1
|
Shan L, Tang X, Liu Y, Qi W, Zhao H, Zang L, Li Y, Li X. Targeting BMPER as a therapeutic strategy for pulmonary arterial hypertension. Cell Signal 2025; 133:111880. [PMID: 40383173 DOI: 10.1016/j.cellsig.2025.111880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/19/2025] [Accepted: 05/15/2025] [Indexed: 05/20/2025]
Abstract
Pulmonary arterial hypertension is a life-threatening condition marked by pulmonary vascular remodeling, leading to increased pulmonary artery pressure and right heart hypertrophy. This chronic process involves excessive proliferation and migration of endothelial and smooth muscle cells. Our research, involving both pulmonary arterial hypertension patients and animal models, reveals reduced BMPER levels in cases of pulmonary arterial hypertension. In vitro mechanistic studies are performed using human pulmonary artery endothelial cells and smooth muscle cells. Additionally, we demonstrate BMPER function in vivo through its overexpression via an adeno-associated virus. Our findings indicate that BMPER can attenuate cell proliferation and migration in endothelial cells by inhibiting the PI3K/AKT signaling pathway. Additionally, BMPER reduces smooth muscle cell proliferation and migration by inhibiting BMP4 activity through a paracrine mechanism. Furthermore, BMPER expression is regulated by the transcription factor ERG. Notably, in vivo overexpression of BMPER significantly alleviates the progression of pulmonary arterial hypertension. In summary, our study identifies BMPER as a novel therapeutic target for pulmonary arterial hypertension and provides new insights into the underlying mechanisms of the disease.
Collapse
Affiliation(s)
- Lina Shan
- Department of Respiratory Disease, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China
| | - Xiaofeng Tang
- Department of Respiratory Medicine, Huanggang Central Hospital, Huanggang 439000, China
| | - Yi Liu
- Department of Critical Care Medicine, Panzhihua Central Hospital, Panzhihua 61700, China
| | - Weiyi Qi
- Department of Respiratory Disease, First Affiliated Hospital of China Medical University, Shen Yang 400042, China
| | - Hainan Zhao
- Department of Nephrology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Lie Zang
- Department of Neurology, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China
| | - Yanwei Li
- Slow Disease Management Center, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China
| | - Xin Li
- Department of Respiratory Disease, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China.
| |
Collapse
|
2
|
Shen Y, Gleghorn JP. Class III Phosphatidylinositol-3 Kinase/Vacuolar Protein Sorting 34 in Cardiovascular Health and Disease. J Cardiovasc Transl Res 2025; 18:392-407. [PMID: 39821606 PMCID: PMC12043424 DOI: 10.1007/s12265-024-10581-z] [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: 06/24/2024] [Accepted: 12/12/2024] [Indexed: 01/19/2025]
Abstract
Phosphatidylinositol-3 kinases (PI3Ks) play a critical role in maintaining cardiovascular health and the development of cardiovascular diseases (CVDs). Specifically, vacuolar Protein Sorting 34 (VPS34) or PIK3C3, the only member of Class III PI3K, plays an important role in CVD progression. The main function of VPS34 is inducing the production of phosphatidylinositol 3-phosphate, which, together with other essential structural and regulatory proteins in forming VPS34 complexes, further regulates the mammalian target of rapamycin activation, autophagy, and endocytosis. VPS34 is found to have crucial functions in the cardiovascular system, including dictating the proliferation and survival of vascular smooth muscle cells and cardiomyocytes and the formation of thrombosis. This review aims to summarize our current knowledge and recent advances in understanding the function and regulation of VPS34 in cardiovascular health and disease. We also discuss the current development of VPS34 inhibitors and their potential to treat CVDs.
Collapse
Affiliation(s)
- Yuanjun Shen
- Departments of Biomedical Engineering, University of Delaware, Newark, DE, USA.
- School of Pharmacy and Pharmceutical Sciences, Binghamton University, Johnson City, NY, USA.
| | - Jason P Gleghorn
- Departments of Biomedical Engineering, University of Delaware, Newark, DE, USA
- Biological Sciences, University of Delaware, Newark, DE, USA
| |
Collapse
|
3
|
Zhou X, Hu Q, Yu M, Li K. Overexpression of Neural Precursor Cell Expressed Developmentally Downregulated 9 (NEDD9) reduces ox-LDL-induced Anoikis in atherosclerotic vascular endothelial cells. IJC HEART & VASCULATURE 2025; 56:101609. [PMID: 39897415 PMCID: PMC11787488 DOI: 10.1016/j.ijcha.2025.101609] [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: 09/29/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 02/04/2025]
Abstract
Objective This study purposes to explore the action of the Anoikis gene in vascular endothelial cell injury, explore diagnostic biomarkers, and provide new insights into potential molecular mechanisms, as well as offer a new perspective for disease detection and treatment. Methods The Anoikis gene set was used for enrichment analysis on the Gene Expression Omnibus (GEO: GSE100927) dataset, to identify the intersection genes related to Atherosclerosis. Further, the expression and pathway enrichment of Anoikis genes in GSE100927 was investigated. The Least Absolute Shrinkage and Selection Operator (LASSO) method for dimensionality reduction modeling was employed to obtain Atherosclerosis-related genes and construct Anoikis score. The NEDD9, FOSB, and ERCC1 expression in ox-LDL-induced the Bend.3 cells was validated by reverse transcription quantitative polymerase chain reaction (RT-qPCR). Overexpression or silencing NEDD9 on Anoikis in ox-LDL and detachment-induced the Bend.3 cells was analyzed by using Cell Counting Kit-8 (CCK8), 5-Ethynyl-2'-deoxyuridine (EdU), and flow cytometry assays. Results Based on Anoikis gene analysis, NFIL3, NR4A3, ADAMTS4, NEDD9, STX17-AS1, and CSF3 were found to be under-expressed, while FOSB and ERCC1 were found to be over-expressed in the atherosclerosis group compared to the normal group. LASSO regression analysis yielded an Anoikis score = -9.522e-01 × NFIL3 - 3.410 × NEDD9 + 2.728e-01 × ADAMTS4 + 1.178 × FOSB + 5.896e-15 × ERCC1 + 1.558e+01. Compared with the blank group, NEDD9, FOSB, and ERCC1 were under-expressed in the ox-LDL intervention group. si-NEDD9 promoted an increase in reactive oxygen species (ROS) and apoptosis levels in the Bend.3 cells intervened by ox-LDL. Transfection with oe-NEDD9 increased the viability of Bend.3 cells induced by the ox-LDL and detachment, while decreasing ROS and apoptosis levels. Conclusion This study developed a reliable atherosclerotic Anoikis model for predicting endothelial cell injury. During Anoikis genes, the overexpression of NEDD9 reduces ox-LDL and detachment-induced endothelial cell Anoikis.
