1
|
Su Y, Lucas R, Fulton DJ, Verin AD. Mechanisms of pulmonary endothelial barrier dysfunction in acute lung injury and acute respiratory distress syndrome. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:80-87. [PMID: 39006829 PMCID: PMC11242916 DOI: 10.1016/j.pccm.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Indexed: 07/16/2024]
Abstract
Endothelial cells (ECs) form a semi-permeable barrier between the interior space of blood vessels and the underlying tissues. Pulmonary endothelial barrier integrity is maintained through coordinated cellular processes involving receptors, signaling molecules, junctional complexes, and protein-regulated cytoskeletal reorganization. In acute lung injury (ALI) or its more severe form acute respiratory distress syndrome (ARDS), the loss of endothelial barrier integrity secondary to endothelial dysfunction caused by severe pulmonary inflammation and/or infection leads to pulmonary edema and hypoxemia. Pro-inflammatory agonists such as histamine, thrombin, bradykinin, interleukin 1β, tumor necrosis factor α, vascular endothelial growth factor, angiopoietin-2, and platelet-activating factor, as well as bacterial toxins and reactive oxygen species, cause dynamic changes in cytoskeletal structure, adherens junction disorganization, and detachment of vascular endothelial cadherin (VE-cadherin) from the actin cytoskeleton, leading to an increase in endothelial permeability. Endothelial interactions with leukocytes, platelets, and coagulation enhance the inflammatory response. Moreover, inflammatory infiltration and the associated generation of pro-inflammatory cytokines during infection cause EC death, resulting in further compromise of the structural integrity of lung endothelial barrier. Despite the use of potent antibiotics and aggressive intensive care support, the mortality of ALI is still high, because the mechanisms of pulmonary EC barrier disruption are not fully understood. In this review, we summarized recent advances in the studies of endothelial cytoskeletal reorganization, inter-endothelial junctions, endothelial inflammation, EC death, and endothelial repair in ALI and ARDS, intending to shed some light on the potential diagnostic and therapeutic targets in the clinical management of the disease.
Collapse
Affiliation(s)
- Yunchao Su
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30912, USA
| | - Rudolf Lucas
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - David J.R. Fulton
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Alexander D. Verin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
2
|
Motta-Ribeiro GC, Winkler T, Costa ELV, de Prost N, Tucci MR, Vidal Melo MF. Worsening of lung perfusion to tissue density distributions during early acute lung injury. J Appl Physiol (1985) 2023; 135:239-250. [PMID: 37289955 PMCID: PMC10393328 DOI: 10.1152/japplphysiol.00028.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 05/12/2023] [Accepted: 06/05/2023] [Indexed: 06/10/2023] Open
Abstract
Lung perfusion magnitude and distribution are essential for oxygenation and, potentially, lung inflammation and protection during acute respiratory distress syndrome (ARDS). Yet, perfusion patterns and their relationship to inflammation are unknown pre-ARDS. We aimed to assess perfusion/density ratios and spatial perfusion-density distributions and associate these to lung inflammation, during early lung injury in large animals at different physiological conditions caused by different systemic inflammation and positive end-expiratory pressure (PEEP) levels. Sheep were protectively ventilated (16-24 h) and imaged for lung density, pulmonary capillary perfusion (13Nitrogen-saline), and inflammation (18F-fluorodeoxyglucose) using positron emission and computed tomography. We studied four conditions: permissive atelectasis (PEEP = 0 cmH2O); and ARDSNet low-stretch PEEP-setting strategy with supine moderate or mild endotoxemia, and prone mild endotoxemia. Perfusion/density heterogeneity increased pre-ARDS in all groups. Perfusion redistribution to density depended on ventilation strategy and endotoxemia level, producing more atelectasis in mild than moderate endotoxemia (P = 0.010) with the oxygenation-based PEEP-setting strategy. The spatial distribution of 18F-fluorodeoxyglucose uptake was related to local Q/D (P < 0.001 for Q/D group interaction). Moderate endotoxemia yielded markedly low/zero perfusion in normal-low density lung, with 13Nitrogen-saline perfusion indicating nondependent capillary obliteration. Prone animals' perfusion was remarkably homogeneously distributed with density. Lung perfusion redistributes heterogeneously to density during pre-ARDS protective ventilation in animals. This is associated with increased inflammation, nondependent capillary obliteration, and lung derecruitment susceptibility depending on endotoxemia level and ventilation strategy.NEW & NOTEWORTHY Perfusion redistribution does not follow lung density redistribution in the first 16-24 h of systemic endotoxemia and protective tidal volume mechanical ventilation. The same oxygenation-based positive end-expiratory pressure (PEEP)-setting strategy can lead at different endotoxemia levels to different perfusion redistributions, PEEP values, and lung aerations, worsening lung biomechanical conditions. During early acute lung injury, regional perfusion-to-tissue density ratio is associated with increased neutrophilic inflammation, and susceptibility to nondependent capillary occlusion and lung derecruitment, potentially marking and/or driving lung injury.
Collapse
Affiliation(s)
- Gabriel C Motta-Ribeiro
- Biomedical Engineering Program, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tilo Winkler
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Eduardo L V Costa
- Divisão de Pneumologia, Faculdade de Medicina, Instituto do Coração (Incor), Hospital das Clínicas, Universidade de São Paulo, São Paulo, Brazil
- Instituto de Ensino e Pesquisa do Hospital Sírio Libanês, São Paulo, Brazil
| | - Nicolas de Prost
- Hôpitaux Universitaires Henri Mondor and Université Paris Est Créteil and INSERM - Unité U955, Créteil, France
| | - Mauro R Tucci
- Divisão de Pneumologia, Faculdade de Medicina, Instituto do Coração (Incor), Hospital das Clínicas, Universidade de São Paulo, São Paulo, Brazil
| | - Marcos F Vidal Melo
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, New York, United States
| |
Collapse
|
3
|
Shi X, Seidle KA, Simms KJ, Dong F, Chilian WM, Zhang P. Endothelial progenitor cells in the host defense response. Pharmacol Ther 2023; 241:108315. [PMID: 36436689 PMCID: PMC9944665 DOI: 10.1016/j.pharmthera.2022.108315] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/15/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022]
Abstract
Extensive injury of endothelial cells in blood vasculature, especially in the microcirculatory system, frequently occurs in hosts suffering from sepsis and the accompanied systemic inflammation. Pathological factors, including toxic components derived from invading microbes, oxidative stress associated with tissue ischemia/reperfusion, and vessel active mediators generated during the inflammatory response, are known to play important roles in mediating endothelial injury. Collapse of microcirculation and tissue edema developed from the failure of endothelial barrier function in vital organ systems, including the lung, brain, and kidney, are detrimental, which often predict fatal outcomes. The host body possesses a substantial capacity for maintaining vascular homeostasis and repairing endothelial damage. Bone marrow and vascular wall niches house endothelial progenitor cells (EPCs). In response to septic challenges, EPCs in their niche environment are rapidly activated for proliferation and angiogenic differentiation. In the meantime, release of EPCs from their niches into the blood stream and homing of these vascular precursors to tissue sites of injury are markedly increased. The recruited EPCs actively participate in host defense against endothelial injury and repair of damage in blood vasculature via direct differentiation into endothelial cells for re-endothelialization as well as production of vessel active mediators to exert paracrine and autocrine effects on angiogenesis/vasculogenesis. In recent years, investigations on significance of EPCs in host defense and molecular signaling mechanisms underlying regulation of the EPC response have achieved substantial progress, which promotes exploration of vascular precursor cell-based approaches for effective prevention and treatment of sepsis-induced vascular injury as well as vital organ system failure.
Collapse
Affiliation(s)
- Xin Shi
- Department of Integrative Medical Sciences, Northeast Ohio Medical University College of Medicine, Rootstown, OH 44272, United States of America
| | - Kelly A Seidle
- Department of Integrative Medical Sciences, Northeast Ohio Medical University College of Medicine, Rootstown, OH 44272, United States of America
| | - Kevin J Simms
- Department of Integrative Medical Sciences, Northeast Ohio Medical University College of Medicine, Rootstown, OH 44272, United States of America
| | - Feng Dong
- Department of Integrative Medical Sciences, Northeast Ohio Medical University College of Medicine, Rootstown, OH 44272, United States of America
| | - William M Chilian
- Department of Integrative Medical Sciences, Northeast Ohio Medical University College of Medicine, Rootstown, OH 44272, United States of America
| | - Ping Zhang
- Department of Integrative Medical Sciences, Northeast Ohio Medical University College of Medicine, Rootstown, OH 44272, United States of America.
| |
Collapse
|
4
|
Wei T, Zhang C, Song Y. Molecular mechanisms and roles of pyroptosis in acute lung injury. Chin Med J (Engl) 2022; 135:2417-2426. [PMID: 36583860 PMCID: PMC9945565 DOI: 10.1097/cm9.0000000000002425] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Indexed: 12/31/2022] Open
Abstract
ABSTRACT Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS), which are characterized by excessive inflammation and accompanied by diffuse injury of alveoli, can result in severe respiratory failures. The morbidity and mortality of patients remain high because the major treatments for ALI/ARDS are mainly supportive due to the lack of effective therapies. Numerous studies have demonstrated that the aggravation of coronavirus disease 2019 (COVID-19) leads to severe pneumonia and even ARDS. Pyroptosis, a biological process identified as a type of programed cell death, is mainly triggered by inflammatory caspase activation and is directly meditated by the gasdermin protein family, as well as being associated with the secretion and release of pro-inflammatory cytokines. Clinical and experimental evidence corroborates that pyroptosis of various cells in the lung, such as immune cells and structural cells, may play an important role in the pathogenesis of "cytokine storms" in ALI/ARDS, including those induced by COVID-19. Here, with a focus on ALI/ARDS and COVID-19, we summarized the recent advances in this field and proposed the theory of an inflammatory cascade in pyroptosis to identify new targets and pave the way for new approaches to treat these diseases.
Collapse
Affiliation(s)
- Tianchang Wei
- Department of Pulmonary Medicine, Shanghai Key Laboratory of Lung Inflammation and Injury, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Cuiping Zhang
- Department of Pulmonary Medicine, Shanghai Key Laboratory of Lung Inflammation and Injury, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yuanlin Song
- Department of Pulmonary Medicine, Shanghai Key Laboratory of Lung Inflammation and Injury, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai 200032, China
- Shanghai Respiratory Research Institute, Shanghai 200032, China
- Jinshan Hospital of Fudan University, Shanghai 201508, China
| |
Collapse
|
5
|
Omar SA, Abdul-Hafez A, Ibrahim S, Pillai N, Abdulmageed M, Thiruvenkataramani RP, Mohamed T, Madhukar BV, Uhal BD. Stem-Cell Therapy for Bronchopulmonary Dysplasia (BPD) in Newborns. Cells 2022; 11:cells11081275. [PMID: 35455954 PMCID: PMC9025385 DOI: 10.3390/cells11081275] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/30/2022] [Accepted: 04/07/2022] [Indexed: 11/17/2022] Open
Abstract
Premature newborns are at a higher risk for the development of respiratory distress syndrome (RDS), acute lung injury (ALI) associated with lung inflammation, disruption of alveolar structure, impaired alveolar growth, lung fibrosis, impaired lung angiogenesis, and development of bronchopulmonary dysplasia (BPD) with severe long-term developmental adverse effects. The current therapy for BPD is limited to supportive care including high-oxygen therapy and pharmacotherapy. Recognizing more feasible treatment options to improve lung health and reduce complications associated with BPD is essential for improving the overall quality of life of premature infants. There is a reduction in the resident stem cells in lungs of premature infants with BPD, which strongly suggests a critical role of stem cells in BPD pathogenesis; this warrants the exploration of the potential therapeutic use of stem-cell therapy. Stem-cell-based therapies have shown promise for the treatment of many pathological conditions including acute lung injury and BPD. Mesenchymal stem cells (MSCs) and MSC-derived extracellular vesicles (EVs) including exosomes are promising and effective therapeutic modalities for the treatment of BPD. Treatment with MSCs and EVs may help to reduce lung inflammation, improve pulmonary architecture, attenuate pulmonary fibrosis, and increase the survival rate.
Collapse
Affiliation(s)
- Said A. Omar
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (A.A.-H.); (S.I.); (N.P.); (M.A.); (R.P.T.); (T.M.); (B.V.M.)
- Regional Neonatal Intensive Care Unit, Sparrow Health System, Lansing, MI 48912, USA
- Correspondence: ; Tel.: +1-517-364-2948
| | - Amal Abdul-Hafez
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (A.A.-H.); (S.I.); (N.P.); (M.A.); (R.P.T.); (T.M.); (B.V.M.)
| | - Sherif Ibrahim
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (A.A.-H.); (S.I.); (N.P.); (M.A.); (R.P.T.); (T.M.); (B.V.M.)
| | - Natasha Pillai
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (A.A.-H.); (S.I.); (N.P.); (M.A.); (R.P.T.); (T.M.); (B.V.M.)
| | - Mohammed Abdulmageed
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (A.A.-H.); (S.I.); (N.P.); (M.A.); (R.P.T.); (T.M.); (B.V.M.)
