1
|
Shin YE, Kim H, Yoo JJ, Kim SG, Kim YS. A Potential Pneumothorax Induced by Immune Checkpoint Inhibitors: A Case Report and Literature Review. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1634. [PMID: 39459421 PMCID: PMC11509409 DOI: 10.3390/medicina60101634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024]
Abstract
Background and Objectives: Immune checkpoint inhibitors (ICIs), which target immune checkpoints in cancer cells, are increasingly used as a mainstay in anticancer treatment. The combination of atezolizumab and bevacizumab is also a first-line treatment for hepatocellular carcinoma (HCC). However, ICIs can cause immune-related adverse events (IrAEs) which range from mild to severe, potentially leading to the need for discontinuing immunotherapy. We report a case of a pneumothorax, a rare side effect caused by IrAEs. Materials and Methods: This paper reports a case of a 78-year-old male HCC patient who developed a recurrent pneumothorax, suspected to be an adverse effect of ICIs. Results: The patient was a current smoker with a 30 pack-year smoking history. Prior to initiating ICIs, a chest CT scan showed mild emphysema and fibrosis attributable to smoking. Following ICI treatment, the patient developed a recurrent pneumothorax. Further tests revealed no underlying cause for the pneumothorax other than the ICIs and smoking, and there were no signs of intrapulmonary metastasis or pneumonitis. Conclusions: When a pneumothorax occurs in a patient undergoing immunotherapy, it is important to consider it as a potential adverse effect of the treatment. Special attention should be given to the possibility that immunotherapy may exacerbate underlying lung conditions. Patients should be advised on the importance of smoking cessation. As there are currently no guidelines for resuming immunotherapy after a pneumothorax, it is crucial to weigh the risks and benefits and consider dose reduction or discontinuation of the medication.
Collapse
Affiliation(s)
| | | | - Jeong-Ju Yoo
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon 14584, Republic of Korea; (Y.-E.S.); (H.K.); (S.G.K.); (Y.S.K.)
| | | | | |
Collapse
|
2
|
Smits AJ, Isebia K, Combee-Duffy C, van der Wal S, Nossent EJ, Boonstra A, Vonk-Noordegraaf A, Bogaard HJ, Serné EH. Low nailfold capillary density in patients with pulmonary arterial hypertension and chronic thromboembolic pulmonary hypertension: biomarker of clinical outcome? Sci Rep 2024; 14:19467. [PMID: 39174569 PMCID: PMC11341751 DOI: 10.1038/s41598-024-69017-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/30/2024] [Indexed: 08/24/2024] Open
Abstract
Nailfold capillary density is lower in patients with pulmonary arterial hypertension (PAH). It is unclear whether this observation signifies a unique systemic manifestation of PAH, or reflects microcirculatory dysfunction secondary to pulmonary hypertension (PH). Capillary density and loop dimensions were measured by nailfold-capillaroscopy (NC) in 30 PAH (23 idiopathic, or iPAH, 7 hereditary, or hPAH), 17 chronic thromboembolic PH (CTEPH) patients and 48 controls. NC-Measurements were repeated after pulmonary endarterectomy (PEA) or balloon pulmonary angioplasty (BPA) in CTEPH patients. We examined whether NC-measurements were related to markers of disease severity and predictive of time to clinical worsening (TTCW) as tested by univariate linear/logistic regression and cox-regression analysis, respectively. Capillary density was significantly lower in PAH (7.5 ± 1.1, p < 0.001) and in CTEPH (8.4 ± 1.5, p < 0.001) compared to asymptomatic controls (10.3 ± 1.0 capillaries/mm). Capillary density was similar in iPAH and hPAH and unrelated to hemodynamics in either PAH or CTEPH. A lower capillary density was predictive of clinical worsening in PAH (p 0.05). After normalization of pulmonary artery pressures by PEA or BPA, capillary density remained reduced in CTEPH patients. Capillary loop apex, capillary and venous- and arterial limb diameter were increased in patients with PAH and CTEPH compared to controls. Nailfold capillary density is reduced to a similar extent in iPAH, hPAH and CTEPH. Normalization of hemodynamics by PEA or BPA does not lead to a restoration of capillary density in CTEPH. Capillary dimensions were increased in both patients with PAH and CTEPH. Lower capillary density was predictive of clinical worsening in PAH. Our findings indicate that a loss of peripheral capillaries is not specific to PAH and is not related to the hemodynamic disturbance per se, but that shared mechanisms may account for a simultaneous development of a systemic microangiopathy and pulmonary vascular remodeling.
Collapse
Affiliation(s)
- A J Smits
- Department of Pulmonology, Amsterdam University Medical Center, De Boelelaan 1119, 1081 HV, Amsterdam, The Netherlands
| | - K Isebia
- Department of Vascular Medicine, Amsterdam University Medical Center, De Boelelaan 1119, 1081 HV, Amsterdam, The Netherlands
| | - C Combee-Duffy
- Department of Vascular Medicine, Amsterdam University Medical Center, De Boelelaan 1119, 1081 HV, Amsterdam, The Netherlands
| | - S van der Wal
- Department of Pulmonology, Amsterdam University Medical Center, De Boelelaan 1119, 1081 HV, Amsterdam, The Netherlands
| | - E J Nossent
- Department of Pulmonology, Amsterdam University Medical Center, De Boelelaan 1119, 1081 HV, Amsterdam, The Netherlands
| | - A Boonstra
- Department of Pulmonology, Amsterdam University Medical Center, De Boelelaan 1119, 1081 HV, Amsterdam, The Netherlands
| | - A Vonk-Noordegraaf
- Department of Pulmonology, Amsterdam University Medical Center, De Boelelaan 1119, 1081 HV, Amsterdam, The Netherlands
| | - H J Bogaard
- Department of Pulmonology, Amsterdam University Medical Center, De Boelelaan 1119, 1081 HV, Amsterdam, The Netherlands
| | - E H Serné
- Department of Vascular Medicine, Amsterdam University Medical Center, De Boelelaan 1119, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
3
|
Bahi M, Li C, Wang G, Korman BD. Systemic Sclerosis-Associated Pulmonary Arterial Hypertension: From Bedside to Bench and Back Again. Int J Mol Sci 2024; 25:4728. [PMID: 38731946 PMCID: PMC11084945 DOI: 10.3390/ijms25094728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/02/2024] [Accepted: 04/11/2024] [Indexed: 05/13/2024] Open
Abstract
Systemic sclerosis (SSc) is a heterogeneous disease characterized by autoimmunity, vasculopathy, and fibrosis which affects the skin and internal organs. One key aspect of SSc vasculopathy is pulmonary arterial hypertension (SSc-PAH) which represents a leading cause of morbidity and mortality in patients with SSc. The pathogenesis of pulmonary hypertension is complex, with multiple vascular cell types, inflammation, and intracellular signaling pathways contributing to vascular pathology and remodeling. In this review, we focus on shared molecular features of pulmonary hypertension and those which make SSc-PAH a unique entity. We highlight advances in the understanding of the clinical and translational science pertinent to this disease. We first review clinical presentations and phenotypes, pathology, and novel biomarkers, and then highlight relevant animal models, key cellular and molecular pathways in pathogenesis, and explore emerging treatment strategies in SSc-PAH.
Collapse
Affiliation(s)
| | | | | | - Benjamin D. Korman
- Division of Allergy, Immunology, and Rheumatology, University of Rochester Medical Center, 601 Elmwood Ave, Box 695, Rochester, NY 14642, USA; (M.B.)
| |
Collapse
|
4
|
Lipskaia L, Breau M, Cayrou C, Churikov D, Braud L, Jacquet J, Born E, Fouillade C, Curras-Alonso S, Bauwens S, Jourquin F, Fiore F, Castellano R, Josselin E, Sánchez-Ferrer C, Giovinazzo G, Lachaud C, Gilson E, Flores I, Londono-Vallejo A, Adnot S, Géli V. mTert induction in p21-positive cells counteracts capillary rarefaction and pulmonary emphysema. EMBO Rep 2024; 25:1650-1684. [PMID: 38424230 PMCID: PMC10933469 DOI: 10.1038/s44319-023-00041-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 03/02/2024] Open
Abstract
Lung diseases develop when telomeres shorten beyond a critical point. We constructed a mouse model in which the catalytic subunit of telomerase (mTert), or its catalytically inactive form (mTertCI), is expressed from the p21Cdkn1a locus. Expression of either TERT or TERTCI reduces global p21 levels in the lungs of aged mice, highlighting TERT non-canonical function. However, only TERT reduces accumulation of very short telomeres, oxidative damage, endothelial cell (ECs) senescence and senile emphysema in aged mice. Single-cell analysis of the lung reveals that p21 (and hence TERT) is expressed mainly in the capillary ECs. We report that a fraction of capillary ECs marked by CD34 and endowed with proliferative capacity declines drastically with age, and this is counteracted by TERT but not TERTCI. Consistently, only TERT counteracts decline of capillary density. Natural aging effects are confirmed using the experimental model of emphysema induced by VEGFR2 inhibition and chronic hypoxia. We conclude that catalytically active TERT prevents exhaustion of the putative CD34 + EC progenitors with age, thus protecting against capillary vessel loss and pulmonary emphysema.
Collapse
Affiliation(s)
- Larissa Lipskaia
- Institute for Lung Health, Justus Liebig University, Giessen, Germany
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, FHU SENEC, AP-HP, 94010, Créteil, and Université Paris-Est Créteil (UPEC), Paris, France
| | - Marielle Breau
- Marseille Cancer Research Centre (CRCM), U1068 INSERM, UMR7258 CNRS, UM105 Aix-Marseille University, Institut Paoli-Calmettes, Ligue Nationale Contre le Cancer (Equipe labellisée), Team Telomeres and Chromatin, Marseille, France
| | - Christelle Cayrou
- Marseille Cancer Research Centre (CRCM), U1068 INSERM, UMR7258 CNRS, UM105 Aix-Marseille University, Institut Paoli-Calmettes, Ligue Nationale Contre le Cancer (Equipe labellisée), Team Telomeres and Chromatin, Marseille, France
| | - Dmitri Churikov
- Marseille Cancer Research Centre (CRCM), U1068 INSERM, UMR7258 CNRS, UM105 Aix-Marseille University, Institut Paoli-Calmettes, Ligue Nationale Contre le Cancer (Equipe labellisée), Team Telomeres and Chromatin, Marseille, France
| | - Laura Braud
- Marseille Cancer Research Centre (CRCM), U1068 INSERM, UMR7258 CNRS, UM105 Aix-Marseille University, Institut Paoli-Calmettes, Ligue Nationale Contre le Cancer (Equipe labellisée), Team Telomeres and Chromatin, Marseille, France
| | - Juliette Jacquet
- Institute for Lung Health, Justus Liebig University, Giessen, Germany
| | - Emmanuelle Born
- Institute for Lung Health, Justus Liebig University, Giessen, Germany
| | - Charles Fouillade
- Institut Curie, Inserm U1021, CNRS UMR 3347, University Paris-Saclay, PSL Research University, Orsay, France
| | - Sandra Curras-Alonso
- Institut Curie, PSL Research University, CNRS UMR3244, Sorbonne Université, Telomeres and Cancer, 75005, Paris, France
| | - Serge Bauwens
- Université Côte d'Azur, CNRS, Inserm, IRCAN, Faculty of Medicine, Nice, France
| | - Frederic Jourquin
- Marseille Cancer Research Centre (CRCM), U1068 INSERM, UMR7258 CNRS, UM105 Aix-Marseille University, Institut Paoli-Calmettes, Ligue Nationale Contre le Cancer (Equipe labellisée), Team Telomeres and Chromatin, Marseille, France
| | - Frederic Fiore
- Centre d'Immunophénomique, Aix Marseille Université, INSERM, CNRS UMR, Marseille, France
| | - Rémy Castellano
- Marseille Cancer Research Centre (CRCM), TrGET Preclinical Platform, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille Université, Marseille, France
| | - Emmanuelle Josselin
- Marseille Cancer Research Centre (CRCM), TrGET Preclinical Platform, Institut Paoli-Calmettes, Inserm, CNRS, Aix Marseille Université, Marseille, France
| | | | - Giovanna Giovinazzo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029, Madrid, Spain
| | - Christophe Lachaud
- Marseille Cancer Research Centre (CRCM), U1068 INSERM, UMR7258 CNRS, UM105 Aix-Marseille University, Institut Paoli-Calmettes, Team DNA Interstrand Crosslink Lesions and Blood Disorders, Marseille, France
| | - Eric Gilson
- Université Côte d'Azur, CNRS, Inserm, IRCAN, Faculty of Medicine, Nice, France
| | - Ignacio Flores
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029, Madrid, Spain
- Centro de Biologia Molecular Severo Ochoa, CSIC-UAM, Cantoblanco, Madrid, Spain
| | - Arturo Londono-Vallejo
- Institut Curie, PSL Research University, CNRS UMR3244, Sorbonne Université, Telomeres and Cancer, 75005, Paris, France
| | - Serge Adnot
- Institute for Lung Health, Justus Liebig University, Giessen, Germany.
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, FHU SENEC, AP-HP, 94010, Créteil, and Université Paris-Est Créteil (UPEC), Paris, France.
| | - Vincent Géli
- Marseille Cancer Research Centre (CRCM), U1068 INSERM, UMR7258 CNRS, UM105 Aix-Marseille University, Institut Paoli-Calmettes, Ligue Nationale Contre le Cancer (Equipe labellisée), Team Telomeres and Chromatin, Marseille, France.
| |
Collapse
|
5
|
Trivedi A, Lu TM, Summers B, Kim K, Rhee AJ, Houghton S, Byers DE, Lis R, Reed HO. Lung lymphatic endothelial cells undergo inflammatory and prothrombotic changes in a model of chronic obstructive pulmonary disease. Front Cell Dev Biol 2024; 12:1344070. [PMID: 38440076 PMCID: PMC10910060 DOI: 10.3389/fcell.2024.1344070] [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: 11/24/2023] [Accepted: 01/29/2024] [Indexed: 03/06/2024] Open
Abstract
The lymphatic vasculature regulates lung homeostasis through drainage of fluid and trafficking of immune cells and plays a key role in the response to lung injury in several disease states. We have previously shown that lymphatic dysfunction occurs early in the pathogenesis of chronic obstructive pulmonary disease (COPD) caused by cigarette smoke (CS) and that this is associated with increased thrombin and fibrin clots in lung lymph. However, the direct effects of CS and thrombin on lymphatic endothelial cells (LECs) in COPD are not entirely clear. Studies of the blood vasculature have shown that COPD is associated with increased thrombin after CS exposure that causes endothelial dysfunction characterized by changes in the expression of coagulation factors and leukocyte adhesion proteins. Here, we determined whether similar changes occur in LECs. We used an in vitro cell culture system and treated human lung microvascular lymphatic endothelial cells with cigarette smoke extract (CSE) and/or thrombin. We found that CSE treatment led to decreased fibrinolytic activity in LECs, which was associated with increased expression of plasminogen activator inhibitor 1 (PAI-1). LECs treated with both CSE and thrombin together had a decreased expression of tissue factor pathway inhibitor (TFPI) and increased expression of adhesion molecules. RNA sequencing of lung LECs isolated from mice exposed to CS also showed upregulation of prothrombotic and inflammatory pathways at both acute and chronic exposure time points. Analysis of publicly available single-cell RNA sequencing of LECs as well as immunohistochemical staining of lung tissue from COPD patients supported these data and showed increased expression of inflammatory markers in LECs from COPD patients compared to those from controls. These studies suggest that in parallel with blood vessels, the lymphatic endothelium undergoes inflammatory changes associated with CS exposure and increased thrombin in COPD. Further research is needed to unravel the mechanisms by which these changes affect lymphatic function and drive tissue injury in COPD.
