1
|
Singh E, Bisserier M. Experimental animal models and patient-derived platforms to bridge preclinical discovery and translational therapeutics in pulmonary arterial hypertension. J Transl Med 2025; 23:665. [PMID: 40528193 DOI: 10.1186/s12967-025-06709-7] [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: 05/09/2025] [Accepted: 06/02/2025] [Indexed: 06/20/2025] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive vascular disease characterized by sustained elevation of pulmonary arterial pressure, endothelial cell dysfunction, and right ventricular failure. A wide range of experimental animal models, including the monocrotaline model, Sugen combined with hypoxia, and pulmonary artery banding in large animals, have been pivotal in uncovering disease mechanisms such as vascular remodeling, metabolic dysregulation, and hypoxia-inducible signaling. More recently, human-based platforms, including induced pluripotent stem cell-derived vascular cells, organ-on-chip systems, and precision-cut lung slices, have emerged as powerful tools to model patient-specific pathophysiology and study pharmacological responses. These systems enable the interrogation of BMPR2 mutations, mitochondrial dysfunction, and sex-specific responses, factors often overlooked in traditional preclinical models. Moreover, integrating these platforms with omics technologies and comorbidity-driven experimental systems addresses key translational gaps. This review provides an overview of animal and human-based models used in PAH research and highlights emerging strategies to enhance their translational relevance. We advocate for a multi-platform and precision medicine-oriented approach that bridges preclinical insights with clinical outcomes to accelerate therapeutic development in PAH.
Collapse
Affiliation(s)
- Elizabeth Singh
- Department of Cell Biology and Anatomy, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY, 10595, USA
- Department of Physiology, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY, 10595, USA
| | - Malik Bisserier
- Department of Cell Biology and Anatomy, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY, 10595, USA.
- Department of Physiology, New York Medical College, 15 Dana Road, BSB 131A, Valhalla, NY, 10595, USA.
| |
Collapse
|
2
|
Zheng X, Zhao J, Jia X, Pan J, Xu S, Wang D, Li J, Ji Y, Zhu Z, Hasnain M, Sui Z, Wang R, Yuan Y. Fucoxanthin Ameliorates Vascular Remodeling via Attenuating Oxidative Stress in Hypoxic Pulmonary Hypertension Rats. J Nutr Biochem 2025:110002. [PMID: 40513841 DOI: 10.1016/j.jnutbio.2025.110002] [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: 10/07/2024] [Revised: 06/09/2025] [Accepted: 06/10/2025] [Indexed: 06/16/2025]
Abstract
Hypoxic pulmonary hypertension (HPH) is a fatal cardiopulmonary disease characterized by pulmonary vascular remodeling, primarily resulting from abnormal proliferation of pulmonary artery smooth muscle cells (PASMCs). Fucoxanthin, a natural carotenoid with potent antioxidant activity, was investigated for its therapeutic potential in HPH, given the critical role of oxidative stress in disease pathogenesis. In this study, Sprague-Dawley rats were exposed to intermittent chronic hypoxia for 4 weeks to mimic severe HPH. The results demonstrated that fucoxanthin significantly reduced the elevated right ventricular systolic pressure (RVSP), alleviated right ventricular hypertrophy, and mitigated pulmonary artery remodeling in the HPH rats. Additionally, fucoxanthin enhanced superoxide dismutase (SOD) activity and glutathione (GSH)/glutathione disulfide (GSSG) ratio while decreasing malondialdehyde (MDA) levels in both lung tissues and serum of HPH rats. In vitro, fucoxanthin inhibited cell proliferation and migration, decreased reactive oxygen species (ROS) production in hypoxia-induced PASMCs, and improved cell viability in hypoxia-induced endothelial cells (ECs). Importantly, fucoxanthin reduced hypoxia-inducible factor 1 alpha (HIF-1α) expression in both lung tissues and PASMCs under hypoxia. Notably, fucoxanthin exhibited effects similar to those of 2-methoxyestradiol (2ME2), an inhibitor of HIF-1α, on cell proliferation and ROS production in hypoxia-induced PASMCs. Moreover, fucoxanthin treatment did not significantly alter HIF-1α expression, cell proliferation, or ROS production after 2ME2 blocked HIF-1α. Collectively, fucoxanthin suppressed hypoxia-induced oxidative stress primarily by regulating the HIF-1α-ROS pathway, thereby alleviating pulmonary remodeling in HPH. Our findings represent a promising therapeutic strategy for HPH by improving pulmonary vascular remodeling.
Collapse
Affiliation(s)
- Xu Zheng
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Jina Zhao
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Xiaoqin Jia
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Jinjin Pan
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Shuo Xu
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Dingyou Wang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Junxia Li
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China; The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuke Ji
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Zhilong Zhu
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Muhammad Hasnain
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Zheng Sui
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China; Department of Vasculocardiology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Rui Wang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China; College of Pharmacy, Dalian Medical University, Dalian, China.
| | - Yuhui Yuan
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.
| |
Collapse
|
3
|
Ohno Y, Fujisawa Y, Yoshikawa T, Seki S, Takenaka D, Fujii K, Ito Y, Kimata H, Akino N, Nagata H, Nomura M, Ueda T, Ozawa Y. Dynamic perfusion area-detector CT in non-small cell lung cancer with progressive fibrosing interstitial lung disease. Eur Radiol 2025:10.1007/s00330-025-11653-7. [PMID: 40402289 DOI: 10.1007/s00330-025-11653-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 03/24/2025] [Accepted: 04/09/2025] [Indexed: 05/23/2025]
Abstract
OBJECTIVES To determine the capability of dynamic contrast-enhanced (CE-) perfusion area-detector CT (ADCT) for detecting pathological structural changes in stage I non-small cell lung cancer (NSCLC) patients. MATERIALS AND METHODS Sixty-three consecutive stage I NSCLC patients with progressive fibrosing interstitial lung disease (PF-ILD) underwent dynamic CE-perfusion ADCT analyzed by dual-input maximum slope (DMS) methods for total, pulmonary arterial and systemic arterial perfusion (TPDMS, PAPDMS and SAPDMS) maps, surgical treatment and pathological examination. Multicentric ROIs were then placed over sites assessed as normal lung, pulmonary emphysema, GGO or reticular pattern without traction bronchiectasis, reticular pattern with traction bronchiectasis and honeycombing in the resected lung. Next, an analysis of variance (ANOVA) followed by Tukey's honest significant difference (HSD) multiple comparison test was performed for a comparison of each of the perfusion parameters for five groups. Finally, discrimination accuracy for evaluation of lung parenchymal change was compared for all indexes and combined methods. RESULTS PAPDMSs of abnormal lungs were significantly lower than that of normal lungs (p < 0.0001). SAPDMSs of normal or emphysematous lungs were significantly lower than those of others (p < 0.0001). SAPDMS of GGO or reticular pattern without traction bronchiectasis was significantly lower than that for reticular pattern with traction bronchiectasis and honeycombing (p < 0.0001). Discrimination accuracy of combined perfusion index was significantly higher than that of each index (p < 0.0001). CONCLUSION Dynamic CE-perfusion ADCT is useful for detecting pathological structural changes in stage I NSCLC patients with PF-ILD. KEY POINTS Question Can dynamic first-pass contrast-enhanced perfusion matrices evaluate parenchymal lung changes and disease severity of parenchymal diseases in stage I non-small cell lung cancer (NSCLC) patients? Findings Perfusion indexes differentiated significantly among normal lung, emphysema, GGO or reticular pattern without traction bronchiectasis, reticular pattern with traction bronchiectasis and honeycombing and significantly improved discrimination accuracy by combined methods. Clinical relevance Dynamic first-pass contrast-enhanced perfusion area-detector CT has the potential to assess underlying pathologies and pulmonary functional changes in stage I non-small cell carcinoma patients with progressive fibrosing interstitial lung disease.
Collapse
Affiliation(s)
- Yoshiharu Ohno
- Department of Diagnostic Radiology, Fujita Health University School of Medicine, Toyoake, Japan.
- Joint Research Laboratory of Advanced Medical Imaging, Fujita Health University School of Mediciine, Toyoake, Japan.
- Division of Functional and Diagnostic Imaging Research, Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan.
| | | | - Takeshi Yoshikawa
- Department of Diagnostic Radiology, Fujita Health University School of Medicine, Toyoake, Japan
- Division of Functional and Diagnostic Imaging Research, Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan
- Division of Diagnostic Radiology, Hyogo Cancer Center, Akashi, Japan
| | - Shinichiro Seki
- Division of Functional and Diagnostic Imaging Research, Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan
- Division of Diagnostic Radiology, Hyogo Cancer Center, Akashi, Japan
| | - Daisuke Takenaka
- Department of Diagnostic Radiology, Fujita Health University School of Medicine, Toyoake, Japan
- Division of Functional and Diagnostic Imaging Research, Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kenji Fujii
- Canon Medical Systems Corporation, Otawara, Japan
| | - Yuya Ito
- Canon Medical Systems Corporation, Otawara, Japan
| | | | | | - Hiroyuki Nagata
- Joint Research Laboratory of Advanced Medical Imaging, Fujita Health University School of Mediciine, Toyoake, Japan
| | - Masahiko Nomura
- Department of Diagnostic Radiology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Takahiro Ueda
- Department of Diagnostic Radiology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Yoshiyuki Ozawa
- Department of Diagnostic Radiology, Fujita Health University School of Medicine, Toyoake, Japan
| |
Collapse
|
4
|
Jensen L, Guo Z, Sun X, Jing X, Yang Y, Cao Y. Angiogenesis, signaling pathways, and animal models. Chin Med J (Engl) 2025; 138:1153-1162. [PMID: 40254738 PMCID: PMC12091601 DOI: 10.1097/cm9.0000000000003561] [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: 06/14/2024] [Indexed: 04/22/2025] Open
Abstract
ABSTRACT The vasculature plays a critical role in homeostasis and health as well as in the development and progression of a wide range of diseases, including cancer, cardiovascular diseases, metabolic diseases (and their complications), chronic inflammatory diseases, ophthalmic diseases, and neurodegenerative diseases. As such, the growth of the vasculature mediates normal development and physiology, as well as disease, when pathologically induced vessels are morphologically and functionally altered owing to an imbalance of angiogenesis-stimulating and angiogenesis-inhibiting factors. This review offers an overview of the angiogenic process and discusses recent findings that provide additional interesting nuances to this process, including the roles of intussusception and angiovasculogenesis, which may hold promise for future therapeutic interventions. In addition, we review the methodology, including those of in vitro and in vivo assays, which has helped build the vast amount of knowledge on angiogenesis available today and identify important remaining knowledge gaps that should be bridged through future research.
Collapse
Affiliation(s)
- Lasse Jensen
- Department of Health, Medical and Caring Sciences, Unit of Diagnostics and Specialist Medicine, Linköping University, Linköping SE-58183, Sweden
| | - Ziheng Guo
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaoting Sun
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vison and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang 325024, China
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna 17165, Sweden
| | - Xu Jing
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna 17165, Sweden
| | - Yunlong Yang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna 17165, Sweden
| |
Collapse
|
5
|
Dai J, Chen H, Fang J, Wu S, Jia Z. Vascular Remodeling: The Multicellular Mechanisms of Pulmonary Hypertension. Int J Mol Sci 2025; 26:4265. [PMID: 40362501 PMCID: PMC12072204 DOI: 10.3390/ijms26094265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/24/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
Pulmonary hypertension (PH) is a serious cardiovascular disease caused by a variety of pathogenic factors, which is characterized by increased pulmonary vascular resistance (PVR) and progressive elevation of mean pulmonary artery pressure (mPAP). This disease can lead to right ventricular hypertrophy and, in severe cases, right heart failure and even death. Vascular remodeling-a pathological modification involving aberrant vasoconstriction, cell proliferation, apoptosis resistance, and inflammation in the pulmonary vascular system-is a significant pathological hallmark of PH and a critical process in its progression. Recent studies have found that vascular remodeling involves the participation of a diversity of cellular pathological alterations, such as the dysfunction of pulmonary artery endothelial cells (PAECs), the proliferation and migration of pulmonary artery smooth muscle cells (PASMCs), the phenotypic differentiation of pulmonary artery fibroblasts, the inflammatory response of immune cells, and pericyte proliferation. This review focuses on the mechanisms and the intercellular crosstalk of these cells in the PH process, emphasizing recent advances in knowledge regarding cellular signaling pathways, inflammatory responses, apoptosis, and proliferation. To develop better treatments, a list of possible therapeutic approaches meant to slow down certain biological functions is provided, with the aim of providing new insights into the treatment of PH by simplifying the intricacies of these complex connections. In this review, comprehensive academic databases such as PubMed, Embase, Web of Science, and Google Scholar were systematically searched to discuss studies relevant to human and animal PH, with a focus on vascular remodeling in PH.
Collapse
Affiliation(s)
- Jinjin Dai
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming 650500, China; (J.D.); (H.C.); (J.F.)
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Kunming 650500, China
- Key Laboratory of Microcosmic Syndrome Differentiation, Education Department of Yunnan, Kunming 650500, China
| | - Hongyang Chen
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming 650500, China; (J.D.); (H.C.); (J.F.)
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Kunming 650500, China
- Key Laboratory of Microcosmic Syndrome Differentiation, Education Department of Yunnan, Kunming 650500, China
| | - Jindong Fang
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming 650500, China; (J.D.); (H.C.); (J.F.)
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Kunming 650500, China
- Key Laboratory of Microcosmic Syndrome Differentiation, Education Department of Yunnan, Kunming 650500, China
| | - Shiguo Wu
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming 650500, China; (J.D.); (H.C.); (J.F.)
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Kunming 650500, China
- Key Laboratory of Microcosmic Syndrome Differentiation, Education Department of Yunnan, Kunming 650500, China
| | - Zhuangzhuang Jia
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming 650500, China; (J.D.); (H.C.); (J.F.)
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Kunming 650500, China
- Key Laboratory of Microcosmic Syndrome Differentiation, Education Department of Yunnan, Kunming 650500, China
| |
Collapse
|
6
|
Jimenez SA, Mendoza FA, Piera-Velazquez S. A review of recent studies on the pathogenesis of Systemic Sclerosis: focus on fibrosis pathways. Front Immunol 2025; 16:1551911. [PMID: 40308583 PMCID: PMC12040652 DOI: 10.3389/fimmu.2025.1551911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 03/07/2025] [Indexed: 05/02/2025] Open
Abstract
Systemic Sclerosis (SSc) is a systemic autoimmune disease of unknown etiology characterized by the development of frequently progressive cutaneous and internal organ fibrosis accompanied by severe vascular alterations. The pathogenesis of SSc is highly complex and, despite extensive investigation, has not been fully elucidated. Numerous studies have suggested that unknown etiologic factors cause multiple alterations in genetically receptive hosts, leading to SSc development and progression. These events may be functionally and pathologically interconnected and include: 1) Structural and functional microvascular and endothelial cell abnormalities; 2) Severe oxidative stress and high reactive oxygen species (3); Frequently progressive cutaneous and visceral fibrosis; 4) Transdifferentiation of various cell types into activated myofibroblasts, the cells ultimately responsible for the fibrotic process; 5) Establishment of a chronic inflammatory process in various affected tissues; 6) Release of cytokines, chemokines, and growth factors from the inflammatory cells; 7) Abnormalities in humoral and cellular immunity with the production of specific autoantibodies; and 8) Epigenetic alterations including changes in multiple non-coding RNAs. These events manifest with different levels of intensity in the affected organs and display remarkable individual variability, resulting in a wide heterogeneity in the extent and severity of clinical manifestations. Here, we will review some of the recent studies related to SSc pathogenesis.
Collapse
Affiliation(s)
- Sergio A. Jimenez
- Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia, PA, United States
| | - Fabian A. Mendoza
- Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia, PA, United States
- Division of Rheumatology, Department of Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - Sonsoles Piera-Velazquez
- Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
7
|
Posey JN, Jordan M, Lewis CV, Sul C, Dobrinskikh E, Swindle D, Denorme F, Irwin D, Di Paola J, Stenmark K, Nozik ES, Delaney C. Nbeal2 knockout mice are not protected against hypoxia-induced pulmonary vascular remodeling and pulmonary hypertension. Blood Adv 2025; 9:1571-1584. [PMID: 39693512 PMCID: PMC11986223 DOI: 10.1182/bloodadvances.2024013880] [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: 06/10/2024] [Revised: 11/05/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
ABSTRACT Inflammation drives the initiation and progression of pulmonary hypertension (PH). Platelets, increasingly recognized as immune cells, are activated and increased in the lungs of patients with PH. Platelet activation leads to the release of α-granule chemokines, many of which are implicated in PH. We hypothesized that hypoxia-induced secretion of platelet α-granule-stored proteins and PH would be prevented in Neurobeachin-like 2 knockout (Nbeal2-/-) α-granule-deficient mice. Wild-type (WT) and Nbeal2-/- mice were maintained in normoxia or exposed to 10% hypobaric hypoxia for 3, 14, 21, or 35 days. We observed macrothrombocytopenia, increased circulating neutrophils and monocytes, and increased lung interstitial macrophages (IMs) in Nbeal2-/- mice at baseline. Hypoxia-induced platelet activation was attenuated, and hypoxia-induced increase in lung platelet factor 4 (PF4) and platelets was delayed in Nbeal2-/- mice compared with in WT mice. Finally, although pulmonary vascular remodeling (PVR) and PH were attenuated at day 21, Nbeal2-/- mice were not protected against hypoxia-induced PVR and PH at day 35. Although this mutation also affected circulating monocytes, neutrophils, and lung IMs, all of which are critical in the development of experimental PH, we gained further support for the role of platelets and α-granule proteins, such as PF4, in PH progression and pathogenesis and made several observations that expand our understanding of α-granule-deficient mice in chronic hypoxia.