Collapse
Affiliation(s)
- Xiaowei Zhou
- Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha 410008, PR China
- National Clinical Research Center for Geriatric Disorders,Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Qinghua Hu
- Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha 410008, PR China
- National Clinical Research Center for Geriatric Disorders,Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Meihong Yu
- Department of Gastroenterology, Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, PR China
- Research Center of Digestive Diseases, Central South University, Changsha 410011, Hunan Province, PR China
- Clinical Research Center of Digestive Diseases of Hunan Province, Changsha 410011, Hunan Province, PR China
| | - Kaixuan Li
- Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha 410008, PR China
- National Clinical Research Center for Geriatric Disorders,Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| |
Collapse
|
4
|
Zhou Q, Huang R, Xiong X, Liang Z, Zhang W. Prediction of pulmonary embolism by an explainable machine learning approach in the real world. Sci Rep 2025; 15:835. [PMID: 39755685 PMCID: PMC11700180 DOI: 10.1038/s41598-024-75435-9] [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: 05/22/2024] [Accepted: 10/04/2024] [Indexed: 01/06/2025] Open
Abstract
In recent years, large amounts of researches showed that pulmonary embolism (PE) has become a common disease, and PE remains a clinical challenge because of its high mortality, high disability, high missed and high misdiagnosed rates. To address this, we employed an artificial intelligence-based machine learning algorithm (MLA) to construct a robust predictive model for PE. We retrospectively analyzed 1480 suspected PE patients hospitalized in West China Hospital of Sichuan University between May 2015 and April 2020. 126 features were screened and diverse MLAs were utilized to craft predictive models for PE. Area under the receiver operating characteristic curves (AUC) were used to evaluate their performance and SHapley Additive exPlanation (SHAP) values were utilized to elucidate the prediction model. Regarding the efficacy of the single model that most accurately predicted the outcome, RF demonstrated the highest efficacy in predicting outcomes, with an AUC of 0.776 (95% CI 0.774-0.778). The SHAP summary plot delineated the positive and negative effects of features attributed to the RF prediction model, including D-dimer, activated partial thromboplastin time (APTT), fibrin and fibrinogen degradation products (FFDP), platelet count, albumin, cholesterol, and sodium. Furthermore, the SHAP dependence plot illustrated the impact of individual features on the RF prediction model. Finally, the MLA based PE predicting model was designed as a web page that can be applied to the platform of clinical management. In this study, PE prediction model was successfully established and designed as a web page, facilitating the optimization of early diagnosis and timely treatment strategies to enhance PE patient outcomes.
Collapse
Affiliation(s)
- Qiao Zhou
- Department of Respiratory and Critical Care Medicine, Changhai Hospital, The Second Military Medical University, Shanghai, People's Republic of China
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Ruichen Huang
- Department of Respiratory and Critical Care Medicine, Changhai Hospital, The Second Military Medical University, Shanghai, People's Republic of China
| | - Xingyu Xiong
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China.
| | - Zongan Liang
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China.
| | - Wei Zhang
- Department of Respiratory and Critical Care Medicine, Changhai Hospital, The Second Military Medical University, Shanghai, People's Republic of China.
| |
Collapse
|
5
|
An Y, Xu M, Yan M, Zhang H, Li C, Wang L, Liu C, Dong H, Chen L, Zhang L, Chen Y, Han X, Li Y, Wang D, Gao C. Erythrophagocytosis-induced ferroptosis contributes to pulmonary microvascular thrombosis and thrombotic vascular remodeling in pulmonary arterial hypertension. J Thromb Haemost 2025; 23:158-170. [PMID: 39357568 DOI: 10.1016/j.jtha.2024.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 08/01/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Whether primary or just as a complication from the progression of pulmonary arterial hypertension (PAH), thrombosis seems to be an important player in this condition. The crosstalk between red blood cells (RBCs) and pulmonary microvascular endothelial cells (PMVECs) and their role in PAH remain undefined. OBJECTIVES The goals of this study were to assess the role of RBC-PMVEC interaction in microvascular thrombosis and thrombotic vascular remodeling under hypoxic conditions. METHODS We established an in vitro hypoxic coincubation model of RBC and PMVEC as well as a hypoxic mouse model. We investigated erythrophagocytosis (EP), ferroptosis, thrombosis tendency, and pulmonary hemodynamics in experimental PAH. RESULTS Increased EP in PMVEC triggered ferroptosis, enhanced procoagulant activity, and exacerbated vessel remodeling under hypoxic conditions. In the PAH mouse model induced by chronic hypoxia, EP-induced ferroptosis followed by upregulated TMEM16F led to a high tendency of thrombus formation and thrombotic vascular remodeling. Inhibition of ferroptosis or silencing of TMEM16F could alleviate hypercoagulable phenotype, reverse right ventricular systolic pressure, right ventricular hypertrophy index, and remodeling of pulmonary vessels. CONCLUSION These results illustrate the pathogenic RBC-PMVEC interactions in PAH. Inhibition EP, ferroptosis, or TMEM16F could be a novel therapeutic target to prevent PAH development and thrombotic complications.
