/FoxO3 signaling pathway might be a target for novel therapeutics in the field of PAH.
Collapse
Affiliation(s)
- Swati Dabral
- Institute of Physiology, University of Würzburg, Würzburg, Germany.
| | - Minhee Noh
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Franziska Werner
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Lisa Krebes
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Katharina Völker
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Christopher Maier
- Department of Thoracic and Cardiovascular Surgery, University hospital Würzburg, Würzburg, Germany
| | - Ivan Aleksic
- Department of Thoracic and Cardiovascular Surgery, University hospital Würzburg, Würzburg, Germany
| | - Tatyana Novoyatleva
- Justus-Liebig-University Giessen (JLU), Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Stefan Hadzic
- Justus-Liebig-University Giessen (JLU), Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Ralph Theo Schermuly
- Justus-Liebig-University Giessen (JLU), Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Vinicio A de Jesus Perez
- Divisions of Pulmonary and Critical Care Medicine and Stanford Cardiovascular Institute, Stanford University, California, USA
| | - Michaela Kuhn
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
23
|
Budhram B, Weatherald J, Humbert M. Pulmonary Hypertension in Connective Tissue Diseases Other than Systemic Sclerosis. Semin Respir Crit Care Med 2024; 45:419-434. [PMID: 38499196 DOI: 10.1055/s-0044-1782217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
Pulmonary hypertension (PH) is a known complication of certain connective tissue diseases (CTDs), with systemic sclerosis (SSc) being the most common in the Western world. However, PH in association with non-SSc CTD such as systemic lupus erythematous, mixed connective tissue disease, and primary Sjögren's syndrome constitutes a distinct subset of patients with inherently different epidemiologic profiles, pathophysiologic mechanisms, clinical features, therapeutic options, and prognostic implications. The purpose of this review is to inform a practical approach for clinicians evaluating patients with non-SSc CTD-associated PH.The development of PH in these patients involves a complex interplay between genetic factors, immune-mediated mechanisms, and endothelial cell dysfunction. Furthermore, the broad spectrum of CTD manifestations can contribute to the development of PH through various pathophysiologic mechanisms, including intrinsic pulmonary arteriolar vasculopathy (pulmonary arterial hypertension, Group 1 PH), left-heart disease (Group 2), chronic lung disease (Group 3), chronic pulmonary artery obstruction (Group 4), and unclear and/or multifactorial mechanisms (Group 5). The importance of diagnosing PH early in symptomatic patients with non-SSc CTD is highlighted, with a review of the relevant biomarkers, imaging, and diagnostic procedures required to establish a diagnosis.Therapeutic strategies for non-SSc PH associated with CTD are explored with an in-depth review of the medical, interventional, and surgical options available to these patients, emphasizing the CTD-specific considerations that guide treatment and aid in prognosis. By identifying gaps in the current literature, we offer insights into future research priorities that may prove valuable for patients with PH associated with non-SSc CTD.
Collapse
Affiliation(s)
- Brandon Budhram
- Division of Respirology, Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jason Weatherald
- Division of Pulmonary Medicine, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Marc Humbert
- Université Paris-Saclay, Inserm UMR_S 999, Service de Pneumologie et Soins Intensifs Respiratoires, European Reference Network for Rare Respiratory Diseases (ERN-LUNG), Hôpital Bicêtre (Assistance Publique Hôpitaux de Paris), Le Kremlin-Bicêtre, France
| |
Collapse
|
24
|
Arif W, Bhimani RK, Ali Shah M, Tausif Z, Nisar U, Kumar R, Bhimani PD, Shoaibullah S, Naveed MA, Raja A, Raja S, Deepak F, Shafique MA, Mustafa MS. Unraveling disparities: Probing gender, race, and geographic inequities in pulmonary heart disease mortality in the United States: An extensive longitudinal examination (1999-2020) leveraging CDC WONDER data. Curr Probl Cardiol 2024; 49:102527. [PMID: 38492618 DOI: 10.1016/j.cpcardiol.2024.102527] [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: 03/13/2024] [Accepted: 03/13/2024] [Indexed: 03/18/2024]
Abstract
This comprehensive study delves into the epidemiological landscape of Pulmonary Heart Disease (PHD) mortality in the United States from 1999 to 2020, leveraging the extensive CDC WONDER database. PHD encompasses conditions affecting the right side of the heart due to lung disorders or elevated pressure in the pulmonary arteries, including pulmonary hypertension, pulmonary embolism, and chronic thromboembolic pulmonary hypertension (CTEPH). Analyzing data from death certificates, demographic characteristics, and geographical segmentation, significant trends emerge. The age-adjusted mortality rates (AAMRs) for PHD-related deaths show a fluctuating pattern, initially decreasing from 1999 to 2006, followed by a steady increase until 2020. Male patients consistently exhibit higher AAMRs than females, with notable disparities observed among racial/ethnic groups and geographic regions. Non-hispanic (NH) Black or African American individuals, residents of specific states like Colorado and the District of Columbia, and those in the Midwest region demonstrate elevated AAMRs. Furthermore, nonmetropolitan areas consistently manifest higher AAMRs than metropolitan areas. These findings underscore the urgent need for intensified prevention and treatment strategies to address the rising mortality associated with PHD, particularly among vulnerable populations. Insights from this study offer valuable guidance for public health initiatives aimed at reducing PHD-related mortality and improving outcomes nationwide.
Collapse
Affiliation(s)
- Waqar Arif
- Dow International Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | | | - Mohsin Ali Shah
- Shaheed Mohtarma Benazir Bhutto Medical College Lyari, Karachi, Pakistan
| | | | - Umer Nisar
- Dow International Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | - Rohet Kumar
- Shaheed Mohtarma Benazir Bhutto Medical College Lyari, Karachi, Pakistan
| | | | - Syed Shoaibullah
- Dow International Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | | | - Adarsh Raja
- Shaheed Mohtarma Benazir Bhutto Medical College Lyari, Karachi, Pakistan.
| | - Sandesh Raja
- Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | - Fnu Deepak
- Shaheed Mohtarma Benazir Bhutto Medical College Lyari, Karachi, Pakistan
| | | | | |
Collapse
|
25
|
Weinstein N, Carlsen J, Schulz S, Stapleton T, Henriksen HH, Travnik E, Johansson PI. A Lifelike guided journey through the pathophysiology of pulmonary hypertension-from measured metabolites to the mechanism of action of drugs. Front Cardiovasc Med 2024; 11:1341145. [PMID: 38845688 PMCID: PMC11153715 DOI: 10.3389/fcvm.2024.1341145] [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: 11/19/2023] [Accepted: 04/12/2024] [Indexed: 06/09/2024] Open
Abstract
Introduction Pulmonary hypertension (PH) is a pathological condition that affects approximately 1% of the population. The prognosis for many patients is poor, even after treatment. Our knowledge about the pathophysiological mechanisms that cause or are involved in the progression of PH is incomplete. Additionally, the mechanism of action of many drugs used to treat pulmonary hypertension, including sotatercept, requires elucidation. Methods Using our graph-powered knowledge mining software Lifelike in combination with a very small patient metabolite data set, we demonstrate how we derive detailed mechanistic hypotheses on the mechanisms of PH pathophysiology and clinical drugs. Results In PH patients, the concentration of hypoxanthine, 12(S)-HETE, glutamic acid, and sphingosine 1 phosphate is significantly higher, while the concentration of L-arginine and L-histidine is lower than in healthy controls. Using the graph-based data analysis, gene ontology, and semantic association capabilities of Lifelike, led us to connect the differentially expressed metabolites with G-protein signaling and SRC. Then, we associated SRC with IL6 signaling. Subsequently, we found associations that connect SRC, and IL6 to activin and BMP signaling. Lastly, we analyzed the mechanisms of action of several existing and novel pharmacological treatments for PH. Lifelike elucidated the interplay between G-protein, IL6, activin, and BMP signaling. Those pathways regulate hallmark pathophysiological processes of PH, including vasoconstriction, endothelial barrier function, cell proliferation, and apoptosis. Discussion The results highlight the importance of SRC, ERK1, AKT, and MLC activity in PH. The molecular pathways affected by existing and novel treatments for PH also converge on these molecules. Importantly, sotatercept affects SRC, ERK1, AKT, and MLC simultaneously. The present study shows the power of mining knowledge graphs using Lifelike's diverse set of data analytics functionalities for developing knowledge-driven hypotheses on PH pathophysiological and drug mechanisms and their interactions. We believe that Lifelike and our presented approach will be valuable for future mechanistic studies of PH, other diseases, and drugs.
