1
|
Pei J, Wang S, Cheng K, Xu S, Zhao X, Zhao K, Luo Y, Li W, Yu J, Liu J. C-X-C-chemokine-receptor-type-4 as a potential target for diagnosis and treatment of acute radiation-induced esophagitis. Int Immunopharmacol 2025; 150:114289. [PMID: 39970707 DOI: 10.1016/j.intimp.2025.114289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 02/01/2025] [Accepted: 02/10/2025] [Indexed: 02/21/2025]
Abstract
PURPOSE Acute radiation-induced esophagitis (ARIE) is a common and dose-limiting adverse reaction associated with radiotherapy for thoracic tumors. mRNA sequencing identified CXCR4 as a potential target for noninvasive imaging of ARIE and significant up-regulation of CXCR4 expression was further confirmed in ARIE experimental animal model and clinical samples. This research investigated the feasibility of targeting CXCR4 for the diagnosis and treatment of ARIE. METHODS ARIE models were established, and magnetic resonance imaging was performed. Dynamic, blocking, histopathological studies, mRNA-sequencing and flow cytometry were conducted. The feasibility of an 18F-labeled polypeptide (QHY-04) targeting CXCR4 for detecting ARIE was validated. CXCR4 blockade using AMD3100 was applied immediately post radiotherapy. RESULTS Increased signal intensity in the esophagus and surrounding tissues was observed in ARIE models, with clinical manifestations confirmed by H&E staining. Immunofluorescence staining demonstrated significant CXCR4 up-regulation. Significantly increased [18F]AlF-NOTA-QHY-04 uptake in the irradiated esophagus was observed via PET imaging. Immune cell infiltration and flow cytometry identified CXCR4-positive neutrophils and monocytes as the primary source of the radiotracer. AMD3100-mediated CXCR4 blockade significantly reduced ARIE. CONCLUSION CXCR4-targeted PET/CT facilitates noninvasive detection of ARIE in experimental animal models. CXCR4 blockade mitigates ARIE, highlighting CXCR4 as a promising theranostic target of ARIE.
Collapse
Affiliation(s)
- Jinli Pei
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Shijie Wang
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Kai Cheng
- Department of PET/CT Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Shengnan Xu
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xinzhi Zhao
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Kunlong Zhao
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yuxi Luo
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Wanhu Li
- Department of PET/CT Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jinming Yu
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jie Liu
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
2
|
Paget JT, Ward JA, McKean AR, Mansfield DC, McLaughlin M, Kyula-Currie JN, Smith HG, Roulstone V, Li C, Zhou Y, Hardiman T, Grigoriadis A, O'Brien Coon D, Irshad S, Melcher AA, Harrington KJ, Khan A. CXCL12-Targeted Immunomodulatory Gene Therapy Reduces Radiation-Induced Fibrosis in Healthy Tissues. Mol Cancer Ther 2025; 24:431-443. [PMID: 39666014 DOI: 10.1158/1535-7163.mct-23-0872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 07/26/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
Radiation-induced fibrosis (RIF) is a progressive pathology deleteriously impacting cancer survivorship. CXCL12 is an immune-stromal signal implicated in fibrosis and innate response. We hypothesized that modulation of CXCL12 would phenotypically mitigate RIF. CXCL12 expression was characterized in a rodent model of RIF and its expression modulated by the intravascular delivery of lentiviral vectors encoding small hairpin RNA to silence (LVShCXCL12) or overexpress (LVOeCXCL12) CXCL12. Multimodal fibrotic outcomes were quantified, and flow cytometry and Y-chromosome lineage-tracking studies performed to examine cellular recruitment and activation after radiotherapy. Whole-tissue RNA sequencing was used to examine matrisomal response. MATBIII tumors were engrafted within tissues with differing levels of CXCL12 expression, and tumoral response to RT was evaluated. CXCL12 was upregulated in irradiated fibroblasts demonstrating DNA damage after radiotherapy, which led to the recruitment of CD68+ macrophages. Silencing CXCL12 with LVShCXCL12 demonstrated reduced RIF phenotype as a result of decreased macrophage recruitment. Transcriptomic profiling identified osteopontin (OPN; SPP1) as being highly differentially expressed in LVShCXCL12-treated tissues. Tumors growing in tissues devoid of CXCL12 expression responded better after RT because of reductions in peritumoral fibrosis as a result of decreased CXCL12 and OPN expression at the tumor/normal tissue interface. This was also associated with greater CD8+ T-cell infiltration in tumors with less fibrosis. Antibody-mediated OPN blockade slowed tumor growth by increased intratumoral CD8+ T-cell activation. The CXCL12/OPN axis is an important node of immune/matrisomal cross-talk in the development of fibrosis. Therapeutic manipulation of this axis may offer greater antitumor efficacy while also reducing adverse effects.
Collapse
Affiliation(s)
- James T Paget
- Chester Beatty Laboratories, Targeted Therapy Team, The Institute of Cancer Research, London, United Kingdom
| | - Joseph A Ward
- Chester Beatty Laboratories, Targeted Therapy Team, The Institute of Cancer Research, London, United Kingdom
| | - Andrew R McKean
- Chester Beatty Laboratories, Targeted Therapy Team, The Institute of Cancer Research, London, United Kingdom
| | - David C Mansfield
- Chester Beatty Laboratories, Targeted Therapy Team, The Institute of Cancer Research, London, United Kingdom
| | - Martin McLaughlin
- Chester Beatty Laboratories, Targeted Therapy Team, The Institute of Cancer Research, London, United Kingdom
| | - Joan N Kyula-Currie
- Chester Beatty Laboratories, Targeted Therapy Team, The Institute of Cancer Research, London, United Kingdom
| | - Henry G Smith
- Chester Beatty Laboratories, Targeted Therapy Team, The Institute of Cancer Research, London, United Kingdom
| | - Victoria Roulstone
- Chester Beatty Laboratories, Targeted Therapy Team, The Institute of Cancer Research, London, United Kingdom
| | - Chunhei Li
- Division of Infection and Immunity, School of Medicine, Systems Immunity University Research Institute, Cardiff University, Cardiff, United Kingdom
| | - You Zhou
- Division of Infection and Immunity, School of Medicine, Systems Immunity University Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Thomas Hardiman
- Cancer Bioinformatics Group, King's College, London, United Kingdom
| | | | - Devin O'Brien Coon
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Johns Hopkins University Whiting School of Engineering, Baltimore, Maryland
| | - Sheeba Irshad
- Division of Cancer and Pharmaceutical Sciences, King's College, London, United Kingdom
| | - Alan A Melcher
- Chester Beatty Laboratories, Targeted Therapy Team, The Institute of Cancer Research, London, United Kingdom
| | - Kevin J Harrington
- Chester Beatty Laboratories, Targeted Therapy Team, The Institute of Cancer Research, London, United Kingdom
| | - Aadil Khan
- Chester Beatty Laboratories, Targeted Therapy Team, The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
3
|
Zhao T, Su Y. Mechanisms and Therapeutic Potential of Myofibroblast Transformation in Pulmonary Fibrosis. JOURNAL OF RESPIRATORY BIOLOGY AND TRANSLATIONAL MEDICINE 2025; 2:10001. [PMID: 40190620 PMCID: PMC11970920 DOI: 10.70322/jrbtm.2025.10001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive, irreversible, and fatal disease with an increasing incidence and limited therapeutic options. It is characterized by the formation and deposition of excess extracellular matrix proteins resulting in the gradual replacement of normal lung architecture by fibrous tissue. The cellular and molecular mechanism of IPF has not been fully understood. A hallmark in IPF is pulmonary fibroblast to myofibroblast transformation (FMT). During excessive lung repair upon exposure to harmful stimuli, lung fibroblasts transform into myofibroblasts under stimulation of cytokines, chemokines, and vesicles from various cells. These mediators interact with lung fibroblasts, initiating multiple signaling cascades, such as TGFβ1, MAPK, Wnt/β-catenin, NF-κB, AMPK, endoplasmic reticulum stress, and autophagy, contributing to lung FMT. Furthermore, single-cell transcriptomic analysis has revealed significant heterogeneity among lung myofibroblasts, which arise from various cell types and are adapted to the altered microenvironment during pathological lung repair. This review provides an overview of recent research on the origins of lung myofibroblasts and the molecular pathways driving their formation, with a focus on the interactions between lung fibroblasts and epithelial cells, endothelial cells, and macrophages in the context of lung fibrosis. Based on these molecular insights, targeting the lung FMT could offer promising avenues for the treatment of IPF.
Collapse
Affiliation(s)
- Tianming Zhao
- Department of Pharmacology & Toxicology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| | - Yunchao Su
- Department of Pharmacology & Toxicology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
- Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30912, USA
| |
Collapse
|
4
|
Russo RC, Ryffel B. The Chemokine System as a Key Regulator of Pulmonary Fibrosis: Converging Pathways in Human Idiopathic Pulmonary Fibrosis (IPF) and the Bleomycin-Induced Lung Fibrosis Model in Mice. Cells 2024; 13:2058. [PMID: 39768150 PMCID: PMC11674266 DOI: 10.3390/cells13242058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and lethal interstitial lung disease (ILD) of unknown origin, characterized by limited treatment efficacy and a fibroproliferative nature. It is marked by excessive extracellular matrix deposition in the pulmonary parenchyma, leading to progressive lung volume decline and impaired gas exchange. The chemokine system, a network of proteins involved in cellular communication with diverse biological functions, plays a crucial role in various respiratory diseases. Chemokine receptors trigger the activation, proliferation, and migration of lung-resident cells, including pneumocytes, endothelial cells, alveolar macrophages, and fibroblasts. Around 50 chemokines can potentially interact with 20 receptors, expressed by both leukocytes and non-leukocytes such as tissue parenchyma cells, contributing to processes such as leukocyte mobilization from the bone marrow, recirculation through lymphoid organs, and tissue influx during inflammation or immune response. This narrative review explores the complexity of the chemokine system in the context of IPF and the bleomycin-induced lung fibrosis mouse model. The goal is to identify specific chemokines and receptors as potential therapeutic targets. Recent progress in understanding the role of the chemokine system during IPF, using experimental models and molecular diagnosis, underscores the complex nature of this system in the context of the disease. Despite advances in experimental models and molecular diagnostics, discovering an effective therapy for IPF remains a significant challenge in both medicine and pharmacology. This work delves into microarray results from lung samples of IPF patients and murine samples at different stages of bleomycin-induced pulmonary fibrosis. By discussing common pathways identified in both IPF and the experimental model, we aim to shed light on potential targets for therapeutic intervention. Dysregulation caused by abnormal chemokine levels observed in IPF lungs may activate multiple targets, suggesting that chemokine signaling plays a central role in maintaining or perpetuating lung fibrogenesis. The highlighted chemokine axes (CCL8-CCR2, CCL19/CCL21-CCR7, CXCL9-CXCR3, CCL3/CCL4/CCL5-CCR5, and CCL20-CCR6) present promising opportunities for advancing IPF treatment research and uncovering new pharmacological targets within the chemokine system.
Collapse
Affiliation(s)
- Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais-UFMG, Belo Horizonte 31270-901, MG, Brazil
| | - Bernhard Ryffel
- Laboratory of Immuno-Neuro Modulation (INEM), UMR7355 Centre National de la Recherche Scientifique (CNRS), University of Orleans, 45071 Orleans, France
| |
Collapse
|
5
|
Xie K, Tan X, Chen Z, Yao Y, Luo J, Ma H, Feng Y, Jiang W. Exploring the Mechanisms and Preventive Strategies for the Progression from Idiopathic Pulmonary Fibrosis to Lung Cancer: Insights from Transcriptomics and Genetic Factors. Biomedicines 2024; 12:2382. [PMID: 39457694 PMCID: PMC11504276 DOI: 10.3390/biomedicines12102382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/06/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Idiopathic pulmonary fibrosis (IPF) leads to excessive fibrous tissue in the lungs, increasing the risk of lung cancer (LC) due to heightened fibroblast activity. Advances in nucleotide point mutation studies offer insights into fibrosis-to-cancer transitions. Methods: A two-sample Mendelian randomization (TSMR) approach was used to explore the causal relationship between IPF and LC. A weighted gene co-expression network analysis (WGCNA) identified shared gene modules related to immunogenic cell death (ICD) from transcriptomic datasets. Machine learning selected key genes, and a multi-layer perceptron (MLP) model was developed for IPF prediction and diagnosis. SMR and PheWAS were used to assess the expression of key genes concerning IPF risk. The impact of core genes on immune cells in the IPF microenvironment was explored, and in vivo experiments were conducted to examine the progression from IPF to LC. Results: The TSMR approach indicated a genetic predisposition for IPF progressing to LC. The predictive model, which includes eight ICD key genes, demonstrated a strong predictive capability (AUC = 0.839). The SMR analysis revealed that the elevated expression of MS4A4A was associated with an increased risk of IPF (OR = 1.275, 95% CI: 1.029-1.579; p = 0.026). The PheWAS did not identify any significant traits linked to MS4A4A expression. The rs9265808 locus in MS4A4A was identified as a susceptibility site for the progression of IPF to LC, with mutations potentially reprogramming lung neutrophils and increasing the LC risk. In vivo studies suggested MS4A4A as a promising therapeutic target. Conclusions: A causal link between IPF and LC was established, an effective prediction model was developed, and MS4A4A was highlighted as a therapeutic target to prevent IPF from progressing to LC.
Collapse
Affiliation(s)
- Kai Xie
- Department of Thoracic and Cardiovascular Surgery, Medical Center of Soochow University, Suzhou 215000, China; (K.X.); (X.T.); (Z.C.); (H.M.)
| | - Xiaoyan Tan
- Department of Thoracic and Cardiovascular Surgery, Medical Center of Soochow University, Suzhou 215000, China; (K.X.); (X.T.); (Z.C.); (H.M.)
| | - Zhe Chen
- Department of Thoracic and Cardiovascular Surgery, Medical Center of Soochow University, Suzhou 215000, China; (K.X.); (X.T.); (Z.C.); (H.M.)
| | - Yu Yao
- Department of Respiratory Medicine, Nanjing University of Chinese Medicine, Nanjing 210000, China;
| | - Jing Luo
- Department of Cardiothoracic Surgery, Medical School of Nanjing University, Nanjing 210002, China;
| | - Haitao Ma
- Department of Thoracic and Cardiovascular Surgery, Medical Center of Soochow University, Suzhou 215000, China; (K.X.); (X.T.); (Z.C.); (H.M.)