Collapse
Affiliation(s)
- Yao An
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Minghui Xu
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Meishan Yan
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Hongyu Zhang
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Caixia Li
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Lifeng Wang
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Caixu Liu
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Haoran Dong
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Li Chen
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Lixin Zhang
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Yingli Chen
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Xu Han
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Yun Li
- Hematology Department, Daqing Oil Field General Hospital, Daqing, China
| | - Dongsheng Wang
- Department of Emergency, the Fifth Affiliated Hospital of Harbin Medical University, Daqing, China
| | - Chunyan Gao
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China.
| |
Collapse
|
6
|
Tsai J, Malik S, Tjen-A-Looi SC. Pulmonary Hypertension: Pharmacological and Non-Pharmacological Therapies. Life (Basel) 2024; 14:1265. [PMID: 39459565 PMCID: PMC11509317 DOI: 10.3390/life14101265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/23/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Pulmonary hypertension (PH) is a severe and chronic disease characterized by increased pulmonary vascular resistance and remodeling, often precipitating right-sided heart dysfunction and death. Although the condition is progressive and incurable, current therapies for the disease focus on multiple different drugs and general supportive therapies to manage symptoms and prolong survival, ranging from medications more specific to pulmonary arterial hypertension (PAH) to exercise training. Moreover, there are multiple studies exploring novel experimental drugs and therapies including unique neurostimulation, to help better manage the disease. Here, we provide a narrative review focusing on current PH treatments that target multiple underlying biochemical mechanisms, including imbalances in vasoconstrictor-vasodilator and autonomic nervous system function, inflammation, and bone morphogenic protein (BMP) signaling. We also focus on the potential of novel therapies for managing PH, focusing on multiple types of neurostimulation including acupuncture. Lastly, we also touch upon the disease's different subgroups, clinical presentations and prognosis, diagnostics, demographics, and cost.
Collapse
Affiliation(s)
- Jason Tsai
- Susan Samueli Integrative Health Institute, College of Health Sciences, University of California-Irvine, Irvine, CA 92617, USA;
| | | | - Stephanie C. Tjen-A-Looi
- Susan Samueli Integrative Health Institute, College of Health Sciences, University of California-Irvine, Irvine, CA 92617, USA;
| |
Collapse
|
7
|
Alladina JW, Giacona FL, Haring AM, Hibbert KA, Medoff BD, Schmidt EP, Thompson T, Maron BA, Alba GA. Circulating Biomarkers of Endothelial Dysfunction Associated With Ventilatory Ratio and Mortality in ARDS Resulting From SARS-CoV-2 Infection Treated With Antiinflammatory Therapies. CHEST CRITICAL CARE 2024; 2:100054. [PMID: 39035722 PMCID: PMC11259037 DOI: 10.1016/j.chstcc.2024.100054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
BACKGROUND The association of plasma biomarkers and clinical outcomes in ARDS resulting from SARS-CoV-2 infection predate the evidence-based use of immunomodulators. RESEARCH QUESTION Which plasma biomarkers are associated with clinical outcomes in patients with ARDS resulting from SARS-CoV-2 infection treated routinely with immunomodulators? STUDY DESIGN AND METHODS We collected plasma from patients with ARDS resulting from SARS-CoV-2 infection within 24 h of admission to the ICU between December 2020 and March 2021 (N = 69). We associated 16 total biomarkers of inflammation (eg, IL-6), coagulation (eg, D-dimer), epithelial injury (eg, surfactant protein D), and endothelial injury (eg, angiopoietin-2) with the primary outcome of in-hospital mortality and secondary outcome of ventilatory ratio (at baseline and day 3). RESULTS Thirty patients (43.5%) died within 60 days. All patients received corticosteroids and 6% also received tocilizumab. Compared with survivors, nonsurvivors demonstrated a higher baseline modified Sequential Organ Failure Assessment score (median, 8.5 [interquartile range (IQR), 7-9] vs 7 [IQR, 5-8]); P = .004), lower Pao2 to Fio2 ratio (median, 153 [IQR, 118-182] vs 184 [IQR, 142-247]; P = .04), and higher ventilatory ratio (median, 2.0 [IQR, 1.9-2.3] vs 1.5 [IQR, 1.4-1.9]; P < .001). No difference was found in inflammatory, coagulation, or epithelial biomarkers between groups. Nonsurvivors showed higher median neural precursor cell expressed, developmentally down-regulated 9 (NEDD9) levels (median, 8.4 ng/mL [IQR, 7.0-11.2 ng/mL] vs 6.9 ng/mL [IQR, 5.5-8.0 ng/mL]; P = .0025), von Willebrand factor domain A2 levels (8.7 ng/mL [IQR, 7.9-9.7 ng/mL] vs 6.5 ng/mL [IQR, 5.7-8.7 ng/mL]; P = .007), angiopoietin-2 levels (9.0 ng/mL [IQR, 7.9-14.1 ng/mL] vs 7.0 ng/mL [IQR, 5.6-10.6 ng/mL]; P = .01), and syndecan-1 levels (15.9 ng/mL [IQR, 14.5-17.5 ng/mL] vs 12.6 ng/mL [IQR, 10.5-16.1 ng/mL]; P = .01). Only NEDD9 level met the adjusted threshold for significance (P < .003). Plasma NEDD9 level was associated with 60-day mortality (adjusted OR, 9.7; 95% CI, 1.6-60.4; P = .015). Syndecan-1 level correlated with both baseline (ρ = 0.4; P = .001) and day 3 ventilatory ratio (ρ = 0.5; P < .001). INTERPRETATION Biomarkers of inflammation, coagulation, and epithelial injury were not associated with clinical outcomes in a small cohort of patients with ARDS uniformly treated with immunomodulators. However, endothelial biomarkers, including plasma NEDD9, were associated with 60-day mortality.
Collapse
Affiliation(s)
- Jehan W Alladina
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Francesca L Giacona
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Alexis M Haring
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Kathryn A Hibbert
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Benjamin D Medoff
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Eric P Schmidt
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Taylor Thompson
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Bradley A Maron
- Department of Medicine; University of Maryland School of Medicine, Baltimore, University of Maryland-Institute for Health Computing, Bethesda, MD
| | - George A Alba
- Division of Pulmonary and Critical Care Medicine, Bethesda, MD, Department of Medicine, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
8
|
Alba GA, Zhou IY, Mascia M, Magaletta M, Alladina JW, Giacona FL, Ginns LC, Caravan P, Maron BA, Montesi SB. Plasma NEDD9 is increased following SARS-CoV-2 infection and associates with indices of pulmonary vascular dysfunction. Pulm Circ 2024; 14:e12356. [PMID: 38500738 PMCID: PMC10946282 DOI: 10.1002/pul2.12356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/31/2023] [Accepted: 03/10/2024] [Indexed: 03/20/2024] Open
Abstract
Compared to healthy volunteers, participants with post-acute sequelae of SARS-CoV-2 infection (PASC) demonstrated increased plasma levels of the prothrombotic protein NEDD9, which associated inversely with indices of pulmonary vascular function. This suggests persistent pulmonary vascular dysfunction may play a role in the pathobiology of PASC.
