1
|
Tourn J, Crescence L, Bruzzese L, Panicot-Dubois L, Dubois C. Cellular and Molecular Mechanisms Leading to Air Travel-Induced Thrombosis. Circ Res 2025; 136:115-134. [PMID: 39745986 DOI: 10.1161/circresaha.124.325208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Venous thromboembolism, characterized by deep vein thrombosis and pulmonary embolism, is the third cardiovascular disease in the world. Deep vein thrombosis occurs when a blood clot forms in areas of impaired blood flow, and it is significantly affected by environmental factors. Local hypoxia, caused by venous stasis, plays a critical role in deep vein thrombosis under normal conditions, and this effect is intensified when the Po2 decreases, such as during air travel or high-altitude exposure. The lower oxygen levels and reduced pressure at high altitudes further contribute to deep vein thrombosis development. These conditions increase the pro-coagulant activity of neutrophils, platelets, and red blood cells, which interact on the surface of activated endothelial cells, promoting clot formation. Understanding the mechanisms involved in thrombus formation when Po2 is reduced, with or without pressure reduction, is crucial for preventing the development of venous thromboembolisms in such conditions and identifying innovative therapeutic targets. This literature review explores the mechanisms involved in thrombus formation related to high-altitude conditions and discusses the pro-coagulant consequences induced by environmental disturbances.
Collapse
Affiliation(s)
- Julie Tourn
- Aix Marseille University, INSERM 1263, INRAE 1260, Center for CardioVascular and Nutrition Research (C2VN), Marseille, France (J.T., L.C., L.B., L.P.-D., C.D.)
| | - Lydie Crescence
- Aix Marseille University, INSERM 1263, INRAE 1260, Center for CardioVascular and Nutrition Research (C2VN), Marseille, France (J.T., L.C., L.B., L.P.-D., C.D.)
- Plateforme Aix Marseille, Plateforme d'Imagerie Vasculaire et de Microscopie Intravitale, C2VN, Marseille, France (L.C., L.B., L.P.-D., C.D.)
| | - Laurie Bruzzese
- Aix Marseille University, INSERM 1263, INRAE 1260, Center for CardioVascular and Nutrition Research (C2VN), Marseille, France (J.T., L.C., L.B., L.P.-D., C.D.)
- Plateforme Aix Marseille, Plateforme d'Imagerie Vasculaire et de Microscopie Intravitale, C2VN, Marseille, France (L.C., L.B., L.P.-D., C.D.)
| | - Laurence Panicot-Dubois
- Aix Marseille University, INSERM 1263, INRAE 1260, Center for CardioVascular and Nutrition Research (C2VN), Marseille, France (J.T., L.C., L.B., L.P.-D., C.D.)
- Plateforme Aix Marseille, Plateforme d'Imagerie Vasculaire et de Microscopie Intravitale, C2VN, Marseille, France (L.C., L.B., L.P.-D., C.D.)
| | - Christophe Dubois
- Aix Marseille University, INSERM 1263, INRAE 1260, Center for CardioVascular and Nutrition Research (C2VN), Marseille, France (J.T., L.C., L.B., L.P.-D., C.D.)
- Plateforme Aix Marseille, Plateforme d'Imagerie Vasculaire et de Microscopie Intravitale, C2VN, Marseille, France (L.C., L.B., L.P.-D., C.D.)
