1
|
Mao Y, Suryawanshi A, Patial S, Saini Y. Airspaces-derived exosomes contain disease-relevant protein signatures in a mouse model of cystic fibrosis (CF)-like mucoinflammatory lung disease. Front Pharmacol 2024; 15:1460692. [PMID: 39386033 PMCID: PMC11461968 DOI: 10.3389/fphar.2024.1460692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 08/26/2024] [Indexed: 10/12/2024] Open
Abstract
Exosomes, membrane-bound extracellular vesicles, ranging from approximately 30-200 nm in diameter, are released by almost all cell types and play critical roles in intercellular communication. In response to the prevailing stress, the exosome-bound protein signatures vary in abundance and composition. To identify the bronchoalveolar lavage fluid (BALF) exosome-bound proteins associated with mucoinflammatory lung disease and to gain insights into their functional implications, we compared BALF exosomes-derived proteins from adult Scnn1b transgenic (Scnn1b-Tg+) and wild type (WT) mice. A total of 3,144 and 3,119 proteins were identified in BALF exosomes from Scnn1b-Tg+ and WT mice, respectively. Using cutoff criteria (Log2 fold-change > 1 and adjusted p-value < 0.05), the comparison of identified proteins revealed 127 and 30 proteins that were significantly upregulated and downregulated, respectively, in Scnn1b-Tg+ versus WT mice. In addition, 52 and 27 proteins were exclusively enriched in Scnn1b-Tg+ and WT mice, respectively. The identified exosome-bound proteins from the homeostatic airspaces of WT mice were mostly relevant to the normal physiological processes. The protein signatures enriched in the BALF exosomes of Scnn1b-Tg+ mice were relevant to macrophage activation and mucoinflammatory processes. Ingenuity pathway analyses revealed that the enriched proteins in Scnn1b-Tg+ mice contributed to the inflammatory responses and antimicrobial defense pathways. Selective proteins including, RETNLA/FIZZ1, LGALS3/Galectin-3, S100A8/MRP8, and CHIL3/YM1 were immunolocalized to specific cell types. The comparative analysis between enriched BALF exosome proteins and previously identified differentially upregulated genes in Scnn1b-Tg+ versus WT mice suggested that the compartment-/cell-specific upregulation in gene expression dictates the enrichment of their respective proteins in the lung airspaces. Taken together, this study demonstrates that the BALF exosome-bound protein signatures reflect disease-relevant disturbances. Our findings suggest that the exosomes carry disease-relevant protein signatures that can be used as a diagnostic as well as predictive biomarkers for mucoinflammatory lung disease.
Collapse
Affiliation(s)
- Yun Mao
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Amol Suryawanshi
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Sonika Patial
- Comparative and Molecular Pathogenesis Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, United States
| | - Yogesh Saini
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
2
|
Kawasaki T, Takeda Y, Kumanogoh A. Proteomics of blood extracellular vesicles in inflammatory respiratory diseases for biomarker discovery and new insights into pathophysiology. Inflamm Regen 2024; 44:38. [PMID: 39294831 PMCID: PMC11409490 DOI: 10.1186/s41232-024-00351-4] [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: 07/09/2024] [Accepted: 09/02/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND Inflammatory respiratory diseases, such as interstitial lung disease (ILD), bronchial asthma (BA), chronic obstructive pulmonary disease (COPD), and respiratory infections, remain significant global health concerns owing to their chronic and severe nature. Emerging as a valuable resource, blood extracellular vesicles (EVs) offer insights into disease pathophysiology and biomarker discovery in these conditions. MAIN BODY This review explores the advancements in blood EV proteomics for inflammatory respiratory diseases, highlighting their potential as non-invasive diagnostic and prognostic tools. Blood EVs offer advantages over traditional serum or plasma samples. Proteomic analyses of blood EVs have revealed numerous biomarkers that can be used to stratify patients, predict disease progression, and identify candidate therapeutic targets. Blood EV proteomics has identified proteins associated with progressive fibrosis in ILD, offering new avenues of treatment. In BA, eosinophil-derived EVs harbor biomarkers crucial for managing eosinophilic inflammation. Research on COPD has also identified proteins that correlate with lung function. Moreover, EVs play a critical role in respiratory infections such as COVID-19, and disease-associated proteins are encapsulated. Thus, proteomic studies have identified key molecules involved in disease severity and immune responses, underscoring their role in monitoring and guiding therapy. CONCLUSION This review highlights the potential of blood EV proteomics as a non-invasive diagnostic and prognostic tool for inflammatory respiratory diseases, providing a promising avenue for improved patient management and therapeutic development.
Collapse
Affiliation(s)
- Takahiro Kawasaki
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
- Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, Japan.
- Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Suita, Osaka, Japan.
| | - Yoshito Takeda
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka, Japan
- Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Suita, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan
- Japan Agency for Medical Research and Development - Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Suita, Osaka, Japan
- Center for Advanced Modalities and DDS (CAMaD), Osaka University, Osaka, Japan
| |
Collapse
|
3
|
Lit KK, Zhirenova Z, Blocki A. Insulin-like growth factor-binding protein 7 (IGFBP7): A microenvironment-dependent regulator of angiogenesis and vascular remodeling. Front Cell Dev Biol 2024; 12:1421438. [PMID: 39045455 PMCID: PMC11263173 DOI: 10.3389/fcell.2024.1421438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 06/10/2024] [Indexed: 07/25/2024] Open
Abstract
Insulin-like Growth Factor-Binding Protein 7 (IGFBP7) is an extracellular matrix (ECM) glycoprotein, highly enriched in activated vasculature during development, physiological and pathological tissue remodeling. Despite decades of research, its role in tissue (re-)vascularization is highly ambiguous, exhibiting pro- and anti-angiogenic properties in different tissue remodeling states. IGFBP7 has multiple binding partners, including structural ECM components, cytokines, chemokines, as well as several receptors. Based on current evidence, it is suggested that IGFBP7's bioactivity is strongly dependent on the microenvironment it is embedded in. Current studies indicate that during physiological angiogenesis, IGFBP7 promotes endothelial cell attachment, luminogenesis, vessel stabilization and maturation. Its effects on other stages of angiogenesis and vessel function remain to be determined. IGFBP7 also modulates the pro-angiogenic properties of other signaling factors, such as VEGF-A and IGF, and potentially acts as a growth factor reservoir, while its actual effects on the factors' signaling may depend on the environment IGFBP7 is embedded in. Besides (re-)vascularization, IGFBP7 clearly promotes progenitor and stem cell commitment and may exhibit anti-inflammatory and anti-fibrotic properties. Nonetheless, its role in inflammation, immunomodulation, fibrosis and cellular senescence is again likely to be context-dependent. Future studies are required to shed more light on the intricate functioning of IGFBP7.
