1
|
Lewis CV, Nguyen TTN, Porfilio TE, Burciaga SD, Posey JN, Jordan M, Colon Hidalgo D, Stenmark KR, Mickael C, Sul C, Oberley-Deegan RE, Delaney C, Nozik ES. Vascular EC-SOD limits the accumulation, proinflammatory profibrotic reprogramming, and hyaluronan binding of interstitial macrophages in hypoxia. Am J Physiol Lung Cell Mol Physiol 2025; 328:L885-L900. [PMID: 40332405 DOI: 10.1152/ajplung.00399.2024] [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: 12/09/2024] [Revised: 03/04/2025] [Accepted: 05/02/2025] [Indexed: 05/08/2025] Open
Abstract
Dysregulated redox signaling contributes to pulmonary hypertension (PH) and vascular depletion of the redox enzyme extracellular superoxide dismutase (EC-SOD) from smooth muscle cells [EC-SOD SMC knockout (KO)] worsens chronic hypoxic PH. Given the important role of macrophages in PH, this study aimed to determine if interstitial macrophages (IMs) and their interactions with hyaluronan (HA), a component of extracellular matrix (ECM), are modulated by vascular EC-SOD. Floxed wild-type, EC-SOD SMC KO, and SOD mimetic- or vehicle-treated mice were exposed to hypobaric hypoxia [∼10% fraction of inspired oxygen ([Formula: see text])], for 4, 14, or 21 days. Using flow cytometry, we demonstrated that the transient increase in IMs at day 4 was exacerbated in EC-SOD SMC KO mice and prevented with SOD mimetic pretreatment. Highlighting the importance of targeting vascular oxidative stress in the early response to hypoxia, pretreatment with a single dose of EC-SOD mimetic decreased right ventricular systolic pressure, right ventricular hypertrophy, and small vessel muscularization at day 21. To assess IM phenotypic reprogramming in hypoxia, RNA-seq was performed on flow-sorted IMs revealing baseline proinflammatory activation and enhanced activation of vascular and ECM remodeling pathways in response to hypoxia in EC-SOD SMC KO IMs compared with controls. To further investigate the ECM remodeling response, we quantified IMs expressing the lymphatic vessel endothelial hyaluronan receptor 1 (Lyve1), and IM-hyaluronan binding. Lyve1+ IMs and Lyve1+ HA+ IMs were increased in response to hypoxia in EC-SOD SMC KO mice and accumulated in the perivascular space of the lung. In conclusion, vascular EC-SOD limits IM accumulation and proinflammatory profibrotic IM signaling, including perivascular accumulation of Lyve1+ IMs and their binding to hyaluronan.NEW & NOTEWORTHY Expression of the redox enzyme EC-SOD limits PH severity. Using vascular-selective EC-SOD depletion and SOD mimetic treatment in chronic hypoxic PH, we demonstrated that EC-SOD limits the hypoxia-induced accumulation of IMs. IMs from mice with low vascular EC-SOD were proinflammatory at baseline and enhanced ECM remodeling pathway activation in response to hypoxia. We identified Lyve1+ IMs as a perivascular, ECM-interacting subset that accumulate in hypoxia and could contribute to vascular remodeling in PH.
