1
|
He X, Cui Y, Li T, Luo L, Zeng Z, Ma Y, Chen Y. PU.1 alleviates the inhibitory effects of cigarette smoke on endothelial progenitor cell function and lung-homing through Wnt/β-catenin and CXCL12/CXCR4 pathways. Tob Induc Dis 2024; 22:TID-22-27. [PMID: 38274000 PMCID: PMC10809061 DOI: 10.18332/tid/174661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 02/18/2023] [Accepted: 10/30/2023] [Indexed: 01/27/2024] Open
Abstract
INTRODUCTION Endothelial progenitor cells (EPCs) dysfunction is involved in the pathogenesis of chronic obstructive pulmonary disease (COPD). The transcription factor PU.1 is essential for the maintenance of stem/progenitor cell homeostasis. However, the role of PU.1 in COPD and its effects on EPC function and lung-homing, remain unclear. This study aimed to explore the protective activity of PU.1 and the underlying mechanisms in a cigarette smoke extract (CSE)-induced emphysema mouse model. METHODS C57BL/6 mice were treated with CSE to establish a murine emphysema model and injected with overexpressed PU.1 or negative control adeno-associated virus. Morphometry of lung slides, lung function, and apoptosis of lung tissues were evaluated. Immunofluorescence co-localization was used to analyze EPCs homing into the lung. Flow cytometry was performed to detect EPC count in lung tissues and bone marrow (BM). The angiogenic ability of BM-derived EPCs cultured in vitro was examined by tube formation assay. We determined the expression levels of PU.1, β-catenin, C-X-C motif ligand 12 (CXCL12), C-X-C motif receptor 4 (CXCR4), stem cell antigen-1 (Sca-1), and stemness genes. RESULTS CSE exposure significantly reduced the expression of PU.1 in mouse lung tissues, BM, and BM-derived EPCs. PU.1 overexpression attenuated CSE-induced emphysematous changes, lung function decline, and apoptosis. In emphysematous mice, PU.1 overexpression markedly reversed the decreased proportion of EPCs in BM and promoted the lung-homing of EPCs. The impaired angiogenic ability of BM-derived EPCs induced by CSE could be restored by the overexpression of PU.1. In addition, PU.1 upregulation evidently reversed the decreased expression of β-catenin, CXCL12, CXCR4, Scal-1, and stemness genes in mouse lung tissues, BM, and BM-derived EPCs after CSE exposure. CONCLUSIONS PU.1 alleviates the inhibitory effects of CSE on EPC function and lung-homing via activating the canonical Wnt/β-catenin pathway and CXCL12/CXCR4 axis. While further research is needed, our research may indicate a potential therapeutic target for COPD patients.
Collapse
Affiliation(s)
- Xue He
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Yanan Cui
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Tiao Li
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Lijuan Luo
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Zihang Zeng
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Yiming Ma
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| | - Yan Chen
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Research Unit of Respiratory Disease, Central South University, Changsha, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, China
| |
Collapse
|
2
|
Zhu X, Fan C, Xiong Z, Chen M, Li Z, Tao T, Liu X. Development and application of oncolytic viruses as the nemesis of tumor cells. Front Microbiol 2023; 14:1188526. [PMID: 37440883 PMCID: PMC10335770 DOI: 10.3389/fmicb.2023.1188526] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/18/2023] [Indexed: 07/15/2023] Open
Abstract
Viruses and tumors are two pathologies that negatively impact human health, but what occurs when a virus encounters a tumor? A global consensus among cancer patients suggests that surgical resection, chemotherapy, radiotherapy, and other methods are the primary means to combat cancer. However, with the innovation and development of biomedical technology, tumor biotherapy (immunotherapy, molecular targeted therapy, gene therapy, oncolytic virus therapy, etc.) has emerged as an alternative treatment for malignant tumors. Oncolytic viruses possess numerous anti-tumor properties, such as directly lysing tumor cells, activating anti-tumor immune responses, and improving the tumor microenvironment. Compared to traditional immunotherapy, oncolytic virus therapy offers advantages including high killing efficiency, precise targeting, and minimal side effects. Although oncolytic virus (OV) therapy was introduced as a novel approach to tumor treatment in the 19th century, its efficacy was suboptimal, limiting its widespread application. However, since the U.S. Food and Drug Administration (FDA) approved the first OV therapy drug, T-VEC, in 2015, interest in OV has grown significantly. In recent years, oncolytic virus therapy has shown increasingly promising application prospects and has become a major research focus in the field of cancer treatment. This article reviews the development, classification, and research progress of oncolytic viruses, as well as their mechanisms of action, therapeutic methods, and routes of administration.