- Regional Neonatal Intensive Care Unit, Sparrow Health System, Lansing, MI 48912, USA
| | - Ranga Prasanth Thiruvenkataramani
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (A.A.-H.); (S.I.); (N.P.); (M.A.); (R.P.T.); (T.M.); (B.V.M.)
- Regional Neonatal Intensive Care Unit, Sparrow Health System, Lansing, MI 48912, USA
| | - Tarek Mohamed
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (A.A.-H.); (S.I.); (N.P.); (M.A.); (R.P.T.); (T.M.); (B.V.M.)
- Regional Neonatal Intensive Care Unit, Sparrow Health System, Lansing, MI 48912, USA
| | - Burra V. Madhukar
- Division of Neonatology, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA; (A.A.-H.); (S.I.); (N.P.); (M.A.); (R.P.T.); (T.M.); (B.V.M.)
| | - Bruce D. Uhal
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA;
| |
Collapse
|
6
|
Xu Q, Chen J, Zhu Y, Xia W, Liu Y, Xu J. JTE-013 Alleviates Inflammatory Injury and Endothelial Dysfunction Induced by Sepsis In Vivo and In Vitro. J Surg Res 2021; 265:323-332. [PMID: 33971464 DOI: 10.1016/j.jss.2021.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 02/09/2021] [Accepted: 03/03/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Nowadays, there is no approved targeted agent for lung injury induced by sepsis. S1PR2 is confirmed to be a promising diagnosis and treatment target. JTE-013 as S1PR2 antagonists may be an agent of great potential. In this research, we sought to determine the functional role of JTE-013 in lung injury induced by sepsis. MATERIALS AND METHODS Seventy-two rats were assigned into normal group, sepsis model group and JTE-013 group. The animal model of lung injury induced by sepsis was constructed by cecal ligation and puncture. The human pulmonary microvascular endothelial cells (HPMECs) were divided into control, LPS and LPS + JTE-013 group. HPMECs induced by LPS served as the cell model of lung injury induced by sepsis. HE staining assay was performed for assessment of the pathological condition and Evans blue was applied for assessment of pulmonary tissue permeability. Wet/dry ratio was measured as indicators of pulmonary edema degree and neutrophil count was measured as indicators of infection status. The levels of inflammatory factors were detected by corresponding kits, cell survival by CCK-8 assay and protein expression level by western blot. RESULTS S1PR2 was highly expressed in vivo model of lung injury induced by sepsis. It was observed that JTE-013 as antagonist of S1PR2 alleviated the lung tissue injury, endothelial dysfunction and pulmonary edema induced by sepsis. In addition, JTE-013 reduced neutrophil count and levels of inflammatory factors. Moreover, results confirmed that JTE-013 enhanced cell viability and mitigated inflammatory response in cell model of sepsis. CONCLUSIONS Overall, JTE-013 as an antagonist of S1PR2 could relieve inflammatory injury and endothelial dysfunction induced by sepsis in vivo and vitro, resulting in attenuation of lung injury. These findings elucidated that JTE-013 may be a promising targeted agent for lung injury induced by sepsis.
Collapse
Affiliation(s)
- Qiumin Xu
- Emergency Department, The Fourth Sanatorium Area of Hangzhou Special Service Sanatorium Center of Air Force, Nanjing City, Jiangsu Province, China
| | - Jiusheng Chen
- Emergency Department, The Fourth Sanatorium Area of Hangzhou Special Service Sanatorium Center of Air Force, Nanjing City, Jiangsu Province, China
| | - Yifan Zhu
- Convalescent Department, The Fourth Sanatorium Area of Hangzhou Special Service Sanatorium Center of Air Force, Nanjing City, Jiangsu Province, China
| | - Wenjuan Xia
- Emergency Department, The Fourth Sanatorium Area of Hangzhou Special Service Sanatorium Center of Air Force, Nanjing City, Jiangsu Province, China
| | - Yong Liu
- Emergency Department, The Fourth Sanatorium Area of Hangzhou Special Service Sanatorium Center of Air Force, Nanjing City, Jiangsu Province, China
| | - Jieying Xu
- Special Functions Section, The Fourth Sanatorium Area of Hangzhou Special Service Sanatorium Center of Air Force, Nanjing City, Jiangsu Province, China.
| |
Collapse
|
7
|
Hoseinnia S, Ghane M, Norouzi J, Hosseini F. Mesenchymal stem cell and endothelial progenitor cells coinjection improves LPS-induced lung injury via Tie2 activation and downregulation of the TLR4/MyD88 pathway. J Cell Biochem 2021; 122:1791-1804. [PMID: 34397115 DOI: 10.1002/jcb.30133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 12/15/2022]
Abstract
Sepsis is one of the most important complications of infection with a high mortality rate. Recently, cell therapy has been widely used to reduce the symptoms of sepsis. It has been previously reported that mesenchymal stem cell (MSC) and endothelial progenitor cells (EPC) therapy have beneficial effects in experimental models of sepsis. The effects of coculture of MSC and EPC have not yet been used to treat sepsis. Therefore, the aim of this study was to investigate the therapeutic potential of EPC + MSC coculture on the residual effects of sepsis in a lipopolysaccharide (LPS)-induced mice model. Coinjections of EPC + MSC significantly enhanced the survival rate of LPS-induced mice, decreased concentrations of pro-inflammatory cytokines, and increased the level of anti-inflammatory cytokine. The LPS-induced mice that were treated with EPC + MSC showed a notable reduction in pulmonary edema, hepatic enzymes, and C-reactive protein level compared with the control group. Our results showed that coinjection of EPC + MSC up and downregulates Tie2 and TLR4/MyD88 signaling pathways in LPS-induced mice, respectively. Also, in vitro study showed that viability, adhesion, and migration in coculture cells is significantly decreased after being induced with 10 μg/ml LPS. Our results showed that LPS impaired the functional activity of the cocultured EPC + MSC via upregulation of the TLR4/MyD88 signaling pathway, which may be associated with decreased pTie2/Tie2 expression. In conclusion, coinjection of EPC and MSC modulated the TLR4/MyD88 signaling pathway that leads to reduce the inflammatory response. This study may provide promising results for the introduction of cocultured cells to manage infectious diseases and balance the immune response through immune regulatory function.
Collapse
Affiliation(s)
- Sadaf Hoseinnia
- Department of Microbiology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Maryam Ghane
- Department of Biology, Islamshahr Branch, Islamic Azad University, Islamshahr, Iran
| | - Jamile Norouzi
- Department of Microbiology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Farzaneh Hosseini
- Department of Microbiology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
8
|
Kolesnichenko OA, Whitsett JA, Kalin TV, Kalinichenko VV. Therapeutic Potential of Endothelial Progenitor Cells in Pulmonary Diseases. Am J Respir Cell Mol Biol 2021; 65:473-488. [PMID: 34293272 DOI: 10.1165/rcmb.2021-0152tr] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Compromised alveolar development and pulmonary vascular remodeling are hallmarks of pediatric lung diseases such as bronchopulmonary dysplasia (BPD) and alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV). Although advances in surfactant therapy, corticosteroids, and anti-inflammatory drugs have improved clinical management of preterm infants, still those who suffer with severe vascular complications lack viable treatment options. Paucity of the alveolar capillary network in ACDMPV causes respiratory distress and leads to mortality in a vast majority of ACDMPV infants. The discovery of endothelial progenitor cells (EPCs) in 1997 brought forth the paradigm of postnatal vasculogenesis and hope for promoting vascularization in fragile patient populations, such as those with BPD and ACDMPV. The identification of diverse EPC populations, both hematopoietic and nonhematopoietic in origin, provided a need to identify progenitor cell selective markers which are linked to progenitor properties needed to develop cell-based therapies. Focusing to the future potential of EPCs for regenerative medicine, this review will discuss various aspects of EPC biology, beginning with the identification of hematopoietic, nonhematopoietic, and tissue-resident EPC populations. We will review knowledge related to cell surface markers, signature gene expression, key transcriptional regulators, and will explore the translational potential of EPCs for cell-based therapy for BPD and ACDMPV. The ability to produce pulmonary EPCs from patient-derived induced pluripotent stem cells (iPSCs) in vitro, holds promise for restoring vascular growth and function in the lungs of patients with pediatric pulmonary disorders.
Collapse
Affiliation(s)
- Olena A Kolesnichenko
- Cincinnati Children's Hospital Medical Center, 2518, Cincinnati, Ohio, United States
| | - Jeffrey A Whitsett
- The Perinatal Institute and Section of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
| | - Tanya V Kalin
- Cincinnati Children\'s Hospital Medical Center, 2518, Pediatrics, Cincinnati, Ohio, United States
| | - Vladimir V Kalinichenko
- Cincinnati Children's Hospital Medical Center, Pediatrics, Division of Pulmonary Biology, Cincinnati, Ohio, United States;
| |
Collapse
|
9
|
Fucoidan inhibits LPS-induced acute lung injury in mice through regulating GSK-3β-Nrf2 signaling pathway. Arch Pharm Res 2020; 43:646-654. [PMID: 32533502 DOI: 10.1007/s12272-020-01234-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 05/12/2020] [Indexed: 10/24/2022]
Abstract
The purpose of this study was to investigate the protective effects of fucoidan on Lipopolysaccharide (LPS)-induced acute lung injury (ALI) in mice. The mice were divided into the control, LPS, and LPS + fucoidan (20, 40, or 80 mg/kg) groups. LPS was given by intracheal instillation and fucoidan was given 1 h before LPS treatment. Myeloperoxidase (MPO) activity, malondialdehyde (MDA), superoxide dismutase (SOD), reactive oxygen species (ROS), glutathione (GSH) contents, and inflammatory cytokine production were detected. The results showed that LPS-induced TNF-α, IL-1β, and IL-6 production, lung wet/dry (W/D) ratio, ROS, MDA content, and MPO activity were suppressed by fucoidan. The levels of SOD and GSH were increased by fucoidan. Meanwhile, LPS-induced nuclear factor kappa-B (NF-κB) activation was dose-dependently attenuated by fucoidan. Furthermore, fucoidan increased the expression of nuclear factor erythroid-2 related factor 2 (Nrf2), Glycogen synthase kinase3β (GSK-3β), and heme oxygenase (HO-1). In vitro, the results demonstrated that fucoidan or GSK-3β inhibitor significantly inhibited LPS-induced TNF-α production in A549 cells. And the inhibition of fucoidan on TNF-α production was blocked by Nrf2 siRNA. This study showed fucoidan protected mice against LPS-induced ALI through inhibiting inflammatory and oxidative responses via regulating GSK-3β-Nrf2 signaling pathway.
Collapse
|
10
|
Patry C, Doniga T, Lenz F, Viergutz T, Weiss C, Tönshoff B, Kalenka A, Yard B, Krebs J, Schaible T, Beck G, Rafat N. Increased mobilization of mesenchymal stem cells in patients with acute respiratory distress syndrome undergoing extracorporeal membrane oxygenation. PLoS One 2020; 15:e0227460. [PMID: 31986159 PMCID: PMC6984734 DOI: 10.1371/journal.pone.0227460] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 12/18/2019] [Indexed: 01/31/2023] Open
Abstract
Background The acute respiratory distress syndrome (ARDS) is characterized by pulmonary epithelial and endothelial barrier dysfunction and injury. In severe forms of ARDS, extracorporeal membrane oxygenation (ECMO) is often the last option for life support. Endothelial progenitor (EPC) and mesenchymal stem cells (MSC) can regenerate damaged endothelium and thereby improve pulmonary endothelial dysfunction. However, we still lack sufficient knowledge about how ECMO might affect EPC- and MSC-mediated regenerative pathways in ARDS. Therefore, we investigated if ECMO impacts EPC and MSC numbers in ARDS patients. Methods Peripheral blood mononuclear cells from ARDS patients undergoing ECMO (n = 16) and without ECMO support (n = 12) and from healthy volunteers (n = 16) were isolated. The number and presence of circulating EPC and MSC was detected by flow cytometry. Serum concentrations of vascular endothelial growth factor (VEGF) and angiopoietin 2 (Ang2) were determined. Results In the ECMO group, MSC subpopulations were higher by 71% compared to the non-ECMO group. Numbers of circulating EPC were not significantly altered. During ECMO, VEGF and Ang2 serum levels remained unchanged compared to the non-ECMO group (p = 0.16), but Ang2 serum levels in non-survivors of ARDS were significantly increased by 100% (p = 0.02) compared to survivors. Conclusions ECMO support in ARDS is specifically associated with an increased number of circulating MSC, most likely due to enhanced mobilization, but not with a higher numbers of EPC or serum concentrations of VEGF and Ang2.