Collapse
Affiliation(s)
- Caitlin V Lewis
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Pediatric Critical Care Medicine, Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado, United States
| | - Thi-Tina N Nguyen
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Pediatric Critical Care Medicine, Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado, United States
| | - Timothy E Porfilio
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Pediatric Critical Care Medicine, Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado, United States
| | - Samuel D Burciaga
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Pediatric Critical Care Medicine, Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado, United States
| | - Janelle N Posey
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Neonatology, Department of Pediatrics, Division of Cardiology, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado, United States
| | - Mariah Jordan
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Neonatology, Department of Pediatrics, Division of Cardiology, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado, United States
| | - Daniel Colon Hidalgo
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Pediatric Critical Care Medicine, Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado, United States
| | - Kurt R Stenmark
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Pediatric Critical Care Medicine, Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado, United States
| | - Claudia Mickael
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, Colorado, United States
| | - Christina Sul
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Pediatric Critical Care Medicine, Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado, United States
| | - Rebecca E Oberley-Deegan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Cassidy Delaney
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Neonatology, Department of Pediatrics, Division of Cardiology, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado, United States
| | - Eva S Nozik
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Pediatric Critical Care Medicine, Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado, United States
| |
Collapse
|
2
|
Mora Massad K, Dai Z, Petrache I, Ventetuolo CE, Lahm T. Lung endothelial cell heterogeneity in health and pulmonary vascular disease. Am J Physiol Lung Cell Mol Physiol 2025; 328:L877-L884. [PMID: 39772753 PMCID: PMC12116231 DOI: 10.1152/ajplung.00296.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/16/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025] Open
Abstract
Lung endothelial cells (ECs) are essential for maintaining organ function and homeostasis. Despite sharing some common features with ECs from organ systems, lung ECs exhibit significant heterogeneity in morphology, function, and gene expression. This heterogeneity is increasingly recognized as a key contributor to the development of pulmonary diseases like pulmonary hypertension (PH). In this mini-review, we explore the evolving understanding of lung EC heterogeneity, particularly through the lens of single-cell RNA sequencing (scRNA-seq) technologies. These advances have provided unprecedented insights into the diverse EC subpopulations, their specific roles, and the disturbances in their homeostatic functions that contribute to PH pathogenesis. In particular, these studies identified novel and functionally distinct cell types such as aerocytes and general capillary ECs that are critical for maintaining lung function in health and disease. In addition, multiple novel pathways and mechanisms have been identified that contribute to aberrant pulmonary vascular remodeling in PH. Emerging techniques like single-nucleus RNA sequencing and spatial transcriptomics have further pushed the field forward by discovering novel disease mediators. As research continues to leverage these advanced techniques, the field is poised to uncover novel EC subtypes and disease mechanisms, paving the way for new therapeutic targets in PH and other lung diseases.
Collapse
Grants
- 4IPA1275127 American Heart Association (AHA)
- R01 HL077328 NHLBI NIH HHS
- P01 HL158507 NHLBI NIH HHS
- Reuben M. Chernaick Fellowship
- R01HL169509 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- I01 BX002042 BLRD VA
- R01 HL141268 NHLBI NIH HHS
- Borstein Family Foundation
- R01HL144727 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- Colorado Pulmonary Vascular Disease Award
- R01 HL170096 NHLBI NIH HHS
- R01HL170096 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL158596 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL144727 NHLBI NIH HHS
- HL077328 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL169509 NHLBI NIH HHS
- R01 HL158596 NHLBI NIH HHS
- R01 HL162794 NHLBI NIH HHS
- R01-HL141268 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL62794 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
Collapse
Affiliation(s)
- Karina Mora Massad
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Zhiyu Dai
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine in St. Louis, Saint Louis, Missouri, United States
| | - Irina Petrache
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado, United States
| | - Corey E Ventetuolo
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Warren Alpert Medical School of Brown University, Providence, Rhode Island, United States
- Department of Health Services, Policy and Practice, Brown University, Providence, Rhode Island, United States
| | - Tim Lahm
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado, United States
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado, United States