Collapse
Affiliation(s)
- Xiao Zhu
- Zhejiang Provincial People's Hospital Affiliated to Hangzhou Medical College, Hangzhou Medical College, Hangzhou, China
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
- Department of Biological and Chemical Sciences, New York Institute of Technology—Manhattan Campus, New York, NY, United States
| | - Chenyang Fan
- Department of Clinical Medicine, Medicine and Technology, School of Zunyi Medical University, Zunyi, China
| | - Zhuolong Xiong
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Mingwei Chen
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Zesong Li
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital(Shenzhen Institute of Translational Medicine), Shenzhen, China
| | - Tao Tao
- Department of Gastroenterology, Zibo Central Hospital, Zibo, China
| | - Xiuqing Liu
- Department of Clinical Laboratory, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| |
Collapse
|
3
|
Wang X, Zhao X, He Z. Mesenchymal stem cell carriers enhance anti-tumor efficacy of oncolytic virotherapy. Oncol Lett 2021; 21:238. [PMID: 33664802 PMCID: PMC7882891 DOI: 10.3892/ol.2021.12499] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 12/09/2020] [Indexed: 12/21/2022] Open
Abstract
Oncolytic viruses (OVs) specifically infect, replicate and eventually destroy tumor cells, with no concomitant toxicity to adjacent normal cells. Furthermore, OVs can regulate tumor microenvironments and stimulate anti-tumor immune responses. Mesenchymal stem cells (MSCs) have inherent tumor tropisms and immunosuppressive functions. MSCs carrying OVs not only protect viruses from clearing by the immune system, but they also deliver the virus to tumor lesions. Equally, cytokines released by MSCs enhance anti-tumor immune responses, suggesting that MSCs carrying OVs may be considered as a promising strategy in enhancing the anti-tumor efficacies of virotherapy. In the present review, preclinical and clinical studies were evaluated and discussed, as well as the effectiveness of MSCs carrying OVs for tumor treatment.
Collapse
Affiliation(s)
- Xianyao Wang
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
- Key Laboratory of Adult Stem Cell Translational Research, Chinese Academy of Medical Sciences, Guiyang, Guizhou 550004, P.R. China
- Department of Immunology, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Xing Zhao
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
- Key Laboratory of Adult Stem Cell Translational Research, Chinese Academy of Medical Sciences, Guiyang, Guizhou 550004, P.R. China
- Department of Immunology, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Zhixu He
- Key Laboratory of Adult Stem Cell Translational Research, Chinese Academy of Medical Sciences, Guiyang, Guizhou 550004, P.R. China
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, P.R. China
| |
Collapse
|
4
|
Hao W, Li M, Pang Y, Du W, Huang X. Increased chemokines levels in patients with chronic obstructive pulmonary disease: correlation with quantitative computed tomography metrics. Br J Radiol 2020; 94:20201030. [PMID: 33237823 DOI: 10.1259/bjr.20201030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE We sought to explore the relationships between multiple chemokines with spirometry, inflammatory mediators and CT findings of emphysema, small airways disease and bronchial wall thickness. METHODS All patients with COPD (n = 65) and healthy control subjects (n = 23) underwent high-resolution CT, with image analysis determining the low attenuation area (LAA), ratio of mean lung attenuation on expiratory and inspiratory scans (E/I MLD) and bronchial wall thickness of inner perimeter of a 10-mm diameter airway (Pi10). At enrollment, subjects underwent pulmonary function studies, chemokines and inflammatory mediators measurements. RESULTS Multiple chemokines (CCL2, CCL3, CCL5, CX3CL1, CXCL8, CXCL9, CXCL10, CXCL11 and CXCL12) and inflammatory mediators (MMP-9, MMP-12, IL-18 and neutrophil count) were markedly increased in the serum of COPD patients compared with healthy controls. There were associations between small airway disease (E/I MLD) and CCL11, CXCL8, CXCL10, CXCL11, CXCL12 and CX3CL1. Especially CXCL8 and CX3CL1 are strongly associated with E/I MLD (r = 0.74, p < 0.001; r = 0.76, p < 0.001, respectively). CXCL8, CXCL12 and CX3CL1 were moderately positively correlated with emphysema (%LAA) (r = 0.49, p < 0.05; r = 0.51, p < 0.05; r = 0.54, p < 0.01, respectively). Bronchial wall thickness (Pi10)showed no significant differences between the COPD and healthy controls,,but there was an association between Pi10 and FEV1% in COPD patients (r=-0.420, p = 0.048). Our statistical results showed that there were not any associations between airway wall thickness (Pi10) and chemokines. CONCLUSION Pulmonary chemokines levels are closely associated with the extent of gas trapping, small airways disease and emphysema identified on high-resolution chest CT scan. ADVANCES IN KNOWLEDGE This study combines quantitative CT analysis with multiplex chemokines and inflammatory mediators to identify a new role of pathological changes in COPD.