| |
Collapse
|
2
|
Li J, Sun L, Zhao Y. Advances in non-coding RNA as a biomarker for obstructive sleep apnoea hypoventilation syndrome. Sleep Breath 2024; 28:1899-1908. [PMID: 39017902 DOI: 10.1007/s11325-024-03109-x] [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: 01/10/2024] [Revised: 06/19/2024] [Accepted: 07/12/2024] [Indexed: 07/18/2024]
Abstract
PURPOSE Obstructive sleep apnoea hypoventilation syndrome (OSAHS) is a common sleep disorder that affects multiple body systems, which in turn is closely associated with cognitive dysfunction, diabetes mellitus, oncological cardiovascular diseases and metabolic disorders. In recent years, non-coding RNA (ncRNA) has emerged as a new opportunity for biomarker discovery. We therefore discuss the research progress and potential role of ncRNAs in obstructive sleep apnea hypoventilation syndrome. METHODS This review systematically searched relevant academic literature from PubMed, Web of Science and other databases. During the retrieval process, a combination of keywords such as "OSAHS", "ncRNA", "lncRNA", "miRAN", "circRNA" was used for search. RESULTS Circulating ncRNA has good area under the ROC curve, sensitivity and specificity in the diagnosis of OSAHS, and has the potential to become a diagnostic marker for OSAHS, while several circulating ncRNAs or circulating ncRNAs in combination with other tests such as the Obstructive Sleep Apnoea Screening Scale have a higher value of application as a test for OSAHS. Further analyses revealed that many circulating ncRNAs were significantly differentially expressed in the serum of OSAHS patients with different very severities, a potential marker for predicting the severity of OSAHS, and that the ncRNA content of patients' serum also had a significant effect during CPAP therapy, suggesting that it may have potential for therapeutic monitoring. Meanwhile, serum ncRNAs from patients have been shown to be effective in the diagnosis of OSAHS complications such as hypertension, Alzheimer's disease, acute myocardial infarction and atherosclerosis. The expression of up- or down-regulated ncRNAs can regulate different signalling pathways, which in turn affects various OSAHS complications such as pulmonary hypertension, diabetes mellitus, and cognitive dysfunction, and is expected to become a new direction for the treatment of these complications. CONCLUSIONS The changes in ncRNA expression in OSAHS patients are expected to be a novel biomarker for the diagnosis and treatment of OSAHS, and can also be used as a potential biomarker for the combination of diabetes mellitus, cardiovascular disease, respiratory disease, and cognitive dysfunction in OSAHS. It is believed that the continuous progress of ncRNA-related research is expected to promote the early detection, diagnosis and treatment of OSAHS and its complications.
Collapse
Affiliation(s)
- Jingli Li
- Kunming University of Science and Technology Affiliated The First People's Hospital of Yunnan Province, Kunming, 650500, Yunnan, China
| | - Limei Sun
- Kunming University of Science and Technology Affiliated Puer City People's Hospital, Puer, 665000, Yunnan, China
| | - Yuan Zhao
- Kunming University of Science and Technology Affiliated Puer City People's Hospital, Puer, 665000, Yunnan, China.
| |
Collapse
|
3
|
Zhang XB, Song Y, Lai YT, Qiu SZ, Hu AK, Li DX, Zheng NS, Zeng HQ, Lin QC. MiR-210-3p enhances intermittent hypoxia-induced tumor progression via inhibition of E2F3. Sleep Breath 2024; 28:607-617. [PMID: 37775619 DOI: 10.1007/s11325-023-02925-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 09/09/2023] [Accepted: 09/15/2023] [Indexed: 10/01/2023]
Abstract
PURPOSE Intermittent hypoxia (IH) is a hallmark of obstructive sleep apnea (OSA), which is related to tumorigenesis and progression. Although micro-ribonucleic acid-210-3p (miR-210-3p) is correlated with hypoxia-induced tumor development, its role in the relationship between IH and tumor function remains poorly understood. The present work focused on elucidating the molecular mechanism through which miR-210-3p drives tumor progression under IH. METHODS MiR-210-3p levels were quantified within tumor samples from patients with lung adenocarcinoma who had or did not have OSA. Correlations between miR-210-3p and polysomnographic variables were analyzed. For in vitro experiments, miR-210-3p was inhibited or overexpressed via transfection under IH conditions. Cell viability, growth, invasion and migration assays were carried out. For in vivo modeling of IH using mouse xenografts, a miR-210-3p antagomir was intratumorally injected, tumor biological behaviors were evaluated, and reverse transcription-quantitative polymerase chain reaction (RT-qPCR), immunohistochemistry and western blot were carried out for detecting miR-210-3p and E2F transcription factor 3 (E2F3) expression. RESULTS For patients with lung adenocarcinoma and OSA, high miR-210-3p levels showed positive relation to polysomnographic variables, such as oxygen desaturation index, apnea-hypopnea index, and proportion of total sleep time with oxygen saturation in arterial blood < 90%. IH enhanced tumor viability, proliferation, migration, and invasion, downregulated E2F3 expression, and increased miR-210-3-p levels. miR-210-3p overexpression induced similar changes. These changes were reversed by miR-210-3p inhibition in vitro or miR-210-3p antagomir through intratumoral injection in vivo. CONCLUSIONS IH-induced tumor development is driven through miR-210-3p by E2F3 suppression. MiR-210-3p represents a potential therapeutic target among patients with concomitant cancer and OSA.