Collapse
Affiliation(s)
- Anjali Trivedi
- Department of Medicine, Division of Pulmonary and Critical Care, Weill Cornell Medicine, New York, NY, United States
| | - Tyler M. Lu
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY, United States
- Molecular and Cellular Biology Program, SUNY Downstate School of Graduate Studies, Brooklyn, NY, United States
| | - Barbara Summers
- Department of Medicine, Division of Pulmonary and Critical Care, Weill Cornell Medicine, New York, NY, United States
| | - Kihwan Kim
- Department of Medicine, Division of Pulmonary and Critical Care, Weill Cornell Medicine, New York, NY, United States
| | - Alexander J. Rhee
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Sean Houghton
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Derek E. Byers
- Department of Medicine, Division of Pulmonary and Critical Care, Washington University School of Medicine, St. Louis, MO, United States
| | - Raphaël Lis
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Hasina Outtz Reed
- Department of Medicine, Division of Pulmonary and Critical Care, Weill Cornell Medicine, New York, NY, United States
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
6
|
Bikou O, Sassi Y. The Sugen/Hypoxia Rat Model for Pulmonary Hypertension and Right Heart Failure. Methods Mol Biol 2024; 2803:163-172. [PMID: 38676892 DOI: 10.1007/978-1-0716-3846-0_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2024]
Abstract
Pulmonary hypertension (PH) is a devastating disease, characterized by complex remodeling of the pulmonary vasculature. PH is classified into five groups based on different etiology, pathology, as well as therapy and prognosis. Animal models are essential for the study of underlying mechanisms, pathophysiology, and preclinical testing of new therapies for PH. The complexity of the disease with different clinical entities dictates the necessity for more than one animal model to resemble PH, as a single model cannot imitate the broad spectrum of human PH.Here we describe a detailed protocol for creating a rat model of PH with right ventricular (RV) failure. Furthermore, we present how to characterize it hemodynamically by invasive measurements of RV and pulmonary arterial (PA) pressures. Animals subjected to this model display severe pulmonary vascular remodeling and RV dysfunction. In this model, rats undergo a single subcutaneous injection of Sugen (SU5416, a vascular endothelial growth factor inhibitor) and are immediately exposed to chronic hypoxia in a hypoxia chamber for 3-6 weeks. This Sugen/Hypoxia rat model resembles Group 1 PH.
Collapse
Affiliation(s)
- Olympia Bikou
- Department of Medicine I, LMU University Hospital, LMU Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich, Germany
| | - Yassine Sassi
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA.
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA.
- Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA, USA.
| |
Collapse
|
7
|
Pham AT, Oliveira AC, Albanna M, Alvarez-Castanon J, Dupee Z, Patel D, Fu C, Mukhsinova L, Nguyen A, Jin L, Bryant AJ. Non-Interferon-Dependent Role of STING Signaling in Pulmonary Hypertension. Arterioscler Thromb Vasc Biol 2024; 44:124-142. [PMID: 37942608 PMCID: PMC10872846 DOI: 10.1161/atvbaha.123.320121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 10/24/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND Patients with constitutive activation of DNA-sensing pathway through stimulator of IFN (interferon) genes (STING), such as those with STING-associated vasculopathy with onset in infancy, develop pulmonary hypertension (PH). However, the role of STING signaling in general PH patients is heretofore undescribed. Here, we seek to investigate the role of STING in PH development. METHODS STING expression in patient lung samples was examined. PH was induced in global STING-deficient mice and global type I IFN receptor 1-deficient mice using bleomycin or chronic hypoxia exposure. PH development was evaluated by right ventricular systolic pressure and Fulton index, with additional histological and flow cytometric analysis. VEGF (vascular endothelial growth factor) expression on murine immune cells was quantified and evaluated with multiplex and flow cytometry. Human myeloid-derived cells were differentiated from peripheral blood mononuclear cells and treated with either STING agonist or STING antagonist for evaluation of VEGF secretion. RESULTS Global STING deficiency protects mice from PH development, and STING-associated PH seems independent of type I IFN signaling. Furthermore, a role for STING-VEGF signaling pathway in PH development was demonstrated, with altered VEGF secretion in murine pulmonary infiltrated myeloid cells in a STING-dependent manner. In addition, pharmacological manipulation of STING in human myeloid-derived cells supports in vivo findings. Finally, a potential role of STING-VEGF-mediated apoptosis in disease development and progression was illustrated, providing a roadmap toward potential therapeutic applications. CONCLUSIONS Overall, these data provide concrete evidence of STING involvement in PH, establishing biological plausibility for STING-related therapies in PH treatment.
Collapse
Affiliation(s)
- Ann T Pham
- Department of Medicine, University of Florida College of Medicine, Gainesville
| | - Aline C Oliveira
- Department of Medicine, University of Florida College of Medicine, Gainesville
| | - Muhammad Albanna
- Department of Medicine, University of Florida College of Medicine, Gainesville
| | | | - Zadia Dupee
- Department of Medicine, University of Florida College of Medicine, Gainesville
| | - Diya Patel
- Department of Medicine, University of Florida College of Medicine, Gainesville
| | - Chunhua Fu
- Department of Medicine, University of Florida College of Medicine, Gainesville
| | - Laylo Mukhsinova
- Department of Medicine, University of Florida College of Medicine, Gainesville
| | - Amy Nguyen
- Department of Medicine, University of Florida College of Medicine, Gainesville
| | - Lei Jin
- Department of Medicine, University of Florida College of Medicine, Gainesville
| | - Andrew J Bryant
- Department of Medicine, University of Florida College of Medicine, Gainesville
| |
Collapse
|
8
|
Mebratu YA, Jones JT, Liu C, Negasi ZH, Rahman M, Rojas-Quintero J, O’Connor GT, Gao W, Dupuis J, Cho MH, Litonjua AA, Randell S, Tesfaigzi Y. Bik promotes proteasomal degradation to control low-grade inflammation. J Clin Invest 2023; 134:e170594. [PMID: 38113109 PMCID: PMC10866658 DOI: 10.1172/jci170594] [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: 03/20/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023] Open
Abstract
Although chronic low-grade inflammation does not cause immediate clinical symptoms, over the longer term, it can enhance other insults or age-dependent damage to organ systems and thereby contribute to age-related disorders, such as respiratory disorders, heart disease, metabolic disorders, autoimmunity, and cancer. However, the molecular mechanisms governing low-level inflammation are largely unknown. We discovered that Bcl-2-interacting killer (Bik) deficiency causes low-level inflammation even at baseline and the development of spontaneous emphysema in female but not male mice. Similarly, a single nucleotide polymorphism that reduced Bik levels was associated with increased inflammation and enhanced decline in lung function in humans. Transgenic expression of Bik in the airways of Bik-deficient mice inhibited allergen- or LPS-induced lung inflammation and reversed emphysema in female mice. Bik deficiency increased nuclear but not cytosolic p65 levels because Bik, by modifying the BH4 domain of Bcl-2, interacted with regulatory particle non-ATPase 1 (RPN1) and RPN2 and enhanced proteasomal degradation of nuclear proteins. Bik deficiency increased inflammation primarily in females because Bcl-2 and Bik levels were reduced in lung tissues and airway cells of female compared with male mice. Therefore, controlling low-grade inflammation by modifying the unappreciated role of Bik and Bcl-2 in facilitating proteasomal degradation of nuclear proteins may be crucial in treating chronic age-related diseases.
Collapse
Affiliation(s)
- Yohannes A. Mebratu
- Brigham and Women’s Hospital, Division of Pulmonary and Critical Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Jane T. Jones
- University Geisel School of Medicine, Department of Microbiology and Immunology, Dartmouth, Hanover, New Hampshire, USA
| | - Congjian Liu
- Brigham and Women’s Hospital, Division of Pulmonary and Critical Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Zerihun H. Negasi
- Brigham and Women’s Hospital, Division of Pulmonary and Critical Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Mizanur Rahman
- Brigham and Women’s Hospital, Division of Pulmonary and Critical Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Joselyn Rojas-Quintero
- Brigham and Women’s Hospital, Division of Pulmonary and Critical Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - George T. O’Connor
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
- National Heart, Lung, and Blood Institute’s (NHLBI’s) Framingham Heart Study, Framingham, Massachusetts, USA
| | - Wei Gao
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
- Department of Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Canada
| | - Michael H. Cho
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Augusto A. Litonjua
- Division of Pediatric Pulmonary Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Scott Randell
- Marsico Lung Institute, UNC School of Medicine, Chapel Hill, North Carolina, USA
| | - Yohannes Tesfaigzi
- Brigham and Women’s Hospital, Division of Pulmonary and Critical Medicine, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Haffez H, Elsayed NA, Ahmed MF, Fatahala SS, Khaleel EF, Badi RM, Elkaeed EB, El Hassab MA, Hammad SF, Eldehna WM, Masurier N, El-Haggar R. Novel N-Arylmethyl-aniline/chalcone hybrids as potential VEGFR inhibitors: synthesis, biological evaluations, and molecular dynamic simulations. J Enzyme Inhib Med Chem 2023; 38:2278022. [PMID: 37982203 PMCID: PMC11003488 DOI: 10.1080/14756366.2023.2278022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 10/25/2023] [Indexed: 11/21/2023] Open
Abstract
Significant advancements have been made in the domain of targeted anticancer therapy for the management of malignancies in recent times. VEGFR-2 is characterised by its pivotal involvement in angiogenesis and subsequent mechanisms that promote tumour cells survival. Herein, novel N-arylmethyl-aniline/chalcone hybrids 5a-5n were designed and synthesised as potential anticancer and VEGFR-2 inhibitors. The anticancer activity was evaluated at the NCI-USA, resulting in the identification of 10 remarkably potent molecules 5a-5j that were further subjected to the five-dose assays. Thereafter, they were explored for their VEGFR-2 inhibitory activity where 5e and 5h emerged as the most potent inhibitors. 5e and 5h induced apoptosis with cell cycle arrest at the SubG0-G1 phase within HCT-116 cells. Moreover, their impact on some key apoptotic genes was assessed, suggesting caspase-dependent apoptosis. Furthermore, molecular docking and molecular dynamics simulations were conducted to explore the binding modes and stability of the protein-ligand complexes.
Collapse
Affiliation(s)
- Hesham Haffez
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo, Ain Helwan, Egypt
- Center of Scientific Excellence “Helwan Structural Biology Research, (HSBR)”, Helwan University, Cairo, Egypt
| | - Nosaiba A. Elsayed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, Cairo, Ain Helwan, Egypt
| | - Marwa F. Ahmed
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, Cairo, Ain Helwan, Egypt
| | - Samar S. Fatahala
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Eman F. Khaleel
- Department of Medical Physiology, College of Medicine, King Khalid University, Asir, Saudi Arabia
| | - Rehab Mustafa Badi
- Department of Medical Physiology, College of Medicine, King Khalid University, Asir, Saudi Arabia
| | - Eslam B. Elkaeed
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Riyadh, Saudi Arabia
| | - Mahmoud A. El Hassab
- Department of Medicinal Chemistry, Faculty of Pharmacy, King Salman International University (KSIU), South Sinai, Egypt
| | - Sherif F. Hammad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, Cairo, Ain Helwan, Egypt
- Medicinal Chemistry Department, PharmD Program, Egypt-Japan University of Science and Technology (E-JUST), New Borg El-Arab City, Egypt Alexandria
| | - Wagdy M. Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Nicolas Masurier
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS, Université de Montpellier, ENSCM, Montpellier, France
| | - Radwan El-Haggar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, Cairo, Ain Helwan, Egypt
| |
Collapse
|
10
|
van Nijnatten J, Faiz A, Timens W, Guryev V, Slebos DJ, Klooster K, Hartman JE, Kole T, Choy DF, Chakrabarti A, Grimbaldeston M, Rosenberger CM, Kerstjens H, Brandsma CA, van den Berge M. A bronchial gene signature specific for severe COPD that is retained in the nose. ERJ Open Res 2023; 9:00354-2023. [PMID: 38020574 PMCID: PMC10680034 DOI: 10.1183/23120541.00354-2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/09/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction A subset of COPD patients develops advanced disease with severe airflow obstruction, hyperinflation and extensive emphysema. We propose that the pathogenesis in these patients differs from mild-moderate COPD and is reflected by bronchial gene expression. The aim of the present study was to identify a unique bronchial epithelial gene signature for severe COPD patients. Methods We obtained RNA sequencing data from bronchial brushes from 123 ex-smokers with severe COPD, 23 with mild-moderate COPD and 23 non-COPD controls. We identified genes specific to severe COPD by comparing severe COPD to non-COPD controls, followed by removing genes that were also differentially expressed between mild-moderate COPD and non-COPD controls. Next, we performed a pathway analysis on these genes and evaluated whether this signature is retained in matched nasal brushings. Results We identified 219 genes uniquely differentially expressed in severe COPD. Interaction network analysis identified VEGFA and FN1 as the key genes with the most interactions. Genes were involved in extracellular matrix regulation, collagen binding and the immune response. Of interest were 10 genes (VEGFA, DCN, SPARC, COL6A2, MGP, CYR61, ANXA6, LGALS1, C1QA and C1QB) directly connected to fibronectin 1 (FN1). Most of these genes were lower expressed in severe COPD and showed the same effect in nasal brushings. Conclusions We found a unique severe COPD bronchial gene signature with key roles for VEGFA and FN1, which was retained in the upper airways. This supports the hypothesis that severe COPD, at least partly, comprises a different pathology and supports the potential for biomarker development based on nasal brushes in COPD.