| |
Collapse
|
3
|
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
|
4
|
Prasad RR, Kumar S, Zhang H, Li M, Hu CJ, Riddle S, McKeon BA, Frid M, Hoetzenecker K, Crnkovic S, Kwapiszewska G, Tuder RM, Stenmark KR. An intracellular complement system drives metabolic and proinflammatory reprogramming of vascular fibroblasts in pulmonary hypertension. JCI Insight 2025; 10:e184141. [PMID: 39946184 PMCID: PMC11949053 DOI: 10.1172/jci.insight.184141] [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: 06/24/2024] [Accepted: 02/12/2025] [Indexed: 03/25/2025] Open
Abstract
The complement system is central to the innate immune response, playing a critical role in proinflammatory and autoimmune diseases such as pulmonary hypertension (PH). Recent discoveries highlight the emerging role of intracellular complement, or the "complosome," in regulating cellular processes such as glycolysis, mitochondrial dynamics, and inflammatory gene expression. This study investigated the hypothesis that intracellular complement proteins C3, CFB, and CFD are upregulated in PH fibroblasts (PH-Fibs) and drive their metabolic and inflammatory states, contributing to PH progression. Our results revealed a pronounced upregulation of CFD, CFB, and C3 in PH-Fibs from human samples and bovine models, both in vivo and in vitro. The finding of elevated levels of C3 activation fragments, including C3b, C3d, and C3a, emphasized enhanced C3 activity. PH-Fibs exhibited notable metabolic reprogramming and increased levels of proinflammatory mediators such as MCP1, SDF1, IL-6, IL-13, and IL-33. Silencing CFD via shRNA reduced CFB activation and C3a production, while normalizing glycolysis, tricarboxylic acid (TCA) cycle activity, and fatty acid metabolism. Metabolomic and gene expression analyses of CFD-knockdown PH-Fibs revealed restored metabolic and inflammatory profiles, underscoring CFD's crucial role in these changes. This study emphasizes the crucial role of intracellular complement in PH pathogenesis, highlighting the potential for complement-targeted therapies in PH.
Collapse
Affiliation(s)
- Ram Raj Prasad
- Cardiovascular and Pulmonary Research Laboratory (CVP), Department of Pediatrics and Medicine, and
| | - Sushil Kumar
- Cardiovascular and Pulmonary Research Laboratory (CVP), Department of Pediatrics and Medicine, and
| | - Hui Zhang
- Cardiovascular and Pulmonary Research Laboratory (CVP), Department of Pediatrics and Medicine, and
| | - Min Li
- Cardiovascular and Pulmonary Research Laboratory (CVP), Department of Pediatrics and Medicine, and
| | - Cheng-Jun Hu
- Department of Craniofacial Biology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Suzette Riddle
- Cardiovascular and Pulmonary Research Laboratory (CVP), Department of Pediatrics and Medicine, and
| | - Brittany A. McKeon
- Cardiovascular and Pulmonary Research Laboratory (CVP), Department of Pediatrics and Medicine, and
| | - M.G. Frid
- Cardiovascular and Pulmonary Research Laboratory (CVP), Department of Pediatrics and Medicine, and
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Slaven Crnkovic
- Ludwig Boltzmann Institute for Lung Vascular Research, Otto Loewi Research Center, Lung Research Cluster, Medical University of Graz, Graz, Austria
- Institute for Lung Health, Cardiopulmonary Institute, Member of the German Center for Lung Research, Justus Liebig University Giessen, Germany
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research, Otto Loewi Research Center, Lung Research Cluster, Medical University of Graz, Graz, Austria
- Institute for Lung Health, Cardiopulmonary Institute, Member of the German Center for Lung Research, Justus Liebig University Giessen, Germany
| | - Rubin M. Tuder
- Cardiovascular and Pulmonary Research Laboratory (CVP), Department of Pediatrics and Medicine, and
- Department of Lung Biology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kurt R. Stenmark
- Cardiovascular and Pulmonary Research Laboratory (CVP), Department of Pediatrics and Medicine, and
| |
Collapse
|
5
|
Forbes LM, Bauer N, Bhadra A, Bogaard HJ, Choudhary G, Goss KN, Gräf S, Heresi GA, Hopper RK, Jose A, Kim Y, Klouda T, Lahm T, Lawrie A, Leary PJ, Leopold JA, Oliveira SD, Prisco SZ, Rafikov R, Rhodes CJ, Stewart DJ, Vanderpool RR, Yuan K, Zimmer A, Hemnes AR, de Jesus Perez VA, Wilkins MR. Precision Medicine for Pulmonary Vascular Disease: The Future Is Now (2023 Grover Conference Series). Pulm Circ 2025; 15:e70027. [PMID: 39749110 PMCID: PMC11693987 DOI: 10.1002/pul2.70027] [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: 10/19/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 01/04/2025] Open
Abstract
Pulmonary vascular disease is not a single condition; rather it can accompany a variety of pathologies that impact the pulmonary vasculature. Applying precision medicine strategies to better phenotype, diagnose, monitor, and treat pulmonary vascular disease is increasingly possible with the growing accessibility of powerful clinical and research tools. Nevertheless, challenges exist in implementing these tools to optimal effect. The 2023 Grover Conference Series reviewed the research landscape to summarize the current state of the art and provide a better understanding of the application of precision medicine to managing pulmonary vascular disease. In particular, the following aspects were discussed: (1) Clinical phenotypes, (2) genetics, (3) epigenetics, (4) biomarker discovery, (5) application of precision biology to clinical trials, (6) the right ventricle (RV), and (7) integrating precision medicine to clinical care. The present review summarizes the content of these discussions and the prospects for the future.