Collapse
Affiliation(s)
- George A. Alba
- Harvard Medical SchoolBostonMassachusettsUSA
- Division of Pulmonary and Critical Care MedicineMassachusetts General HospitalBostonMassachusettsUSA
| | - Iris Y. Zhou
- Harvard Medical SchoolBostonMassachusettsUSA
- Department of Radiology, Athinoula A. Martinos Center for Biomedical ImagingMassachusetts General HospitalBostonMassachusettsUSA
- Institute for Innovation in ImagingMassachusetts General HospitalBostonMassachusettsUSA
| | - Molly Mascia
- Harvard Medical SchoolBostonMassachusettsUSA
- Division of Pulmonary and Critical Care MedicineMassachusetts General HospitalBostonMassachusettsUSA
| | - Michael Magaletta
- Department of Radiology, Athinoula A. Martinos Center for Biomedical ImagingMassachusetts General HospitalBostonMassachusettsUSA
| | - Jehan W. Alladina
- Harvard Medical SchoolBostonMassachusettsUSA
- Division of Pulmonary and Critical Care MedicineMassachusetts General HospitalBostonMassachusettsUSA
| | - Francesca L. Giacona
- Division of Pulmonary and Critical Care MedicineMassachusetts General HospitalBostonMassachusettsUSA
| | - Leo C. Ginns
- Harvard Medical SchoolBostonMassachusettsUSA
- Division of Pulmonary and Critical Care MedicineMassachusetts General HospitalBostonMassachusettsUSA
| | - Peter Caravan
- Harvard Medical SchoolBostonMassachusettsUSA
- Department of Radiology, Athinoula A. Martinos Center for Biomedical ImagingMassachusetts General HospitalBostonMassachusettsUSA
- Institute for Innovation in ImagingMassachusetts General HospitalBostonMassachusettsUSA
| | - Bradley A. Maron
- Division of Cardiovascular MedicineBrigham and Women's HospitalBostonMassachusettsUSA
- Department of MedicineUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Sydney B. Montesi
- Harvard Medical SchoolBostonMassachusettsUSA
- Division of Pulmonary and Critical Care MedicineMassachusetts General HospitalBostonMassachusettsUSA
- Institute for Innovation in ImagingMassachusetts General HospitalBostonMassachusettsUSA
| |
Collapse
|
9
|
Johnson S, Sommer N, Cox-Flaherty K, Weissmann N, Ventetuolo CE, Maron BA. Pulmonary Hypertension: A Contemporary Review. Am J Respir Crit Care Med 2023; 208:528-548. [PMID: 37450768 PMCID: PMC10492255 DOI: 10.1164/rccm.202302-0327so] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 07/14/2023] [Indexed: 07/18/2023] Open
Abstract
Major advances in pulmonary arterial hypertension, pulmonary hypertension (PH) associated with lung disease, and chronic thromboembolic PH cast new light on the pathogenetic mechanisms, epidemiology, diagnostic approach, and therapeutic armamentarium for pulmonary vascular disease. Here, we summarize key basic, translational, and clinical PH reports, emphasizing findings that build on current state-of-the-art research. This review includes cutting-edge progress in translational pulmonary vascular biology, with a guide to the diagnosis of patients in clinical practice, incorporating recent PH definition revisions that continue emphasis on early detection of disease. PH management is reviewed including an overview of the evolving considerations for the approach to treatment of PH in patients with cardiopulmonary comorbidities, as well as a discussion of the groundbreaking sotatercept data for the treatment of pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Shelsey Johnson
- The Pulmonary Center, Division of Pulmonary, Allergy, Sleep and Critical Care, Boston University School of Medicine, Boston, Massachusetts
- Department of Pulmonary and Critical Care Medicine and
| | - Natascha Sommer
- Excellence Cluster Cardiopulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University, Giessen, Germany
| | | | - Norbert Weissmann
- Excellence Cluster Cardiopulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University, Giessen, Germany
| | - Corey E. Ventetuolo
- Department of Medicine and
- Department of Health Services, Policy and Practice, Brown University, Providence, Rhode Island
| | - Bradley A. Maron
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, and Harvard Medical School, Boston, Massachusetts
- Department of Cardiology and Department of Pulmonary, Allergy, Sleep, and Critical Care Medicine, VA Boston Healthcare System, Boston, Massachusetts
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland; and
- The University of Maryland-Institute for Health Computing, Bethesda, Maryland
| |
Collapse
|
10
|
Wei H, Xiao X, Zeng S, Liu Y, Liu X, Zeng T, Xu P, Xia W, Guo L, Hong S, Lv W, Chen Y, Xu R. Alterations in factors associated with diabetic retinopathy combined with thrombosis: A review. Medicine (Baltimore) 2023; 102:e34373. [PMID: 37543800 PMCID: PMC10403020 DOI: 10.1097/md.0000000000034373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 05/17/2023] [Accepted: 06/26/2023] [Indexed: 08/07/2023] Open
Abstract
Diabetic retinopathy (DR) is one of the most common and serious microvascular complications of diabetes mellitus, the incidence of which has been increasing annually, and it is the main cause of vision loss in diabetic patients and a common cause of blindness. It is now found that thrombosis plays a crucial role in the disease progression in DR patients, and the final vision loss in DR may be related to the occurrence of thrombosis in the retinal vessels, which is dominated by abnormal endothelial cell function, together with platelet dysfunction, imbalance of coagulation and fibrinolytic function, and related alterations of inflammatory factors leading to the main cause of thrombotic disease in DR patients. In this review, we examine the role between DR and thrombosis and the association of each factor, including endothelial dysfunction; platelet dysfunction; coagulation-fibrinolytic imbalance; and alterations in inflammatory factors.