Collapse
Affiliation(s)
- Kwok Keung Lit
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine (CNRM), Hong Kong Science Park, Shatin, Hong Kong SAR, China
| | - Zhamilya Zhirenova
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine (CNRM), Hong Kong Science Park, Shatin, Hong Kong SAR, China
| | - Anna Blocki
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine (CNRM), Hong Kong Science Park, Shatin, Hong Kong SAR, China
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| |
Collapse
|
4
|
Chu M, Wang R, Jing X, Li D, Fu G, Deng J, Xu Z, Zhao J, Liu Z, Fan Q, Pei L, Zeng Z, Liu C, Chen Z, Lu J, Liu XA. Conventional and multi-omics assessments of subacute inhalation toxicity due to propylene glycol and vegetable glycerin aerosol produced by electronic cigarettes. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 271:116002. [PMID: 38277972 DOI: 10.1016/j.ecoenv.2024.116002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 01/09/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024]
Abstract
Propylene glycol (PG) and vegetable glycerin (VG) are the most common solvents used in electronic cigarette liquids. No long-term inhalation toxicity assessments have been performed combining conventional and multi-omics approaches on the potential respiratory effects of the solvents in vivo. In this study, the systemic toxicity of aerosol generated from a ceramic heating coil-based e-cigarette was evaluated. First, the aerosol properties were characterized, including carbonyl emissions, the particle size distribution, and aerosol temperatures. To determine toxicological effects, rats were exposed, through their nose only, to filtered air or a propylene glycol (PG)/ glycerin (VG) (50:50, %W/W) aerosol mixture at the target concentration of 3 mg/L for six hours daily over a continuous 28-day period. Compared with the air group, female rats in the PG/VG group exhibited significantly lower body weights during both the exposure period and recovery period, and this was linked to a reduced food intake. Male rats in the PG/VG group also experienced a significant decline in body weight during the exposure period. Importantly, rats exposed to the PG/VG aerosol showed only minimal biological effects compared to those with only air exposure, with no signs of toxicity. Moreover, the transcriptomic, proteomic, and metabolomic analyses of the rat lung tissues following aerosol exposure revealed a series of candidate pathways linking aerosol inhalation to altered lung functions, especially the inflammatory response and disease. Dysregulated pathways of arachidonic acids, the neuroactive ligand-receptor interaction, and the hematopoietic cell lineage were revealed through integrated multi-omics analysis. Therefore, our integrated multi-omics approach offers novel systemic insights and early evidence of environmental-related health hazards associated with an e-cigarette aerosol using two carrier solvents in a rat model.
Collapse
Affiliation(s)
- Ming Chu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China; Laboratory of Life and Health Sciences, Shenzhen First union Technology Co., Ltd, Shenzhen 518103, China
| | - Ruoxi Wang
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Xiaoyuan Jing
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Ding Li
- Laboratory of Life and Health Sciences, Shenzhen First union Technology Co., Ltd, Shenzhen 518103, China; Laboratory of Life and Health Sciences, Shenzhen Health Union Biotechnology Co., Ltd, Shenzhen 518103, China
| | - Guofeng Fu
- Laboratory of Life and Health Sciences, Shenzhen First union Technology Co., Ltd, Shenzhen 518103, China
| | - Jingjing Deng
- Laboratory of Life and Health Sciences, Shenzhen Health Union Biotechnology Co., Ltd, Shenzhen 518103, China
| | - Zhibin Xu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Jing Zhao
- Laboratory of Life and Health Sciences, Shenzhen Health Union Biotechnology Co., Ltd, Shenzhen 518103, China
| | - Zhang Liu
- Laboratory of Life and Health Sciences, Shenzhen Health Union Biotechnology Co., Ltd, Shenzhen 518103, China
| | - Qiming Fan
- Guangdong Zhongke EnHealth Science and Technology Co., Ltd. Foshan 528000, China
| | - Lanjie Pei
- Hubei Provincial Key Laboratory for Applied Toxicology, Hubei Provincial Center for Disease Control and Prevention, Wuhan 430079, China
| | - Zhi Zeng
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Chuan Liu
- Laboratory of Life and Health Sciences, Shenzhen First union Technology Co., Ltd, Shenzhen 518103, China
| | - Zuxin Chen
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS); Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Jin Lu
- Laboratory of Life and Health Sciences, Shenzhen First union Technology Co., Ltd, Shenzhen 518103, China; Laboratory of Life and Health Sciences, Shenzhen Health Union Biotechnology Co., Ltd, Shenzhen 518103, China.
| | - Xin-An Liu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
5
|
Escarrer-Garau G, Martín-Medina A, Truyols-Vives J, Gómez-Bellvert C, Elowsson L, Westergren-Thorsson G, Molina-Molina M, Mercader-Barceló J, Sala-Llinàs E. In Vivo and In Vitro Pro-Fibrotic Response of Lung-Resident Mesenchymal Stem Cells from Patients with Idiopathic Pulmonary Fibrosis. Cells 2024; 13:160. [PMID: 38247851 PMCID: PMC10814068 DOI: 10.3390/cells13020160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/23/2024] Open
Abstract
Lung-resident mesenchymal stem cells (LR-MSC) are thought to participate in idiopathic pulmonary fibrosis (IPF) by differentiating into myofibroblasts. On the other hand, LR-MSC in IPF patients present senescence-related features. It is unclear how they respond to a profibrotic environment. Here, we investigated the profibrotic response of LR-MSC isolated from IPF and control (CON) patients. LR-MSC were inoculated in mice 48 h after bleomycin (BLM) instillation to analyze their contribution to lung damage. In vitro, LR-MSC were exposed to TGFβ. Mice inoculated with IPF LR-MSC exhibited worse maintenance of their body weight. The instillation of either IPF or CON LR-MSC sustained BLM-induced histological lung damage, bronchoalveolar lavage fluid cell count, and the expression of the myofibroblast marker, extracellular matrix (ECM) proteins, and proinflammatory cytokines in the lungs. In vitro, IPF LR-MSC displayed higher basal protein levels of aSMA and fibronectin than CON LR-MSC. However, the TGFβ response in the expression of TGFβ, aSMA, and ECM genes was attenuated in IPF LR-MSC. In conclusion, IPF LR-MSC have acquired myofibroblastic features, but their capacity to further respond to profibrotic stimuli seems to be attenuated. In an advanced stage of the disease, LR-MSC may participate in disease progression owing to their limited ability to repair epithelial damage.