Collapse
Affiliation(s)
- Lindsay M. Forbes
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of ColoradoAuroraColoradoUSA
| | - Natalie Bauer
- Department of PharmacologyCollege of Medicine, University of South AlabamaMobileAlabamaUSA
- Department of Physiology and Cell BiologyUniversity of South AlabamaMobileAlabamaUSA
| | - Aritra Bhadra
- Department of PharmacologyCollege of Medicine, University of South AlabamaMobileAlabamaUSA
- Center for Lung BiologyCollege of Medicine, University of South AlabamaMobileAlabamaUSA
| | - Harm J. Bogaard
- Department of Pulmonary MedicineAmsterdam UMCAmsterdamNetherlands
| | - Gaurav Choudhary
- Division of CardiologyWarren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
- Lifespan Cardiovascular InstituteRhode Island and Miriam HospitalsProvidenceRhode IslandUSA
- Department of CardiologyProvidence VA Medical CenterProvidenceRhode IslandUSA
| | - Kara N. Goss
- Department of Medicine and PediatricsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Stefan Gräf
- Division of Computational Genomics and Genomic Medicine, Department of MedicineUniversity of Cambridge, Victor Phillip Dahdaleh Heart & Lung Research InstituteCambridgeUK
| | | | - Rachel K. Hopper
- Department of PediatricsStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Arun Jose
- Division of Pulmonary, Critical Care, and Sleep MedicineUniversity of CincinnatiCincinnatiOhioUSA
| | - Yunhye Kim
- Division of Pulmonary MedicineBoston Children's HospitalBostonMAUSA
| | - Timothy Klouda
- Division of Pulmonary MedicineBoston Children's HospitalBostonMAUSA
| | - Tim Lahm
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of ColoradoAuroraColoradoUSA
- Division of Pulmonary, Critical Care, and Sleep MedicineNational Jewish HealthDenverColoradoUSA
- Pulmonary and Critical Care SectionRocky Mountain Regional VA Medical CenterDenverColoradoUSA
| | - Allan Lawrie
- National Heart and Lung InstituteImperial College LondonLondonUK
| | - Peter J. Leary
- Departments of Medicine and EpidemiologyUniversity of WashingtonSeattleWashingtonUSA
| | - Jane A. Leopold
- Division of Cardiovascular MedicineBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Suellen D. Oliveira
- Department of Anesthesiology, Department of Physiology and BiophysicsUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - Sasha Z. Prisco
- Division of CardiovascularLillehei Heart Institute, University of MinnesotaMinneapolisMinnesotaUSA
| | - Ruslan Rafikov
- Department of MedicineIndiana UniversityIndianapolisIndianaUSA
| | | | - Duncan J. Stewart
- Ottawa Hospital Research InstituteFaculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | | | - Ke Yuan
- Division of Pulmonary MedicineBoston Children's HospitalBostonMAUSA
| | - Alexsandra Zimmer
- Department of MedicineBrown UniversityProvidenceRhode IslandUSA
- Lifespan Cardiovascular InstituteRhode Island HospitalProvidenceRhode IslandUSA
| | - Anna R. Hemnes
- Division of Allergy, Pulmonary and Critical Care MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Vinicio A. de Jesus Perez
- Division of Pulmonary and Critical Care MedicineStanford University Medical CenterStanfordCaliforniaUSA
| | | |
Collapse
|
6
|
Otálora-Otálora BA, Payán-Gómez C, López-Rivera JJ, Pedroza-Aconcha NB, Arboleda-Mojica SL, Aristizábal-Guzmán C, Isaza-Ruget MA, Álvarez-Moreno CA. Interplay of Transcriptomic Regulation, Microbiota, and Signaling Pathways in Lung and Gut Inflammation-Induced Tumorigenesis. Cells 2024; 14:1. [PMID: 39791702 PMCID: PMC11720097 DOI: 10.3390/cells14010001] [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: 12/02/2024] [Revised: 12/15/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025] Open
Abstract