Collapse
Affiliation(s)
- Janelle N. Posey
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
- Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Mariah Jordan
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
- Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Caitlin V. Lewis
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
- Section of Pediatric Critical Care, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Christina Sul
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
- Section of Pediatric Critical Care, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Evgenia Dobrinskikh
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
- Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Delaney Swindle
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Frederik Denorme
- Department of Emergency Medicine, Washington University School of Medicine, St. Louis, MO
| | - David Irwin
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Jorge Di Paola
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Washington University in St. Louis, St. Louis, MO
| | - Kurt Stenmark
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
- Section of Pediatric Critical Care, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Eva S. Nozik
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
- Section of Pediatric Critical Care, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Cassidy Delaney
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, CO
- Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
8
|
Peers de Nieuwburgh M, Hunt M, Chandrasekaran P, Vincent TL, Hayes KB, Randazzo IR, Gunder M, De Bie FR, Colson A, Lu M, Wen H, Michki SN, Rychik J, Debiève F, Katzen J, Young LR, Davey MG, Flake AW, Gaynor JW, Frank DB. Chronic Hypoxia in an EXTrauterine Environment for Neonatal Development Impairs Lung Development. Am J Respir Cell Mol Biol 2025; 72:441-452. [PMID: 39453404 DOI: 10.1165/rcmb.2024-0012oc] [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: 01/11/2024] [Accepted: 10/23/2024] [Indexed: 10/26/2024] Open
Abstract
Severe fetal hypoxia poses a significant risk to lung development, resulting in severe postnatal complications. Existing chronic hypoxia animal models lack the ability to achieve pathologically reduced fetal oxygen without compromising animal development, placental blood flow, or maternal health. Using an established model of isolated chronic hypoxia involving the Extrauterine Environment for Neonatal Development, we are able to investigate the direct impact of fetal hypoxia on lung development. Oxygen delivery to preterm fetal lambs (105-110 d gestational age) delivered by cesarean section was reduced, and animals were supported using the Extrauterine Environment for Neonatal Development through the canalicular or saccular stage of lung development. Fetal lambs in hypoxic conditions showed significant growth restriction compared with their normoxic counterparts. We also observed modest aberrant vascular remodeling in the saccular group after hypoxic conditions, with decreased macrovessel numbers and microvascular endothelial cell numbers and increased peripheral vessel muscularization. In addition, fetal hypoxia resulted in enlarged distal airspaces and decreased septal wall volume. Moreover, there was a reduction in mature SFTPB (surfactant protein B) and processed SFTPC protein expression concomitant with a decrease in alveolar type 2 cell number. These findings demonstrate that maternally independent fetal hypoxia predominantly affects distal airway development, alveolar type 2 cell number, and surfactant production, with mild effects on the vasculature.
Collapse
Affiliation(s)
| | - Mallory Hunt
- Division of Cardiovascular Surgery, Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Tiffany L Vincent
- Division of Pulmonology and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | | | - Arthur Colson
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, and
- Service d'Obstétrique, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | | | | | - Sylvia N Michki
- Division of Cardiology
- Division of Pulmonology and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Fréderic Debiève
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, and
- Service d'Obstétrique, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Jeremy Katzen
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Lisa R Young
- Division of Pulmonology and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | | |
Collapse
|
9
|
Yang X, Liu H, Wu X. High-altitude pulmonary hypertension: a comprehensive review of mechanisms and management. Clin Exp Med 2025; 25:79. [PMID: 40063280 PMCID: PMC11893705 DOI: 10.1007/s10238-025-01577-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 01/26/2025] [Indexed: 03/14/2025]
Abstract
High-altitude pulmonary hypertension (HAPH) is characterized by an increase in pulmonary artery pressure due to prolonged exposure to hypoxic environment at high altitudes. The development of HAPH involves various factors such as pressure changes, inflammation, oxidative stress, gene regulation, and signal transduction. The pathophysiological mechanisms of this condition operate at molecular, cellular, and genetic levels. Diagnosis of HAPH often relies on echocardiography, cardiac catheterization, and other methods to assess pulmonary artery pressure and its impact on cardiac function. Treatment options for HAPH encompass both nondrug and drug therapies. While advancements have been made in understanding the pathological mechanisms through research on animal models and clinical trials, there are still limitations to be addressed. Future research should focus on exploring molecular targets, personalized medicine, long-term management strategies, and interdisciplinary approaches. By leveraging advanced technologies like systems biology, omics technology, big data, and artificial intelligence, a comprehensive analysis of HAPH pathogenesis can lead to the identification of new treatment targets and strategies, ultimately enhancing patient quality of life and prognosis. Furthermore, research on health monitoring and preventive measures for populations living at high altitudes should be intensified to reduce the incidence and mortality of HAPH.
Collapse
Affiliation(s)
- Xitong Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China
- The First Affiliated Hospital of Dali University, Dali, Yunnan, China
| | - Hong Liu
- The First Affiliated Hospital of Dali University, Dali, Yunnan, China
| | - Xinhua Wu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China.
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China.
- The First Affiliated Hospital of Dali University, Dali, Yunnan, China.
| |
Collapse
|
10
|
Wang Y, Wang Y, Zhang W. Dysregulation of Mitochondrial in Pulmonary Hypertension-Related Right Ventricular Remodeling: Pathophysiological Features and Targeting Drugs. Rev Cardiovasc Med 2025; 26:25781. [PMID: 40160582 PMCID: PMC11951289 DOI: 10.31083/rcm25781] [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: 07/21/2024] [Revised: 11/14/2024] [Accepted: 11/29/2024] [Indexed: 04/02/2025] Open
Abstract
Pulmonary hypertension (PH) is a life-threatening condition characterized by right ventricular (RV) remodeling, which is a major determinant of patient survival. The progression of right ventricular remodeling is significantly influenced by mitochondrial dysfunction, providing profound insights into vascular health and cardiovascular risk. In this review, we discuss the molecular targets, pathophysiological characteristics, and potential mechanisms underlying mitochondrial dysfunction in PH, encompassing disturbances in mitochondrial dynamics, inflammation, and dysregulation of mitochondrial energy metabolism. Finally, we review the primary therapeutic targets currently utilized to address cardiac dysfunction resulting from mitochondrial damage. Hopefully, this might inspire novel approaches to the management of cardiovascular disorders.
Collapse
Affiliation(s)
- Yuehan Wang
- Departments of Pharmacy, The Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China
- Huankui Academy, Nanchang University, 330036 Nanchang, Jiangxi, China
| | - Yingzhuo Wang
- The First Clinical Medical College, Nanchang University, 330036 Nanchang, Jiangxi, China
| | - Weifang Zhang
- Departments of Pharmacy, The Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China
- Huankui Academy, Nanchang University, 330036 Nanchang, Jiangxi, China
| |
Collapse
|
11
|
Cieri IF, Rodriguez Alvarez AA, Patel S, Boya M, Nurko A, Teeple W, Dua A. TEG-Guided Anticoagulation Assessment in Deep Vein Arterialization: A Prospective Analysis. Ann Vasc Surg 2025; 112:287-297. [PMID: 39733999 DOI: 10.1016/j.avsg.2024.12.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/11/2024] [Accepted: 12/18/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND Deep vein arterialization (DVA) is an innovative surgical technique aimed at enhancing blood flow in compromised limbs facing amputation. Maintenance of flow postrevascularization is crucial to limb salvage. As this is a new technique, no standardized thromboprophylaxis regime is currently established, and postprocedure thromboprophylaxis is at the discretion of the proceduralist. This study aims to evaluate coagulation profiles using viscoelastic studies in peripheral artery disease patients who underwent DVA, assessing the impact of various postprocedure thromboprophylaxis regimens. METHODS Patients (aged > 60 years) undergoing DVA were prospectively evaluated using thromboelastography at baseline, 1, 3, and 6 months (2020-2024). Postprocedure thromboprophylaxis included mono antiplatelet therapy (MAPT), MAPT + direct oral anticoagulant (DOAC), dual antiplatelet therapy (DAPT), or DAPT + DOAC. Coagulation profiles were analyzed using descriptive statistics. RESULTS Among 16 patients (mean age 66.6 years, 75% male/Caucasian), hypertension and hyperlipidemia were present in 91%, and diabetes in 88%. The DAPT + DOAC group showed consistently superior platelet inhibition with the lowest adenosine diphosphate maximum amplitude values throughout baseline (35.65 mm vs. 42.2-65.03 mm in other groups), 1 month (26.7 mm vs. 32.14-69.4 mm), 3 months (27.36 mm vs. 32.2-39.97 mm), and 6 months (43.7 mm vs. 50.2-50.5 mm). MAPT demonstrated the slowest clot strengthening (citrated kaolin angle 65.25° vs. 68.7-71.55°). CONCLUSION Thromboelastography with platelet mapping demonstrated enhanced platelet inhibition and reduced clot formation in the DAPT + DOAC group, suggesting the importance of coagulation monitoring.
Collapse
Affiliation(s)
- Isabella F Cieri
- Division of Vascular and Endovascular Surgery, Massachusetts General Hospital, Boston, MA
| | | | - Shiv Patel
- Division of Vascular and Endovascular Surgery, Massachusetts General Hospital, Boston, MA
| | - Mounika Boya
- Division of Vascular and Endovascular Surgery, Massachusetts General Hospital, Boston, MA
| | - Andrea Nurko
- Division of Vascular and Endovascular Surgery, Massachusetts General Hospital, Boston, MA
| | - William Teeple
- Division of Vascular and Endovascular Surgery, Massachusetts General Hospital, Boston, MA
| | - Anahita Dua
- Division of Vascular and Endovascular Surgery, Massachusetts General Hospital, Boston, MA.
| |
Collapse
|
12
|
Oğuz M, Uzun M, Yılmaz İ, Erdem A, Demirci M, Duran NE. Pulmonary arterial stiffness and vascular tone in pulmonary hypertension: Insights from waveform-derived reflection index and hemodynamic correlations. Heart Lung 2025; 70:50-56. [PMID: 39571493 DOI: 10.1016/j.hrtlng.2024.11.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/04/2024] [Accepted: 11/11/2024] [Indexed: 02/24/2025]
Abstract
BACKGROUND Pulmonary hypertension (PH) involves increased arterial stiffness and reduced vascular tone, affecting pulmonary arterial wave reflections. The Reflection Index (RI) may provide insights into these changes. OBJECTIVE This study examines the utility of RI in PH patients by correlating it with key right heart catheterization (RHC) parameters. METHODS Patients who underwent RHC with a preliminary diagnosis of PH, including those with normal RHC findings and those diagnosed with Group 1 and Group 4 PH, were included in the study. RI was defined as the ratio of systolic to diastolic pressure differences from pulmonary arterial waveforms and compared with hemodynamic, clinical, and echocardiographic parameters. RESULTS The study included 115 patients (mean age 53.92 ± 16.43 years; 43.5% male). RI showed significant correlations with key RHC parameters, such as sPAP (r=0.359, p<0.001), dPAP (r=0.322, p<0.001), mPAP (r=0.339, p<0.001), PVR (r=0.431, p<0.001), and pSO2 (r=-0.243, p=0.011). Among echocardiographic measures, RI correlated with TRV (r=0.377, p<0.001) and echo sPAP (r=0.359, p<0.001). In multivariable analysis, RI (OR:1.032, p=0.003) and NT-proBNP (OR:1.004, p=0.049) remained significant predictors of PH. ROC analysis demonstrated the moderate predictive power for RI (AUC=0.806, p<0.001), with 76.4% sensitivity and 78.5% specificity at a cut-off of 232.05. CONCLUSION RI is a valuable parameter for assessing pulmonary arterial stiffness and vascular tone in patients with PAH and CTEPH. Significant correlations were observed with key hemodynamic parameters, including PVR and mPAP. Additionally, RI demonstrated moderate predictive power for PH. These findings highlight the potential of RI as an independent marker of vascular health, providing direct insights into the pulmonary arterial bed.
Collapse
Affiliation(s)
- Mustafa Oğuz
- Department of Cardiology, Sultan II. Abdulhamid Han Training and Research Hospital, Tibbiye Street, Istanbul 34668, Turkey.
| | - Mehmet Uzun
- Department of Cardiology, Sultan II. Abdulhamid Han Training and Research Hospital, Tibbiye Street, Istanbul 34668, Turkey
| | - İrem Yılmaz
- Department of Cardiology, Sultan II. Abdulhamid Han Training and Research Hospital, Tibbiye Street, Istanbul 34668, Turkey
| | - Almina Erdem
- Department of Cardiology, Sultan II. Abdulhamid Han Training and Research Hospital, Tibbiye Street, Istanbul 34668, Turkey
| | - Murat Demirci
- Department of Cardiology, Marmara University Medical School, Istanbul, Turkey
| | - Nilüfer Ekşi Duran
- Department of Cardiology, Sultan II. Abdulhamid Han Training and Research Hospital, Tibbiye Street, Istanbul 34668, Turkey
| |
Collapse
|
13
|
Nguyen TTN, Lewis CV, Hidalgo DC, Posey JN, Jordan M, Porfilio TE, Grayck MR, Wright CJ, Delaney C, Nozik ES. A maternal hypoxia mouse model to study the effect of late gestational hypoxia on offspring lung outcomes. Front Physiol 2025; 16:1513703. [PMID: 40084179 PMCID: PMC11904635 DOI: 10.3389/fphys.2025.1513703] [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: 10/18/2024] [Accepted: 02/10/2025] [Indexed: 03/16/2025] Open
Abstract
Extremely preterm birth predisposes infants to bronchopulmonary dysplasia and associated pulmonary hypertension (PH). High altitude exposure during pregnancy has also been shown to worsen infant lung and pulmonary vascular outcomes. Animal models addressing the mechanisms for how maternal hypoxia impacts postnatal and adult lung and pulmonary vascular outcomes are lacking and development of a model to address this gap would enable new mechanistic studies. We hypothesize that late gestational hypoxia disrupts lung and pulmonary vascular development in the offspring, leading to abrupted lung development and PH in adulthood. Pregnant wild-type mice were exposed to hypobaric hypoxia at 505 mmHg, from day 16.5 of gestation until birth. Lung and pulmonary vascular outcomes were measured in juvenile and mature offspring. We found that late gestational hypoxia resulted in abrupted alveolar and pulmonary vascular development in juvenile offspring and that adult offspring showed persistent abrupted alveolar development as well as PH. This striking model will provide a new opportunity to determine mechanisms responsible for poor outcomes secondary to maternal hypoxia and assess important factors that increase susceptibility to adult diseases in former preterm infants.
Collapse
Affiliation(s)
- Thi-Tina N. Nguyen
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Pediatrics, Division of Pediatric Critical Care, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Caitlin V. Lewis
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Pediatrics, Division of Pediatric Critical Care, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Daniel Colon Hidalgo
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Medicine, Division of Pulmonary and Critical Care, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Janelle N. Posey
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Pediatrics, Division of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Mariah Jordan
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Pediatrics, Division of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Timothy E. Porfilio
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Pediatrics, Division of Pediatric Critical Care, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Maya R. Grayck
- Department of Pediatrics, Division of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Clyde J. Wright
- Department of Pediatrics, Division of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Cassidy Delaney
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Pediatrics, Division of Neonatology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Eva S. Nozik
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Pediatrics, Division of Pediatric Critical Care, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
14
|
Ramachandra AB, Sharma P, De Man R, Nikola F, Guerrera N, Doddaballapur P, Cavinato C, Choi R, Raredon MSB, Szafron JM, Zhuang ZW, Barnthaler T, Justet A, Akingbesote ND, Abu Hussein NS, Diggs L, Perry RJ, Adams TS, Singh I, Kaminski N, Yan X, Tellides G, Humphrey JD, Manning EP. Hypoxia-Induced Cardiopulmonary Remodeling and Recovery: Critical Roles of the Proximal Pulmonary Artery, Macrophages, and Exercise. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.15.638455. [PMID: 40027757 PMCID: PMC11870459 DOI: 10.1101/2025.02.15.638455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Hypoxemia impairs cardiopulmonary function. We investigated pulmonary artery remodeling in mice exposed to chronic hypoxia for up to five weeks and quantified associated changes in cardiac and lung function, without or with subsequent normoxic recovery in the absence or presence of exercise or pharmacological intervention. Hypoxia-induced stiffening of the proximal pulmonary artery stemmed primarily from remodeling of the adventitial collagen, which resulted in part from altered inter-cellular signaling associated with phenotypic changes in the mural smooth muscle cells and macrophages. Such stiffening appeared to precede and associate with both right ventricular and lung dysfunction, with changes emerging to similar degrees regardless of the age of onset of hypoxia during postnatal development. Key homeostatic target values of the wall mechanics were recovered by the pulmonary arteries with normoxic recovery while other values recovered only partially. Overall cardiopulmonary dysfunction due to hypoxia was similarly only partially reversible. Remodeling of the cardiopulmonary system due to hypoxia is a complex, multi-scale process that involves maladaptations of the proximal pulmonary artery.