Collapse
Affiliation(s)
| | - Aina Martín-Medina
- iRESPIRE Research Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
| | - Joan Truyols-Vives
- MolONE Research Group, University of the Balearic Islands (UIB), 07122 Palma, Spain
| | | | - Linda Elowsson
- Lung Biology, Department of Experimental Medical Science, Lund University, 08908 Lund, Sweden
| | | | - Maria Molina-Molina
- ILD Unit, Respiratory Department, University Hospital of Bellvitge-Bellvitge Biomedical Research Institute (IDIBELL), 08908 Hospitalet de Llobregat, Barcelona, Spain
- Centre of Biomedical Research Network in Respiratory Diseases (CIBERES), 28029 Madrid, Spain
| | - Josep Mercader-Barceló
- MolONE Research Group, University of the Balearic Islands (UIB), 07122 Palma, Spain
- iRESPIRE Research Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
- Centre of Biomedical Research Network in Respiratory Diseases (CIBERES), 28029 Madrid, Spain
| | - Ernest Sala-Llinàs
- iRESPIRE Research Group, Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
- Centre of Biomedical Research Network in Respiratory Diseases (CIBERES), 28029 Madrid, Spain
| |
Collapse
|
6
|
Li L, Huang J, Liu Y. The extracellular matrix glycoprotein fibrillin-1 in health and disease. Front Cell Dev Biol 2024; 11:1302285. [PMID: 38269088 PMCID: PMC10806136 DOI: 10.3389/fcell.2023.1302285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/19/2023] [Indexed: 01/26/2024] Open
Abstract
Fibrillin-1 (FBN1) is a large, cysteine-rich, calcium binding extracellular matrix glycoprotein encoded by FBN1 gene. It serves as a structural component of microfibrils and provides force-bearing mechanical support in elastic and nonelastic connective tissue. As such, mutations in the FBN1 gene can cause a wide variety of genetic diseases such as Marfan syndrome, an autosomal dominant disorder characterized by ocular, skeletal and cardiovascular abnormalities. FBN1 also interacts with numerous microfibril-associated proteins, growth factors and cell membrane receptors, thereby mediating a wide range of biological processes such as cell survival, proliferation, migration and differentiation. Dysregulation of FBN1 is involved in the pathogenesis of many human diseases, such as cancers, cardiovascular disorders and kidney diseases. Paradoxically, both depletion and overexpression of FBN1 upregulate the bioavailability and signal transduction of TGF-β via distinct mechanisms in different settings. In this review, we summarize the structure and expression of FBN1 and present our current understanding of the functional role of FBN1 in various human diseases. This knowledge will allow to develop better strategies for therapeutic intervention of FBN1 related diseases.
Collapse
Affiliation(s)
- Li Li
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Junxin Huang
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| | - Youhua Liu
- State Key Laboratory of Organ Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- National Clinical Research Center of Kidney Disease, Guangdong Provincial Institute of Nephrology, Guangzhou, China
| |
Collapse
|
7
|
Mismetti V, Delavenne X, Montani D, Bezzeghoud S, Delezay O, Hodin S, Launay D, Marchand-Adam S, Nunes H, Ollier E, Reynaud-Gaubert M, Pastre J, Traclet J, Quetant S, Zeghmar S, Bertoletti L, Cottin V. Proteomic biomarkers for survival in systemic sclerosis-associated pulmonary hypertension. Respir Res 2023; 24:273. [PMID: 37936223 PMCID: PMC10629050 DOI: 10.1186/s12931-023-02578-0] [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: 07/11/2023] [Accepted: 10/24/2023] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND Interstitial lung disease (ILD) and pulmonary hypertension (PH) represent the major causes of mortality in systemic sclerosis (SSc). Patients with systemic sclerosis and combined PH and ILD (SSc-PH-ILD) generally have a poor prognosis. Predictors of survival and of potential benefit of treatment are lacking in patients with SSc-PH-ILD. OBJECTIVE To identify specific plasma protein expression patterns associated with survival in patients with SSc-PH-ILD. MATERIALS AND METHODS Post-hoc analysis of a prospective multicenter French study in patients with PH-ILD. An untargeted proteomic analysis using mass spectrometry was performed to identify plasma protein changes associated with long-term overall survival in patients with SSc-PH-ILD. RESULTS Thirty two patients were included in the analysis, of whom 13 died during follow-up (median survival: 76.5 months). At baseline, survivors had less severe hemodynamic impairment [pulmonary vascular resistance of 4.4 Wood Units (IQR 3-5.2) vs. 6.2 Wood Units (IQR 4.2-10.7)] and higher carbon monoxide diffusing capacity [median 39% (IQR 35-44%) vs. 25% (IQR 22-30.5%)], than the 13 patients who died. Seven proteins, associated with haemostasis and fibrosis, were differentially expressed according to patients' survival. In the survivor group, two proteins were increased (ADAMTS13, SERPIND1) and five were decreased (PTGDS, OLFM1, C7, IGFBP7, FBN1) compared to the non-survivor groups. CONCLUSION The prognosis of SSc-PH-ILD patients is poor. This proteomic approach found 7 plasma proteins (involved in haemostasis and fibrosis pathways) associated with survival. These potential biomarkers may be good candidates to prognostic enrichment.
Collapse
Affiliation(s)
- Valentine Mismetti
- Université Jean Monnet Saint-Étienne, Mines Saint-Etienne, INSERM, SAINBIOSE U1059, 42023, Saint-Etienne, France
| | - Xavier Delavenne
- Université Jean Monnet Saint-Étienne, Mines Saint-Etienne, INSERM, SAINBIOSE U1059, 42023, Saint-Etienne, France
| | - David Montani
- Service de Pneumologie et Soins Intensifs Thoraciques, Centre de Référence de L'Hypertension Pulmonaire, INSERM U999 Centre de Compétence Maladies Pulmonaires Rares-OrphaLung Hôpital de Bicêtre, Le Kremlin Bicêtre, Paris, France
| | - Souad Bezzeghoud
- Service de Médecine Vasculaire et Thérapeutique, INSERM, CIC-1408, CHU de Saint-Etienne, Saint-Etienne, France
| | - Olivier Delezay
- Université Jean Monnet Saint-Étienne, Mines Saint-Etienne, INSERM, SAINBIOSE U1059, 42023, Saint-Etienne, France
| | - Sophie Hodin
- Université Jean Monnet Saint-Étienne, Mines Saint-Etienne, INSERM, SAINBIOSE U1059, 42023, Saint-Etienne, France
| | - David Launay
- Service de Médecine Interne et d'Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes et Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO) CHU Lille, Lille, France
| | | | - Hilario Nunes
- Service de Pneumologie, Centre de Référence des Maladies Pulmonaires Rares, Hôpital Avicenne, INSERM U1272, Université Sorbonne Paris Nord, Bobigny, France
| | - Edouard Ollier
- Université Jean Monnet Saint-Étienne, Mines Saint-Etienne, INSERM, SAINBIOSE U1059, 42023, Saint-Etienne, France
| | - Martine Reynaud-Gaubert
- Centre de Compétence des Maladies Pulmonaires Rares (OrphaLung), Service de Pneumologie et Transplantation Pulmonaire, CHU Nord, AP-HM, Aix Marseille Université, Marseille, France
| | - Jean Pastre
- Service de Pneumologie et Soins Intensifs Centre de Compétence Maladie Pulmonaire Rare (OrphaLung) Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Julie Traclet
- National Reference Center for Rare Pulmonary Diseases, Department of Respiratory Diseases, Hospices Civils de Lyon, Louis Pradel Hospital, 69677, Lyon, France
| | | | - Sabrina Zeghmar
- National Reference Center for Rare Pulmonary Diseases, Department of Respiratory Diseases, Hospices Civils de Lyon, Louis Pradel Hospital, 69677, Lyon, France
| | - Laurent Bertoletti
- Service de Médecine Vasculaire et Thérapeutique, INSERM, UMR1059, Université Jean-Monnet, INSERM, CIC-1408, CHU de Saint-Etienne, INNOVTE, 42055, Saint-Etienne, France.
