1
|
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
|
2
|
Heydari K, Johnson C, Diane Cooper I, Hersi K, Tanba C, Sun J, Solomon MA, Elinoff JM. Flow Cytometric Determination of Circulating Progenitor Cells in Patients With Pulmonary Arterial Hypertension: A Systematic Review. Pulm Circ 2025; 15:e70065. [PMID: 40125448 PMCID: PMC11925724 DOI: 10.1002/pul2.70065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 03/25/2025] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by progressive narrowing and obliteration of distal, pre-capillary pulmonary vessels. Yet, noninvasive biomarkers that reflect this disease-defining process are lacking. A systematic review of PAH studies that measured circulating progenitor cells (CPCs) or circulating endothelial cells (CECs) in PAH by flow cytometry was performed to understand how future studies, leveraging state-of-the-art single-cell analyses, can advance the field. The study was conducted in accordance with the Preferred Reporting Items for Systematic Reviews. Of the 2422 studies identified, 20 met inclusion criteria. Nineteen studies measured CPCs by flow cytometry, only one study examined CECs. A total of 647 PAH patients were included across all 19 CPC studies. Marker schemes chosen to define CPCs, and the methods of flow cytometry used, varied significantly across studies. Meta-analysis of a subgroup of CPC studies (n = 8) similarly identified a significant amount of heterogeneity even amongst studies using the same marker scheme. In conclusion, a systematic review of CPC studies in PAH patients reveals the limitations of the current literature. Future studies should include contemporary risk assessments, disease duration, reporting of comorbid conditions, and serial sampling over time. Furthermore, methods that incorporate best practices for detecting rare cell populations by flow cytometry are essential and should be reported in sufficient detail in future publications. With the emergence of single-cell technologies, future studies of circulating progenitor and endothelial cells in PAH remain relevant and may incorporate several insights from the current review to build upon the existing literature.
Collapse
Affiliation(s)
- Kimia Heydari
- Critical Care Medicine DepartmentNIH Clinical Center, National Institutes of HealthBethesdaMarylandUSA
| | - Carrie Johnson
- Critical Care Medicine DepartmentNIH Clinical Center, National Institutes of HealthBethesdaMarylandUSA
| | | | - Kadija Hersi
- National Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Carl Tanba
- Department of Pulmonary and Critical Care MedicineTufts Medical CenterBostonMarylandUSA
| | - Junfeng Sun
- Critical Care Medicine DepartmentNIH Clinical Center, National Institutes of HealthBethesdaMarylandUSA
| | - Michael A. Solomon
- Critical Care Medicine DepartmentNIH Clinical Center, National Institutes of HealthBethesdaMarylandUSA
- National Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Jason M. Elinoff
- Critical Care Medicine DepartmentNIH Clinical Center, National Institutes of HealthBethesdaMarylandUSA
- National Heart Lung and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
3
|
Wang S, Awad KS, Chen LY, Siddique MAH, Ferreyra GA, Wang CL, Joseph T, Yu ZX, Takeda K, Demirkale CY, Zhao YY, Elinoff JM, Danner RL. Endothelial PHD2 deficiency induces apoptosis resistance and inflammation via AKT activation and AIP1 loss independent of HIF2α. Am J Physiol Lung Cell Mol Physiol 2024; 327:L503-L519. [PMID: 39159362 PMCID: PMC11482463 DOI: 10.1152/ajplung.00077.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/16/2024] [Accepted: 08/09/2024] [Indexed: 08/21/2024] Open
Abstract
In hypoxic and pseudohypoxic rodent models of pulmonary hypertension (PH), hypoxia-inducible factor (HIF) inhibition attenuates disease initiation. However, HIF activation alone, due to genetic alterations or use of inhibitors of prolyl hydroxylase domain (PHD) enzymes, has not been definitively shown to cause PH in humans, indicating the involvement of other mechanisms. Given the association between endothelial cell dysfunction and PH, the effects of pseudohypoxia and its underlying pathways were investigated in primary human lung endothelial cells. PHD2 silencing or inhibition, while activating HIF2α, induced apoptosis-resistance and IFN/STAT activation in endothelial cells, independent of HIF signaling. Mechanistically, PHD2 deficiency activated AKT and ERK, inhibited JNK, and reduced AIP1 (ASK1-interacting protein 1), all independent of HIF2α. Like PHD2, AIP1 silencing affected these same kinase pathways and produced a similar dysfunctional endothelial cell phenotype, which was partially reversed by AKT inhibition. Consistent with these in vitro findings, AIP1 protein levels in lung endothelial cells were decreased in Tie2-Cre/Phd2 knockout mice compared with wild-type controls. Lung vascular endothelial cells from patients with pulmonary arterial hypertension (PAH) showed IFN/STAT activation. Lung tissue from both SU5416/hypoxia PAH rats and patients with PAH all showed AKT activation and dysregulated AIP1 expression. In