- Department of The First Clinical, Medical College of Soochow University, Suzhou 215006, China
| | - Yu Feng
- Department of The First Clinical, Medical College of Soochow University, Suzhou 215006, China
| | - Wei Jiang
- Department of Thoracic and Cardiovascular Surgery, Medical Center of Soochow University, Suzhou 215000, China; (K.X.); (X.T.); (Z.C.); (H.M.)
| |
Collapse
|
6
|
Chaudary N, Hill RP, Milosevic M. Targeting the CXCL12/CXCR4 pathway to reduce radiation treatment side effects. Radiother Oncol 2024; 194:110194. [PMID: 38447871 DOI: 10.1016/j.radonc.2024.110194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/08/2024]
Abstract
High precision, image-guided radiotherapy (RT) has increased the therapeutic ratio, enabling higher tumor and lower normal tissue doses, leading to improved patient outcomes. Nevertheless, some patients remain at risk of developing serious side effects.In many clinical situations, the radiation tolerance of normal tissues close to the target volume limits the dose that can safely be delivered and thus the potential for tumor control and cure. This is particularly so in patients being re-treated for tumor progression or a second primary tumor within a previous irradiated volume, scenarios that are becoming more frequent in clinical practice.Various normal tissue 'radioprotective' drugs with the potential to reduce side effects have been studied previously. Unfortunately, most have failed to impact clinical practice because of lack of therapeutic efficacy, concern about concurrent tumor protection or excessive drug-related toxicity. This review highlights the evidence indicating that targeting the CXCL12/CXCR4 pathway can mitigate acute and late RT-induced injury and reduce treatment side effects in a manner that overcomes these previous translational challenges. Pre-clinical studies involving a broad range of normal tissues commonly affected in clinical practice, including skin, lung, the gastrointestinal tract and brain, have shown that CXCL12 signalling is upregulated by RT and attracts CXCR4-expressing inflammatory cells that exacerbate acute tissue injury and late fibrosis. These studies also provide convincing evidence that inhibition of CXCL12/CXCR4 signalling during or after RT can reduce or prevent RT side effects, warranting further evaluation in clinical studies. Greater dialogue with the pharmaceutical industry is needed to prioritize the development and availability of CXCL12/CXCR4 inhibitors for future RT studies.
Collapse
Affiliation(s)
- Naz Chaudary
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Richard P Hill
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Michael Milosevic
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
7
|
Wang X, Zhang X, Liu Z, Zhao N, Li X, Su P, Zheng G, Zhang X, Wang H, Zhang Y. Naringenin nanoparticles targeting cyclin B1 suppress the progression of rheumatoid arthritis-associated lung cancer by inhibiting fibroblast-to-myofibroblast transition. Int J Biochem Cell Biol 2024; 169:106557. [PMID: 38460905 DOI: 10.1016/j.biocel.2024.106557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/28/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024]
Abstract
There is growing evidence of an elevated risk of lung cancer in patients with rheumatoid arthritis. The poor prognosis of rheumatoid arthritis-associated lung cancer and the lack of therapeutic options pose an even greater challenge to the clinical management of patients. This study aimed to identify potential molecular targets associated with the progression of rheumatoid arthritis-associated lung cancer and examine the efficacy of naringenin nanoparticles targeting cyclin B1. Mendelian randomizatio analysis revealed that rheumatoid arthritis has a positive correlation with the risk of lung cancer. Cyclin B1 was significantly upregulated in patients with rheumatoid arthritis-associated lung cancer and was significantly overexpressed in synovial tissue fibroblasts. Furthermore, the overexpression of cyclin B1 in rheumatoid arthritis fibroblast-like synoviocytes, which promotes their proliferation and fibroblast-to-myofibroblast transition, can significantly contribute to the growth and infiltration of lung cancer cells. Importantly, our prepared naringenin nanoparticles targeting cyclin B1 effectively attenuated proliferation and fibroblast-to-myofibroblast transition by blocking cells at the G2/M phase. In vivo experiments, naringenin nanoparticles targeting cyclin B1 significantly alleviated the development of collagen-induced arthritis and lung orthotopic tumors. Collectively, our results reveal that naringenin nanoparticles targeting cyclin B1 can suppress the progression of rheumatoid arthritis-associated lung cancer by inhibiting fibroblast-to-myofibroblast transition. These findings provide new insights into the treatment of rheumatoid arthritis-associated lung cancer therapy.
Collapse
Affiliation(s)
- Xilong Wang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012, China
| | - Xiaoyu Zhang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Zhipu Liu
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012, China
| | - Na Zhao
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012, China
| | - Xiaohan Li
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012, China
| | - Peng Su
- Department of Pathology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Guixi Zheng
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012, China
| | - Xin Zhang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012, China
| | - Hongxing Wang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012, China.
| | - Yi Zhang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012, China.
| |
Collapse
|
8
|
Sharma P, Otto M. Multifunctional nanocomposites modulating the tumor microenvironment for enhanced cancer immunotherapy. Bioact Mater 2024; 31:440-462. [PMID: 37701452 PMCID: PMC10494322 DOI: 10.1016/j.bioactmat.2023.08.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/09/2023] [Accepted: 08/28/2023] [Indexed: 09/14/2023] Open
Abstract
Cancer immunotherapy has gained momentum for treating malignant tumors over the past decade. Checkpoint blockade and chimeric antigen receptor cell therapy (CAR-T) have shown considerable potency against liquid and solid cancers. However, the tumor microenvironment (TME) is highly immunosuppressive and hampers the effect of currently available cancer immunotherapies on overall treatment outcomes. Advancements in the design and engineering of nanomaterials have opened new avenues to modulate the TME. Progress in the current nanocomposite technology can overcome immunosuppression and trigger robust immunotherapeutic responses by integrating synergistic functions of different molecules. We will review recent advancements in nanomedical applications and discuss specifically designed nanocomposites modulating the TME for cancer immunotherapy. In addition, we provide information on the current landscape of clinical-stage nanocomposites for cancer immunotherapy.
Collapse
Affiliation(s)
- Prashant Sharma
- Department of Child Health, University of Arizona College of Medicine-Phoenix, ABC1 Building, 425 N 5th Street, Phoenix, AZ, 85004, USA
| | - Mario Otto
- Department of Child Health, University of Arizona College of Medicine-Phoenix, ABC1 Building, 425 N 5th Street, Phoenix, AZ, 85004, USA
- Center for Cancer and Blood Disorders (CCBD), Phoenix Children's, 1919 E Thomas Rd, Phoenix, AZ, 85016, USA
| |
Collapse
|
9
|
Eyraud E, Maurat E, Sac-Epée JM, Henrot P, Zysman M, Esteves P, Trian T, Dupuy JW, Leipold A, Saliba AE, Begueret H, Girodet PO, Thumerel M, Hustache-Castaing R, Marthan R, Levet F, Vallois P, Contin-Bordes C, Berger P, Dupin I. Short-range interactions between fibrocytes and CD8 + T cells in COPD bronchial inflammatory response. eLife 2023; 12:RP85875. [PMID: 37494277 PMCID: PMC10371228 DOI: 10.7554/elife.85875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023] Open
Abstract
Bronchi of chronic obstructive pulmonary disease (COPD) are the site of extensive cell infiltration, allowing persistent contact between resident cells and immune cells. Tissue fibrocytes interaction with CD8+ T cells and its consequences were investigated using a combination of in situ, in vitro experiments and mathematical modeling. We show that fibrocytes and CD8+ T cells are found in the vicinity of distal airways and that potential interactions are more frequent in tissues from COPD patients compared to those of control subjects. Increased proximity and clusterization between CD8+ T cells and fibrocytes are associated with altered lung function. Tissular CD8+ T cells from COPD patients promote fibrocyte chemotaxis via the CXCL8-CXCR1/2 axis. Live imaging shows that CD8+ T cells establish short-term interactions with fibrocytes, that trigger CD8+ T cell proliferation in a CD54- and CD86-dependent manner, pro-inflammatory cytokines production, CD8+ T cell cytotoxic activity against bronchial epithelial cells and fibrocyte immunomodulatory properties. We defined a computational model describing these intercellular interactions and calibrated the parameters based on our experimental measurements. We show the model's ability to reproduce histological ex vivo characteristics, and observe an important contribution of fibrocyte-mediated CD8+ T cell proliferation in COPD development. Using the model to test therapeutic scenarios, we predict a recovery time of several years, and the failure of targeting chemotaxis or interacting processes. Altogether, our study reveals that local interactions between fibrocytes and CD8+ T cells could jeopardize the balance between protective immunity and chronic inflammation in the bronchi of COPD patients.
Collapse
Affiliation(s)
- Edmée Eyraud
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC1401, Proteomics Facility, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Pessac, France
| | - Elise Maurat
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC1401, Proteomics Facility, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Pessac, France
| | - Jean-Marc Sac-Epée
- Univ-Lorraine, Institut Elie Cartan de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Pauline Henrot
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC1401, Proteomics Facility, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Pessac, France
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Würzburg, Germany
| | - Maeva Zysman
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC1401, Proteomics Facility, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Pessac, France
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Würzburg, Germany
| | - Pauline Esteves
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC1401, Proteomics Facility, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Pessac, France
| | - Thomas Trian
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC1401, Proteomics Facility, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Pessac, France
| | - Jean-William Dupuy
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC1401, Proteomics Facility, Pessac, France
| | - Alexander Leipold
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Würzburg, Germany
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Würzburg, Germany
| | - Hugues Begueret
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC1401, Proteomics Facility, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Pessac, France
- CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Pessac, France
| | - Pierre-Olivier Girodet
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC1401, Proteomics Facility, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Pessac, France
- CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Pessac, France
| | - Matthieu Thumerel
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC1401, Proteomics Facility, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Pessac, France
- CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Pessac, France
| | - Romain Hustache-Castaing
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC1401, Proteomics Facility, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Pessac, France
- CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Pessac, France
| | - Roger Marthan
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC1401, Proteomics Facility, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Pessac, France
- CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Pessac, France
| | - Florian Levet
- Univ. Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, Bordeaux, France
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, Bordeaux, France
| | - Pierre Vallois
- Univ-Lorraine, Institut Elie Cartan de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Cécile Contin-Bordes
- CNRS, UMR5164 ImmunoConcEpT, Université de Bordeaux, Bordeaux, France
- CHU de Bordeaux, Laboratoire d'Immunologie et Immunogénétique, Bordeaux, France
| | - Patrick Berger
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Département de Pharmacologie, CIC1401, Proteomics Facility, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Pessac, France
- CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Pessac, France
| | - Isabelle Dupin
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Pessac, France
| |
Collapse
|
10
|
Gomez-Manjarres DC, Axell-House DB, Patel DC, Odackal J, Yu V, Burdick MD, Mehrad B. Sirolimus suppresses circulating fibrocytes in idiopathic pulmonary fibrosis in a randomized controlled crossover trial. JCI Insight 2023; 8:e166901. [PMID: 36853800 PMCID: PMC10243828 DOI: 10.1172/jci.insight.166901] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/23/2023] [Indexed: 03/01/2023] Open
Abstract
BACKGROUNDFibrocytes are BM-derived circulating cells that traffic to the injured lungs and contribute to fibrogenesis. The mTOR inhibitor, sirolimus, inhibits fibrocyte CXCR4 expression, reducing fibrocyte traffic and attenuating lung fibrosis in animal models. We sought to test the hypothesis that short-term treatment with sirolimus reduces the concentration of CXCR4+ circulating fibrocytes in patients with idiopathic pulmonary fibrosis (IPF).METHODSWe conducted a short-term randomized double-blind placebo-controlled crossover pilot trial to assess the safety and tolerability of sirolimus in IPF. Participants were randomly assigned to sirolimus or placebo for approximately 6 weeks, and after a 4-week washout, they were assigned to the alternate treatment. Toxicity, lung function, and the concentration of circulating fibrocytes were measured before and after each treatment.RESULTSIn the 28 study participants, sirolimus resulted in a statistically significant 35% decline in the concentration of total fibrocytes, 34% decline in CXCR4+ fibrocytes, and 42% decline in fibrocytes expressing α-smooth muscle actin, but no significant change in these populations occurred on placebo. Respiratory adverse events occurred more frequently during treatment with placebo than sirolimus; the incidence of adverse events and drug tolerability did not otherwise differ during therapy with drug and placebo. Lung function was unaffected by either treatment, with the exception of a small decline in gas transfer during treatment with placebo.CONCLUSIONAs compared with placebo, short-term treatment with sirolimus resulted in reduction of circulating fibrocyte concentrations in participants with IPF, with an acceptable safety profile.TRIAL REGISTRATIONClinicalTrials.gov, accession no. NCT01462006.FUNDINGNIH R01HL098329 and American Heart Association 18TPA34170486.
Collapse
Affiliation(s)
- Diana C. Gomez-Manjarres
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, Florida, USA
| | - Dierdre B. Axell-House
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Divya C. Patel
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, Florida, USA
| | - John Odackal
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Victor Yu
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Marie D. Burdick
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, Florida, USA
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Borna Mehrad
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, Florida, USA
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
11
|
Ahmedy OA, Kamel MW, Abouelfadl DM, Shabana ME, Sayed RH. Berberine attenuates epithelial mesenchymal transition in bleomycin-induced pulmonary fibrosis in mice via activating A 2aR and mitigating the SDF-1/CXCR4 signaling. Life Sci 2023; 322:121665. [PMID: 37028546 DOI: 10.1016/j.lfs.2023.121665] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/15/2023] [Accepted: 03/31/2023] [Indexed: 04/09/2023]
Abstract
AIMS Berberine is endowed with anti-oxidant, anti-inflammatory and anti-fibrotic effects. This study explored the role of adenosine A2a receptor (A2aR) activation and SDF-1/CXCR4 signaling suppression in the protective effects of berberine in bleomycin-induced pulmonary fibrosis in mice. MAIN METHODS Pulmonary fibrosis was generated in mice by injecting bleomycin (40 U/kg, i.p.) on days 0, 3, 7, 10 and 14. Mice were treated with berberine (5 mg/kg, i.p.) from day 15 to day 28. KEY FINDINGS Severe lung fibrosis and increased collagen content were observed in the bleomycin-challenged mice. Pulmonary A2aR downregulation was documented in bleomycin-induced pulmonary fibrosis animals and was accompanied by enhanced expression of SDF-1/CXCR4. Moreover, TGF-β1elevation and pSmad2/3 overexpression were reported in parallel with enhanced epithelial mesenchymal transition (EMT) markers expression, vimentin and α-SMA. Besides, bleomycin significantly elevated the inflammatory and pro-fibrogenic mediator NF-κB p65, TNF-α and IL-6. Furthermore, bleomycin administration induced oxidative stress as depicted by decreased Nrf2, SOD, GSH and catalase levels. Interestingly, berberine administration markedly ameliorated the fibrotic changes in lungs by modulating the purinergic system through the inhibition of A2aR downregulation, mitigating EMT and effectively suppressing inflammation and oxidative stress. Strikingly, A2aR blockade by SCH 58261, impeded the pulmonary protective effect of berberine. SIGNIFICANCE These findings indicated that berberine could attenuate the pathological processes of bleomycin-induced pulmonary fibrosis at least partially via upregulating A2aR and mitigating the SDF-1/CXCR4 related pathway, suggesting A2aR as a potential therapeutic target for the management of pulmonary fibrosis.