Collapse
Affiliation(s)
- Christian Patry
- Department of Pediatrics I, University Children’s Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Thalia Doniga
- Department of Neonatology, University Children’s Hospital Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Franziska Lenz
- Department of Anaesthesiology and Intensive Care Medicine, University Medical Center Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Tim Viergutz
- Department of Anaesthesiology and Intensive Care Medicine, University Medical Center Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Christel Weiss
- Department of Medical Statistics and Biomathematics, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Burkhard Tönshoff
- Department of Pediatrics I, University Children’s Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Armin Kalenka
- Department of Anaesthesiology and Intensive Care Medicine, University Medical Center Mannheim, University of Heidelberg, Heidelberg, Germany
- Department of Anaesthesiology and Intensive Care Medicine, Hospital Bergstraße, Heppenheim, Germany
| | - Benito Yard
- Department of Medicine V, University Medical Center Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Jörg Krebs
- Department of Anaesthesiology and Intensive Care Medicine, University Medical Center Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Thomas Schaible
- Department of Neonatology, University Children’s Hospital Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Grietje Beck
- Department of Anaesthesiology and Intensive Care Medicine, Dr. Horst-Schmidt Clinic, Wiesbaden, Germany
| | - Neysan Rafat
- Department of Pediatrics I, University Children’s Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
- Department of Neonatology, University Children’s Hospital Mannheim, University of Heidelberg, Heidelberg, Germany
- Department of Pharmaceutical Sciences, Bahá'í Institute of Higher Education (BIHE), Teheran, Iran
- * E-mail:
| |
Collapse
|
11
|
Liu A, Zhang X, He H, Zhou L, Naito Y, Sugita S, Lee JW. Therapeutic potential of mesenchymal stem/stromal cell-derived secretome and vesicles for lung injury and disease. Expert Opin Biol Ther 2019; 20:125-140. [PMID: 31701782 DOI: 10.1080/14712598.2020.1689954] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: The acute respiratory distress syndrome (ARDS) is a devastating clinical condition common in patients with respiratory failure. Based largely on numerous preclinical studies and recent Phase I/II clinical trials, administration of stem cells, specifically mesenchymal stem or stromal cells (MSC), as a therapeutic for acute lung injury (ALI) holds great promise. However, concern for the use of stem cells, specifically the risk of iatrogenic tumor formation, remains unresolved. Accumulating evidence now suggest that stem cell-derived conditioned medium (CM) and/or extracellular vesicles (EV) might constitute compelling alternatives.Areas covered: The current review focuses on the preclinical studies testing MSC CM and/or EV as treatment for ALI and other inflammatory lung diseases.Expert opinion: Clinical application of MSC or their secreted CM may be limited by the cost of growing enough cells, the logistic of MSC storage, and the lack of standardization of what constitutes MSC CM. However, the clinical application of MSC EV remains promising, primarily due to the ability of EV to maintain the functional phenotype of the parent cell as a therapeutic. However, utilization of MSC EV will also require large-scale production, the cost of which may be prohibitive unless the potency of the EV can be increased.
Collapse
Affiliation(s)
- Airan Liu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xiwen Zhang
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Hongli He
- Departments of Anesthesiology and Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Li Zhou
- Departments of Anesthesiology and Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Yoshifumi Naito
- Departments of Anesthesiology and Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Shinji Sugita
- Departments of Anesthesiology and Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Jae-Woo Lee
- Departments of Anesthesiology and Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| |
Collapse
|
12
|
Bui KCT, Kim R, Abbasi A, Nguyen M, Villosis MF, Chen Q. Erythropoietin treatment is associated with a reduction in moderate to severe bronchopulmonary dysplasia in preterm infants. A regional retrospective study. Early Hum Dev 2019; 137:104831. [PMID: 31374455 DOI: 10.1016/j.earlhumdev.2019.104831] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/21/2019] [Accepted: 07/24/2019] [Indexed: 11/26/2022]
Abstract
UNLABELLED Erythropoietin treatment is associated with a reduction in moderate to severe bronchopulmonary dysplasia in preterm infants. A regional retrospective study. OBJECTIVE To determine whether premature infants treated with erythropoietin (Epo) in the neonatal period for anemia had a lower incidence of bronchopulmonary dysplasia (BPD), defined as oxygen need at 36 weeks postmenstrual age, and lower rehospitalization rates in the first year of life than infants not exposed. METHODS Retrospective study of a population of infants born at 23 to 32 weeks gestational age, between January 2009 and December 2014, with birthweight ≤1500 g. Patient characteristics, and risk factors for BPD were compared between patients who received erythropoietin, and those not exposed. To examine the association between the outcomes of BPD at 36 weeks PMA, rehospitalization, and erythropoietin treatment, we performed a propensity score (PS) analysis using inverse probability of treatment weighted (IPTW) approach. For comparison, we conducted a logistic regression adjusting for the same covariates used to generate PS using the original population. RESULTS The study population included 1821 preterm infants: 928 received Epo and 893 did not. Epo treatment was associated with a reduction in BPD (18.8% versus 25.9%, p < 0.01) at 36 weeks PMA and reduced median length of stay with lowest BPD rate with Epo initiation before 2 weeks of age. There was no difference in rehospitalization rates in the first year of life. CONCLUSION Erythropoietin treatment was associated with a reduction in BPD but not in rehospitalization rate in the first year of life.
Collapse
Affiliation(s)
- Kim Chi T Bui
- Department of Pediatrics, Kaiser Permanente Los Angeles, United States of America.
| | - Romina Kim
- Department of Pediatrics, Kaiser Permanente Los Angeles, United States of America
| | | | | | | | - Qiaoling Chen
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, United States of America
| |
Collapse
|
13
|
Early Prediction of Persistent Organ Failure by Circulating Endothelial Progenitor Cells in Patients With Acute Pancreatitis. Shock 2019; 50:265-272. [PMID: 29200137 DOI: 10.1097/shk.0000000000001065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Increased circulating endothelial progenitor cells (cEPC) have been observed in patients with vascular injury associated with sepsis and acute lung injury. However, a role for cEPC in severe acute pancreatitis (SAP) remains unclear. We therefore conducted a prospective study to study whether the quantities of cEPC can predict persistent organ failure (POF) in patients with predicted SAP. METHODS A total of 42 predicted SAP patients who were admitted within 24 h after symptom onset and 10 healthy control subjects were enrolled in our study. The proportions of cEPC were analyzed based on flow cytometry simultaneously. Vascular endothelial growth factor (VEGF) levels were measured by enzyme-linked immunosorbent assay. RESULTS The percentage of cEPC was significantly higher in patients with predicted SAP compared with healthy controls. Similarly, the levels of VEGF in peripheral blood were also significantly higher in predicted SAP patients than in the controls. Notably, patients with POF had lower proportion of cEPC compared with patients with transient organ failure (TOF). In contrast, patients with POF had a significantly higher level of VEGF compared with TOF. Of note, the percentages of cEPC were significantly inversely correlated with disease severity scores. More importantly, cEPC showed an excellent discriminative power for predicting POF among predicted SAP patients, whereas plasma VEGF and disease severity scores showed moderate accuracy in predicting future POF. CONCLUSIONS Peripheral EPC as a novel biomarker is elevated and may aid to predict the development of POF in patients with predicted SAP.
Collapse
|
14
|
Yang N, Tian H, Zhan E, Zhai L, Jiao P, Yao S, Lu G, Mu Q, Wang J, Zhao A, Zhou Y, Qin S. Reverse-D-4F improves endothelial progenitor cell function and attenuates LPS-induced acute lung injury. Respir Res 2019; 20:131. [PMID: 31242908 PMCID: PMC6595601 DOI: 10.1186/s12931-019-1099-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 06/13/2019] [Indexed: 12/22/2022] Open
Abstract
Background Patients with acute lung injury (ALI) have increased levels of pro-inflammatory mediators, which impair endothelial progenitor cell (EPC) function. Increasing the number of EPC and alleviating EPC dysfunction induced by pro-inflammatory mediators play important roles in suppressing ALI development. Because the high density lipoprotein reverse-D-4F (Rev-D4F) improves EPC function, we hypothesized that it might repair lipopolysaccharide (LPS)-induced lung damage by improving EPC numbers and function in an LPS-induced ALI mouse model. Methods LPS was used to induce ALI in mice, and then the mice received intraperitoneal injections of Rev-D4F. Immunohistochemical staining, flow cytometry, MTT, transwell, and western blotting were used to assess the effect of Rev-D4F on repairment of lung impairment, and improvement of EPC numbers and function, as well as the signaling pathways involved. Results Rev-D4F inhibits LPS-induced pulmonary edema and decreases plasma levels of the pro-inflammatory mediators TNF-α and ET-1 in ALI mice. Rev-D4F inhibited infiltration of red and white blood cells into the interstitial space, reduced lung injury-induced inflammation, and restored injured pulmonary capillary endothelial cells. In addition, Rev-D4F increased numbers of circulating EPC, stimulated EPC differentiation, and improved EPC function impaired by LPS. Rev-D4F also acted via a PI3-kinase-dependent mechanism to restore levels of phospho-AKT, eNOS, and phospho-eNOS suppressed by LPS. Conclusions These findings indicate that Rev-D4F has an important vasculoprotective role in ALI by improving the EPC numbers and functions, and Rev-D4F reverses LPS-induced EPC dysfuncion partially through PI3K/AKT/eNOS signaling pathway. Electronic supplementary material The online version of this article (10.1186/s12931-019-1099-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nana Yang
- Experimental Center for Medical Research, Weifang Medical University, Weifang City, People's Republic of China.
| | - Hua Tian
- Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Shandong First Medical University, Tai-an City, People's Republic of China
| | - Enxin Zhan
- Institute of Preschool Education, Jinan Preschool Education College, Jinan City, People's Republic of China
| | - Lei Zhai
- Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Shandong First Medical University, Tai-an City, People's Republic of China
| | - Peng Jiao
- Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Shandong First Medical University, Tai-an City, People's Republic of China
| | - Shutong Yao
- Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Shandong First Medical University, Tai-an City, People's Republic of China
| | - Guohua Lu
- Experimental Center for Medical Research, Weifang Medical University, Weifang City, People's Republic of China
| | - Qingjie Mu
- Experimental Center for Medical Research, Weifang Medical University, Weifang City, People's Republic of China
| | - Juan Wang
- Department of Pharmaceutical Sciences, Binzhou Medical College, Yantai City, People's Republic of China
| | - Aihua Zhao
- Department of Emergency Medicine, the second Affiliated Hospital of Shandong First Medical University, Tai-an City, People's Republic of China
| | - Yadong Zhou
- Department of Emergency Medicine, the second Affiliated Hospital of Shandong First Medical University, Tai-an City, People's Republic of China
| | - Shucun Qin
- Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Shandong First Medical University, Tai-an City, People's Republic of China. .,Heart Center of Shandong First Medical University, Tai-an City, People's Republic of China.
| |
Collapse
|
15
|
Intravenous delivery of granulocyte-macrophage colony stimulating factor impairs survival in lipopolysaccharide-induced sepsis. PLoS One 2019; 14:e0218602. [PMID: 31220157 PMCID: PMC6586330 DOI: 10.1371/journal.pone.0218602] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 06/05/2019] [Indexed: 01/11/2023] Open
Abstract
Background Cell-based therapies with bone marrow-derived progenitor cells (BMDPC) lead to an improved clinical outcome in animal sepsis models. In the present study we evaluated the ability of granulocyte macrophage-colony stimulating factor (GM-CSF) to mobilize BMDPC in a lipopolysaccharide (LPS)-induced sepsis model and thereby its potential as a novel treatment strategy. Methods Male Wistar rats received LPS (25μg/kg/h for 4 days) intravenously and were subsequently treated with GM-CSF 12.5μg/kg (0h,24h,48h,72h). As control groups, rats were infused with sodium chloride or GM-CSF only. Clinical and laboratory parameters, proinflammatory plasma cytokines as well as BMDPC counts were analyzed. Cytokine release by isolated peripheral blood mononuclear cells from rat spleen upon incubation with LPS, GM-CSF and a combination of both were investigated in vitro. Results In vivo, rats receiving both LPS and GM-CSF, showed a reduced weight loss and increased mobilization of BMDPC. At the same time, this regime resulted in an increased release of proinflammatory cytokines (IL-6, IL-8) and a significantly increased mortality. In vitro, the combination of LPS and GM-CSF showed a significantly increased IL-6 release upon incubation compared to incubation with LPS or GM-CSF alone. Conclusions GM-CSF did not have a beneficial effect on the clinical course in our LPS-induced sepsis model. It synergistically promoted inflammation with LPS and probably thereby impaired survival.