Collapse
|
15
|
Buncha V, Lang L, Fopiano KA, Ilatovskaya DV, Kapuku G, Verin AD, Bagi Z. Endothelial cell-selective adhesion molecule deficiency exhibits increased pulmonary vascular resistance due to impaired endothelial nitric oxide signaling. Am J Physiol Heart Circ Physiol 2025; 328:H283-H293. [PMID: 39740345 DOI: 10.1152/ajpheart.00593.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/02/2024] [Accepted: 12/19/2024] [Indexed: 01/02/2025]
Abstract
Endothelial cell-selective adhesion molecule (ESAM) is a member of tight junction molecules, highly abundant in the heart and the lung, and plays a role in regulating endothelial cell permeability. We previously reported that mice with genetic ESAM deficiency (ESAM-/-) exhibit coronary microvascular dysfunction leading to the development of left ventricular diastolic dysfunction. Here, we hypothesize that ESAM-/- mice display impairments in the pulmonary vasculature, affecting the overall pulmonary vascular resistance (PVR). We utilized ESAM-/- mice and employed isolated, ventilated, and perfused whole lung preparation to assess PVR independently of cardiac function. PVR was assessed in response to stepwise increases in flow, and also in response to perfusion of the endothelium-dependent agonist, bradykinin, the thromboxane analog, U46619, and the nitric oxide (NO) donor sodium nitroprusside (SNP). We found that PVR, at every applied flow rate, is significantly elevated in ESAM-/- mice compared with WT mice. Bradykinin-induced reduction in PVR and U46619-induced increase in PVR were both diminished in ESAM-/- mice, whereas SNP-induced responses were similar in wild-type (WT) and ESAM-/- mice. Inhibition of NO synthase with N(ω)-nitro-l-arginine methyl ester increased agonist-induced PVR in WT but not in ESAM-/- mice. Pulmonary arteries isolated from ESAM-/- mice exhibited a reduced level of phospho-Ser473-Akt and phospho-Ser1177-eNOS. Furthermore, in human lung microvascular endothelial cells cultured under flow conditions, we found that siRNA-mediated knockdown of ESAM impaired fluid shear stress-induced endothelial cell alignment. Thus, we suggest that ESAM plays an important role in the endothelium-dependent, flow/shear stress- and vasoactive agonist-stimulated, and NO-mediated maintenance of PVR in mice.NEW & NOTEWORTHY Our study reveals a novel role for ESAM in contributing to the maintenance of pulmonary vascular resistance under normal physiological conditions. Employing mice with global genetic deficiency of ESAM and using isolated whole lung preparation, we show significant impairments in nitric oxide-mediated pulmonary artery function. In vitro cell culture studies demonstrate impaired fluid shear stress-induced cell alignment in human lung endothelial cells after siRNA-mediated ESAM knockdown.
Collapse
Affiliation(s)
- Vadym Buncha
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Liwei Lang
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Katie Anne Fopiano
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Daria V Ilatovskaya
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Gaston Kapuku
- Department of Medicine, Georgia Prevention Institute, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Alexander D Verin
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Zsolt Bagi
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| |
Collapse
|
16
|
Zhang JJ, Ye XR, Liu XS, Zhang HL, Qiao Q. Impact of sodium-glucose cotransporter-2 inhibitors on pulmonary vascular cell function and arterial remodeling. World J Cardiol 2025; 17:101491. [PMID: 39866213 PMCID: PMC11755123 DOI: 10.4330/wjc.v17.i1.101491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/02/2024] [Accepted: 12/17/2024] [Indexed: 01/21/2025] Open
Abstract
Sodium-glucose cotransporter-2 (SGLT-2) inhibitors represent a cutting-edge class of oral antidiabetic therapeutics that operate through selective inhibition of glucose reabsorption in proximal renal tubules, consequently augmenting urinary glucose excretion and attenuating blood glucose levels. Extensive clinical investigations have demonstrated their profound cardiovascular efficacy. Parallel basic science research has elucidated the mechanistic pathways through which diverse SGLT-2 inhibitors beneficially modulate pulmonary vascular cells and arterial remodeling. Specifically, these inhibitors exhibit promising potential in enhancing pulmonary vascular endothelial cell function, suppressing pulmonary smooth muscle cell proliferation and migration, reversing pulmonary arterial remodeling, and maintaining hemodynamic equilibrium. This comprehensive review synthesizes current literature to delineate the mechanisms by which SGLT-2 inhibitors enhance pulmonary vascular cell function and reverse pulmonary remodeling, thereby offering novel therapeutic perspectives for pulmonary vascular diseases.
Collapse
Affiliation(s)
- Jing-Jing Zhang
- Chinese Academy Medical Sciences, Fuwai Yunnan Hospital, Kunming 650000, Yunnan Province, China
- Kunming Medical University, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming 650000, Yunnan Province, China
| | - Xue-Rui Ye
- Chinese Academy Medical Sciences, Fuwai Yunnan Hospital, Kunming 650000, Yunnan Province, China
- Kunming Medical University, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming 650000, Yunnan Province, China
| | - Xue-Song Liu
- Department of Biochemistry, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Hao-Ling Zhang
- Department of Biomedical Science, Advanced Medical and Dental Institute, University Sains Malaysia, Penang 13200, Malaysia
| | - Qian Qiao
- Chinese Academy Medical Sciences, Fuwai Yunnan Hospital, Kunming 650000, Yunnan Province, China
- Kunming Medical University, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming 650000, Yunnan Province, China.
| |
Collapse
|
17
|
Farha S, Madden E, Trotter D, Zlojutro V, Kirkness JP, Fouras A, Erzurum S, Asosingh K. Quantification of the pulmonary vasculature in mice under chronic hypoxia with contrast-free pulmonary angiography in vivo imaging. J Appl Physiol (1985) 2025; 138:318-325. [PMID: 39717944 DOI: 10.1152/japplphysiol.00279.2024] [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: 04/15/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 12/25/2024] Open
Abstract
Innovative advancements in preclinical imaging have led to the development of cone-beam computed tomography (CBCT) combined with contrast-free pulmonary angiography (CFPA), a novel lung scanning technology capable of assessing lung function and pulmonary vascular morphology. This cutting-edge approach integrates CBCT to provide detailed quantification of the pulmonary vascular tree. The application of this technique to image and quantify changes in the pulmonary vascular tree of mice exposed to chronic hypoxia has not been investigated. In this study, we investigated the feasibility of utilizing CFPA for imaging changes in the murine lung vascular bed under chronic hypoxia and assessed whether vascular metrics correlate with hematologic parameters and/or right ventricular pressure and mass. Our results revealed a significant increase in hemoglobin and total pulmonary vascular blood volume, as well as total pulmonary vessel length following exposure to chronic hypoxia. The pulmonary vascular blood volume and total vessel length strongly correlated with hemoglobin. There was also an increase in pulmonary arterial pressure and right ventricular mass under hypoxia that was linked to the hematological response and the changes in the pulmonary vascular bed. These findings highlight the application of preclinical CBCT and CFPA imaging as a valuable tool for visual and quantitative analysis of the pulmonary vasculature in preclinical models of chronic hypoxia and its potential use in investigating other pulmonary vasculopathies.NEW & NOTEWORTHY Cone-beam computed tomography (CBCT) combined with contrast-free pulmonary angiography (CFPA) is a novel lung scanning technology capable of assessing pulmonary vascular morphology in vivo in murine models.
Collapse
Affiliation(s)
- Samar Farha
- Pulmonary Medicine, Cleveland Clinic, Cleveland, Ohio, United States
| | - Evan Madden
- Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, United States
| | - Dylan Trotter
- Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, United States
| | - Violetta Zlojutro
- Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, United States
| | - Jason P Kirkness
- Miller School of Medicine, University of Miami, Miami, Florida, United States
| | | | - Serpil Erzurum
- Pulmonary Medicine, Cleveland Clinic, Cleveland, Ohio, United States
- Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, United States
| | - Kewal Asosingh
- Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, United States
| |
Collapse
|
18
|
Zhu H, Li C, Hu F, Wu L, Wu L, Zhou M, Liu W, Dai A. RO4929097 inhibits NICD3 to alleviate pulmonary hypertension via blocking Notch3/HIF-2α/FoxM1 signaling pathway. In Vitro Cell Dev Biol Anim 2025; 61:107-116. [PMID: 39621175 DOI: 10.1007/s11626-024-00976-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 08/04/2024] [Indexed: 02/06/2025]
Abstract
Pulmonary hypertension (PH) is a condition in which the smooth muscle cells (SMCs) in the pulmonary arteries multiply excessively, causing the arteries to narrow. This can ultimately result in right heart failure and premature death. Notch3 is an important factor involved in pulmonary vascular remodeling in PH. RO4929097, as a γ-secretase inhibitor that inhibits Notch3 signaling pathway, may be a potential drug for the treatment of PH, but its feasibility and related mechanism of action need to be further investigated. In vitro modeling by hypoxic incubation of human pulmonary artery SMCs (HPASMCs). RO4929097 and plasmids including overexpression-NICD3 (oe-NICD3) and NICD3 small interfering RNA (siRNA) were used to alter the expression of NICD3, and HIF-2α inhibitor PT-2385 was used to alter the expression of HIF-2α. Western blot, EdU incorporation assay was used to investigate the alteration of NICD3, HIF-2α, FoxM1 protein expression, and cell proliferation. The severity of PH in rats was assessed by measuring the weight ratio of right ventricle (RV) to left ventricle (LV) and septum (S) (RV/[LV + S]) and hematoxylin-eosin (H&E) staining of lung tissues in a hypoxia-induced PH rat model. We first determined that hypoxia induction for 48 h had the strongest induction of NICD3 and Notch3 in HPASMCs, and the strongest inhibition by 10 μM RO4929097. Treatment of HPASMCs under hypoxic conditions with RO4929097 inhibited hypoxia-induced expression of NICD3, HIF-2α, FoxM1, and proliferation of HPASMCs. The inhibitory effect of RO4929097 was reversed after overexpression of NICD3 in HPASMCs. Further, we found that PT-2385 reversed the promotional effect of overexpression of NICD3 on the proliferation of HPASMCs. In vivo experiments, hypoxia-induced PH rats treated with RO4929097 showed a reduction in right ventricular hypertrophy index (RVHI) and a return to normal pulmonary artery morphology, indicating a reduction in the severity of PH. Our data suggest that RO4929097 regulates the Notch3/HIF-2α/FoxM1 signaling pathway by inhibiting the expression of NICD3, thereby inhibiting hypoxia-induced proliferation of HPASMCs. In vivo experiments also confirmed that RO4929097 could alleviate PH as a potential therapeutic strategy.
Collapse
Affiliation(s)
- Hao Zhu
- Department of Respiratory and Critical Care Medicine, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, China
| | - Cheng Li
- Department of Respiratory and Critical Care Medicine, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, China
| | - Fang Hu
- Department of Respiratory and Critical Care Medicine, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, China
| | - Lifu Wu
- Department of Respiratory and Critical Care Medicine, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, China
| | - Ling Wu
- Department of Respiratory and Critical Care Medicine, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, China
| | - Meihua Zhou
- Department of Respiratory and Critical Care Medicine, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, China
| | - Wei Liu
- Department of Respiratory and Critical Care Medicine, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, China.
| | - Aiguo Dai
- Department of Respiratory and Critical Care Medicine, Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, 410016, Hunan, China.
- Department of Respiratory Diseases, Medical School, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| |
Collapse
|
19
|
Shirota Y, Ohmori S, Engel JD, Moriguchi T. GATA2 participates in protection against hypoxia-induced pulmonary vascular remodeling. PLoS One 2024; 19:e0315446. [PMID: 39739870 DOI: 10.1371/journal.pone.0315446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/25/2024] [Indexed: 01/02/2025] Open
Abstract
The vascular endothelium is vital for cardio-pulmonary homeostasis and, thus, plays a crucial role in preventing life-threatening lung diseases. The transcription factor GATA2 is essential for hematopoiesis and maintaining vascular integrity. Heterozygous mutations in GATA2 can lead to a primary immunodeficiency syndrome with pulmonary manifestations. Some GATA2 haploinsufficient patients develop pulmonary hypertension (PH), characterized by vascular remodeling and occlusion of small pulmonary arteries. However, the mechanism underlying pulmonary vascular remodeling in GATA2 haploinsufficient patients remain unclear. To understand how GATA2 deficiency affects pulmonary artery homeostasis, we applied a chronic hypoxia-mediated PH model using inducible systemic Gata2 conditionally deficient (G2-CKO) mice. The G2-CKO mice exhibited augmented pulmonary vascular remodeling, with enhanced α-smooth muscle actin accumulation and increased apoptotic cells in the vascular wall upon chronic hypoxia. Transcript analysis and chromatin immunoprecipitation assays using mouse pulmonary vascular endothelial cells revealed that GATA2 directly regulates the expression of G6pdx (a crucial cytoprotective enzyme) and Bmp4 (a growth factor that mediates vascular homeostasis). These results suggest that GATA2-deficient lungs are vulnerable to the hypoxic stress due to a diminished cellular protective response, making G2-CKO mice more prone to vascular remodeling upon chronic hypoxia. These findings provide insights into the mechanisms underlying GATA2-haploinsufficiency-related pulmonary hypertension.
Collapse
Affiliation(s)
- Yuko Shirota
- Division of Hematology and Rheumatology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Shin'ya Ohmori
- Department of Pharmacy, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki, Japan
| | - James Douglas Engel
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, United States of America
| | - Takashi Moriguchi
- Division of Medical Biochemistry, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| |
Collapse
|
20
|
Williams J, Iheagwam FN, Maroney SP, Schmitt LR, Brown RD, Krafsur GM, Frid MG, McCabe MC, Gandjeva A, Williams KJ, Luyendyk JP, Saviola AJ, Tuder RM, Stenmark K, Hansen KC. A bovine model of hypoxia-induced pulmonary hypertension reveals a gradient of immune and matrisome response with a complement signature found in circulation. Am J Physiol Cell Physiol 2024; 327:C1666-C1680. [PMID: 39495247 PMCID: PMC11684870 DOI: 10.1152/ajpcell.00274.2024] [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: 04/29/2024] [Revised: 08/19/2024] [Accepted: 09/11/2024] [Indexed: 11/05/2024]
Abstract
Pulmonary hypertension (PH) is a progressive vascular disease characterized by vascular remodeling, stiffening, and luminal obstruction, driven by dysregulated cell proliferation, inflammation, and extracellular matrix (ECM) alterations. Despite the recognized contribution of ECM dysregulation to PH pathogenesis, the precise molecular alterations in the matrisome remain poorly understood. In this study, we employed a matrisome-focused proteomics approach to map the protein composition in a young bovine calf model of acute hypoxia-induced PH. Our findings reveal distinct alterations in the matrisome along the pulmonary vascular axis, with the most prominent changes observed in the main pulmonary artery. Key alterations included a strong immune response and wound repair signature, characterized by increased levels of complement components, coagulation cascade proteins, and provisional matrix markers. In addition, we observed upregulation of ECM-modifying enzymes, growth factors, and core ECM proteins implicated in vascular stiffening, such as collagens, periostin, tenascin-C, and fibrin(ogen). Notably, these alterations correlated with increased mean pulmonary arterial pressure and vascular remodeling. In the plasma, we identified increased levels of complement components, indicating a systemic inflammatory response accompanying the vascular remodeling. Our findings shed light on the dynamic matrisome remodeling in early-stage PH, implicating a wound-healing trajectory with distinct patterns from the main pulmonary artery to the distal vasculature. This study provides novel insights into the immune cell infiltration and matrisome alterations associated with PH pathogenesis and highlights potential biomarkers and therapeutic targets within the matrisome landscape.NEW & NOTEWORTHY Extensive immune cell infiltration and matrisome alterations associated with hypoxia-induced pulmonary hypertension in a large mammal model. Matrisome components correlate with increased resistance to identify candidate alterations that drive biomechanical manifestations of the disease.