Collapse
Affiliation(s)
- Jos van Nijnatten
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands
- University of Groningen University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
- University of Technology Sydney, Respiratory Bioinformatics and Molecular Biology, Sydney, NSW, Australia
| | - Alen Faiz
- University of Groningen University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
- University of Technology Sydney, Respiratory Bioinformatics and Molecular Biology, Sydney, NSW, Australia
- University of Groningen University Medical Center Groningen, Department of Pulmonary Diseases, Groningen, the Netherlands
| | - Wim Timens
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands
- University of Groningen University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
| | - Victor Guryev
- University of Groningen University Medical Center Groningen, European Research Institute for the Biology of Ageing, Groningen, the Netherlands
| | - Dirk-Jan Slebos
- University of Groningen University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
- University of Groningen University Medical Center Groningen, Department of Pulmonary Diseases, Groningen, the Netherlands
| | - Karin Klooster
- University of Groningen University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
- University of Groningen University Medical Center Groningen, Department of Pulmonary Diseases, Groningen, the Netherlands
| | - Jorine E. Hartman
- University of Groningen University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
- University of Groningen University Medical Center Groningen, Department of Pulmonary Diseases, Groningen, the Netherlands
| | - Tessa Kole
- University of Groningen University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
- University of Groningen University Medical Center Groningen, Department of Pulmonary Diseases, Groningen, the Netherlands
| | | | | | | | | | - Huib Kerstjens
- University of Groningen University Medical Center Groningen, Department of Pulmonary Diseases, Groningen, the Netherlands
| | - Corry-Anke Brandsma
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands
- University of Groningen University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
- These authors contributed equally
| | - Maarten van den Berge
- University of Groningen University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, the Netherlands
- University of Groningen University Medical Center Groningen, Department of Pulmonary Diseases, Groningen, the Netherlands
- These authors contributed equally
| |
Collapse
|
11
|
Korde A, Haslip M, Pednekar P, Khan A, Chioccioli M, Mehta S, Lopez-Giraldez F, Bermejo S, Rojas M, Dela Cruz C, Matthay MA, Pober JS, Pierce RW, Takyar SS. MicroRNA-1 protects the endothelium in acute lung injury. JCI Insight 2023; 8:e164816. [PMID: 37737266 PMCID: PMC10561733 DOI: 10.1172/jci.insight.164816] [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: 08/31/2022] [Accepted: 08/10/2023] [Indexed: 09/23/2023] Open
Abstract
Acute lung injury (ALI) and its most severe form, acute respiratory distress syndrome (ARDS), cause severe endothelial dysfunction in the lung, and vascular endothelial growth factor (VEGF) is elevated in ARDS. We found that the levels of a VEGF-regulated microRNA, microRNA-1 (miR-1), were reduced in the lung endothelium after acute injury. Pulmonary endothelial cell-specific (EC-specific) overexpression of miR-1 protected the lung against cell death and barrier dysfunction in both murine and human models and increased the survival of mice after pneumonia-induced ALI. miR-1 had an intrinsic protective effect in pulmonary and other types of ECs; it inhibited apoptosis and necroptosis pathways and decreased capillary leak by protecting adherens and tight junctions. Comparative gene expression analysis and RISC recruitment assays identified miR-1 targets in the context of injury, including phosphodiesterase 5A (PDE5A), angiopoietin-2 (ANGPT2), CNKSR family member 3 (CNKSR3), and TNF-α-induced protein 2 (TNFAIP2). We validated miR-1-mediated regulation of ANGPT2 in both mouse and human ECs and found that in a 119-patient pneumonia cohort, miR-1 correlated inversely with ANGPT2. These findings illustrate a previously unknown role of miR-1 as a cytoprotective orchestrator of endothelial responses to acute injury with prognostic and therapeutic potential.
Collapse
Affiliation(s)
- Asawari Korde
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Maria Haslip
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Prachi Pednekar
- Department of Medicine, Yale New Haven Hospital, New Haven, Connecticut, USA
| | | | - Maurizio Chioccioli
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sameet Mehta
- Department of Genetics, Yale University School Medicine, New Haven, Connecticut, USA
| | | | - Santos Bermejo
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Mauricio Rojas
- Division of Pulmonary, Critical Care and Sleep Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Charles Dela Cruz
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Michael A. Matthay
- Cardiovascular Research Institute, Department of Medicine and Anesthesiology, UCSF, San Francisco, California, USA
| | | | | | - Shervin S. Takyar
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
12
|
Haraguchi T, Matsuoka S, Yagihashi K, Matsushita S, Yamashiro T, Kobayashi Y, Mimura H. Quantitative Computed Tomography Analysis of the Longitudinal Change Between Centrilobular and Paraseptal Emphysema Subtypes: A Retrospective Study. J Comput Assist Tomogr 2023; 47:746-752. [PMID: 37707404 DOI: 10.1097/rct.0000000000001482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
OBJECTIVE This study aimed to investigate the difference between the extent of centrilobular emphysema (CLE) and paraseptal emphysema (PSE) on follow-up chest CT scans and their relationship to the cross-sectional area (CSA) of small pulmonary vessels. METHODS Sixty-two patients (36 CLE and 26 PSE) who underwent 2 chest CT scans were enrolled in this study. The percentage of low attenuation volume (%LAV) and total CSA of the small pulmonary vessels <5 mm 2 (%CSA < 5) were measured at the 2 time points. Analysis of the initial %CSA < 5 and the change in the %LAV and %CSA < 5 on follow-up imaging was performed. RESULTS The initial %CSA < 5 was not significantly different between the CLE and the PSE groups (CLE, 0.66 vs. PSE, 0.71; P = 0.78). There was no significant difference in the longitudinal change in the %LAV between the 2 groups (CLE, -0.048% vs. PSE, 0.005%; P = 0.26). The longitudinal change in the %CSA < 5 in patients with PSE significantly decreased compared with those with CLE (CLE, 0.025% vs. PSE, -0.018%; P = 0.02). CONCLUSIONS The longitudinal change in the %CSA < 5 was significantly different for patients with CLE and PSE, demonstrating an important pathophysiological difference between the subtypes.
Collapse
Affiliation(s)
| | - Shin Matsuoka
- From the Department of Radiology, St. Marianna University School of Medicine, Sugao 2-16-1, Miyamae-ku, Kawasaki, Japan
| | - Kunihiro Yagihashi
- From the Department of Radiology, St. Marianna University School of Medicine, Sugao 2-16-1, Miyamae-ku, Kawasaki, Japan
| | | | | | | | - Hidefumi Mimura
- From the Department of Radiology, St. Marianna University School of Medicine, Sugao 2-16-1, Miyamae-ku, Kawasaki, Japan
| |
Collapse
|
13
|
Borek I, Birnhuber A, Voelkel NF, Marsh LM, Kwapiszewska G. The vascular perspective on acute and chronic lung disease. J Clin Invest 2023; 133:e170502. [PMID: 37581311 PMCID: PMC10425217 DOI: 10.1172/jci170502] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2023] Open
Abstract
The pulmonary vasculature has been frequently overlooked in acute and chronic lung diseases, such as acute respiratory distress syndrome (ARDS), pulmonary fibrosis (PF), and chronic obstructive pulmonary disease (COPD). The primary emphasis in the management of these parenchymal disorders has largely revolved around the injury and aberrant repair of epithelial cells. However, there is increasing evidence that the vascular endothelium plays an active role in the development of acute and chronic lung diseases. The endothelial cell network in the capillary bed and the arterial and venous vessels provides a metabolically highly active barrier that controls the migration of immune cells, regulates vascular tone and permeability, and participates in the remodeling processes. Phenotypically and functionally altered endothelial cells, and remodeled vessels, can be found in acute and chronic lung diseases, although to different degrees, likely because of disease-specific mechanisms. Since vascular remodeling is associated with pulmonary hypertension, which worsens patient outcomes and survival, it is crucial to understand the underlying vascular alterations. In this Review, we describe the current knowledge regarding the role of the pulmonary vasculature in the development and progression of ARDS, PF, and COPD; we also outline future research directions with the hope of facilitating the development of mechanism-based therapies.
Collapse
Affiliation(s)
- Izabela Borek
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Anna Birnhuber
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Otto Loewi Research Center, Division of Physiology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Norbert F. Voelkel
- Pulmonary Medicine Department, University of Amsterdam Medical Centers, Amsterdam, Netherlands
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Leigh M. Marsh
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Otto Loewi Research Center, Division of Physiology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Otto Loewi Research Center, Division of Physiology and Pathophysiology, Medical University of Graz, Graz, Austria
- Institute for Lung Health, German Lung Center (DZL), Cardiopulmonary Institute, Giessen, Germany
| |
Collapse
|
14
|
Kondo T, Fujiwara K, Nakagawa M, Fujimoto K, Yumita S, Ishino T, Ogawa K, Iwanaga T, Koroki K, Kanzaki H, Inoue M, Kobayashi K, Kiyono S, Nakamura M, Kanogawa N, Ogasawara S, Nakamoto S, Chiba T, Kato J, Kato N. Estimation of the effect of atezolizumab plus bevacizumab on pulmonary arterial hypertension using computed tomography in HCC patients. Sci Rep 2023; 13:11524. [PMID: 37460776 DOI: 10.1038/s41598-023-38377-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/07/2023] [Indexed: 07/20/2023] Open
Abstract
The effect of the combination of atezolizumab and bevacizumab (Atez/Bev) for hepatocellular carcinoma (HCC) on pulmonary arterial hypertension (PAH) is unknown. Estimation of PAH by using computed tomography (CT) has recently been proposed. Thus, we aimed to estimate the effect of Atez/Bev on PAH using CT. Altogether, 113 patients who received Atez/Bev for HCC were enrolled. Probable PAH was defined as the diameter of the main pulmonary artery (mPA-D) ≥ 33 mm, whereas suspicious PAH was defined as mPA-D ≥ 29 mm or mPA-D/the diameter of the ascending aorta (aAo-D) ≥ 1.0. Before treatment, probable/suspicious PAH were diagnosed in 7 (6.7%)/22 (21.0%) patients, respectively. mPA-D and mPA-D/aAo-D significantly increased after induction of Atez/Bev. The increment of mPA-D was correlated with the occurrence of post-treatment respiratory/heart failure. In analysis of 55 patients who underwent CT at 3 months after the last dose of Atez/Bev, mPA-D and mPA-D/aAo-D significantly decreased. However, in the group with continuous treatment of other molecular-targeted drugs after Atez/Bev, mPA-D and mPA-D/aAo-D showed no significant change. In conclusion, PAH may not be a rare complication in patients with HCC and should be managed carefully because of the possible negative effect of Atez/Bev on PAH.
Collapse
Affiliation(s)
- Takayuki Kondo
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba, 260-8670, Japan.
- Ultrasound Center, Chiba University Hospital, Chiba, Japan.
| | - Kisako Fujiwara
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Miyuki Nakagawa
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Kentaro Fujimoto
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Sae Yumita
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Takamasa Ishino
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Keita Ogawa
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Terunao Iwanaga
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Keisuke Koroki
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Hiroaki Kanzaki
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Masanori Inoue
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Kazufumi Kobayashi
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Soichiro Kiyono
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Masato Nakamura
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Naoya Kanogawa
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Sadahisa Ogasawara
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Shingo Nakamoto
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Tetsuhiro Chiba
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Jun Kato
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba, 260-8670, Japan
| | - Naoya Kato
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba, 260-8670, Japan
- Ultrasound Center, Chiba University Hospital, Chiba, Japan
| |
Collapse
|
15
|
Upadhyay P, Wu CW, Pham A, Zeki AA, Royer CM, Kodavanti UP, Takeuchi M, Bayram H, Pinkerton KE. Animal models and mechanisms of tobacco smoke-induced chronic obstructive pulmonary disease (COPD). JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2023; 26:275-305. [PMID: 37183431 PMCID: PMC10718174 DOI: 10.1080/10937404.2023.2208886] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is the third leading cause of death worldwide, and its global health burden is increasing. COPD is characterized by emphysema, mucus hypersecretion, and persistent lung inflammation, and clinically by chronic airflow obstruction and symptoms of dyspnea, cough, and fatigue in patients. A cluster of pathologies including chronic bronchitis, emphysema, asthma, and cardiovascular disease in the form of hypertension and atherosclerosis variably coexist in COPD patients. Underlying causes for COPD include primarily tobacco use but may also be driven by exposure to air pollutants, biomass burning, and workplace related fumes and chemicals. While no single animal model might mimic all features of human COPD, a wide variety of published models have collectively helped to improve our understanding of disease processes involved in the genesis and persistence of COPD. In this review, the pathogenesis and associated risk factors of COPD are examined in different mammalian models of the disease. Each animal model included in this review is exclusively created by tobacco smoke (TS) exposure. As animal models continue to aid in defining the pathobiological mechanisms of and possible novel therapeutic interventions for COPD, the advantages and disadvantages of each animal model are discussed.
Collapse
Affiliation(s)
- Priya Upadhyay
- Center for Health and the Environment, University of California, Davis, Davis, CA 95616 USA
| | - Ching-Wen Wu
- Center for Health and the Environment, University of California, Davis, Davis, CA 95616 USA
| | - Alexa Pham
- Center for Health and the Environment, University of California, Davis, Davis, CA 95616 USA
| | - Amir A. Zeki
- Department of Internal Medicine; Division of Pulmonary, Critical Care, and Sleep Medicine, Center for Comparative Respiratory Biology and Medicine, School of Medicine; University of California, Davis, School of Medicine; U.C. Davis Lung Center; Davis, CA USA
| | - Christopher M. Royer
- California National Primate Research Center, University of California, Davis, Davis, CA 95616 USA
| | - Urmila P. Kodavanti
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Minoru Takeuchi
- Department of Animal Medical Science, Kyoto Sangyo University, Kyoto, Japan
| | - Hasan Bayram
- Koc University Research Center for Translational Medicine (KUTTAM), School of Medicine, Istanbul, Turkey
| | - Kent E. Pinkerton
- Center for Health and the Environment, University of California, Davis, Davis, CA 95616 USA
| |
Collapse
|
16
|
Kreniske JS, Kaner RJ, Glesby MJ. Pathogenesis and management of emphysema in people with HIV. Expert Rev Respir Med 2023; 17:873-887. [PMID: 37848398 PMCID: PMC10872640 DOI: 10.1080/17476348.2023.2272702] [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: 03/04/2023] [Accepted: 10/16/2023] [Indexed: 10/19/2023]
Abstract
INTRODUCTION Since early in the HIV epidemic, emphysema has been identified among people with HIV (PWH) and has been associated with increased mortality. Smoking cessation is key to risk reduction. Health maintenance for PWH and emphysema should ensure appropriate vaccination and lung cancer screening. Treatment should adhere to inhaler guidelines for the general population, but inhaled corticosteroid (ICS) should be used with caution. Frontiers in treatment include targeted therapeutics. Major knowledge gaps exist in the epidemiology of and optimal care for PWH and emphysema, particularly in low and middle-income countries (LMIC). AREAS COVERED Topics addressed include risk factors, pathogenesis, current treatment and prevention strategies, and frontiers in research. EXPERT OPINION There are limited data on the epidemiology of emphysema in LMIC, where more than 90% of deaths from COPD occur and where the morbidity of HIV is most heavily concentrated. The population of PWH is aging, and age-related co-morbidities such as emphysema will only increase in salience. Over the next 5 years, the authors anticipate novel trials of targeted therapy for emphysema specific to PWH, and we anticipate a growing body of evidence to inform optimal clinical care for lung health among PWH in LMIC.