Collapse
Affiliation(s)
- Omaima A Ahmedy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, 11562 Cairo, Egypt.
| | - Marwa W Kamel
- Department of Cancer Biology, Pharmacology Unit, National Cancer Institute, Cairo University, 11796, Egypt
| | - Dalia M Abouelfadl
- Department of Pathology, Medical and Clinical Studies, Research Institute, National Research Center, Egypt
| | - Marwa E Shabana
- Department of Pathology, Medical and Clinical Studies, Research Institute, National Research Center, Egypt
| | - Rabab H Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, 11562 Cairo, Egypt
| |
Collapse
|
12
|
Islam MA, Kibria MK, Hossen MB, Reza MS, Tasmia SA, Tuly KF, Mosharof MP, Kabir SR, Kabir MH, Mollah MNH. Bioinformatics-based investigation on the genetic influence between SARS-CoV-2 infections and idiopathic pulmonary fibrosis (IPF) diseases, and drug repurposing. Sci Rep 2023; 13:4685. [PMID: 36949176 PMCID: PMC10031699 DOI: 10.1038/s41598-023-31276-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 03/09/2023] [Indexed: 03/24/2023] Open
Abstract
Some recent studies showed that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections and idiopathic pulmonary fibrosis (IPF) disease might stimulate each other through the shared genes. Therefore, in this study, an attempt was made to explore common genomic biomarkers for SARS-CoV-2 infections and IPF disease highlighting their functions, pathways, regulators and associated drug molecules. At first, we identified 32 statistically significant common differentially expressed genes (cDEGs) between disease (SARS-CoV-2 and IPF) and control samples of RNA-Seq profiles by using a statistical r-package (edgeR). Then we detected 10 cDEGs (CXCR4, TNFAIP3, VCAM1, NLRP3, TNFAIP6, SELE, MX2, IRF4, UBD and CH25H) out of 32 as the common hub genes (cHubGs) by the protein-protein interaction (PPI) network analysis. The cHubGs regulatory network analysis detected few key TFs-proteins and miRNAs as the transcriptional and post-transcriptional regulators of cHubGs. The cDEGs-set enrichment analysis identified some crucial SARS-CoV-2 and IPF causing common molecular mechanisms including biological processes, molecular functions, cellular components and signaling pathways. Then, we suggested the cHubGs-guided top-ranked 10 candidate drug molecules (Tegobuvir, Nilotinib, Digoxin, Proscillaridin, Simeprevir, Sorafenib, Torin 2, Rapamycin, Vancomycin and Hesperidin) for the treatment against SARS-CoV-2 infections with IFP diseases as comorbidity. Finally, we investigated the resistance performance of our proposed drug molecules compare to the already published molecules, against the state-of-the-art alternatives publicly available top-ranked independent receptors by molecular docking analysis. Molecular docking results suggested that our proposed drug molecules would be more effective compare to the already published drug molecules. Thus, the findings of this study might be played a vital role for diagnosis and therapies of SARS-CoV-2 infections with IPF disease as comorbidity risk.
Collapse
Affiliation(s)
- Md Ariful Islam
- Bioinformatics Lab(Dry), Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Md Kaderi Kibria
- Bioinformatics Lab(Dry), Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Md Bayazid Hossen
- Bioinformatics Lab(Dry), Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Md Selim Reza
- Bioinformatics Lab(Dry), Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Samme Amena Tasmia
- Bioinformatics Lab(Dry), Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Khanis Farhana Tuly
- Bioinformatics Lab(Dry), Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Md Parvez Mosharof
- Bioinformatics Lab(Dry), Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
- School of Business, University of Southern Queensland, Toowoomba, QLD, 4350, Australia
| | - Syed Rashel Kabir
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Md Hadiul Kabir
- Bioinformatics Lab(Dry), Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Md Nurul Haque Mollah
- Bioinformatics Lab(Dry), Department of Statistics, University of Rajshahi, Rajshahi, 6205, Bangladesh.
| |
Collapse
|
13
|
Zhang Y, Wu Z, Lu S, Lin M, Yue X, Wang Z, Cai B. Time-Series Expression Profile Analysis of Post-Traumatic Joint Contracture in Rats at the Early Stages of the Healing Process. J Inflamm Res 2023; 16:1169-1181. [PMID: 36945316 PMCID: PMC10024884 DOI: 10.2147/jir.s400557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 03/09/2023] [Indexed: 03/17/2023] Open
Abstract
Objective This study aimed to characterize the gene expression profile at the early stages of the healing process of post-traumatic joint contracture (PTJC). Methods Twelve rats were used for PTJC model establishment and were divided into four groups according to the sampling time: S0d, S3d, S7d and S2w. Transcriptome sequencing was performed on fibrotic joint capsule samples in four groups followed by bioinformatics analyses including differentially expressed genes (DEGs) screening, Short Time-series Expression Miner (STEM) analysis, network construction, and pathway analysis. Five important genes were validated by qRT-PCR. Results A total of 1171, 1052 and 793 DEGs were screened in S3d vs S0d, S7d vs S0d, and S2w vs S0d comparison groups, respectively. A total of 383 overlapping genes were screened out, which were significantly enriched in some inflammatory functions and pathways. Through STEM analysis, three clusters were identified, including 105, 57 and 57 DEGs, respectively. Then, based on the cluster genes, 10 genes, such as Il6, Timp1, Cxcl1, Cxcr4 and Mmp3, were further selected after PPI and pathway analyses. The expression levels of Il6, Timp1, Cxcl1, Cxcr4 and Mmp3 were validated by qRT-PCR. Conclusion The present study screened out several genes with significant changes in expression levels at the early stages of the healing process in PTJC, such as Il6, Timp1, Cxcl1, Cxcr4 and Mmp3. Our study offers a valuable contribution to the understanding pathomechanism of PTJC.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Rehabilitation Medicine, Hainan Western Central Hospital, Danzhou, Hainan, People’s Republic of China
- Department of Rehabilitation Medicine, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Department of Oral Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, Shanghai, People’s Republic of China
- Correspondence: Yuxin Zhang; Bin Cai, Department of Rehabilitation Medicine, Hainan Western Central Hospital, No. 2, Fubo East Road, Nada Town, Danzhou, Hainan, 571700, People’s Republic of China, Tel +86-21-53315248, Email ;
| | - Zhigang Wu
- Department of Rehabilitation Medicine, Hainan Western Central Hospital, Danzhou, Hainan, People’s Republic of China
| | - Shenji Lu
- Department of Rehabilitation Medicine, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Minghui Lin
- Department of Rehabilitation Medicine, Hainan Western Central Hospital, Danzhou, Hainan, People’s Republic of China
| | - Xiaokun Yue
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Zengguang Wang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Bin Cai
- Department of Rehabilitation Medicine, Hainan Western Central Hospital, Danzhou, Hainan, People’s Republic of China
- Department of Rehabilitation Medicine, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
14
|
Septembre-Malaterre A, Boina C, Douanier A, Gasque P. Deciphering the Antifibrotic Property of Metformin. Cells 2022; 11:cells11244090. [PMID: 36552855 PMCID: PMC9777391 DOI: 10.3390/cells11244090] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/14/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Fibrosis is a chronic progressive and incurable disease leading to organ dysfunction. It is characterized by the accumulation of extracellular matrix proteins produced by mesenchymal stem cells (MSCs) differentiating into myofibroblasts. Given the complexity of its pathophysiology, the search for effective treatments for fibrosis is of paramount importance. Metformin, a structural dimethyl analog of the galegine guanide extracted from the "French Lilac" (Fabaceae Galega officinalis), is the most widely used antidiabetic drug, recently recognized for its antifibrotic effects through ill-characterized mechanisms. The in vitro model of TGF-β1-induced fibrosis in human primary pulmonary mesenchymal stem cells (HPMSCs), identified as CD248+ and CD90+ cells, was used to study the effects of metformin extracts. These effects were tested on the expression of canonical MSC differentiation markers, immune/inflammatory factors and antioxidative stress molecules using qRT-PCR (mRNA, miRNA), immunofluorescence and ELISA experiments. Interestingly, metformin is able to reduce/modulate the expression of different actors involved in fibrosis. Indeed, TGF-β1 effects were markedly attenuated by metformin, as evidenced by reduced expression of three collagen types and Acta2 mRNAs. Furthermore, metformin attenuated the effects of TGF-β1 on the expression of PDGF, VEGF, erythropoietin, calcitonin and profibrotic miRs, possibly by controlling the expression of several key TGF/Smad factors. The expression of four major fibrogenic MMPs was also reduced by metformin treatment. In addition, metformin controlled MSC differentiation into lipofibroblasts and osteoblasts and had the ability to restore redox balance via the Nox4/Nrf2, AMP and Pi3K pathways. Overall, these results show that metformin is a candidate molecule for antifibrotic effect and/or aiming to combat the development of chronic inflammatory diseases worldwide.
Collapse
Affiliation(s)
- Axelle Septembre-Malaterre
- Unité de Recherche, EPI ‘Etudes en Pharmaco-Immunologie’, Université de la Réunion, Allée des Topazes, CS11021, 97400 Saint Denis, France
- Laboratoire D’immunologie Clinique et Expérimentale de la Zone de L’océan Indien (LICE-OI), CHU La Réunion Site Félix Guyon Allée des Topazes, CS11021, 97400 Saint Denis, France
- Correspondence:
| | - Chailas Boina
- Unité de Recherche, EPI ‘Etudes en Pharmaco-Immunologie’, Université de la Réunion, Allée des Topazes, CS11021, 97400 Saint Denis, France
- Laboratoire D’immunologie Clinique et Expérimentale de la Zone de L’océan Indien (LICE-OI), CHU La Réunion Site Félix Guyon Allée des Topazes, CS11021, 97400 Saint Denis, France
| | - Audrey Douanier
- Unité de Recherche, EPI ‘Etudes en Pharmaco-Immunologie’, Université de la Réunion, Allée des Topazes, CS11021, 97400 Saint Denis, France
- Laboratoire D’immunologie Clinique et Expérimentale de la Zone de L’océan Indien (LICE-OI), CHU La Réunion Site Félix Guyon Allée des Topazes, CS11021, 97400 Saint Denis, France
| | - Philippe Gasque
- Unité de Recherche, EPI ‘Etudes en Pharmaco-Immunologie’, Université de la Réunion, Allée des Topazes, CS11021, 97400 Saint Denis, France
- Laboratoire D’immunologie Clinique et Expérimentale de la Zone de L’océan Indien (LICE-OI), CHU La Réunion Site Félix Guyon Allée des Topazes, CS11021, 97400 Saint Denis, France
| |
Collapse
|
15
|
Peng W, Kepsch A, Kracht TO, Hasan H, Wijayarathna R, Wahle E, Pleuger C, Bhushan S, Günther S, Kauerhof AC, Planinić A, Fietz D, Schuppe HC, Wygrecka M, Loveland KL, Ježek D, Meinhardt A, Hedger MP, Fijak M. Activin A and CCR2 regulate macrophage function in testicular fibrosis caused by experimental autoimmune orchitis. Cell Mol Life Sci 2022; 79:602. [PMID: 36434305 PMCID: PMC9700630 DOI: 10.1007/s00018-022-04632-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/21/2022] [Accepted: 11/13/2022] [Indexed: 11/27/2022]
Abstract
Experimental autoimmune-orchitis (EAO), a rodent model of chronic testicular inflammation and fibrosis, replicates pathogenic changes seen in some cases of human spermatogenic disturbances. During EAO, increased levels of pro-inflammatory and pro-fibrotic mediators such as TNF, CCL2, and activin A are accompanied by infiltration of leukocytes into the testicular parenchyma. Activin A levels correlate with EAO severity, while elevated CCL2 acting through its receptor CCR2 mediates leukocyte trafficking and recruits macrophages. CCR2 + CXCR4 + macrophages producing extracellular matrix proteins contribute widely to fibrogenesis. Furthermore, testicular macrophages (TMs) play a critical role in organ homeostasis. Therefore, we aimed to investigate the role of the activin A/CCL2-CCR2/macrophage axis in the development of testicular fibrosis. Following EAO induction, we observed lower levels of organ damage, collagen deposition, and leukocyte infiltration (including fibronectin+, collagen I+ and CXCR4+ TMs) in Ccr2-/- mice than in WT mice. Furthermore, levels of Il-10, Ccl2, and the activin A subunit Inhba mRNAs were lower in Ccr2-/- EAO testes. Notably, fibronectin+ TMs were also present in biopsies from patients with impaired spermatogenesis and fibrotic alterations. Overexpression of the activin A antagonist follistatin reduced tissue damage and collagen I+ TM accumulation in WT EAO testes, while treating macrophages with activin A in vitro increased the expression of Ccr2, Fn1, Cxcr4, and Mmp2 and enhanced migration along a CCL2 gradient; these effects were abolished by follistatin. Taken together, our data indicate that CCR2 and activin A promote fibrosis during testicular inflammation by regulating macrophage function. Inhibition of CCR2 or activin A protects against damage progression, offering a promising avenue for therapeutic intervention.
Collapse
Affiliation(s)
- Wei Peng
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
| | - Artem Kepsch
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
| | - Till O Kracht
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
| | - Hiba Hasan
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
| | - Rukmali Wijayarathna
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Eva Wahle
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
| | - Christiane Pleuger
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
| | - Sudhanshu Bhushan
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
| | - Stefan Günther
- ECCPS Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - A Christine Kauerhof
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Ana Planinić
- Department of Histology and Embryology, School of Medicine, University of Zagreb, Zagreb, Croatia
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Daniela Fietz
- Department of Veterinary Anatomy, Histology and Embryology, Justus Liebig University of Giessen, Giessen, Germany
| | - Hans-Christian Schuppe
- Department of Urology, Paediatric Urology and Andrology, Justus Liebig University of Giessen, Giessen, Germany
| | - Małgorzata Wygrecka
- Center for Infection and Genomics of the Lung, German Center for Lung Research, University of Giessen and Marburg Lung Center, Giessen, Germany
| | - Kate L Loveland
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Davor Ježek
- Department of Histology and Embryology, School of Medicine, University of Zagreb, Zagreb, Croatia
- Centre of Excellence for Reproductive and Regenerative Medicine, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Andreas Meinhardt
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Mark P Hedger
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Monika Fijak
- Department of Anatomy and Cell Biology, Justus Liebig University of Giessen, Aulweg 123, 35392, Giessen, Germany.
| |
Collapse
|
16
|
Agbogan VA, Gastineau P, Tejerina E, Karray S, Zavala F. CpG-Activated Regulatory B-Cell Progenitors Alleviate Murine Graft-Versus-Host-Disease. Front Immunol 2022; 13:790564. [PMID: 35479094 PMCID: PMC9035844 DOI: 10.3389/fimmu.2022.790564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Development of Graft Versus Host Disease (GVHD) represents a major impediment in allogeneic hematopoietic stem cell transplantation (HSCT). The observation that the presence of bone marrow and circulating hematogones correlated with reduced GVHD risks prompted us to evaluate whether B-cell progenitors, which provide protection in various autoimmune disease models following activation with the TLR-9 agonist CpG (CpG-proBs), could likewise reduce this allogeneic disorder. In a murine model of GVHD that recapitulates an initial phase of acute GVHD followed by a phase of chronic sclerodermatous GVHD, we found that CpG-proBs, adoptively transferred during the initial phase of disease, reduced the diarrhea score and mostly prevented cutaneous fibrosis. Progenitors migrated to the draining lymph nodes and to the skin where they mainly differentiated into follicular B cells. CpG activation and IFN-γ expression were required for the protective effect, which resulted in reduced CD4+ T-cell-derived production of critical cytokines such as TGF-β, IL-13 and IL-21. Adoptive transfer of CpG-proBs increased the T follicular regulatory to T follicular helper (Tfr/Tfh) ratio. Moreover, CpG-proBs privileged the accumulation of IL-10-positive CD8+ T cells, B cells and dendritic cells in the skin. However, CpG-proBs did not improve survival. Altogether, our findings support the notion that adoptively transferred CpG-proBs exert immunomodulating effect that alleviates symptoms of GVHD but require additional anti-inflammatory strategy to improve survival.