| | - Vincent Cottin
- National Reference Center for Rare Pulmonary Diseases, Department of Respiratory Diseases, Hospices Civils de Lyon, Louis Pradel Hospital, 69677, Lyon, France
- Univ Lyon, INRA, UMR754, 69008, Lyon, France
| |
Collapse
|
8
|
Chang J, Zou S, Xiao Y, Zhu D. Identification and validation of targets of swertiamarin on idiopathic pulmonary fibrosis through bioinformatics and molecular docking-based approach. BMC Complement Med Ther 2023; 23:352. [PMID: 37798725 PMCID: PMC10557187 DOI: 10.1186/s12906-023-04171-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/15/2023] [Indexed: 10/07/2023] Open
Abstract
BACKGROUND Swertiamarin is the main hepatoprotective component of Swertiapatens and has anti-inflammatory and antioxidation effects. Our previous study showed that it was a potent inhibitor of idiopathic pulmonary fibrosis (IPF) and can regulate the expressions of α-smooth muscle actin (α-SMA) and epithelial cadherin (E-cadherin), two markers of the TGF-β/Smad (transforming growth factor beta/suppressor of mothers against decapentaplegic family) signaling pathway. But its targets still need to be investigated. The main purpose of this study is to identify the targets of swertiamarin. METHODS GEO2R was used to analyze the differentially expressed genes (DEGs) of GSE10667, GSE110147, and GSE71351 datasets from the Gene Expression Omnibus (GEO) database. The DEGs were then enriched with Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis for their biological functions and annotated terms. The protein-protein interaction (PPI) network was constructed to identify hub genes. The identified hub genes were predicted for their bindings to swertiamarin by molecular docking (MD) and validated by experiments. RESULTS 76 upregulated and 27 downregulated DEGs were screened out. The DEGs were enriched in the biological function of cellular component (CC) and 7 cancer-related signaling pathways. Three hub genes, i.e., LOX (lysyl oxidase), COL5A2 (collagen type V alpha 2 chain), and CTGF (connective tissue growth factor) were selected, virtually tested for the interactions with swertiamarin by MD, and validated by in vitro experiments. CONCLUSION LOX, COL5A2, and CTGF were identified as the targets of swertiamarin on IPF.
Collapse
Affiliation(s)
- Jun Chang
- College of Life Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China.
| | - Shaoqing Zou
- College of Life Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Yiwen Xiao
- College of Life Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Du Zhu
- College of Life Science, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China.
| |
Collapse
|
9
|
Kalia V, Baccarelli AA, Happel C, Hollander JA, Jukic AM, McAllister KA, Menon R, Merrick BA, Milosavljevic A, Ravichandran LV, Roth ME, Subramanian A, Tyson FL, Worth L, Shaughnessy DT. Seminar: Extracellular Vesicles as Mediators of Environmental Stress in Human Disease. ENVIRONMENTAL HEALTH PERSPECTIVES 2023; 131:104201. [PMID: 37861803 PMCID: PMC10588739 DOI: 10.1289/ehp12980] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 09/27/2023] [Accepted: 09/28/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND Extracellular vesicles (EVs), membrane-bound particles containing a variety of RNA types, DNA, proteins, and other macromolecules, are now appreciated as an important means of communication between cells and tissues, both in normal cellular physiology and as a potential indicator of cellular stress, environmental exposures, and early disease pathogenesis. Extracellular signaling through EVs is a growing field of research for understanding fundamental mechanisms of health and disease and for the potential for biomarker discovery and therapy development. EVs are also known to play important roles in mediating the effects of exposure to environmental stress. OBJECTIVES This seminar addresses the application of new tools and approaches for EV research, developed in part through the National Institutes of Health (NIH) Extracellular RNA Communication Program, and reflects presentations and discussions from a workshop held 27-28 September 2021 by the National Institute of Environmental Health Sciences (NIEHS) and the National Center for Advancing Translational Sciences (NCATS) on "Extracellular Vesicles, Exosomes, and Cell-Cell Signaling in Response to Environmental Stress." The panel of experts discussed current research on EVs and environmental exposures, highlighted recent advances in EV isolation and characterization, and considered research gaps and opportunities toward identifying and characterizing the roles for EVs in environmentally related diseases, as well as the current challenges and opportunities in this field. DISCUSSION The authors discuss the application of new experimental models, particularly organ-on-chip (OOC) systems and in vitro approaches and how these have the potential to extend findings in population-based studies of EVs in exposure-related diseases. Given the complex challenges of identifying cell-specific EVs related to environmental exposures, as well as the general heterogeneity and variability in EVs in blood and other accessible biological samples, there is a critical need for rigorous reporting of experimental methods and validation studies. The authors note that these efforts, combined with cross-disciplinary approaches, would ensure that future research efforts in environmental health studies on EV biomarkers are rigorous and reproducible. https://doi.org/10.1289/EHP12980.