Collapse
Affiliation(s)
- Jonah S. Kreniske
- Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, USA
| | - Robert J. Kaner
- Division of Pulmonary and Critical Care Medicine, Weill Cornell Medical College, USA
- Department of Genetic Medicine, Weill Cornell Medical College, USA
| | - Marshall J. Glesby
- Division of Infectious Diseases, Weill Cornell Medical College, USA
- Department of Population Health Sciences, Weill Cornell Medical College, USA
| |
Collapse
|
17
|
Curtis JL. Understanding COPD Etiology, Pathophysiology, and Definition. Respir Care 2023; 68:859-870. [PMID: 37353333 PMCID: PMC10289621 DOI: 10.4187/respcare.10873] [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] [Indexed: 06/25/2023]
Abstract
COPD, one of the leading worldwide health problems, currently lacks truly disease-modifying medical therapies applicable to most patients. Developing such novel therapies has been hampered by the marked heterogeneity of phenotypes between individuals with COPD. Such heterogeneity suggests that, rather than a single cause (particularly just direct inhalation of tobacco products), development and progression of COPD likely involve both complex gene-by-environment interactions to multiple inhalational exposures and a variety of molecular pathways. However, there has been considerable recent progress toward understanding how specific pathological processes can lead to discrete COPD phenotypes, particularly that of small airways disease. Advances in imaging techniques that correlate to specific types of histological damage, and in the immunological mechanisms of lung damage in COPD, hold promise for development of personalized therapies. At the same time, there is growing recognition that the current diagnostic criteria for COPD, based solely on spirometry, exclude large numbers of individuals with very similar disease manifestations. This concise review summarizes current understanding of the etiology and pathophysiology of COPD and provides background explaining the increasing calls to expand the diagnostic criteria used to diagnose COPD and some challenges in doing so.
Collapse
Affiliation(s)
- Jeffrey L Curtis
- Medical Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan; Division of Pulmonary and Critical Care Medicine, Michigan Medicine, Ann Arbor, Michigan; and Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
18
|
Miklós Z, Horváth I. The Role of Oxidative Stress and Antioxidants in Cardiovascular Comorbidities in COPD. Antioxidants (Basel) 2023; 12:1196. [PMID: 37371927 DOI: 10.3390/antiox12061196] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/28/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Oxidative stress driven by several environmental and local airway factors associated with chronic obstructive bronchiolitis, a hallmark feature of COPD, plays a crucial role in disease pathomechanisms. Unbalance between oxidants and antioxidant defense mechanisms amplifies the local inflammatory processes, worsens cardiovascular health, and contributes to COPD-related cardiovascular dysfunctions and mortality. The current review summarizes recent developments in our understanding of different mechanisms contributing to oxidative stress and its countermeasures, with special attention to those that link local and systemic processes. Major regulatory mechanisms orchestrating these pathways are also introduced, with some suggestions for further research in the field.
Collapse
Affiliation(s)
- Zsuzsanna Miklós
- National Korányi Institute for Pulmonology, Korányi F. Street 1, H-1121 Budapest, Hungary
| | - Ildikó Horváth
- National Korányi Institute for Pulmonology, Korányi F. Street 1, H-1121 Budapest, Hungary
- Department of Pulmonology, University of Debrecen, Nagyerdei krt 98, H-4032 Debrecen, Hungary
| |
Collapse
|
19
|
Jandl K, Radic N, Zeder K, Kovacs G, Kwapiszewska G. Pulmonary vascular fibrosis in pulmonary hypertension - The role of the extracellular matrix as a therapeutic target. Pharmacol Ther 2023; 247:108438. [PMID: 37210005 DOI: 10.1016/j.pharmthera.2023.108438] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/03/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
Pulmonary hypertension (PH) is a condition characterized by changes in the extracellular matrix (ECM) deposition and vascular remodeling of distal pulmonary arteries. These changes result in increased vessel wall thickness and lumen occlusion, leading to a loss of elasticity and vessel stiffening. Clinically, the mechanobiology of the pulmonary vasculature is becoming increasingly recognized for its prognostic and diagnostic value in PH. Specifically, the increased vascular fibrosis and stiffening resulting from ECM accumulation and crosslinking may be a promising target for the development of anti- or reverse-remodeling therapies. Indeed, there is a huge potential in therapeutic interference with mechano-associated pathways in vascular fibrosis and stiffening. The most direct approach is aiming to restore extracellular matrix homeostasis, by interference with its production, deposition, modification and turnover. Besides structural cells, immune cells contribute to the level of ECM maturation and degradation by direct cell-cell contact or the release of mediators and proteases, thereby opening a huge avenue to target vascular fibrosis via immunomodulation approaches. Indirectly, intracellular pathways associated with altered mechanobiology, ECM production, and fibrosis, offer a third option for therapeutic intervention. In PH, a vicious cycle of persistent activation of mechanosensing pathways such as YAP/TAZ initiates and perpetuates vascular stiffening, and is linked to key pathways disturbed in PH, such as TGF-beta/BMPR2/STAT. Together, this complexity of the regulation of vascular fibrosis and stiffening in PH allows the exploration of numerous potential therapeutic interventions. This review discusses connections and turning points of several of these interventions in detail.
Collapse
Affiliation(s)
- Katharina Jandl
- Division of Pharmacology, Otto Loewi Research Center, Medical University Graz, Graz, Austria; Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Graz, Austria.
| | - Nemanja Radic
- Division of Physiology, Otto Loewi Research Center, Medical University Graz, Graz, Austria
| | - Katarina Zeder
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Gabor Kovacs
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Graz, Austria; Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Graz, Austria; Division of Physiology, Otto Loewi Research Center, Medical University Graz, Graz, Austria; Institute for Lung Health, Member of the German Lung Center (DZL), Giessen, Germany
| |
Collapse
|
20
|
Kotlyarov S. The Role of Smoking in the Mechanisms of Development of Chronic Obstructive Pulmonary Disease and Atherosclerosis. Int J Mol Sci 2023; 24:8725. [PMID: 37240069 PMCID: PMC10217854 DOI: 10.3390/ijms24108725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/05/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Tobacco smoking is a major cause of chronic obstructive pulmonary disease (COPD) and atherosclerotic cardiovascular disease (ASCVD). These diseases share common pathogenesis and significantly influence each other's clinical presentation and prognosis. There is increasing evidence that the mechanisms underlying the comorbidity of COPD and ASCVD are complex and multifactorial. Smoking-induced systemic inflammation, impaired endothelial function and oxidative stress may contribute to the development and progression of both diseases. The components present in tobacco smoke can have adverse effects on various cellular functions, including macrophages and endothelial cells. Smoking may also affect the innate immune system, impair apoptosis, and promote oxidative stress in the respiratory and vascular systems. The purpose of this review is to discuss the importance of smoking in the mechanisms underlying the comorbid course of COPD and ASCVD.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
21
|
Blackburn JB, Li NF, Bartlett NW, Richmond BW. An update in club cell biology and its potential relevance to chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol 2023; 324:L652-L665. [PMID: 36942863 PMCID: PMC10110710 DOI: 10.1152/ajplung.00192.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 03/10/2023] [Accepted: 03/16/2023] [Indexed: 03/23/2023] Open
Abstract
Club cells are found in human small airways where they play an important role in immune defense, xenobiotic metabolism, and repair after injury. Over the past few years, data from single-cell RNA sequencing (scRNA-seq) studies has generated new insights into club cell heterogeneity and function. In this review, we integrate findings from scRNA-seq experiments with earlier in vitro, in vivo, and microscopy studies and highlight the many ways club cells contribute to airway homeostasis. We then discuss evidence for loss of club cells or club cell products in the airways of patients with chronic obstructive pulmonary disease (COPD) and discuss potential mechanisms through which this might occur.
Collapse
Affiliation(s)
- Jessica B Blackburn
- Department of Veterans Affairs Medical Center, Nashville, Tennessee, United States
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
| | - Ngan Fung Li
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States
| | - Nathan W Bartlett
- Viral Immunology and Respiratory Disease Group, University of Newcastle, Callaghan, New South Wales, Australia
| | - Bradley W Richmond
- Department of Veterans Affairs Medical Center, Nashville, Tennessee, United States
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, United States
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States
| |
Collapse
|
22
|
Myronenko O, Foris V, Crnkovic S, Olschewski A, Rocha S, Nicolls MR, Olschewski H. Endotyping COPD: hypoxia-inducible factor-2 as a molecular "switch" between the vascular and airway phenotypes? Eur Respir Rev 2023; 32:220173. [PMID: 36631133 PMCID: PMC9879331 DOI: 10.1183/16000617.0173-2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 11/08/2022] [Indexed: 01/13/2023] Open
Abstract
COPD is a heterogeneous disease with multiple clinical phenotypes. COPD endotypes can be determined by different expressions of hypoxia-inducible factors (HIFs), which, in combination with individual susceptibility and environmental factors, may cause predominant airway or vascular changes in the lung. The pulmonary vascular phenotype is relatively rare among COPD patients and characterised by out-of-proportion pulmonary hypertension (PH) and low diffusing capacity of the lung for carbon monoxide, but only mild-to-moderate airway obstruction. Its histologic feature, severe remodelling of the small pulmonary arteries, can be mediated by HIF-2 overexpression in experimental PH models. HIF-2 is not only involved in the vascular remodelling but also in the parenchyma destruction. Endothelial cells from human emphysema lungs express reduced HIF-2α levels, and the deletion of pulmonary endothelial Hif-2α leads to emphysema in mice. This means that both upregulation and downregulation of HIF-2 have adverse effects and that HIF-2 may represent a molecular "switch" between the development of the vascular and airway phenotypes in COPD. The mechanisms of HIF-2 dysregulation in the lung are only partly understood. HIF-2 levels may be controlled by NAD(P)H oxidases via iron- and redox-dependent mechanisms. A better understanding of these mechanisms may lead to the development of new therapeutic targets.
Collapse
Affiliation(s)
- Oleh Myronenko
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Vasile Foris
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Slaven Crnkovic
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Division of Physiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, Graz, Austria
| | - Sonia Rocha
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular, and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Mark R Nicolls
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Horst Olschewski
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| |
Collapse
|
23
|
Lu YS, Chen YJ, Lee CL, Kuo FY, Tseng YH, Chen CH. Effects of photobiomodulation as an adjunctive treatment in chronic obstructive pulmonary disease: a narrative review. Lasers Med Sci 2023; 38:56. [PMID: 36707463 PMCID: PMC9883131 DOI: 10.1007/s10103-022-03661-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 11/29/2022] [Indexed: 01/29/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a disease characterized by chronic airway inflammation and remodeling and lung parenchymal inflammation and destruction, which result in many pulmonary and extrapulmonary manifestations. The anti-inflammatory effect of photobiomodulation (PBM) has been reported in previous studies. This review was conducted to evaluate the direct effect of PBM on lung inflammation in COPD. The other effects of PBM on modulation of peripheral and respiratory muscle metabolism and angiogenesis in lung tissues were also discussed. The databases of PubMed, Cochrane Library, and Google Scholar were searched to find the relevant studies. Keywords included PBM and related terms, COPD-related signs, and lung inflammation. A total of 12 articles were selected and reviewed in this study. Based on the present review, PBM is helpful in reducing lung inflammation through decreasing the inflammatory cytokines and chemokines at multiple levels and increasing anti-inflammatory cytokines. In addition, PBM also improves both peripheral and respiratory muscle metabolism and promote angiogenesis. This review demonstrated that PBM is a promising adjunctive treatment modality for COPD management which merits further validation.
Collapse
Affiliation(s)
- Yen-Sen Lu
- Department of General Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Yi-Jen Chen
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Physical Medicine and Rehabilitation, Kaohsiung Municipal Siaogang Hospital, Kaohsiung, Taiwan
- Department of Physical Medicine and Rehabilitation, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Ling Lee
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Physical Medicine and Rehabilitation, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan
| | - Fang-Yu Kuo
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Physical Medicine and Rehabilitation, Kaohsiung Municipal Siaogang Hospital, Kaohsiung, Taiwan
| | - Yu-Hsuan Tseng
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chia-Hsin Chen
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.
- Department of Physical Medicine and Rehabilitation, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
24
|
Zhang L, Jiang F, Xie Y, Mo Y, Zhang X, Liu C. Diabetic endothelial microangiopathy and pulmonary dysfunction. Front Endocrinol (Lausanne) 2023; 14:1073878. [PMID: 37025413 PMCID: PMC10071002 DOI: 10.3389/fendo.2023.1073878] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 02/17/2023] [Indexed: 04/08/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a widespread metabolic condition with a high global morbidity and mortality rate that affects the whole body. Their primary consequences are mostly caused by the macrovascular and microvascular bed degradation brought on by metabolic, hemodynamic, and inflammatory variables. However, research in recent years has expanded the target organ in T2DM to include the lung. Inflammatory lung diseases also impose a severe financial burden on global healthcare. T2DM has long been recognized as a significant comorbidity that influences the course of various respiratory disorders and their disease progress. The pathogenesis of the glycemic metabolic problem and endothelial microangiopathy of the respiratory disorders have garnered more attention lately, indicating that the two ailments have a shared history. This review aims to outline the connection between T2DM related endothelial cell dysfunction and concomitant respiratory diseases, including Coronavirus disease 2019 (COVID-19), asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF).
Collapse
Affiliation(s)
- Lanlan Zhang
- Department of Respiratory and Critical Care Medicine, Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Lanlan Zhang, ; Xin Zhang, ; Chuntao Liu,
| | - Faming Jiang
- Department of Respiratory and Critical Care Medicine, Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China
| | - Yingying Xie
- Department of Nephrology, Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yan Mo
- Department of Neurology Medicine, The Aviation Industry Corporation of China (AVIC) 363 Hospital, Chengdu, China
| | - Xin Zhang
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Lanlan Zhang, ; Xin Zhang, ; Chuntao Liu,
| | - Chuntao Liu
- Department of Respiratory and Critical Care Medicine, Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Lanlan Zhang, ; Xin Zhang, ; Chuntao Liu,
| |
Collapse
|
25
|
Chen Q, Lin J, Deng Z, Qian W. Exosomes derived from human umbilical cord mesenchymal stem cells protect against papain-induced emphysema by preventing apoptosis through activating VEGF-VEGFR2-mediated AKT and MEK/ERK pathways in rats. Regen Ther 2022; 21:216-224. [PMID: 36092502 PMCID: PMC9420880 DOI: 10.1016/j.reth.2022.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 06/25/2022] [Accepted: 07/07/2022] [Indexed: 12/04/2022] Open
Abstract
Introduction Chronic obstructive pulmonary disease (COPD) is a leading cause of high mortality and heavy burden in the world. Unfortunately, emphysema, as an important component of COPD, has no curative treatments currently. Recently, human umbilical cord mesenchymal stem cells-derived exosomes (hUCMSC-Ex) constitute a promising alternative approach for tissue regeneration and repair. However, the roles of hUCMSC-Ex in emphysema and its mechanism are largely unknown. Here, we investigated the effect and the action mechanism of hUCMSC-Ex in repairing emphysema induced by papain in rats. Methods SD rats were used to establish a papain-induced emphysema model and estimate the effect and mechanism of hUCMSC-Ex treatment. H&E staining and mean linear intercept (MLI) were used to evaluate the hUCMSC-Ex effect on emphysema. Western blotting, TUNEL and miRNA-seq were used to investigate the molecular mechanisms of hUCMSC-Ex treatment in models of papain-induced emphysema. Results Papain treatment led to typical emphysema, while hUCMSC-Ex reversed emphysematous changes effectively. Apoptosis of endothelial cells and other types of cells were observed in models, while hUCMSC-Ex effectively prevented their apoptosis. hUCMSC-Ex repressed active caspase-3, activated VEGF-VEGFR2-mediated AKT pathway and MEK/ERK pathway in emphysematous lungs. Notably, several miRNAs, such as hsa-miR-10a-5p and hsa-miR-146a-5p, were target related to the roles of hUCMSC-Ex in papain-induced emphysema through VEGF-VEGFR2-mediated AKT and MEK/ERK pathways. Conclusions hUCMSC-Ex effectively rescued the papain-induced emphysema injury through VEGF-VEGFR2-mediated AKT pathway and MEK/ERK pathway. Exosomes from human umbilical cord mesenchymal stem cells (hUCMSC-Ex) pro protect against papain-injured emphysema in rats. hUCMSC-Ex prevent lung cells apoptosis by activating VEGF-VEGFR2-mediated AKT and MEK/ERK pathways. Several miRNAs, such as hsa-miR-10a-5p, were target related to the roles of hUCMSC-Ex.