Inflammation can positively and negatively affect tumorigenesis based on the duration, scope, and sequence of related events through the regulation of signaling pathways. A transcriptomic analysis of five pulmonary arterial hypertension, twelve Crohn's disease, and twelve ulcerative colitis high throughput sequencing datasets using R language specialized libraries and gene enrichment analyses identified a regulatory network in each inflammatory disease. IRF9 and LINC01089 in pulmonary arterial hypertension are related to the regulation of signaling pathways like MAPK, NOTCH, human papillomavirus, and hepatitis c infection. ZNF91 and TP53TG1 in Crohn's disease are related to the regulation of PPAR, MAPK, and metabolic signaling pathways. ZNF91, VDR, DLEU1, SATB2-AS1, and TP53TG1 in ulcerative colitis are related to the regulation of PPAR, AMPK, and metabolic signaling pathways. The activation of the transcriptomic network and signaling pathways might be related to the interaction of the characteristic microbiota of the inflammatory disease, with the lung and gut cell receptors present in membrane rafts and complexes. The transcriptomic analysis highlights the impact of several coding and non-coding RNAs, suggesting their relationship with the unlocking of cell phenotypic plasticity for the acquisition of the hallmarks of cancer during lung and gut cell adaptation to inflammatory phenotypes.
Collapse
Affiliation(s)
| | - César Payán-Gómez
- Dirección Académica, Universidad Nacional de Colombia, Sede de La Paz, La Paz 202017, Colombia; (C.P.-G.); (N.B.P.-A.)
| | - Juan Javier López-Rivera
- Grupo de Investigación INPAC, Specialized Laboratory, Clinica Universitaria Colombia, Clínica Colsanitas S.A., Bogotá 111321, Colombia;
| | - Natalia Belén Pedroza-Aconcha
- Dirección Académica, Universidad Nacional de Colombia, Sede de La Paz, La Paz 202017, Colombia; (C.P.-G.); (N.B.P.-A.)
| | | | - Claudia Aristizábal-Guzmán
- Grupo de Investigación INPAC, Unidad de Investigación, Fundación Universitaria Sanitas, Bogotá 110131, Colombia;
| | - Mario Arturo Isaza-Ruget
- Keralty, Sanitas International Organization, Grupo de Investigación INPAC, Fundación Universitaria Sanitas, Bogotá 110131, Colombia;
| | - Carlos Arturo Álvarez-Moreno
- Infectious Diseases Department, Clinica Universitaria Colombia, Clínica Colsanitas S.A., Bogotá 111321, Colombia;
| |
Collapse
|
7
|
Erratum: Digital Spatial Profiling Identifies Distinct Molecular Signatures of Vascular Lesions in Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 2024; 210:1279-1280. [PMID: 39545753 PMCID: PMC11568431 DOI: 10.1164/rccm.v210erratum7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024] Open
|
8
|
Andruska AM, Cantu Valadez R, Ichimura K, Chu P, Zhang T, Schimmel K, Wang L, Cao A, Aldred MA, Spiekerkoetter E. Increased BMP-Responsive Transcription Factors in Distinct Endothelial and Mesenchymal Cells in PAH. Am J Respir Crit Care Med 2024; 211:127-130. [PMID: 39499865 PMCID: PMC11755370 DOI: 10.1164/rccm.202405-1039le] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 10/30/2024] [Indexed: 11/07/2024] Open
Affiliation(s)
| | | | - Kenzo Ichimura
- Stanford University, Department of Medicine, Stanford, California, United States
| | - Pauline Chu
- Stanford University, Stanford, California, United States
| | - Tianyi Zhang
- Stanford University, Stanford, California, United States
| | - Katharina Schimmel
- Stanford University, Department of Medicine, Division of Pulmonary, Allergy and Critical Care, Stanford, California, United States
| | - Lingli Wang
- Stanford University School of Medicine, Pediatrics, Stanford, California, United States
| | - Aiqin Cao
- Stanford University School of Medicine, Pediatrics, Stanford, California, United States
| | - Micheala A Aldred
- Indiana University School of Medicine, Dept of Medicine, Indianapolis, Indiana, United States
| | - Edda Spiekerkoetter
- Stanford University, Pulmonary and Critcal Care, Stanford, California, United States;
| |
Collapse
|
9
|
Guignabert C, Aman J, Bonnet S, Dorfmüller P, Olschewski AJ, Pullamsetti S, Rabinovitch M, Schermuly RT, Humbert M, Stenmark KR. Pathology and pathobiology of pulmonary hypertension: current insights and future directions. Eur Respir J 2024; 64:2401095. [PMID: 39209474 PMCID: PMC11533988 DOI: 10.1183/13993003.01095-2024] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 06/08/2024] [Indexed: 09/04/2024]