Collapse
Affiliation(s)
- Nathan Weinstein
- CAG Center for Endotheliomics, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Jørn Carlsen
- CAG Center for Endotheliomics, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Sebastian Schulz
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Timothy Stapleton
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Hanne H. Henriksen
- CAG Center for Endotheliomics, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Evelyn Travnik
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Pär Ingemar Johansson
- CAG Center for Endotheliomics, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
26
|
Awad KS, Wang S, Dougherty EJ, Keshavarz A, Demirkale CY, Yu ZX, Miller L, Elinoff JM, Danner RL. BMPR2 Loss Activates AKT by Disrupting DLL4/NOTCH1 and PPARγ Signaling in Pulmonary Arterial Hypertension. Int J Mol Sci 2024; 25:5403. [PMID: 38791441 PMCID: PMC11121464 DOI: 10.3390/ijms25105403] [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: 04/16/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive cardiopulmonary disease characterized by pathologic vascular remodeling of small pulmonary arteries. Endothelial dysfunction in advanced PAH is associated with proliferation, apoptosis resistance, and endothelial to mesenchymal transition (EndoMT) due to aberrant signaling. DLL4, a cell membrane associated NOTCH ligand, plays a pivotal role maintaining vascular integrity. Inhibition of DLL4 has been associated with the development of pulmonary hypertension, but the mechanism is incompletely understood. Here we report that BMPR2 silencing in pulmonary artery endothelial cells (PAECs) activated AKT and suppressed the expression of DLL4. Consistent with these in vitro findings, increased AKT activation and reduced DLL4 expression was found in the small pulmonary arteries of patients with PAH. Increased NOTCH1 activation through exogenous DLL4 blocked AKT activation, decreased proliferation and reversed EndoMT. Exogenous and overexpression of DLL4 induced BMPR2 and PPRE promoter activity, and BMPR2 and PPARG mRNA in idiopathic PAH (IPAH) ECs. PPARγ, a nuclear receptor associated with EC homeostasis, suppressed by BMPR2 loss was induced and activated by DLL4/NOTCH1 signaling in both BMPR2-silenced and IPAH ECs, reversing aberrant phenotypic changes, in part through AKT inhibition. Directly blocking AKT or restoring DLL4/NOTCH1/PPARγ signaling may be beneficial in preventing or reversing the pathologic vascular remodeling of PAH.
Collapse
MESH Headings
- Humans
- Proto-Oncogene Proteins c-akt/metabolism
- Signal Transduction
- Bone Morphogenetic Protein Receptors, Type II/metabolism
- Bone Morphogenetic Protein Receptors, Type II/genetics
- PPAR gamma/metabolism
- PPAR gamma/genetics
- Receptor, Notch1/metabolism
- Receptor, Notch1/genetics
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Endothelial Cells/metabolism
- Adaptor Proteins, Signal Transducing/metabolism
- Adaptor Proteins, Signal Transducing/genetics
- Calcium-Binding Proteins/metabolism
- Calcium-Binding Proteins/genetics
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/genetics
- Pulmonary Arterial Hypertension/pathology
- Male
- Cell Proliferation
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/pathology
- Female
- Cells, Cultured
Collapse
Affiliation(s)
- Keytam S. Awad
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD 20892, USA; (S.W.); (E.J.D.); (A.K.); (C.Y.D.); (L.M.); (R.L.D.)
| | - Shuibang Wang
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD 20892, USA; (S.W.); (E.J.D.); (A.K.); (C.Y.D.); (L.M.); (R.L.D.)
| | - Edward J. Dougherty
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD 20892, USA; (S.W.); (E.J.D.); (A.K.); (C.Y.D.); (L.M.); (R.L.D.)
| | - Ali Keshavarz
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD 20892, USA; (S.W.); (E.J.D.); (A.K.); (C.Y.D.); (L.M.); (R.L.D.)
| | - Cumhur Y. Demirkale
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD 20892, USA; (S.W.); (E.J.D.); (A.K.); (C.Y.D.); (L.M.); (R.L.D.)
| | - Zu Xi Yu
- Critical Care Medicine and Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA; (Z.X.Y.); (J.M.E.)
| | - Latonia Miller
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD 20892, USA; (S.W.); (E.J.D.); (A.K.); (C.Y.D.); (L.M.); (R.L.D.)
| | - Jason M. Elinoff
- Critical Care Medicine and Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA; (Z.X.Y.); (J.M.E.)
| | - Robert L. Danner
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD 20892, USA; (S.W.); (E.J.D.); (A.K.); (C.Y.D.); (L.M.); (R.L.D.)
- Critical Care Medicine and Pulmonary Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA; (Z.X.Y.); (J.M.E.)
| |
Collapse
|
27
|
Kazemi N, Bordbar A, Bavarsad SS, Ghasemi P, Bakhshi M, Rezaeeyan H. Molecular Insights into the Relationship Between Platelet Activation and Endothelial Dysfunction: Molecular Approaches and Clinical Practice. Mol Biotechnol 2024; 66:932-947. [PMID: 38184492 DOI: 10.1007/s12033-023-01010-8] [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: 08/27/2023] [Accepted: 11/27/2023] [Indexed: 01/08/2024]
Abstract
Platelets are one of the coagulation cells. When platelet activation occurs, many mediators are released and affect endothelial cells (ECs) and lead to endothelial dysfunction (ED). ED plays an important role in the pathogenesis of many diseases, including cardiovascular disease (CVD). Platelet are of important factors in ED. The release of mediators by platelets causes the stimulation of inflammatory pathways, oxidative stress, and apoptosis, which ultimately result in ED.On the other hand, platelet activation in CVD patients can be associated with a bad prognosis. Platelet activation can increase the level of markers such as p-selectin in the serum. Also, in this study, we have discussed the role of platelet as a diagnostic factor, as well as its use as a treatment option. In addition, we discussed some of the molecular pathways that are used to target platelet activation.
Collapse
Affiliation(s)
- Niloufar Kazemi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization (IBTO), Tehran, Iran
| | - Armin Bordbar
- Department of Cardiology, Musavi Hospital, School of Medicine, Zanjan University of Medical Science, Zanjan, Iran
| | | | - Parisa Ghasemi
- Research Committee, Medical School, Arak University of Medical Sciences, Arak, Iran
| | - Maryam Bakhshi
- Islamic Azad University of Najaf Abad, Affiliated Hospitals, Isfahan, Iran
| | - Hadi Rezaeeyan
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization (IBTO), Tehran, Iran.
| |
Collapse
|
28
|
Correale M, Chirivì F, Bevere EML, Tricarico L, D’Alto M, Badagliacca R, Brunetti ND, Vizza CD, Ghio S. Endothelial Function in Pulmonary Arterial Hypertension: From Bench to Bedside. J Clin Med 2024; 13:2444. [PMID: 38673717 PMCID: PMC11051060 DOI: 10.3390/jcm13082444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/09/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
Pulmonary arterial hypertension is a complex pathology whose etiology is still not completely well clarified. The pathogenesis of pulmonary arterial hypertension involves different molecular mechanisms, with endothelial dysfunction playing a central role in disease progression. Both individual genetic predispositions and environmental factors seem to contribute to its onset. To further understand the complex relationship between endothelial and pulmonary hypertension and try to contribute to the development of future therapies, we report a comprehensive and updated review on endothelial function in pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Michele Correale
- Cardiothoracic Department, Policlinico Riuniti University Hospital, 71100 Foggia, Italy;
| | - Francesco Chirivì
- Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy; (F.C.); (E.M.L.B.); (N.D.B.)
| | - Ester Maria Lucia Bevere
- Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy; (F.C.); (E.M.L.B.); (N.D.B.)
| | - Lucia Tricarico
- Cardiothoracic Department, Policlinico Riuniti University Hospital, 71100 Foggia, Italy;
| | - Michele D’Alto
- Department of Cardiology, A.O.R.N. dei Colli, Monaldi Hospital, University of Campania L. ‘Vanvitelli’, 80133 Naples, Italy;
| | - Roberto Badagliacca
- Department of Clinical, Anesthesiological and Cardiovascular Sciences, I School of Medicine, Sapienza University of Rome, 00185 Rome, Italy; (R.B.); (C.D.V.)
| | - Natale D. Brunetti
- Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy; (F.C.); (E.M.L.B.); (N.D.B.)
| | - Carmine Dario Vizza
- Department of Clinical, Anesthesiological and Cardiovascular Sciences, I School of Medicine, Sapienza University of Rome, 00185 Rome, Italy; (R.B.); (C.D.V.)
| | - Stefano Ghio
- Division of Cardiology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| |
Collapse
|
29
|
Liu W, Xiu L, Zhou M, Li T, Jiang N, Wan Y, Qiu C, Li J, Hu W, Zhang W, Wu J. The Critical Role of the Shroom Family Proteins in Morphogenesis, Organogenesis and Disease. PHENOMICS (CHAM, SWITZERLAND) 2024; 4:187-202. [PMID: 38884059 PMCID: PMC11169129 DOI: 10.1007/s43657-023-00119-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 06/18/2024]
Abstract
The Shroom (Shrm) family of actin-binding proteins has a unique and highly conserved Apx/Shrm Domain 2 (ASD2) motif. Shroom protein directs the subcellular localization of Rho-associated kinase (ROCK), which remodels the actomyosin cytoskeleton and changes cellular morphology via its ability to phosphorylate and activate non-muscle myosin II. Therefore, the Shrm-ROCK complex is critical for the cellular shape and the development of many tissues, including the neural tube, eye, intestines, heart, and vasculature system. Importantly, the structure and expression of Shrm proteins are also associated with neural tube defects, chronic kidney disease, metastasis of carcinoma, and X-link mental retardation. Therefore, a better understanding of Shrm-mediated signaling transduction pathways is essential for the development of new therapeutic strategies to minimize damage resulting in abnormal Shrm proteins. This paper provides a comprehensive overview of the various Shrm proteins and their roles in morphogenesis and disease.