Collapse
Affiliation(s)
- Qin Chen
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Jiang Lin
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Zhaoqun Deng
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Wei Qian
- Department of Otolaryngology-Head and Neck Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
- Corresponding author. Department of Otolaryngology-Head and Neck Surgery, Affiliated People's Hospital of Jiangsu University, Dianli RD. 8, Zhenjiang, Jiangsu Province, 212002, China.
| |
Collapse
|
26
|
Celli B, Fabbri L, Criner G, Martinez FJ, Mannino D, Vogelmeier C, Montes de Oca M, Papi A, Sin DD, Han MK, Agusti A. Definition and Nomenclature of Chronic Obstructive Pulmonary Disease: Time for Its Revision. Am J Respir Crit Care Med 2022; 206:1317-1325. [PMID: 35914087 PMCID: PMC9746870 DOI: 10.1164/rccm.202204-0671pp] [Citation(s) in RCA: 142] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 08/01/2022] [Indexed: 12/24/2022] Open
Affiliation(s)
- Bartolome Celli
- Pulmonary Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Leonardo Fabbri
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Gerard Criner
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Fernando J Martinez
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York
| | - David Mannino
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Claus Vogelmeier
- Pulmonary and Critical Care Medicine, Department of Medicine, University Medical Center University of Marburg, German Center for Lung Research (DZL), Philipps University Marburg, Marburg, Germany
| | - Maria Montes de Oca
- Hospital Universitario de Caracas, Universidad Central de Venezuela and Centro Médico de Caracas, Caracas, Venezuela
| | - Alberto Papi
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Don D Sin
- Division of Respiratory Medicine, Centre for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - MeiLan K Han
- University of Michigan Health System, Ann Arbor, Michigan; and
| | - Alvar Agusti
- Cátedra Salud Respiratoria, Universitat de Barcelona; Respiratory Institute, Hospital Clinic, Barcelona; IDIBAPS, CIBERES, Barcelona, Spain
| |
Collapse
|
27
|
Jiang T, Hu W, Zhang S, Ren C, Lin S, Zhou Z, Wu H, Yin J, Tan L. Fibroblast growth factor 10 attenuates chronic obstructive pulmonary disease by protecting against glycocalyx impairment and endothelial apoptosis. Respir Res 2022; 23:269. [PMID: 36183124 PMCID: PMC9526324 DOI: 10.1186/s12931-022-02193-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 09/22/2022] [Indexed: 11/23/2022] Open
Abstract
Background The defects and imbalance in lung repair and structural maintenance contribute to the pathogenesis of chronic obstructive pulmonary diseases (COPD), yet the molecular mechanisms that regulate lung repair process are so far incompletely understood. We hypothesized that cigarette smoking causes glycocalyx impairment and endothelial apoptosis in COPD, which could be repaired by the stimulation of fibroblast growth factor 10 (FGF10)/FGF receptor 1 (FGFR1) signaling. Methods We used immunostaining (immunohistochemical [IHC] and immunofluorescence [IF]) and enzyme-linked immunosorbent assay (ELISA) to detect the levels of glycocalyx components and endothelial apoptosis in animal models and in patients with COPD. We used the murine emphysema model and in vitro studies to determine the protective and reparative role of FGF10/FGFR1. Results Exposure to cigarette smoke caused endothelial glycocalyx impairment and emphysematous changes in murine models and human specimens. Pretreatment of FGF10 attenuated the development of emphysema and the shedding of glycocalyx components induced by CSE in vivo. However, FGF10 did not attenuate the emphysema induced by endothelial-specific killing peptide CGSPGWVRC-GG-D(KLAKLAK)2. Mechanistically, FGF10 alleviated smoke-induced endothelial apoptosis and glycocalyx repair through FGFR1/ERK/SOX9/HS6ST1 signaling in vitro. FGF10 was shown to repair pulmonary glycocalyx injury and endothelial apoptosis, and attenuate smoke-induced COPD through FGFR1 signaling. Conclusions Our results suggest that FGF10 may serve as a potential therapeutic strategy against COPD via endothelial repair and glycocalyx reconstitution. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-02193-5. It is the first time to prove the confirm the endothelial glycocalyx impairment in COPD. FGF10 attenuates the development of emphysema and the shedding of glycocalyx induced by CSE in vivo. FGF10 alleviates smoke-induced endothelial apoptosis and glycocalyx repair through FGFR1/ERK/SOX9/HS6ST1 signaling.
Collapse
Affiliation(s)
- Tian Jiang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai, 200032, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Key Laboratory of Lung Inflammation and Injury, Shanghai, 200032, China
| | - Weiping Hu
- Department of Critical Care and Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Shaoyuan Zhang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai, 200032, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Key Laboratory of Lung Inflammation and Injury, Shanghai, 200032, China
| | - Changhao Ren
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai, 200032, China
| | - Siyun Lin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai, 200032, China
| | - Zhenyu Zhou
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hao Wu
- Department of Clinical Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jun Yin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai, 200032, China. .,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China. .,Key Laboratory of Lung Inflammation and Injury, Shanghai, 200032, China.
| | - Lijie Tan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai, 200032, China. .,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China. .,Key Laboratory of Lung Inflammation and Injury, Shanghai, 200032, China.
| |
Collapse
|
28
|
Huang X, Yin W, Shen M, Wang X, Ren T, Wang L, Liu M, Guo Y. Contributions of Emphysema and Functional Small Airway Disease on Intrapulmonary Vascular Volume in COPD. Int J Chron Obstruct Pulmon Dis 2022; 17:1951-1961. [PMID: 36045693 PMCID: PMC9423118 DOI: 10.2147/copd.s368974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 08/16/2022] [Indexed: 11/23/2022] Open
Abstract
Background Previous studies have demonstrated that there is a certain correlation between emphysema and changes in pulmonary small blood vessels in patients with chronic obstructive pulmonary disease (COPD), but most of them were limited to the investigation of the inspiratory phase. The emphysema indicators need to be further optimized. Based on the parametric response mapping (PRM) method, this study aimed to investigate the effect of emphysema and functional small airway disease on intrapulmonary vascular volume (IPVV). Methods This retrospective study enrolled 63 healthy subjects and 47 COPD patients, who underwent both inspiratory and expiratory CT scans of the chest and pulmonary function tests (PFTs). Inspiratory and expiratory IPVV were measured by using an automatic pulmonary vessels integration segmentation approach, the ratio of emphysema volume (Emph%), functional small airway disease volume (fsAD%), and normal areas volume (Normal%) were quantified by the PRM method for biphasic CT scans. The participants were grouped according to PFTs. Analysis of variance (ANOVA) and Kruskal–Wallis H-test were used to analyze the differences in indicators between different groups. Then, Spearman’s rank correlation coefficients were used to analyze the correlation between Emph%, fsAD%, Normal%, PFTs, and IPVV. Finally, multiple linear regression was applied to analyze the effects of Emph% and fsAD% on IPVV. Results Differences were found in age, body mass index (BMI), smoking index, FEV1%, FEV1/forced vital capacity (FVC), expiratory IPVV, IPVV relative value, IPVV difference value, Emph%, fsAD%, and Normal% between the groups (P<0.05). A strong correlation was established between the outcomes of PFTs and quantitative CT indexes. Finally, the effect of Emph% was more significant than that of fsAD% on expiratory IPVV, IPVV difference value, and IPVV relative value. Conclusion IPVV may have a potential value in assessing COPD severity and is significantly affected by emphysema.
Collapse
Affiliation(s)
- Xiaoqi Huang
- Department of Radiology, Yan'an University Affiliated Hospital, Yan'an, People's Republic of China
| | - Weiling Yin
- Department of Radiology, Yan'an University Affiliated Hospital, Yan'an, People's Republic of China
| | - Min Shen
- Department of Radiology, Yan'an University Affiliated Hospital, Yan'an, People's Republic of China
| | - Xionghui Wang
- Department of Radiology, Yan'an University Affiliated Hospital, Yan'an, People's Republic of China
| | - Tao Ren
- Department of Radiology, Yan'an University Affiliated Hospital, Yan'an, People's Republic of China
| | - Lei Wang
- Department of Radiology, Yan'an University Affiliated Hospital, Yan'an, People's Republic of China
| | - Min Liu
- Department of Radiology, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Youmin Guo
- Department of Radiology, Yan'an University Affiliated Hospital, Yan'an, People's Republic of China
| |
Collapse
|
29
|
MacLean MR, Fanburg B, Hill N, Lazarus HM, Pack TF, Palacios M, Penumatsa KC, Wring SA. Serotonin and Pulmonary Hypertension; Sex and Drugs and ROCK and Rho. Compr Physiol 2022; 12:4103-4118. [PMID: 36036567 DOI: 10.1002/cphy.c220004] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Serotonin is often referred to as a "happy hormone" as it maintains good mood, well-being, and happiness. It is involved in communication between nerve cells and plays a role in sleeping and digestion. However, too much serotonin can have pathogenic effects and serotonin synthesis is elevated in pulmonary artery endothelial cells from patients with pulmonary arterial hypertension (PAH). PAH is characterized by elevated pulmonary pressures, right ventricular failure, inflammation, and pulmonary vascular remodeling; serotonin has been shown to be associated with these pathologies. The rate-limiting enzyme in the synthesis of serotonin in the periphery of the body is tryptophan hydroxylase 1 (TPH1). TPH1 expression and serotonin synthesis are elevated in pulmonary artery endothelial cells in patients with PAH. The serotonin synthesized in the pulmonary arterial endothelium can act on the adjacent pulmonary arterial smooth muscle cells (PASMCs), adventitial macrophages, and fibroblasts, in a paracrine fashion. In humans, serotonin enters PASMCs cells via the serotonin transporter (SERT) and it can cooperate with the 5-HT1B receptor on the plasma membrane; this activates both contractile and proliferative signaling pathways. The "serotonin hypothesis of pulmonary hypertension" arose when serotonin was associated with PAH induced by diet pills such as fenfluramine, aminorex, and chlorphentermine; these act as indirect serotonergic agonists causing the release of serotonin from platelets and cells through the SERT. Here the role of serotonin in PAH is reviewed. Targeting serotonin synthesis or signaling is a promising novel alternative approach which may lead to novel therapies for PAH. © 2022 American Physiological Society. Compr Physiol 12: 1-16, 2022.
Collapse
Affiliation(s)
- Margaret R MacLean
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland
| | - Barry Fanburg
- Pulmonary, Critical Care and Sleep Division, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | - Nicolas Hill
- Pulmonary, Critical Care and Sleep Division, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | | | | | | | - Krishna C Penumatsa
- Pulmonary, Critical Care and Sleep Division, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | | |
Collapse
|
30
|
Della Rocca Y, Fonticoli L, Rajan TS, Trubiani O, Caputi S, Diomede F, Pizzicannella J, Marconi GD. Hypoxia: molecular pathophysiological mechanisms in human diseases. J Physiol Biochem 2022; 78:739-752. [PMID: 35870078 PMCID: PMC9684243 DOI: 10.1007/s13105-022-00912-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 07/14/2022] [Indexed: 12/01/2022]
Abstract
Abstract
Hypoxia, a low O2 tension, is a fundamental feature that occurs in physiological events as well as pathophysiological conditions, especially mentioned for its role in the mechanism of angiogenesis, glucose metabolism, and cell proliferation/survival. The hypoxic state through the activation of specific mechanisms is an aggravating circumstance commonly noticed in multiple sclerosis, cancer, heart disease, kidney disease, liver disease, lung disease, and in inflammatory bowel disease. On the other hand, hypoxia could play a key role in tissue regeneration and repair of damaged tissues, especially by acting on specific tissue stem cells, but their features may result as a disadvantage when it is concerned for neoplastic stem cells. Furthermore, hypoxia could also have a potential role in tissue engineering and regenerative medicine due to its capacity to improve the performance of biomaterials. The current review aims to highlight the hypoxic molecular mechanisms reported in different pathological conditions to provide an overview of hypoxia as a therapeutic agent in regenerative and molecular therapy.
Graphical abstract
Collapse
Affiliation(s)
- Ylenia Della Rocca
- Department of Innovative Technologies in Medicine & Dentistry, University "G. d'Annunzio" Chieti-Pescara, Chieti, Italy
| | - Luigia Fonticoli
- Department of Innovative Technologies in Medicine & Dentistry, University "G. d'Annunzio" Chieti-Pescara, Chieti, Italy
| | | | - Oriana Trubiani
- Department of Innovative Technologies in Medicine & Dentistry, University "G. d'Annunzio" Chieti-Pescara, Chieti, Italy
| | - Sergio Caputi
- Department of Innovative Technologies in Medicine & Dentistry, University "G. d'Annunzio" Chieti-Pescara, Chieti, Italy
| | - Francesca Diomede
- Department of Innovative Technologies in Medicine & Dentistry, University "G. d'Annunzio" Chieti-Pescara, Chieti, Italy.
| | - Jacopo Pizzicannella
- Cardiology Intensive Care Unit, "Ss. Annunziata" Hospital, ASL02 Lanciano-Vasto-Chieti, Chieti, Italy
| | - Guya Diletta Marconi
- Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio" Chieti-Pescara, Chieti, Italy
| |
Collapse
|
31
|
Rehman S, Ullah H, Nagarajan JS, Sukaina M, Ghafoor B, Shaik Masthan S, Shahrukh S, Min Allah H, Qureshi MH. Pneumothorax as a Complication of Bevacizumab-Containing Chemotherapy: A Systematic Review of Case Reports. Cureus 2022; 14:e27338. [PMID: 36043019 PMCID: PMC9414787 DOI: 10.7759/cureus.27338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2022] [Indexed: 12/21/2022] Open
Abstract
Bevacizumab is a monoclonal anti-vascular endothelial growth factor (VEGF) antibody that binds to and makes all of the VEGF isoforms inactive, and thus prevents angiogenesis, development, and the spread of the tumor. The most reported side effects after administering bevacizumab include bleeding, high blood pressure, heart failure, proteinuria, thrombosis, and gastrointestinal perforation. Pneumothorax has rarely been reported as a complication of bevacizumab, but with an unclear mechanism. This article aims to explore the occurrence of pneumothorax as a side effect after using bevacizumab through a systematic review of current case reports published on the topic. A literature search was conducted using PubMed, Google Scholar, ScienceDirect, and Directory of Open Access through the utilization of appropriate keywords, and case reports were selected based on predefined inclusion and exclusion criteria. Our results encompass five case reports that were further evaluated for demographic, clinical, and treatment parameters. This systematic review concludes that pneumothorax can occur after bevacizumab-containing chemotherapy although this side effect is relatively rare. Awareness regarding this possible side effect can assist clinicians during their practice in considering pneumothorax as a possible differential diagnosis when encountering patients presenting with pulmonary symptoms after starting bevacizumab-containing chemotherapy; hence, timely diagnosis and treatment can save a life.