Collapse
Affiliation(s)
- Viviane A. Agbogan
- Université Paris Cité, INSERM U1151, CNRS UMR8152, Institut Necker Enfants Malades (INEM), Paris, France
| | - Pauline Gastineau
- Université Paris Cité, INSERM U1151, CNRS UMR8152, Institut Necker Enfants Malades (INEM), Paris, France
| | - Emmanuel Tejerina
- Université Paris Cité, INSERM U1151, CNRS UMR8152, Institut Necker Enfants Malades (INEM), Paris, France
| | - Saoussen Karray
- Université Paris Cité, INSERM U976, Institut de Recherche Saint-Louis (IRSL), Hôpital Saint-Louis, Paris, France
| | - Flora Zavala
- Université Paris Cité, INSERM U1151, CNRS UMR8152, Institut Necker Enfants Malades (INEM), Paris, France
- *Correspondence: Flora Zavala, ; orcid.org/0000-0002-2338-6802
| |
Collapse
|
17
|
Dominguez EC, Phandthong R, Nguyen M, Ulu A, Guardado S, Sveiven S, Talbot P, Nordgren TM. Aspirin-Triggered Resolvin D1 Reduces Chronic Dust-Induced Lung Pathology without Altering Susceptibility to Dust-Enhanced Carcinogenesis. Cancers (Basel) 2022; 14:1900. [PMID: 35454807 PMCID: PMC9032113 DOI: 10.3390/cancers14081900] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/28/2022] [Accepted: 04/01/2022] [Indexed: 02/05/2023] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide, with increased risk being associated with unresolved or chronic inflammation. Agricultural and livestock workers endure significant exposure to agricultural dusts on a routine basis; however, the chronic inflammatory and carcinogenic effects of these dust exposure is unclear. We have developed a chronic dust exposure model of lung carcinogenesis in which mice were intranasally challenged three times a week for 24 weeks, using an aqueous dust extract (HDE) made from dust collected in swine confinement facilities. We also treated mice with the omega-3-fatty acid lipid mediator, aspirin-triggered resolvin D1 (AT-RvD1) to provide a novel therapeutic strategy for mitigating the inflammatory and carcinogenic effects of HDE. Exposure to HDE resulted in significant immune cell influx into the lungs, enhanced lung tumorigenesis, severe tissue pathogenesis, and a pro-inflammatory and carcinogenic gene signature, relative to saline-exposed mice. AT-RvD1 treatment mitigated the dust-induced inflammatory response but did not protect against HDE + NNK-enhanced tumorigenesis. Our data suggest that chronic HDE exposure induces a significant inflammatory and pro-carcinogenic response, whereas treatment with AT-RvD1 dampens the inflammatory responses, providing a strong argument for the therapeutic use of AT-RvD1 to mitigate chronic inflammation.
Collapse
Affiliation(s)
- Edward C. Dominguez
- Environmental Toxicology Graduate Program, University of California Riverside, Riverside, CA 92521, USA; (E.C.D.); (P.T.)
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA 92521, USA; (M.N.); (A.U.); (S.G.); (S.S.)
| | - Rattapol Phandthong
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA 92521, USA;
| | - Matthew Nguyen
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA 92521, USA; (M.N.); (A.U.); (S.G.); (S.S.)
| | - Arzu Ulu
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA 92521, USA; (M.N.); (A.U.); (S.G.); (S.S.)
| | - Stephanie Guardado
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA 92521, USA; (M.N.); (A.U.); (S.G.); (S.S.)
| | - Stefanie Sveiven
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA 92521, USA; (M.N.); (A.U.); (S.G.); (S.S.)
| | - Prue Talbot
- Environmental Toxicology Graduate Program, University of California Riverside, Riverside, CA 92521, USA; (E.C.D.); (P.T.)
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA 92521, USA;
| | - Tara M. Nordgren
- Environmental Toxicology Graduate Program, University of California Riverside, Riverside, CA 92521, USA; (E.C.D.); (P.T.)
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA 92521, USA; (M.N.); (A.U.); (S.G.); (S.S.)
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
18
|
Qiu L, Gong G, Wu W, Li N, Li Z, Chen S, Li P, Chen T, Zhao H, Hu C, Fang Z, Wang Y, Liu H, Cui P, Zhang G. A novel prognostic signature for idiopathic pulmonary fibrosis based on five-immune-related genes. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1570. [PMID: 34790776 PMCID: PMC8576669 DOI: 10.21037/atm-21-4545] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/02/2021] [Indexed: 01/04/2023]
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a highly fatal lung disease of unknown etiology with a median survival after diagnosis of only 2–3 years. Its poor prognosis is due to the limited therapy options available as well as the lack of effective prognostic indicators. This study aimed to construct a novel prognostic signature for IPF to assist in the personalized management of IPF patients during treatment. Methods Differentially-expressed genes (DEGs) in IPF patients versus healthy individuals were analyzed using the “limma” package of R software. Immune-related genes (IRGs) were obtained from the ImmPort database. Univariate Cox regression analysis was adopted to screen significantly prognostic IRGs for IPF patients. Multiple Cox regression analysis was used to identify optimal prognostic IRGs and construct a prognostic signature. Results Compared with healthy individuals, there were a total of 52 prognosis-related DEGs in the bronchoalveolar lavage (BAL) samples of IPF patients, of which 37 genes were identified as IRGs. Of these, five genes (CXCL14, SLC40A1, RNASE3, CCR3, and RORA) were significantly associated with overall survival (OS) in IPF patients, and were utilized for establishment of the prognostic signature. IPF patients were divided into high- and low-risk groups based on the prognostic signature. Marked differences in the OS probability were observed between high- and low-risk IPF patients. The area under curves (AUCs) of the receiver operating characteristic (ROC) curve for the prognostic signature in the training and validation cohorts were 0.858 and 0.837, respectively. The expression levels between RNASE3 and SLC40A1 (P<0.01, r=0.394), between RORA and CXCL14 (P<0.01, r=−0.355), between CCR3 and CXCL14 (P<0.01, r=0.258), as well as between RNASE3 and CCR3 (P<0.01, r=0.293) were significantly correlated. Conclusions We developed a validated and reproducible IRG-based prognostic signature that should be helpful in the personalized management of patients with IPF, providing new insights into the relationship between the immune system and IPF.
Collapse
Affiliation(s)
- Lingxiao Qiu
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China.,Henan Provincial Medical Key Laboratory for Interstitial Lung Disease and Lung Transplantation, Zhengzhou, China
| | - Gencheng Gong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenjuan Wu
- Department of Geriatric Medicine, Henan Provincial People's Hospital, Zhengzhou, China
| | - Nana Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhaonan Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shanshan Chen
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Provincial Medical Key Laboratory for Interstitial Lung Disease and Lung Transplantation, Zhengzhou, China
| | - Ping Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Provincial Medical Key Laboratory for Interstitial Lung Disease and Lung Transplantation, Zhengzhou, China.,Zhengzhou Key Laboratory for Chronic Respiratory Disease, Zhengzhou, China
| | - Tengfei Chen
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huasi Zhao
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunling Hu
- Department of Respiratory Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zeming Fang
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongping Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Panpan Cui
- School of Nursing and Heath, Zhengzhou University, Zhengzhou, China
| | - Guojun Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Provincial Medical Key Laboratory for Interstitial Lung Disease and Lung Transplantation, Zhengzhou, China.,Zhengzhou Key Laboratory for Chronic Respiratory Disease, Zhengzhou, China
| |
Collapse
|
19
|
Campbell CA, Burdick MD, Strieter RM. Systemic Fibrocyte Levels and Keloid Expression of the Chemoattractant CXCL12 Are Upregulated Compared With Patients With Normal Scar. Ann Plast Surg 2021; 87:150-155. [PMID: 34253698 DOI: 10.1097/sap.0000000000002929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Fibrocytes are bone marrow mesenchymal precursors with a surface phenotype compatible with leukocytes, fibroblasts, and hematopoietic progenitors that have been shown to traffic to wound healing sites in response to described chemokine pathways. Keloids are focal fibrotic responses to cutaneous trauma characterized by disordered collagen, which may be associated with elevated systemic fibrocyte levels and/or wound bed chemokine expression. METHODS Blood specimens from patients with longstanding keloids and those who form grossly normal scars were assayed by fluorescence activated cell sorting analysis for fibrocytes (CD45+, Col I+). The expression of the fibrocyte chemotactic cell surface marker CXCR4, intracellular markers of fibroblast differentiation (pSMAD2/3), and plasma levels of the CXCR4 cognate CXCL12 were compared. Keloid specimens and grossly normal scars were excised, and local expression of CXCL12 was assayed. RESULTS Keloid-forming patients demonstrated a significantly greater number of circulating fibrocytes (17.4 × 105 cells/mL) than control patients (1.01 × 105 cells/mL, P = 0.004). The absolute number of fibrocytes expressing CXCR4 was significantly greater (P = 0.012) in keloid-forming patients. Systemic CXCL12 levels were insignificantly greater in keloid-forming patients than controls. Keloid specimens had significantly greater CXCL12 expression (529.3 pg/mL) than normal scar (undetectable). CONCLUSIONS Systemic fibrocyte levels and the CXCR4/CXCL12 biologic axis responsible for fibrocyte trafficking to areas of regional fibrosis were both upregulated in patients who form keloids compared with controls. Keloids persistently expressed CXLC12, which serves both as the main chemoattractant for fibrocytes and a downstream mediator for local inflammation, suggesting a role for this biologic axis in keloid formation and possibly recurrence.
Collapse
Affiliation(s)
| | | | - Robert M Strieter
- Novartis Institutes for BioMedical Research, Cambridge, United Kingdom
| |
Collapse
|
20
|
van der Ploeg EA, Melgert BN, Burgess JK, Gan CT. The potential of biomarkers of fibrosis in chronic lung allograft dysfunction. Transplant Rev (Orlando) 2021; 35:100626. [PMID: 33992914 DOI: 10.1016/j.trre.2021.100626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/30/2021] [Accepted: 05/01/2021] [Indexed: 11/27/2022]
Abstract
Chronic lung allograft dysfunction (CLAD) is the major long-term cause of morbidity and mortality after lung transplantation. Both bronchiolitis obliterans syndrome and restrictive lung allograft syndrome, two main types of CLAD, lead to fibrosis in either the small airways or alveoli and pleura. Pathological pathways in CLAD and other types of fibrosis, for example idiopathic pulmonary fibrosis, are assumed to overlap and therefore fibrosis biomarkers could aid in the early detection of CLAD. These biomarkers could help to differentiate between different phenotypes of CLAD and could, in comparison to biomarkers of inflammation, possibly distinguish an infectious event from CLAD when a decline in lung function is present. This review gives an overview of known CLAD specific biomarkers, describes new promising fibrosis biomarkers currently investigated in other types of fibrosis, and discusses the possible use of these fibrosis biomarkers for CLAD.
Collapse
Affiliation(s)
- Eline A van der Ploeg
- University of Groningen, University Medical Centre Groningen, Department of Pulmonary Medicine, PO Box 30. 001, 9700, RB, Groningen, the Netherlands.
| | - Barbro N Melgert
- University of Groningen, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, PO box 196, 9700, AD, Groningen, the Netherlands; University of Groningen, University Medical Centre Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, PO Box 30.001, 9700, RB, Groningen, the Netherlands.
| | - Janette K Burgess
- University of Groningen, University Medical Centre Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, PO Box 30.001, 9700, RB, Groningen, the Netherlands; University of Groningen, University Medical Centre Groningen, Department of Pathology and Medical Biology, PO Box 30.001, 9700, RB, Groningen, the Netherlands.
| | - C Tji Gan
- University of Groningen, University Medical Centre Groningen, Department of Pulmonary Medicine, PO Box 30. 001, 9700, RB, Groningen, the Netherlands.
| |
Collapse
|
21
|
Li Z, Liu Y, Fang X, Shu Z. Nanomaterials Enhance the Immunomodulatory Effect of Molecular Targeted Therapy. Int J Nanomedicine 2021; 16:1631-1661. [PMID: 33688183 PMCID: PMC7935456 DOI: 10.2147/ijn.s290346] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/23/2021] [Indexed: 01/22/2023] Open
Abstract
Molecular targeted therapy, a tumor therapy strategy that inhibits specific oncogenic targets, has been shown to modulate the immune response. In addition to directly inhibiting the proliferation and metastasis of tumor cells, molecular targeted drugs can activate the immune system through a variety of mechanisms, including by promoting tumor antigen processing and presentation, increasing intratumoral T cell infiltration, enhancing T cell activation and function, and attenuating the immunosuppressive effect of the tumor microenvironment. However, poor water solubility, insufficient accumulation at the tumor site, and nonspecific targeting of immune cells limit their application. To this end, a variety of nanomaterials have been developed to overcome these obstacles and amplify the immunomodulatory effects of molecular targeted drugs. In this review, we summarize the impact of molecular targeted drugs on the antitumor immune response according to their mechanisms, highlight the advantages of nanomaterials in enhancing the immunomodulatory effect of molecular targeted therapy, and discuss the current challenges and future prospects.