Collapse
Affiliation(s)
- Wanling Liu
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, 200438 China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Lei Xiu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Mingzhe Zhou
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, 200438 China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Tao Li
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, 200438 China
| | - Ning Jiang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Yanmin Wan
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, 200438 China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Chao Qiu
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, 200438 China
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Jian Li
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, 200438 China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200438 China
| | - Wei Hu
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, 200438 China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200438 China
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Monglia University, Hohhot, 010030 China
| | - Wenhong Zhang
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, 200438 China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200438 China
- Shanghai Huashen Institute of Microbes and Infections, Shanghai, 200052 China
| | - Jing Wu
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, 200438 China
- Shanghai Huashen Institute of Microbes and Infections, Shanghai, 200052 China
| |
Collapse
|
30
|
Dignam JP, Sharma S, Stasinopoulos I, MacLean MR. Pulmonary arterial hypertension: Sex matters. Br J Pharmacol 2024; 181:938-966. [PMID: 37939796 DOI: 10.1111/bph.16277] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 11/10/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a complex disease of multifactorial origin. While registries have demonstrated that women are more susceptible to the disease, females with PAH have superior right ventricle (RV) function and a better prognosis than their male counterparts, a phenomenon referred to as the 'estrogen paradox'. Numerous pre-clinical studies have investigated the involvement of sex hormones in PAH pathobiology, often with conflicting results. However, recent advances suggest that abnormal estrogen synthesis, metabolism and signalling underpin the sexual dimorphism of this disease. Other sex hormones, such as progesterone, testosterone and dehydroepiandrosterone may also play a role. Several non-hormonal factor including sex chromosomes and epigenetics have also been implicated. Though the underlying pathophysiological mechanisms are complex, several compounds that modulate sex hormones levels and signalling are under investigation in PAH patients. Further elucidation of the estrogen paradox will set the stage for the identification of additional therapeutic targets for this disease.
Collapse
Affiliation(s)
- Joshua P Dignam
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Smriti Sharma
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Ioannis Stasinopoulos
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, UK
| | - Margaret R MacLean
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| |
Collapse
|
31
|
Novara ME, Di Martino E, Stephens B, Nayrouz M, Vitulo P, Carollo A, Provenzani A. Future Perspectives of Pulmonary Arterial Hypertension: A Review of Novel Pipeline Treatments and Indications. Drugs R D 2024; 24:13-28. [PMID: 38514585 PMCID: PMC11035521 DOI: 10.1007/s40268-024-00453-x] [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] [Accepted: 01/07/2024] [Indexed: 03/23/2024] Open
Abstract
Pulmonary arterial hypertension is characterized by elevated blood pressure and pathological changes in the pulmonary arterioles, leading to the development of right-heart failure and potentially fatal outcomes if left untreated. This review aims to provide an overview of novel drugs or formulations and new drug indications for pulmonary arterial hypertension that are currently in phases II-III of randomized controlled trials, and describe the rationale for the use of these targeted therapies, as well as their efficacy, safety profile, and impact on quality of life and survival. The literature research was conducted using data from ClinicalTrials.gov for the period between 1 January 2016 up to 31 December 2022. The population of interest includes individuals aged ≥ 18 years who have been diagnosed with pulmonary arterial hypertension. The review selection criteria included trials with recruiting, enrolling by invitation, active, terminated or completed status in 2022 and 2023. A total of 24 studies were selected for evaluation based on the inclusion and exclusion criteria. This review summarizes the updated information from randomized clinical trials involving novel therapies for pulmonary arterial hypertension. However, larger clinical trials are required to validate their clinical safety and effects. In the future, clinicians should choose therapies based on the patient's individual situation and requirements when developing treatment strategies.
Collapse
Affiliation(s)
- Maria Eugenia Novara
- Clinical Pharmacy Service, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (ISMETT), Palermo, Italy
| | - Enrica Di Martino
- Clinical Pharmacy Service, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (ISMETT), Palermo, Italy
| | - Brandon Stephens
- School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Mary Nayrouz
- School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Patrizio Vitulo
- Pneumology Unit, Department for the Treatment and Study of Cardiothoracic Diseases and Cardiothoracic Transplantation, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (ISMETT), Palermo, Italy
| | - Anna Carollo
- Clinical Pharmacy Service, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (ISMETT), Palermo, Italy
| | - Alessio Provenzani
- Clinical Pharmacy Service, Mediterranean Institute for Transplantation and Advanced Specialized Therapies (ISMETT), Palermo, Italy.
| |
Collapse
|
32
|
Park JM, Seo YS, Kim SH, Kim HY, Kim MS, Lee MY. Impact of inhalation exposure to cigarette smoke on the pathogenesis of pulmonary hypertension primed by monocrotaline in rats. J Appl Toxicol 2024; 44:470-483. [PMID: 37876240 DOI: 10.1002/jat.4555] [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: 07/03/2023] [Revised: 09/26/2023] [Accepted: 09/26/2023] [Indexed: 10/26/2023]
Abstract
Extensive, long-term exposure to cigarette smoke (CS) was recently suggested to be a risk factor for pulmonary hypertension, although further validation is required. The vascular effects of CS share similarities with the etiology of pulmonary hypertension, including vascular inflammation and remodeling. Thus, we examined the influence of CS exposure on the pathogenesis of monocrotaline (MCT)-induced pulmonary hypertension, hypothesizing that smoking might accelerate the development of primed pulmonary hypertension. CS was generated from 3R4F reference cigarettes, and rats were exposed to CS by inhalation at total particulate matter concentrations of 100-300 μg/L for 4 h/day, 7 days/week for 4 weeks. Following 1 week of initial exposure, rats received 60 mg/kg MCT and were sacrificed and analyzed after an additional 3 weeks of exposure. MCT induced hypertrophy in pulmonary arterioles and increased the Fulton index, a measure of right ventricular hypertrophy. Additional CS exposure exacerbated arteriolar hypertrophy but did not further elevate the Fulton index. No significant alterations were observed in levels of endothelin-1 and vascular endothelial growth factor, or in hematological and serum biochemical parameters. Short-term inhalation exposure to CS exacerbated arteriolar hypertrophy in the lung, although this effect did not directly aggravate the overworked heart under the current experimental conditions.
Collapse
Affiliation(s)
- Jung-Min Park
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Yoon-Seok Seo
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Sung-Hwan Kim
- Inhalation Toxicology Research Group, Korea Institute of Toxicology, Jeongeup-si, Jeollabuk-do, Republic of Korea
| | - Hyeon-Young Kim
- Inhalation Toxicology Research Group, Korea Institute of Toxicology, Jeongeup-si, Jeollabuk-do, Republic of Korea
| | - Min-Seok Kim
- Inhalation Toxicology Research Group, Korea Institute of Toxicology, Jeongeup-si, Jeollabuk-do, Republic of Korea
| | - Moo-Yeol Lee
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Goyang-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
33
|
Dang ZC, Yang Z, Liu S, Du GM, Jin L, Zhao ZZ. Efficacy of Sildenafil on healthy humans in high‑altitude hypoxia at rest and during exercise: A meta‑analysis. Exp Ther Med 2024; 27:88. [PMID: 38274336 PMCID: PMC10809317 DOI: 10.3892/etm.2024.12376] [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: 05/30/2023] [Accepted: 10/23/2023] [Indexed: 01/27/2024] Open
Abstract
The current meta-analysis aimed to fully evaluate the efficacy of Sildenafil in healthy humans at different altitudes, focusing on echocardiographic and hemodynamic parameters. Relevant studies were retrieved from the Cochrane, Embase and PubMed databases. Odds ratios (OR) were determined for dichotomous data and weighted mean differences with 95% confidence intervals (CIs) for continuous data. A total of 16 RCTs were included in the current meta-analysis. Short-term treatment with Sildenafil significantly elevated resting heart rate (P<0.01) at altitudes <4,000 meters. No significant differences in heart rate were observed between the Sildenafil and placebo groups at rest and during exercise at an altitude of >4,000 meters (P>0.05). Sildenafil improved resting cardiac output at an altitude of >5,000 meters (P<0.01) and exercising arterial oxygen saturation at <4,000 meters (P<0.01). Sildenafil reduced resting pulmonary artery systolic pressure (PASP) at altitudes >4,000 meters (P<0.01) and exercising PASP at altitudes >5,000 meters (P<0.01). Therefore, Sildenafil efficacy in healthy humans with high-altitude hypoxia is related to altitude and rest or exercise.