Collapse
Affiliation(s)
- Jason Williams
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, Colorado, United States
| | - Franklyn N Iheagwam
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, Colorado, United States
| | - Sean P Maroney
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, Colorado, United States
| | - Lauren R Schmitt
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, Colorado, United States
| | - R Dale Brown
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Greta M Krafsur
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Maria G Frid
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Maxwell C McCabe
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, Colorado, United States
| | - Aneta Gandjeva
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Kurt J Williams
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan, United States
| | - James P Luyendyk
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan, United States
| | - Anthony J Saviola
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, Colorado, United States
| | - Rubin M Tuder
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Program in Translational Lung Research, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver, Denver, Colorado, United States
| | - Kurt Stenmark
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Colorado, Aurora, Colorado, United States
| |
Collapse
|
21
|
Azaredo Raposo M, Inácio Cazeiro D, Guimarães T, Lousada N, Freitas C, Brito J, Martins S, Resende C, Dorfmüller P, Luís R, Moreira S, Alves da Silva P, Moita L, Oliveira M, Pinto FJ, Plácido R. Pulmonary arterial hypertension: Navigating the pathways of progress in diagnosis, treatment, and patient care. Rev Port Cardiol 2024; 43:699-719. [PMID: 38972452 DOI: 10.1016/j.repc.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/01/2024] [Accepted: 03/10/2024] [Indexed: 07/09/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a form of precapillary pulmonary hypertension caused by a complex process of endothelial dysfunction and vascular remodeling. If left untreated, this progressive disease presents with symptoms of incapacitating fatigue causing marked loss of quality of life, eventually culminating in right ventricular failure and death. Patient management is complex and based on accurate diagnosis, risk stratification, and treatment initiation, with close monitoring of response and disease progression. Understanding the underlying pathophysiology has enabled the development of multiple drugs directed at different targets in the pathological chain. Vasodilator therapy has been the mainstay approach for the last few years, significantly improving quality of life, functional status, and survival. Recent advances in therapies targeting dysfunctional pathways beyond endothelial dysfunction may address the fundamental processes underlying the disease, raising the prospect of increasingly effective options for this high-risk group of patients with a historically poor prognosis.
Collapse
Affiliation(s)
- Miguel Azaredo Raposo
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, CAML, CCUL, Faculty of Medicine, Lisbon, Portugal
| | - Daniel Inácio Cazeiro
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, CAML, CCUL, Faculty of Medicine, Lisbon, Portugal
| | - Tatiana Guimarães
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, CAML, CCUL, Faculty of Medicine, Lisbon, Portugal
| | - Nuno Lousada
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, CAML, CCUL, Faculty of Medicine, Lisbon, Portugal
| | - Céline Freitas
- Association for Research and Development of Faculty of Medicine (AIDFM), Cardiovascular Research Support Unit (GAIC), Lisbon, Portugal
| | - Joana Brito
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, CAML, CCUL, Faculty of Medicine, Lisbon, Portugal
| | - Susana Martins
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, CAML, CCUL, Faculty of Medicine, Lisbon, Portugal
| | - Catarina Resende
- Rheumatology Department, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
| | - Peter Dorfmüller
- Department of Pathology, University Hospital of Giessen and Marburg, Giessen, Germany; Institute for Lung Health, Giessen, Germany
| | - Rita Luís
- Pathology Department, Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal; Pathology Department, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal; Pathology Institute, Faculdade Medicina Universidade Lisboa, Lisbon, Portugal
| | - Susana Moreira
- Pulmonology Department, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
| | - Pedro Alves da Silva
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, CAML, CCUL, Faculty of Medicine, Lisbon, Portugal
| | - Luís Moita
- Innate Immunity and Inflammation Laboratory, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | | | - Fausto J Pinto
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, CAML, CCUL, Faculty of Medicine, Lisbon, Portugal
| | - Rui Plácido
- Cardiology Department, Centro Hospitalar Universitário Lisboa Norte, CAML, CCUL, Faculty of Medicine, Lisbon, Portugal; Cardiology Department, CUF Tejo, Lisbon, Portugal.
| |
Collapse
|
22
|
Suzuki Y, Kawasaki T, Tatsumi K, Okaya T, Sato S, Shimada A, Misawa T, Hatano R, Morimoto C, Kasuya Y, Hasegawa Y, Ohara O, Suzuki T. Transcriptome Analysis of Fibroblasts in Hypoxia-Induced Vascular Remodeling: Functional Roles of CD26/DPP4. Int J Mol Sci 2024; 25:12599. [PMID: 39684311 DOI: 10.3390/ijms252312599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/13/2024] [Accepted: 11/17/2024] [Indexed: 12/18/2024] Open
Abstract
In hypoxic pulmonary hypertension (PH), pulmonary vascular remodeling is characterized by the emergence of activated adventitial fibroblasts, leading to medial smooth muscle hyperplasia. Previous studies have suggested that CD26/dipeptidyl peptidase-4 (DPP4) plays a crucial role in the pathobiological processes in lung diseases. However, its role in pulmonary fibroblasts in hypoxic PH remains unknown. Therefore, we aimed to clarify the mechanistic role of CD26/DPP4 in lung fibroblasts in hypoxic PH. Dpp4 knockout (Dpp4 KO) and wild-type (WT) mice were exposed to hypoxia for 4 weeks. The degree of PH severity and medial wall thickness was augmented in Dpp4 KO mice compared with that in WT mice, suggesting that CD26/DPP4 plays a suppressive role in the development of hypoxic PH. Transcriptome analysis of human lung fibroblasts cultured under hypoxic conditions revealed that TGFB2, TGFB3, and TGFA were all upregulated as differentially expressed genes after DPP4 knockdown with small interfering RNA treatment. These results suggest that CD26/DPP4 plays a suppressive role in TGFβ signal-regulated fibroblast activation under hypoxic conditions. Therefore, CD26/DPP4 may be a potential therapeutic target in patients with PH associated with chronic hypoxia.
Collapse
Affiliation(s)
- Yuri Suzuki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Takeshi Kawasaki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Tadasu Okaya
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Shun Sato
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Ayako Shimada
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Tomoko Misawa
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
- Synergy Institute for Futuristic Mucosal Vaccine Research and Development, Chiba University, Chiba 260-8670, Japan
| | - Ryo Hatano
- Department of Therapy Development and Innovation for Immune disorders and Cancers, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Chikao Morimoto
- Department of Therapy Development and Innovation for Immune disorders and Cancers, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Yoshitoshi Kasuya
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
- Department of Pharmacology, Faculty of Pharmacy, Juntendo University, Chiba 279-0013, Japan
| | - Yoshinori Hasegawa
- Department of Applied Genomics, Kazusa DNA Research Institute, Chiba 292-0818, Japan
| | - Osamu Ohara
- Department of Applied Genomics, Kazusa DNA Research Institute, Chiba 292-0818, Japan
| | - Takuji Suzuki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
- Synergy Institute for Futuristic Mucosal Vaccine Research and Development, Chiba University, Chiba 260-8670, Japan
| |
Collapse
|
23
|
Laban H, Siegmund S, Schlereth K, Trogisch FA, Ablieh A, Brandenburg L, Weigert A, De La Torre C, Mogler C, Hecker M, Kuebler WM, Korff T. Nuclear factor of activated T-cells 5 is indispensable for a balanced adaptive transcriptional response of lung endothelial cells to hypoxia. Cardiovasc Res 2024; 120:1590-1606. [PMID: 39107245 DOI: 10.1093/cvr/cvae151] [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: 04/04/2024] [Revised: 05/07/2024] [Accepted: 06/01/2024] [Indexed: 08/09/2024] Open
Abstract
AIMS Chronic hypoxia causes detrimental structural alterations in the lung, which may cause pulmonary hypertension and are partially mediated by the endothelium. While its relevance for the development of hypoxia-associated lung diseases is well known, determinants controlling the initial adaptation of the lung endothelium to hypoxia remain largely unexplored. METHODS AND RESULTS We revealed that hypoxia activates the transcription factor nuclear factor of activated T-cells 5 (NFAT5) and studied its regulatory function in murine lung endothelial cells (MLECs). EC-specific knockout of Nfat5 (Nfat5(EC)-/-) in mice exposed to normobaric hypoxia (10% O2) for 21 days promoted vascular fibrosis and aggravated the increase in pulmonary right ventricular systolic pressure as well as right ventricular dysfunction as compared with control mice. Microarray- and single-cell RNA-sequencing-based analyses revealed an impaired growth factor-, energy-, and protein-metabolism-associated gene expression in Nfat5-deficient MLEC after exposure to hypoxia for 7 days. Specifically, loss of NFAT5 boosted the expression and release of platelet-derived growth factor B (Pdgfb)-a hypoxia-inducible factor 1 alpha (HIF1α)-regulated driver of vascular smooth muscle cell (VSMC) growth-in capillary MLEC of hypoxia-exposed Nfat5(EC)-/- mice, which was accompanied by intensified VSMC coverage of distal pulmonary arteries. CONCLUSION Collectively, our study shows that early and transient subpopulation-specific responses of MLEC to hypoxia may determine the degree of organ dysfunction in later stages. In this context, NFAT5 acts as a protective transcription factor required to rapidly adjust the endothelial transcriptome to cope with hypoxia. Specifically, NFAT5 restricts HIF1α-mediated Pdgfb expression and consequently limits muscularization and resistance of the pulmonary vasculature.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Adaptation, Physiological
- Cell Hypoxia
- Cells, Cultured
- Disease Models, Animal
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Gene Expression Regulation
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/physiopathology
- Hypoxia/metabolism
- Hypoxia/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Lung/metabolism
- Lung/blood supply
- Lung/pathology
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/physiopathology
- Signal Transduction
- Transcription Factors/metabolism
- Transcription Factors/genetics
- Transcription, Genetic
- Vascular Remodeling
- Ventricular Function, Right
Collapse
Affiliation(s)
- Hebatullah Laban
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology, Heidelberg University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung e.V. (DZHK), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Sophia Siegmund
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology, Heidelberg University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| | - Katharina Schlereth
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| | - Felix A Trogisch
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
- Department of Cardiovascular Physiology and Cardiac Imaging Center, Core Facility Platform Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Alia Ablieh
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology, Heidelberg University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| | - Lennart Brandenburg
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology, Heidelberg University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| | - Andreas Weigert
- Institute of Biochemistry I Pathobiochemistry, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany
| | - Carolina De La Torre
- NGS Core Facility, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Carolin Mogler
- Institute of Pathology, School of Medicine, Technical University Munich, Munich, Germany
| | - Markus Hecker
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology, Heidelberg University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Thomas Korff
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology, Heidelberg University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| |
Collapse
|
24
|
Niihori M, James J, Varghese MV, McClain N, Lawal OS, Philip RC, Baggett BK, Goncharov DA, de Jesus Perez V, Goncharova EA, Rafikov R, Rafikova O. Mitochondria as a primary determinant of angiogenic modality in pulmonary arterial hypertension. J Exp Med 2024; 221:e20231568. [PMID: 39320470 PMCID: PMC11452743 DOI: 10.1084/jem.20231568] [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: 08/31/2023] [Revised: 03/27/2024] [Accepted: 08/19/2024] [Indexed: 09/26/2024] Open
Abstract
Impaired pulmonary angiogenesis plays a pivotal role in the progression of pulmonary arterial hypertension (PAH) and patient mortality, yet the molecular mechanisms driving this process remain enigmatic. Our study uncovered a striking connection between mitochondrial dysfunction (MD), caused by a humanized mutation in the NFU1 gene, and severely disrupted pulmonary angiogenesis in adult lungs. Restoring the bioavailability of the NFU1 downstream target, lipoic acid (LA), alleviated MD and angiogenic deficiency and rescued the progressive PAH phenotype in the NFU1G206C model. Notably, significant NFU1 expression and signaling insufficiencies were also identified in idiopathic PAH (iPAH) patients' lungs, emphasizing this study's relevance beyond NFU1 mutation cases. The remarkable improvement in mitochondrial function of PAH patient-derived pulmonary artery endothelial cells (PAECs) following LA supplementation introduces LA as a potential therapeutic approach. In conclusion, this study unveils a novel role for MD in dysregulated pulmonary angiogenesis and PAH manifestation, emphasizing the need to correct MD in PAH patients with unrecognized NFU1/LA deficiency.
Collapse
Affiliation(s)
- Maki Niihori
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, IN, USA
| | - Joel James
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, IN, USA
| | - Mathews V. Varghese
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, IN, USA
| | - Nolan McClain
- Department of Medicine, University of Arizona, Tucson, AZ, USA
| | - Odunayo Susan Lawal
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, IN, USA
| | - Rohit C. Philip
- Department of Electrical and Computer Engineering, University of Arizona College of Engineering, Tucson, AZ, USA
- Department of Medical Imaging, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Brenda K. Baggett
- The University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| | - Dmitry A. Goncharov
- Division of Pulmonary, Critical Care and Sleep Medicine, Lung Center, University of California, Davis School of Medicine, Davis, CA, USA
| | - Vinicio de Jesus Perez
- Division of Pulmonary and Critical Care Medicine, Stanford University Medical Center, Stanford, CA, USA
| | - Elena A. Goncharova
- Division of Pulmonary, Critical Care and Sleep Medicine, Lung Center, University of California, Davis School of Medicine, Davis, CA, USA
| | - Ruslan Rafikov
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, IN, USA
| | - Olga Rafikova
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, IN, USA
| |
Collapse
|
25
|
Wang H, Gao Y, Bai J, Liu H, Li Y, Zhang J, Ma C, Zhao X, Zhang L, Wan K, Zhu D. CircLMBR1 inhibits phenotypic transformation of hypoxia-induced pulmonary artery smooth muscle via the splicing factor PUF60. Eur J Pharmacol 2024; 980:176855. [PMID: 39059570 DOI: 10.1016/j.ejphar.2024.176855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/04/2024] [Accepted: 07/24/2024] [Indexed: 07/28/2024]
Abstract
Phenotypic transformation of pulmonary artery smooth muscle cells (PASMCs) contributes to vascular remodeling in hypoxic pulmonary hypertension (PH). Recent studies have suggested that circular RNAs (circRNAs) may play important roles in the vascular remodeling of hypoxia-induced PH. However, whether circRNAs cause pulmonary vascular remodeling by regulating the phenotypic transformation in PH has not been investigated. Microarray and RT-qPCR analysis identified that circLMBR1, a novel circRNA, decreased in mouse lung tissues of the hypoxia-SU5416 PH model, as well as in human PASMCs and mouse PASMCs exposed to hypoxia. Overexpression of circLMBR1 in the Semaxinib (SU5416) mouse model ameliorated hypoxia-induced PH and vascular remodeling in the lungs. Notably, circLMBR1 was mainly distributed in the nucleus and bound to the splicing factor PUF60. CircLMBR1 suppressed the phenotypic transformation of human PASMCs and vascular remodeling by inhibiting PUF60 expression. Furthermore, we identified U2AF65 as the downstream regulatory factor of PUF60. U2AF65 directly interacted with the pre-mRNA of the contractile phenotype marker smooth muscle protein 22-α (SM22α) and inhibited its splicing. Meanwhile, hypoxia exposure increased the formation of the PUF60-U2AF65 complex, thereby inhibiting SM22α production and inducing the transition of human PASMCs from a contractile phenotype to a synthetic phenotype. Overall, our results verified the important role of circLMBR1 in the pathological process of PH. We also proposed a new circLMBR1/PUF60-U2AF65/pre-SM22α pathway that could regulate the phenotypic transformation and proliferation of human PASMCs. This study may provide new perspectives for the diagnosis and treatment of PH.