Abstract
In recent years, major advances have been made in the understanding of the cellular and molecular mechanisms driving pulmonary vascular remodelling in various forms of pulmonary hypertension, including pulmonary arterial hypertension, pulmonary hypertension associated with left heart disease, pulmonary hypertension associated with chronic lung disease and hypoxia, and chronic thromboembolic pulmonary hypertension. However, the survival rates for these different forms of pulmonary hypertension remain unsatisfactory, underscoring the crucial need to more effectively translate innovative scientific knowledge into healthcare interventions. In these proceedings of the 7th World Symposium on Pulmonary Hypertension, we delve into recent developments in the field of pathology and pathophysiology, prioritising them while questioning their relevance to different subsets of pulmonary hypertension. In addition, we explore how the latest omics and other technological advances can help us better and more rapidly understand the myriad basic mechanisms contributing to the initiation and progression of pulmonary vascular remodelling. Finally, we discuss strategies aimed at improving patient care, optimising drug development, and providing essential support to advance research in this field.
Collapse
Affiliation(s)
- Christophe Guignabert
- Université Paris-Saclay, Hypertension Pulmonaire: Physiopathology and Innovation Thérapeutique, HPPIT, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, HPPIT, Le Kremlin-Bicêtre, France
| | - Jurjan Aman
- Department of Pulmonary Medicine, Amsterdam UMC, VU University Medical Center, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Sébastien Bonnet
- Pulmonary Hypertension research group, Centre de Recherche de l'Institut de Cardiologie et de Pneumologie de Québec, Quebec City, QC, Canada
- Department of Medicine, Université Laval, Quebec City, QC, Canada
| | - Peter Dorfmüller
- Department of Pathology, University Hospital Giessen/Marburg, Giessen, Germany
| | - Andrea J Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, Graz, Austria
| | - Soni Pullamsetti
- Max Planck Institute for Heart and Lung Research Bad Nauheim, Bad Nauheim, Germany
- Department of Internal Medicine, German Center for Lung Research (DZL) Cardio-Pulmonary Institute (CPI)
- Universities of Giessen and Marburg Lung Centre, Member of the German Center for Lung Research (DZL), Justus-Liebig University Giessen, Giessen, Germany
| | - Marlene Rabinovitch
- BASE Initiative, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Ralph T Schermuly
- Department of Internal Medicine, German Center for Lung Research (DZL) Cardio-Pulmonary Institute (CPI)
| | - Marc Humbert
- Université Paris-Saclay, Hypertension Pulmonaire: Physiopathology and Innovation Thérapeutique, HPPIT, Faculté de Médecine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, HPPIT, Le Kremlin-Bicêtre, France
- Department of Respiratory and Intensive Care Medicine, Assistance Publique Hôpitaux de Paris, Hôpital Bicêtre, ERN-LUNG, Le Kremlin-Bicêtre, France
| | - Kurt R Stenmark
- Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, University of Colorado, Denver, CO, USA
| |
Collapse
|
10
|
Mickael C, Sanders LA, Lee MH, Kumar R, Fonseca-Balladares D, Gandjeva A, Cautivo-Reyes K, Kassa B, Kumar S, Irwin D, Swindle D, Phang T, Stearman RS, Molofsky AB, McKee AS, Stenmark KR, Graham BB, Tuder RM. Classical dendritic cells contribute to hypoxia-induced pulmonary hypertension. FASEB J 2024; 38:e70015. [PMID: 39212294 PMCID: PMC11462638 DOI: 10.1096/fj.202400338rr] [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: 02/13/2024] [Revised: 08/06/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