Collapse
Affiliation(s)
- Zhan-Cui Dang
- Department of Public Health, Medical College, Qinghai University, Xining, Qinghai 810000, P.R. China
| | - Zhiquan Yang
- Department of Rehabilitation, Women and Children's Hospital of Qinghai Province, Xining, Qinghai 810000, P.R. China
| | - Shou Liu
- Department of Public Health, Medical College, Qinghai University, Xining, Qinghai 810000, P.R. China
| | - Guo-Mei Du
- Department of Physical Examination, Qinghai Red Cross Hospital, Xining, Qinghai 810000, P.R. China
| | - Linde Jin
- Department of Cardiology, Qinghai Provincial People's Hospital, Xining, Qinghai 810000, P.R. China
| | - Zhong-Zhi Zhao
- Department of Endemic Disease Control, Qinghai Provincial Institute for Endemic Disease Prevention and Control, Xining, Qinghai 811602, P.R. China
| |
Collapse
|
34
|
Pawar SG, Khan N, Salam A, Joshi M, Saravanan PB, Pandey S. The association of Pulmonary Hypertension and right ventricular systolic function - updates in diagnosis and treatment. Dis Mon 2024; 70:101635. [PMID: 37734967 DOI: 10.1016/j.disamonth.2023.101635] [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] [Indexed: 09/23/2023]
Abstract
Right ventricular (RV) systolic function is an essential but neglected component in cardiac evaluation, and its importance to the contribution to overall cardiac function is undermined. It is not only sensitive to the effect of left heart valve disease but is also more sensitive to changes in pressure overload than the left ventricle. Pulmonary Hypertension is the common and well-recognized complication of RV systolic dysfunction. It is also the leading cause of pulmonary valve disease and right ventricular dysfunction. Patients with a high pulmonary artery pressure (PAP) and a low RV ejection fraction have a seven-fold higher risk of death than heart failure patients with a normal PAP and RV ejection fraction. Furthermore, it is an independent predictor of survival in these patients. In this review, we examine the association of right ventricular systolic function with Pulmonary Hypertension by focusing on various pathological and clinical manifestations while assessing their impact. We also explore new 2022 ESC/ERS guidelines for diagnosing and treating right ventricular dysfunction in Pulmonary Hypertension.
Collapse
Affiliation(s)
| | - Nida Khan
- Jinnah Sindh Medical University, Pakistan
| | - Ajal Salam
- Government Medical College Kottayam, Kottayam, Kerala, India
| | - Muskan Joshi
- Tbilisi State Medical University, Tbilisi, Georgia
| | | | | |
Collapse
|
35
|
Santamans AM, Cicuéndez B, Mora A, Villalba-Orero M, Rajlic S, Crespo M, Vo P, Jerome M, Macías Á, López JA, Leiva M, Rocha SF, León M, Rodríguez E, Leiva L, Pintor Chocano A, García Lunar I, García-Álvarez A, Hernansanz-Agustín P, Peinado VI, Barberá JA, Ibañez B, Vázquez J, Spinelli JB, Daiber A, Oliver E, Sabio G. MCJ: A mitochondrial target for cardiac intervention in pulmonary hypertension. SCIENCE ADVANCES 2024; 10:eadk6524. [PMID: 38241373 PMCID: PMC10798563 DOI: 10.1126/sciadv.adk6524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 12/19/2023] [Indexed: 01/21/2024]
Abstract
Pulmonary hypertension (PH) can affect both pulmonary arterial tree and cardiac function, often leading to right heart failure and death. Despite the urgency, the lack of understanding has limited the development of effective cardiac therapeutic strategies. Our research reveals that MCJ modulates mitochondrial response to chronic hypoxia. MCJ levels elevate under hypoxic conditions, as in lungs of patients affected by COPD, mice exposed to hypoxia, and myocardium from pigs subjected to right ventricular (RV) overload. The absence of MCJ preserves RV function, safeguarding against both cardiac and lung remodeling induced by chronic hypoxia. Cardiac-specific silencing is enough to protect against cardiac dysfunction despite the adverse pulmonary remodeling. Mechanistically, the absence of MCJ triggers a protective preconditioning state mediated by the ROS/mTOR/HIF-1α axis. As a result, it preserves RV systolic function following hypoxia exposure. These discoveries provide a potential avenue to alleviate chronic hypoxia-induced PH, highlighting MCJ as a promising target against this condition.
Collapse
Affiliation(s)
- Ayelén M. Santamans
- Cardiovascular Risk Factors and Brain Function Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Beatriz Cicuéndez
- Cardiovascular Risk Factors and Brain Function Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Alfonso Mora
- Cardiovascular Risk Factors and Brain Function Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Molecular Oncology Programme, Organ crosstalk in metabolic diseases groupOrgan crosstalk in metabolic diseases group, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - María Villalba-Orero
- Cardiovascular Risk Factors and Brain Function Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Departamento de Medicina y Cirugía Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Sanela Rajlic
- Department of Cardiothoracic and Vascular Surgery, University of Medicine Mainz, 55131 Mainz, Germany
- Department of Cardiology, Department of Cardiology, Molecular Cardiology, University Medical Center, 55131 Mainz, Germany
| | - María Crespo
- Cardiovascular Risk Factors and Brain Function Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Paula Vo
- Program in Molecular Medicine, UMass Chan Medical School, Worcester MA 01605
| | - Madison Jerome
- Program in Molecular Medicine, UMass Chan Medical School, Worcester MA 01605
| | - Álvaro Macías
- Cardiovascular Risk Factors and Brain Function Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Juan Antonio López
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Novel mechanisms of Atherocleroclerosis Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Magdalena Leiva
- Department of Immunology, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Susana F. Rocha
- Cardiovascular Risk Factors and Brain Function Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Marta León
- Cardiovascular Risk Factors and Brain Function Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Elena Rodríguez
- Cardiovascular Risk Factors and Brain Function Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Molecular Oncology Programme, Organ crosstalk in metabolic diseases groupOrgan crosstalk in metabolic diseases group, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Luis Leiva
- Cardiovascular Risk Factors and Brain Function Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Molecular Oncology Programme, Organ crosstalk in metabolic diseases groupOrgan crosstalk in metabolic diseases group, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Aránzazu Pintor Chocano
- Cardiovascular Risk Factors and Brain Function Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Inés García Lunar
- Cardiovascular Risk Factors and Brain Function Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Cardiology Department, University Hospital La Moraleja, Madrid, Spain
| | - Ana García-Álvarez
- Cardiovascular Risk Factors and Brain Function Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Cardiology Department, Hospital Clínic Barcelona-IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Pablo Hernansanz-Agustín
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Víctor I. Peinado
- Department of Experimental Pathology, Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC-IDIBAPS), Barcelona, Spain
- Department of Pulmonary Medicine, Hospital Clínic, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| | - Joan Albert Barberá
- Department of Pulmonary Medicine, Hospital Clínic, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| | - Borja Ibañez
- Cardiovascular Risk Factors and Brain Function Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Cardiology Department, IIS-Fundación Jiménez Díaz Hospital, Madrid, Spain
| | - Jesús Vázquez
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Novel mechanisms of Atherocleroclerosis Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Jessica B. Spinelli
- Program in Molecular Medicine, UMass Chan Medical School, Worcester MA 01605
- UMass Chan Medical School Cancer Center, Worcester MA 01605
| | - Andreas Daiber
- Department of Cardiothoracic and Vascular Surgery, University of Medicine Mainz, 55131 Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany
| | - Eduardo Oliver
- Cardiovascular Risk Factors and Brain Function Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Centro de Investigaciones biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| | - Guadalupe Sabio
- Cardiovascular Risk Factors and Brain Function Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Molecular Oncology Programme, Organ crosstalk in metabolic diseases groupOrgan crosstalk in metabolic diseases group, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| |
Collapse
|
36
|
Satawiriya M, Khongphatthanayothin A, Limsuwan A. Reversible severe pulmonary hypertension related to scurvy in children. BMC Cardiovasc Disord 2024; 24:24. [PMID: 38172747 PMCID: PMC10765653 DOI: 10.1186/s12872-023-03629-6] [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: 08/26/2023] [Accepted: 11/22/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Severe pulmonary hypertension (PH) in childhood is rare and can manifest as a life-threatening episode. We present 2 children with restrictive dietary habits with severe pulmonary hypertension secondary to scurvy and iron deficiency anemia with treatment and outcome. CASE PRESENTATION The first case is a 2-year-old boy who presented with vomiting, diarrhea, and fever. After rehydration, he had recurrent episodes of hypotension with intermittent abdominal pain. Fluid resuscitation and inotropic medication were given. Then he suddenly collapsed. After 4-min cardiopulmonary resuscitation, his hemodynamic was stabilized. Most of the medical workup was unremarkable except for PH from the echocardiogram with estimated systolic pulmonary artery pressure (PAP) at 67 mmHg. Transient PH was diagnosed, and milrinone was prescribed. Since he had restrictive dietary habits and sclerotic rim at epiphysis in chest films, his vitamin C level was tested and reported low-level result. The second case is a 6-year-old boy with acute dyspnea, a month of low-grade fever, mild cyanosis, and a swollen left knee. Echocardiogram indicated moderate TR with estimated systolic PAP at 56 mmHg (systolic blood pressure 90 mmHg). Milrinone was given. Right cardiac catheterization showed PAP 66/38 (mean 50) mmHg and PVRi 5.7 WU.m2. Other medical conditions causing PH were excluded. With a history of improper dietary intake and clinical suspicion of scurvy, vitamin C was tested and reported undetectable level. Administration of vitamin C in both cases rapidly reversed pulmonary hypertension. CONCLUSION Pediatric PH related to vitamin C deficiency can manifest with a wide range of symptoms, varying from mild and nonspecific to severe life-threatening episodes characterized by pulmonary hypertensive crises. PH associated with scurvy is entirely reversible with appropriate investigation, diagnosis, and treatment. Our report highlights the importance of considering nutritional deficiencies as potential confounding factors in pediatric PH, emphasizing the need for comprehensive evaluation and management of these patients.