Collapse
MESH Headings
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/drug effects
- Animals
- Humans
- Mice
- Vascular Remodeling/drug effects
- Vascular Remodeling/genetics
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Phenotype
- RNA, Circular/genetics
- RNA, Circular/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Male
- Splicing Factor U2AF/genetics
- Splicing Factor U2AF/metabolism
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/genetics
- Hypoxia/metabolism
- Hypoxia/genetics
- Mice, Inbred C57BL
- Cell Hypoxia
- Indoles/pharmacology
- Pyrroles
Collapse
Affiliation(s)
- Hongdan Wang
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Pharmacy, Harbin Medical University, Harbin, 150081, PR China
| | - Yupei Gao
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Pharmacy, Harbin Medical University, Harbin, 150081, PR China
| | - June Bai
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Pharmacy, Harbin Medical University, Harbin, 150081, PR China
| | - Huiyu Liu
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Pharmacy, Harbin Medical University, Harbin, 150081, PR China
| | - Yiying Li
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Pharmacy, Harbin Medical University, Harbin, 150081, PR China
| | - Junting Zhang
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Pharmacy, Harbin Medical University, Harbin, 150081, PR China
| | - Cui Ma
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, 163319, PR China
| | - Xijuan Zhao
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, 163319, PR China
| | - Lixin Zhang
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, 163319, PR China
| | - Kuiyu Wan
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China
| | - Daling Zhu
- Central Laboratory of Harbin Medical University (Daqing), Daqing, 163319, PR China; College of Pharmacy, Harbin Medical University, Harbin, 150081, PR China; Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, Harbin Medical University, Harbin, 150081, PR China.
| |
Collapse
|
26
|
Peng S, Liang Y, Zhu H, Wang Y, Li Y, Zhao Z, Li Y, Zhuang R, Huang L, Zhang X, Guo Z. A nitroreductase responsive probe for early diagnosis of pulmonary fibrosis disease. Redox Biol 2024; 75:103294. [PMID: 39096854 PMCID: PMC11345524 DOI: 10.1016/j.redox.2024.103294] [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: 06/20/2024] [Revised: 07/17/2024] [Accepted: 07/28/2024] [Indexed: 08/05/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a serious interstitial lung disease. However, the definitive diagnosis of IPF is impeded by the limited capabilities of current diagnostic methods, which may fail to capture the optimal timing for treatment. The main goal of this study is to determine the feasibility of a nitroreductase (NTR) responsive probe, 18F-NCRP, for early detection and deterioration monitoring of IPF. 18F-NCRP was obtained with high radiochemical purity (>95 %). BLM-injured mice were established by intratracheal instillation with bleomycin (BLM) and characterized through histological analysis. Longitudinal PET/CT imaging, biodistribution study and in vitro autoradiography were performed. The correlations between the uptake of 18F-NCRP and mean lung density (tested by CT), as well as histopathological characteristics were analyzed. In PET imaging study, 18F-NCRP exhibited promising efficacy in monitoring the progression of IPF, which was earlier than CT. The ratio of uptake in BLM-injured lung to control lung increased from 1.4-fold on D15 to 2.2-fold on D22. Biodistribution data showed a significant lung uptake of 18F-NCRP in BLM-injured mice. There was a strong positive correlation between the 18F-NCRP uptake in the BLM-injured lungs and the histopathological characteristics. Given that, 18F-NCRP PET imaging of NTR, a promising biomarker for investigating the underlying pathogenic mechanism of IPF, is attainable as well as desirable, which might lay the foundation for establishing an NTR-targeted imaging evaluation system of IPF.
Collapse
Affiliation(s)
- Shilan Peng
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang an Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China
| | - Yuanyuan Liang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang an Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China
| | - Haotian Zhu
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang an Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China
| | - Yike Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang an Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China
| | - Yun Li
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang an Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China
| | - Zuoquan Zhao
- Theranostics and Translational Research Center, Institute of Clinical Medicine, Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Yesen Li
- Department of Nuclear Medicine & Minnan PET Center, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, China
| | - Rongqiang Zhuang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang an Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China
| | - Lumei Huang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang an Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China.
| | - Xianzhong Zhang
- Theranostics and Translational Research Center, Institute of Clinical Medicine, Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| | - Zhide Guo
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang an Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Rd, Xiamen, 361102, China.
| |
Collapse
|
27
|
Wan JJ, Yi J, Wang FY, Li X, Zhang C, Song L, Dai AG. Role of mitophagy in pulmonary hypertension: Targeting the mechanism and pharmacological intervention. Mitochondrion 2024; 78:101928. [PMID: 38992857 DOI: 10.1016/j.mito.2024.101928] [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: 11/20/2023] [Revised: 05/29/2024] [Accepted: 07/07/2024] [Indexed: 07/13/2024]
Abstract
Mitophagy, a crucial pathway in eukaryotic cells, selectively eliminates dysfunctional mitochondria, thereby maintaining cellular homeostasis via mitochondrial quality control. Pulmonary hypertension (PH) refers to a pathological condition where pulmonary arterial pressure is abnormally elevated due to various reasons, and the underlying pathogenesis remains elusive. This article examines the molecular mechanisms underlying mitophagy, emphasizing its role in PH and the progress in elucidating related molecular signaling pathways. Additionally, it highlights current drug regulatory pathways, aiming to provide novel insights into the prevention and treatment of pulmonary hypertension.
Collapse
Affiliation(s)
- Jia-Jing Wan
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Jian Yi
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410021, Hunan, China
| | - Fei-Ying Wang
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Xia Li
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Chao Zhang
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Lan Song
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Ai-Guo Dai
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China; Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410021, Hunan, China.
| |
Collapse
|
28
|
de la Cruz-Thea B, Natali L, Ho-Xuan H, Bruckmann A, Coll-Bonfill N, Strieder N, Peinado VI, Meister G, Musri MM. Differentiation and Growth-Arrest-Related lncRNA ( DAGAR): Initial Characterization in Human Smooth Muscle and Fibroblast Cells. Int J Mol Sci 2024; 25:9497. [PMID: 39273443 PMCID: PMC11394763 DOI: 10.3390/ijms25179497] [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: 07/26/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Vascular smooth muscle cells (SMCs) can transition between a quiescent contractile or "differentiated" phenotype and a "proliferative-dedifferentiated" phenotype in response to environmental cues, similar to what in occurs in the wound healing process observed in fibroblasts. When dysregulated, these processes contribute to the development of various lung and cardiovascular diseases such as Chronic Obstructive Pulmonary Disease (COPD). Long non-coding RNAs (lncRNAs) have emerged as key modulators of SMC differentiation and phenotypic changes. In this study, we examined the expression of lncRNAs in primary human pulmonary artery SMCs (hPASMCs) during cell-to-cell contact-induced SMC differentiation. We discovered a novel lncRNA, which we named Differentiation And Growth Arrest-Related lncRNA (DAGAR) that was significantly upregulated in the quiescent phenotype with respect to proliferative SMCs and in cell-cycle-arrested MRC5 lung fibroblasts. We demonstrated that DAGAR expression is essential for SMC quiescence and its knockdown hinders SMC differentiation. The treatment of quiescent SMCs with the pro-inflammatory cytokine Tumor Necrosis Factor (TNF), a known inducer of SMC dedifferentiation and proliferation, elicited DAGAR downregulation. Consistent with this, we observed diminished DAGAR expression in pulmonary arteries from COPD patients compared to non-smoker controls. Through pulldown experiments followed by mass spectrometry analysis, we identified several proteins that interact with DAGAR that are related to cell differentiation, the cell cycle, cytoskeleton organization, iron metabolism, and the N-6-Methyladenosine (m6A) machinery. In conclusion, our findings highlight DAGAR as a novel lncRNA that plays a crucial role in the regulation of cell proliferation and SMC differentiation. This paper underscores the potential significance of DAGAR in SMC and fibroblast physiology in health and disease.
Collapse
MESH Headings
- Humans
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Fibroblasts/metabolism
- Cell Differentiation/genetics
- Myocytes, Smooth Muscle/metabolism
- Cell Proliferation/genetics
- Pulmonary Artery/metabolism
- Pulmonary Artery/cytology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/cytology
- Pulmonary Disease, Chronic Obstructive/metabolism
- Pulmonary Disease, Chronic Obstructive/genetics
- Pulmonary Disease, Chronic Obstructive/pathology
- Cells, Cultured
Collapse
Affiliation(s)
- Benjamin de la Cruz-Thea
- Mercedes and Martin Ferreyra Medical Research Institute, National Council for Scientific and Technical Research, National University of Córdoba (INIMEC-CONICET-UNC), Córdoba 5016, Argentina
| | - Lautaro Natali
- Mercedes and Martin Ferreyra Medical Research Institute, National Council for Scientific and Technical Research, National University of Córdoba (INIMEC-CONICET-UNC), Córdoba 5016, Argentina
| | - Hung Ho-Xuan
- Regensburg Center for Biochemistry (RCB), Laboratory for RNA Biology, University of Regensburg, 93053 Regensburg, Germany
| | - Astrid Bruckmann
- Regensburg Center for Biochemistry (RCB), Laboratory for RNA Biology, University of Regensburg, 93053 Regensburg, Germany
| | - Núria Coll-Bonfill
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, MO 63104, USA
| | - Nicholas Strieder
- NGS-Core, LIT-Leibniz-Institute for Immunotherapy, 93053 Regensburg, Germany
| | - Víctor I Peinado
- Department of Experimental Pathology, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
- Department of Pulmonary Medicine, Hospital Clínic, Biomedical Research Institut August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain
- Biomedical Research Networking Center in Respiratory Diseases (CIBERES), 28029 Madrid, Spain
| | - Gunter Meister
- Regensburg Center for Biochemistry (RCB), Laboratory for RNA Biology, University of Regensburg, 93053 Regensburg, Germany
| | - Melina M Musri
- Mercedes and Martin Ferreyra Medical Research Institute, National Council for Scientific and Technical Research, National University of Córdoba (INIMEC-CONICET-UNC), Córdoba 5016, Argentina
| |
Collapse
|
29
|
Zhang Y, Wang J, Zhang M, Li X, Zhang F, Zhou M, Yang K, Chen W, Ding H, Tan X, Zhang Q, Qiao Z. Study on the Regulatory Mechanism of the PDK1-Mediated TGF-β/Smad Signaling Pathway in Hypoxia-Induced Yak Lungs. Animals (Basel) 2024; 14:2422. [PMID: 39199957 PMCID: PMC11350703 DOI: 10.3390/ani14162422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/10/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024] Open
Abstract
The aim of this study was to investigate the effects of hypoxia-induced phenotype, glucose metabolism, ROS levels, and the PDK1-mediated regulation of TGF-β/Smad signaling in yellow cattles, yaks, and those overexpressing PDK1 PASMCs using growth curves, flow cytometry, scratch experiments, glucose and lactic acid assays, RT-qPCR, and Western blotting. The results showed that hypoxia significantly promoted proliferation, migration, antiapoptosis, ROS levels, glucose consumption, and lactate production in yellow cattle PASMCs (p < 0.05), and the cells were dedifferentiated from the contractile phenotype; conversely, hypoxia had no significant effect on yak PASMCs (p > 0.05). PDK1 overexpression significantly promoted proliferation, antiapoptosis, glucose consumption, and lactate production in yak PASMCs under normoxia and hypoxia (p < 0.05), decreased their migration levels under hypoxia (p < 0.05), and dedifferentiated the contractile phenotype of the cells. Overexpression of PDK1 in yak PASMCs is detrimental to their adaptation to hypoxic environments. Yak PASMCs adapted to the effects of hypoxia on lung tissue by downregulating the expression of genes related to the PDK1 and TGF-β/Smad signaling pathways. Taken together, the regulation of PDK1-mediated TGF-β/Smad signaling may be involved in the process of yaks' adaptation to the hypoxic environment of the plateau, reflecting the good adaptive ability of yaks. The present study provides basic information to further elucidate the mechanism of PDK1-mediated TGF-β/Smad signaling induced by hypoxia in the lungs of yaks, as well as target genes for the treatment of plateau diseases in humans and animals.
Collapse
Affiliation(s)
- Yiyang Zhang
- Engineering Research Center of Key Technology and Industrialization of Cell-Based Vaccine, Ministry of Education, Northwest Minzu University, Lanzhou 730030, China; (Y.Z.); (X.L.); (F.Z.); (M.Z.); (Z.Q.)
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (M.Z.); (W.C.); (H.D.); (X.T.)
| | - Jun Wang
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (M.Z.); (W.C.); (H.D.); (X.T.)
| | - Meng Zhang
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (M.Z.); (W.C.); (H.D.); (X.T.)
| | - Xiaoyun Li
- Engineering Research Center of Key Technology and Industrialization of Cell-Based Vaccine, Ministry of Education, Northwest Minzu University, Lanzhou 730030, China; (Y.Z.); (X.L.); (F.Z.); (M.Z.); (Z.Q.)
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (M.Z.); (W.C.); (H.D.); (X.T.)
| | - Fan Zhang
- Engineering Research Center of Key Technology and Industrialization of Cell-Based Vaccine, Ministry of Education, Northwest Minzu University, Lanzhou 730030, China; (Y.Z.); (X.L.); (F.Z.); (M.Z.); (Z.Q.)
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (M.Z.); (W.C.); (H.D.); (X.T.)
| | - Manlin Zhou
- Engineering Research Center of Key Technology and Industrialization of Cell-Based Vaccine, Ministry of Education, Northwest Minzu University, Lanzhou 730030, China; (Y.Z.); (X.L.); (F.Z.); (M.Z.); (Z.Q.)
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (M.Z.); (W.C.); (H.D.); (X.T.)
| | - Kun Yang
- Engineering Research Center of Key Technology and Industrialization of Cell-Based Vaccine, Ministry of Education, Northwest Minzu University, Lanzhou 730030, China; (Y.Z.); (X.L.); (F.Z.); (M.Z.); (Z.Q.)
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (M.Z.); (W.C.); (H.D.); (X.T.)
| | - Weiji Chen
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (M.Z.); (W.C.); (H.D.); (X.T.)
| | - Haie Ding
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (M.Z.); (W.C.); (H.D.); (X.T.)
| | - Xiao Tan
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (M.Z.); (W.C.); (H.D.); (X.T.)
| | - Qian Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China;
| | - Zilin Qiao
- Engineering Research Center of Key Technology and Industrialization of Cell-Based Vaccine, Ministry of Education, Northwest Minzu University, Lanzhou 730030, China; (Y.Z.); (X.L.); (F.Z.); (M.Z.); (Z.Q.)
- Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou 730030, China
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730030, China; (J.W.); (M.Z.); (W.C.); (H.D.); (X.T.)
| |
Collapse
|
30
|
Gerasimovskaya E, Patil RS, Davies A, Maloney ME, Simon L, Mohamed B, Cherian-Shaw M, Verin AD. Extracellular purines in lung endothelial permeability and pulmonary diseases. Front Physiol 2024; 15:1450673. [PMID: 39234309 PMCID: PMC11372795 DOI: 10.3389/fphys.2024.1450673] [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: 06/17/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
The purinergic signaling system is an evolutionarily conserved and critical regulatory circuit that maintains homeostatic balance across various organ systems and cell types by providing compensatory responses to diverse pathologies. Despite cardiovascular diseases taking a leading position in human morbidity and mortality worldwide, pulmonary diseases represent significant health concerns as well. The endothelium of both pulmonary and systemic circulation (bronchial vessels) plays a pivotal role in maintaining lung tissue homeostasis by providing an active barrier and modulating adhesion and infiltration of inflammatory cells. However, investigations into purinergic regulation of lung endothelium have remained limited, despite widespread recognition of the role of extracellular nucleotides and adenosine in hypoxic, inflammatory, and immune responses within the pulmonary microenvironment. In this review, we provide an overview of the basic aspects of purinergic signaling in vascular endothelium and highlight recent studies focusing on pulmonary microvascular endothelial cells and endothelial cells from the pulmonary artery vasa vasorum. Through this compilation of research findings, we aim to shed light on the emerging insights into the purinergic modulation of pulmonary endothelial function and its implications for lung health and disease.
Collapse
Affiliation(s)
| | - Rahul S. Patil
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Adrian Davies
- Department of Internal Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - McKenzie E. Maloney
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Office of Academic Affairs, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Liselle Simon
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Basmah Mohamed
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Mary Cherian-Shaw
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Alexander D. Verin
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
31
|
He J, Wang K, Wang B, Cui Y, Zhang Q. Effect of the TGF-β/BMP Signaling Pathway on the Proliferation of Yak Pulmonary Artery Smooth Muscle Cells under Hypoxic Conditions. Animals (Basel) 2024; 14:2072. [PMID: 39061534 PMCID: PMC11274247 DOI: 10.3390/ani14142072] [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: 05/27/2024] [Revised: 07/06/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
To survive in low-oxygen environments, yaks effectively avoid hypoxia-induced pulmonary arterial hypertension through vascular remodeling. The TGF-β/BMP signaling pathway plays a key role in maintaining the homeostasis of pulmonary artery smooth muscle cells (PASMCs). However, little is known about the molecular regulatory mechanisms by which the TGF-β/BMP signaling pathway contributes to the proliferation of yak PASMCs. In this study, yak PASMCs were cultured in vitro, and a hypoxia model was constructed to investigate the effect of TGFβ/BMP signaling on yak PASMC proliferation. Hypoxia treatment increased the proliferation of yak PASMCs significantly. As the duration of hypoxia increased, the expression levels of TGF-β1 and the phosphorylation levels of Smad2/3 were upregulated significantly. The BMP signaling pathway was transiently activated by hypoxia, with increases in BMPR2 expression and Smad1/5 phosphorylation, and these changes were gradually reversed with prolonged hypoxia exposure. In addition, exogenous TGF-β1 activated the TGF-β signaling pathway, increased the phosphorylation levels of the downstream proteins Smad2 and Smad3, and increased the proliferation and migration rates of yak PASMCs significantly. Finally, treatment with noggin (an inhibitor of BMP signaling) significantly reduced BMPR2 protein expression levels and Smad1/5 phosphorylation levels and increased yak PASMC proliferation and migration rates. In summary, these results revealed that under hypoxic conditions, the dynamic regulation of the TGF-β/BMP signaling pathway promotes the proliferation of yak PASMCs.
Collapse
Affiliation(s)
- Junfeng He
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (B.W.); (Y.C.); (Q.Z.)
| | - Kejin Wang
- Gansu Provincial Center for Disease Control and Prevention, Lanzhou 730070, China;
| | - Biao Wang
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (B.W.); (Y.C.); (Q.Z.)
| | - Yan Cui
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (B.W.); (Y.C.); (Q.Z.)