Collapse
Affiliation(s)
- Vrinda Kalia
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Andrea A. Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Christine Happel
- National Center for Advancing Translational Sciences, National Institutes of Health (NIH), U.S. Department of Health and Human Services (DHHS), Bethesda, Maryland, USA
| | - Jonathan A. Hollander
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Anne Marie Jukic
- Division of Intramural Research, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Kimberly A. McAllister
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Ramkumar Menon
- Department of Obstetrics and Gynecology, Division of Basic Science and Translational Research, University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Bruce A. Merrick
- Division of Translational Toxicology, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | | | - Lingamanaidu V. Ravichandran
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Matthew E. Roth
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Anita Subramanian
- Division of Intramural Research, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Frederick L. Tyson
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Leroy Worth
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Daniel T. Shaughnessy
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, Research Triangle Park, North Carolina, USA
| |
Collapse
|
10
|
Chen Y, Li X, Fan X. Integrated proteomics and metabolomics reveal variations in pulmonary fibrosis development and the potential therapeutic effect of Shuangshen Pingfei formula. JOURNAL OF ETHNOPHARMACOLOGY 2023; 303:115894. [PMID: 36356715 DOI: 10.1016/j.jep.2022.115894] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 10/18/2022] [Accepted: 10/29/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shuangshen Pingfei formula (SSPF), a Chinese medicine prescription, has been prescribed to alleviate PF. However, little is known about the molecular mechanism underlying PF progression and the regulatory mechanism in SSPF. AIMS OF THE STUDY To discriminate the molecular alterations underlying the development of pulmonary fibrosis (PF) and reveal the regulatory mechanism of Shuangshen Pingfei formula (SSPF). MATERIALS AND METHODS An integrated analysis of a time-course pathology combined with proteomics and metabolomics was performed to investigate changes in body weight, survival rate, lung coefficient, histopathology, proteins, and metabolites of lung tissues at different time points upon bleomycin (BLM) exposure and SSPF treatment. RESULTS The results showed that PF progression was characterized by gradually aggravated fibrosis accompanied by inflammation with extended exposure (7, 14, and 21 days). SSPF significantly attenuated lung fibrosis, as evidenced by increased weight, and reduced lung coefficients and fibrosis scores. Moreover, 368 common differentially expressed proteins (DEPs) were identified, and 102 DEPs were continuously and monotonically upregulated via proteomics among the three BLM treatments. The DEPs were principally involved in extracellular matrix (ECM) remodeling and arginine and proline (AP) metabolic reprogramming. Additionally, metabolomics analyses revealed that BLM exposure mainly affected six metabolism pathways, including 34 differentially regulated metabolites (DRMs). Furthermore, correlation analysis found that several DEPs and DRMs, including L-ornithine, S-adenosyl-L-methionine, ARG, and AOC1, were associated with arginine and proline metabolism, and 8,9-EET, 8,9-DHET, CYP2B, etc., were involved in arachidonic acid (AA) metabolism, suggesting that these two pathways play a critical role in the development of fibrosis. After SSPF treatment, the related protein expression and metabolic disorders were regulated, implying that SSPF provides potential solutions to target these pathways for benefit in the treatment of PF. CONCLUSION Our data suggest that ECM remodeling, and metabolic reprogramming of AP and AA are distinctive features of PF development. Simultaneously, we confirmed that SSPF could effectively regulate metabolic disorders, indicating its potential clinical application for PF therapy. Our findings using multiple approaches provide a molecular-scale perspective on the mechanisms of PF progression and the amelioration of SSPF.
Collapse
Affiliation(s)
- Yeqing Chen
- College of Integrated Chinese and Western Medicine, College of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Xiaolin Li
- College of Integrated Chinese and Western Medicine, College of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xinsheng Fan
- College of Integrated Chinese and Western Medicine, College of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
11
|
Sun X, Ding T, Wang B, Chang Z, Fei H, Geng L, Wang Y. Identification of lncRNA-miRNA-mRNA networks in circulating exosomes as potential biomarkers for systemic sclerosis. Front Med (Lausanne) 2023; 10:1111812. [PMID: 36873898 PMCID: PMC9977830 DOI: 10.3389/fmed.2023.1111812] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/18/2023] [Indexed: 02/18/2023] Open
Abstract
Objective This study aimed to analyze potential biomarkers for systemic sclerosis (SSc) by constructing lncRNA-miRNA-mRNA networks in circulating exosomes (cirexos). Materials and methods Differentially expressed mRNAs (DEmRNAs) and lncRNAs (DElncRNAs) in SSc cirexos were screened using high-throughput sequencing and detected with real-time quantitative PCR (RT-qPCR). Differentially expressed genes (DEGs) were analyzed using the DisGeNET, GeneCards, GSEA4.2.3, Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases. Receiver operating characteristic (ROC) curves, correlation analyses, and a double-luciferase reporter gene detection assay were used to analyze competing endogenous RNA (ceRNA) networks and clinical data. Results In this study, 286 DEmRNAs and 192 DElncRNAs were screened, of which 18 DEGs were the same as the SSc-related genes. The main SSc-related pathways included extracellular matrix (ECM) receptor interaction, local adhesion, platelet activation, and IgA production by the intestinal immune network. A hub gene, COL1A1, was obtained by a protein-protein interaction (PPI) network. Four ceRNA networks were predicted through Cytoscape. The relative expression levels of COL1A1, ENST0000313807, and NON-HSAT194388.1 were significantly higher in SSc, while the relative expression levels of hsa-miR-29a-3p, hsa-miR-29b-3p, and hsa-miR-29c-3p were significantly lower in SSc (P < 0.05). The ROC curve showed that the ENST00000313807-hsa-miR-29a-3p-COL1A1 network as a combined biomarker of SSc is more valuable than independent diagnosis, and that it is correlated with high-resolution CT (HRCT), Scl-70, C-reactive protein (CRP), Ro-52, IL-10, IgM, lymphocyte percentage, neutrophil percentage, albumin divided by globulin, urea, and RDW-SD (P < 0.05). Double-luciferase reporter gene detection showed that ENST00000313807 interacts with hsa-miR-29a-3p, which interacts with COL1A1. Conclusion The ENST00000313807-hsa-miR-29a-3p-COL1A1 network in plasma cirexos represents a potential combined biomarker for the clinical diagnosis and treatment of SSc.