Collapse
|
16
|
Aly H, Mansi Y, Ez El Din Z, Gabr Metwally H, Sabry A. Mesenchymal stromal cells and TGF-β1 in tracheal aspirate of premature infants: early predictors for bronchopulmonary dysplasia? J Perinat Med 2019; 47:470-477. [PMID: 30789824 DOI: 10.1515/jpm-2018-0305] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 01/10/2019] [Indexed: 11/15/2022]
Abstract
Background The pathogenesis of bronchopulmonary dysplasia (BPD) includes arrest of alveolar septation and enhanced fibrosis. We hypothesized that mesenchymal stromal cells (MSC) and transforming growth factor-β1 (TGF-β1) in tracheal aspirates of mechanically ventilated premature infants differ in BPD and non-BPD infants. Methods Tracheal aspirates were collected during the first week of life. Mononuclear cells were separated, cultured and immunophenotyped by flow cytometry. MSCs colony/cluster ratio was calculated as an index for dysplastic potentials. TGF-β1 was assessed by enzyme-linked immunosorbent assay (ELISA). Setting: Neonatal intensive care unit. Patients Premature infants at risk for BPD. Results A total of 121 preterm infants were enrolled; 27 of them died and among the 94 survivors 23 infants had BPD. MSCs were identified in younger [gestational age (GA): 30.9±1.7 vs. 31.8±1.8, P=0.025] and smaller [birth weight (BW): 1.3±0.28 vs. 1.44±0.37 kg, P=0.04] infants with lower Apgar scores. The recovery rate of MSCs in BPD and non-BPD groups did not differ. BPD group had significantly smaller colony/cluster ratio compared to non-BPD (0.97 vs. 4.25, P=0.002). TGF-β1 was significantly greater in BPD infants (4173.9±864.3 vs. 3705.8±540.5 pg/mL, P=0.021). Conclusion Infants with BPD had different MSCs morphology and greater TGF-β1 expression. The pathogenesis for these morphological changes of resident lung MSCs needs further studying.
Collapse
Affiliation(s)
- Hany Aly
- Department of Neonatology, Cleveland Clinic Children's Hospital, 9500 Euclid Avenue, M31-37, Cleveland, OH 44195, USA
| | - Yasmeen Mansi
- Division of Neonatology, Department of Pediatrics, Cairo University Children's Hospital, Cairo, Egypt
| | - Zahraa Ez El Din
- Division of Neonatology, Department of Pediatrics, Cairo University Children's Hospital, Cairo, Egypt
| | - Hala Gabr Metwally
- Division of Hematology, Department of Pediatrics, Cairo University Children's Hospital, Cairo, Egypt
| | - Amira Sabry
- Division of Neonatology, Department of Pediatrics, Cairo University Children's Hospital, Cairo, Egypt
| |
Collapse
|
17
|
Qiu Y, Chen C, Zhang J, Chen M, Gong H, Gong L, Du L, Wang R. VEGF attenuates lung injury by inducing homing of CD133+ progenitors via VEGFR1. Biochem Biophys Res Commun 2019; 511:650-657. [DOI: 10.1016/j.bbrc.2019.02.071] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 02/14/2019] [Indexed: 12/22/2022]
|
18
|
Sueblinvong V, Weiss DJ. Acute Lung Injury: Endothelial Progenitor Cells to the Rescue? Am J Med Sci 2019; 357:1-2. [PMID: 30446164 DOI: 10.1016/j.amjms.2018.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/01/2018] [Accepted: 10/04/2018] [Indexed: 11/18/2022]
Affiliation(s)
- Viranuj Sueblinvong
- Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University School of Medicine, Atlanta, Georgia.
| | - Daniel J Weiss
- Pulmonary and Critical Care Medicine, University of Vermont College of Medicine, Burlington, Vermont
| |
Collapse
|
19
|
Rafat N, Patry C, Sabet U, Viergutz T, Weiss C, Tönshoff B, Beck G, Schaible T. Endothelial Progenitor and Mesenchymal Stromal Cells in Newborns With Congenital Diaphragmatic Hernia Undergoing Extracorporeal Membrane Oxygenation. Front Pediatr 2019; 7:490. [PMID: 31824902 PMCID: PMC6882772 DOI: 10.3389/fped.2019.00490] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 11/07/2019] [Indexed: 02/03/2023] Open
Abstract
Background: Endothelial progenitor (EPC) and mesenchymal stromal cells (MSC) can regenerate damaged endothelium and thereby improve pulmonary endothelial dysfunction. We do not know, how extracorporeal membrane oxygenation (ECMO) might affect EPC- and MSC-mediated regenerative pathways in patients with congenital diaphragmatic hernia (CDH). Therefore, we investigated, if ECMO support impacts EPC and MSC numbers in CDH patients. Methods: Peripheral blood mononuclear cells from newborns with ECMO-dependent (n = 18) and ECMO-independent CDH (n = 12) and from healthy controls (n = 12) were isolated. The numbers of EPC and MSC were identified by flowcytometry. Serum levels of vascular endothelial growth factor (VEGF) and angiopoietin (Ang)-2 were determined. Results: EPC and MSC were elevated in newborns with CDH. ECMO-dependent infants had higher EPC subpopulation counts (2,1-7,6-fold) before treatment compared to ECMO-independent infants. In the disease course, EPC and MSC subpopulation counts in ECMO-dependent infants were lower than before ECMO initiation. During ECMO, VEGF serum levels were significantly reduced (by 90.5%) and Ang2 levels significantly increased (by 74.8%). Conclusions: Our data suggest that ECMO might be associated with a rather impaired mobilization of EPC and MSC and with a depression of VEGF serum levels in newborns with CDH.
Collapse
Affiliation(s)
- Neysan Rafat
- Department of Neonatology, University Children's Hospital Mannheim, University of Heidelberg, Mannheim, Germany.,Department of Pediatrics I, University Children's Hospital Heidelberg, Heidelberg, Germany.,Department of Pharmaceutical Sciences, Bahá'í Institute of Higher Education (BIHE), Teheran, Iran
| | - Christian Patry
- Department of Pediatrics I, University Children's Hospital Heidelberg, Heidelberg, Germany
| | - Ursula Sabet
- Department of Pediatrics I, University Children's Hospital Heidelberg, Heidelberg, Germany
| | - Tim Viergutz
- Department of Anesthesiology and Critical Care Medicine, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Christel Weiss
- Department for Medical Statistics and Biomathematics, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Burkhard Tönshoff
- Department of Pediatrics I, University Children's Hospital Heidelberg, Heidelberg, Germany
| | - Grietje Beck
- Department of Anesthesiology, Helios Dr. Horst-Schmidt Clinic, Wiesbaden, Germany
| | - Thomas Schaible
- Department of Neonatology, University Children's Hospital Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
20
|
Nejad-Moghaddam A, Tahmasbpour E, Sohrabiyan M, Jafari H, Ghanei M. Stem cells therapy: a review on approaches that can be used for treatment of respiratory failures in sulfur mustard-injured patients. Immunopharmacol Immunotoxicol 2018; 40:359-367. [PMID: 30488735 DOI: 10.1080/08923973.2018.1510961] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 07/06/2018] [Indexed: 12/28/2022]
Abstract
Sulfur mustard (SM) is a toxic agent which causes severe abnormalities in an airway system such as necrosis and inflammation, oxidative stress, chronic bronchitis, shortness of breath, and chronic obstructive pulmonary disease. Although possible mechanisms of SM toxicity have been extensively considered, there is still need to find an appropriate clinical treatment to decrease chronic lung injuries caused by SM. Due to extensive progresses and achievement in tissue repairing through stem cells therapy, the importance of cell therapy for the treatment of lung injuries has been increased. However, several factors such as types of stem cells, necessary conditions for growth and proliferation of stem cells, and their homing into the target tissues are considered as the most important problems in this issue. Mesenchymal stem cells (MSCs) are a class of multipotent stem cells with proliferative and self-renewal capacity which are able to differentiate into different cell lines such as lung epithelial cells. They have a potential repairing and immune modulatory properties which make them as a good candidate for the regeneration of bronchioles tract in SM-exposed patients. Unlike chemical drugs, the differentiation and high-level safety properties of MSCs can be considered as a new strategy for the treatment of SM-injured patients with pulmonary complications. This review aims to consider the therapeutic effects of MSCs in the treatment of SM-induced pulmonary injuries in both animals and humans.
Collapse
Affiliation(s)
- Amir Nejad-Moghaddam
- a Marine Medicine Research Center , Baqiyatallah University of Medical Sciences , Tehran , Iran
| | - Eisa Tahmasbpour
- b Laboratory of Regenerative Medicine & Biomedical Innovations , Pasteur Institute of Iran , Tehran , Iran
| | - Milad Sohrabiyan
- c Chemical Injuries Research Center, Systems Biology and Poisonings Institute , Baqiyatallah University of Medical Sciences , Tehran , Iran
| | - Hosein Jafari
- a Marine Medicine Research Center , Baqiyatallah University of Medical Sciences , Tehran , Iran
| | - Mostafa Ghanei
- c Chemical Injuries Research Center, Systems Biology and Poisonings Institute , Baqiyatallah University of Medical Sciences , Tehran , Iran
| |
Collapse
|
21
|
Horie S, Gonzalez HE, Laffey JG, Masterson CH. Cell therapy in acute respiratory distress syndrome. J Thorac Dis 2018; 10:5607-5620. [PMID: 30416812 DOI: 10.21037/jtd.2018.08.28] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is driven by a severe pro-inflammatory response resulting in lung damage, impaired gas exchange and severe respiratory failure. There is no specific treatment that effectively improves outcome in ARDS. However, in recent years, cell therapy has shown great promise in preclinical ARDS studies. A wide range of cells have been identified as potential candidates for use, among these are mesenchymal stromal cells (MSCs), which are adult multi-lineage cells that can modulate the immune response and enhance repair of damaged tissue. The therapeutic potential of MSC therapy for sepsis and ARDS has been demonstrated in multiple in vivo models. The therapeutic effect of these cells seems to be due to two different mechanisms; direct cellular interaction, and paracrine release of different soluble products such as extracellular vesicles (EVs)/exosomes. Different approaches have also been studied to enhance the therapeutic effect of these cells, such as the over-expression of anti-inflammatory or pro-reparative molecules. Several clinical trials (phase I and II) have already shown safety of MSCs in ARDS and other diseases. However, several translational issues still need to be addressed, such as the large-scale production of cells, and their potentiality and variability, before the therapeutic potential of stem cells therapies can be realized.
Collapse
Affiliation(s)
- Shahd Horie
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland
| | - Hector Esteban Gonzalez
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland
| | - John G Laffey
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland.,Department of Anesthesia and Intensive Care Medicine, Galway University Hospitals, SAOLTA Hospital Group, Ireland
| | - Claire H Masterson
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
22
|
Abstract
This study investigated the effects of glutamine (GLN) administration on circulating endothelial progenitor cells (EPCs) and lung angiopoietin (Ang) gene expressions in polymicrobial sepsis. Mice were randomly assigned to a normal control group (NC), septic saline group (SS), and septic GLN group (SG). All mice were fed with a chow diet. Sepsis was induced by cecal ligation and puncture (CLP). The mice in SS group were injected with saline, whereas SG group administered 0.75 g GLN/kg body weight once via tail vein 1 h after CLP. Mice were killed 24 and 48 h after CLP. Their blood and lungs were collected for further analysis. The results showed that, compared with normal mice, sepsis resulted in higher C-X-C motif chemokine-12, vascular endothelial growth factor, nitric oxide levels, and a higher circulating EPC percentage. In addition, inflammatory cytokine concentrations and Ang-2 gene expression were upregulated in lung tissues. GLN administration enhanced the mobilization of EPC, and downregulated inflammatory cytokine production and the Ang-2 gene expressions in lungs. Histopathological findings showed that the extent of inflammatory lesions of the lung alveolar was less severe in the SG group than the SS group after CLP. Our results suggest that a single dose of intravenous GLN administration after initiation of sepsis promotes the mobilization of circulating EPC, and modulates the balance of Ang-Tie2 axis that may improve the vascular function, ameliorate inflammation, and protect lung injury against polymicrobial sepsis.
Collapse
|
23
|
Dhindsa DS, Khambhati J, Sandesara PB, Eapen DJ, Quyyumi AA. Biomarkers to Predict Cardiovascular Death. Card Electrophysiol Clin 2017; 9:651-664. [PMID: 29173408 DOI: 10.1016/j.ccep.2017.07.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
This article reviews biomarkers that have been shown to identify subjects at increased risk for cardiovascular death within the general population, in those with established coronary artery disease, and in those with heart failure. Use of biomarkers for risk stratification for sudden cardiac death continues to evolve. It seems that a multimarker strategy for risk stratification using simple measures of circulating proteins and usual clinical risk factors, particularly in patients with known coronary artery disease, can be used to identify patients at near-term risk of death. Whether similar strategies in the general population will prove to be cost-effective needs to be investigated.