- Gansu Province Livestock Embryo Engineering Research Center, Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China
| | - Qian Zhang
- Laboratory of Animal Anatomy & Tissue Embryology, Department of Basic Veterinary Medicine, Faculty of Veterinary Medicine, Gansu Agricultural University, Lanzhou 730070, China; (B.W.); (Y.C.); (Q.Z.)
| |
Collapse
|
32
|
Shen H, Gao Y, Ge D, Tan M, Yin Q, Wei TYW, He F, Lee TY, Li Z, Chen Y, Yang Q, Liu Z, Li X, Chen Z, Yang Y, Zhang Z, Thistlethwaite PA, Wang J, Malhotra A, Yuan JXJ, Shyy JYJ, Gong K. BRCC3 Regulation of ALK2 in Vascular Smooth Muscle Cells: Implication in Pulmonary Hypertension. Circulation 2024; 150:132-150. [PMID: 38557054 PMCID: PMC11230848 DOI: 10.1161/circulationaha.123.066430] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 02/28/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND An imbalance of antiproliferative BMP (bone morphogenetic protein) signaling and proliferative TGF-β (transforming growth factor-β) signaling is implicated in the development of pulmonary arterial hypertension (PAH). The posttranslational modification (eg, phosphorylation and ubiquitination) of TGF-β family receptors, including BMPR2 (bone morphogenetic protein type 2 receptor)/ALK2 (activin receptor-like kinase-2) and TGF-βR2/R1, and receptor-regulated Smads significantly affects their activity and thus regulates the target cell fate. BRCC3 modifies the activity and stability of its substrate proteins through K63-dependent deubiquitination. By modulating the posttranslational modifications of the BMP/TGF-β-PPARγ pathway, BRCC3 may play a role in pulmonary vascular remodeling, hence the pathogenesis of PAH. METHODS Bioinformatic analyses were used to explore the mechanism by which BRCC3 deubiquitinates ALK2. Cultured pulmonary artery smooth muscle cells (PASMCs), mouse models, and specimens from patients with idiopathic PAH were used to investigate the rebalance between BMP and TGF-β signaling in regulating ALK2 phosphorylation and ubiquitination in the context of pulmonary hypertension. RESULTS BRCC3 was significantly downregulated in PASMCs from patients with PAH and animals with experimental pulmonary hypertension. BRCC3, by de-ubiquitinating ALK2 at Lys-472 and Lys-475, activated receptor-regulated Smad1/5/9, which resulted in transcriptional activation of BMP-regulated PPARγ, p53, and Id1. Overexpression of BRCC3 also attenuated TGF-β signaling by downregulating TGF-β expression and inhibiting phosphorylation of Smad3. Experiments in vitro indicated that overexpression of BRCC3 or the de-ubiquitin-mimetic ALK2-K472/475R attenuated PASMC proliferation and migration and enhanced PASMC apoptosis. In SM22α-BRCC3-Tg mice, pulmonary hypertension was ameliorated because of activation of the ALK2-Smad1/5-PPARγ axis in PASMCs. In contrast, Brcc3-/- mice showed increased susceptibility of experimental pulmonary hypertension because of inhibition of the ALK2-Smad1/5 signaling. CONCLUSIONS These results suggest a pivotal role of BRCC3 in sustaining pulmonary vascular homeostasis by maintaining the integrity of the BMP signaling (ie, the ALK2-Smad1/5-PPARγ axis) while suppressing TGF-β signaling in PASMCs. Such rebalance of BMP/TGF-β pathways is translationally important for PAH alleviation.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- Activin Receptors, Type II/metabolism
- Activin Receptors, Type II/genetics
- Bone Morphogenetic Protein Receptors, Type II/metabolism
- Bone Morphogenetic Protein Receptors, Type II/genetics
- Cell Proliferation
- Cells, Cultured
- Disease Models, Animal
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/pathology
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- PPAR gamma/metabolism
- PPAR gamma/genetics
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/pathology
- Pulmonary Arterial Hypertension/genetics
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Signal Transduction
- Ubiquitination
- Vascular Remodeling
Collapse
Affiliation(s)
- Hui Shen
- Department of Cardiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Institute of Cardiovascular Disease, Yangzhou Key Lab of Innovation Frontiers in Cardiovascular Disease, China (H.S., Y.G., D.G., M.T., Q. Yin, Z.L., X.L., Z.C., Y.Y., Z.Z., K.G.)
| | - Ya Gao
- Department of Cardiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Institute of Cardiovascular Disease, Yangzhou Key Lab of Innovation Frontiers in Cardiovascular Disease, China (H.S., Y.G., D.G., M.T., Q. Yin, Z.L., X.L., Z.C., Y.Y., Z.Z., K.G.)
| | - Dedong Ge
- Department of Cardiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Institute of Cardiovascular Disease, Yangzhou Key Lab of Innovation Frontiers in Cardiovascular Disease, China (H.S., Y.G., D.G., M.T., Q. Yin, Z.L., X.L., Z.C., Y.Y., Z.Z., K.G.)
| | - Meng Tan
- Department of Cardiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Institute of Cardiovascular Disease, Yangzhou Key Lab of Innovation Frontiers in Cardiovascular Disease, China (H.S., Y.G., D.G., M.T., Q. Yin, Z.L., X.L., Z.C., Y.Y., Z.Z., K.G.)
| | - Qing Yin
- Department of Cardiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Institute of Cardiovascular Disease, Yangzhou Key Lab of Innovation Frontiers in Cardiovascular Disease, China (H.S., Y.G., D.G., M.T., Q. Yin, Z.L., X.L., Z.C., Y.Y., Z.Z., K.G.)
| | - Tong-You Wade Wei
- Division of Cardiology (T.-Y.W.W., J.Y.-J.S.), University of California, San Diego, La Jolla
| | - Fangzhou He
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, China (F.H.)
| | - Tzong-Yi Lee
- Warshel Institute for Computational Biology, School of Medicine, Chinese University of Hong Kong, Shenzhen, China (T.-Y.L., Z.L.)
| | - Zhongyan Li
- Warshel Institute for Computational Biology, School of Medicine, Chinese University of Hong Kong, Shenzhen, China (T.-Y.L., Z.L.)
| | - Yuqin Chen
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, China (Y.C., Q. Yang, J.W.)
| | - Qifeng Yang
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, China (Y.C., Q. Yang, J.W.)
| | - Zhangyu Liu
- Department of Cardiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Institute of Cardiovascular Disease, Yangzhou Key Lab of Innovation Frontiers in Cardiovascular Disease, China (H.S., Y.G., D.G., M.T., Q. Yin, Z.L., X.L., Z.C., Y.Y., Z.Z., K.G.)
| | - Xinxin Li
- Department of Cardiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Institute of Cardiovascular Disease, Yangzhou Key Lab of Innovation Frontiers in Cardiovascular Disease, China (H.S., Y.G., D.G., M.T., Q. Yin, Z.L., X.L., Z.C., Y.Y., Z.Z., K.G.)
| | - Zixuan Chen
- Department of Cardiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Institute of Cardiovascular Disease, Yangzhou Key Lab of Innovation Frontiers in Cardiovascular Disease, China (H.S., Y.G., D.G., M.T., Q. Yin, Z.L., X.L., Z.C., Y.Y., Z.Z., K.G.)
| | - Yi Yang
- Department of Cardiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Institute of Cardiovascular Disease, Yangzhou Key Lab of Innovation Frontiers in Cardiovascular Disease, China (H.S., Y.G., D.G., M.T., Q. Yin, Z.L., X.L., Z.C., Y.Y., Z.Z., K.G.)
| | - Zhengang Zhang
- Department of Cardiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Institute of Cardiovascular Disease, Yangzhou Key Lab of Innovation Frontiers in Cardiovascular Disease, China (H.S., Y.G., D.G., M.T., Q. Yin, Z.L., X.L., Z.C., Y.Y., Z.Z., K.G.)
| | - Patricia A Thistlethwaite
- Department of Medicine, Division of Cardiothoracic Surgery (P.A.T.), University of California, San Diego, La Jolla
| | - Jian Wang
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, China (Y.C., Q. Yang, J.W.)
- Guangzhou National Laboratory, Guangzhou International Bio Island, China (J.W.)
| | - Atul Malhotra
- Division of Pulmonary and Critical Care Medicine (A.M.), University of California, San Diego, La Jolla
| | - Jason X-J Yuan
- Division of Pulmonary, Critical Care and Sleep Medicine (J.X.-J.Y.), University of California, San Diego, La Jolla
| | - John Y-J Shyy
- Division of Cardiology (T.-Y.W.W., J.Y.-J.S.), University of California, San Diego, La Jolla
| | - Kaizheng Gong
- Department of Cardiology, Affiliated Hospital of Yangzhou University, Yangzhou University, Institute of Cardiovascular Disease, Yangzhou Key Lab of Innovation Frontiers in Cardiovascular Disease, China (H.S., Y.G., D.G., M.T., Q. Yin, Z.L., X.L., Z.C., Y.Y., Z.Z., K.G.)
| |
Collapse
|
33
|
Mao Z, Zheng P, Zhu X, Wang L, Zhang F, Liu H, Li H, Zhou L, Liu W. Obstructive sleep apnea hypopnea syndrome and vascular lesions: An update on what we currently know. Sleep Med 2024; 119:296-311. [PMID: 38723575 DOI: 10.1016/j.sleep.2024.05.010] [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: 03/21/2024] [Revised: 04/23/2024] [Accepted: 05/02/2024] [Indexed: 06/18/2024]
Abstract
Obstructive sleep apnea-hypopnea syndrome (OSAHS) is the most prevalent sleep and respiratory disorder. This syndrome can induce severe cardiovascular and cerebrovascular complications, and intermittent hypoxia is a pivotal contributor to this damage. Vascular pathology is closely associated with the impairment of target organs, marking a focal point in current research. Vascular lesions are the fundamental pathophysiological basis of multiorgan ailments and indicate a shared pathogenic mechanism among common cardiovascular and cerebrovascular conditions, suggesting their importance as a public health concern. Increasing evidence shows a strong correlation between OSAHS and vascular lesions. Previous studies predominantly focused on the pathophysiological alterations in OSAHS itself, such as intermittent hypoxia and fragmented sleep, leading to vascular disruptions. This review aims to delve deeper into the vascular lesions affected by OSAHS by examining the microscopic pathophysiological mechanisms involved. Emphasis has been placed on examining how OSAHS induces vascular lesions through disruptions in the endothelial barrier, metabolic dysregulation, cellular phenotype alterations, neuroendocrine irregularities, programmed cell death, vascular inflammation, oxidative stress and epigenetic modifications. This review examines the epidemiology and associated risk factors for OSAHS and vascular diseases and subsequently describes the existing evidence on vascular lesions induced by OSAHS in the cardiovascular, cerebrovascular, retinal, renal and reproductive systems. A detailed account of the current research on the pathophysiological mechanisms mediating vascular lesions caused by OSAHS is provided, culminating in a discussion of research advancements in therapeutic modalities to mitigate OSAHS-related vascular lesions and the implications of these treatment strategies.
Collapse
Affiliation(s)
- Zhenyu Mao
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pengdou Zheng
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyan Zhu
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingling Wang
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengqin Zhang
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiguo Liu
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hai Li
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Ling Zhou
- Department of Respiratory and Critical Care Medicine, National Health Committee (NHC) Key Laboratory of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Wei Liu
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China.
| |
Collapse
|
34
|
Ahmed ASI, Blood AB, Zhang L. Hypoxia-induced pulmonary hypertension in adults and newborns: implications for drug development. Drug Discov Today 2024; 29:104015. [PMID: 38719143 PMCID: PMC11936511 DOI: 10.1016/j.drudis.2024.104015] [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: 02/15/2024] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024]
Abstract
Chronic hypoxia-induced pulmonary hypertension (CHPH) presents a complex challenge, characterized by escalating pulmonary vascular resistance and remodeling, threatening both newborns and adults with right heart failure. Despite advances in understanding the pathobiology of CHPH, its molecular intricacies remain elusive, particularly because of the multifaceted nature of arterial remodeling involving the adventitia, media, and intima. Cellular imbalance arises from hypoxia-induced mitochondrial disturbances and oxidative stress, reflecting the diversity in pulmonary hypertension (PH) pathology. In this review, we highlight prominent mechanisms causing CHPH in adults and newborns, and emerging therapeutic targets of potential pharmaceuticals.
Collapse
Affiliation(s)
- Abu Shufian Ishtiaq Ahmed
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| | - Arlin B Blood
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| |
Collapse
|
35
|
Gao Z, Santos RB, Rupert J, Van Drunen R, Yu Y, Eckel‐Mahan K, Kolonin MG. Endothelial-specific telomerase inactivation causes telomere-independent cell senescence and multi-organ dysfunction characteristic of aging. Aging Cell 2024; 23:e14138. [PMID: 38475941 PMCID: PMC11296101 DOI: 10.1111/acel.14138] [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: 11/02/2023] [Revised: 01/31/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
It has remained unclear how aging of endothelial cells (EC) contributes to pathophysiology of individual organs. Cell senescence results in part from inactivation of telomerase (TERT). Here, we analyzed mice with Tert knockout specifically in EC. Tert loss in EC induced transcriptional changes indicative of senescence and tissue hypoxia in EC and in other cells. We demonstrate that EC-Tert-KO mice have leaky blood vessels. The blood-brain barrier of EC-Tert-KO mice is compromised, and their cognitive function is impaired. EC-Tert-KO mice display reduced muscle endurance and decreased expression of enzymes responsible for oxidative metabolism. Our data indicate that Tert-KO EC have reduced mitochondrial content and function, which results in increased dependence on glycolysis. Consistent with this, EC-Tert-KO mice have metabolism changes indicative of increased glucose utilization. In EC-Tert-KO mice, expedited telomere attrition is observed for EC of adipose tissue (AT), while brain and skeletal muscle EC have normal telomere length but still display features of senescence. Our data indicate that the loss of Tert causes EC senescence in part through a telomere length-independent mechanism undermining mitochondrial function. We conclude that EC-Tert-KO mice is a model of expedited vascular senescence recapitulating the hallmarks aging, which can be useful for developing revitalization therapies.
Collapse
Affiliation(s)
- Zhanguo Gao
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Rafael Bravo Santos
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Joseph Rupert
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Rachel Van Drunen
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Yongmei Yu
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Kristin Eckel‐Mahan
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Mikhail G. Kolonin
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| |
Collapse
|
36
|
Shen T, Shi J, Zhao X, Fu L, Wang N, Zheng X, Chen Y, Li M, Ma C, Liu P, Zhu D. Presenilin 1 Is a Therapeutic Target in Pulmonary Hypertension and Promotes Vascular Remodeling. Am J Respir Cell Mol Biol 2024; 70:468-481. [PMID: 38381098 DOI: 10.1165/rcmb.2022-0426oc] [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: 11/02/2022] [Accepted: 02/20/2024] [Indexed: 02/22/2024] Open
Abstract
Small muscular pulmonary artery remodeling is a dominant feature of pulmonary arterial hypertension (PAH). PSEN1 affects angiogenesis, cancer, and Alzheimer's disease. We aimed to determine the role of PSEN1 in the pathogenesis of vascular remodeling in pulmonary hypertension (PH). Hemodynamics and vascular remodeling in the Psen1-knockin and smooth muscle-specific Psen1-knockout mice were assessed. The functional partners of PSEN1 were predicted by bioinformatics analysis and biochemical experiments. The therapeutic effect of PH was evaluated by administration of the PSEN1-specific inhibitor ELN318463. We discovered that both the mRNA and protein levels of PSEN1 were increased over time in hypoxic rats, monocrotaline rats, and Su5416/hypoxia mice. Psen1 transgenic mice were highly susceptible to PH, whereas smooth muscle-specific Psen1-knockout mice were resistant to hypoxic PH. STRING analysis showed that Notch1/2/3, β-catenin, Cadherin-1, DNER (delta/notch-like epidermal growth factor-related receptor), TMP10, and ERBB4 appeared to be highly correlated with PSEN1. Immunoprecipitation confirmed that PSEN1 interacts with β-catenin and DNER, and these interactions were suppressed by the catalytic PSEN1 mutations D257A, D385A, and C410Y. PSEN1 was found to mediate the nuclear translocation of the Notch1 intracellular domains and activated RBP-Jκ. Octaarginine-coated liposome-mediated pharmacological inhibition of PSEN1 significantly prevented and reversed the pathological process in hypoxic and monocrotaline-induced PH. PSEN1 essentially drives the pathogenesis of PAH and interacted with the noncanonical Notch ligand DNER. PSEN1 can be used as a promising molecular target for treating PAH. PSEN1 inhibitor ELN318463 can prevent and reverse the progression of PH and can be developed as a potential anti-PAH drug.