Collapse
Affiliation(s)
- Haiyan Wei
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Xiaoping Xiao
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, P.R. China
- Provincial Key Laboratory of Low-Carbon Solid Waste Recycling, Gannan Normal University, Ganzhou, P. R. China
| | - Shuqin Zeng
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Ye Liu
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Xiaofang Liu
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Tianyu Zeng
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Pengxiang Xu
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Wenyan Xia
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Li Guo
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Shihua Hong
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Weiming Lv
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Yijian Chen
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Rong Xu
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| |
Collapse
|
11
|
Elhage Hassan M, Vinales J, Perkins S, Sandesara P, Aggarwal V, Jaber WA. Pathogenesis, Diagnosis, and Management of Chronic Thromboembolic Pulmonary Hypertension. Interv Cardiol Clin 2023; 12:e37-e49. [PMID: 38964822 DOI: 10.1016/j.iccl.2024.04.003] [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: 07/06/2024]
Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) is thought to occur as a sequelae of thromboembolic processes in the pulmonary vasculature. The pathophysiology of CTEPH is multifactorial, including impaired fibrinolysis, endothelial dysregulation, and hypoxic adaptations. The diagnosis of CTEPH is typically delayed considering the nonspecific nature of the symptoms, lack of screening, and relatively low incidence. Diagnostic tools include ventilation-perfusion testing, echocardiography, cardiac catheterization, and pulmonary angiography. The only potentially curative treatment for CTEPH is pulmonary endarterectomy However, approximately 40% of patients are inoperable. Currently, only Riociguat is Food and Drug Administration approved specifically for CTEPH, with additional drug trials underway.
Collapse
Affiliation(s)
- Malika Elhage Hassan
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1364 Clifton Road Northeast Suite F606, Atlanta, GA 30322, USA
| | - Jorge Vinales
- Department of Medicine, University of Michigan Medical School, 1301 Catherine Street, Ann Arbor, MI 48109, USA
| | - Sidney Perkins
- Department of Internal Medicine, University of Michigan Medical School, 1500 E Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Pratik Sandesara
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1364 Clifton Road Northeast Suite F606, Atlanta, GA 30322, USA
| | - Vikas Aggarwal
- Department of Cardiology, Henry Ford Medical Center, 2799 W Grand Blvd, K-2 Cath Admin Suite, Detroit, MI 48206, USA
| | - Wissam A Jaber
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1364 Clifton Road Northeast Suite F606, Atlanta, GA 30322, USA.
| |
Collapse
|
12
|
Ye Y, Xu Q, Wuren T. Inflammation and immunity in the pathogenesis of hypoxic pulmonary hypertension. Front Immunol 2023; 14:1162556. [PMID: 37215139 PMCID: PMC10196112 DOI: 10.3389/fimmu.2023.1162556] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 04/25/2023] [Indexed: 05/24/2023] Open
Abstract
Hypoxic pulmonary hypertension (HPH) is a complicated vascular disorder characterized by diverse mechanisms that lead to elevated blood pressure in pulmonary circulation. Recent evidence indicates that HPH is not simply a pathological syndrome but is instead a complex lesion of cellular metabolism, inflammation, and proliferation driven by the reprogramming of gene expression patterns. One of the key mechanisms underlying HPH is hypoxia, which drives immune/inflammation to mediate complex vascular homeostasis that collaboratively controls vascular remodeling in the lungs. This is caused by the prolonged infiltration of immune cells and an increase in several pro-inflammatory factors, which ultimately leads to immune dysregulation. Hypoxia has been associated with metabolic reprogramming, immunological dysregulation, and adverse pulmonary vascular remodeling in preclinical studies. Many animal models have been developed to mimic HPH; however, many of them do not accurately represent the human disease state and may not be suitable for testing new therapeutic strategies. The scientific understanding of HPH is rapidly evolving, and recent efforts have focused on understanding the complex interplay among hypoxia, inflammation, and cellular metabolism in the development of this disease. Through continued research and the development of more sophisticated animal models, it is hoped that we will be able to gain a deeper understanding of the underlying mechanisms of HPH and implement more effective therapies for this debilitating disease.
Collapse
Affiliation(s)
- Yi Ye
- Research Center for High Altitude Medicine, Qinghai University, Xining, China
- High-Altitude Medicine Key Laboratory of the Ministry of Education, Xining, China
- Qinghai Provincial Key Laboratory for Application of High-Altitude Medicine, Xining, China
- Qinghai-Utah Key Laboratory of High-Altitude Medicine, Xining, China
| | - Qiying Xu
- Research Center for High Altitude Medicine, Qinghai University, Xining, China
- High-Altitude Medicine Key Laboratory of the Ministry of Education, Xining, China
- Qinghai Provincial Key Laboratory for Application of High-Altitude Medicine, Xining, China
- Qinghai-Utah Key Laboratory of High-Altitude Medicine, Xining, China
| | - Tana Wuren
- Research Center for High Altitude Medicine, Qinghai University, Xining, China
- High-Altitude Medicine Key Laboratory of the Ministry of Education, Xining, China
- Qinghai Provincial Key Laboratory for Application of High-Altitude Medicine, Xining, China
- Qinghai-Utah Key Laboratory of High-Altitude Medicine, Xining, China
| |
Collapse
|
13
|