Collapse
Affiliation(s)
- Xiaolin Sun
- The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China
| | - Tiantian Ding
- The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China
| | - Baoyue Wang
- The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China
| | - Zhifang Chang
- The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China
| | - Hongchang Fei
- The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China
| | - Lixia Geng
- The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China
| | - Yongfu Wang
- The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China
| |
Collapse
|
12
|
McNamee N, de la Fuente LR, Santos-Martinez MJ, O'Driscoll L. Proteomics profiling identifies extracellular vesicles' cargo associated with tumour cell induced platelet aggregation. BMC Cancer 2022; 22:1023. [PMID: 36171564 PMCID: PMC9520807 DOI: 10.1186/s12885-022-10068-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 08/23/2022] [Indexed: 11/21/2022] Open
Abstract
Background Cancer patients have an increased risk of developing venous thromboembolism, with up to 30% dying within a month of their development. Some cancer cells are known to induce platelet aggregation, and this interaction is understood to contribute to thrombosis and haematogenous metastasis. Many researchers have reported on extracellular vesicles (EVs) released from platelets. However, less is known about how cancer cells’ EVs may affect platelet function. Here EVs released by triple-negative breast cancer (TNBC) cell line variants were extensively investigated in this regard. Methods EVs were separated from conditioned media of TNBC Hs578T and Hs578Ts(i)8 cells using filtration and ultracentrifugation and were characterised by nanoparticle tracking analysis, immunoblots, and transmission electron microscopy. Blood samples from consenting donors were procured, and their platelets collected by differential centrifugation. Light transmission aggregometry and optical microscopy evaluated the potential interaction of TNBC cells and their EVs with platelets. Global proteomic analysis was performed on the EVs, by in-solution digestion and mass spectrometry. Data analysis included the use of Perseus, FunRich, and Vesiclepedia. Immunoblotting was used as a secondary method to investigate some key EV cargo proteins identified by the global proteomics approach. Results Both TNBC cell variants induced platelet aggregation. Increasing cell numbers significantly reduced the time taken for platelet aggregation to occur. EVs released by the cells also resulted in platelet aggregation. The time to induce platelet aggregation was EV dose-dependent. Proteomics profiling and immunoblotting of the EVs’ cargo identified candidate proteins (including uPAR and PDGFRβ) that may be involved during this process. Conclusions TNBC cells induce platelet aggregation. Furthermore, the cell-free EVs induced this undesirable effect. A number of EV cargo proteins were identified that may be relevant as therapeutic targets. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-10068-7.
Collapse
Affiliation(s)
- Niamh McNamee
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland.,Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - Laura Rodriguez de la Fuente
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland.,Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Maria Jose Santos-Martinez
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland.,Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland. .,Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland. .,Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
13
|
Negrete-García MC, de Jesús Ramos-Abundis J, Alvarado-Vasquez N, Montes-Martínez E, Montaño M, Ramos C, Sommer B. Exosomal Micro-RNAs as Intercellular Communicators in Idiopathic Pulmonary Fibrosis. Int J Mol Sci 2022; 23:11047. [PMID: 36232350 PMCID: PMC9569972 DOI: 10.3390/ijms231911047] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/09/2022] [Accepted: 09/14/2022] [Indexed: 12/12/2022] Open
Abstract
Communication between neighboring or distant cells is made through a complex network that includes extracellular vesicles (EVs). Exosomes, which are a subgroup of EVs, are released from most cell types and have been found in biological fluids such as urine, plasma, and airway secretions like bronchoalveolar lavage (BAL), nasal lavage, saliva, and sputum. Mainly, the cargo exosomes are enriched with mRNAs and microRNAs (miRNAs), which can be transferred to a recipient cell consequently modifying and redirecting its biological function. The effects of miRNAs derive from their role as gene expression regulators by repressing or degrading their target mRNAs. Nowadays, various types of research are focused on evaluating the potential of exosomal miRNAs as biomarkers for the prognosis and diagnosis of different pathologies. Nevertheless, there are few reports on their role in the pathophysiology of idiopathic pulmonary fibrosis (IPF), a chronic lung disease characterized by progressive lung scarring with no cure. In this review, we focus on the role and effect of exosomal miRNAs as intercellular communicators in the onset and progression of IPF, as well as discussing their potential utility as therapeutic agents for the treatment of this disease.
Collapse
Affiliation(s)
- María Cristina Negrete-García
- Molecular Biology Laboratory, Department of Research in Pulmonary Fibrosis, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Calzada de Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico
| | - Javier de Jesús Ramos-Abundis
- Molecular Biology Laboratory, Department of Research in Pulmonary Fibrosis, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Calzada de Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico
- Higher School of Medicine Instituto Politécnico Nacional, Salvador Díaz Mirón esquina Plan de San Luis S/N, Miguel Hidalgo, Casco de Santo Tomás, Mexico City 11340, Mexico
| | - Noé Alvarado-Vasquez
- Biochemistry Department, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Calzada de Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico
| | - Eduardo Montes-Martínez
- Molecular Biology Laboratory, Department of Research in Pulmonary Fibrosis, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Calzada de Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico
| | - Martha Montaño
- Cell Biology Laboratory, Department of Research in Pulmonary Fibrosis, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Calzada de Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico
| | - Carlos Ramos
- Cell Biology Laboratory, Department of Research in Pulmonary Fibrosis, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Calzada de Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico
| | - Bettina Sommer
- Bronchial Hyperreactivity Research Department, National Institute of Respiratory Diseases “Ismael Cosío Villegas” Calzada de Tlalpan 4502, Col. Sección XVI, Mexico City 14080, Mexico
| |
Collapse
|
14
|
Demais V, Pohl A, Wunderlich KA, Pfaller AM, Kaplan L, Barthélémy A, Dittrich R, Puig B, Giebel B, Hauck SM, Pfrieger FW, Grosche A. Release of VAMP5-positive extracellular vesicles by retinal Müller glia in vivo. J Extracell Vesicles 2022; 11:e12254. [PMID: 36043482 PMCID: PMC9428896 DOI: 10.1002/jev2.12254] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/25/2022] [Accepted: 07/18/2022] [Indexed: 11/11/2022] Open
Abstract
Cell-cell interactions in the central nervous system are based on the release of molecules mediating signal exchange and providing structural and trophic support through vesicular exocytosis and the formation of extracellular vesicles. The specific mechanisms employed by each cell type in the brain are incompletely understood. Here, we explored the means of communication used by Müller cells, a type of radial glial cells in the retina, which forms part of the central nervous system. Using immunohistochemical, electron microscopic, and molecular analyses, we provide evidence for the release of distinct extracellular vesicles from endfeet and microvilli of retinal Müller cells in adult mice in vivo. We identify VAMP5 as a Müller cell-specific SNARE component that is part of extracellular vesicles and responsive to ischemia, and we reveal differences between the secretomes of immunoaffinity-purified Müller cells and neurons in vitro. Our findings suggest extracellular vesicle-based communication as an important mediator of cellular interactions in the retina.