Collapse
Affiliation(s)
- Zhongmin Li
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| | - Yilun Liu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| | - Xuedong Fang
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| | - Zhenbo Shu
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| |
Collapse
|
22
|
Yang F, Hou ZF, Zhu HY, Chen XX, Li WY, Cao RS, Li YX, Chen R, Zhang W. Catalpol Protects Against Pulmonary Fibrosis Through Inhibiting TGF-β1/Smad3 and Wnt/β-Catenin Signaling Pathways. Front Pharmacol 2021; 11:594139. [PMID: 33584272 PMCID: PMC7878558 DOI: 10.3389/fphar.2020.594139] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/29/2020] [Indexed: 12/30/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal lung disease characterized by fibroblast proliferation and extracellular matrix remodeling; however, the molecular mechanisms underlying its occurrence and development are not yet fully understood. Despite it having a variety of beneficial pharmacological activities, the effects of catalpol (CAT), which is extracted from Rehmannia glutinosa, in IPF are not known. In this study, the differentially expressed genes, proteins, and pathways of IPF in the Gene Expression Omnibus database were analyzed, and CAT was molecularly docked with the corresponding key proteins to screen its pharmacological targets, which were then verified using an animal model. The results show that collagen metabolism imbalance, inflammatory response, and epithelial-mesenchymal transition (EMT) are the core processes in IPF, and the TGF-β1/Smad3 and Wnt/β-catenin pathways are the key signaling pathways for the development of pulmonary fibrosis. Our results also suggest that CAT binds to TGF-βR1, Smad3, Wnt3a, and GSK-3β through hydrogen bonds, van der Waals bonds, and other interactions to downregulate the expression and phosphorylation of Smad3, Wnt3a, GSK-3β, and β-catenin, inhibit the expression of cytokines, and reduce the degree of oxidative stress in lung tissue. Furthermore, CAT can inhibit the EMT process and collagen remodeling by downregulating fibrotic biomarkers and promoting the expression of epithelial cadherin. This study elucidates several key processes and signaling pathways involved in the development of IPF, and suggests the potential value of CAT in the treatment of IPF.
Collapse
Affiliation(s)
- Fan Yang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhen-Feng Hou
- College of Life Sciences, Shandong Normal University, Jinan, China
| | - Hao-Yue Zhu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiao-Xuan Chen
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wan-Yang Li
- School of Public Health, Xiangya Medical College, Central South University, Changsha, China
| | - Ren-Shuang Cao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yu-Xuan Li
- Second School of Clinical Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ru Chen
- Biomedical Research Institute of Fudan University, Shanghai, China
| | - Wei Zhang
- Department of Pulmonary Diseases, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
23
|
Liao J, Zhang Z, Yuan Q, Liu Q, Kuang J, Fang Y, Hu X. A lncRNA Gpr137b-ps/miR-200a-3p/CXCL14 axis modulates hepatic stellate cell (HSC) activation. Toxicol Lett 2021; 336:21-31. [PMID: 33069761 DOI: 10.1016/j.toxlet.2020.10.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 08/31/2020] [Accepted: 10/02/2020] [Indexed: 01/08/2023]
Abstract
Hepatic fibrosis is the wound healing response upon the liver tissue damage caused by multiple stimuli. Targeting activated hepatic stellate cells (HSCs), the major extracellular matrix (ECM)-producing cells within the damaged liver, has been regarded as one of the main treatments for hepatic fibrosis. In the present study, we performed preliminary bioinformatics analysis attempting to identify possible factors related to hepatic fibrosis and found that lncRNA G protein-coupled receptor 137B (Gpr137b-ps) and C-X-C motif chemokine ligand 14 (CXCL14) showed to be markedly upregulated within carbon tetrachloride (CCl4)-caused hepatic fibrotic mice tissue samples and activated HSCs. CXCL14 The silencing of lncRNA Gpr137b-ps or CXCL14 alone could significantly improve CCl4-induced fibrotic changes in mice liver in vivo and collagen I and III release by HSCs and HSC proliferation in vitro. miR-200a-3p directly targeted lncRNA Gpr137b-ps and CXCL14, respectively. LncRNA Gpr137b-ps relieved miR-200a-3p-induced inhibition on CXCL14 expression via acting as a ceRNA. In HSCs, the effects of lncRNA Gpr137b-ps silencing on collagen I and III release by HSCs and HSC proliferation were significantly reversed by miR-200a-3p inhibition, and the effects of miR-200a-3p inhibition were reversed by CXCL14 silencing. In conclusion, we demonstrated a lncRNA Gpr137b-ps/miR-200a-3p/CXCL14 axis that modulates HSC activation and might exert an effect on the pathogenesis of liver fibrosis.
Collapse
Affiliation(s)
- Jinmao Liao
- Department of Hepatopathy, The Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, Hunan, China
| | - Zheng Zhang
- Department of Hepatopathy, The Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, Hunan, China
| | - Qi Yuan
- Department of Hepatopathy, The Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, Hunan, China
| | - Qiong Liu
- Department of Hepatopathy, The Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, Hunan, China
| | - Jia Kuang
- Department of Hepatopathy, The Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, Hunan, China
| | - Yuan Fang
- Department of Hepatopathy, The Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, Hunan, China
| | - Xiaoxuan Hu
- Department of Hepatopathy, The Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, Hunan, China.
| |
Collapse
|
24
|
Odackal J, Yu V, Gomez-Manjerres D, Field JJ, Burdick MD, Mehrad B. Circulating fibrocytes as prognostic biomarkers of autoimmune interstitial lung disease. ERJ Open Res 2020; 6:00481-2020. [PMID: 33263049 PMCID: PMC7682700 DOI: 10.1183/23120541.00481-2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 08/29/2020] [Indexed: 12/14/2022] Open
Abstract
Background Autoimmunity is a common cause of pulmonary fibrosis and can present either as a manifestation of an established connective tissue disease or as the recently described entity of interstitial pneumonia with autoimmune features. The rate of progression and responsiveness to immunosuppression in these illnesses are difficult to predict. Circulating fibrocytes are bone marrow-derived progenitor cells that home to injured tissues and contribute to lung fibrogenesis. We sought to test the hypothesis that the blood fibrocyte concentration predicts outcome and treatment responsiveness in autoimmune interstitial lung diseases. Methods We compared the concentration of circulating fibrocytes in 50 subjects with autoimmune interstitial lung disease and 26 matched healthy controls and assessed the relationship between serial peripheral blood fibrocyte concentrations and clinical outcomes over a median of 6.25 years. Results As compared to controls, subjects with autoimmune interstitial lung disease had higher circulating concentrations of total fibrocytes, the subset of activated fibrocytes, and fibrocytes with activation of PI3K/AKT/mTOR, transforming growth factor-β (TGF-β) receptor and interleukin (IL)-4/IL-13 receptor signalling pathways. Over the follow-up period, there were episodes of marked elevation in the concentration of circulating fibrocytes in subjects with autoimmune interstitial lung disease but not controls. Initiation of immunosuppressive therapy was associated with a decline in the concentration of circulating fibrocytes. For each 100 000 cells·mL−1 increase in peak concentration of circulating fibrocytes, we found a 5% increase in odds of death or lung function decline. Conclusion In patients with autoimmune interstitial lung disease, circulating fibrocytes may represent a biomarker of outcome and treatment response. Autoimmune diseases are common causes of pulmonary fibrosis. The blood concentration of fibrocytes, cells involved in formation of scar tissue, predicts outcomes and response to immunosuppression in these patients.https://bit.ly/35bel62
Collapse
Affiliation(s)
- John Odackal
- Dept of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Victor Yu
- Dept of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Diana Gomez-Manjerres
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Joshua J Field
- Medical Sciences Institute and Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI, USA
| | - Marie D Burdick
- Dept of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA.,Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Borna Mehrad
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
25
|
Diao X. CXCR4 polymeric nanocomplex-mediated pulmonary delivery of siPD-L1: A novel procedure to enhance immunotherapy. Thorac Cancer 2020; 11:2753-2754. [PMID: 32893969 DOI: 10.1111/1759-7714.13627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 11/29/2022] Open
Affiliation(s)
- Xiayao Diao
- Department of Urology, Sun Yat-sen Memorial Hospital, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant, Tumor Epigenetics and Gene Regulation, Sun Yat-sen, Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
26
|
CXCR4 + cells are increased in lung tissue of patients with idiopathic pulmonary fibrosis. Respir Res 2020; 21:221. [PMID: 32843095 PMCID: PMC7449054 DOI: 10.1186/s12931-020-01467-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/23/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND CXCR4, a transmembrane-receptor located on epithelial cells that is activated by CXCL12, may have a role in IPF via migration of CXCR4+ fibrocytes to the lung. However, its expression has not been fully characterised in idiopathic pulmonary fibrosis (IPF) or other fibrotic interstitial lung diseases (ILDs). CXCL12 is constitutively expressed in the bone marrow, and levels of CXCR4 regulate control of this signalling pathway. The aim of this study was to profile the expression of CXCR4 in lung tissue and peripheral circulation of patients with IPF and other fibrotic ILDs. METHODS Expression of CXCR4 on peripheral blood mononuclear cells (PBMCs) was examined by flow cytometry in 20 patients with IPF and 10 age-matched non-disease control (NDC) donors. Levels of CXCL12 in human plasma were measured by ELISA. Expression of CXCR4, CXCL12, CD45, and e-cadherin was assessed in IPF (n = 10), other fibrotic ILD (n = 8) and NDC (n = 10) lung tissue by multiplex immunohistochemistry (OPAL) and slides were scanned using a Vectra 3 scanner. Cells were quantified with computer automated histological analysis software (HALO). RESULTS In blood, the number of CXCR4+ cells was lower but the level of CXCL12 was higher in patients with IPF compared to NDC donors. Elevated CXCR4 expression was detected in lung tissue from patients with IPF and other fibrotic ILDs compared to NDC. There were higher levels of CXCR4+/e-cadherin+/CXCL12+ (epithelial) cells in IPF lung tissue compared to NDC, but there was no difference in the numbers of CXCR4+/CD45+/CXCL12+ (myeloid) cells between the two groups. CONCLUSIONS This report demonstrates that CXCR4 is overexpressed not only in IPF but also in other ILDs and expression is particularly prominent within both honeycomb cysts and distal airway epithelium. This observation supports the hypothesis that CXCR4 may drive tissue fibrosis through binding its specific ligand CXCL12. Although CXCR4 expressing cells could be either of epithelial or myeloid origin it appears that the former is more prominent in IPF lung tissue. Further characterization of the cells of the honeycomb cyst may lead to a better understanding of the fibrogenic processes in IPF and other end-stage fibrotic ILDs.
Collapse
|
27
|
Lin CM, Alrbiaan A, Odackal J, Zhang Z, Scindia Y, Sung SSJ, Burdick MD, Mehrad B. Circulating fibrocytes traffic to the lung in murine acute lung injury and predict outcomes in human acute respiratory distress syndrome: a pilot study. Mol Med 2020; 26:52. [PMID: 32460694 PMCID: PMC7251319 DOI: 10.1186/s10020-020-00176-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 04/27/2020] [Indexed: 02/06/2023] Open
Abstract
Background Fibrosis is an integral component of the pathogenesis of acute lung injury and is associated with poor outcomes in patients with acute respiratory distress syndrome (ARDS). Fibrocytes are bone marrow-derived cells that traffic to injured tissues and contribute to fibrosis; hence their concentration in the peripheral blood has the potential to serve as a biomarker of lung fibrogenesis. We therefore sought to test the hypothesis that the concentration and phenotype of circulating fibrocytes in patients with ARDS predicts clinical outcomes. Methods For the animal studies, C57Bl/6 mice were infected with experimental Klebsiella pneumoniae in a model of acute lung injury; one-way ANOVA was used to compare multiple groups and two-way ANOVA was used to compare two groups over time. For the human study, 42 subjects with ARDS and 12 subjects with pneumonia (without ARDS) were compared to healthy controls. Chi-squared or Fisher’s exact test were used to compare binary outcomes. Survival data was expressed using a Kaplan-Meier curve and compared by log-rank test. Univariable and multivariable logistic regression were used to predict death. Results In mice with acute lung injury caused by Klebsiella pneumonia, there was a time-dependent increase in lung soluble collagen that correlated with sequential expansion of fibrocytes in the bone marrow, blood, and then lung compartments. Correspondingly, when compared via cross-sectional analysis, the initial concentration of blood fibrocytes was elevated in human subjects with ARDS or pneumonia as compared to healthy controls. In addition, fibrocytes from subjects with ARDS displayed an activated phenotype and on serial measurements, exhibited intermittent episodes of markedly elevated concentration over a median of 1 week. A peak concentration of circulating fibrocytes above a threshold of > 4.8 × 106 cells/mL cells correlated with mortality that was independent of age, ratio of arterial oxygen concentration to the fraction of inspired oxygen, and vasopressor requirement. Conclusions Circulating fibrocytes increase in a murine model of acute lung injury and elevation in the number of these cells above a certain threshold is correlated with mortality in human ARDS. Therefore, these cells may provide a useful and easily measured biomarker to predict outcomes in these patients.
Collapse
Affiliation(s)
- Christine M Lin
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, 1600 SW Archer Road, Box 100225, Gainesville, FL, 32610-0225, USA
| | - Abdullah Alrbiaan
- Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - John Odackal
- Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Zhimin Zhang
- Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Yogesh Scindia
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, 1600 SW Archer Road, Box 100225, Gainesville, FL, 32610-0225, USA
| | - Sun-Sang J Sung
- Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Marie D Burdick
- Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Borna Mehrad
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, 1600 SW Archer Road, Box 100225, Gainesville, FL, 32610-0225, USA.
| |
Collapse
|
28
|
Li Z, Wang Y, Shen Y, Qian C, Oupicky D, Sun M. Targeting pulmonary tumor microenvironment with CXCR4-inhibiting nanocomplex to enhance anti-PD-L1 immunotherapy. SCIENCE ADVANCES 2020; 6:eaaz9240. [PMID: 32440550 PMCID: PMC7228744 DOI: 10.1126/sciadv.aaz9240] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 03/02/2020] [Indexed: 05/19/2023]
Abstract
Anti-programmed cell death 1 ligand 1 (PD-L1) therapy is extraordinarily effective in select patients with cancer. However, insufficient lymphocytic infiltration, weak T cell-induced inflammation, and immunosuppressive cell accumulation in the tumor microenvironment (TME) may greatly diminish the efficacy. Here, we report development of the FX@HP nanocomplex composed of fluorinated polymerized CXCR4 antagonism (FX) and paclitaxel-loaded human serum albumin (HP) for pulmonary delivery of anti-PD-L1 small interfering RNA (siPD-L1) to treat orthotopic lung tumors. FX@HP induced T cell infiltration, increased expression of calreticulin on tumor cells, and reduced the myeloid-derived suppressor cells/regulatory T cells in the TME, thereby acting synergistically with siPD-L1 for effective immunotherapy. Our work suggests that the CXCR4-inhibiting nanocomplex decreases tumor fibrosis, facilitates T cell infiltration and relieves immunosuppression to modulate the immune process to improve the objective response rate of anti-PD-L1 immunotherapy.
Collapse
Affiliation(s)
- Zhaoting Li
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yixin Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yuexin Shen
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Chenggen Qian
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - David Oupicky
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Minjie Sun
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
29
|
Wang XA, Griffiths K, Foley M. Emerging Role of CXCR4 in Fibrosis. ANTI-FIBROTIC DRUG DISCOVERY 2020:211-234. [DOI: 10.1039/9781788015783-00211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Recent evidence has shown that the chemokine receptor CXCR4 and its natural chemokine ligand CXCL12 promote pro-inflammatory responses in a variety of situations and this axis has emerged as a central player in tissue fibrosis. Although its role as a co-receptor for human immunodeficiency virus (HIV) and a key player in various cancers has been well established, the role of CXCR4 in various types of fibrosis has emerged only recently. This review will explore the involvement of CXCR4 in the development of fibrosis, focusing mainly on lung, kidney and eye fibrosis.