Maron BA, Kleiner DE, Arons E, Wertheim BM, Sharma NS, Haley KJ, Samokhin AO, Rowin EJ, Maron MS, Rosing DR, Maron BJ. Evidence of Advanced Pulmonary Vascular Remodeling in Obstructive Hypertrophic Cardiomyopathy With Pulmonary Hypertension. Chest 2023; 163:678-686. [PMID: 36243062 PMCID: PMC9993337 DOI: 10.1016/j.chest.2022.09.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/28/2022] [Accepted: 09/29/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Elevated mean pulmonary artery pressure (mPAP) is common in patients with hypertrophic cardiomyopathy (HCM) and heart failure symptoms. However, dynamic left ventricular (LV) outflow tract obstruction may confound interpretation of pulmonary hypertension (PH) pathophysiologic features in HCM when relying on resting invasive hemodynamic data alone. RESEARCH QUESTION Do structural changes to the lung vasculature clarify PH pathophysiologic features in patients with HCM with progressive heart failure? STUDY DESIGN AND METHODS Clinical data and ultrarare lung autopsy specimens were acquired retrospectively from the National Institutes of Health (1975-1992). Patients were included based on the availability of lung tissue and recorded mPAP. Discarded tissue from rejected lung donors served as control specimens. Histomorphology was performed on pulmonary arterioles and veins. Comparisons were calculated using the Student t test and Mann-Whitney U test; Pearson correlation was used to assess association between morphometric measurements and HCM cardiac and hemodynamic measurements. RESULTS The HCM cohort (n = 7; mean ± SD age, 43 ± 18 years; 71% men) showed maximum mean ± SD LV wall thickness of 25 ± 2.8 mm, mean ± SD outflow tract gradient of 90 ± 30 mm Hg, median mPAP of 25 mm Hg (interquartile range [IQR], 6 mm Hg), median pulmonary artery wedge pressure (PAWP) of 16 mm Hg (IQR, 4 mm Hg), and median pulmonary vascular resistance of 1.8 Wood units (WU; IQR, 2.4 WU). Compared with control samples (n = 5), patients with HCM showed greater indexed pulmonary arterial hypertrophy (20.7 ± 7.2% vs 49.7 ± 12%; P < .001) and arterial wall fibrosis (11.5 ± 3.4 mm vs 21.0 ± 4.7 mm; P < .0001), which correlated with mPAP (r = 0.84; P = .018), PAWP (r = 0.74; P = .05), and LV outflow tract gradient (r = 0.78; P = .035). Compared with control samples, pulmonary vein thickness was increased by 2.9-fold (P = .008) in the HCM group, which correlated with mPAP (r = 0.81; P = .03) and LV outflow tract gradient (r = 0.83; P = .02). INTERPRETATION To the best of our knowledge, these data demonstrate for the first time that in patients with obstructive HCM, heart failure is associated with pathogenic pulmonary vascular remodeling even when mPAP is elevated only mildly. These observations clarify PH pathophysiologic features in HCM, with future implications for clinical strategies that mitigate outflow tract obstruction.
Collapse
Affiliation(s)
- Bradley A Maron
- Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA.
| | - David E Kleiner
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD
| | - Elena Arons
- Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Bradley M Wertheim
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Nirmal S Sharma
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Kathleen J Haley
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Andriy O Samokhin
- Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Ethan J Rowin
- HCM Center, Lahey Hospital and Medical Center, Burlington, MA
| | - Martin S Maron
- HCM Center, Lahey Hospital and Medical Center, Burlington, MA
| | - Douglas R Rosing
- Cardiovascular Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Barry J Maron
- HCM Center, Lahey Hospital and Medical Center, Burlington, MA
| |
Collapse
|
14
|
Hansmann G, Chouvarine P, Diekmann F, Giera M, Ralser M, Mülleder M, von Kaisenberg C, Bertram H, Legchenko E, Hass R. Human umbilical cord mesenchymal stem cell-derived treatment of severe pulmonary arterial hypertension. NATURE CARDIOVASCULAR RESEARCH 2022; 1:568-576. [PMID: 39195868 PMCID: PMC11358026 DOI: 10.1038/s44161-022-00083-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 05/09/2022] [Indexed: 08/29/2024]
Abstract
Here we report application of human umbilical cord mesenchymal stem cell (HUCMSC)-derived therapy for pulmonary arterial hypertension (PAH). A 3-year-old female presented with heritable PAH associated with hereditary hemorrhagic telangiectasia and was treated for 6 months with serial intravascular infusions of conditioned media (CM) from allogenic HUCMSCs. The treatment markedly improved clinical and hemodynamic parameters and decreased blood plasma markers of vascular fibrosis, injury and inflammation. A comparative analysis of single-cell RNA sequencing data collected from three HUCMSCs and two human umbilical vein endothelial cell (HUVEC) controls identified eight common cell clusters, all of which indicated regenerative potential specific for HUCMSCs. The properties of HUCMSCs were validated by untargeted label-free quantitation of the cell and CM proteome, suggesting increased activity of regeneration, autophagy and anti-inflammation pathways and mitochondrial function. Prostaglandin analysis demonstrated increased HUCMSC secretion of prostaglandin E2, known for its regenerative capacity. Additional prospective clinical studies are warranted to confirm and further explore the benefits of HUCMSC-derived therapy for PAH.
Collapse
Affiliation(s)
- Georg Hansmann
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany.
- European Pediatric Pulmonary Vascular Disease Network, Berlin, Germany.
| | - Philippe Chouvarine
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany
- European Pediatric Pulmonary Vascular Disease Network, Berlin, Germany
| | - Franziska Diekmann
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany
- European Pediatric Pulmonary Vascular Disease Network, Berlin, Germany
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Markus Ralser
- Department of Biochemistry, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Mülleder
- Department of Biochemistry, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Constantin von Kaisenberg
- Departments of Obstetrics, Gynecology and Reproductive Medicine, Hannover Medical School, Hannover, Germany
| | - Harald Bertram
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany
| | - Ekaterina Legchenko
- Department of Pediatric Cardiology and Critical Care, Hannover Medical School, Hannover, Germany
- European Pediatric Pulmonary Vascular Disease Network, Berlin, Germany
| | - Ralf Hass
- Biochemistry and Tumor Biology Lab, Department of Gynecology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
15
|
Rhodes CJ, Sweatt AJ, Maron BA. Harnessing Big Data to Advance Treatment and Understanding of Pulmonary Hypertension. Circ Res 2022; 130:1423-1444. [PMID: 35482840 PMCID: PMC9070103 DOI: 10.1161/circresaha.121.319969] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Pulmonary hypertension is a complex disease with multiple causes, corresponding to phenotypic heterogeneity and variable therapeutic responses. Advancing understanding of pulmonary hypertension pathogenesis is likely to hinge on integrated methods that leverage data from health records, imaging, novel molecular -omics profiling, and other modalities. In this review, we summarize key data sets generated thus far in the field and describe analytical methods that hold promise for deciphering the molecular mechanisms that underpin pulmonary vascular remodeling, including machine learning, network medicine, and functional genetics. We also detail how genetic and subphenotyping approaches enable earlier diagnosis, refined prognostication, and optimized treatment prediction. We propose strategies that identify functionally important molecular pathways, bolstered by findings across multi-omics platforms, which are well-positioned to individualize drug therapy selection and advance precision medicine in this highly morbid disease.