Collapse
Affiliation(s)
- Xiao-Bin Zhang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, No.201, Hubin South Road, Siming District, Xiamen, Fujian, People's Republic of China.
- The Third Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, People's Republic of China.
| | - Yang Song
- Ningde Food and Drug Inspection Testing Center, Ningde, Fujian, People's Republic of China
| | - Yan-Ting Lai
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, No.201, Hubin South Road, Siming District, Xiamen, Fujian, People's Republic of China
| | - Shao-Zhao Qiu
- Department of Anesthesiology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - An-Ke Hu
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, No.201, Hubin South Road, Siming District, Xiamen, Fujian, People's Republic of China
| | - Dai-Xi Li
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, No.201, Hubin South Road, Siming District, Xiamen, Fujian, People's Republic of China
| | - Nai-Shan Zheng
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, No.201, Hubin South Road, Siming District, Xiamen, Fujian, People's Republic of China
| | - Hui-Qing Zeng
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, No.201, Hubin South Road, Siming District, Xiamen, Fujian, People's Republic of China
| | - Qi-Chang Lin
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, No. 20, Chazhong Road, Taijiang District, Fuzhou, Fujian, People's Republic of China.
| |
Collapse
|
4
|
Recoquillon S, Ali S, Justeau G, Riou J, Martinez MC, Andriantsitohaina R, Gagnadoux F, Trzepizur W. Small Extracellular Vesicles Harboring PD-L1 in Obstructive Sleep Apnea. Int J Mol Sci 2024; 25:3208. [PMID: 38542182 PMCID: PMC10970061 DOI: 10.3390/ijms25063208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 02/29/2024] [Accepted: 03/06/2024] [Indexed: 04/04/2024] Open
Abstract
Obstructive sleep apnea syndrome (OSA) has been associated with increased cancer incidence and aggressiveness. One hypothesis to support this association is the implication of immune response, particularly the programmed cell death pathway, formed by the receptor PD-1 and its ligand PD-L1. Recent studies have shown dysregulation of this pathway in severe OSA patients. It has also been shown that small extracellular vesicles (sEVs) carrying PD-L1 induce lymphocyte dysfunction. Thus, the aim of our study was to analyze the expression of PD-L1 on sEVs of OSA patients and to evaluate the role of sEVs on lymphocyte activation and cytotoxicity. Circulating sEVs were isolated from OSA patients and the control group. Lymphocytes were isolated from the control group. Circulating sEVs were characterized by western blot, nanotracking analysis, and flow cytometry and were incubated with lymphocytes. Our results show no differences in the quantity and composition of sEVs in OSA patients and no significant effects of sEVs in OSA patients on lymphocyte activation and cytotoxicity. These results suggest that OSA does not modify PD-L1 expression on sEVs, which does not contribute to dysregulation of cytotoxic lymphocytes.