Collapse
Affiliation(s)
- Marin Satawiriya
- Division of Pediatric Cardiology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama 6 Rd, Rachathewi, Bangkok, 10400, Thailand
| | - Apichai Khongphatthanayothin
- Bangkok Heart Hospital, Bangkok, Thailand
- Center of Excellence in Arrhythmia Research Chulalongkorn University, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Alisa Limsuwan
- Division of Pediatric Cardiology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama 6 Rd, Rachathewi, Bangkok, 10400, Thailand.
| |
Collapse
|
37
|
Liu X, Li X, Duan J, Zhang R, Zhang H, Wang W, Shi B, Zhou H, Li G. The percentage of circulating fibrocytes is associated with increased morbidity of pulmonary hypertension in patients on hemodialysis. Semin Dial 2024; 37:43-51. [PMID: 36693653 DOI: 10.1111/sdi.13139] [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: 10/26/2022] [Accepted: 01/07/2023] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Pulmonary hypertension (PH) is highly prevalent in patients receiving dialysis. The precise mechanisms underlying PH in hemodialysis (HD) patients have not been adequately addressed. Emerging experimental evidence indicates that circulating fibrocytes may contribute significantly to this process. METHODS We measured the proportion of circulating fibrocytes using flow cytometry analysis and prospectively analyzed patients during HD from February 1, 2017, to February 1, 2022. Then we investigated correlations between circulating fibrocytes, inflammation cytokines, PH, and their affective factors that predict the prognosis of HD patients. RESULTS The cohort included 192 patients. During a follow-up of 5 years, we registered 66 all-cause deaths, and 11 patients received kidney transplantation. The incidence of PH among HD patients was 30.9%. We found that the circulating fibrocyte level significantly correlated with pulmonary arterial systolic pressure (r = 0.412, p < 0.05). In the multiple logistic regression analysis, the percentage of circulating fibrocytes was an independent predictor of PH (odds ratio [OR]: 2.080, 95% confidence interval [CI]: 1.539-2.812, p < 0.001). Controlling for confounding covariates in the multivariate Cox regression models, the presence of PH conferred an increased risk of all-cause mortality in HD patients [hazard ratio (HR): 2.183, 95% CI:1.257-3.788, p = 0.006]. CONCLUSION The prevalence of PH was high in HD patients and was associated with higher all-cause mortality. Higher circulating fibrocyte level was an independent predictor of the presence of PH; these fibrocytes may serve as early detection markers and novel therapeutic targets.
Collapse
Affiliation(s)
- Xing Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Xinjian Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Junying Duan
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Ruining Zhang
- Department of Kidney Disease and Blood Purification, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Haipeng Zhang
- Department of Clinical Laboratory, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Weiding Wang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Bingshuo Shi
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Hong Zhou
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
38
|
Bickel MA, Sherry DM, Bullen EC, Vance ML, Jones KL, Howard EW, Conley SM. Microvascular smooth muscle cells exhibit divergent phenotypic switching responses to platelet-derived growth factor and insulin-like growth factor 1. Microvasc Res 2024; 151:104609. [PMID: 37716411 PMCID: PMC10842624 DOI: 10.1016/j.mvr.2023.104609] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/18/2023] [Accepted: 09/09/2023] [Indexed: 09/18/2023]
Abstract
OBJECTIVE Vascular smooth muscle cell (VSMC) phenotypic switching is critical for normal vessel formation, vascular stability, and healthy brain aging. Phenotypic switching is regulated by mediators including platelet derived growth factor (PDGF)-BB, insulin-like growth factor (IGF-1), as well as transforming growth factor-β (TGF-β) and endothelin-1 (ET-1), but much about the role of these factors in microvascular VSMCs remains unclear. METHODS We used primary rat microvascular VSMCs to explore PDGF-BB- and IGF-1-induced phenotypic switching. RESULTS PDGF-BB induced an early proliferative response, followed by formation of polarized leader cells and rapid, directionally coordinated migration. In contrast, IGF-1 induced cell hypertrophy, and only a small degree of migration by unpolarized cells. TGF-β and ET-1 selectively inhibit PDGF-BB-induced VSMC migration primarily by repressing migratory polarization and formation of leader cells. Contractile genes were downregulated by both growth factors, while other genes were differentially regulated by PDGF-BB and IGF-1. CONCLUSIONS These studies indicate that PDGF-BB and IGF-1 stimulate different types of microvascular VSMC phenotypic switching characterized by different modes of cell migration. Our studies are consistent with a chronic vasoprotective role for IGF-1 in VSMCs in the microvasculature while PDGF is more involved in VSMC proliferation and migration in response to acute activities such as neovascularization. Better understanding of the nuances of the phenotypic switching induced by these growth factors is important for our understanding of a variety of microvascular diseases.
Collapse
Affiliation(s)
- Marisa A Bickel
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America
| | - David M Sherry
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America; Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America; Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America
| | - Elizabeth C Bullen
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America
| | - Michaela L Vance
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America
| | - Ken L Jones
- Bioinformatic Solutions, LLC, Sheridan, WY 82801, United States of America
| | - Eric W Howard
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America
| | - Shannon M Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America.
| |
Collapse
|
39
|
Schulze KM, Horn AG, Weber RE, Behnke BJ, Poole DC, Musch TI. Pulmonary hypertension alters blood flow distribution and impairs the hyperemic response in the rat diaphragm. Front Physiol 2023; 14:1281715. [PMID: 38187132 PMCID: PMC10766809 DOI: 10.3389/fphys.2023.1281715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 12/07/2023] [Indexed: 01/09/2024] Open
Abstract
Pulmonary hypertension (PH) is characterized by pulmonary vascular remodeling, respiratory muscle and cardiac impairments, and exercise intolerance. Specifically, impaired gas exchange increases work of the diaphragm; however, compromised contractile function precludes the diaphragm from meeting the increased metabolic demand of chronic hyperventilation in PH. Given that muscle contractile function is in part, dependent upon adequate blood flow (Q ˙ ), diaphragmatic dysfunction may be predicated by an inability to match oxygen delivery with oxygen demand. We hypothesized that PH rats would demonstrate a decreased hyperemic response to contractions compared to healthy controls. Methods: Sprague-Dawley rats were randomized into healthy (HC, n = 7) or PH (n = 7) groups. PH rats were administered monocrotaline (MCT) while HC rats received vehicle. Disease progression was monitored via echocardiography. Regional and total diaphragm blood flow and vascular conductance at baseline and during 3 min of electrically-stimulated contractions were determined using fluorescent microspheres. Results: PH rats displayed morphometric and echocardiographic criteria for disease (i.e., acceleration time/ejection time, right ventricular hypertrophy). In all rats, total costal diaphragm Q ˙ increased during contractions and did not differ between groups. In HC rats, there was a greater increase in medial costal Q ˙ compared to PH rats (55% ± 3% vs. 44% ± 4%, p < 0.05), who demonstrated a redistribution of Q ˙ to the ventral costal region. Conclusion: These findings support a redistribution of regional diaphragm perfusion and an impaired medial costal hyperemic response in PH, suggesting that PH alters diaphragm vascular function and oxygen delivery, providing a potential mechanism for PH-induced diaphragm contractile dysfunction.