Collapse
Affiliation(s)
- Shafi Rehman
- Internal Medicine, Lady Reading Hospital, Peshawar, PAK
| | - Hameed Ullah
- Internal Medicine, Hayatabad Medical Complex, Peshawar, Peshawar, PAK
| | | | | | - Bushra Ghafoor
- Research, University of Texas Southwestern Medical Center, Dallas, USA
- Graduate Medical Education, Dera Ghazi Khan Medical College, Dera Ghazi Khan, PAK
- Internal Medicine, Nishtar Medical University, Multan, PAK
| | | | - Shazmah Shahrukh
- Internal Medicine, Dow University of Health Sciences, Civil Hospital Karachi, Karachi, PAK
| | - Hassan Min Allah
- Internal Medicine, Multan Medical and Dental College, Multan, PAK
| | | |
Collapse
|
32
|
Tsikis ST, Fligor SC, Hirsch TI, Pan A, Yu LJ, Kishikawa H, Joiner MM, Mitchell PD, Puder M. Lipopolysaccharide-induced murine lung injury results in long-term pulmonary changes and downregulation of angiogenic pathways. Sci Rep 2022; 12:10245. [PMID: 35715592 PMCID: PMC9205148 DOI: 10.1038/s41598-022-14618-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 06/09/2022] [Indexed: 11/18/2022] Open
Abstract
Acute respiratory distress syndrome is the most severe form of acute lung injury (ALI) and is associated with significant mortality. Lipopolysaccharide (LPS)-induced injury is a valuable murine model of ALI but there is a paucity of data on lung regeneration and the role of angiogenic signaling involving vascular endothelial growth factor (VEGF). Eight-week-old male C57BL/6J mice were randomized to receive intratracheal instillation of either LPS or isovolumetric phosphate buffered saline as a vehicle control. Mice were observed at a single follow-up time-point that was either short-term (24 h or 4 days) or long-term (7 days or 4 weeks). On pulmonary function testing, LPS-treated mice had increased compliance at 4 weeks post-instillation, which correlated with decreased vascularization and with time-dependent, progressive decrease in alveolarization. Treadmill exercise tolerance testing demonstrated impaired performance at 24 h, 4 days and 4 weeks following LPS exposure. On lung protein analysis, LPS instillation decreased VEGF expression at up to 4 weeks, and decreased activation of its key receptor, VEGFR2 at 7 days and 4 weeks post-instillation. Together, these data provide insight on long-term pulmonary functional outcomes 4 weeks after ALI and identify angiogenic proteins as possible therapeutic targets following lung injury.
Collapse
Affiliation(s)
- S T Tsikis
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - S C Fligor
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - T I Hirsch
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - A Pan
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - L J Yu
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - H Kishikawa
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - M M Joiner
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA
| | - P D Mitchell
- Institutional Centers for Clinical and Translational Research, Boston Children's Hospital, Boston, MA, 02115, USA
| | - M Puder
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Ave, Fegan 3, Boston, MA, 02115, USA.
| |
Collapse
|
33
|
Wang N, Guo Z, Gong X, Kang S, Cui Z, Yuan Y. A Nomogram for Predicting the Risk of Pulmonary Hypertension for Patients with Chronic Obstructive Pulmonary Disease. Int J Gen Med 2022; 15:5751-5762. [PMID: 35770051 PMCID: PMC9234502 DOI: 10.2147/ijgm.s363035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 06/14/2022] [Indexed: 12/29/2022] Open
Abstract
Background Pulmonary hypertension (PH) is a life-threatening complication of chronic obstructive pulmonary disease (COPD). Timely diagnosis of PH in COPD patients is vital to achieve proper treatment; however, there is no algorithm to identify those at high risk. We aimed to develop a predictive model for PH in patients with COPD that provides individualized risk estimates. Methods A total of 527 patients with COPD who were admitted to our hospital between May 2019 and December 2020 were retrospectively enrolled in this study. Using echocardiographic results as a standard, patients were stratified into a moderate- or high-PH probability group and a low-PH probability group. They were randomly grouped into either the training set (n = 368 patients) or validation set (n = 159 patients) in a ratio of 7:3. We utilized the least absolute shrinkage and selection operator (LASSO) regression model to select the feature variables. The characteristic variables selected in the LASSO regression were analyzed using multivariable logistic regression to construct the predictive model. The predictive model was displayed using a nomogram. We used the receiver operating characteristic curve, calibration curve, and clinical decision curve analysis (DCA) to evaluate model performance, and internal validation was assessed. Results The predictive factors included in the prediction model were Global Initiative for Chronic Obstructive Lung Disease (GOLD) stage, emphysema, PaCO2, NT-pro-BNP, red blood cell (RBC) distribution width-standard deviation (RDW-SD), and neutrophil/lymphocyte ratio (NLR). The predictive model yielded an area under the curve (AUC) of 0.770 (95% confidence interval [CI], 0.719–0.820); in the internal validation, the AUC was 0.741 (95% CI, 0.659–0.823). The predictive model was well calibrated, and the DCA showed that the proposed nomogram had strong clinical applicability. Conclusion This study showed that a simple nomogram could be used to calculate the risk of PH in patients with COPD which can be useful for the individualized clinical management of COPD patients who may be occur with PH. Further studies need to be confirmed by larger sample sizes and validated in the stable COPD population.
Collapse
Affiliation(s)
- Ning Wang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
- Department of Respiratory and Critical Care Medicine, Hengshui People’s Hospital, Hengshui, People’s Republic of China
| | - Zhenjiang Guo
- Department of Gastrointestinal Surgery, Hengshui People’s Hospital, Hengshui, People’s Republic of China
| | - Xiaowei Gong
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Shiwei Kang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Zhaobo Cui
- Department of Respiratory and Critical Care Medicine, Hengshui People’s Hospital, Hengshui, People’s Republic of China
| | - Yadong Yuan
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, People’s Republic of China
- Correspondence: Yadong Yuan, Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, No. 215 Heping West Road, Shijiazhuang, Hebei, 050000, People’s Republic of China, Tel/Fax +86-311-66003989, Email
| |
Collapse
|
34
|
Angiogenesis, Lymphangiogenesis, and Inflammation in Chronic Obstructive Pulmonary Disease (COPD): Few Certainties and Many Outstanding Questions. Cells 2022; 11:cells11101720. [PMID: 35626756 PMCID: PMC9139415 DOI: 10.3390/cells11101720] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/20/2022] [Accepted: 05/21/2022] [Indexed: 02/07/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by chronic inflammation, predominantly affecting the lung parenchyma and peripheral airways, that results in progressive and irreversible airflow obstruction. COPD development is promoted by persistent pulmonary inflammation in response to several stimuli (e.g., cigarette smoke, bacterial and viral infections, air pollution, etc.). Angiogenesis, the formation of new blood vessels, and lymphangiogenesis, the formation of new lymphatic vessels, are features of airway inflammation in COPD. There is compelling evidence that effector cells of inflammation (lung-resident macrophages and mast cells and infiltrating neutrophils, eosinophils, basophils, lymphocytes, etc.) are major sources of a vast array of angiogenic (e.g., vascular endothelial growth factor-A (VEGF-A), angiopoietins) and/or lymphangiogenic factors (VEGF-C, -D). Further, structural cells, including bronchial and alveolar epithelial cells, endothelial cells, fibroblasts/myofibroblasts, and airway smooth muscle cells, can contribute to inflammation and angiogenesis in COPD. Although there is evidence that alterations of angiogenesis and, to a lesser extent, lymphangiogenesis, are associated with COPD, there are still many unanswered questions.
Collapse
|
35
|
Nitta NA, Sato T, Komura M, Yoshikawa H, Suzuki Y, Mitsui A, Kuwasaki E, Takahashi F, Kodama Y, Seyama K, Takahashi K. Exposure to the heated tobacco product IQOS generates apoptosis-mediated pulmonary emphysema in murine lungs. Am J Physiol Lung Cell Mol Physiol 2022; 322:L699-L711. [PMID: 35380471 DOI: 10.1152/ajplung.00215.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 03/21/2022] [Accepted: 03/31/2022] [Indexed: 11/22/2022] Open
Abstract
Pulmonary emphysema is predominantly caused by chronic exposure to cigarette smoke (CS). Novel tobacco substitutes, such as heated tobacco products (HTPs), have emerged as healthier alternatives to cigarettes. IQOS, the most popular HTP in Japan, is advertised as harmless compared with conventional cigarettes. Although some studies have reported its toxicity, few in vivo studies have been conducted. Here, 12-wk-old C57BL6/J male mice were divided into three groups and exposed to air (as control), IQOS aerosol, or CS for 6 mo. After exposure, the weight gain was significantly suppressed in the IQOS and CS groups compared with the control (-4.93 g; IQOS vs. air and -5.504 g; CS vs. air). The serum cotinine level was significantly higher in the IQOS group than in the control group. The neutrophils and lymphocyte count increased in the bronchoalveolar lavage fluid of the IQOS and CS groups compared with those in the control group. Chronic IQOS exposure induced pulmonary emphysema similar to that observed in the CS group. Furthermore, expression levels of the genes involved in the apoptosis-related pathways were significantly upregulated in the lungs of the IQOS-exposed mice. Cytochrome c, cleaved caspase-3, and cleaved poly (ADP-ribose) polymerase-1 were overexpressed in the IQOS group compared with the control. Single-stranded DNA and TdT-mediated dUTP nick-end labeling-positive alveolar septal cell count significantly increased in the IQOS group compared with the control. In conclusion, chronic exposure to IQOS aerosol induces pulmonary emphysema predominantly via apoptosis-related pathways. This suggests that HTPs are not completely safe tobacco products.
Collapse
Affiliation(s)
- Naoko Arano Nitta
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tadashi Sato
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Moegi Komura
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hitomi Yoshikawa
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yohei Suzuki
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Aki Mitsui
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Eriko Kuwasaki
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Fumiyuki Takahashi
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuzo Kodama
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kuniaki Seyama
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kazuhisa Takahashi
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
36
|
Abstract
Pulmonary hypertension (PH) describes heterogeneous population of patients with a mean pulmonary arterial pressure >20 mm Hg. Rarely, PH presents as a primary disorder but is more commonly part of a complex phenotype associated with comorbidities. Regardless of the cause, PH reduces life expectancy and impacts quality of life. The current clinical classification divides PH into 1 of 5 diagnostic groups to assign treatment. There are currently no pharmacological cures for any form of PH. Animal models are essential to help decipher the molecular mechanisms underlying the disease, to assign genotype-phenotype relationships to help identify new therapeutic targets, and for clinical translation to assess the mechanism of action and putative efficacy of new therapies. However, limitations inherent of all animal models of disease limit the ability of any single model to fully recapitulate complex human disease. Within the PH community, we are often critical of animal models due to the perceived low success upon clinical translation of new drugs. In this review, we describe the characteristics, advantages, and disadvantages of existing animal models developed to gain insight into the molecular and pathological mechanisms and test new therapeutics, focusing on adult forms of PH from groups 1 to 3. We also discuss areas of improvement for animal models with approaches combining several hits to better reflect the clinical situation and elevate their translational value.
Collapse
Affiliation(s)
- Olivier Boucherat
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC, Canada
- Department of Medicine, Université Laval, Québec, QC, Canada
| | - Vineet Agrawal
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Allan Lawrie
- Dept of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK & Insigneo institute for in silico medicine, Sheffield, UK
| | - Sebastien Bonnet
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC, Canada
- Department of Medicine, Université Laval, Québec, QC, Canada
| |
Collapse
|
37
|
Cheng Y, Gu W, Zhang G, Guo X. Notch1 activation of Jagged1 contributes to differentiation of mesenchymal stem cells into endothelial cells under cigarette smoke extract exposure. BMC Pulm Med 2022; 22:139. [PMID: 35410206 PMCID: PMC9004089 DOI: 10.1186/s12890-022-01913-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 03/23/2022] [Indexed: 11/30/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) have shown therapeutic potential for engraftment to, differentiation into, endothelial cells (ECs). However, low-efficiency yields hinder their use as ECs for therapeutic vascularization. Methods The Notch1 signaling pathway is key to optimal pulmonary development. Recent evidence has shown that this pathway participated in angiogenesis. Herein, we found that in MSCs, Jagged1 was a target for Notch 1, resulting in a positive feedback loop that propagated a wave of ECs differentiation. Results In vitro, Jagged1 was found to be activated by Notch1 in MSCs, resulting in the RBP-Jκ-dependent expression of Jagged1 mRNA, a response that was blocked by Notch1 inhibition. Notch1 promoted the formation of cord-like structures on Matrigel. However, cigarette smoke extract inhibited this process, compared to that in control groups. Moreover, Notch1-overexpressing cells upregulated the expressing of HIF-1α gene. The HIF-1α was an angiogenic factor that clustered with Notch1, underscoring the critical role of Notch1 pathway in vessel assembly. Interestingly, this was abrogated by incubation with Notch1 shRNA. Conclusions Notch signaling pathway promotes differentiation of MSCs in to ECs. It also regulates angiogenesis and transcription of specific markers on ECs. These results provide a mechanism that regulates differentiation of MSCs into ECs phenotypes. Supplementary Information The online version contains supplementary material available at 10.1186/s12890-022-01913-3.
Collapse
Affiliation(s)
- Yi Cheng
- Department of Respiratory Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 KongJiang Road, Shanghai, 200092, China
| | - Wen Gu
- Department of Respiratory Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 KongJiang Road, Shanghai, 200092, China
| | - Guorui Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuejun Guo
- Department of Respiratory Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 KongJiang Road, Shanghai, 200092, China.
| |
Collapse
|
38
|
Yu H, Lin Y, Zhong Y, Guo X, Lin Y, Yang S, Liu J, Xie X, Sun Y, Wang D, Li B, Ran P, Dai J. Impaired AT2 to AT1 cell transition in PM2.5-induced mouse model of chronic obstructive pulmonary disease. Respir Res 2022; 23:70. [PMID: 35337337 PMCID: PMC8957194 DOI: 10.1186/s12931-022-01996-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/16/2022] [Indexed: 12/12/2022] Open
Abstract
Background Particular matter 2.5 (PM2.5) is one of the most important air pollutant, and it is positively associated with the development of chronic obstructive pulmonary disease (COPD). However, the precise underlying mechanisms through which PM2.5 promotes the development of COPD remains largely unknown.