Collapse
Affiliation(s)
- TingTing Shen
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
- Central Laboratory of Harbin Medical University-Daqing, College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, China; and
| | - JiuCheng Shi
- Central Laboratory of Harbin Medical University-Daqing, College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, China; and
| | - XiJuan Zhao
- Central Laboratory of Harbin Medical University-Daqing, College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, China; and
| | - Li Fu
- Central Laboratory of Harbin Medical University-Daqing, College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, China; and
| | - Na Wang
- Central Laboratory of Harbin Medical University-Daqing, College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, China; and
| | - XiaoDong Zheng
- Central Laboratory of Harbin Medical University-Daqing, College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, China; and
| | - YingLi Chen
- Central Laboratory of Harbin Medical University-Daqing, College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, China; and
| | - MingHui Li
- Central Laboratory of Harbin Medical University-Daqing, College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, China; and
| | - Cui Ma
- Central Laboratory of Harbin Medical University-Daqing, College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, China; and
| | - PiXu Liu
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - DaLing Zhu
- Central Laboratory of Harbin Medical University-Daqing, College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing, China; and
- College of Pharmacy, Harbin Medical University, Harbin, China
| |
Collapse
|
37
|
Zhang H, Li M, Hu CJ, Stenmark KR. Fibroblasts in Pulmonary Hypertension: Roles and Molecular Mechanisms. Cells 2024; 13:914. [PMID: 38891046 PMCID: PMC11171669 DOI: 10.3390/cells13110914] [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: 04/26/2024] [Revised: 05/17/2024] [Accepted: 05/22/2024] [Indexed: 06/20/2024] Open
Abstract
Fibroblasts, among the most prevalent and widely distributed cell types in the human body, play a crucial role in defining tissue structure. They do this by depositing and remodeling extracellular matrixes and organizing functional tissue networks, which are essential for tissue homeostasis and various human diseases. Pulmonary hypertension (PH) is a devastating syndrome with high mortality, characterized by remodeling of the pulmonary vasculature and significant cellular and structural changes within the intima, media, and adventitia layers. Most research on PH has focused on alterations in the intima (endothelial cells) and media (smooth muscle cells). However, research over the past decade has provided strong evidence of the critical role played by pulmonary artery adventitial fibroblasts in PH. These fibroblasts exhibit the earliest, most dramatic, and most sustained proliferative, apoptosis-resistant, and inflammatory responses to vascular stress. This review examines the aberrant phenotypes of PH fibroblasts and their role in the pathogenesis of PH, discusses potential molecular signaling pathways underlying these activated phenotypes, and highlights areas of research that merit further study to identify promising targets for the prevention and treatment of PH.
Collapse
Affiliation(s)
- Hui Zhang
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Min Li
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Cheng-Jun Hu
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Craniofacial Biology, University of Colorado School of Dental Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kurt R. Stenmark
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
38
|
Xie J, Lin H, Zuo A, Shao J, Sun W, Wang S, Song J, Yao W, Luo Y, Sun J, Wang M. The JMJD family of histone demethylase and their intimate links to cardiovascular disease. Cell Signal 2024; 116:111046. [PMID: 38242266 DOI: 10.1016/j.cellsig.2024.111046] [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: 02/23/2023] [Revised: 01/05/2024] [Accepted: 01/11/2024] [Indexed: 01/21/2024]
Abstract
The incidence rate and mortality rate of cardiovascular disease rank first in the world. It is associated with various high-risk factors, and there is no single cause. Epigenetic modifications, such as DNA methylation or histone modification, actively participate in the initiation and development of cardiovascular diseases. Histone lysine methylation is a type of histone post-translational modification. The human Jumonji C domain (JMJD) protein family consists of more than 30 members. JMJD proteins participate in many key nuclear processes and play a key role in the specific regulation of gene expression, DNA damage and repair, and DNA replication. Importantly, increasing evidence shows that JMJD proteins are abnormally expressed in cardiovascular diseases, which may be a potential mechanism for the occurrence and development of these diseases. Here, we discuss the key roles of JMJD proteins in various common cardiovascular diseases. This includes histone lysine demethylase, which has been studied in depth, and less-studied JMJD members. Furthermore, we focus on the epigenetic changes induced by each JMJD member, summarize recent research progress, and evaluate their relationship with cardiovascular diseases and therapeutic potential.
Collapse
Affiliation(s)
- Jiarun Xie
- Department of Traditional Chinese Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Haoyu Lin
- Department of Traditional Chinese Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Anna Zuo
- Department of Traditional Chinese Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Junqiao Shao
- Department of Traditional Chinese Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Wei Sun
- Department of Traditional Chinese Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Shaoting Wang
- Department of Traditional Chinese Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Jianda Song
- Department of Traditional Chinese Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Wang Yao
- Department of Traditional Chinese Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Yanyu Luo
- Department of Traditional Chinese Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Jia Sun
- Department of Traditional Chinese Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| | - Ming Wang
- Department of Traditional Chinese Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
39
|
Dai ZK, Chen YC, Hsieh SL, Yeh JL, Hsu JH, Wu BN. The Xanthine Derivative KMUP-1 Inhibits Hypoxia-Induced TRPC1 Expression and Store-Operated Ca 2+ Entry in Pulmonary Arterial Smooth Muscle Cells. Pharmaceuticals (Basel) 2024; 17:440. [PMID: 38675401 PMCID: PMC11053947 DOI: 10.3390/ph17040440] [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: 02/23/2024] [Revised: 03/17/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Exposure to hypoxia results in the development of pulmonary arterial hypertension (PAH). An increase in the intracellular Ca2+ concentration ([Ca2+]i) in pulmonary artery smooth muscle cells (PASMCs) is a major trigger for pulmonary vasoconstriction and proliferation. This study investigated the mechanism by which KMUP-1, a xanthine derivative with phosphodiesterase inhibitory activity, inhibits hypoxia-induced canonical transient receptor potential channel 1 (TRPC1) protein overexpression and regulates [Ca2+]i through store-operated calcium channels (SOCs). Ex vivo PASMCs were cultured from Sprague-Dawley rats in a modular incubator chamber under 1% O2/5% CO2 for 24 h to elucidate TRPC1 overexpression and observe the Ca2+ release and entry. KMUP-1 (1 μM) inhibited hypoxia-induced TRPC family protein encoded for SOC overexpression, particularly TRPC1. KMUP-1 inhibition of TRPC1 protein was restored by the protein kinase G (PKG) inhibitor KT5823 (1 μM) and the protein kinase A (PKA) inhibitor KT5720 (1 μM). KMUP-1 attenuated protein kinase C (PKC) activator phorbol 12-myristate 13-acetate (PMA, 1 μM)-upregulated TRPC1. We suggest that the effects of KMUP-1 on TRPC1 might involve activating the cyclic guanosine monophosphate (cGMP)/PKG and cyclic adenosine monophosphate (cAMP)/PKA pathways and inhibiting the PKC pathway. We also used Fura 2-acetoxymethyl ester (Fura 2-AM, 5 μM) to measure the stored calcium release from the sarcoplasmic reticulum (SR) and calcium entry through SOCs in hypoxic PASMCs under treatment with thapsigargin (1 μM) and nifedipine (5 μM). In hypoxic conditions, store-operated calcium entry (SOCE) activity was enhanced in PASMCs, and KMUP-1 diminished this activity. In conclusion, KMUP-1 inhibited the expression of TRPC1 protein and the activity of SOC-mediated Ca2+ entry upon SR Ca2+ depletion in hypoxic PASMCs.
Collapse
Affiliation(s)
- Zen-Kong Dai
- Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Z.-K.D.); (J.-H.H.)
- Division of Pediatric Cardiology and Pulmonology, Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Yi-Chen Chen
- Department of Pharmacology, Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-C.C.); (J.-L.Y.)
| | - Su-Ling Hsieh
- Department of Pharmacy, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
| | - Jwu-Lai Yeh
- Department of Pharmacology, Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-C.C.); (J.-L.Y.)
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Jong-Hau Hsu
- Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Z.-K.D.); (J.-H.H.)
- Division of Pediatric Cardiology and Pulmonology, Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Bin-Nan Wu
- Department of Pharmacology, Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-C.C.); (J.-L.Y.)
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| |
Collapse
|
40
|
Listyoko AS, Okazaki R, Harada T, Inui G, Yamasaki A. Impact of obesity on airway remodeling in asthma: pathophysiological insights and clinical implications. FRONTIERS IN ALLERGY 2024; 5:1365801. [PMID: 38562155 PMCID: PMC10982419 DOI: 10.3389/falgy.2024.1365801] [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: 01/05/2024] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
The prevalence of obesity among asthma patients has surged in recent years, posing a significant risk factor for uncontrolled asthma. Beyond its impact on asthma severity and patients' quality of life, obesity is associated with reduced lung function, increased asthma exacerbations, hospitalizations, heightened airway hyperresponsiveness, and elevated asthma-related mortality. Obesity may lead to metabolic dysfunction and immune dysregulation, fostering chronic inflammation characterized by increased pro-inflammatory mediators and adipocytokines, elevated reactive oxygen species, and reduced antioxidant activity. This chronic inflammation holds the potential to induce airway remodeling in individuals with asthma and obesity. Airway remodeling encompasses structural and pathological changes, involving alterations in the airway's epithelial and subepithelial layers, hyperplasia and hypertrophy of airway smooth muscle, and changes in airway vascularity. In individuals with asthma and obesity, airway remodeling may underlie heightened airway hyperresponsiveness and increased asthma severity, ultimately contributing to the development of persistent airflow limitation, declining lung function, and a potential increase in asthma-related mortality. Despite efforts to address the impact of obesity on asthma outcomes, the intricate mechanisms linking obesity to asthma pathophysiology, particularly concerning airway remodeling, remain incompletely understood. This comprehensive review discusses current research investigating the influence of obesity on airway remodeling, to enhance our understanding of obesity's role in the context of asthma airway remodeling.
Collapse
Affiliation(s)
- Aditya Sri Listyoko
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
- Pulmonology and Respiratory Medicine Department, Faculty of Medicine, Brawijaya University-Dr. Saiful Anwar General Hospital, Malang, Indonesia
| | - Ryota Okazaki
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Tomoya Harada
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Genki Inui
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Akira Yamasaki
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| |
Collapse
|
41
|
Lin C, Yang H, Luo Q, Liu Q. FAK mediates hypoxia-induced pulmonary artery smooth muscle cell proliferation by modulating mitochondrial transcription termination factor 1/cyclin D1. Clin Transl Sci 2024; 17:e13767. [PMID: 38488492 PMCID: PMC10941516 DOI: 10.1111/cts.13767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 02/05/2024] [Accepted: 02/20/2024] [Indexed: 03/18/2024] Open
Abstract
This study aimed to investigate the mechanism of FAK-dependent hypoxia-induced proliferation on human pulmonary artery smooth muscle cells (HPASMCs). Primary HPASMCs were isolated and cultured in vitro under normal and hypoxia conditions to assess cell proliferation with cell counting kit-8. FAK and mitochondrial transcription termination factor 1 (mTERF1) were silenced with siRNA, mRNA, and protein levels of FAK, mTERF1, and cyclin D1 were determined. HPASMC proliferation increased under hypoxia compared to normal conditions. Knocking down FAK or mTERF1 with siRNA led to decreased cell proliferation under both normal and hypoxia conditions. FAK knockdown led to the reduction of both mTERF1 and cyclin D1 expressions under the hypoxia conditions, whereas mTERF1 knockdown led to the downregulation of cyclin D1 expression but not FAK expression under the same condition. However, under normal conditions, knocking down either FAK or mTERF1 had no impact on cyclin D1 expression. These results suggested that FAK may regulate the mTERF1/cyclin D1 signaling pathway to modulate cell proliferation in hypoxia.
Collapse
Affiliation(s)
- Chunlong Lin
- Department of Respiratory and Critical Care MedicineYueyang People's Hospital of Hunan Normal UniversityYueyangChina
| | - Hui Yang
- Department of Respiratory and Critical Care MedicineYueyang People's Hospital of Hunan Normal UniversityYueyangChina
| | - Qiong Luo
- Department of Respiratory and Critical Care MedicineYueyang People's Hospital of Hunan Normal UniversityYueyangChina
| | - Qi Liu
- Department of Respiratory and Critical Care MedicineYueyang People's Hospital of Hunan Normal UniversityYueyangChina
| |
Collapse
|
42
|
Truong LN, Wilson Santos E, Zheng YM, Wang YX. Rieske Iron-Sulfur Protein Mediates Pulmonary Hypertension Following Nicotine/Hypoxia Coexposure. Am J Respir Cell Mol Biol 2024; 70:193-202. [PMID: 38029303 PMCID: PMC10914767 DOI: 10.1165/rcmb.2023-0181oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 11/29/2023] [Indexed: 12/01/2023] Open
Abstract
The high mortality rate in patients with chronic obstructive pulmonary disease (COPD) may be due to pulmonary hypertension (PH). These diseases are highly associated with cigarette smoke and its key component nicotine. Here, we created a novel animal model of PH using coexposure to nicotine (or cigarette smoke) and hypoxia. This heretofore unreported model showed significant early-onset pulmonary vasoremodeling and PH. Using newly generated mice with complementary smooth muscle-specific Rieske iron-sulfur protein (RISP) gene knockout and overexpression, we demonstrate that RISP is critically involved in promoting pulmonary vasoremodeling and PH, which are implemented by oxidative ataxia telangiectasia-mutated-mediated DNA damage and NF-κB-dependent inflammation in a reciprocal positive mechanism. Together, our findings establish for the first time an animal model of hypoxia-induced early-onset PH in which mitochondrial RISP-dependent DNA damage and NF-κB inflammation play critical roles in vasoremodeling. Specific therapeutic targets for RISP and related oxidative stress-associated signaling pathways may create unique and effective treatments for PH, chronic obstructive pulmonary disease, and their complications.
Collapse
Affiliation(s)
- Lillian N Truong
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Ed Wilson Santos
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Yun-Min Zheng
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| | - Yong-Xiao Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York
| |
Collapse
|
43
|
Chen J, Song M, Qian D, Liu L, Yang K, Shou Y, Zhao H, Zhang L. Atorvastatin rescues pulmonary artery hypertension by inhibiting the AKT/ERK-dependent PDGF-BB/HIF-1α axis. Panminerva Med 2024; 66:4-9. [PMID: 33908728 DOI: 10.23736/s0031-0808.20.03910-5] [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: 11/08/2022]
Abstract
BACKGROUND The aim of this study is to explore the role of atorvastatin in rescuing pulmonary artery hypertension (PAH) by inhibiting the AKT/ERK-dependent PDGF-BB/HIF-1α axis. METHODS PAH model in rats was established by MCT induction, followed by Atorvastatin intervention. Pulmonary hemodynamic measurement and pulmonary morphological evaluation in rats were conducted. Human pulmonary artery smooth muscle cells (hPASMCs) were subjected to hypoxic exposure or PDGF-BB treatment, followed by atorvastatin induction. Relative levels of HIF-1α, p-ERK and p-Akt were detected. Viability and apoptosis were respectively determined by cell counting kit-8 (CCK-8) assay and flow cytometry. RESULTS Atorvastatin protected PAH-induced increases in RVSP and Fulton's index in rats. Meanwhile, it inhibited vascular remodeling following PAH by downregulating HIF-1α and PDGF-BB. Hypoxia or PDGF-BB treatment in hPASMCs resulted in upregulation of p-ERK and p-Akt, and viability increase, which were partially abolished by Atorvastatin intervention. In addition, atorvastatin triggered apoptosis in hypoxia or PDGF-BB-induced hPASMCs. CONCLUSIONS Atorvastatin inhibits the activation of HIF-1α and proliferative ability, and triggers apoptosis in hPASMCs exposed to hypoxia or PDGF-BB treatment through inactivating the AKT/ERK pathway.