Collapse
Affiliation(s)
- Devinder S Dhindsa
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road Northeast, Suite 507, Atlanta, GA 30322, USA
| | - Jay Khambhati
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road Northeast, Suite 507, Atlanta, GA 30322, USA
| | - Pratik B Sandesara
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road Northeast, Suite 507, Atlanta, GA 30322, USA
| | - Danny J Eapen
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road Northeast, Suite 507, Atlanta, GA 30322, USA
| | - Arshed A Quyyumi
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 1462 Clifton Road Northeast, Suite 507, Atlanta, GA 30322, USA.
| |
Collapse
|
24
|
Gazdhar A, Ravikumar P, Pastor J, Heller M, Ye J, Zhang J, Moe OW, Geiser T, Hsia CCW. Alpha-Klotho Enrichment in Induced Pluripotent Stem Cell Secretome Contributes to Antioxidative Protection in Acute Lung Injury. Stem Cells 2017; 36:616-625. [PMID: 29226550 DOI: 10.1002/stem.2752] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 11/07/2017] [Accepted: 11/21/2017] [Indexed: 02/06/2023]
Abstract
Induced pluripotent stem cells (iPSCs) have been reported to alleviate organ injury, although the mechanisms of action remain unclear and administration of intact cells faces many limitations. We hypothesized that cell-free conditioned media (CM) containing the secretome of iPSCs possess antioxidative constituents that can alleviate pulmonary oxidant stress damage. We derived iPSCs from human dermal fibroblasts and harvested the CM. Addition of iPSC CM to cultured human alveolar type-1 epithelial cells mitigated hyperoxia-induced depletion of endogenous total antioxidant capacity while tracheal instillation of iPSC CM into adult rat lungs enhanced hyperoxia-induced increase in TAC. In both the in vitro and in vivo models, iPSC CM ameliorated oxidative damage to DNA, lipid, and protein, and activated the nuclear factor (erythroid 2)-related factor 2 (Nrf2) network of endogenous antioxidant proteins. Compared with control fibroblast-conditioned or cell-free media, iPSC CM is highly enriched with αKlotho at a concentration up to more than 10-fold of that in normal serum. αKlotho is an essential antioxidative cell maintenance and protective factor and an activator of the Nrf2 network. Immunodepletion of αKlotho reduced iPSC CM-mediated cytoprotection by ∼50%. Thus, the abundant αKlotho content significantly contributes to iPSC-mediated antioxidation and cytoprotection. Results uncover a major mechanism of iPSC action, suggest a fundamental role of αKlotho in iPSC maintenance, and support the translational potential of airway delivery of cell-free iPSC secretome for protection against lung injury. The targeted cell-free secretome-based approach may also be applicable to the amelioration of injury in other organs. Stem Cells 2018;36:616-625.
Collapse
Affiliation(s)
- Amiq Gazdhar
- Department of Pulmonary Medicine, University Hospital, Bern, Switzerland.,Department of Clinical Research, University Hospital, Bern, Switzerland
| | - Priya Ravikumar
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Johanne Pastor
- Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Manfred Heller
- Department of Clinical Research, University Hospital, Bern, Switzerland
| | - Jianfeng Ye
- Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jianning Zhang
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Orson W Moe
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Charles and Jane Pak Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Thomas Geiser
- Department of Pulmonary Medicine, University Hospital, Bern, Switzerland.,Department of Clinical Research, University Hospital, Bern, Switzerland
| | - Connie C W Hsia
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
25
|
Wang Y, Liu HN, Zhen Z, Yiu KH, Tse HF, Pelekos G, Tonetti M, Jin L. Periodontal treatment modulates gene expression of endothelial progenitor cells in diabetic patients. J Clin Periodontol 2017; 44:1253-1263. [DOI: 10.1111/jcpe.12806] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/18/2017] [Indexed: 11/28/2022]
Affiliation(s)
- Yi Wang
- Discipline of Periodontology; Faculty of Dentistry; The University of Hong Kong; Hong Kong SAR China
| | - Hin Nam Liu
- Discipline of Periodontology; Faculty of Dentistry; The University of Hong Kong; Hong Kong SAR China
| | - Zhe Zhen
- Division of Cardiology; Department of Medicine; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong SAR China
| | - Kai Hang Yiu
- Division of Cardiology; Department of Medicine; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong SAR China
| | - Hung Fat Tse
- Division of Cardiology; Department of Medicine; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong SAR China
| | - George Pelekos
- Discipline of Periodontology; Faculty of Dentistry; The University of Hong Kong; Hong Kong SAR China
| | - Maurizio Tonetti
- Discipline of Periodontology; Faculty of Dentistry; The University of Hong Kong; Hong Kong SAR China
| | - Lijian Jin
- Discipline of Periodontology; Faculty of Dentistry; The University of Hong Kong; Hong Kong SAR China
| |
Collapse
|
26
|
Yu M, Wang C, Zeng G, Zeng G, Zhou L, Chen T, Tan X, Wang Y. Toll‑like receptor 4 is expressed and functional in late endothelial progenitor cells. Mol Med Rep 2017; 16:5549-5554. [PMID: 28849058 DOI: 10.3892/mmr.2017.7291] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 07/07/2017] [Indexed: 02/05/2023] Open
Abstract
It has been previously demonstrated that lipopolysaccharides (LPS) inhibit the viability, migration, adhesion and in vitro angiogenesis of late endothelial progenitor cells (EPCs). However, the mechanisms underlying this LPS‑induced impairment of late EPC functional activity are unknown. The aim of the present study was to investigate whether Toll‑like receptor 4 (TLR4) is expressed and functional on late EPCs, using late EPCs of 3‑5 passages. Cells were deprived of serum for 24 h prior to experiments and incubated with 10 µg/ml LPS for 24 h with or without pretreatment with 2 µg/ml TLR4 signaling inhibitor CLI‑095 for 30 min. The viability, migration, adhesion and in vitro angiogenesis, as well as the expression of silent information regulator 1 (SIRT1), in late EPCs were evaluated. Treatment with 10 µg/ml LPS decreased the viability, migration and adhesion abilities, and in vitro angiogenesis of late EPCs. Pretreatment with the TLR4 signaling inhibitor reversed this LPS‑induced dysfunction of late EPCs. LPS downregulated the expression of SIRT1 protein, however, blocking TLR4 attenuated the effect of LPS on SIRT1 expression. Therefore, the results of the present study indicate that LPS impaired the functional activity of late EPCs via TLR4, which may be associated with decreased SIRT1 expression.
Collapse
Affiliation(s)
- Min Yu
- Department of Cardiology, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Cantian Wang
- Department of Cardiology, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Guoning Zeng
- Department of Cardiology, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Guoning Zeng
- Department of Cardiology, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Lihong Zhou
- Department of Cardiology, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Tingting Chen
- Department of Cardiology, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Xuerui Tan
- Department of Cardiology, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Yanping Wang
- Department of Cardiology, The First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| |
Collapse
|
27
|
Inhibition of Murine Pulmonary Microvascular Endothelial Cell Apoptosis Promotes Recovery of Barrier Function under Septic Conditions. Mediators Inflamm 2017; 2017:3415380. [PMID: 28250575 PMCID: PMC5303866 DOI: 10.1155/2017/3415380] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 10/25/2016] [Accepted: 12/20/2016] [Indexed: 12/13/2022] Open
Abstract
Sepsis is characterized by injury of the pulmonary microvasculature and the pulmonary microvascular endothelial cells (PMVEC), leading to barrier dysfunction and acute respiratory distress syndrome (ARDS). Our recent work identified a strong correlation between PMVEC apoptosis and microvascular leak in septic mice in vivo, but the specific role of apoptosis in septic PMVEC barrier dysfunction remains unclear. Thus, we hypothesize that PMVEC apoptosis is likely required for PMVEC barrier dysfunction under septic conditions in vitro. Septic stimulation (mixture of tumour necrosis factor α, interleukin 1β, and interferon γ [cytomix]) of isolated murine PMVEC resulted in a significant loss of barrier function as early as 4 h after stimulation, which persisted until 24 h. PMVEC apoptosis, as reflected by caspase activation, DNA fragmentation, and loss of membrane polarity, was first apparent at 8 h after cytomix. Pretreatment of PMVEC with the pan-caspase inhibitor Q-VD significantly decreased septic PMVEC apoptosis and was associated with reestablishment of PMVEC barrier function at 16 and 24 h after stimulation but had no effect on septic PMVEC barrier dysfunction over the first 8 h. Collectively, our data suggest that early septic murine PMVEC barrier dysfunction driven by proinflammatory cytokines is not mediated through apoptosis, but PMVEC apoptosis contributes to late septic PMVEC barrier dysfunction.
Collapse
|
28
|
Gumina DL, Black CP, Balasubramaniam V, Winn VD, Baker CD. Umbilical Cord Blood Circulating Progenitor Cells and Endothelial Colony-Forming Cells Are Decreased in Preeclampsia. Reprod Sci 2016; 24:1088-1096. [PMID: 27879452 DOI: 10.1177/1933719116678692] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Preeclampsia (PE) is a pregnancy-specific disease characterized by the new onset of hypertension and proteinuria. Mothers with PE are known to develop endothelial dysfunction, but its effect on infants has been understudied, as newborns are often asymptomatic. Recent studies indicate that infants born from preeclamptic pregnancies develop endothelial dysfunction including higher blood pressure during childhood and an increased risk of stroke later in life. We hypothesize that PE reduces the number and function of fetal angiogenic progenitor cells and may contribute to this increased risk. We quantified 2 distinct types of angiogenic progenitors, pro-angiogenic circulating progenitor cells (CPCs) and endothelial colony-forming cells (ECFCs), from the umbilical cord blood of preeclamptic pregnancies and normotensive controls. Pro-angiogenic and nonangiogenic CPCs were enumerated via flow cytometry and ECFCs by cell culture. Additionally, we studied the growth, migration, and tube formation of ECFCs from PE and gestational age-matched normotensive control pregnancies. We found that PE resulted in decreased cord blood pro-angiogenic CPCs and ECFCs. Nonangiogenic CPCs were also decreased. Preeclamptic ECFCs demonstrated decreased growth and migration but formed tube-like structures in vitro similar to controls. Our results suggest that the preeclamptic environment alters the number and function of angiogenic progenitor cells and may increase the risk of later vascular disease.
Collapse
Affiliation(s)
- Diane L Gumina
- 1 Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Claudine P Black
- 2 Department of Pediatrics, Pediatric Heart Lung Center, University of Colorado School of Medicine, Aurora, CO, USA
| | - Vivek Balasubramaniam
- 3 Pediatric Pulmonology, Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Virginia D Winn
- 1 Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
- 4 Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
- Authors contributed equally to the article
| | - Christopher D Baker
- 2 Department of Pediatrics, Pediatric Heart Lung Center, University of Colorado School of Medicine, Aurora, CO, USA
- Authors contributed equally to the article
| |
Collapse
|
29
|
Siavashi V, Asadian S, Taheri-Asl M, Babaei H, Keshavarz S, Bazaei M, Nassiri SM. The Improvement of Respiratory Performance After Phototherapy-Induced EPC Mobilization in Preterm Infants With RDS. J Cell Biochem 2016; 118:594-604. [DOI: 10.1002/jcb.25745] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 09/19/2016] [Indexed: 01/03/2023]
Affiliation(s)
- Vahid Siavashi
- Department of Clinical Pathology; Faculty of Veterinary Medicine; University of Tehran; Tehran Iran
| | - Simin Asadian
- Imam Reza Hospitals; Kermanshah University of Medical Sciences; Kermanshah Iran
| | - Masoud Taheri-Asl
- Departments of Radiology; Faculty of Paramedicine; AJA University of Medical Sciences; Tehran Iran
| | - Homa Babaei
- Department of Pediatrics; Imam Reza Hospital; Kermanshah University of Medical Science; Kermanshah Iran
| | | | - Mohammad Bazaei
- Department of Clinical Pathology; Faculty of Veterinary Medicine; University of Tehran; Tehran Iran
| | - Seyed Mahdi Nassiri
- Department of Clinical Pathology; Faculty of Veterinary Medicine; University of Tehran; Tehran Iran
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW Acute respiratory distress syndrome (ARDS) is a devastating disease process with a 40% mortality rate, and for which there is no therapy. Stem cells are an exciting potential therapy for ARDS, and are currently the subject of intensive ongoing research efforts. We review data concerning the therapeutic promise of cell-based therapies for ARDS. RECENT FINDINGS Recent experimental studies suggest that cell-based therapies, particularly mesenchymal stem/stromal cells (MSCs), endothelial progenitor cells, and embryonic or induced pluripotent stem cells all offer considerable promise for ARDS. Of these cell types, mesenchymal stromal cells offer the greatest potential for allogeneic therapy, given the large body of preclinical data supporting their use, and the advanced state of our understanding of their diverse mechanisms of action. Although other stem cells such as EPCs also have therapeutic potential, greater barriers exist, particularly the requirement for autologous EPC therapy. Other stem cells, such as ESCs and iPSCs, are at an earlier stage in the translational process, but offer the hope of directly replacing injured lung tissue. Ultimately, lung-derived stem cells may offer the greatest hope for lung diseases, given their homeostatic role in replacing and repairing damaged native lung tissues.MSCs are currently in early phase clinical trials, the results of which will be of critical importance to subsequent translational efforts for MSCs in ARDS. A number of translational challenges exist, including minimizing variability in cell batches, developing standard tests for cell potency, and producing large amounts of clinical-grade cells for use in patients. SUMMARY Cell-based therapies, particularly MSCs, offer considerable promise for the treatment of ARDS. Overcoming translational challenges will be important to fully realizing their therapeutic potential for ARDS.