conclusion, PHD2 deficiency in lung vascular endothelial cells drives an apoptosis-resistant and inflammatory phenotype, mediated by AKT activation and AIP1 loss independent of HIF signaling. Targeting these pathways, including PHD2, AKT, and AIP1, holds the potential for developing new treatments for endothelial dysfunction in PH.NEW & NOTEWORTHY HIF activation alone does not conclusively lead to human PH, suggesting that HIF-independent signaling may also contribute to hypoxia-induced PH. This study demonstrated that PHD2 silencing-induced pseudohypoxia in human lung endothelial cells suppresses apoptosis and activates STAT, effects that persist despite HIF2α inhibition or knockdown and are attributed to AKT and ERK activation, JNK inhibition, and AIP1 loss. These findings align with observations in lung endothelial cells and tissues from PAH rodent models and patients.
Collapse
Affiliation(s)
- Shuibang Wang
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, United States
| | - Keytam S Awad
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, United States
| | - Li-Yuan Chen
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, United States
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Mohammad A H Siddique
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, United States
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Gabriela A Ferreyra
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, United States
| | - Caroline L Wang
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, United States
| | - Thea Joseph
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, United States
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Zu-Xi Yu
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Kazuyo Takeda
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Cumhur Y Demirkale
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, United States
| | - You-Yang Zhao
- Section for Injury Repair and Regeneration, Stanley Manne Children Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, United States
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States
| | - Jason M Elinoff
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, United States
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States
| | - Robert L Danner
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
4
|
Hemnes A, Fortune N, Simon K, Trenary IA, Shay S, Austin E, Young JD, Britain E, West J, Talati M. A multimodal approach identifies lactate as a central feature of right ventricular failure that is detectable in human plasma. Front Med (Lausanne) 2024; 11:1387195. [PMID: 39346939 PMCID: PMC11428650 DOI: 10.3389/fmed.2024.1387195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 08/23/2024] [Indexed: 10/01/2024] Open
Abstract
Background In PAH metabolic abnormalities in multiple pathways are well-recognized features of right ventricular dysfunction, however, prior work has focused mainly on the use of a single "omic" modality to describe a single deranged pathway. We integrated metabolomic and epigenomic data using transcriptomics in failing and non-failing RVs from a rodent model to provide novel mechanistic insight and translated these findings to accessible human specimens by correlation with plasma from PAH patients. Methods Study was conducted in a doxycycline-inducible BMPR2 mutant mouse model of RV failure. Plasma was collected from controls and PAH patients. Transcriptomic and metabolomic analyses were done on mouse RV tissue and human plasma. For mouse RV, we layered metabolomic and transcriptomic data for multiple metabolic pathways and compared our findings with metabolomic and transcriptomic data obtained for human plasma. We confirmed our key findings in cultured cardiomyocyte cells with BMPR2 mutation. Results In failing mouse RVs, (1) in the glycolysis pathway, glucose is converted to lactate via aerobic glycolysis, but may also be utilized for glycogen, fatty acid, and nucleic acid synthesis, (2) in the fatty acid pathway, FAs are accumulated in the cytoplasm because the transfer of FAs to mitochondria is reduced, however, the ß-oxidation pathway is likely to be functional. (3) the TCA cycle is altered at multiple checkpoints and accumulates citrate, and the glutaminolysis pathway is not activated. In PAH patients, plasma metabolic and transcriptomic data indicated that unlike in the failing BMPR2 mutant RV, expression of genes and metabolites measured for the glycolysis pathway, FA pathway, TCA cycle, and glutaminolysis pathway were increased. Lactate was the only metabolite that was increased both in RV and circulation. We confirmed using a stable isotope of lactate that cultured cardiomyocytes with mutant BMPR2 show a modest increase in endogenous lactate, suggesting a possibility of an increase in lactate production by cardiomyocytes in failing BMPR2 mutant RV. Conclusion In the failing RV with mutant BMPR2, lactate is produced by RV cardiomyocytes and may be secreted out, thereby increasing lactate in circulation. Lactate can potentially serve as a marker of RV dysfunction in PAH, which warrants investigation.