Collapse
Affiliation(s)
- Xilun Anthony Wang
- The Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University Bundoora Melbourne 3086 Australia
| | - Katherine Griffiths
- The Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University Bundoora Melbourne 3086 Australia
| | - Michael Foley
- The Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University Bundoora Melbourne 3086 Australia
- AdAlta Limited 15/2 Park Drive Bundoora 3083 Australia
| |
Collapse
|
30
|
Li F, Xu X, Geng J, Wan X, Dai H. The autocrine CXCR4/CXCL12 axis contributes to lung fibrosis through modulation of lung fibroblast activity. Exp Ther Med 2020; 19:1844-1854. [PMID: 32104240 PMCID: PMC7027131 DOI: 10.3892/etm.2020.8433] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 10/30/2019] [Indexed: 02/06/2023] Open
Abstract
The C-X-C Motif Chemokine Receptor 4/C-X-C Motif Chemokine Ligand 12 (CXCR4/CXCL12) axis has been implicated in the pathogenesis of pulmonary fibrosis. However, the mechanisms governing this remain to be determined. The current study demonstrated that human lung fibroblasts (HLFs) exhibit high CXCL12 expression and also exhibit high expression of its corresponding receptor CXCR4. Exogenous CXCL12 was revealed to significantly promote the migration and proliferation of HLFs, and potentiate CXCR4 expression. These effects were demonstrated to be inhibited by AMD3100, which is an antagonist of CXCR4. Lung and bronchoalveolar lavage fluid CXCR4 and CXCL12 expression was upregulated by in vivo bleomycin administration, which was partially inhibited by pre-treatment with AMD3100. AMD3100 also reduced lung collagen content in the bleomycin model. Inhibiting CXCR4 was indicated to ameliorate the lung compliance and resistance of pulmonary fibrosis. In conclusion, the results of the present study suggested that autocrine CXCR4/CXCL12 axis is an important mechanism underlying the pathogenesis of idiopathic pulmonary fibrosis, and may serve as a potential therapeutic target that can be used in the treatment of pulmonary disease.
Collapse
Affiliation(s)
- Fei Li
- Department of Pulmonary and Critical Care Medicine, Beijing An-Zhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Xuefeng Xu
- Department of Pulmonary and Critical Care Medicine, Beijing An-Zhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Jing Geng
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Capital Medical University, National Clinical Research Center for Respiratory Diseases, Beijing 100029, P.R. China
| | - Xuan Wan
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Huaping Dai
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Capital Medical University, National Clinical Research Center for Respiratory Diseases, Beijing 100029, P.R. China
| |
Collapse
|
31
|
Nicastro M, Vescovini R, Maritati F, Palmisano A, Urban ML, Incerti M, Fenaroli P, Peyronel F, Benigno GD, Mangieri D, Volpi R, Becchi G, Romagnani P, Corradi D, Vaglio A. Fibrocytes in Chronic Periaortitis: A Novel Mechanism Linking Inflammation and Fibrosis. Arthritis Rheumatol 2019; 71:1913-1922. [DOI: 10.1002/art.41024] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 06/18/2019] [Indexed: 12/23/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Paola Romagnani
- University of Florence and Meyer Children's Hospital Florence Italy
| | | | - Augusto Vaglio
- University of Florence and Meyer Children's Hospital Florence Italy
| |
Collapse
|
32
|
Majka SM, Rojas M, Petrache I, Foronjy RF. Mesenchymal Regulation of the Microvascular Niche in Chronic Lung Diseases. Compr Physiol 2019; 9:1431-1441. [PMID: 31688970 DOI: 10.1002/cphy.c180043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The adult lung is comprised of diverse vascular, epithelial, and mesenchymal progenitor cell populations that reside in distinct niches. Mesenchymal progenitor cells (MPCs) are intimately associated with both the epithelium and the vasculature, and new evidence is emerging to describe their functional roles in these niches. Also emerging, following lineage analysis and single cell sequencing, is a new understanding of the diversity of mesenchymal cell subpopulations in the lung. However, several gaps in knowledge remain, including how newly defined MPC lineages interact with cells in the vascular niche and the role of adult lung MPCs during lung repair and regeneration following injury, especially in chronic lung diseases. Here we summarize how the current evidence on MPC regulation of the microvasculature during tissue homeostasis and injury may inform studies on understanding their role in chronic lung disease pathogenesis or repair. © 2019 American Physiological Society. Compr Physiol 9:1431-1441, 2019.
Collapse
Affiliation(s)
- Susan M Majka
- Department of Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, National Jewish Health, Denver, Colorado, USA
| | - Mauricio Rojas
- McGowan Institute for Regenerative Medicine, Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Irina Petrache
- Department of Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, National Jewish Health, Denver, Colorado, USA
| | - Robert F Foronjy
- Division of Pulmonary and Critical Care Medicine, SUNY Downstate Medical Center, Brooklyn, New York, USA
| |
Collapse
|
33
|
Cao J, Zhu W, Yu D, Pan L, Zhong L, Xiao Y, Gao Y, Jiao Y, Zhang Q, Ji J, Yang H, Zhang S, Cao J. The Involvement of SDF-1α/CXCR4 Axis in Radiation-Induced Acute Injury and Fibrosis of Skin. Radiat Res 2019; 192:410-421. [PMID: 31390312 DOI: 10.1667/rr15384.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Radiation-induced acute skin injury and consequent fibrosis are common complications of cancer radiotherapy and radiation accidents. Stromal cell-derived factor-1α (SDF-1α) and its receptor, CXC chemokine receptor 4 (CXCR4) have been shown to be involved in multiple cellular events. However, the role of SDF-1α/CXCR4 axis in radiation-induced acute injury and fibrosis of skin has not been reported. In this study, we found that the expression of SDF-1α and CXCR4 was significantly increased in irradiated skin tissues of humans, monkeys and rats, compared to their nonirradiated counterparts. Mice with keratinocyte-specific ablation of CXCR4 showed less severe skin damage than wild-type mice after receiving a 35 Gy dose of radiation. Consistently, subcutaneous injection of AMD3100, an FDA approved SDF-1α/CXCR4 inhibitor, attenuated skin injury and fibrosis induced by exposure to radiation in a rat model. Mechanically, the SDF-1α/CXCR4 axis promotes pro-fibrotic TGF-b/Smad signaling through the PI3K-MAPK signaling cascade in human keratinocyte HaCaT cells and skin fibroblast WS1 cells. AMD3100 inhibited Smad2 nuclear translocation and transcriptional activity of Smad2/3 induced by radiation, which suppressed the pro-fibrotic TGF-b/Smad signaling pathway activated by exposure. Taken together, these findings demonstrate the involvement of SDF-1α/CXCR4 axis in radiation-induced acute injury and fibrosis of skin, and indicate that AMD3100 would be an effective countermeasure against these diseases.
Collapse
Affiliation(s)
- Jinming Cao
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China.,State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Wei Zhu
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China.,State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Daojiang Yu
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.,Departments of Plastic Surgery
| | - Lu Pan
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China.,State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Li Zhong
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China.,State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Yuji Xiao
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China.,State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Yiying Gao
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China.,State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Yang Jiao
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China.,State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Qi Zhang
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China.,State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Jiang Ji
- Departments of Dermatology, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Hongying Yang
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China.,State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Shuyu Zhang
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.,Department of Oncology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213100, China.,Department of Second Affiliated Hospital of Chengdu Medical College (China National Nuclear Corporation 416 Hospital), Chengdu 610051, China
| | - Jianping Cao
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China.,State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| |
Collapse
|
34
|
Li L, Li Q, Wei L, Wang Z, Ma W, Liu F, Shen Y, Zhang S, Zhang X, Li H, Qian Y. Chemokine (C‐X‐C motif) ligand 14 contributes to lipopolysaccharide‐induced fibrogenesis in mouse L929 fibroblasts via modulating PPM1A. J Cell Biochem 2019; 120:13372-13381. [PMID: 30920024 DOI: 10.1002/jcb.28612] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 01/13/2019] [Accepted: 01/25/2019] [Indexed: 01/02/2023]
Affiliation(s)
- Li Li
- Department of Respiratory Disease Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine Shanghai China
| | - Qiuhong Li
- Department of Respiratory Medicine Shanghai Pulmonary Hospital, School of Medicine, Tongji University Shanghai China
| | - Lei Wei
- Department of Respiratory Disease Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine Shanghai China
| | - Zhongfu Wang
- Department of Respiratory Disease Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine Shanghai China
| | - Wei Ma
- Department of Respiratory Disease Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine Shanghai China
| | - Fangying Liu
- Department of Respiratory Disease Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine Shanghai China
| | - Yanhua Shen
- Department of Respiratory Disease Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine Shanghai China
| | - Shanfang Zhang
- Department of Respiratory Disease Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine Shanghai China
| | - Xiulian Zhang
- Department of Respiratory Disease Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine Shanghai China
| | - Huiping Li
- Department of Respiratory Medicine Shanghai Pulmonary Hospital, School of Medicine, Tongji University Shanghai China
| | - Yechang Qian
- Department of Respiratory Disease Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine Shanghai China
| |
Collapse
|
35
|
HIV-1 Protein gp120 Induces Mouse Lung Fibroblast-to-Myofibroblast Transdifferentiation via CXCR4 Activation. Am J Med Sci 2019; 357:483-491. [PMID: 31000424 DOI: 10.1016/j.amjms.2019.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 01/09/2019] [Accepted: 03/14/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND Individuals with HIV have ∼2-fold increased risk of developing pulmonary fibrosis. The mechanism(s) by which this occurs has yet to be determined. HIV-1 protein gp120 activates CXCR4 in the lymphocyte, promoting a variety of intracellular signaling pathways including those common to TGFβ1 associated with lung fibroblast-to-myofibroblast transdifferentiation. We hypothesized that gp120 promotes pulmonary fibrotic changes via activation of CXCR4 in the lung fibroblast. METHODS Mouse primary lung fibroblasts (PLFs) were cultured ± gp120, then analyzed for α-SMA expression and stress fiber formation. In parallel, PLFs were cultured ± gp120 ± AMD3100 (a CXCR4 antagonist), and α-SMA, pan and phospho-Akt, and total and phospho-MAPK (or ERK1/2) protein expression was quantified. Finally, lungs and PLFs from wild-type and HIV-1 transgenic mice were analyzed for hydroxyproline and α-SMA content. RESULTS gp120 treatment increased α-SMA expression and myofibroblast differentiation in PLFs. gp120 treatment activated phosphorylation of ERK1/2, but not PI3K-Akt. Pretreatment with AMD3100 inhibited gp120-induced ERK1/2 phosphorylation and gp120-induced α-SMA expression. In parallel, there was a significant increase in hydroxyproline content in lungs from older HIV-1 transgenic mice and a >3-fold increase in α-SMA expression in PLFs isolated from HIV-1 transgenic mice. CONCLUSIONS gp120 induces α-SMA expression and fibroblast-to-myofibroblast transdifferentiation by activating the CXCR4-ERK1/2 signaling pathway in mouse PLFs. Lungs of older HIV-1 transgenic mice contain higher hydroxyproline content and their PLFs have a striking increase in α-SMA expression. These results suggest a mechanism by which individuals with HIV are at increased risk of developing pulmonary fibrotic changes as they age.
Collapse
|
36
|
Tang M, Yang Y, Yu J, Qiu J, Chen P, Wu Y, Wang Q, Xu Z, Ge J, Yu K, Zhuang J. Tetramethylpyrazine in a Murine Alkali-Burn Model Blocks NFκB/NRF-1/CXCR4-Signaling-Induced Corneal Neovascularization. Invest Ophthalmol Vis Sci 2019; 59:2133-2141. [PMID: 29801148 DOI: 10.1167/iovs.17-23712] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Tetramethylpyrazine (TMP) is the active ingredient extracted from the Chinese herb Chuanxiong. The purpose of our study was to identify the mechanism of therapeutic TMP suppression of pathologic chemokine receptor 4 (CXCR4) transcription. Methods C57BL/6J mice with alkali-burned corneas were treated with either TMP eye drops (1.5 mg/mL) or PBS. Corneal neovascularization (CNV) was measured and a clinical assessment was made by slit lamp microscopy. Expression of CXCR4 and the transcription factors nuclear respiratory factor-1 (NRF-1), nuclear factor kappa B (NFκB), forkhead box C1, and yin yang 1 were tracked by real-time RT-PCR and immunofluorescence staining of murine corneas. Western blot, real-time PCR, and immunofluorescence evaluated expression of related genes in human umbilical vein endothelial cells (HUVECs) after 200-μmol/L TMP treatment. In addition, plasmid transfection and chromatin immunoprecipitation assays elucidated the relationship among NRF-1, NFκB, and CXCR4. Results Corneas treated with TMP had smaller areas of neovascularization and scored better in clinical assessments. Injured corneas showed significantly elevated expressions of NRF-1, NFκB, and CXCR4 that were normalized in vivo by TMP treatment. Similarly, in HUVECs in vitro, TMP decreased expression of NRF-1, NFκB, and CXCR4. Overexpression of NFκB or NRF-1 raised the expression of CXCR4 in HUVECs, but not synergistically. Chromatin immunoprecipitation assays detected only NRF-1 bound to the CXCR4 promoter region, suggesting NFκB controls CXCR4 expression by upregulating NRF-1. Together, our data suggest TMP downregulates CXCR4 by repressing NRF-1 expression in CNV, likely indirectly by downregulating NFκB. Conclusions Our results implicate a novel mechanism wherein TMP inhibits neovascularization via an NFκB/NRF-1/CXCR4 circuit.
Collapse
Affiliation(s)
- Mingjun Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ying Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jingzhi Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jin Qiu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Pei Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yihui Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Qiyun Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Zhuojun Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jian Ge
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Keming Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jing Zhuang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
37
|
Chen G, Bracamonte-Baran W, Diny NL, Hou X, Talor MV, Fu K, Liu Y, Davogustto G, Vasquez H, Taegtmeyer H, Frazier OH, Waisman A, Conway SJ, Wan F, Čiháková D. Sca-1 + cardiac fibroblasts promote development of heart failure. Eur J Immunol 2018; 48:1522-1538. [PMID: 29953616 DOI: 10.1002/eji.201847583] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 05/09/2018] [Accepted: 06/25/2018] [Indexed: 12/19/2022]
Abstract
The causative effect of GM-CSF produced by cardiac fibroblasts to development of heart failure has not been shown. We identified the pathological GM-CSF-producing cardiac fibroblast subset and the specific deletion of IL-17A signaling to these cells attenuated cardiac inflammation and heart failure. We describe here the CD45- CD31- CD29+ mEF-SK4+ PDGFRα+ Sca-1+ periostin+ (Sca-1+ ) cardiac fibroblast subset as the main GM-CSF producer in both experimental autoimmune myocarditis and myocardial infarction mouse models. Specific ablation of IL-17A signaling to Sca-1+ periostin+ cardiac fibroblasts (PostnCre Il17rafl/fl ) protected mice from post-infarct heart failure and death. Moreover, PostnCre Il17rafl/fl mice had significantly fewer GM-CSF-producing Sca-1+ cardiac fibroblasts and inflammatory Ly6Chi monocytes in the heart. Sca-1+ cardiac fibroblasts were not only potent GM-CSF producers, but also exhibited plasticity and switched their cytokine production profiles depending on local microenvironments. Moreover, we also found GM-CSF-positive cardiac fibroblasts in cardiac biopsy samples from heart failure patients of myocarditis or ischemic origin. Thus, this is the first identification of a pathological GM-CSF-producing cardiac fibroblast subset in human and mice hearts with myocarditis and ischemic cardiomyopathy. Sca-1+ cardiac fibroblasts direct the type of immune cells infiltrating the heart during cardiac inflammation and drive the development of heart failure.