Collapse
|
31
|
Yen YT, Roan JN, Fang SY, Chang SW, Tseng YL, Lam CF. Autologous endothelial progenitor cells improve allograft survival in porcine lung transplantation with prolonged ischemia. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:277. [PMID: 27570771 DOI: 10.21037/atm.2016.06.22] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND As endothelial progenitor cells (EPCs) attenuated acute lung injury (ALI) in rabbit model, we hypothesized that autologous EPCs preserved lung graft function during the acute reperfusion period of lung transplantation and tested the therapeutic potential of EPCs in a porcine model of lung transplantation with prolonged graft ischemia. METHODS Day-7 EPCs isolated from the recipient subjects or plain culture media were administered into the left pulmonary artery immediately before restoration of pulmonary blood flow in a porcine lung allotransplantation model, with the transplantation surgeons blinded to the content of injection. Hemodynamics and arterial blood gas were recorded, and the right pulmonary artery was occluded 30 min after reperfusion to evaluate the lung graft function. The lung grafts were sectioned for histological examination at the end of experiments. The total ischemic time for lung graft was approximately 14 h. RESULTS All animals receiving plain medium died within 40 min after reperfusion, but 3 out of 5 (60%) piglets receiving EPCs survived up to 4 h after diversion of the entire cardiac output into the lung graft (P<0.01). The donor body weight, recipient body weight, cold ischemic time, and time for anastomosis were comparable between the EPC and control group (P=0.989, 0.822, 0.843, and 0.452, respectively). The mean aortic pressure decreased, and the cardiac output and mean pulmonary artery pressure elevated after right pulmonary artery occlusion. All these parameters were gradually compensated in the EPC group but decompensated in the control group. Better preservation of gas exchange function, reduced thrombi formation in the terminal pulmonary arterioles, and attenuated interstitial hemorrhage of the lung graft were observed in the EPC group. CONCLUSIONS We concluded autologous EPCs significantly enhanced the function of lung allograft and improved survival in a porcine model of lung transplantation with prolonged ischemia.
Collapse
Affiliation(s)
- Yi-Ting Yen
- Division of Thoracic Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medical College, National Cheng Kung University, Tainan, Taiwan;; Institube of Clinical Medicine, College of Medical College, National Cheng Kung University, Tainan, Taiwan
| | - Jun-Neng Roan
- Institube of Clinical Medicine, College of Medical College, National Cheng Kung University, Tainan, Taiwan;; Division of Cardiovascular Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medical College, National Cheng Kung University, Tainan, Taiwan
| | - Shih-Yuan Fang
- Department of Anesthesiology, National Cheng Kung University Hospital, College of Medical College, National Cheng Kung University, Tainan, Taiwan
| | - Shi-Wei Chang
- Department of Anesthesiology, National Cheng Kung University Hospital, College of Medical College, National Cheng Kung University, Tainan, Taiwan
| | - Yau-Lin Tseng
- Division of Thoracic Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medical College, National Cheng Kung University, Tainan, Taiwan
| | - Chen-Fuh Lam
- Department of Anesthesiology, National Cheng Kung University Hospital, College of Medical College, National Cheng Kung University, Tainan, Taiwan;; Department of Anesthesiology, Buddhist Tzu Chi General Hospital and Tzu Chi University School of Medicine, Hualien, Taiwan
| |
Collapse
|
32
|
Müller-Redetzky H, Lienau J, Suttorp N, Witzenrath M. Therapeutic strategies in pneumonia: going beyond antibiotics. Eur Respir Rev 2016; 24:516-24. [PMID: 26324814 DOI: 10.1183/16000617.0034-2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Dysregulation of the innate immune system drives lung injury and its systemic sequelae due to breakdown of vascular barrier function, harmful hyperinflammation and microcirculatory failure, which contribute to the unfavourable outcome of patients with severe pneumonia. A variety of promising therapeutic targets have been identified and numerous innovative therapeutic approaches demonstrated to improve lung injury in experimental preclinical studies. However, at present specific preventive or curative strategies for the treatment of lung failure in pneumonia in addition to antibiotics are still missing. The aim of this mini-review is to give a short overview of some, but not all, adjuvant therapeutic strategies for pneumonia and its most important complications, sepsis and acute respiratory distress syndrome, and briefly discuss future perspectives.
Collapse
Affiliation(s)
- Holger Müller-Redetzky
- Dept of Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jasmin Lienau
- Dept of Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Norbert Suttorp
- Dept of Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Martin Witzenrath
- Dept of Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
33
|
Sekine A, Nishiwaki T, Nishimura R, Kawasaki T, Urushibara T, Suda R, Suzuki T, Takayanagi S, Terada J, Sakao S, Tada Y, Iwama A, Tatsumi K. Prominin-1/CD133 expression as potential tissue-resident vascular endothelial progenitor cells in the pulmonary circulation. Am J Physiol Lung Cell Mol Physiol 2016; 310:L1130-42. [PMID: 27059286 DOI: 10.1152/ajplung.00375.2014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 03/31/2016] [Indexed: 11/22/2022] Open
Abstract
Pulmonary vascular endothelial cells could contribute to maintain homeostasis in adult lung vasculature. "Tissue-resident" endothelial progenitor cells (EPCs) play pivotal roles in postnatal vasculogenesis, vascular repair, and tissue regeneration; however, their local pulmonary counterparts remain to be defined. To determine whether prominin-1/CD133 expression can be a marker of tissue-resident vascular EPCs in the pulmonary circulation, we examined the origin and characteristics of prominin-1/CD133-positive (Prom1(+)) PVECs considering cell cycle status, viability, histological distribution, and association with pulmonary vascular remodeling. Prom1(+) PVECs exhibited high steady-state transit through the cell cycle compared with Prom1(-) PVECs and exhibited homeostatic cell division as assessed using the label dilution method and mice expressing green fluorescent protein. In addition, Prom1(+) PVECs showed more marked expression of putative EPC markers and drug resistance genes as well as highly increased activation of aldehyde dehydrogenase compared with Prom1(-) PVECs. Bone marrow reconstitution demonstrated that tissue-resident cells were the source of >98% of Prom1(+) PVECs. Immunofluorescence analyses revealed that Prom1(+) PVECs preferentially resided in the arterial vasculature, including the resistant vessels of the lung. The number of Prom1(+) PVECs was higher in developing postnatal lungs. Sorted Prom1(+) PVECs gave rise to colonies and formed fine vascular networks compared with Prom1(-) PVECs. Moreover, Prom1(+) PVECs increased in the monocrotaline and the Su-5416 + hypoxia experimental models of pulmonary vascular remodeling. Our findings indicated that Prom1(+) PVECs exhibited the phenotype of tissue-resident EPCs. The unique biological characteristics of Prom1(+) PVECs predominantly contribute to neovasculogenesis and maintenance of homeostasis in pulmonary vascular tissues.
Collapse
Affiliation(s)
- Ayumi Sekine
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan; and
| | - Tetsu Nishiwaki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan; and
| | - Rintaro Nishimura
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan; and
| | - Takeshi Kawasaki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan; and
| | - Takashi Urushibara
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan; and
| | - Rika Suda
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan; and
| | - Toshio Suzuki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan; and
| | - Shin Takayanagi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan; and
| | - Jiro Terada
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan; and
| | - Seiichiro Sakao
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan; and
| | - Yuji Tada
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan; and
| | - Atsushi Iwama
- Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan; and
| |
Collapse
|
34
|
Suzuki T, Tada Y, Nishimura R, Kawasaki T, Sekine A, Urushibara T, Kato F, Kinoshita T, Ikari J, West J, Tatsumi K. Endothelial-to-mesenchymal transition in lipopolysaccharide-induced acute lung injury drives a progenitor cell-like phenotype. Am J Physiol Lung Cell Mol Physiol 2016; 310:L1185-98. [PMID: 27106288 DOI: 10.1152/ajplung.00074.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 04/17/2016] [Indexed: 12/15/2022] Open
Abstract
Pulmonary vascular endothelial function may be impaired by oxidative stress in endotoxemia-derived acute lung injury. Growing evidence suggests that endothelial-to-mesenchymal transition (EndMT) could play a pivotal role in various respiratory diseases; however, it remains unclear whether EndMT participates in the injury/repair process of septic acute lung injury. Here, we analyzed lipopolysaccharide (LPS)-treated mice whose total number of pulmonary vascular endothelial cells (PVECs) transiently decreased after production of reactive oxygen species (ROS), while the population of EndMT-PVECs significantly increased. NAD(P)H oxidase inhibition suppressed EndMT of PVECs. Most EndMT-PVECs derived from tissue-resident cells, not from bone marrow, as assessed by mice with chimeric bone marrow. Bromodeoxyuridine-incorporation assays revealed higher proliferation of capillary EndMT-PVECs. In addition, EndMT-PVECs strongly expressed c-kit and CD133. LPS loading to human lung microvascular endothelial cells (HMVEC-Ls) induced reversible EndMT, as evidenced by phenotypic recovery observed after removal of LPS. LPS-induced EndMT-HMVEC-Ls had increased vasculogenic ability, aldehyde dehydrogenase activity, and expression of drug resistance genes, which are also fundamental properties of progenitor cells. Taken together, our results demonstrate that LPS induces EndMT of tissue-resident PVECs during the early phase of acute lung injury, partly mediated by ROS, contributing to increased proliferation of PVECs.
Collapse
Affiliation(s)
- Toshio Suzuki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan; and
| | - Yuji Tada
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan; and
| | - Rintaro Nishimura
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan; and
| | - Takeshi Kawasaki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan; and
| | - Ayumi Sekine
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan; and
| | - Takashi Urushibara
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan; and
| | - Fumiaki Kato
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan; and
| | - Taku Kinoshita
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan; and
| | - Jun Ikari
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan; and
| | - James West
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan; and
| |
Collapse
|
35
|
Fujinaga H, Fujinaga H, Watanabe N, Kato T, Tamano M, Terao M, Takada S, Ito Y, Umezawa A, Kuroda M. Cord blood-derived endothelial colony-forming cell function is disrupted in congenital diaphragmatic hernia. Am J Physiol Lung Cell Mol Physiol 2016; 310:L1143-54. [PMID: 27130531 DOI: 10.1152/ajplung.00357.2015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 04/22/2016] [Indexed: 01/07/2023] Open
Abstract
Vascular growth is necessary for normal lung development. Although endothelial progenitor cells (EPCs) play an important role in vascularization, little is known about EPC function in congenital diaphragmatic hernia (CDH), a severe neonatal condition that is associated with pulmonary hypoplasia. We hypothesized that the function of endothelial colony-forming cells (ECFCs), a type of EPC, is impaired in CDH. Cord blood (CB) was collected from full-term CDH patients and healthy controls. We assessed CB progenitor cell populations as well as plasma vascular endothelial growth factor (VEGF) and stromal cell-derived factor 1α (SDF1α) levels. CB ECFC clonogenicity; growth kinetics; migration; production of VEGF, SDF1α, and nitric oxide (NO); vasculogenic capacity; and mRNA expression of VEGF-A, fms-related tyrosine kinase 1 (FLT1), kinase insert domain receptor (KDR), nitric oxide synthase (NOS) 1-3, SDF1, and chemokine (C-X-C motif) receptor 4 (CXCR4) were also assessed. Compared with controls, CB ECFCs were decreased in CDH. CDH ECFCs had reduced potential for self-renewal, clonogenicity, proliferation, and migration. Their capacity for NO production was enhanced but their response to VEGF was blunted in CDH ECFCs. In vivo potential for de novo vasculogenesis was reduced in CDH ECFCs. There was no difference in CB plasma VEGF and SDF1α concentrations, VEGF and SDF1α production by ECFCs, and ECFC mRNA expression of VEGF-A, FLT1, KDR, NOS1-3, SDF1, and CXCR4 between CDH and control subjects. In conclusion, CB ECFC function is disrupted in CDH, but these changes may be caused by mechanisms other than alteration of VEGF-NO and SDF1-CXCR4 signaling.