Collapse
Affiliation(s)
- Anna Hemnes
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Niki Fortune
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Katie Simon
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Irina A Trenary
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
| | - Sheila Shay
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Eric Austin
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jamey D Young
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| | - Evan Britain
- Department of Cardiology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - James West
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Megha Talati
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
5
|
Peng Q, Arulsamy K, Lu YW, Wu H, Zhu B, Singh B, Cui K, Wylie-Sears J, Li K, Wong S, Cowan DB, Aikawa M, Wang DZ, Bischoff J, Chen K, Chen H. Novel Role of Endothelial CD45 in Regulating Endothelial-to-Mesenchymal Transition in Atherosclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.03.610974. [PMID: 39282400 PMCID: PMC11398423 DOI: 10.1101/2024.09.03.610974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
Background Protein-tyrosine-phosphatase CD45 is exclusively expressed in all nucleated cells of the hematopoietic system but is rarely expressed in endothelial cells. Interestingly, our recent study indicated that activation of the endogenous CD45 promoter in human endothelial colony forming cells (ECFCs) induced expression of multiple EndoMT marker genes. However, the detailed molecular mechanisms underlying CD45 that drive EndoMT and the therapeutic potential of manipulation of CD45 expression in atherosclerosis are entirely unknown. Method We generated a tamoxifen-inducible EC-specific CD45 deficient mouse strain (EC-iCD45KO) in an ApoE-deficient (ApoE-/-) background and fed with a Western diet (C57BL/6) for atherosclerosis and molecular analyses. We isolated and enriched mouse aortic endothelial cells with CD31 beads to perform single-cell RNA sequencing. Biomedical, cellular, and molecular approaches were utilized to investigate the role of endothelial CD45-specific deletion in the prevention of EndoMT in ApoE-/- model of atherosclerosis. Results Single-cell RNA sequencing revealed that loss of endothelial CD45 inhibits EndoMT marker expression and transforming growth factor-β signaling in atherosclerotic mice. which is associated with the reductions of lesions in the ApoE-/- mouse model. Mechanistically, the loss of endothelial cell CD45 results in increased KLF2 expression, which inhibits transforming growth factor-β signaling and EndoMT. Consistently, endothelial CD45 deficient mice showed reduced lesion development, plaque macrophages, and expression of cell adhesion molecules when compared to ApoE-/- controls. Conclusions These findings demonstrate that the loss of endothelial CD45 protects against EndoMT-driven atherosclerosis, promoting KLF2 expression while inhibiting TGFβ signaling and EndoMT markers. Thus, targeting endothelial CD45 may be a novel therapeutic strategy for EndoMT and atherosclerosis.