Collapse
Affiliation(s)
- Sylvain Recoquillon
- SFR ICAT, Team Carme, MitoVasc Laboratory, UMR CNRS 6015 INSERM 1083, University of Angers, 49000 Angers, France; (S.R.); (F.G.)
| | - Sakina Ali
- INSERM 1063, University of Angers, 49045 Angers, France;
| | - Grégoire Justeau
- Department of Respiratory and Sleep Medicine, Angers University Hospital, 49100 Angers, France;
| | - Jérémie Riou
- Delegation for Clinical Research and Innovation, Angers University Hospital, 49100 Angers, France;
| | - M. Carmen Martinez
- PhyMedExp, Montpellier University, INSERM, CNRS, CHRU Montpellier, 34295 Montpellier, France; (M.C.M.); (R.A.)
| | - Ramaroson Andriantsitohaina
- PhyMedExp, Montpellier University, INSERM, CNRS, CHRU Montpellier, 34295 Montpellier, France; (M.C.M.); (R.A.)
| | - Frédéric Gagnadoux
- SFR ICAT, Team Carme, MitoVasc Laboratory, UMR CNRS 6015 INSERM 1083, University of Angers, 49000 Angers, France; (S.R.); (F.G.)
- Department of Respiratory and Sleep Medicine, Angers University Hospital, 49100 Angers, France;
| | - Wojciech Trzepizur
- SFR ICAT, Team Carme, MitoVasc Laboratory, UMR CNRS 6015 INSERM 1083, University of Angers, 49000 Angers, France; (S.R.); (F.G.)
- Department of Respiratory and Sleep Medicine, Angers University Hospital, 49100 Angers, France;
| |
Collapse
|
5
|
Liu W, Zhu Q, Li X, Wang Y, Zhao C, Ma C. Effects of obstructive sleep apnea on myocardial injury and dysfunction: a review focused on the molecular mechanisms of intermittent hypoxia. Sleep Breath 2024; 28:41-51. [PMID: 37548920 DOI: 10.1007/s11325-023-02893-2] [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: 11/18/2022] [Revised: 06/08/2023] [Accepted: 07/26/2023] [Indexed: 08/08/2023]
Abstract
Obstructive sleep apnea (OSA) is characterized by intermittent hypoxia (IH) and is strongly associated with adverse cardiovascular outcomes. Myocardial injury and dysfunction have been commonly observed in clinical practice, particularly in patients with severe OSA. However, the underlying mechanisms remain obscure. In this review, we summarized the molecular mechanisms by which IH impact on myocardial injury and dysfunction. In brief, IH-induced cardiomyocyte death proceeds through the regulation of multiple biological processes, including differentially expressed transcription factors, alternative epigenetic programs, and altered post-translational modification. Besides cell death, various cardiomyocyte injuries, such as endoplasmic reticulum stress, occurs with IH. In addition to the direct effects on cardiomyocytes, IH has been found to deteriorate myocardial blood and energy supply by affecting the microvascular structure and disrupting glucose and lipid metabolism. For better diagnosis and treatment of OSA, further studies on the molecular mechanisms of IH-induced myocardial injury and dysfunction are essential.
Collapse
Affiliation(s)
- Wen Liu
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning Province, China
- Clinical Medical Research Center of Imaging in Liaoning Province, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Qing Zhu
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning Province, China
- Clinical Medical Research Center of Imaging in Liaoning Province, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Xinxin Li
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning Province, China
- Clinical Medical Research Center of Imaging in Liaoning Province, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Yonghuai Wang
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning Province, China
- Clinical Medical Research Center of Imaging in Liaoning Province, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Cuiting Zhao
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning Province, China
- Clinical Medical Research Center of Imaging in Liaoning Province, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning Province, China
| | - Chunyan Ma
- Department of Cardiovascular Ultrasound, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning Province, China.