Collapse
Affiliation(s)
- Wendong Hao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'anJiaotong University, Xi'an, China.,Department of Respiratory Medicine, The Affiliated Hospital of Yan'an University, Yan'an, China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'anJiaotong University, Xi'an, China
| | - Yamei Pang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'anJiaotong University, Xi'an, China
| | - Weiping Du
- Clinical Laboratory Diagnosis Department, The Affiliated Hospital of Yan'an University, Yan'an, China
| | - Xiaoqi Huang
- Department of Radiology, The Affiliated Hospital of Yan'an University, Yan'an, China
| |
Collapse
|
5
|
Sperelakis I, Tsitoura E, Koutoulaki C, Mastrodimou S, Tosounidis TH, Spandidos DA, Antoniou KM, Kontakis G. Influence of reaming intramedullary nailing on MSC population after surgical treatment of patients with long bone fracture. Mol Med Rep 2020; 22:2521-2527. [PMID: 32705190 PMCID: PMC7411410 DOI: 10.3892/mmr.2020.11320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 07/09/2020] [Indexed: 11/22/2022] Open
Abstract
Reamed intramedullary nailing (RIN) is a surgical method of choice for treatment of diaphyseal fractures. This procedure affects the biological environment of bone tissue locally and systemically. This study investigated the influence of RIN on mesenchymal stem cells (MSCs) in patients with long bone fractures. The axis of C-X-C motif chemokine receptor 4 (CXCR4)/stromal cell-derived factor 1 (SDF-1) was selected since it is considered as major pathway for MSC homing and migration. Iliac crest bone marrow (IC-BM) samples and blood samples were collected at two different time points. One sample was collected before the RIN (BN) and the other immediately after RIN (AN). BM-MSCs were cultured and RT-qPCR was performed for CXCR4 mRNA levels and ELISA for the SDF-1 sera levels. The experimental study revealed that there was a correlation between the increase of SDF-1 levels in peripheral blood and a decrease in the levels of CXCR4 in MSCs in the IC-BM following RIN. The levels of SDF-1 showed a significant increase in the sera of patients after RIN. In conclusion, the present study is the first providing evidence of the effects of RIN on MSC population via the CXCR4/SDF-1 axis. The levels of serum SDF-1 factor were elevated after RIN while increased levels of SDF-1 in peripheral blood were inversely correlated with the mRNA levels of CXCR4 on BM-MSCs after RIN. Therefore, this study contributes to enlighten the systematic effects of RIN on the population of MSCs at a cellular level.