Collapse
Affiliation(s)
- Christopher J Rhodes
- Department of Medicine, National Heart and Lung Institute, Imperial College London, United Kingdom (C.J.R.)
| | - Andrew J Sweatt
- Department of Medicine, National Heart and Lung Institute, Imperial College London, United Kingdom (C.J.R.)
| | - Bradley A Maron
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (B.A.M.).,Division of Cardiology, VA Boston Healthcare System, West Roxbury, MA (B.A.M.)
| |
Collapse
|
16
|
Rizzo AN, Yuan JX. NEDD9 provides mechanistic insight into the coagulopathy of COVID19. Pulm Circ 2022; 12:e12087. [PMID: 35599982 PMCID: PMC9111000 DOI: 10.1002/pul2.12087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 04/26/2022] [Indexed: 11/11/2022] Open
Affiliation(s)
- Alicia N. Rizzo
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine University of Colorado Aurora CO 80045
| | - Jason X.‐J. Yuan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine University of California, San Diego La Jolla CA 92093
| |
Collapse
|
17
|
Alba GA, Samokhin AO, Wang R, Wertheim BM, Haley KJ, Padera RF, Vargas SO, Rosas IO, Hariri LP, Shih A, Thompson BT, Mitchell RN, Maron BA. Pulmonary endothelial NEDD9 and the prothrombotic pathophenotype of acute respiratory distress syndrome due to SARS-CoV-2 infection. Pulm Circ 2022; 12:e12071. [PMID: 35599981 PMCID: PMC9111030 DOI: 10.1002/pul2.12071] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 12/15/2022] Open
Abstract
The pathobiology of in situ pulmonary thrombosis in acute respiratory distress syndrome (ARDS) due to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection is incompletely characterized. In human pulmonary artery endothelial cells (HPAECs), hypoxia increases neural precursor cell expressed, developmentally downregulated 9 (NEDD9) and induces expression of a prothrombotic NEDD9 peptide (N9P) on the extracellular plasma membrane surface. We hypothesized that the SARS-CoV-2-ARDS pathophenotype involves increased pulmonary endothelial N9P. Paraffin-embedded autopsy lung specimens were acquired from patients with SARS-CoV-2-ARDS (n = 13), ARDS from other causes (n = 10), and organ donor controls (n = 5). Immunofluorescence characterized the expression of N9P, fibrin, and transcription factor 12 (TCF12), a putative binding target of SARS-CoV-2 and known transcriptional regulator of NEDD9. We performed RNA-sequencing on normal HPAECs treated with normoxia or hypoxia (0.2% O2) for 24 h. Immunoprecipitation-liquid chromatography-mass spectrometry (IP-LC-MS) profiled protein-protein interactions involving N9P relevant to thrombus stabilization. Hypoxia increased TCF12 messenger RNA significantly compared to normoxia in HPAECs in vitro (+1.19-fold, p = 0.001; false discovery rate = 0.005), and pulmonary endothelial TCF12 expression was increased threefold in SARS-CoV-2-ARDS versus donor control lungs (p < 0.001). Compared to donor controls, pulmonary endothelial N9P-fibrin colocalization was increased in situ in non-SARS-CoV-2-ARDS and SARS-CoV-2-ARDS decedents (3.7 ± 1.2 vs. 10.3 ± 3.2 and 21.8 ± 4.0 arb. units, p < 0.001). However, total pulmonary endothelial N9P was increased significantly only in SARS-CoV-2-ARDS versus donor controls (15 ± 4.2 vs. 6.3 ± 0.9 arb. units, p < 0.001). In HPAEC plasma membrane isolates, IP-LC-MS identified a novel protein-protein interaction between NEDD9 and the β3-subunit of the αvβ3-integrin, which regulates fibrin anchoring to endothelial cells. In conclusion, lethal SARS-CoV-2-ARDS is associated with increased pulmonary endothelial N9P expression and N9P-fibrin colocalization in situ. Further investigation is needed to determine the pathogenetic and potential therapeutic relevance of N9P to the thrombotic pathophenotype of SARS-CoV-2-ARDS.
Collapse
Affiliation(s)
- George A. Alba
- Division of Pulmonary and Critical Care MedicineMassachusetts General HospitalBostonMassachusettsUSA
| | - Andriy O. Samokhin
- Division of Cardiovascular MedicineBrigham and Women's HospitalBostonMassachusettsUSA
| | - Rui‐Sheng Wang
- Division of Cardiovascular MedicineBrigham and Women's HospitalBostonMassachusettsUSA
| | - Bradley M. Wertheim
- Division of Pulmonary and Critical Care MedicineBrigham and Women's HospitalBostonMassachusettsUSA
| | - Kathleen J. Haley
- Division of Pulmonary and Critical Care MedicineBrigham and Women's HospitalBostonMassachusettsUSA
| | - Robert F. Padera
- Department of PathologyBrigham and Women's HospitalBostonMassachusettsUSA
| | - Sara O. Vargas
- Department of PathologyBoston Children's HospitalBostonMassachusettsUSA
| | - Ivan O. Rosas
- Division of Pulmonary and Critical Care MedicineBaylor College of MedicineHoustonTexasUSA
| | - Lida P. Hariri
- Division of Pulmonary and Critical Care MedicineMassachusetts General HospitalBostonMassachusettsUSA
- Department of PathologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Angela Shih
- Department of PathologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Boyd Taylor Thompson
- Division of Pulmonary and Critical Care MedicineMassachusetts General HospitalBostonMassachusettsUSA
| | | | - Bradley A. Maron
- Division of Cardiovascular MedicineBrigham and Women's HospitalBostonMassachusettsUSA
| |
Collapse
|
18
|
Alba GA, Atri D, Darbha S, Singh I, Tapson VF, Lewis MI, Chun HJ, Yu YR, Maron BA, Rajagopal S. Chronic Thromboembolic Pulmonary Hypertension: the Bench. Curr Cardiol Rep 2021; 23:141. [PMID: 34410515 DOI: 10.1007/s11886-021-01572-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/27/2021] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Chronic thromboembolic pulmonary hypertension (CTEPH) is an uncommon complication of acute pulmonary embolism (PE), in which the red, platelet-rich thrombus does not resolve but forms into an organized yellow, fibrotic scar-like obstruction in the pulmonary vasculature. Here we review the pathobiology of CTEPH. RECENT FINDINGS Our current knowledge has predominantly been informed by studies of human samples and animal models that are inherently limited in their ability to recapitulate all aspects of the disease. These studies have identified alterations in platelet biology and inflammation in the formation of a scar-like thrombus that comprised endothelial cells, myofibroblasts, and immune cells, along with a small vessel pulmonary arterial hypertension-like vasculopathy. The development of CTEPH-specific therapies is currently hindered by a limited knowledge of its pathobiology. The development of new CTEPH medical therapies will require new insights into its pathobiology that bridge the gap from bench to bedside.