Collapse
Affiliation(s)
- Qianman Peng
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School; Boston, MA, 02115, USA
| | - Kulandaisamy Arulsamy
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School; Boston, MA, USA
| | - Yao Wei Lu
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School; Boston, MA, 02115, USA
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School; Boston, MA, USA
| | - Hao Wu
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School; Boston, MA, 02115, USA
| | - Bo Zhu
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School; Boston, MA, 02115, USA
| | - Bandana Singh
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School; Boston, MA, 02115, USA
| | - Kui Cui
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School; Boston, MA, 02115, USA
| | - Jill Wylie-Sears
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School; Boston, MA, 02115, USA
| | - Kathryn Li
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School; Boston, MA, 02115, USA
| | - Scott Wong
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School; Boston, MA, 02115, USA
| | - Douglas B. Cowan
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School; Boston, MA, 02115, USA
| | - Masanori Aikawa
- Brigham and Women’s Hospital, Harvard Medical School; Boston, MA, 02115, USA
| | - Da-Zhi Wang
- Center for Regenerative Medicine and USF Health Heart Institute, Department of Internal Medicine, University of South Florida, Tampa
| | - Joyce Bischoff
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School; Boston, MA, 02115, USA
| | - Kaifu Chen
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School; Boston, MA, USA
| | - Hong Chen
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School; Boston, MA, 02115, USA
| |
Collapse
|
6
|
Tang B, Vadgama A, Redmann B, Hong J. Charting the cellular landscape of pulmonary arterial hypertension through single-cell omics. Respir Res 2024; 25:192. [PMID: 38702687 PMCID: PMC11067161 DOI: 10.1186/s12931-024-02823-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 04/25/2024] [Indexed: 05/06/2024] Open
Abstract
This review examines how single-cell omics technologies, particularly single-cell RNA sequencing (scRNAseq), enhance our understanding of pulmonary arterial hypertension (PAH). PAH is a multifaceted disorder marked by pulmonary vascular remodeling, leading to high morbidity and mortality. The cellular pathobiology of this heterogeneous disease, involving various vascular and non-vascular cell types, is not fully understood. Traditional PAH studies have struggled to resolve the complexity of pathogenic cell populations. scRNAseq offers a refined perspective by detailing cellular diversity within PAH, identifying unique cell subsets, gene networks, and molecular pathways that drive the disease. We discuss significant findings from recent literature, summarizing how scRNAseq has shifted our understanding of PAH in human, rat, and mouse models. This review highlights the insights gained into cellular phenotypes, gene expression patterns, and novel molecular targets, and contemplates the challenges and prospective paths for research. We propose ways in which single-cell omics could inform future research and translational efforts to combat PAH.
Collapse
Affiliation(s)
- Brian Tang
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine at UCLA, University of California, 200 UCLA Medical Plaza, Suite 365-B, Box 951693, Los Angeles, CA, 90095, USA
| | - Arjun Vadgama
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine at UCLA, University of California, 200 UCLA Medical Plaza, Suite 365-B, Box 951693, Los Angeles, CA, 90095, USA
| | - Bryce Redmann
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine at UCLA, University of California, 200 UCLA Medical Plaza, Suite 365-B, Box 951693, Los Angeles, CA, 90095, USA
| | - Jason Hong
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine at UCLA, University of California, 200 UCLA Medical Plaza, Suite 365-B, Box 951693, Los Angeles, CA, 90095, USA.