Collapse
Affiliation(s)
- Hideshi Fujinaga
- Department of Molecular Pathology, Tokyo Medical University, Tokyo, Japan; Department of Reproductive Biology, National Institute for Child Health and Development, Tokyo, Japan; Division of Neonatology, Center for Maternal-Fetal and Neonatal Medicine, National Center for Child Health and Development, Tokyo, Japan;
| | - Hiroko Fujinaga
- Department of Reproductive Biology, National Institute for Child Health and Development, Tokyo, Japan
| | - Nobuyuki Watanabe
- Department of Human Genetics, National Institute for Child Health and Development, Tokyo, Japan; and
| | - Tomoko Kato
- Department of Systems BioMedicine, National Institute for Child Health and Development, Tokyo, Japan
| | - Moe Tamano
- Department of Systems BioMedicine, National Institute for Child Health and Development, Tokyo, Japan
| | - Miho Terao
- Department of Systems BioMedicine, National Institute for Child Health and Development, Tokyo, Japan
| | - Shuji Takada
- Department of Systems BioMedicine, National Institute for Child Health and Development, Tokyo, Japan
| | - Yushi Ito
- Division of Neonatology, Center for Maternal-Fetal and Neonatal Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Akihiro Umezawa
- Department of Reproductive Biology, National Institute for Child Health and Development, Tokyo, Japan
| | - Masahiko Kuroda
- Department of Molecular Pathology, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
36
|
Millar FR, Summers C, Griffiths MJ, Toshner MR, Proudfoot AG. The pulmonary endothelium in acute respiratory distress syndrome: insights and therapeutic opportunities. Thorax 2016; 71:462-73. [DOI: 10.1136/thoraxjnl-2015-207461] [Citation(s) in RCA: 162] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 02/12/2016] [Indexed: 01/23/2023]
|
37
|
Lu CL, Leu JG, Liu WC, Zheng CM, Lin YF, Shyu JF, Wu CC, Lu KC. Endothelial Progenitor Cells Predict Long-Term Mortality in Hemodialysis Patients. Int J Med Sci 2016; 13:240-247. [PMID: 26941585 PMCID: PMC4773289 DOI: 10.7150/ijms.14209] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 01/22/2016] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND The endothelial progenitor cells (EPCs) dysfunction is a critical event in the initiation of atherosclerotic plaque development and the level of circulating EPCs can be considered a biomarker of cardiovascular events. The level and functional change in EPCs has been investigated in hemodialysis patients, but the effect of absolute number of EPCs on risk of death has not yet been explored. We hypothesized that the number of EPCs predicted death from cardiovascular and all-cause mortality in hemodialysis patients. METHODS We evaluate the association between endothelial progenitor cells and clinical outcome in 154 patients on maintenance hemodialysis. The blood sample was drawn at the time of patient enrollment and EPCs were identified by flow cytometry using triple staining for CD34/CD133/KDR. RESULTS The median duration of follow-up was 4.19 years. There were 79 (51.3%) deaths during the follow-up period, 41 of whom died due to a confirmed cardiovascular cause. The cumulative survival was greater in the high-EPC group than the low-EPC group for all-cause and cardiovascular mortality. Decreased EPCs levels were associated with a significant increase in the risk of cardiovascular and all-cause mortality after adjusting for age, gender, current smokers, diabetes mellitus, and hypertension. CONCLUSIONS The level of circulating EPCs independently predicts the clinical outcome in patients on maintenance hemodialysis. Thus, the EPCs levels may be a useful predictive tool for evaluating the risk of death in maintenance hemodialysis patients.
Collapse
Affiliation(s)
- Chien-Lin Lu
- 1. Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- 3. Division of Nephrology, Department of Medicine, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Jyh-Gang Leu
- 2. School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei, Taiwan
- 3. Division of Nephrology, Department of Medicine, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Wen-Chih Liu
- 1. Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- 4. Division of Nephrology, Department of Internal Medicine, Yonghe Cardinal Tien Hospital, New Taipei City, Taiwan
| | - Cai-Mei Zheng
- 1. Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- 5. Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taiwan
- 6. Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University
| | - Yuh-Feng Lin
- 1. Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- 5. Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taiwan
| | - Jia-Fwu Shyu
- 7. Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Chia-Chao Wu
- 8. Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Kuo-Cheng Lu
- 9. Department of Medicine, Cardinal Tien Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| |
Collapse
|
38
|
Flamini V, Jiang WG, Lane J, Cui YX. Significance and therapeutic implications of endothelial progenitor cells in angiogenic-mediated tumour metastasis. Crit Rev Oncol Hematol 2016; 100:177-89. [PMID: 26917455 DOI: 10.1016/j.critrevonc.2016.02.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 11/30/2015] [Accepted: 02/15/2016] [Indexed: 01/16/2023] Open
Abstract
Cancer conveys profound social and economic consequences throughout the world. Metastasis is responsible for approximately 90% of cancer-associated mortality and, when it occurs, cancer becomes almost incurable. During metastatic dissemination, cancer cells pass through a series of complex steps including the establishment of tumour-associated angiogenesis. The human endothelial progenitor cells (hEPCs) are a cell population derived from the bone marrow which are required for endothelial tubulogenesis and neovascularization. They also express abundant inflammatory cytokines and paracrine angiogenic factors. Clinically hEPCs are highly correlated with relapse, disease progression, metastasis and treatment response in malignancies such as breast cancer, ovarian cancer and non-small-cell lung carcinoma. It has become evident that the hEPCs are involved in the angiogenesis-required progression and metastasis of tumours. However, it is not clear in what way the signalling pathways, controlling the normal cellular function of human BM-derived EPCs, are hijacked by aggressive tumour cells to facilitate tumour metastasis. In addition, the actual roles of hEPCs in tumour angiogenesis-mediated metastasis are not well characterised. In this paper we reviewed the clinical relevance of the hEPCs with cancer diagnosis, progression and prognosis. We further summarised the effects of tumour microenvironment on the hEPCs and underlying mechanisms. We also hypothesized the roles of altered hEPCs in tumour angiogenesis and metastasis. We hope this review may enhance our understanding of the interaction between hEPCs and tumour cells thus aiding the development of cellular-targeted anti-tumour therapies.
Collapse
Affiliation(s)
- Valentina Flamini
- Cardiff China Medical Research Collaborative, School of Medicine, Cardiff University, UK
| | - Wen G Jiang
- Cardiff China Medical Research Collaborative, School of Medicine, Cardiff University, UK
| | - Jane Lane
- Cardiff China Medical Research Collaborative, School of Medicine, Cardiff University, UK
| | - Yu-Xin Cui
- Cardiff China Medical Research Collaborative, School of Medicine, Cardiff University, UK.
| |
Collapse
|
39
|
Lehle K, Friedl L, Wilm J, Philipp A, Müller T, Lubnow M, Schmid C. Accumulation of Multipotent Progenitor Cells on Polymethylpentene Membranes During Extracorporeal Membrane Oxygenation. Artif Organs 2015; 40:577-85. [PMID: 26510997 DOI: 10.1111/aor.12599] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Multipotent progenitor cells were mobilized during pediatric extracorporeal membrane oxygenation (ECMO). We hypothesize that these cells also adhered onto polymethylpentene (PMP) fibers within the membrane oxygenator (MO) during adult ECMO support. Mononuclear cells were removed from the surface of explanted PMP-MOs (n = 16). Endothelial-like outgrowth and mesenchymal-like cells were characterized by flow cytometric analysis using different surface markers. Spindle-shaped attaching cells were identified early, but without proliferative activity. After long-term cultivation palisading type or cobblestone-type outgrowth cells with high proliferative activity appeared and were characterized as (i) leukocytoid CD45+/CD31+ (CD133+/VEGFR-II+/CD90+/CD14+/CD146dim/CD105dim); (ii) endothelial-like CD45-/CD31+ (VEGF-RII+/CD146+/CD105+/CD133-/CD14-/CD90-); and (iii) mesenchymal-like cells CD45-/CD31- (CD105+/CD90+/CD133dim/VEGFR-II-/CD146-/CD14-). The distribution of the cell populations depended on the MO and cultivation time. Endothelial-like cells formed capillary-like structures and did uptake Dil-acetylated low-density lipoprotein. Endothelial- and mesenchymal-like cells adhered on the surface of PMP-MOs. Further research is needed to identify the clinical relevance of these cells.
Collapse
Affiliation(s)
- Karla Lehle
- Department of Cardiothoracic Surgery, University Medical Center, Regensburg, Germany
| | - Lucas Friedl
- Department of Cardiothoracic Surgery, University Medical Center, Regensburg, Germany
| | - Julius Wilm
- Department of Cardiothoracic Surgery, University Medical Center, Regensburg, Germany
| | - Alois Philipp
- Department of Cardiothoracic Surgery, University Medical Center, Regensburg, Germany
| | - Thomas Müller
- Department of Internal Medicine II, University Medical Center, Regensburg, Germany
| | - Matthias Lubnow
- Department of Internal Medicine II, University Medical Center, Regensburg, Germany
| | - Christof Schmid
- Department of Cardiothoracic Surgery, University Medical Center, Regensburg, Germany
| |
Collapse
|
40
|
Kawasaki T, Nishiwaki T, Sekine A, Nishimura R, Suda R, Urushibara T, Suzuki T, Takayanagi S, Terada J, Sakao S, Tatsumi K. Vascular Repair by Tissue-Resident Endothelial Progenitor Cells in Endotoxin-Induced Lung Injury. Am J Respir Cell Mol Biol 2015; 53:500-12. [PMID: 25719275 DOI: 10.1165/rcmb.2014-0185oc] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Vascular disruption is one of the pathological hallmarks in acute respiratory distress syndrome. Bone marrow (BM)-derived circulating endothelial progenitor cells (EPCs) and lung tissue-resident EPCs have been considered to play a pivotal role in pulmonary vascular repair; however, which population is predominant in local pulmonary vasculogenesis remains to be clarified. We therefore examined the origin of EPCs participating in the regenerative process of pulmonary vascular endothelial cells (PVECs) in experimental acute respiratory distress syndrome. Lung samples from mice administered LPS intratracheally were investigated for cell dynamics and EPC functions. Quantitative flow cytometric analysis demonstrated that the number of PVECs decreased by roughly 20% on Day 1 and then recovered on Day 7 of LPS challenge. Bromodeoxyuridine-incorporation assays and immunofluorescence microscopy demonstrated that proliferating PVECs preferentially located in the capillary vessels. Experiments using BM chimera mice revealed that most of the regenerating PVECs were tissue-resident cells, and BM-derived cells hardly engrafted as PVECs. The population of circulating putative phenotypical EPCs decreased during the first week after LPS challenge. The regenerating PVECs were characterized by high colony-forming and vasculogenic capacities, intracellular reactive oxygen species scavenging and aldehyde dehydrogenase activites, and enhanced gene expression of Abcb1b (a drug-resistant gene), suggesting that the population of PVECs included tissue-resident EPCs activated during regenerative process of PVECs. The proliferating PVECs expressed CD34, Flk-1/KDR, and c-kit more strongly and Prom1/CD133 less strongly on the surface than nonproliferating PVECs. Our findings indicated that lung tissue-resident EPCs predominantly contribute to pulmonary vascular repair after endotoxin-induced injury.
Collapse
Affiliation(s)
- Takeshi Kawasaki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tetsu Nishiwaki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Ayumi Sekine
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Rintaro Nishimura
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Rika Suda
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takashi Urushibara
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toshio Suzuki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Shin Takayanagi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Jiro Terada
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Seiichiro Sakao
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
41
|
Song J, Lu H, Zheng X, Huang X. Effects of Vascular Endothelial Growth Factor in Recovery Phase of Acute Lung Injury in Mice. Lung 2015; 193:1029-36. [PMID: 26415949 DOI: 10.1007/s00408-015-9803-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 09/18/2015] [Indexed: 02/07/2023]
Abstract
AIM To test the hypothesis that exogenous administration of vascular endothelial growth factor (VEGF) promotes lung repair in acute lung injury (ALI). METHODS ALI was induced by intranasal lipopolysaccharide (LPS) administration in mice, followed by different treatment protocols for 7 days in 3 groups (n = 6, each) including the LPS, the VEGF and the anti-VEGF group. At day 7, peripheral blood and lungs were collected. Lung wet-to-dry (W/D) ratio and lung injury score were measured. Immunohistochemistry assay was employed to detect the number of pulmonary vessels. Circulating endothelial progenitor cells (EPCs) was detected using flow cytometric analysis, and the apoptosis of lung cells was determined by TUNEL staining. RESULTS VEGF treatment reduced W/D ratio and pulmonary neutrophil infiltration in the VEGF group compared with the LPS group. The treatment of VEGF increased the number of pulmonary vessels, and significantly increased the number of circulating EPC cells. Moreover, administration of VEGF decreased the percentage of apoptotic cells in the VEGF group. CONCLUSIONS Our results suggest that VEGF may contribute to vascular endothelial repair and function as a protective factor against ALI.
Collapse
Affiliation(s)
- Junfeng Song
- Department of Pediatrics, Hangzhou First People's Hospital, 261 Huansha Road, Hangzhou, 310006, China.
| | - Hui Lu
- Department of Pediatrics, Hangzhou First People's Hospital, 261 Huansha Road, Hangzhou, 310006, China
| | - Xuyang Zheng
- Department of Pediatrics, Hangzhou First People's Hospital, 261 Huansha Road, Hangzhou, 310006, China
| | - Xianmei Huang
- Department of Pediatrics, Hangzhou First People's Hospital, 261 Huansha Road, Hangzhou, 310006, China
| |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW The prognosis of patients with respiratory failure in the ICU remains poor, while current therapeutic approaches are aimed at minimizing ventilator-induced lung injury. Stem cell-based therapies have the potential to transform respiratory failure treatment by achieving lung repair. The purpose of this article is to critically review the large body of clinical and experimental work performed with respect to the use of stem/progenitor cells in respiratory failure, and to discuss current challenges and future directions. RECENT FINDINGS Since the initial report of cell therapy for lung injury in 2005, numerous preclinical and clinical studies have been performed that support the ability of various stem cell populations to improve physiologic lung function and reduce inflammation in both infective and sterile acute respiratory distress syndrome. Nevertheless, many important issues (e.g., mechanism of action, long-term engraftment, optimal cell type, dose, route of administration) remain to be resolved. SUMMARY Cell-based therapeutics hold promise, particularly for acute respiratory distress syndrome, and early preclinical testing has been encouraging. To advance clinical testing of cell therapies in respiratory failure, and to help ensure that this approach will facilitate bench-to-bedside and bedside-to-bench discoveries, parallel paths of basic and clinical research are needed, including measures of cell therapy effectiveness in vivo and in vitro.