Collapse
Affiliation(s)
- Guobao Chen
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | | | - Nicola L Diny
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Xuezhou Hou
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Monica V Talor
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Kai Fu
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Yue Liu
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Giovanni Davogustto
- Department of Internal Medicine, Division of Cardiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Hernan Vasquez
- Department of Internal Medicine, Division of Cardiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Heinrich Taegtmeyer
- Department of Internal Medicine, Division of Cardiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - O Howard Frazier
- Texas Heart Institute, CHI St. Luke's Health - Baylor St. Luke's Medical Center, MC 2-114A, PO Box 20345, Houston, TX, USA
| | - Ari Waisman
- Institute for Molecular Medicine, University of Mainz, Mainz, Germany
| | - Simon J Conway
- Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Fengyi Wan
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniela Čiháková
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.,W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
38
|
Tai W, Zhou Z, Zheng B, Li J, Ding J, Wu H, Gao L, Dong Z. Inhibitory effect of circulating fibrocytes on injury repair in acute lung injury/acute respiratory distress syndrome mice model. J Cell Biochem 2018; 119:7982-7990. [PMID: 29323734 DOI: 10.1002/jcb.26664] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 01/09/2018] [Indexed: 11/06/2022]
Abstract
The study was aimed to explore the functions of circulating fibrocytes (CFs) on injury repair in acute lung injury/acute respiratory distress syndrome (ALI/ARDS) mice model and its clinical value as a biomarker for ALI/ARDS. ALI/ARDS mice model was established by intratracheal instillation of lipopolysaccharide (LPS). Mononuclear cells were isolated from peripheral blood of ALI/ARDS model and flow cytometry was used to measure CFs defined as cells positive for CD45 and collagen-1. Histological changes of lung tissues were evaluated by H&E staining and Masson's trichrome staining. The correlations of CFs counts with damnification of lung tissue and the severity of pulmonary fibrosis were evaluated by Pearson correlation analyses. Western blot was used to detect the protein expression of collagen-1. ELISA was applied to determine cytokine CXCL12 concentration. Clinical relevance between CFs and ALI/ARDS was investigated. The greater number of CFs in the ALI/ARDS group implied higher degree of lung injury and more severe pulmonary fibrosis. The protein expression of collagen-1 and concentration of cytokine CXCL12 in ALI/ARDS group were higher than that in control group. Clinical and prognostic analysis revealed the higher injury degree and death rates in ALI/ARDS group than those in control group, and identified a greater severity and mortality for patients with ARDS than those with ALI. ROC curve analysis indicated the counts of CFs greater than 5.85% can predict death rates with AUC = 0.928. CFs had an inhibitory effect on injury repair in ALI/ARDS mice model. This might be unfavorable as a clinical marker for progression of ALI/ARDS.
Collapse
Affiliation(s)
- Wenlin Tai
- Department of Clinical Laboratory, Yunnan Molecular Diagnostic Center, The 2nd Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zeping Zhou
- Department of Hematology, The 2nd Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Boyang Zheng
- Department of Clinical Laboratory, Yunnan Molecular Diagnostic Center, The 2nd Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jinyu Li
- Department of Clinical Laboratory, Yunnan Molecular Diagnostic Center, The 2nd Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jiawei Ding
- Department of Clinical Laboratory, Yunnan Molecular Diagnostic Center, The 2nd Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Hanxin Wu
- Department of Clinical Laboratory, Yunnan Molecular Diagnostic Center, The 2nd Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Ling Gao
- Department of Clinical Laboratory, Yunnan Molecular Diagnostic Center, The 2nd Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zhaoxing Dong
- Department of Respiratory, The 2nd Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
39
|
Habiel DM, Espindola MS, Coelho AL, Hogaboam CM. Modeling Idiopathic Pulmonary Fibrosis in Humanized Severe Combined Immunodeficient Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:891-903. [PMID: 29378172 PMCID: PMC5954978 DOI: 10.1016/j.ajpath.2017.12.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 12/11/2017] [Accepted: 12/28/2017] [Indexed: 12/17/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fibrotic lung disease of unknown etiopathogenesis with limited therapeutic options. IPF is characterized by an abundance of fibroblasts and loss of epithelial progenitors, which cumulates in unrelenting fibrotic lung remodeling and loss of normal oxygenation. IPF has been challenging to model in rodents; nonetheless, mouse models of lung fibrosis provide clues as to the natural progression of lung injury and remodeling, but many have not been useful in predicting efficacy of therapeutics in clinical IPF. We provide a detailed methodologic description of various iterations of humanized mouse models, initiated by the i.v. injection of cells from IPF lung biopsy or explants specimens into severe combined immunodeficiency (SCID)/beige or nonobese diabetic SCID γ mice. Unlike cells from normal lung samples, IPF cells promote persistent, nonresolving lung remodeling in SCID mice. Finally, we provide examples and discuss potential advantages and pitfalls of human-specific targeting approaches in a humanized SCID model of pulmonary fibrosis.
Collapse
Affiliation(s)
- David M Habiel
- Women's Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California.
| | - Milena S Espindola
- Women's Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Ana L Coelho
- Women's Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Cory M Hogaboam
- Women's Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California.
| |
Collapse
|
40
|
Griffiths K, Habiel DM, Jaffar J, Binder U, Darby WG, Hosking CG, Skerra A, Westall GP, Hogaboam CM, Foley M. Anti-fibrotic Effects of CXCR4-Targeting i-body AD-114 in Preclinical Models of Pulmonary Fibrosis. Sci Rep 2018; 8:3212. [PMID: 29453386 PMCID: PMC5816662 DOI: 10.1038/s41598-018-20811-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 01/24/2018] [Indexed: 01/19/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic fibrotic lung disease that is prevalent in individuals >50 years of age, with a median survival of 3–5 years and limited therapeutic options. The disease is characterized by collagen deposition and remodeling of the lung parenchyma in a process that is thought to be driven by collagen-expressing immune and structural cells. The G-protein coupled C-X-C chemokine receptor 4, CXCR4, is a candidate therapeutic target for IPF owing to its role in the recruitment of CXCR4+ fibrocytes from the bone marrow to fibrotic lung tissue and its increased expression levels by structural cells in fibrotic lung tissue. We have engineered a novel fully human single domain antibody “i-body” called AD-114 that binds with high affinity to human CXCR4. We demonstrate here that AD-114 inhibits invasive wound healing and collagen 1 secretion by human IPF fibroblasts but not non-diseased control lung fibroblasts. Furthermore, in a murine bleomycin model of pulmonary fibrosis, AD-114 reduced the accumulation of fibrocytes (CXCR4+/Col1+/CD45+) in fibrotic murine lungs and ameliorated the degree of lung injury. Collectively, these studies demonstrate that AD-114 holds promise as a new biological therapeutic for the treatment of IPF.
Collapse
Affiliation(s)
- K Griffiths
- AdAlta Limited, La Trobe University, 15/2 Park Drive, Bundoora, 3083, Australia.,The Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Melbourne, 3086, Australia
| | - D M Habiel
- Cedars-Sinai, Medical Centre, Los Angeles, CA, 90048, USA
| | - J Jaffar
- Department of Respiratory Medicine, Alfred Hospital and Monash University, Melbourne, Victoria, 3000, Australia
| | - U Binder
- XL-protein GmbH, Lise-Meitner-Str. 30, 85354, Freising, Germany
| | - W G Darby
- AdAlta Limited, La Trobe University, 15/2 Park Drive, Bundoora, 3083, Australia.,The Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Melbourne, 3086, Australia
| | - C G Hosking
- AdAlta Limited, La Trobe University, 15/2 Park Drive, Bundoora, 3083, Australia.,The Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Melbourne, 3086, Australia
| | - A Skerra
- XL-protein GmbH, Lise-Meitner-Str. 30, 85354, Freising, Germany
| | - G P Westall
- Department of Respiratory Medicine, Alfred Hospital and Monash University, Melbourne, Victoria, 3000, Australia
| | - C M Hogaboam
- Cedars-Sinai, Medical Centre, Los Angeles, CA, 90048, USA
| | - M Foley
- AdAlta Limited, La Trobe University, 15/2 Park Drive, Bundoora, 3083, Australia. .,The Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Melbourne, 3086, Australia.
| |
Collapse
|
41
|
Yao Y, Zheng Z, Song Q. Mesenchymal stem cells: A double-edged sword in radiation-induced lung injury. Thorac Cancer 2017; 9:208-217. [PMID: 29235254 PMCID: PMC5792737 DOI: 10.1111/1759-7714.12573] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/10/2017] [Accepted: 11/10/2017] [Indexed: 01/06/2023] Open
Abstract
Radiation therapy is an important treatment modality for multiple thoracic malignancies. However, radiation‐induced lung injury (RILI), which is the term generally used to describe damage to the lungs caused by exposure to ionizing radiation, remains a critical issue affecting both tumor control and patient quality of life. Despite tremendous effort, there is no current consensus regarding the optimal treatment approach for RILI. Because of a number of functional advantages, including self‐proliferation, multi‐differentiation, injury foci chemotaxis, anti‐inflammation, and immunomodulation, mesenchymal stem cells (MSCs) have been a focus of research for many years. Accumulating evidence indicates the therapeutic potential of transplantation of MSCs derived from adipose tissue, umbilical cord blood, and bone marrow for inflammatory diseases, including RILI. However, reports have also shown that MSCs, including fibrocytes, lung hematopoietic progenitor cells, and ABCG2+ MSCs, actually enhance the progression of lung injuries. These contradictory results suggest that MSCs may have dual effects and that caution should be taken when using MSCs to treat RILI. In this review, we present and discuss recent evidence of the double‐edged function of MSCs and provide comments on the prospects of these findings.
Collapse
Affiliation(s)
- Yi Yao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhongliang Zheng
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Qibin Song
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
42
|
Ishida Y, Kimura A, Nosaka M, Kuninaka Y, Hemmi H, Sasaki I, Kaisho T, Mukaida N, Kondo T. Essential involvement of the CX3CL1-CX3CR1 axis in bleomycin-induced pulmonary fibrosis via regulation of fibrocyte and M2 macrophage migration. Sci Rep 2017; 7:16833. [PMID: 29203799 PMCID: PMC5714949 DOI: 10.1038/s41598-017-17007-8] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 11/20/2017] [Indexed: 02/06/2023] Open
Abstract
The potential role of macrophages in pulmonary fibrosis (PF) prompted us to evaluate the roles of CX3CR1, a chemokine receptor abundantly expressed in macrophages during bleomycin (BLM)-induced PF. Intratracheal BLM injection induced infiltration of leukocytes such as macrophages into the lungs, which eventually resulted in fibrosis. CX3CR1 expression was mainly detected in the majority of macrophages and in a small portion of α-smooth muscle actin-positive cells in the lungs, while CX3CL1 was expressed in macrophages. BLM-induced fibrotic changes in the lungs were reduced without any changes in the number of leukocytes in Cx3cr1 -/- mice, as compared with those in the wild-type (WT) mice. However, intrapulmonary CX3CR1+ macrophages displayed pro-fibrotic M2 phenotypes; lack of CX3CR1 skewed their phenotypes toward M1 in BLM-challenged lungs. Moreover, fibrocytes expressed CX3CR1, and were increased in BLM-challenged WT lungs. The number of intrapulmonary fibrocytes was decreased in Cx3cr1 -/- mice. Thus, locally-produced CX3CL1 can promote PF development primarily by attracting CX3CR1-expressing M2 macrophages and fibrocytes into the lungs.
Collapse
Affiliation(s)
- Yuko Ishida
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Akihiko Kimura
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Mizuho Nosaka
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yumi Kuninaka
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Hiroaki Hemmi
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Izumi Sasaki
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Tsuneyasu Kaisho
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Naofumi Mukaida
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Toshikazu Kondo
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan.
| |
Collapse
|
43
|
Circulating fibrocytes as biomarkers of impaired lung function in adults with sickle cell disease. Blood Adv 2017; 1:2217-2224. [PMID: 29296869 DOI: 10.1182/bloodadvances.2017010777] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 09/19/2017] [Indexed: 01/01/2023] Open
Abstract
Lung injury and fibrosis are common in patients with sickle cell disease (SCD). Fibrocytes, a population of circulating, bone marrow-derived cells, have been linked to development and progression of tissue fibrogenesis and have been implicated in the development of lung fibrosis in preclinical models of SCD. We tested the hypothesis that the levels and activation state of circulating fibrocytes during steady state are associated with abnormal pulmonary function in adults with SCD. In a prospective cohort of steady-state adults with SCD and healthy age- and race-matched control participants, we measured the concentration and activation state of circulating fibrocytes and assessed pulmonary phenotype with pulmonary function tests (PFTs), a respiratory questionnaire, 6-minute walk test, high-resolution chest computed tomography scan, and echocardiogram. Seventy-one adults with SCD and 26 healthy African American control participants were examined. Compared with control participants, patients with SCD demonstrated higher levels of circulating fibrocytes, a significant proportion of which expressed the activation marker α-smooth muscle actin. Within patients with SCD, elevated absolute concentrations of circulating fibrocytes were strongly and independently associated with impaired lung physiology, as measured by PFTs. We conclude that elevated circulating fibrocytes are associated with lung disease in adults with SCD during steady state, consistent with a role for these cells in pathogenesis of lung fibrosis in this disease. Circulating fibrocytes may represent a novel biomarker for progressive pulmonary fibrosis in patients with SCD.
Collapse
|
44
|
Chiang HY, Chu PH, Lee TH. R1R2 peptide ameliorates pulmonary fibrosis in mice through fibrocyte migration and differentiation. PLoS One 2017; 12:e0185811. [PMID: 28968441 PMCID: PMC5624629 DOI: 10.1371/journal.pone.0185811] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 09/19/2017] [Indexed: 02/06/2023] Open
Abstract
Circulating fibrocytes play a key role in the pathogenesis of pulmonary fibrosis. Fibrocytes are bone marrow-derived leukocytes, which enter the lungs in response to their chemoattractant CXCL12 and differentiate into fibroblasts or myofibroblasts, leading to excess deposition of the collagen-rich extracellular matrix. Matrix metalloproteinase (MMP)-9 and MMP-2, secreted by fibrocytes, degrade the subendothelial basement membrane and promote fibrocyte influx into the lungs. Here, we demonstrate that R1R2, a novel peptide derived from the bacterial adhesin SFS, attenuates pulmonary fibrosis by preventing the differentiation of fibrocytes into myofibroblasts and by reducing the invasion of fibrocytes through basement membrane-like proteins. Moreover, our findings reveal dual regulation of R1R2 on MMP-9 through reduced enzymatic activity on gelatin and increased cleavage of CXCL12. These data suggest that R1R2 has potent anti-fibrotic effects against pulmonary fibrosis.