Pulmonary hypertension (PH) is a chronic and progressive disease with significant morbidity and mortality. It is characterized by remodeled pulmonary vessels associated with perivascular and intravascular accumulation of inflammatory cells. Although there is compelling evidence that bone marrow-derived cells, such as macrophages and T cells, cluster in the vicinity of pulmonary vascular lesions in humans and contribute to PH development in different animal models, the role of dendritic cells in PH is less clear. Dendritic cells' involvement in PH is likely since they are responsible for coordinating innate and adaptive immune responses. We hypothesized that dendritic cells drive hypoxic PH. We demonstrate that a classical dendritic cell (cDC) subset (cDC2) is increased and activated in wild-type mouse lungs after hypoxia exposure. We observe significant protection after the depletion of cDCs in ZBTB46 DTR chimera mice before hypoxia exposure and after established hypoxic PH. In addition, we find that cDC depletion is associated with a reduced number of two macrophage subsets in the lung (FolR2+ MHCII+ CCR2+ and FolR2+ MHCII+ CCR2-). We found that depleting cDC2s, but not cDC1s, was protective against hypoxic PH. Finally, proof-of-concept studies in human lungs show increased perivascular cDC2s in patients with Idiopathic Pulmonary Arterial Hypertension (IPAH). Our data points to an essential role of cDCs, particularly cDC2s, in the pathophysiology of experimental PH.
Collapse
Grants
- R01 HL142701 NHLBI NIH HHS
- R01 HL161004 NHLBI NIH HHS
- R01 AI162806 NIAID NIH HHS
- R01HL142701 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01AI162806 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- American Thoracic Society (ATS)
- K01 HL161024 NHLBI NIH HHS
- K08HL168310 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- K01HL161024 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL135872 NHLBI NIH HHS
- W81XWH2210457 U.S. Department of Defense (DOD)
- Cardiovascular Medical Research and Education Fund (CMREF)
- Actelion Pharmaceuticals (Actelion Pharmaceuticals Ltd)
- R25HL146166 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R25 HL146166 NHLBI NIH HHS
- R01NS126765 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- K08 HL168310 NHLBI NIH HHS
- United Therapeutics Corporation (Uni Ther)
- R01 NS126765 NINDS NIH HHS
- 19CDA34730030 American Heart Association (AHA)
- R01HL135872 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- PO1HL152961 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL158076 NHLBI NIH HHS
- R01 H161004 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P01 HL152961 NHLBI NIH HHS
Collapse
Affiliation(s)
- Claudia Mickael
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado, USA
- Cardiovascular Research Laboratories, University of Colorado, Aurora, Colorado, USA
| | - Linda A. Sanders
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado, USA
- Cardiovascular Research Laboratories, University of Colorado, Aurora, Colorado, USA
| | - Michael H. Lee
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Lung Biology Center, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Rahul Kumar
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Lung Biology Center, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Dara Fonseca-Balladares
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Lung Biology Center, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Aneta Gandjeva
- Cardiovascular Research Laboratories, University of Colorado, Aurora, Colorado, USA
| | - Kelly Cautivo-Reyes
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
- Gilead Sciences, Foster City, California, USA
| | - Biruk Kassa
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Lung Biology Center, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Sushil Kumar
- Cardiovascular Research Laboratories, University of Colorado, Aurora, Colorado, USA
| | - David Irwin
- Cardiovascular Research Laboratories, University of Colorado, Aurora, Colorado, USA