Collapse
|
43
|
Patry C, Orfanos SE, Rafat N. Translational research in ARDS patients: new biological phenotypes. Intensive Care Med 2015. [PMID: 26224249 DOI: 10.1007/s00134-015-4005-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Christian Patry
- Department of Pediatrics I, University Children's Hospital, University of Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Stylianos E Orfanos
- Second Department of Critical Care, Attikon University Hospital, University of Athens Medical School, 1, Rimini St., 12462, Haidari, Athens, Greece
| | - Neysan Rafat
- Department of Pediatrics I, University Children's Hospital, University of Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany.
| |
Collapse
|
44
|
Preconditioning allows engraftment of mouse and human embryonic lung cells, enabling lung repair in mice. Nat Med 2015; 21:869-79. [PMID: 26168294 DOI: 10.1038/nm.3889] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 05/26/2015] [Indexed: 02/07/2023]
Abstract
Repair of injured lungs represents a longstanding therapeutic challenge. We show that human and mouse embryonic lung tissue from the canalicular stage of development (20-22 weeks of gestation for humans, and embryonic day 15-16 (E15-E16) for mouse) are enriched with progenitors residing in distinct niches. On the basis of the marked analogy to progenitor niches in bone marrow (BM), we attempted strategies similar to BM transplantation, employing sublethal radiation to vacate lung progenitor niches and to reduce stem cell competition. Intravenous infusion of a single cell suspension of canalicular lung tissue from GFP-marked mice or human fetal donors into naphthalene-injured and irradiated syngeneic or SCID mice, respectively, induced marked long-term lung chimerism. Donor type structures or 'patches' contained epithelial, mesenchymal and endothelial cells. Transplantation of differentially labeled E16 mouse lung cells indicated that these patches were probably of clonal origin from the donor. Recipients of the single cell suspension transplant exhibited marked improvement in lung compliance and tissue damping reflecting the energy dissipation in the lung tissues. Our study provides proof of concept for lung reconstitution by canalicular-stage human lung cells after preconditioning of the pulmonary niche.
Collapse
|
45
|
Erythropoietin promotes the protective properties of transplanted endothelial progenitor cells against acute lung injury via PI3K/Akt pathway. Shock 2015; 42:327-36. [PMID: 25051281 DOI: 10.1097/shk.0000000000000216] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Accumulating evidences have indicted the participation and repairing effects of endothelial progenitor cells (EPCs) on acute lung injury (ALI). Researchers have also revealed that erythropoietin (EPO) may exert multiple effects on EPCs including proliferative and adhesive properties. The present study was designed to investigate whether EPO can promote the healing efficiency of transplanted EPCs against ALI and the potential mechanism. METHODS Endothelial progenitor cells were derived from bone marrow mononuclear cells of BALB/c mice. In our in vitro studies, we tested the proproliferative and antiapoptotic effects of EPO on cultured EPCs. In our in vivo studies, we induced BALB/c mice ALI model by intratracheal instillation of lipopolysaccharide and treated with/without intravenous injection of enhanced green fluorescent protein-EPCs harvested from enhanced green fluorescent protein-BALB/c mice. Mice that received EPC transplantation either underwent EPO administration or not. The effects of EPO and EPC treatment on promoting pulmonary endothelial repair, decreasing pulmonary capillary permeability, alleviating pulmonary inflammation, improving gas exchange, and promoting pulmonary vessel angiogenesis were tested. The potential mechanisms that mediate EPO functions on EPC were also investigated. RESULTS Our studies revealed a significant antiapoptotic property of EPO on cultured EPCs as well as its promotion on EPC proliferation. We also found the severity of ALI was reduced by EPC therapy, and the protective effects of EPCs were highly enhanced when combined with EPO administration. However, all these stimulating effects of EPO on EPCs were consisted with the expression of phospho-Akt and were abrogated by PI3K inhibitors. CONCLUSIONS Transplanted EPCs directly incorporated into the injured pulmonary vessels and maintain the integrity of pulmonary endothelium. Erythropoietin improved the survival and proliferation of transplanted EPCs and recruited them to the injured sites to exert their repairing functions. PI3K/Akt pathway mediated EPO's functions on EPCs. The combination of EPO and EPC treatment may be a promising cell-based therapy for ALI patients.
Collapse
|
46
|
Mao SZ, Ye X, Liu G, Song D, Liu SF. Resident Endothelial Cells and Endothelial Progenitor Cells Restore Endothelial Barrier Function After Inflammatory Lung Injury. Arterioscler Thromb Vasc Biol 2015; 35:1635-1644. [PMID: 25977568 DOI: 10.1161/atvbaha.115.305519] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 04/29/2015] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Disruption of endothelial barrier integrity is a characteristic of many inflammatory conditions. However, the origin and function of endothelial cells (ECs) restoring endothelial barrier function remain unknown. This study defined the roles of resident ECs (RECs) and bone marrow-derived endothelial progenitor cells (BMDEPCs) in endothelial barrier restoration after endotoxemic lung injury. APPROACH AND RESULTS We generated mice that enable to quantify proliferating RECs or BMDEPCs and also to study the causal link between REC or BMDEPC proliferation and endothelial barrier restoration. Using these mouse models, we showed that endothelial barrier restoration was associated with increased REC and BMDEPC proliferation. RECs and BMDEPCs participate in barrier repair. Immunofluorescence staining demonstrated that RECs proliferate in situ on endothelial layer and that BMDEPCs are engrafted into endothelial layer of lung microvessels at the active barrier repair phase. In lungs, 8 weeks after lipopolysaccharide-induced injury, the number of REC-derived ECs (CD45(-)/CD31(+)/BrdU(+)/rtTA(+)) or BMDEPC-derived ECs (CD45(-)/CD31(+)/eNOS(+)/GFP(+)) increased by 22- or 121-fold, respectively. The suppression of REC or BMDEPC proliferation by blocking REC or BMDEPC intrinsic nuclear factor-κB at the barrier repair phase was associated with an augmented endothelial permeability and impeded endothelial barrier recovery. RECs and BMDEPCs contributed differently to endothelial barrier repair. In lungs, 8 weeks after lipopolysaccharide-induced injury, REC-derived ECs constituted 22%, but BMDEPC-derived ECs constituted only 3.7% of the total new ECs. CONCLUSIONS REC is a major and BMDEPC is a complementary source of new ECs in endothelial barrier restoration. RECs and BMDEPCs play important roles in endothelial barrier restoration after inflammatory lung injury.
Collapse
Affiliation(s)
- Sun-Zhong Mao
- Centers for Heart and Lung Research, and Pulmonary and Critical Care Medicine, the Feinstein Institute for Medical Research, Manhasset, NY 11030, U.S.A.,Institute of Hypoxia Medicine, Wenzhou Medical University, Wenzhou 325035, China
| | - Xiaobing Ye
- Centers for Heart and Lung Research, and Pulmonary and Critical Care Medicine, the Feinstein Institute for Medical Research, Manhasset, NY 11030, U.S.A
| | - Gang Liu
- Centers for Heart and Lung Research, and Pulmonary and Critical Care Medicine, the Feinstein Institute for Medical Research, Manhasset, NY 11030, U.S.A
| | - Dongmei Song
- Centers for Heart and Lung Research, and Pulmonary and Critical Care Medicine, the Feinstein Institute for Medical Research, Manhasset, NY 11030, U.S.A
| | - Shu Fang Liu
- Centers for Heart and Lung Research, and Pulmonary and Critical Care Medicine, the Feinstein Institute for Medical Research, Manhasset, NY 11030, U.S.A.,Institute of Hypoxia Medicine, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
47
|
Zhao YD, Huang X, Yi F, Dai Z, Qian Z, Tiruppathi C, Tran K, Zhao YY. Endothelial FoxM1 mediates bone marrow progenitor cell-induced vascular repair and resolution of inflammation following inflammatory lung injury. Stem Cells 2015; 32:1855-64. [PMID: 24578354 DOI: 10.1002/stem.1690] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 01/25/2014] [Accepted: 01/30/2014] [Indexed: 01/08/2023]
Abstract
Adult stem cell treatment is a potential novel therapeutic approach for acute respiratory distress syndrome. Given the extremely low rate of cell engraftment, it is believed that these cells exert their beneficial effects via paracrine mechanisms. However, the endogenous mediator(s) in the pulmonary vasculature remains unclear. Using the mouse model with endothelial cell (EC)-restricted disruption of FoxM1 (FoxM1 CKO), here we show that endothelial expression of the reparative transcriptional factor FoxM1 is required for the protective effects of bone marrow progenitor cells (BMPC) against LPS-induced inflammatory lung injury and mortality. BMPC treatment resulted in rapid induction of FoxM1 expression in wild type (WT) but not FoxM1 CKO lungs. BMPC-induced inhibition of lung vascular injury, resolution of lung inflammation, and survival, as seen in WT mice, were abrogated in FoxM1 CKO mice following LPS challenge. Mechanistically, BMPC treatment failed to induce lung EC proliferation in FoxM1 CKO mice, which was associated with impaired expression of FoxM1 target genes essential for cell cycle progression. We also observed that BMPC treatment enhanced endothelial barrier function in WT but not in FoxM1-deficient EC monolayers. Restoration of β-catenin expression in FoxM1-deficient ECs normalized endothelial barrier enhancement in response to BMPC treatment. These data demonstrate the requisite role of endothelial FoxM1 in the mechanism of BMPC-induced vascular repair to restore vascular integrity and accelerate resolution of inflammation, thereby promoting survival following inflammatory lung injury.
Collapse
Affiliation(s)
- Yidan D Zhao
- Department of Pharmacology, The University of Illinois College of Medicine, Chicago, Illinois, USA; Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, Illinois, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Moussa MD, Santonocito C, Fagnoul D, Donadello K, Pradier O, Gaussem P, De Backer D, Vincent JL. Evaluation of endothelial damage in sepsis-related ARDS using circulating endothelial cells. Intensive Care Med 2015; 41:231-8. [PMID: 25510299 DOI: 10.1007/s00134-014-3589-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 11/27/2014] [Indexed: 02/07/2023]
Abstract
PURPOSE Endothelial cell activation and dysfunction are involved in the pathophysiology of ARDS. Circulating endothelial cells (CECs) may be a useful marker of endothelial dysfunction and damage but have been poorly studied in ARDS. We hypothesized that the CEC count may be elevated in patients with sepsis-related ARDS compared to those with sepsis without ARDS. METHODS ARDS was defined according to the Berlin consensus definition. The study population included 17 patients with moderate or severe ARDS, 9 with mild ARDS, 13 with sepsis and no ARDS, 13 non-septic patients, and 12 healthy volunteers. Demographic, hemodynamic, and prognostic variables, including PaO(2)/FiO(2) ratio, 28-day survival, blood lactate, APACHE II, and SOFA score, were recorded. CECs were counted in arterial blood samples using the reference CD146 antibody-based immunomagnetic isolation and UEA1-FITC staining method. Measurements were performed 12-24 h after diagnosis of ARDS and repeated daily for 3 days. RESULTS The median day-1 CEC count was significantly higher in patients with moderate or severe ARDS than in mild ARDS or septic-control patients [27.2 (18.3-49.4) vs. 17.4 (11-24.5) cells/ml (p < 0.034), and 18.4 (9.1-31) cells/ml (p < 0.035), respectively]. All septic patients (with or without ARDS) had higher day-1 CEC counts than the non-septic patients [19.6 (14.2-30.6) vs. 10.8 (5.7-13.2) cells/ml, p = 0.002]. CONCLUSION The day-1 CEC count was significantly higher in ARDS patients than in other critically ill patients, and in moderate or severe ARDS patients compared to those with milder disease, making it a potentially useful marker of ARDS severity.
Collapse
Affiliation(s)
- Mouhamed Djahoum Moussa
- Department of Intensive Care, Erasme University Hospital, Université Libre de Bruxelles (ULB), 808 Route de Lennik, 1070, Brussels, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Müller-Redetzky HC, Lienau J, Witzenrath M. The Lung Endothelial Barrier in Acute Inflammation. THE VERTEBRATE BLOOD-GAS BARRIER IN HEALTH AND DISEASE 2015. [PMCID: PMC7123850 DOI: 10.1007/978-3-319-18392-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
50
|
Baskir R, Majka S. Pulmonary Vascular Remodeling by Resident Lung Stem and Progenitor Cells. LUNG STEM CELLS IN THE EPITHELIUM AND VASCULATURE 2015. [DOI: 10.1007/978-3-319-16232-4_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|