| |
Collapse
|
7
|
Hang C, Zu L, Luo X, Wang Y, Yan L, Zhang Z, Le K, Huang Y, Ye L, Ying Y, Chen K, Xu X, Lv Q, Du L. Ddx5 Targeted Epigenetic Modification of Pericytes in Pulmonary Hypertension After Intrauterine Growth Restriction. Am J Respir Cell Mol Biol 2024; 70:400-413. [PMID: 38301267 DOI: 10.1165/rcmb.2023-0244oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 02/01/2024] [Indexed: 02/03/2024] Open
Abstract
Newborns with intrauterine growth restriction (IUGR) have a higher likelihood of developing pulmonary arterial hypertension (PAH) in adulthood. Although there is increasing evidence suggesting that pericytes play a role in regulating myofibroblast transdifferentiation and angiogenesis in malignant and cardiovascular diseases, their involvement in the pathogenesis of IUGR-related pulmonary hypertension and the underlying mechanisms remain incompletely understood. To address this issue, a study was conducted using a Sprague-Dawley rat model of IUGR-related pulmonary hypertension. Our investigation revealed increased proliferation and migration of pulmonary microvascular pericytes in IUGR-related pulmonary hypertension, accompanied by weakened endothelial-pericyte interactions. Through whole-transcriptome sequencing, Ddx5 (DEAD-box protein 5) was identified as one of the hub genes in pericytes. DDX5, a member of the RNA helicase family, plays a role in the regulation of ATP-dependent RNA helicase activities and cellular function. MicroRNAs have been implicated in the pathogenesis of PAH, and microRNA-205 (miR-205) regulates cell proliferation, migration, and angiogenesis. The results of dual-luciferase reporter assays confirmed the specific binding of miR-205 to Ddx5. Mechanistically, miR-205 negatively regulates Ddx5, leading to the degradation of β-catenin by inhibiting the phosphorylation of Gsk3β at serine 9. In vitro experiments showed the addition of miR-205 effectively ameliorated pericyte dysfunction. Furthermore, in vivo experiments demonstrated that miR-205 agomir could ameliorate pulmonary hypertension. Our findings indicated that the downregulation of miR-205 expression mediates pericyte dysfunction through the activation of Ddx5. Therefore, targeting the miR-205/Ddx5/p-Gsk3β/β-catenin axis could be a promising therapeutic approach for IUGR-related pulmonary hypertension.
Collapse
Affiliation(s)
| | - Lu Zu
- Department of Neonatology and
| | - Xiaofei Luo
- Department of Pediatrics, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China; and
| | - Yu Wang
- Department of Neonatology and
| | - Lingling Yan
- Department of Pediatrics, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China; and
| | | | - Kaixing Le
- Academy of Pediatrics, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | | | | | | | | | - Xuefeng Xu
- Department of Rheumatology, Immunology, and Allergy, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang Province, People's Republic of China
| | | | | |
Collapse
|
8
|
Golino JL, Bian J, Wang X, Fu J, Zhu XB, Yeo J, Kelly M, Escorcia FE, Cam M, Xie C. Single-cell RNA sequencing reveals cancer stem-like cells and dynamics in tumor microenvironment during cholangiocarcinoma progression. Front Cell Dev Biol 2023; 11:1250215. [PMID: 38020927 PMCID: PMC10667919 DOI: 10.3389/fcell.2023.1250215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Cholangiocarcinoma is a malignancy of the bile ducts that is driven by activities of cancer stem-like cells and characterized by a heterogeneous tumor microenvironment. To better understand the transcriptional profiles of cancer stem-like cells and dynamics in the tumor microenvironment during the progression of cholangiocarcinoma, we performed single-cell RNA analysis on cells collected from three different timepoints of tumorigenesis in a YAP/AKT mouse model. Bulk RNA sequencing data from TCGA (The Cancer Genome Atlas program) and ICGC cohorts were used to verify and support the finding. In vitro and in vivo experiments were performed to assess the stemness of cancer stem-like cells. We identified Tm4sf1high malignant cells as cancer stem-like cells. Across timepoints of cholangiocarcinoma formation in YAP/AKT mice, we found dynamic change in cancer stem-like cell/stromal/immune cell composition. Nevertheless, the dynamic interaction among cancer stem-like cells, immune cells, and stromal cells at different timepoints was elaborated. Collectively, these data serve as a useful resource for better understanding cancer stem-like cell and malignant cell heterogeneity, stromal cell remodeling, and immune cell reprogramming. It also sheds new light on transcriptomic dynamics during cholangiocarcinoma progression at single-cell resolution.
Collapse
Affiliation(s)
- Jihye L. Golino
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jing Bian
- CCR Collaborative Bioinformatics Resource, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Xin Wang
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jianyang Fu
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Xiao Bin Zhu
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Julie Yeo
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Michael Kelly
- Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | - Freddy E. Escorcia
- Molecular Imaging Branch, Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
- NCI CCR Liver Cancer Program, Bethesda, MD, United States
| | - Maggie Cam
- CCR Collaborative Bioinformatics Resource, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Changqing Xie
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
- NCI CCR Liver Cancer Program, Bethesda, MD, United States
| |
Collapse
|