Collapse
Affiliation(s)
- Jianfei Chen
- Department of Cardiology, Banan People's Hospital of Chongqing, Chongqing, China
| | - Mingbao Song
- Department of Cardiology, Kangxin Hospital of Chongqing, Chongqing, China
| | - Dehui Qian
- Department of Cardiology, Xingqiao Hospital, Chongqing, China
| | - Linqiong Liu
- Department of Cardiology, Banan People's Hospital of Chongqing, Chongqing, China
| | - Kun Yang
- Department of Cardiology, Banan People's Hospital of Chongqing, Chongqing, China
| | - Yunfeng Shou
- Department of Cardiology, Banan People's Hospital of Chongqing, Chongqing, China
| | - Hanru Zhao
- Department of Cardiology, Banan People's Hospital of Chongqing, Chongqing, China
| | - Li Zhang
- Department of Pathology, Southwest Hospital, Chongqing, China -
| |
Collapse
|
44
|
Haider MA, Pearce KJ, Chesler NC, Hill NA, Olufsen MS. Application and reduction of a nonlinear hyperelastic wall model capturing ex vivo relationships between fluid pressure, area, and wall thickness in normal and hypertensive murine left pulmonary arteries. INTERNATIONAL JOURNAL FOR NUMERICAL METHODS IN BIOMEDICAL ENGINEERING 2024; 40:e3798. [PMID: 38214099 DOI: 10.1002/cnm.3798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 08/10/2023] [Accepted: 11/26/2023] [Indexed: 01/13/2024]
Abstract
Pulmonary hypertension is a cardiovascular disorder manifested by elevated mean arterial blood pressure (>20 mmHg) together with vessel wall stiffening and thickening due to alterations in collagen, elastin, and smooth muscle cells. Hypoxia-induced (type 3) pulmonary hypertension can be studied in animals exposed to a low oxygen environment for prolonged time periods leading to biomechanical alterations in vessel wall structure. This study introduces a novel approach to formulating a reduced order nonlinear elastic structural wall model for a large pulmonary artery. The model relating blood pressure and area is calibrated using ex vivo measurements of vessel diameter and wall thickness changes, under controlled pressure conditions, in left pulmonary arteries isolated from control and hypertensive mice. A two-layer, hyperelastic, and anisotropic model incorporating residual stresses is formulated using the Holzapfel-Gasser-Ogden model. Complex relations predicting vessel area and wall thickness with increasing blood pressure are derived and calibrated using the data. Sensitivity analysis, parameter estimation, subset selection, and physical plausibility arguments are used to systematically reduce the 16-parameter model to one in which a much smaller subset of identifiable parameters is estimated via solution of an inverse problem. Our final reduced one layer model includes a single set of three elastic moduli. Estimated ranges of these parameters demonstrate that nonlinear stiffening is dominated by elastin in the control animals and by collagen in the hypertensive animals. The pressure-area relation developed in this novel manner has potential impact on one-dimensional fluids network models of vessel wall remodeling in the presence of cardiovascular disease.
Collapse
Affiliation(s)
- Mansoor A Haider
- Department of Mathematics, North Carolina State University, Raleigh, North Carolina, USA
| | - Katherine J Pearce
- Department of Mathematics, North Carolina State University, Raleigh, North Carolina, USA
| | - Naomi C Chesler
- Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center & Department of Biomedical Engineering, University of California, Irvine (UCI), Irvine, California, USA
| | - Nicholas A Hill
- School of Mathematics and Statistics, University of Glasgow, Glasgow, UK
| | - Mette S Olufsen
- Department of Mathematics, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
45
|
Li S, Lyu Q, Shi Q, Bai Y, Ren X, Ma J. Intermittent short-duration reoxygenation relieves high-altitude pulmonary hypertension via NOX4/H2O2/PPAR-γ axis. Clin Sci (Lond) 2024; 138:103-115. [PMID: 38237016 PMCID: PMC10830432 DOI: 10.1042/cs20231508] [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: 11/21/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 02/03/2024]
Abstract
High-altitude pulmonary hypertension (HAPH) is a severe and progressive disease that can lead to right heart failure. Intermittent short-duration reoxygenation at high altitude is effective in alleviating HAPH; however, the underlying mechanisms are unclear. In the present study, a simulated 5,000-m hypoxia rat model and hypoxic cultured pulmonary artery smooth muscle cells (PASMCs) were used to evaluate the effect and mechanisms of intermittent short-duration reoxygenation. The results showed that intermittent 3-h/per day reoxygenation (I3) effectively attenuated chronic hypoxia-induced pulmonary hypertension and reduced the content of H2O2 and the expression of NADPH oxidase 4 (NOX4) in lung tissues. In combination with I3, while the NOX inhibitor apocynin did not further alleviate HAPH, the mitochondrial antioxidant MitoQ did. Furthermore, in PASMCs, I3 attenuated hypoxia-induced PASMCs proliferation and reversed the activated HIF-1α/NOX4/PPAR-γ axis under hypoxia. Targeting this axis offset the protective effect of I3 on hypoxia-induced PASMCs proliferation. The present study is novel in revealing a new mechanism for preventing HAPH and provides insights into the optimization of intermittent short-duration reoxygenation.
Collapse
Affiliation(s)
- Shaohua Li
- Department of Aerospace Physiology, Air Force Medical University, Xi’an 710032, China
| | - Qiang Lyu
- Department of Nephrology, Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing 100853, China
| | - Qixin Shi
- Department of Aerospace Physiology, Air Force Medical University, Xi’an 710032, China
| | - Yungang Bai
- Department of Aerospace Physiology, Air Force Medical University, Xi’an 710032, China
| | - Xinling Ren
- Department of Respiratory and Critical Care Medicine, Shenzhen University General Hospital, Shenzhen 518071, China
| | - Jin Ma
- Department of Aerospace Physiology, Air Force Medical University, Xi’an 710032, China
| |
Collapse
|
46
|
Kostyunina DS, Pakhomov NV, Jouida A, Dillon E, Baugh JA, McLoughlin P. Transcriptomics and proteomics revealed sex differences in human pulmonary microvascular endothelial cells. Physiol Genomics 2024; 56:194-220. [PMID: 38047313 DOI: 10.1152/physiolgenomics.00051.2023] [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: 06/01/2023] [Revised: 11/09/2023] [Accepted: 11/30/2023] [Indexed: 12/05/2023] Open
Abstract
Marked sexual dimorphism is displayed in the onset and progression of pulmonary hypertension (PH). Females more commonly develop pulmonary arterial hypertension, yet females with pulmonary arterial hypertension and other types of PH have better survival than males. Pulmonary microvascular endothelial cells play a crucial role in pulmonary vascular remodeling and increased pulmonary vascular resistance in PH. Given this background, we hypothesized that there are sex differences in the pulmonary microvascular endothelium basally and in response to hypoxia that are independent of the sex hormone environment. Human pulmonary microvascular endothelial cells (HPMECs) from healthy male and female donors, cultured under physiological shear stress, were analyzed using RNA sequencing and label-free quantitative proteomics. Gene set enrichment analysis identified a number of sex-different pathways in both normoxia and hypoxia, including pathways that regulate cell proliferation. In vitro, the rate of proliferation in female HPMECs was lower than in male HPMECs, a finding that supports the omics results. Interestingly, thrombospondin-1, an inhibitor of proliferation, was more highly expressed in female cells than in male cells. These results demonstrate, for the first time, important differences between female and male HPMECs that persist in the absence of sex hormone differences and identify novel pathways for further investigation that may contribute to sexual dimorphism in pulmonary hypertensive diseases.NEW & NOTEWORTHY There is marked sexual dimorphism in the development and progression of pulmonary hypertension. We show differences in RNA and protein expression between female and male human pulmonary microvascular endothelial cells grown under conditions of physiological shear stress, which identify sex-different cellular pathways both in normoxia and hypoxia. Importantly, these differences were detected in the absence of sex hormone differences. The pathways identified may provide novel targets for the development of sex-specific therapies.
Collapse
Affiliation(s)
- Daria S Kostyunina
- School of Medicine, University College Dublin, Dublin, Ireland
- Conway Institute, University College Dublin, Dublin, Ireland
| | - Nikolai V Pakhomov
- School of Medicine, University College Dublin, Dublin, Ireland
- Conway Institute, University College Dublin, Dublin, Ireland
| | - Amina Jouida
- School of Medicine, University College Dublin, Dublin, Ireland
- Conway Institute, University College Dublin, Dublin, Ireland
| | - Eugene Dillon
- Conway Institute, University College Dublin, Dublin, Ireland
| | - John A Baugh
- School of Medicine, University College Dublin, Dublin, Ireland
- Conway Institute, University College Dublin, Dublin, Ireland
| | - Paul McLoughlin
- School of Medicine, University College Dublin, Dublin, Ireland
- Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
47
|
Richalet JP, Hermand E, Lhuissier FJ. Cardiovascular physiology and pathophysiology at high altitude. Nat Rev Cardiol 2024; 21:75-88. [PMID: 37783743 DOI: 10.1038/s41569-023-00924-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/16/2023] [Indexed: 10/04/2023]
Abstract
Oxygen is vital for cellular metabolism; therefore, the hypoxic conditions encountered at high altitude affect all physiological functions. Acute hypoxia activates the adrenergic system and induces tachycardia, whereas hypoxic pulmonary vasoconstriction increases pulmonary artery pressure. After a few days of exposure to low oxygen concentrations, the autonomic nervous system adapts and tachycardia decreases, thereby protecting the myocardium against high energy consumption. Permanent exposure to high altitude induces erythropoiesis, which if excessive can be deleterious and lead to chronic mountain sickness, often associated with pulmonary hypertension and heart failure. Genetic factors might account for the variable prevalence of chronic mountain sickness, depending on the population and geographical region. Cardiovascular adaptations to hypoxia provide a remarkable model of the regulation of oxygen availability at the cellular and systemic levels. Rapid exposure to high altitude can have adverse effects in patients with cardiovascular diseases. However, intermittent, moderate hypoxia might be useful in the management of some cardiovascular disorders, such as coronary heart disease and heart failure. The aim of this Review is to help physicians to understand the cardiovascular responses to hypoxia and to outline some recommendations that they can give to patients with cardiovascular disease who wish to travel to high-altitude destinations.
Collapse
Affiliation(s)
- Jean-Paul Richalet
- Hypoxie et Poumon, Université Sorbonne Paris Nord, INSERM U1272, Paris, France.
| | - Eric Hermand
- Unité de Recherche Pluridisciplinaire Sport Santé Société, ULR 7369-URePSSS, Université Littoral Côte d'Opale, Université Artois, Université Lille, CHU Lille, Dunkirk, France
| | | |
Collapse
|
48
|
Lee D, Lee H, Jo HN, Yun E, Kwon BS, Kim J, Lee A. Endothelial periostin regulates vascular remodeling by promoting endothelial dysfunction in pulmonary arterial hypertension. Anim Cells Syst (Seoul) 2024; 28:1-14. [PMID: 38186856 PMCID: PMC10769143 DOI: 10.1080/19768354.2023.2300437] [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: 12/05/2023] [Accepted: 12/20/2023] [Indexed: 01/09/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by vascular remodeling associated with extracellular matrix (ECM) deposition, vascular cell hyperproliferation, and neointima formation in the small pulmonary artery. Endothelial dysfunction is considered a key feature in the initiation of vascular remodeling. Although vasodilators have been used for the treatment of PAH, it remains a life-threatening disease. Therefore, it is necessary to identify novel therapeutic targets for PAH treatment. Periostin (POSTN) is a secretory ECM protein involved in physiological and pathological processes, such as tissue remodeling, cell adhesion, migration, and proliferation. Although POSTN has been proposed as a potential target for PAH treatment, its role in endothelial cells has not been fully elucidated. Here, we demonstrated that POSTN upregulation correlates with PAH by analyzing a public microarray conducted on the lung tissues of patients with PAH and biological experimental results from in vivo and in vitro models. Moreover, POSTN overexpression leads to ECM deposition and endothelial abnormalities such as migration. We found that PAH-associated endothelial dysfunction is mediated at least in part by the interaction between POSTN and integrin-linked protein kinase (ILK), followed by activation of nuclear factor-κB signaling. Silencing POSTN or ILK decreases PAH-related stimuli-induced ECM accumulation and attenuates endothelial abnormalities. In conclusion, our study suggests that POSTN serves as a critical regulator of PAH by regulating vascular remodeling, and targeting its role as a potential therapeutic strategy for PAH.
Collapse
Affiliation(s)
- Dawn Lee
- Division of Biological Sciences, Sookmyung Women’s University, Seoul, Republic of Korea
| | - Heeyoung Lee
- Division of Biological Sciences, Sookmyung Women’s University, Seoul, Republic of Korea
| | - Ha-neul Jo
- Division of Biological Sciences, Sookmyung Women’s University, Seoul, Republic of Korea
| | - Eunsik Yun
- Division of Biological Sciences, Sookmyung Women’s University, Seoul, Republic of Korea
| | - Byung Su Kwon
- Department of Obstetrics and Gynecology, School of Medicine, Kyung Hee University Medical Center, Kyung Hee University, Seoul, Republic of Korea
| | - Jongmin Kim
- Division of Biological Sciences, Sookmyung Women’s University, Seoul, Republic of Korea
- Research Institute for Women’s Health, Sookmyung Women’s University, Seoul, Republic of Korea
| | - Aram Lee
- Division of Biological Sciences, Sookmyung Women’s University, Seoul, Republic of Korea
- Research Institute for Women’s Health, Sookmyung Women’s University, Seoul, Republic of Korea
| |
Collapse
|
49
|
Yegambaram M, Sun X, Lu Q, Jin Y, Ornatowski W, Soto J, Aggarwal S, Wang T, Tieu K, Gu H, Fineman JR, Black SM. Mitochondrial hyperfusion induces metabolic remodeling in lung endothelial cells by modifying the activities of electron transport chain complexes I and III. Free Radic Biol Med 2024; 210:183-194. [PMID: 37979892 PMCID: PMC12051485 DOI: 10.1016/j.freeradbiomed.2023.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/02/2023] [Accepted: 11/11/2023] [Indexed: 11/20/2023]
Abstract
OBJECTIVE Pulmonary hypertension (PH) is a progressive disease with vascular remodeling as a critical structural alteration. We have previously shown that metabolic reprogramming is an early initiating mechanism in animal models of PH. This metabolic dysregulation has been linked to remodeling the mitochondrial network to favor fission. However, whether the mitochondrial fission/fusion balance underlies the metabolic reprogramming found early in PH development is unknown. METHODS Utilizing a rat early model of PH, in conjunction with cultured pulmonary endothelial cells (PECs), we utilized metabolic flux assays, Seahorse Bioassays, measurements of electron transport chain (ETC) complex activity, fluorescent microscopy, and molecular approaches to investigate the link between the disruption of mitochondrial dynamics and the early metabolic changes that occur in PH. RESULTS We observed increased fusion mediators, including Mfn1, Mfn2, and Opa1, and unchanged fission mediators, including Drp1 and Fis1, in a two-week monocrotaline-induced PH animal model (early-stage PH). We were able to establish a connection between increases in fusion mediator Mfn1 and metabolic reprogramming. Using an adenoviral expression system to enhance Mfn1 levels in pulmonary endothelial cells and utilizing 13C-glucose labeled substrate, we found increased production of 13C lactate and decreased TCA cycle metabolites, revealing a Warburg phenotype. The use of a 13C5-glutamine substrate showed evidence that hyperfusion also induces oxidative carboxylation. The increase in glycolysis was linked to increased hypoxia-inducible factor 1α (HIF-1α) protein levels secondary to the disruption of cellular bioenergetics and higher levels of mitochondrial reactive oxygen species (mt-ROS). The elevation in mt-ROS correlated with attenuated ETC complexes I and III activities. Utilizing a mitochondrial-targeted antioxidant to suppress mt-ROS, limited HIF-1α protein levels, which reduced cellular glycolysis and reestablished mitochondrial membrane potential. CONCLUSIONS Our data connects mitochondrial fusion-mediated mt-ROS to the Warburg phenotype in early-stage PH development.
Collapse
Affiliation(s)
- Manivannan Yegambaram
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Xutong Sun
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Qing Lu
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Yan Jin
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA
| | | | - Jamie Soto
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA
| | - Saurabh Aggarwal
- Department of Cellular Biology & Pharmacology, Howard Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA; Department of Cellular Biology & Pharmacology, Howard Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Kim Tieu
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Haiwei Gu
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Jeffrey R Fineman
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, 94143, USA; Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Stephen M Black
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA; Department of Cellular Biology & Pharmacology, Howard Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA.
| |
Collapse
|
50
|
Katz MG, Hadas Y, Shtraizent N, Ravvin S, Madjarov JM, Eliyahu E. Unilateral Lung Removal in Combination with Monocrotaline or SU5416 in Rodents: A Reliable Model to Mimic the Pathology of the Human Pulmonary Hypertension. Methods Mol Biol 2024; 2803:173-185. [PMID: 38676893 DOI: 10.1007/978-1-0716-3846-0_13] [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: 04/29/2024]
Abstract
Pulmonary hypertension (PH) is a chronic and progressive disorder characterized by elevated mean pulmonary arterial pressure, pulmonary vascular remodeling, and the development of concentric laminar intimal fibrosis with plexiform lesions. While rodent models have been developed to study PH, they have certain deficiencies and do not entirely replicate the human disease due to the heterogeneity of PH pathology. Therefore, combined models are necessary to study PH. Recent studies have shown that altered pulmonary blood flow is a significant trigger in the development of vascular remodeling and neointimal lesions. One of the most promising rodent models for increased pulmonary flow is the combination of unilateral left pneumonectomy with a "second hit" of monocrotaline (MCT) or SU5416. The removal of one lung in this model forces blood to circulate only in the other lung and induces increased and turbulent pulmonary blood flow. This increased vascular flow leads to progressive remodeling and occlusion of small pulmonary arteries. The second hit by MCT or SU5416 leads to endothelial cell dysfunction, resulting in severe PH and the development of plexiform arteriopathy.
Collapse
Affiliation(s)
- Michael G Katz
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Cardiovascular Surgery and Pediatric Cardiac Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Yoav Hadas
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Shana Ravvin
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jeko M Madjarov
- Atrium Health Sanger Heart and Vascular Institute, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Efrat Eliyahu
- Department of Genetics and Genomic Sciences, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn School for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|