Collapse
Affiliation(s)
- Kiana M. Schulze
- Department of Kinesiology, Kansas State University, Manhattan, KS, United States
| | - Andrew G. Horn
- Department of Kinesiology, Kansas State University, Manhattan, KS, United States
| | - Ramona E. Weber
- Department of Kinesiology, Kansas State University, Manhattan, KS, United States
| | - Bradley J. Behnke
- Department of Kinesiology, Kansas State University, Manhattan, KS, United States
| | - David C. Poole
- Department of Kinesiology, Kansas State University, Manhattan, KS, United States
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS, United States
| | - Timothy I. Musch
- Department of Kinesiology, Kansas State University, Manhattan, KS, United States
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS, United States
| |
Collapse
|
40
|
Wu YC, Wang WT, Yang MC, Su YT, Yeh JL, Hsu JH, Wu JR. The novel roles of YULINK in the migration, proliferation and glycolysis of pulmonary arterial smooth muscle cells: implications for pulmonary arterial hypertension. Biol Res 2023; 56:66. [PMID: 38057829 DOI: 10.1186/s40659-023-00480-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: 08/25/2023] [Accepted: 11/22/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND Abnormal remodeling of the pulmonary vasculature, characterized by the proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs) along with dysregulated glycolysis, is a pathognomonic feature of pulmonary arterial hypertension (PAH). YULINK (MIOS, Entrez Gene: 54468), a newly identified gene, has been recently shown to possess pleiotropic physiologic functions. This study aims to determine novel roles of YULINK in the regulation of PAH-related pathogenesis, including PASMC migration, proliferation and glycolysis. RESULTS Our results utilized two PAH-related cell models: PASMCs treated with platelet-derived growth factor (PDGF) and PASMCs harvested from monocrotaline (MCT)-induced PAH rats (PAH-PASMCs). YULINK modulation, either by knockdown or overexpression, was found to influence PASMC migration and proliferation in both models. Additionally, YULINK was implicated in glycolytic processes, impacting glucose uptake, glucose transporter 1 (GLUT1) expression, hexokinase II (HK-2) expression, and pyruvate production in PASMCs. Notably, YULINK and GLUT1 were observed to colocalize on PASMC membranes under PAH-related pathogenic conditions. Indeed, increased YULINK expression was also detected in the pulmonary artery of human PAH specimen. Furthermore, YULINK inhibition led to the suppression of platelet-derived growth factor receptor (PDGFR) and the phosphorylation of focal adhesion kinase (FAK), phosphoinositide 3-kinase (PI3K), and protein kinase B (AKT) in both cell models. These findings suggest that the effects of YULINK are potentially mediated through the PI3K-AKT signaling pathway. CONCLUSIONS Our findings indicate that YULINK appears to play a crucial role in the migration, proliferation, and glycolysis in PASMCs and therefore positioning it as a novel promising therapeutic target for PAH.
Collapse
Affiliation(s)
- Yi-Chia Wu
- Division of Plastic Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Plastic Surgery, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, 80145, Taiwan
- Department of Surgery, School of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Wei-Ting Wang
- Division of Plastic Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Ming-Chun Yang
- Department of Pediatrics, E-Da Hospital/I-Shou University, No. 1, Yi-Da Road, Jiao-Su Village, Yan-Chao District, Kaohsiung, 82445, Taiwan
- Department of Pediatrics, School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Yu-Tsun Su
- Department of Pediatrics, E-Da Hospital/I-Shou University, No. 1, Yi-Da Road, Jiao-Su Village, Yan-Chao District, Kaohsiung, 82445, Taiwan
- Department of Pediatrics, School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Jwu-Lai Yeh
- Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jong-Hau Hsu
- Division of Pediatric Cardio-Pulmonology, Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jiunn-Ren Wu
- Department of Pediatrics, E-Da Hospital/I-Shou University, No. 1, Yi-Da Road, Jiao-Su Village, Yan-Chao District, Kaohsiung, 82445, Taiwan.
| |
Collapse
|
41
|
Liu Y, Zhu L, Ming Y, Wu Z, Zhang L, Chen Q, Qi Y. A role of TRIM59 in pulmonary hypertension: modulating the protein ubiquitylation modification. J Transl Med 2023; 21:821. [PMID: 37978515 PMCID: PMC10655329 DOI: 10.1186/s12967-023-04712-4] [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: 09/25/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Pulmonary hypertension (PH), an infrequent disease, is characterized by excessive pulmonary vascular remodeling and proliferation of pulmonary artery smooth muscle cells (PASMCs). However, its underlying molecular mechanisms remain unclear. Uncovering its molecular mechanisms will be beneficial to the treatment of PH. METHODS Differently expressed genes (DEGs) in the lung tissues of PH patients were analyzed with a GEO dataset GSE113439. From these DEGs, we focused on TRIM59 which was highly expressed in PH patients. Subsequently, the expression of TRIM59 in the pulmonary arteries of PH patients, lung tissues of PH rat model and PASMCs cultured in a hypoxic condition was verified by quantitative real-time PCR (qPCR), western blot and immunohistochemistry. Furthermore, the role of TRIM59 in PAMSC proliferation and pathological changes in PH rats was assessed via gain-of-function and loss-of-function experiments. In addition, the transcriptional regulation of YAP1/TEAD4 on TRIM59 was confirmed by qPCR, western blot, luciferase reporter assay, ChIP and DNA pull-down. In order to uncover the underlying mechanisms of TRIM59, a protein ubiquitomics and a CoIP- HPLC-MS/MS were companied to identify the direct targets of TRIM59. RESULTS TRIM59 was highly expressed in the pulmonary arteries of PH patients and lung tissues of PH rats. Over-expression of TRIM59 accelerated the proliferation of PASMCs, while TRIM59 silencing resulted in the opposite results. Moreover, TRIM59 silencing mitigated the injuries in heart and lung and attenuated pulmonary vascular remodeling during PH. In addition, its transcription was positively regulated by YAP1/TEAD4. Then we further explored the underlying mechanisms of TRIM59 and found that TRIM59 overexpression resulted in an altered ubiquitylation of proteins. Accompanied with the results of CoIP- HPLC-MS/MS, 34 proteins were identified as the direct targets of TRIM59. CONCLUSION TRIM59 was highly expressed in PH patients and promoted the proliferation of PASMCs and pulmonary vascular remodeling, thus contributing to the pathogenesis of PH. It is indicated that TRIM59 may become a potential target for PH treatment.
Collapse
Affiliation(s)
- Yingli Liu
- Department of Pulmonary and Critical Care Medicine, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| | - Li Zhu
- Department of Pulmonary and Critical Care Medicine, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Yue Ming
- Department of Pulmonary and Critical Care Medicine, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| | - Zhuhua Wu
- Department of Pulmonary and Critical Care Medicine, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| | - Lili Zhang
- Department of Pulmonary and Critical Care Medicine, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| | - Qi Chen
- Department of Pulmonary and Critical Care Medicine, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, People's Republic of China
| | - Yong Qi
- Department of Pulmonary and Critical Care Medicine, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Henan University People's Hospital, Zhengzhou, People's Republic of China.
| |
Collapse
|
42
|
Ye L, Wang B, Xu H, Zhang X. The Emerging Therapeutic Role of Prostaglandin E2 Signaling in Pulmonary Hypertension. Metabolites 2023; 13:1152. [PMID: 37999248 PMCID: PMC10672796 DOI: 10.3390/metabo13111152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/28/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023] Open
Abstract
Mild-to-moderate pulmonary hypertension (PH) is a common complication of chronic obstructive pulmonary disease (COPD). It is characterized by narrowing and thickening of the pulmonary arteries, resulting in increased pulmonary vascular resistance (PVR) and ultimately leading to right ventricular dysfunction. Pulmonary vascular remodeling in COPD is the main reason for the increase of pulmonary artery pressure (PAP). The pathogenesis of PH in COPD is complex and multifactorial, involving chronic inflammation, hypoxia, and oxidative stress. To date, prostacyclin and its analogues are widely used to prevent PH progression in clinical. These drugs have potent anti-proliferative, anti-inflammatory, and stimulating endothelial regeneration properties, bringing therapeutic benefits to the slowing, stabilization, and even some reversal of vascular remodeling. As another well-known and extensively researched prostaglandins, prostaglandin E2 (PGE2) and its downstream signaling have been found to play an important role in various biological processes. Emerging evidence has revealed that PGE2 and its receptors (i.e., EP1-4) are involved in the regulation of pulmonary vascular homeostasis and remodeling. This review focuses on the research progress of the PGE2 signaling pathway in PH and discusses the possibility of treating PH based on the PGE2 signaling pathway.