Collapse
Affiliation(s)
- Valerie Demais
- Plateforme Imagerie In Vitro, CNRS UAR 3156, NeuropôleUniversity of StrasbourgStrasbourgFrance
| | - Anne Pohl
- Department of Physiological GenomicsBioMedical Center BMCLudwig‐Maximilian UniversityPlanegg‐MartinsriedGermany
- Institute of Human GeneticsUniversity of RegensburgRegensburgGermany
| | - Kirsten A. Wunderlich
- Department of Physiological GenomicsBioMedical Center BMCLudwig‐Maximilian UniversityPlanegg‐MartinsriedGermany
| | - Anna M. Pfaller
- Department of Physiological GenomicsBioMedical Center BMCLudwig‐Maximilian UniversityPlanegg‐MartinsriedGermany
| | - Lew Kaplan
- Department of Physiological GenomicsBioMedical Center BMCLudwig‐Maximilian UniversityPlanegg‐MartinsriedGermany
| | - Amelie Barthélémy
- Centre National de la Recherche ScientifiqueUniversité de StrasbourgInstitut des Neurosciences Cellulaires et IntégrativesStrasbourgFrance
| | - Robin Dittrich
- Institute for Transfusion MedicineUniversity Hospital EssenUniversity of Duisburg‐EssenEssenGermany
| | - Berta Puig
- Neurology DepartmentExperimental Research in Stroke and Inflammation (ERSI)University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Bernd Giebel
- Institute for Transfusion MedicineUniversity Hospital EssenUniversity of Duisburg‐EssenEssenGermany
| | - Stefanie M. Hauck
- Metabolomics and Proteomics Core and Research Unit Protein ScienceHelmholtz‐Zentrum MünchenMünchenGermany
| | - Frank W. Pfrieger
- Plateforme Imagerie In Vitro, CNRS UAR 3156, NeuropôleUniversity of StrasbourgStrasbourgFrance
- Centre National de la Recherche ScientifiqueUniversité de StrasbourgInstitut des Neurosciences Cellulaires et IntégrativesStrasbourgFrance
| | - Antje Grosche
- Department of Physiological GenomicsBioMedical Center BMCLudwig‐Maximilian UniversityPlanegg‐MartinsriedGermany
| |
Collapse
|
15
|
Velázquez-Enríquez JM, Ramírez-Hernández AA, Navarro LMS, Reyes-Avendaño I, González-García K, Jiménez-Martínez C, Castro-Sánchez L, Sánchez-Chino XM, Vásquez-Garzón VR, Baltiérrez-Hoyos R. Proteomic Analysis Reveals Differential Expression Profiles in Idiopathic Pulmonary Fibrosis Cell Lines. Int J Mol Sci 2022; 23:ijms23095032. [PMID: 35563422 PMCID: PMC9105114 DOI: 10.3390/ijms23095032] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/18/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, irreversible lung disorder of unknown cause. This disease is characterized by profibrotic activation of resident pulmonary fibroblasts resulting in aberrant deposition of extracellular matrix (ECM) proteins. However, although much is known about the pathophysiology of IPF, the cellular and molecular processes that occur and allow aberrant fibroblast activation remain an unmet need. To explore the differentially expressed proteins (DEPs) associated with aberrant activation of these fibroblasts, we used the IPF lung fibroblast cell lines LL97A (IPF-1) and LL29 (IPF-2), compared to the normal lung fibroblast cell line CCD19Lu (NL-1). Protein samples were quantified and identified using a label-free quantitative proteomic analysis approach by liquid chromatography-tandem mass spectrometry (LC-MS/MS). DEPs were identified after pairwise comparison, including all experimental groups. Gene Ontology (GO) enrichment analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG), and Protein–Protein Interaction (PPI) network construction were used to interpret the proteomic data. Eighty proteins expressed exclusively in the IPF-1 and IPF-2 clusters were identified. In addition, 19 proteins were identified up-regulated in IPF-1 and 10 in IPF-2; 10 proteins were down-regulated in IPF-1 and 2 in IPF-2 when compared to the NL-1 proteome. Using the search tool for retrieval of interacting genes/proteins (STRING) software, a PPI network was constructed between the DEPs and the 80 proteins expressed exclusively in the IPF-2 and IPF-1 clusters, containing 115 nodes and 136 edges. The 10 hub proteins present in the IPP network were identified using the CytoHubba plugin of the Cytoscape software. GO and KEGG pathway analyses showed that the hub proteins were mainly related to cell adhesion, integrin binding, and hematopoietic cell lineage. Our results provide relevant information on DEPs present in IPF lung fibroblast cell lines when compared to the normal lung fibroblast cell line that could play a key role during IPF pathogenesis.
Collapse
Affiliation(s)
- Juan Manuel Velázquez-Enríquez
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68120, Mexico; (J.M.V.-E.); (A.A.R.-H.); (I.R.-A.); (K.G.-G.)
| | - Alma Aurora Ramírez-Hernández
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68120, Mexico; (J.M.V.-E.); (A.A.R.-H.); (I.R.-A.); (K.G.-G.)
| | | | - Itayetzi Reyes-Avendaño
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68120, Mexico; (J.M.V.-E.); (A.A.R.-H.); (I.R.-A.); (K.G.-G.)
| | - Karina González-García
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68120, Mexico; (J.M.V.-E.); (A.A.R.-H.); (I.R.-A.); (K.G.-G.)
| | - Cristian Jiménez-Martínez
- Departamento de Ingeniería Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Adolfo López Mateos, Zacatenco, Av. Wilfrido Massieu Esq. Cda. Miguel Stampa S/N, Alcaldía Gustavo A. Madero, Mexico City 07738, Mexico;
| | - Luis Castro-Sánchez
- Conacyt-Centro Universitario de Investigaciones Biomédicas “CUIB”, Universidad de Colima, Colima 28045, Mexico;
| | - Xariss Miryam Sánchez-Chino
- Catedra-Conacyt, Departamento de Salud El Colegio de La Frontera Sur, Unidad Villahermosa, Tabasco 86280, Mexico;
| | | | - Rafael Baltiérrez-Hoyos
- Conacyt-Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68120, Mexico;
- Correspondence:
| |
Collapse
|
16
|
Jin Y, Qi G, Shou Y, Li D, Liu Y, Guan H, Zhang Q, Chen S, Luo J, Xu L, Li C, Ma W, Chen N, Zheng Y, Yu D. High throughput data-based, toxicity pathway-oriented development of a quantitative adverse outcome pathway network linking AHR activation to lung damages. JOURNAL OF HAZARDOUS MATERIALS 2022; 425:128041. [PMID: 34906874 DOI: 10.1016/j.jhazmat.2021.128041] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/05/2021] [Accepted: 12/06/2021] [Indexed: 06/14/2023]
Abstract
The quantitative adverse outcome pathway (qAOP) is proposed to inform dose-responses at multiple biological levels for the purpose of toxicity prediction. So far, qAOP models concerning human health are scarce. Previously, we proposed 5 key molecular pathways that led aryl hydrogen receptor (AHR) activation to lung damages. The present study assembled an AOP network based on the gene expression signatures of these toxicity pathways, and validated the network using publicly available high throughput data combined with machine learning models. In addition, the AOP network was quantitatively evaluated with omics approaches and bioassays, using 16HBE-CYP1A1 cells exposed to benzo(a)pyrene (BaP), a prototypical AHR activator. Benchmark dose (BMD) analysis of transcriptomics revealed that AHR gene held the lowest BMD value, whereas AHR pathway held the lowest point of departure (PoD) compared to the other 4 pathways. Targeted bioassays were further performed to quantitatively understand the cellular responses, including ROS generation, DNA damage, interleukin-6 production, and extracellular matrix increase marked by collagen expression. Eventually, response-response relationships were plotted using nonlinear model fitting. The present study developed a highly reliable AOP model concerning human health, and validated as well as quantitatively evaluated it, and such a method is likely to be adoptable for risk assessment.