Collapse
Affiliation(s)
- George A Alba
- Division of Pulmonary and Critical Care, Massachusetts General Hospital, Boston, MA, USA
| | - Deepak Atri
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Sriranjani Darbha
- College of Natural Sciences, The University of Texas, Austin, TX, USA
| | - Inderjit Singh
- Division of Pulmonary, Critical Care, and Sleep Medicine, Yale New Haven Hospital and Yale School of Medicine, New Haven, CT, USA
| | - Victor F Tapson
- Division of Pulmonary and Critical Care Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michael I Lewis
- Division of Pulmonary and Critical Care Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hyung J Chun
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, USA
| | - Yen-Rei Yu
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Bradley A Maron
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Section of Cardiology, Veterans Affairs Boston Healthcare System, Boston, MA, USA
| | - Sudarshan Rajagopal
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
19
|
Wang J, Yang K, Yuan JXJ. NEDD9, a Hypoxia-upregulated Mediator for Pathogenic Platelet-Endothelial Cell Interaction in Pulmonary Hypertension. Am J Respir Crit Care Med 2021; 203:1455-1458. [PMID: 33770456 PMCID: PMC8483222 DOI: 10.1164/rccm.202101-0007ed] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- Jian Wang
- Department of Medicine University of California, San Diego La Jolla, California and.,State Key Laboratory of Respiratory Disease The First Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangdong, China
| | - Kai Yang
- State Key Laboratory of Respiratory Disease The First Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangdong, China
| | - Jason X-J Yuan
- Department of Medicine University of California, San Diego La Jolla, California and
| |
Collapse
|
20
|
Saito J, Kojima T, Tanifuji S, Kato Y, Oka S, Ichikawa Y, Miyagi E, Tachibana T, Asou T, Yokoyama U. Transcriptome Analysis Reveals Differential Gene Expression between the Closing Ductus Arteriosus and the Patent Ductus Arteriosus in Humans. J Cardiovasc Dev Dis 2021; 8:jcdd8040045. [PMID: 33923468 PMCID: PMC8073410 DOI: 10.3390/jcdd8040045] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/08/2021] [Accepted: 04/14/2021] [Indexed: 12/14/2022] Open
Abstract
The ductus arteriosus (DA) immediately starts closing after birth. This dynamic process involves DA-specific properties, including highly differentiated smooth muscle, sparse elastic fibers, and intimal thickening (IT). Although several studies have demonstrated DA-specific gene expressions using animal tissues and human fetuses, the transcriptional profiles of the closing DA and the patent DA remain largely unknown. We performed transcriptome analysis using four human DA samples. The three closing DA samples exhibited typical DA morphology, but the patent DA exhibited aorta-like elastic lamellae and poorly formed IT. A cluster analysis revealed that samples were clearly divided into two major clusters, the closing DA and patent DA clusters, and showed distinct gene expression profiles in IT and the tunica media of the closing DA samples. Cardiac neural crest-related genes such as JAG1 were highly expressed in the tunica media and IT of the closing DA samples compared to the patent DA sample. Abundant protein expressions of jagged 1 and the differentiated smooth muscle marker calponin were observed in the closing DA samples but not in the patent DA sample. Second heart field-related genes such as ISL1 were enriched in the patent DA sample. These data indicate that the patent DA may have different cell lineages compared to the closing DA.
Collapse
Affiliation(s)
- Junichi Saito
- Department of Physiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan; (J.S.); (T.K.); (S.T.); (Y.K.); (S.O.)
| | - Tomoyuki Kojima
- Department of Physiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan; (J.S.); (T.K.); (S.T.); (Y.K.); (S.O.)
- Department of Obstetrics and Gynecology, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan;
| | - Shota Tanifuji
- Department of Physiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan; (J.S.); (T.K.); (S.T.); (Y.K.); (S.O.)
| | - Yuko Kato
- Department of Physiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan; (J.S.); (T.K.); (S.T.); (Y.K.); (S.O.)
| | - Sayuki Oka
- Department of Physiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan; (J.S.); (T.K.); (S.T.); (Y.K.); (S.O.)
| | - Yasuhiro Ichikawa
- Department of Cardiovascular Surgery, Kanagawa Children’s Medical Center, 2-138-4 Mutsukawa, Minami-ku, Yokohama, Kanagawa 232-8555, Japan; (Y.I.); (T.T.); (T.A.)
| | - Etsuko Miyagi
- Department of Obstetrics and Gynecology, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan;
| | - Tsuyoshi Tachibana
- Department of Cardiovascular Surgery, Kanagawa Children’s Medical Center, 2-138-4 Mutsukawa, Minami-ku, Yokohama, Kanagawa 232-8555, Japan; (Y.I.); (T.T.); (T.A.)
| | - Toshihide Asou
- Department of Cardiovascular Surgery, Kanagawa Children’s Medical Center, 2-138-4 Mutsukawa, Minami-ku, Yokohama, Kanagawa 232-8555, Japan; (Y.I.); (T.T.); (T.A.)
| | - Utako Yokoyama
- Department of Physiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan; (J.S.); (T.K.); (S.T.); (Y.K.); (S.O.)
- Correspondence: ; Tel.: +81-3-3351-6141
| |
Collapse
|