Methods Mouse alveolar destruction were determined by histological analysis of lung tissues and lung function test. Alveolar type II cells (AT2) to alveolar type I cells (AT1) transition in PM2.5-induced COPD mouse model was confirmed via immunofluorescence staining and qPCR analysis. The differentially expressed genes in PM2.5-induced COPD mouse model were identified by RNA-sequencing of alveolar epithelial organoids and generated by bioinformatics analysis. Results In this study, we found that 6 months exposure of PM2.5 induced a significantly decreased pulmonary compliance and resulted in pulmonary emphysema in mice. We showed that PM2.5 exposure significantly reduced the AT2 to AT1 cell transition in vitro and in vivo. In addition, we found a reduced expression of the intermediate AT2-AT1 cell process marker claudin 4 (CLDN4) at day 4 of differentiation in mouse alveolar organoids treated with PM2.5, suggesting that PM2.5 exposure inhibited AT2 cells from entering the transdifferentiation process. RNA-sequencing of mouse alveolar organoids showed that several key signaling pathways that involved in the AT2 to AT1 cell transition were significantly altered including the Wnt signaling, MAPK signaling and signaling pathways regulating pluripotency of stem cells following PM2.5 exposure. Conclusions In summary, these data demonstrate a critical role of AT2 to AT1 cell transition in PM2.5-induced COPD mouse model and reveal the signaling pathways that potentially regulate AT2 to AT1 cell transition during this process. Our findings therefore advance the current knowledge of PM2.5-induced COPD and may lead to a novel therapeutic strategy to treat this disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-01996-w.
Collapse
Affiliation(s)
- Hongjiao Yu
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, People's Republic of China
| | - Yingnan Lin
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, People's Republic of China
| | - Yue Zhong
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, People's Republic of China
| | - Xiaolan Guo
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, People's Republic of China
| | - Yuyin Lin
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, People's Republic of China
| | - Siqi Yang
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, People's Republic of China
| | - Jinglin Liu
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, People's Republic of China
| | - Xinran Xie
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, People's Republic of China
| | - Yaowei Sun
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, People's Republic of China
| | - Dong Wang
- State Key Lab of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People's Republic of China
| | - Bing Li
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, People's Republic of China
| | - Pixin Ran
- State Key Lab of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People's Republic of China.
| | - Jianwei Dai
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, People's Republic of China. .,State Key Lab of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People's Republic of China. .,The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Qingyuan, 511500, People's Republic of China.
| |
Collapse
|
39
|
Sauler M, McDonough JE, Adams TS, Kothapalli N, Barnthaler T, Werder RB, Schupp JC, Nouws J, Robertson MJ, Coarfa C, Yang T, Chioccioli M, Omote N, Cosme C, Poli S, Ayaub EA, Chu SG, Jensen KH, Gomez JL, Britto CJ, Raredon MSB, Niklason LE, Wilson AA, Timshel PN, Kaminski N, Rosas IO. Characterization of the COPD alveolar niche using single-cell RNA sequencing. Nat Commun 2022; 13:494. [PMID: 35078977 PMCID: PMC8789871 DOI: 10.1038/s41467-022-28062-9] [Citation(s) in RCA: 86] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 12/14/2021] [Indexed: 12/16/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a leading cause of death worldwide, however our understanding of cell specific mechanisms underlying COPD pathobiology remains incomplete. Here, we analyze single-cell RNA sequencing profiles of explanted lung tissue from subjects with advanced COPD or control lungs, and we validate findings using single-cell RNA sequencing of lungs from mice exposed to 10 months of cigarette smoke, RNA sequencing of isolated human alveolar epithelial cells, functional in vitro models, and in situ hybridization and immunostaining of human lung tissue samples. We identify a subpopulation of alveolar epithelial type II cells with transcriptional evidence for aberrant cellular metabolism and reduced cellular stress tolerance in COPD. Using transcriptomic network analyses, we predict capillary endothelial cells are inflamed in COPD, particularly through increased CXCL-motif chemokine signaling. Finally, we detect a high-metallothionein expressing macrophage subpopulation enriched in advanced COPD. Collectively, these findings highlight cell-specific mechanisms involved in the pathobiology of advanced COPD.
Collapse
Affiliation(s)
- Maor Sauler
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA.
| | - John E McDonough
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA.
| | - Taylor S Adams
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Neeharika Kothapalli
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Thomas Barnthaler
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Rhiannon B Werder
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA, 02118, USA
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
- QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Jonas C Schupp
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Respiratory Medicine, Hannover Medical School and Biomedical Research in End-stage and Obstructive Lung Disease Hannover, German Lung Research Center (DZL), Hannover, Germany
| | - Jessica Nouws
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Matthew J Robertson
- Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Cristian Coarfa
- Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Tao Yang
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Maurizio Chioccioli
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Norihito Omote
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Carlos Cosme
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Sergio Poli
- Department of Internal Medicine, Mount Sinai Medical Center, Miami, FL, USA
| | - Ehab A Ayaub
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sarah G Chu
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Jose L Gomez
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Clemente J Britto
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Micha Sam B Raredon
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, USA
| | - Laura E Niklason
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Andrew A Wilson
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA, 02118, USA
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | | | - Naftali Kaminski
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Ivan O Rosas
- Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
40
|
Zhang X, Jin H, Huang X, Chaurasiya B, Dong D, Shanley TP, Zhao YY. Robust genome editing in adult vascular endothelium by nanoparticle delivery of CRISPR-Cas9 plasmid DNA. Cell Rep 2022; 38:110196. [PMID: 34986352 PMCID: PMC8769807 DOI: 10.1016/j.celrep.2021.110196] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 07/16/2021] [Accepted: 12/10/2021] [Indexed: 12/25/2022] Open
Abstract
Vascular endothelium plays a crucial role in vascular homeostasis and tissue fluid balance. To target endothelium for robust genome editing, we developed poly(ethylene glycol) methyl ether-block-poly(lactide-co-glycolide) (PEG-b-PLGA) copolymer-based nanoparticle formulated with polyethyleneimine. A single i.v. administration of mixture of nanoparticles and plasmid DNA expressing Cas9 controlled by CDH5 promoter and guide RNA (U6 promoter) induced highly efficient genome editing in endothelial cells (ECs) of the vasculatures, including lung, heart, aorta, and peripheral vessels in adult mice. Western blotting and immunofluorescent staining demonstrated an ∼80% decrease of protein expression selectively in ECs, resulting in a phenotype similar to that of genetic knockout mice. Nanoparticle delivery of plasmid DNA could induce genome editing of two genes or genome editing and transgene expression in ECs simultaneously. Thus, nanoparticle delivery of plasmid DNA is a powerful tool to rapidly and efficiently alter expression of gene(s) in ECs for cardiovascular research and potential gene therapy.
Collapse
Affiliation(s)
- Xianming Zhang
- Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA; Department of Pediatrics, Division of Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Hua Jin
- Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA; Department of Pediatrics, Division of Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Xiaojia Huang
- Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA; Department of Pediatrics, Division of Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Birendra Chaurasiya
- Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA; Department of Pediatrics, Division of Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Daoyin Dong
- Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA; Department of Pediatrics, Division of Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Thomas P Shanley
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - You-Yang Zhao
- Program for Lung and Vascular Biology, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA; Department of Pediatrics, Division of Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
41
|
Ghio AJ, Pavlisko EN, Roggli VL, Todd NW, Sangani RG. Cigarette Smoke Particle-Induced Lung Injury and Iron Homeostasis. Int J Chron Obstruct Pulmon Dis 2022; 17:117-140. [PMID: 35046648 PMCID: PMC8763205 DOI: 10.2147/copd.s337354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 12/06/2021] [Indexed: 11/23/2022] Open
Abstract
It is proposed that the mechanistic basis for non-neoplastic lung injury with cigarette smoking is a disruption of iron homeostasis in cells after exposure to cigarette smoke particle (CSP). Following the complexation and sequestration of intracellular iron by CSP, the host response (eg, inflammation, mucus production, and fibrosis) attempts to reverse a functional metal deficiency. Clinical manifestations of this response can present as respiratory bronchiolitis, desquamative interstitial pneumonitis, pulmonary Langerhans’ cell histiocytosis, asthma, pulmonary hypertension, chronic bronchitis, and pulmonary fibrosis. If the response is unsuccessful, the functional deficiency of iron progresses to irreversible cell death evident in emphysema and bronchiectasis. The subsequent clinical and pathological presentation is a continuum of lung injuries, which overlap and coexist with one another. Designating these non-neoplastic lung injuries after smoking as distinct disease processes fails to recognize shared relationships to each other and ultimately to CSP, as well as the common mechanistic pathway (ie, disruption of iron homeostasis).
Collapse
Affiliation(s)
- Andrew J Ghio
- Human Studies Facility, US Environmental Protection Agency, Chapel Hill, NC, 27514, USA
- Correspondence: Andrew J Ghio Human Studies Facility, US Environmental Protection Agency, 104 Mason Farm Road, Chapel Hill, NC, USA Email
| | | | | | - Nevins W Todd
- Department of Medicine, University of Maryland, Baltimore, MD, 21201, USA
| | - Rahul G Sangani
- Department of Medicine, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
42
|
Tsikis ST, Hirsch TI, Fligor SC, Quigley M, Puder M. Targeting the lung endothelial niche to promote angiogenesis and regeneration: A review of applications. Front Mol Biosci 2022; 9:1093369. [PMID: 36601582 PMCID: PMC9807216 DOI: 10.3389/fmolb.2022.1093369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Lung endothelial cells comprise the pulmonary vascular bed and account for the majority of cells in the lungs. Beyond their role in gas exchange, lung ECs form a specialized microenvironment, or niche, with important roles in health and disease. In early development, progenitor ECs direct alveolar development through angiogenesis. Following birth, lung ECs are thought to maintain their regenerative capacity despite the aging process. As such, harnessing the power of the EC niche, specifically to promote angiogenesis and alveolar regeneration has potential clinical applications. Here, we focus on translational research with applications related to developmental lung diseases including pulmonary hypoplasia and bronchopulmonary dysplasia. An overview of studies examining the role of ECs in lung regeneration following acute lung injury is also provided. These diseases are all characterized by significant morbidity and mortality with limited existing therapeutics, affecting both young children and adults.
Collapse
Affiliation(s)
- Savas T Tsikis
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Thomas I Hirsch
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Scott C Fligor
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Mikayla Quigley
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Mark Puder
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
43
|
Ren J, Lock MC, Darby JRT, Orgeig S, Holman SL, Quinn M, Seed M, Muhlhausler BS, McMillen IC, Morrison JL. PPARγ activation in late gestation does not promote surfactant maturation in the fetal sheep lung. J Dev Orig Health Dis 2021; 12:963-974. [PMID: 33407953 DOI: 10.1017/s204017442000135x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Respiratory distress syndrome results from inadequate functional pulmonary surfactant and is a significant cause of mortality in preterm infants. Surfactant is essential for regulating alveolar interfacial surface tension, and its synthesis by Type II alveolar epithelial cells is stimulated by leptin produced by pulmonary lipofibroblasts upon activation by peroxisome proliferator-activated receptor γ (PPARγ). As it is unknown whether PPARγ stimulation or direct leptin administration can stimulate surfactant synthesis before birth, we examined the effect of continuous fetal administration of either the PPARγ agonist, rosiglitazone (RGZ; Study 1) or leptin (Study 2) on surfactant protein maturation in the late gestation fetal sheep lung. We measured mRNA expression of genes involved in surfactant maturation and showed that RGZ treatment reduced mRNA expression of LPCAT1 (surfactant phospholipid synthesis) and LAMP3 (marker for lamellar bodies), but did not alter mRNA expression of PPARγ, surfactant proteins (SFTP-A, -B, -C, and -D), PCYT1A (surfactant phospholipid synthesis), ABCA3 (phospholipid transportation), or the PPARγ target genes SPHK-1 and PAI-1. Leptin infusion significantly increased the expression of PPARγ and IGF2 and decreased the expression of SFTP-B. However, mRNA expression of the majority of genes involved in surfactant synthesis was not affected. These results suggest a potential decreased capacity for surfactant phospholipid and protein production in the fetal lung after RGZ and leptin administration, respectively. Therefore, targeting PPARγ may not be a feasible mechanistic approach to promote lung maturation.
Collapse
Affiliation(s)
- Jiaqi Ren
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
- Hospital for Sick Children, Toronto, ON, Canada
| | - Mitchell C Lock
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Jack R T Darby
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Sandra Orgeig
- Cancer Research Institute, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Stacey L Holman
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Megan Quinn
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Mike Seed
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Hospital for Sick Children, Toronto, ON, Canada
| | | | - I Caroline McMillen
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| |
Collapse
|
44
|
Aloufi N, Alluli A, Eidelman DH, Baglole CJ. Aberrant Post-Transcriptional Regulation of Protein Expression in the Development of Chronic Obstructive Pulmonary Disease. Int J Mol Sci 2021; 22:ijms222111963. [PMID: 34769392 PMCID: PMC8584689 DOI: 10.3390/ijms222111963] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is an incurable and prevalent respiratory disorder that is characterized by chronic inflammation and emphysema. COPD is primarily caused by cigarette smoke (CS). CS alters numerous cellular processes, including the post-transcriptional regulation of mRNAs. The identification of RNA-binding proteins (RBPs), microRNAs (miRNAs), and long non-coding RNAs (lncRNAs) as main factors engaged in the regulation of RNA biology opens the door to understanding their role in coordinating physiological cellular processes. Dysregulation of post-transcriptional regulation by foreign particles in CS may lead to the development of diseases such as COPD. Here we review current knowledge about post-transcriptional events that may be involved in the pathogenesis of COPD.
Collapse
Affiliation(s)
- Noof Aloufi
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada; (N.A.); (A.A.)
- Department of Medical Laboratory Technology, Applied Medical Science, Taibah University, Universities Road, Medina P.O. Box 344, Saudi Arabia
| | - Aeshah Alluli
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada; (N.A.); (A.A.)
| | - David H. Eidelman
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada;
| | - Carolyn J. Baglole
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada; (N.A.); (A.A.)