| | - Delaney Swindle
- Cardiovascular Research Laboratories, University of Colorado, Aurora, Colorado, USA
| | - Tzu Phang
- Section of Hematology, Oncology, and Bone Marrow Transplantation-Cellular Therapeutics (BMT-CT), Department of Pediatrics, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Robert S. Stearman
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ari B. Molofsky
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Amy S. McKee
- Department of Medicine, Division of Clinical Immunology, University of Colorado, Aurora, Colorado, USA
- Department of Microbiology and Immunology and ClinImmune Cell and Gene Therapy, University of Colorado, Aurora, Colorado, USA
| | - Kurt R. Stenmark
- Cardiovascular Research Laboratories, University of Colorado, Aurora, Colorado, USA
| | - Brian B. Graham
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Lung Biology Center, Zuckerberg San Francisco General Hospital, San Francisco, California, USA
| | - Rubin M. Tuder
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, Colorado, USA
- Cardiovascular Research Laboratories, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
11
|
Rafikov R, de Jesus Perez V, Dekan A, Kudryashova TV, Rafikova O. Deciphering the Complexities of Pulmonary Hypertension: The Emergent Role of Single-Cell Omics. Am J Respir Cell Mol Biol 2024; 72:32-40. [PMID: 39141563 PMCID: PMC11707669 DOI: 10.1165/rcmb.2024-0145ps] [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: 03/27/2024] [Accepted: 08/14/2024] [Indexed: 08/16/2024] Open
Abstract
Expanding upon the critical advancements brought forth by single-cell omics in pulmonary hypertension (PH) research, this review delves deep into how these technologies have been piloted in a new era of understanding this complex disease. By leveraging the power of single cell transcriptomics (scRNA-seq), researchers can now dissect the complicated cellular ecosystem of the lungs, examining the key players such as endothelial cells, smooth muscle cells, pericytes, and immune cells, and their unique roles in the pathogenesis of PH. This more granular view is beyond the limitations of traditional bulk analysis, allowing for the identification of novel therapeutic targets previously obscured in the aggregated data. Connectome analysis based on single-cell omics of the cells involved in pathological changes can reveal a clearer picture of the cellular interactions and transitions in the cellular subtypes. Furthermore, the review acknowledges the challenges that lie ahead, including the need for enhancing the resolution of scRNA-seq to capture even finer details of cellular changes, overcoming logistical barriers in processing human tissue samples, and the necessity of integrating diverse omics approaches to fully comprehend the molecular underpinnings of PH. The promise of these single-cell technologies is immense, offering the potential for targeted drug development and the discovery of biomarkers for early diagnosis and disease monitoring. Through these advancements, the field moves closer to realizing the goal of precision medicine for patients with PH.
Collapse
Affiliation(s)
- Ruslan Rafikov
- Indiana University School of Medicine, Indianapolis, Indiana, United States;
| | | | - Aleksandr Dekan
- Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Tatiana V Kudryashova
- University of Pittsburgh Department of Medicine, Pittsburgh, Pennsylvania, United States
| | - Olga Rafikova
- Indiana University Purdue University at Indianapolis, Indianapolis, Indiana, United States
| |
Collapse
|
12
|
Oldham WM. Exploring the Molecular Signatures of Pulmonary Vascular Lesions. Am J Respir Crit Care Med 2024; 210:256-257. [PMID: 38780073 PMCID: PMC11348971 DOI: 10.1164/rccm.202404-0695ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024] Open
Affiliation(s)
- William M Oldham
- Department of Medicine Brigham and Women's Hospital and Harvard Medical School Boston, Massachusetts
| |
Collapse
|