Collapse
Affiliation(s)
- Hou-Yu Chiang
- Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Pao-Hsien Chu
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ting-Hein Lee
- Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- * E-mail:
| |
Collapse
|
45
|
Just SA, Lindegaard H, Hejbøl EK, Davidsen JR, Bjerring N, Hansen SWK, Schrøder HD, Hansen IMJ, Barington T, Nielsen C. Fibrocyte measurement in peripheral blood correlates with number of cultured mature fibrocytes in vitro and is a potential biomarker for interstitial lung disease in Rheumatoid Arthritis. Respir Res 2017; 18:141. [PMID: 28720095 PMCID: PMC5516315 DOI: 10.1186/s12931-017-0623-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 07/13/2017] [Indexed: 01/08/2023] Open
Abstract
Background Interstitial lung disease (ILD) can be a severe extra-articular disease manifestation in Rheumatoid Arthritis (RA). A potential role of fibrocytes in RA associated ILD (RA-ILD) has not previously been described. We present a modified faster method for measuring circulating fibrocytes, without intracellular staining. The results are compared to the traditional culture method, where the number of monocytes that differentiate into mature fibrocytes in vitro are counted. The results are following compared to disease activity in patients with severe asthma, ILD, RA (without diagnosed ILD) and RA with verified ILD (RA-ILD). Method CD45+ CD34+ CD11b+ (7-AAD− CD3− CD19− CD294−) cells were isolated by cell sorting and stained for pro-collagen type 1. Thirty-nine patients (10 RA, 9 ILD and 10 with severe asthma, 10 with RA-ILD) and 10 healthy controls (HC) were included. Current medication, disease activity, pulmonary function test and radiographic data were collected. Circulating fibrocytes were quantified by flow cytometry. Peripheral blood mononuclear cells were isolated and cultured for 5 days and the numbers of mature fibrocytes were counted. Results 90.2% (mean, SD = 1.5%) of the sorted cells were pro-collagen type 1 positive and thereby fulfilled the criteria for being circulating fibrocytes. The ILD and RA-ILD groups had increased levels of circulating fibrocytes compared to HC (p < 0.05). Levels of circulating fibrocytes correlated overall to number of monocytes that subsequently in vitro differentiated to mature fibrocytes (r = 0.81, p < 0.001). RA patients with pathologically reduced diffusion capacity for carbon monoxide adjusted for hemoglobin (DLCOc) in both the RA and in the combined RA + RA-ILD group, had significantly higher levels of both circulating and number of cultured mature fibrocytes (both p < 0.05). In both groups, the level of circulating fibrocytes and number of mature fibrocytes in culture also correlated to a reduction in DLCOc (r = −0.61 an r = −0.58 both p < 0.05). Conclusions We presented a fast and valid method for measuring circulating fibrocytes using flow cytometry on lysed peripheral blood. Further, we showed for the first time, that the level of circulating fibrocytes correlated with the number of peripheral blood mononuclear cells, that differentiated into mature fibrocytes in vitro. Reduced DLCOc was correlated with high levels of circulating and mature fibrocytes in RA, which have not been reported previously. In such, this study suggests that fibrocytes may exhibit an important role in the pathogenesis of RA-ILD, which requires further clarification in future studies. Trial registration ClinicalTrials.gov:NCT02711657, registered 13/3–2016, retrospectively registered. Electronic supplementary material The online version of this article (doi:10.1186/s12931-017-0623-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Hanne Lindegaard
- Department Rheumatology, Odense University Hospital, Odense, Denmark
| | | | - Jesper Rømhild Davidsen
- South Danish Center for Interstitial Lung Diseases, Odense University Hospital, Odense, Denmark
| | - Niels Bjerring
- Department Respiratory Medicine, Odense University Hospital, Odense, Denmark
| | | | | | | | - Torben Barington
- Department Clinical Immunology, Odense University Hospital, Odense, Denmark.,Odense Patient data Explorative Network (OPEN), Odense University Hospital, Odense, Denmark
| | - Christian Nielsen
- Department Clinical Immunology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
46
|
Li X, Yue S, Luo Z. Mesenchymal stem cells in idiopathic pulmonary fibrosis. Oncotarget 2017; 8:102600-102616. [PMID: 29254275 PMCID: PMC5731985 DOI: 10.18632/oncotarget.18126] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 05/07/2017] [Indexed: 12/21/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a major cause of respiratory failure in critically ill patients and common outcome of various lung interstitial diseases. Its mortality remains high, and no effective pharmacotherapy, in addition to artificial ventilation and transplantation, exists. As such, the administration of mesenchymal stem or stromal cells (MSCs) is currently investigated as a new therapeutic method for pulmonary fibrosis. Clinical trials on MSC-based therapy as a potential treatment for lung injury and fibrosis are also performed. MSCs can migrate to injured sites and secrete multiple paracrine factors and then regulate endothelial and epithelial permeability, decrease inflammation, enhance tissue repair, and inhibit bacterial growth. In this review, recent studies on stem cells, particularly MSCs, involved in alleviating lung inflammation and fibrosis and their potential MSC-induced mechanisms, including migration and differentiation, soluble factor and extracellular vesicle secretion, and endogenous regulatory functions, were summarized.
Collapse
Affiliation(s)
- Xiaohong Li
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Shaojie Yue
- Department of Neonatology, Xiangya Hospital, Central South University, Changsha, China
| | - Ziqiang Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
47
|
Burgess JK, Mauad T, Tjin G, Karlsson JC, Westergren-Thorsson G. The extracellular matrix - the under-recognized element in lung disease? J Pathol 2016; 240:397-409. [PMID: 27623753 PMCID: PMC5129494 DOI: 10.1002/path.4808] [Citation(s) in RCA: 192] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 08/17/2016] [Accepted: 09/05/2016] [Indexed: 12/11/2022]
Abstract
The lung is composed of airways and lung parenchyma, and the extracellular matrix (ECM) contains the main building blocks of both components. The ECM provides physical support and stability to the lung, and as such it has in the past been regarded as an inert structure. More recent research has provided novel insights revealing that the ECM is also a bioactive environment that orchestrates the cellular responses in its environs. Changes in the ECM in the airway or parenchymal tissues are now recognized in the pathological profiles of many respiratory diseases, including asthma, chronic obstructive pulmonary disease (COPD), and idiopathic pulmonary fibrosis (IPF). Only recently have we begun to investigate whether these ECM changes result from the disease process, or whether they constitute a driving factor that orchestrates the pathological outcomes. This review summarizes our current knowledge of the alterations in the ECM in asthma, COPD, and IPF, and the contributions of these alterations to the pathologies. Emerging data suggest that alterations in the composition, folding or rigidity of ECM proteins may alter the functional responses of cells within their environs, and in so doing change the pathological outcomes. These characteristics highlight potential avenues for targeting lung pathologies in the future. This may ultimately contribute to a better understanding of chronic lung diseases, and novel approaches for finding therapeutic solutions. © 2016 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Janette K Burgess
- University of Groningen, University Medical Centre Groningen, GRIAC Research Institute, Department of Pathology and Medical Biology, Groningen, The Netherlands.,Respiratory Cellular and Molecular Biology Group, Woolcock Institute of Medical Research, The University of Sydney, Glebe, NSW, Australia.,Discipline of Pharmacology, The University of Sydney, NSW, Australia.,Central Clinical School, The University of Sydney, NSW, Australia
| | - Thais Mauad
- Department of Pathology, São Paulo University Medical School, São Paulo, Brazil
| | - Gavin Tjin
- Respiratory Cellular and Molecular Biology Group, Woolcock Institute of Medical Research, The University of Sydney, Glebe, NSW, Australia.,Central Clinical School, The University of Sydney, NSW, Australia
| | - Jenny C Karlsson
- Lung Biology, Department of Experimental Medical Sciences, Medical Faculty, Lund University, Lund, Sweden
| | | |
Collapse
|
48
|
Fernando R, Atkins SJ, Smith TJ. Intersection of Chemokine and TSH Receptor Pathways in Human Fibrocytes: Emergence of CXCL-12/CXCR4 Cross Talk Potentially Relevant to Thyroid-Associated Ophthalmopathy. Endocrinology 2016; 157:3779-3787. [PMID: 27471912 PMCID: PMC5045511 DOI: 10.1210/en.2016-1382] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Fibrocytes are monocyte progenitor cells that have been implicated in normal and pathological tissue remodeling. Among the prominent chemokine receptors expressed by these cells is CXC motif receptor 4 (CXCR4), which, with its cognate ligand CXCL motif ligand 12 (CXCL-12), directs fibrocytes to sites of fibrosis. Fibrocytes have been implicated in the pathogenesis of thyroid-associated ophthalmopathy, the ocular manifestation of Graves' disease (GD), by virtue of their unique accumulation as CD34+ orbital fibroblasts (OFs). Fibrocytes also express high levels of functional TSH receptor (TSHR). Here, we determined CXCL-12 and CXCR4 expression in fibrocytes and GD-OF and whether that pathway interacts with TSHR. CXCL-12 is highly expressed in GD-OF, whereas CXCR4 levels are dramatically higher in fibrocytes. Levels of these proteins are differentially regulated by TSH in a cell type-specific manner. Further, CXCL-12 enhances the induction by TSH of IL-6 in fibrocytes but attenuates this induction in GD-OF. In contrast, in pure CD34+ OF, the interplay between TSH and CXCL-12 reverts to that observed in fibrocytes. Our results indicate that CXCL-12 enhances TSH actions in fibrocytes but inhibits them in GD-OF, a dichotomy imposed by factors emanating from CD34- OF. They also suggest a potentially important modulatory role for CD34- OF in determining the factors that influence pathological TSHR signaling in the TAO orbit.
Collapse
Affiliation(s)
- Roshini Fernando
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center and Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48105
| | - Stephen J Atkins
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center and Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48105
| | - Terry J Smith
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center and Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48105
| |
Collapse
|
49
|
Keeley EC, Schutt RC, Marinescu MA, Burdick MD, Strieter RM, Mehrad B. Circulating fibrocytes as predictors of adverse events in unstable angina. Transl Res 2016; 172:73-83.e1. [PMID: 27012475 PMCID: PMC4866880 DOI: 10.1016/j.trsl.2016.02.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 02/05/2016] [Accepted: 02/28/2016] [Indexed: 11/22/2022]
Abstract
Half of the patients who present with unstable angina (UA) develop recurrent symptoms over the subsequent year. Identification of patients destined to develop such adverse events would be clinically valuable, but current tools do not allow for this discrimination. Fibrocytes are bone marrow-derived progenitor cells that co-express markers of leukocytes and fibroblasts and are released into the circulation in the context of tissue injury. We hypothesized that, in patients with UA, the number of circulating fibrocytes predicts subsequent adverse events. We enrolled 55 subjects with UA, 18 with chronic stable angina, and 22 controls and correlated their concentration of circulating fibrocytes to clinical events (recurrent angina, myocardial infarction, revascularization, or death) over the subsequent year. Subjects with UA had a >2-fold higher median concentration of both total and activated fibrocytes compared with subjects with chronic stable angina and controls. In UA subjects, the concentration of total fibrocytes identified those who developed recurrent angina requiring revascularization (time-dependent area under the curve 0.85) and was superior to risk stratification using thrombolysis in myocardial infarction risk score and N-terminal pro B-type natriuretic peptide levels (area under the curve, 0.53 and 0.56, respectively, P < 0.001). After multivariable adjustment for thrombolysis in myocardial infarction predicted death, MI, or recurrent ischemia, total fibrocyte level was associated with recurrent angina (hazard ratio, 1.016 per 10,000 cells/mL increase; 95% confidence interval, 1.007-1.024; P < 0.001). Circulating fibrocytes are elevated in patients with UA and successfully risk stratify them for adverse clinical outcomes. Fibrocytes may represent a novel biomarker of outcome in this population.
Collapse
Affiliation(s)
- Ellen C Keeley
- Department of Medicine, University of Virginia, Charlottesville, Va; Division of Cardiology, University of Virginia, Charlottesville, Va.
| | - Robert C Schutt
- Houston Methodist DeBakey Heart & Vascular Center, Houston, Tex
| | - Mark A Marinescu
- Department of Medicine, University of Virginia, Charlottesville, Va
| | - Marie D Burdick
- Department of Medicine, University of Virginia, Charlottesville, Va; Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, Va
| | - Robert M Strieter
- Department of Medicine, University of Virginia, Charlottesville, Va; Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, Va
| | - Borna Mehrad
- Department of Medicine, University of Virginia, Charlottesville, Va; Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, Va; The Carter Center for Immunology, University of Virginia, Charlottesville, Va
| |
Collapse
|
50
|
Horowitz JC, Osterholzer JJ, Marazioti A, Stathopoulos GT. "Scar-cinoma": viewing the fibrotic lung mesenchymal cell in the context of cancer biology. Eur Respir J 2016; 47:1842-54. [PMID: 27030681 DOI: 10.1183/13993003.01201-2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 02/28/2016] [Indexed: 12/22/2022]
Abstract
Lung cancer and pulmonary fibrosis are common, yet distinct, pathological processes that represent urgent unmet medical needs. Striking clinical and mechanistic parallels exist between these distinct disease entities. The goal of this article is to examine lung fibrosis from the perspective of cancer-associated phenotypic hallmarks, to discuss areas of mechanistic overlap and distinction, and to highlight profibrotic mechanisms that contribute to carcinogenesis. Ultimately, we speculate that such comparisons might identify opportunities to leverage our current understanding of the pathobiology of each disease process in order to advance novel therapeutic approaches for both. We anticipate that such "outside the box" concepts could be translated to a more precise and individualised approach to fibrotic diseases of the lung.
Collapse
Affiliation(s)
- Jeffrey C Horowitz
- Division of Pulmonary and Critical Care Medicine, Dept of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - John J Osterholzer
- Division of Pulmonary and Critical Care Medicine, Dept of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Antonia Marazioti
- Laboratory for Molecular Respiratory Carcinogenesis, Dept of Physiology, Faculty of Medicine, University of Patras, Rio, Greece
| | - Georgios T Stathopoulos
- Laboratory for Molecular Respiratory Carcinogenesis, Dept of Physiology, Faculty of Medicine, University of Patras, Rio, Greece Comprehensive Pneumology Center and Institute for Lung Biology and Disease, University Hospital, Ludwig-Maximilians University and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|