Collapse
Affiliation(s)
- Lan Ye
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116041, China;
| | - Bing Wang
- Department of Endocrinology and Metabolism, The Central Hospital of Dalian University of Technology, Dalian 116000, China;
| | - Hu Xu
- Health Science Center, East China Normal University, Shanghai 200241, China
| | - Xiaoyan Zhang
- Health Science Center, East China Normal University, Shanghai 200241, China
| |
Collapse
|
43
|
Wen S, Unuma K, Funakoshi T, Aki T, Uemura K. Cocaine induces vascular smooth muscle cells proliferation via DRP1-mediated mitochondrial fission and PI3K/HIF-1α signaling. Biochem Biophys Res Commun 2023; 676:30-35. [PMID: 37481940 DOI: 10.1016/j.bbrc.2023.07.020] [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: 07/02/2023] [Accepted: 07/11/2023] [Indexed: 07/25/2023]
Abstract
Long-term cocaine abuse is associated with cardiovascular and pulmonary vascular complications. The vascular toxicity of cocaine can lead to vascular remodeling characterized by excessive proliferation of vascular smooth muscle cells. Though hypoxia-inducible factor (HIF) signaling and mitochondrial fission have been suggested to play essential roles in the pathogenesis of hypoxia-induced vascular remodeling, pathogenetic mechanism for cocaine-related vascular remodeling remains to be elucidated. In this study, we explore the effect of cocaine on the proliferation of vascular smooth muscle cells by in vitro experiments. The findings indicated that the cocaine-induced vascular smooth muscle cell hyperproliferation is achieved by enhancing DRP1-mediated mitochondrial fission and activating PI3K/HIF-1α signaling. Current findings suggested that mitochondrial fission would play a pivotal role in cocaine-related vascular remodeling and would be helpful in understanding the vascular toxicity of cocaine.
Collapse
Affiliation(s)
- Shuheng Wen
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kana Unuma
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takeshi Funakoshi
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Toshihiko Aki
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.
| | - Koichi Uemura
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
44
|
Esparham A, Shoar S, Mehri A, Modukuru VR. Bariatric Surgery and Cardiovascular Disease Risk in Patients with Pulmonary Hypertension: A Propensity Score Matched Analysis of US National Inpatient Sample. Obes Surg 2023; 33:3230-3236. [PMID: 37639208 DOI: 10.1007/s11695-023-06799-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/14/2023] [Accepted: 08/18/2023] [Indexed: 08/29/2023]
Abstract
PURPOSE Previous research has suggested the ameliorating effect of bariatric surgery (BaS) on patients with pulmonary hypertension (PH), but there is a lack of data on the effect of bariatric surgery on the odds of cardiovascular diseases in PH patients. The current study aims to evaluate the association of BaS and coronary artery diseases (CAD), heart failure with reduced ejection fraction (HFrEF), heart failure with preserved ejection fraction (HFpEF), cardiac valve diseases, cardiac rhythm disorders, acute pulmonary embolism, and in-hospital mortality in patients with PH. METHODS The national inpatient sample (NIS) data from 2016 to 2019 were analyzed by using ICD-10 codes. A propensity score matching in a 3:1 ratio was performed to match the BaS and non-BaS groups. RESULTS A total of 3605 patients with a history of BaS and 501419 patients without a history of BaS were included. After propensity matching, BaS was independently associated with a lower CAD hospital admission and a lower rate of in-hospital mortality. On the contrary, BaS was associated with a higher prevalence of atrial fibrillation (AF) and acute pulmonary embolism in patients with PH. HFpEF, HFrEF, other cardiac rhythm disorders, complete heart block, cardiac valve diseases, and ischemic stroke were not significantly different between the two groups in patients with PH. CONCLUSION BaS is independently associated with a reduced rate of in-hospital mortality and CAD hospital admission in patients with PH. However, the risk of atrial fibrillation and acute pulmonary embolism was higher in these patients.
Collapse
Affiliation(s)
- Ali Esparham
- Student Research Committee, College of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Saeed Shoar
- Department of Clinical Research, ScientificWriting Corp, Houston, TX, USA
| | - Ali Mehri
- Endoscopic and Minimally Invasive Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Venkat R Modukuru
- Bariatric and Metabolic Surgery Program, Newark Beth Israel Medical Center, RWJ Barnabas Health, Rutgers NJ Medical School, Newark, NJ, USA
| |
Collapse
|
45
|
Braga CL, Santos RT, da Silva CM, de Novaes Rocha N, Felix NS, Medeiros M, Melo MM, Silva JD, Teixeira DE, Neves CC, Rocco PRM, Cruz FF, Silva PL. Therapeutic effects of hypoxia-preconditioned bone marrow-derived mesenchymal stromal cells and their extracellular vesicles in experimental pulmonary arterial hypertension. Life Sci 2023; 329:121988. [PMID: 37517581 DOI: 10.1016/j.lfs.2023.121988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/17/2023] [Accepted: 07/27/2023] [Indexed: 08/01/2023]
Abstract
AIMS To evaluate BM-MSCs and their extracellular vesicles (EVs) preconditioned with hypoxia or normoxia in experimental pulmonary arterial hypertension (PAH). MAIN METHODS BM-MSCs were isolated and cultured under normoxia (MSC-N, 21%O2) or hypoxia (MSC-H, 1%O2) for 48 h. EVs were then isolated from MSCs under normoxia (EV-N) or hypoxia (EV-H). PAH was induced in male Wistar rats (n = 35) with monocrotaline (60 mg/kg); control animals (CTRL, n = 7) were treated with saline. On day 14, PAH animals received MSCs or EVs under normoxia or hypoxia, intravenously (n = 7/group). On day 28, right ventricular systolic pressure (RVSP), pulmonary acceleration time (PAT)/pulmonary ejection time (PET), and right ventricular hypertrophy (RVH) index were evaluated. Perivascular collagen content, vascular wall thickness, and endothelium-mesenchymal transition were analyzed. KEY FINDINGS PAT/PET was lower in the PAH group (0.26 ± 0.02, P < 0.001) than in CTRLs (0.43 ± 0.02) and only increased in the EV-H group (0.33 ± 0.03, P = 0.014). MSC-N (32 ± 6 mmHg, P = 0.036), MSC-H (31 ± 3 mmHg, P = 0.019), EV-N (27 ± 4 mmHg, P < 0.001), and EV-H (26 ± 5 mmHg, P < 0.001) reduced RVSP compared with the PAH group (39 ± 4 mmHg). RVH was higher in the PAH group than in CTRL and reduced after all therapies. All therapies decreased perivascular collagen fiber content, vascular wall thickness, and the expression of endothelial markers remained unaltered; only MSC-H and EV-H decreased expression of mesenchymal markers in pulmonary arterioles. SIGNIFICANCE MSCs and EVs, under normoxia or hypoxia, reduced right ventricular hypertrophy, perivascular collagen, and vessel wall thickness. Under hypoxia, MSCs and EVs were more effective at improving endothelial to mesenchymal transition in experimental PAH.
Collapse
Affiliation(s)
- Cássia Lisboa Braga
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Renata Trabach Santos
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Carla Medeiros da Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Nazareth de Novaes Rocha
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil; Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niteroi, Brazil
| | - Nathane Santanna Felix
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Mayck Medeiros
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Monique Martins Melo
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Johnatas Dutra Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Douglas Esteves Teixeira
- Laboratory of Biochemistry and Cell Signaling, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, RJ, Brazil
| | - Celso Caruso Neves
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil; Laboratory of Biochemistry and Cell Signaling, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, RJ, Brazil
| | - Patricia Rieken Macedo Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Fernanda Ferreira Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Pedro Leme Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil.
| |
Collapse
|
46
|
ElHady AK, El-Gamil DS, Abdel-Halim M, Abadi AH. Advancements in Phosphodiesterase 5 Inhibitors: Unveiling Present and Future Perspectives. Pharmaceuticals (Basel) 2023; 16:1266. [PMID: 37765073 PMCID: PMC10536424 DOI: 10.3390/ph16091266] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/25/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Phosphodiesterase 5 (PDE5) inhibitors presented themselves as important players in the nitric oxide/cGMP pathway, thus exerting a profound impact on various physiological and pathological processes. Beyond their well-known efficacy in treating male erectile dysfunction (ED) and pulmonary arterial hypertension (PAH), a plethora of studies have unveiled their significance in the treatment of a myriad of other diseases, including cognitive functions, heart failure, multiple drug resistance in cancer therapy, immune diseases, systemic sclerosis and others. This comprehensive review aims to provide an updated assessment of the crucial role played by PDE5 inhibitors (PDE5-Is) as disease-modifying agents taking their limiting side effects into consideration. From a medicinal chemistry and drug discovery perspective, the published PDE5-Is over the last 10 years and their binding characteristics are systemically discussed, and advancement in properties is exposed. A persistent challenge encountered with these agents lies in their limited isozyme selectivity; considering this obstacle, this review also highlights the breakthrough development of the recently reported PDE5 allosteric inhibitors, which exhibit an unparalleled level of selectivity that was rarely achievable by competitive inhibitors. The implications and potential impact of these novel allosteric inhibitors are meticulously explored. Additionally, the concept of multi-targeted ligands is critically evaluated in relation to PDE5-Is by inspecting the broader spectrum of their molecular interactions and effects. The objective of this review is to provide insight into the design of potent, selective PDE5-Is and an overview of their biological function, limitations, challenges, therapeutic potentials, undergoing clinical trials, future prospects and emerging uses, thus guiding upcoming endeavors in both academia and industry within this domain.