Collapse
Affiliation(s)
- Yuan Jin
- School of Public Health, Qingdao University, Qingdao, China
| | - Guangshuai Qi
- School of Public Health, Qingdao University, Qingdao, China
| | - Yingqing Shou
- School of Public Health, Qingdao University, Qingdao, China
| | - Daochuan Li
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Yuzhen Liu
- School of Public Health, Qingdao University, Qingdao, China
| | - Heyuan Guan
- School of Public Health, Qingdao University, Qingdao, China
| | - Qianqian Zhang
- School of Public Health, Qingdao University, Qingdao, China
| | - Shen Chen
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Jiao Luo
- School of Public Health, Qingdao University, Qingdao, China
| | - Lin Xu
- School of Public Health, Qingdao University, Qingdao, China
| | - Chuanhai Li
- School of Public Health, Qingdao University, Qingdao, China
| | - Wanli Ma
- School of Public Health, Qingdao University, Qingdao, China
| | - Ningning Chen
- School of Public Health, Qingdao University, Qingdao, China
| | - Yuxin Zheng
- School of Public Health, Qingdao University, Qingdao, China
| | - Dianke Yu
- School of Public Health, Qingdao University, Qingdao, China.
| |
Collapse
|
17
|
Perryman L, Gray SG. Fibrosis in Mesothelioma: Potential Role of Lysyl Oxidases. Cancers (Basel) 2022; 14:981. [PMID: 35205728 PMCID: PMC8870010 DOI: 10.3390/cancers14040981] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 12/13/2022] Open
Abstract
Immunotherapies (such as checkpoint inhibitors) and standard chemotherapies (such as cisplatin) have limitations in the successful treatment of malignant pleural mesothelioma (MPM). Fibrosis is the accumulation of collagen in the extracellular matrix (ECM) of tissues, making them denser than that of healthy tissues and thereby affecting drug delivery and immune cell infiltration. Moreover, fibrosis severely affects the patient's breathing and quality of life. The production of collagen and its assembly is highly regulated by various enzymes such as lysyl oxidases. Many solid tumors aberrantly express the family of lysyl oxidases (LOX/LOXL). This review examines how LOX/LOXLs were found to be dysregulated in noncancerous and cancerous settings, discusses their roles in solid tumor fibrosis and pathogenesis and explores the role of fibrosis in the development and poor clinical outcomes of patients with MPM. We examine the current preclinical status of drugs targeting LOX/LOXLs and how the incorporation of such drugs may have therapeutic benefits in the treatment and management of patients with MPM.
Collapse
Affiliation(s)
- Lara Perryman
- Drug Discovery Department, Pharmaxis Ltd., Sydney, NSW 2086, Australia;
| | - Steven G. Gray
- Thoracic Oncology, Labmed Directorate, St James’s Hospital, D08 RX0X Dublin, Ireland
| |
Collapse
|
18
|
Ramírez-Hernández AA, Velázquez-Enríquez JM, Santos-Álvarez JC, López-Martínez A, Reyes-Jiménez E, Carrasco-Torres G, González-García K, Vásquez-Garzón VR, Baltierrez-Hoyos R. The Role of Extracellular Vesicles in Idiopathic Pulmonary Fibrosis Progression: An Approach on Their Therapeutics Potential. Cells 2022; 11:cells11040630. [PMID: 35203281 PMCID: PMC8870588 DOI: 10.3390/cells11040630] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 02/07/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fibrosing interstitial lung disease of unknown etiology. Different types of cells are involved in fibrogenesis, which is persistently physical and molecular stimulation, either directly or by interacting with bioactive molecules and extracellular vesicles (EVs). Current evidence suggests that EVs play an essential role in IPF development. EVs are released by a variety of cells, including fibroblasts, epithelial cells, and alveolar macrophages. In addition, EVs can transport bioactive molecules, such as lipids, proteins, and nucleic acids, which play a pivotal role in cellular communication. Several proposed mechanisms show that an acceptor cell can capture, absorb, or interact with EVs through direct fusion with the plasma membrane, ligand–receptor interaction, and endocytotic process, modifying the target cell. During fibrogenesis, the release of EVs is deregulated, increases the EVs amount, and the cargo content is modified. This alteration is closely associated with the maintenance of the fibrotic microenvironment. This review summarizes the current data on the participation of EVs secreted by the cells playing a critical role in IPF pathogenesis.
Collapse
Affiliation(s)
- Alma Aurora Ramírez-Hernández
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Mexico; (A.A.R.-H.); (J.M.V.-E.); (J.C.S.-Á.); (A.L.-M.); (E.R.-J.); (K.G.-G.)
| | - Juan Manuel Velázquez-Enríquez
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Mexico; (A.A.R.-H.); (J.M.V.-E.); (J.C.S.-Á.); (A.L.-M.); (E.R.-J.); (K.G.-G.)
| | - Jovito Cesar Santos-Álvarez
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Mexico; (A.A.R.-H.); (J.M.V.-E.); (J.C.S.-Á.); (A.L.-M.); (E.R.-J.); (K.G.-G.)
| | - Armando López-Martínez
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Mexico; (A.A.R.-H.); (J.M.V.-E.); (J.C.S.-Á.); (A.L.-M.); (E.R.-J.); (K.G.-G.)
| | - Edilburga Reyes-Jiménez
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Mexico; (A.A.R.-H.); (J.M.V.-E.); (J.C.S.-Á.); (A.L.-M.); (E.R.-J.); (K.G.-G.)
| | - Gabriela Carrasco-Torres
- Departamento de Nanociencias y Nanotecnología, Centro de Investigación y de Estudios Avanzados del IPN, Av. IPN 2508, la laguna Ticomán, Ciudad de Mexico 07360, Mexico;
| | - Karina González-García
- Laboratorio de Fibrosis y Cáncer, Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Mexico; (A.A.R.-H.); (J.M.V.-E.); (J.C.S.-Á.); (A.L.-M.); (E.R.-J.); (K.G.-G.)
| | - Verónica Rocío Vásquez-Garzón
- CONACYT-Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Mexico;
| | - Rafael Baltierrez-Hoyos
- CONACYT-Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca de Juárez 68120, Mexico;
- Correspondence:
| |
Collapse
|