Collapse
Affiliation(s)
- Ioannis Sperelakis
- Department of Orthopedics and Traumatology, University of Crete School of Medicine, 71003 Heraklion, Greece
| | - Eliza Tsitoura
- Department of Respiratory Medicine, University General Hospital of Heraklion, Laboratory of Molecular and Cellular Pneumonology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Chara Koutoulaki
- Department of Respiratory Medicine, University General Hospital of Heraklion, Laboratory of Molecular and Cellular Pneumonology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Semeli Mastrodimou
- Department of Respiratory Medicine, University General Hospital of Heraklion, Laboratory of Molecular and Cellular Pneumonology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Theodoros H Tosounidis
- Department of Orthopedics and Traumatology, University of Crete School of Medicine, 71003 Heraklion, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Katerina M Antoniou
- Department of Respiratory Medicine, University General Hospital of Heraklion, Laboratory of Molecular and Cellular Pneumonology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - George Kontakis
- Department of Orthopedics and Traumatology, University of Crete School of Medicine, 71003 Heraklion, Greece
| |
Collapse
|
6
|
Abstract
As the prevalence and impact of lung diseases continue to increase worldwide, new therapeutic strategies are desperately needed. Advances in lung-regenerative medicine, a broad field encompassing stem cells, cell-based therapies, and a range of bioengineering approaches, offer new insights into and new techniques for studying lung physiology and pathophysiology. This provides a platform for the development of novel therapeutic approaches. Applicability to chronic obstructive pulmonary disease of recent advances and applications in cell-based therapies, predominantly those with mesenchymal stromal cell-based approaches, and bioengineering approaches for lung diseases are reviewed.
Collapse
|
7
|
Chemokines in COPD: From Implication to Therapeutic Use. Int J Mol Sci 2019; 20:ijms20112785. [PMID: 31174392 PMCID: PMC6600384 DOI: 10.3390/ijms20112785] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/03/2019] [Accepted: 06/03/2019] [Indexed: 02/07/2023] Open
Abstract
: Chronic Obstructive Pulmonary Disease (COPD) represents the 3rd leading cause of death in the world. The underlying pathophysiological mechanisms have been the focus of extensive research in the past. The lung has a complex architecture, where structural cells interact continuously with immune cells that infiltrate into the pulmonary tissue. Both types of cells express chemokines and chemokine receptors, making them sensitive to modifications of concentration gradients. Cigarette smoke exposure and recurrent exacerbations, directly and indirectly, impact the expression of chemokines and chemokine receptors. Here, we provide an overview of the evidence regarding chemokines involvement in COPD, and we hypothesize that a dysregulation of this tightly regulated system is critical in COPD evolution, both at a stable state and during exacerbations. Targeting chemokines and chemokine receptors could be highly attractive as a mean to control both chronic inflammation and bronchial remodeling. We present a special focus on the CXCL8-CXCR1/2, CXCL9/10/11-CXCR3, CCL2-CCR2, and CXCL12-CXCR4 axes that seem particularly involved in the disease pathophysiology.
Collapse
|
8
|
Coppolino I, Ruggeri P, Nucera F, Cannavò MF, Adcock I, Girbino G, Caramori G. Role of Stem Cells in the Pathogenesis of Chronic Obstructive Pulmonary Disease and Pulmonary Emphysema. COPD 2018; 15:536-556. [DOI: 10.1080/15412555.2018.1536116] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Irene Coppolino
- Dipartimento di Scienze Biomediche, Unità Operativa Complessa di Pneumologia, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, Messina, Italy
| | - Paolo Ruggeri
- Dipartimento di Scienze Biomediche, Unità Operativa Complessa di Pneumologia, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, Messina, Italy
| | - Francesco Nucera
- Dipartimento di Scienze Biomediche, Unità Operativa Complessa di Pneumologia, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, Messina, Italy
| | - Mario Francesco Cannavò
- Dipartimento di Scienze Biomediche, Unità Operativa Complessa di Pneumologia, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, Messina, Italy
| | - Ian Adcock
- Airways Disease Section, National Heart and Lung Institute, Royal Brompton Hospital Biomedical Research Unit, Imperial College, London, UK
| | - Giuseppe Girbino
- Dipartimento di Scienze Biomediche, Unità Operativa Complessa di Pneumologia, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, Messina, Italy
| | - Gaetano Caramori