Collapse
Affiliation(s)
- Ahmed K. ElHady
- School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, New Administrative Capital, Cairo 11865, Egypt;
| | - Dalia S. El-Gamil
- Department of Chemistry, Faculty of Pharmacy, Ahram Canadian University, Cairo 12451, Egypt;
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt;
| | - Ashraf H. Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt;
| |
Collapse
|
47
|
Saint-Martin Willer A, Santos-Gomes J, Adão R, Brás-Silva C, Eyries M, Pérez-Vizcaino F, Capuano V, Montani D, Antigny F. Physiological and pathophysiological roles of the KCNK3 potassium channel in the pulmonary circulation and the heart. J Physiol 2023; 601:3717-3737. [PMID: 37477289 DOI: 10.1113/jp284936] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 07/04/2023] [Indexed: 07/22/2023] Open
Abstract
Potassium channel subfamily K member 3 (KCNK3), encoded by the KCNK3 gene, is part of the two-pore domain potassium channel family, constitutively active at resting membrane potentials in excitable cells, including smooth muscle and cardiac cells. Several physiological and pharmacological mediators, such as intracellular signalling pathways, extracellular pH, hypoxia and anaesthetics, regulate KCNK3 channel function. Recent studies show that modulation of KCNK3 channel expression and function strongly influences pulmonary vascular cell and cardiomyocyte function. The altered activity of KCNK3 in pathological situations such as atrial fibrillation, pulmonary arterial hypertension and right ventricular dysfunction demonstrates the crucial role of KCNK3 in cardiovascular homeostasis. Furthermore, loss of function variants of KCNK3 have been identified in patients suffering from pulmonary arterial hypertension and atrial fibrillation. This review focuses on current knowledge of the role of the KCNK3 channel in pulmonary circulation and the heart, in healthy and pathological conditions.
Collapse
Affiliation(s)
- Anaïs Saint-Martin Willer
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 'Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique', Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Joana Santos-Gomes
- Cardiovascular R&D Centre-UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Rui Adão
- Cardiovascular R&D Centre-UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
- CIBER Enfermedades Respiratorias (Ciberes), Madrid, Spain
| | - Carmen Brás-Silva
- Cardiovascular R&D Centre-UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Mélanie Eyries
- Département de génétique, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France
| | - Francisco Pérez-Vizcaino
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Madrid, Spain
- CIBER Enfermedades Respiratorias (Ciberes), Madrid, Spain
| | - Véronique Capuano
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 'Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique', Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - David Montani
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 'Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique', Hôpital Marie Lannelongue, Le Plessis-Robinson, France
- Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Fabrice Antigny
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999 'Hypertension Pulmonaire: Physiopathologie et Innovation Thérapeutique', Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| |
Collapse
|
48
|
Ye C, Yuan L, Wu K, Shen B, Zhu C. Association between systemic immune-inflammation index and chronic obstructive pulmonary disease: a population-based study. BMC Pulm Med 2023; 23:295. [PMID: 37563621 PMCID: PMC10416535 DOI: 10.1186/s12890-023-02583-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/25/2023] [Indexed: 08/12/2023] Open
Abstract
BACKGROUND The Systemic Immune-Inflammation Index (SII) is a quantitative measurement of the systemic immune-inflammatory response in the human body. The SII has been shown to have prognostic value in various clinical settings, including critical illness, sepsis, and cancer. Its role in chronic obstructive pulmonary disease (COPD) remains unclear and requires further investigation. METHODS We analyzed demographic data from 16,636 participants in the National Health and Nutrition Examination Survey. Logistic regression analysis was performed to assess the correlation between COPD, lung function, chronic respiratory symptoms and SII. We used Cox proportional hazards (PH) model to analyze the relationship between SII and mortality in COPD patients and healthy individuals. We used propensity score matching (PSM) method to match the COPD population with similar baseline levels with the normal population to further analyze the correlation between SII and COPD. RESULTS We recruited 16,636 participants, ages 40 and above, for the study. A multivariable logistic regression analysis revealed that a higher SII level was independently associated with an elevated likelihood of COPD (Odds Ratio (OR) = 1.449; 95% Confidence Interval (CI): 1.252-1.676, P < 0.0001) after controlling for all other factors. Results of subgroup analysis showed a significant positive correlation between SII and COPD in different age groups, gender, Body Mass Index, smoking status, and those with a history of hypertension. The SII index had positive correlation with COPD after PSM (OR = 1.673; 95%CI: 1.443-1.938). After full adjustment, an increase in the SII is associated with a higher all-cause mortality rate. The hazard ratio (HR) with a 95% CI in the general population, COPD patients, and healthy individuals are 1.161 (1.088, 1.239), 1.282 (1.060, 1.550), and 1.129 (1.055, 1.207), respectively. CONCLUSIONS Higher SII levels are linked to higher prevalence of COPD. COPD patients with a higher SII levels have a higher risk of all-cause mortality. Additional large-scale, long-term studies are necessary to confirm these results.
Collapse
Affiliation(s)
- Chenglin Ye
- Department of Clinical Laboratory, institute of translational medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, PR China
| | - Li Yuan
- Department of Clinical Laboratory, Zhongnan Hospital of Wuhan University, Wuhan, 430060, Hubei, PR China
| | - Kailang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, PR China
| | - Bingzheng Shen
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, PR China
| | - Chengliang Zhu
- Department of Clinical Laboratory, institute of translational medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, PR China.
| |
Collapse
|
49
|
Huang H, Lin D, Hu L, Wang J, Yu Y, Yu Y, Li K, Chen F. RNA Binding Protein Quaking Promotes Hypoxia-induced Smooth Muscle Reprogramming in Pulmonary Hypertension. Am J Respir Cell Mol Biol 2023; 69:159-171. [PMID: 37146099 DOI: 10.1165/rcmb.2022-0349oc] [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: 09/06/2022] [Accepted: 05/04/2023] [Indexed: 05/07/2023] Open
Abstract
Pulmonary hypertension (PH) is a devastating disease characterized by progressive increases in pulmonary vascular resistance and remodeling, which eventually leads to right ventricular failure and death. The aim of this study was to identify novel molecular mechanisms involved in the hyperproliferation of pulmonary artery smooth muscle cells (PASMCs) in PH. In this study, we first demonstrated that the mRNA and protein expression amounts of QKI (Quaking), an RNA-binding protein, were elevated in human and rodent PH lung and pulmonary artery tissues and hypoxic human PASMCs. QKI deficiency attenuated PASMC proliferation in vitro and vascular remodeling in vivo. Next, we elucidated that QKI increases STAT3 (signal transducer and activator of transcription 3) mRNA stability by binding to its 3' untranslated region. QKI inhibition reduced STAT3 expression and alleviated PASMC proliferation in vitro. Moreover, we also observed that the upregulated expression of STAT3 promoted PASMC proliferation in vitro and in vivo. In addition, as a transcription factor, STAT3 bound to microRNA (miR)-146b promoter to enhance its expression. We further showed that miR-146b promoted the proliferation of smooth muscle cells by inhibiting STAT1 and TET2 (Tet methylcytosine dioxygenase 2) during pulmonary vascular remodeling. This study has demonstrated new mechanistic insights into hypoxic reprogramming that arouses vascular remodeling, thus providing proof of concept for targeting vascular remodeling by directly modulating the QKI-STAT3-miR-146b pathway in PH.
Collapse
Affiliation(s)
| | | | - Li Hu
- Department of Forensic Medicine and
| | - Jie Wang
- Department of Forensic Medicine and
| | | | | | - Kai Li
- Department of Forensic Medicine and
| | - Feng Chen
- Department of Forensic Medicine and
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
50
|
Tannu M, Krasuski RA. The Evolution of Pulmonary Artery Denervation for Treatment of Pulmonary Arterial Hypertension. Interv Cardiol Clin 2023; 12:381-391. [PMID: 37290841 DOI: 10.1016/j.iccl.2023.03.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive, life-limiting disease. Despite significant medical progress over the last three decades, the prognosis of PAH remains poor. PAH is associated with sympathetic nervous system over-stimulation and baroreceptor-mediated vasoconstriction, leading to pathologic pulmonary artery (PA) and right ventricular remodeling. PA denervation is a minimally-invasive intervention that ablates local sympathetic nerve fibers and baroreceptors to modulate pathologic vasoconstriction. Preliminary animal and clinical studies have shown improvements in short-term pulmonary hemodynamics and PA remodeling. However, future studies are needed to elucidate appropriate patient selection, timing of intervention, and long-term efficacy before integration into standard of care.
Collapse
Affiliation(s)
- Manasi Tannu
- Division of Cardiology, Duke University Health System, DUMC 3012, Durham, NC 27710, USA
| | - Richard A Krasuski
- Division of Cardiology, Duke University Health System, DUMC 3012, Durham, NC 27710, USA.
| |
Collapse
|