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada;
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC H3G 1Y6, Canada
- Correspondence:
| |
Collapse
|
45
|
Ren J, Darby JRT, Lock MC, Holman SL, Saini BS, Bradshaw EL, Orgeig S, Perumal SR, Wiese MD, Macgowan CK, Seed M, Morrison JL. Impact of maternal late gestation undernutrition on surfactant maturation, pulmonary blood flow and oxygen delivery measured by magnetic resonance imaging in the sheep fetus. J Physiol 2021; 599:4705-4724. [PMID: 34487347 DOI: 10.1113/jp281292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 09/01/2021] [Indexed: 11/08/2022] Open
Abstract
Restriction of fetal substrate supply has an adverse effect on surfactant maturation in the lung and thus affects the transition from in utero placental oxygenation to pulmonary ventilation ex utero. The effects on surfactant maturation are mediated by alteration in mechanisms regulating surfactant protein and phospholipid synthesis. This study aimed to determine the effects of late gestation maternal undernutrition (LGUN) and LGUN plus fetal glucose infusion (LGUN+G) compared to Control on surfactant maturation and lung development, and the relationship with pulmonary blood flow and oxygen delivery ( D O 2 ) measured by magnetic resonance imaging (MRI) with molecules that regulate lung development. LGUN from 115 to 140 days' gestation significantly decreased fetal body weight, which was normalized by glucose infusion. LGUN and LGUN+G resulted in decreased fetal plasma glucose concentration, with no change in fetal arterial P O 2 compared to control. There was no effect of LGUN and LGUN+G on the mRNA expression of surfactant proteins (SFTP) and genes regulating surfactant maturation in the fetal lung. However, blood flow in the main pulmonary artery was significantly increased in LGUN, despite no change in blood flow in the left or right pulmonary artery and D O 2 to the fetal lung. There was a negative relationship between left pulmonary artery flow and D O 2 to the left lung with SFTP-B and GLUT1 mRNA expression, while their relationship with VEGFR2 was positive. These results suggest that increased pulmonary blood flow measured by MRI may have an adverse effect on surfactant maturation during fetal lung development. KEY POINTS: Maternal undernutrition during gestation alters fetal lung development by impacting surfactant maturation. However, the direction of change remains controversial. We examined the effects of maternal late gestation maternal undernutrition (LGUN) on maternal and fetal outcomes, signalling pathways involved in fetal lung development, pulmonary haemodynamics and oxygen delivery in sheep using a combination of molecular and magnetic resonance imaging (MRI) techniques. LGUN decreased fetal plasma glucose concentration without affecting arterial P O 2 . Surfactant maturation was not affected; however, main pulmonary artery blood flow was significantly increased in the LGUN fetuses. This is the first study to explore the relationship between in utero MRI measures of pulmonary haemodynamics and lung development. Across all treatment groups, left pulmonary artery blood flow and oxygen delivery were negatively correlated with surfactant protein B mRNA and protein expression in late gestation.
Collapse
Affiliation(s)
- Jiaqi Ren
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia.,Translational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jack R T Darby
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Mitchell C Lock
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Stacey L Holman
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Brahmdeep S Saini
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Division of Cardiology, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
| | - Emma L Bradshaw
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Sandra Orgeig
- UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Sunthara R Perumal
- Preclinical Imaging & Research Laboratories, South Australian Health & Medical Research Institute, Adelaide, Australia
| | - Michael D Wiese
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Mike Seed
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Division of Cardiology, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
46
|
Hisata S, Racanelli AC, Kermani P, Schreiner R, Houghton S, Palikuqi B, Kunar B, Zhou A, McConn K, Capili A, Redmond D, Nolan DJ, Ginsberg M, Ding BS, Martinez FJ, Scandura JM, Cloonan SM, Rafii S, Choi AM. Reversal of emphysema by restoration of pulmonary endothelial cells. J Exp Med 2021; 218:e20200938. [PMID: 34287647 PMCID: PMC8298104 DOI: 10.1084/jem.20200938] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 12/03/2020] [Accepted: 05/17/2021] [Indexed: 12/11/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is marked by airway inflammation and airspace enlargement (emphysema) leading to airflow obstruction and eventual respiratory failure. Microvasculature dysfunction is associated with COPD/emphysema. However, it is not known if abnormal endothelium drives COPD/emphysema pathology and/or if correcting endothelial dysfunction has therapeutic potential. Here, we show the centrality of endothelial cells to the pathogenesis of COPD/emphysema in human tissue and using an elastase-induced murine model of emphysema. Airspace disease showed significant endothelial cell loss, and transcriptional profiling suggested an apoptotic, angiogenic, and inflammatory state. This alveolar destruction was rescued by intravenous delivery of healthy lung endothelial cells. Leucine-rich α-2-glycoprotein-1 (LRG1) was a driver of emphysema, and deletion of Lrg1 from endothelial cells rescued vascular rarefaction and alveolar regression. Hence, targeting endothelial cell biology through regenerative methods and/or inhibition of the LRG1 pathway may represent strategies of immense potential for the treatment of COPD/emphysema.
Collapse
Affiliation(s)
- Shu Hisata
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY
- Division of Pulmonary Medicine, Department of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Alexandra C. Racanelli
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY
- New York Presbyterian Hospital/Weill Cornell Medical Center, New York, NY
| | - Pouneh Kermani
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Ryan Schreiner
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Sean Houghton
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Brisa Palikuqi
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Balvir Kunar
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Aiyuan Zhou
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY
- Department of Respiratory and Critical Care Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Keith McConn
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Allyson Capili
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY
| | - David Redmond
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY
| | | | | | - Bi-Sen Ding
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Fernando J. Martinez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Joseph M. Scandura
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Suzanne M. Cloonan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland and Tallaght University Hospital, Dublin, Ireland
| | - Shahin Rafii
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Augustine M.K. Choi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY
- New York Presbyterian Hospital/Weill Cornell Medical Center, New York, NY
| |
Collapse
|
47
|
Pistenmaa CL, Nardelli P, Ash SY, Come CE, Diaz AA, Rahaghi FN, Barr RG, Young KA, Kinney GL, Simmons JP, Wade RC, Wells JM, Hokanson JE, Washko GR, San José Estépar R. Pulmonary Arterial Pruning and Longitudinal Change in Percent Emphysema and Lung Function: The Genetic Epidemiology of COPD Study. Chest 2021; 160:470-480. [PMID: 33607083 PMCID: PMC8411454 DOI: 10.1016/j.chest.2021.01.084] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/28/2020] [Accepted: 01/23/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Pulmonary endothelial damage has been shown to precede the development of emphysema in animals, and vascular changes in humans have been observed in COPD and emphysema. RESEARCH QUESTION Is intraparenchymal vascular pruning associated with longitudinal progression of emphysema on CT imaging or decline in lung function over 5 years? STUDY DESIGN AND METHODS The Genetic Epidemiology of COPD Study enrolled ever smokers with and without COPD from 2008 through 2011. The percentage of emphysema-like lung, or "percent emphysema," was assessed at baseline and after 5 years on noncontrast CT imaging as the percentage of lung voxels < -950 Hounsfield units. An automated CT imaging-based tool assessed and classified intrapulmonary arteries and veins. Spirometry measures are postbronchodilator. Pulmonary arterial pruning was defined as a lower ratio of small artery volume (< 5 mm2 cross-sectional area) to total lung artery volume. Mixed linear models included demographics, anthropomorphics, smoking, and COPD, with emphysema models also adjusting for CT imaging scanner and lung function models adjusting for clinical center and baseline percent emphysema. RESULTS At baseline, the 4,227 participants were 60 ± 9 years of age, 50% were women, 28% were Black, 47% were current smokers, and 41% had COPD. Median percent emphysema was 2.1 (interquartile range, 0.6-6.3) and progressed 0.24 percentage points/y (95% CI, 0.22-0.26 percentage points/y) over 5.6 years. Mean FEV1 to FVC ratio was 68.5 ± 14.2% and declined 0.26%/y (95% CI, -0.30 to -0.23%/y). Greater pulmonary arterial pruning was associated with more rapid progression of percent emphysema (0.11 percentage points/y per 1-SD increase in arterial pruning; 95% CI, 0.09-0.16 percentage points/y), including after adjusting for baseline percent emphysema and FEV1. Arterial pruning also was associated with a faster decline in FEV1 to FVC ratio (-0.04%/y per 1-SD increase in arterial pruning; 95% CI, -0.008 to -0.001%/y). INTERPRETATION Pulmonary arterial pruning was associated with faster progression of percent emphysema and more rapid decline in FEV1 to FVC ratio over 5 years in ever smokers, suggesting that pulmonary vascular differences may be relevant in disease progression. TRIAL REGISTRY ClinicalTrials.gov; No.: NCT00608764; URL: www.clinicaltrials.gov.
Collapse
Affiliation(s)
| | - P Nardelli
- Department of Radiology, Brigham and Women's Hospital, Boston, MA
| | - S Y Ash
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - C E Come
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - A A Diaz
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - F N Rahaghi
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - R G Barr
- Departments of Medicine and Epidemiology, Columbia University, New York, NY
| | - K A Young
- Department of Epidemiology, Colorado School of Public Health, University of Colorado, Denver, CO
| | - G L Kinney
- Department of Epidemiology, Colorado School of Public Health, University of Colorado, Denver, CO
| | - J P Simmons
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - R C Wade
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - J M Wells
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - J E Hokanson
- Department of Epidemiology, Colorado School of Public Health, University of Colorado, Denver, CO
| | - G R Washko
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | | |
Collapse
|
48
|
Green CE, Clarke J, Bicknell R, Turner AM. Pulmonary MicroRNA Changes Alter Angiogenesis in Chronic Obstructive Pulmonary Disease and Lung Cancer. Biomedicines 2021; 9:830. [PMID: 34356894 PMCID: PMC8301412 DOI: 10.3390/biomedicines9070830] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/05/2021] [Accepted: 07/07/2021] [Indexed: 12/12/2022] Open
Abstract
The pulmonary endothelium is dysfunctional in chronic obstructive pulmonary disease (COPD), a known risk factor for lung cancer. The pulmonary endothelium is altered in emphysema, which is disproportionately affected by cancers. Gene and microRNA expression differs between COPD and non-COPD lung. We hypothesised that the alteration in microRNA expression in the pulmonary endothelium contributes to its dysfunction. A total of 28 patients undergoing pulmonary resection were recruited and endothelial cells were isolated from healthy lung and tumour. MicroRNA expression was compared between COPD and non-COPD patients. Positive findings were confirmed by quantitative polymerase chain reaction (qPCR). Assays assessing angiogenesis and cellular migration were conducted in Human Umbilical Vein Endothelial Cells (n = 3-4) transfected with microRNA mimics and compared to cells transfected with negative control RNA. Expression of miR-181b-3p, miR-429 and miR-23c (all p < 0.05) was increased in COPD. Over-expression of miR-181b-3p was associated with reduced endothelial sprouting (p < 0.05). miR-429 was overexpressed in lung cancer as well and exhibited a reduction in tubular formation. MicroRNA-driven changes in the pulmonary endothelium thus represent a novel mechanism driving emphysema. These processes warrant further study to determine if they may be therapeutic targets in COPD and lung cancer.
Collapse
Affiliation(s)
- Clara E. Green
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Joseph Clarke
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (J.C.); (R.B.)
| | - Roy Bicknell
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (J.C.); (R.B.)
| | - Alice M. Turner
- Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK;
| |
Collapse
|
49
|
Brügger M, Démoulins T, Barut GT, Zumkehr B, Oliveira Esteves BI, Mehinagic K, Haas Q, Schögler A, Rameix-Welti MA, Eléouët JF, Moehrlen U, Marti TM, Schmid RA, Summerfield A, Posthaus H, Ruggli N, Hall SRR, Alves MP. Pulmonary mesenchymal stem cells are engaged in distinct steps of host response to respiratory syncytial virus infection. PLoS Pathog 2021; 17:e1009789. [PMID: 34320038 PMCID: PMC8351988 DOI: 10.1371/journal.ppat.1009789] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 08/09/2021] [Accepted: 07/08/2021] [Indexed: 02/06/2023] Open
Abstract
Lung-resident (LR) mesenchymal stem and stromal cells (MSCs) are key elements of the alveolar niche and fundamental regulators of homeostasis and regeneration. We interrogated their function during virus-induced lung injury using the highly prevalent respiratory syncytial virus (RSV) which causes severe outcomes in infants. We applied complementary approaches with primary pediatric LR-MSCs and a state-of-the-art model of human RSV infection in lamb. Remarkably, RSV-infection of pediatric LR-MSCs led to a robust activation, characterized by a strong antiviral and pro-inflammatory phenotype combined with mediators related to T cell function. In line with this, following in vivo infection, RSV invades and activates LR-MSCs, resulting in the expansion of the pulmonary MSC pool. Moreover, the global transcriptional response of LR-MSCs appears to follow RSV disease, switching from an early antiviral signature to repair mechanisms including differentiation, tissue remodeling, and angiogenesis. These findings demonstrate the involvement of LR-MSCs during virus-mediated acute lung injury and may have therapeutic implications.
Collapse
Affiliation(s)
- Melanie Brügger
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Thomas Démoulins
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - G. Tuba Barut
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Beatrice Zumkehr
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Blandina I. Oliveira Esteves
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Kemal Mehinagic
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Quentin Haas
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Aline Schögler
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Marie-Anne Rameix-Welti
- Université Paris-Saclay, INSERM, Université de Versailles St. Quentin, UMR 1173 (2I), Versailles, France
| | | | - Ueli Moehrlen
- Pediatric Surgery, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Thomas M. Marti
- Department of Biomedical Research, University of Bern, Bern, Switzerland
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ralph A. Schmid
- Department of Biomedical Research, University of Bern, Bern, Switzerland
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Artur Summerfield
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Horst Posthaus
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Nicolas Ruggli
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Sean R. R. Hall
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Gillies McIndoe Research Institute, Wellington, New Zealand
| | - Marco P. Alves
- Institute of Virology and Immunology, University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
50
|
Yuan K, Agarwal S, Chakraborty A, Condon DF, Patel H, Zhang S, Huang F, Mello SA, Kirk OI, Vasquez R, de Jesus Perez VA. Lung Pericytes in Pulmonary Vascular Physiology and Pathophysiology. Compr Physiol 2021; 11:2227-2247. [PMID: 34190345 PMCID: PMC10507675 DOI: 10.1002/cphy.c200027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pericytes are mesenchymal-derived mural cells localized within the basement membrane of pulmonary and systemic capillaries. Besides structural support, pericytes control vascular tone, produce extracellular matrix components, and cytokines responsible for promoting vascular homeostasis and angiogenesis. However, pericytes can also contribute to vascular pathology through the production of pro-inflammatory and pro-fibrotic cytokines, differentiation into myofibroblast-like cells, destruction of the extracellular matrix, and dissociation from the vessel wall. In the lung, pericytes are responsible for maintaining the integrity of the alveolar-capillary membrane and coordinating vascular repair in response to injury. Loss of pericyte communication with alveolar capillaries and a switch to a pro-inflammatory/pro-fibrotic phenotype are common features of lung disorders associated with vascular remodeling, inflammation, and fibrosis. In this article, we will address how to differentiate pericytes from other cells, discuss the molecular mechanisms that regulate the interactions of pericytes and endothelial cells in the pulmonary circulation, and the experimental tools currently used to study pericyte biology both in vivo and in vitro. We will also discuss evidence that links pericytes to the pathogenesis of clinically relevant lung disorders such as pulmonary hypertension, idiopathic lung fibrosis, sepsis, and SARS-COVID. Future studies dissecting the complex interactions of pericytes with other pulmonary cell populations will likely reveal critical insights into the origin of pulmonary diseases and offer opportunities to develop novel therapeutics to treat patients afflicted with these devastating disorders. © 2021 American Physiological Society. Compr Physiol 11:2227-2247, 2021.
Collapse
Affiliation(s)
- Ke Yuan
- Division of Respiratory Diseases Research, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Stuti Agarwal
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Ananya Chakraborty
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - David F. Condon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Hiral Patel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Serena Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Flora Huang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Salvador A. Mello
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | | | - Rocio Vasquez
- University of Central Florida, Orlando, Florida, USA
| | - Vinicio A. de Jesus Perez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| |
Collapse
|