- Dipartimento di Scienze Biomediche, Unità Operativa Complessa di Pneumologia, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, Messina, Italy
| |
Collapse
|
9
|
Barwinska D, Oueini H, Poirier C, Albrecht ME, Bogatcheva NV, Justice MJ, Saliba J, Schweitzer KS, Broxmeyer HE, March KL, Petrache I. AMD3100 ameliorates cigarette smoke-induced emphysema-like manifestations in mice. Am J Physiol Lung Cell Mol Physiol 2018; 315:L382-L386. [PMID: 29745251 DOI: 10.1152/ajplung.00185.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
We have shown that cigarette smoke (CS)-induced pulmonary emphysema-like manifestations are preceded by marked suppression of the number and function of bone marrow hematopoietic progenitor cells (HPCs). To investigate whether a limited availability of HPCs may contribute to CS-induced lung injury, we used a Food and Drug Administration-approved antagonist of the interactions of stromal cell-derived factor 1 (SDF-1) with its chemokine receptor CXCR4 to promote intermittent HPC mobilization and tested its ability to limit emphysema-like injury following chronic CS. We administered AMD3100 (5mg/kg) to mice during a chronic CS exposure protocol of up to 24 wk. AMD3100 treatment did not affect either lung SDF-1 levels, which were reduced by CS, or lung inflammatory cell counts. However, AMD3100 markedly improved CS-induced bone marrow HPC suppression and significantly ameliorated emphysema-like end points, such as alveolar airspace size, lung volumes, and lung static compliance. These results suggest that antagonism of SDF-1 binding to CXCR4 is associated with protection of both bone marrow and lungs during chronic CS exposure, thus encouraging future studies of potential therapeutic benefit of AMD3100 in emphysema.
Collapse
Affiliation(s)
- Daria Barwinska
- Department of Cellular and Integrative Physiology, Indiana University , Indianapolis, Indiana.,Indiana Center for Vascular Biology and Medicine, Indiana University , Indianapolis, Indiana.,Vascular and Cardiac Center for Adult Stem Cell Therapy Signature Center, Indiana University, Purdue University , Indianapolis, Indiana.,Roudebush Veterans Affairs Medical Center, Indiana University , Indianapolis, Indiana.,Division of Nephrology, Department of Medicine, Indiana University , Indianapolis, Indiana
| | - Houssam Oueini
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Indiana University , Indianapolis, Indiana
| | - Christophe Poirier
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Indiana University , Indianapolis, Indiana
| | - Marjorie E Albrecht
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Indiana University , Indianapolis, Indiana
| | - Natalia V Bogatcheva
- Indiana Center for Vascular Biology and Medicine, Indiana University , Indianapolis, Indiana.,Vascular and Cardiac Center for Adult Stem Cell Therapy Signature Center, Indiana University, Purdue University , Indianapolis, Indiana.,Roudebush Veterans Affairs Medical Center, Indiana University , Indianapolis, Indiana.,Division of Cardiology, Department of Medicine, Indiana University , Indianapolis, Indiana
| | - Matthew J Justice
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Indiana University , Indianapolis, Indiana.,Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
| | - Jacob Saliba
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Indiana University , Indianapolis, Indiana
| | - Kelly S Schweitzer
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Indiana University , Indianapolis, Indiana.,Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
| | - Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University , Indianapolis, Indiana
| | - Keith L March
- Indiana Center for Vascular Biology and Medicine, Indiana University , Indianapolis, Indiana.,Vascular and Cardiac Center for Adult Stem Cell Therapy Signature Center, Indiana University, Purdue University , Indianapolis, Indiana.,Roudebush Veterans Affairs Medical Center, Indiana University , Indianapolis, Indiana.,Division of Cardiology, Department of Medicine, Indiana University , Indianapolis, Indiana.,Division of Cardiovascular Medicine and Center for Regenerative Medicine, University of Florida , Gainesville, Florida
| | - Irina Petrache
- Indiana Center for Vascular Biology and Medicine, Indiana University , Indianapolis, Indiana.,Vascular and Cardiac Center for Adult Stem Cell Therapy Signature Center, Indiana University, Purdue University , Indianapolis, Indiana.,Roudebush Veterans Affairs Medical Center, Indiana University , Indianapolis, Indiana.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Indiana University , Indianapolis, Indiana.,Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado.,Department of Medicine, University of Colorado , Denver, Colorado
| |
Collapse
|