- Clinical Medical Research Center of Imaging in Liaoning Province, The First Hospital of China Medical University, No. 155 NanjingBei Street, Heping District, Shenyang, 110001, Liaoning Province, China.
| |
Collapse
|
6
|
Schaubmayr W, Hochreiter B, Hunyadi-Gulyas E, Riegler L, Schmidt K, Tiboldi A, Moser B, Klein KU, Krenn K, Scharbert G, Mohr T, Schmid JA, Spittler A, Tretter V. The Proteome of Extracellular Vesicles Released from Pulmonary Microvascular Endothelium Reveals Impact of Oxygen Conditions on Biotrauma. Int J Mol Sci 2024; 25:2415. [PMID: 38397093 PMCID: PMC10889365 DOI: 10.3390/ijms25042415] [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: 01/22/2024] [Revised: 02/09/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
The lung can experience different oxygen concentrations, low as in hypoxia, high as under supplemental oxygen therapy, or oscillating during intermittent hypoxia as in obstructive sleep apnea or intermittent hypoxia/hyperoxia due to cyclic atelectasis in the ventilated patient. This study aimed to characterize the oxygen-condition-specific protein composition of extracellular vesicles (EVs) released from human pulmonary microvascular endothelial cells in vitro to decipher their potential role in biotrauma using quantitative proteomics with bioinformatic evaluation, transmission electron microscopy, flow cytometry, and non-activated thromboelastometry (NATEM). The release of vesicles enriched in markers CD9/CD63/CD81 was enhanced under intermittent hypoxia, strong hyperoxia and intermittent hypoxia/hyperoxia. Particles with exposed phosphatidylserine were increased under intermittent hypoxia. A small portion of vesicles were tissue factor-positive, which was enhanced under intermittent hypoxia and intermittent hypoxia/hyperoxia. EVs from treatment with intermittent hypoxia induced a significant reduction of Clotting Time in NATEM analysis compared to EVs isolated after normoxic exposure, while after intermittent hypoxia/hyperoxia, tissue factor in EVs seems to be inactive. Gene set enrichment analysis of differentially expressed genes revealed that EVs from individual oxygen conditions potentially induce different biological processes such as an inflammatory response under strong hyperoxia and intermittent hypoxia/hyperoxia and enhancement of tumor invasiveness under intermittent hypoxia.
Collapse
Affiliation(s)
- Wolfgang Schaubmayr
- Department of Anesthesia, General Intensive Care and Pain Medicine, Medical University of Vienna, 1090 Vienna, Austria (B.H.); (K.K.)
| | - Beatrix Hochreiter
- Department of Anesthesia, General Intensive Care and Pain Medicine, Medical University of Vienna, 1090 Vienna, Austria (B.H.); (K.K.)
| | - Eva Hunyadi-Gulyas
- Laboratory of Proteomics Research, HUN-REN Biological Research Centre, 6726 Szeged, Hungary;
| | - Louise Riegler
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Katy Schmidt
- Core Facility of Cell Imaging and Ultrastructure Research, University of Vienna, 1090 Vienna, Austria
| | - Akos Tiboldi
- Department of Anesthesia, General Intensive Care and Pain Medicine, Medical University of Vienna, 1090 Vienna, Austria (B.H.); (K.K.)
| | - Bernhard Moser
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Klaus U. Klein
- Department of Anesthesia, General Intensive Care and Pain Medicine, Medical University of Vienna, 1090 Vienna, Austria (B.H.); (K.K.)
| | - Katharina Krenn
- Department of Anesthesia, General Intensive Care and Pain Medicine, Medical University of Vienna, 1090 Vienna, Austria (B.H.); (K.K.)
| | - Gisela Scharbert
- Department of Anesthesia, General Intensive Care and Pain Medicine, Medical University of Vienna, 1090 Vienna, Austria (B.H.); (K.K.)
| | - Thomas Mohr
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Johannes A. Schmid
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, 1090 Vienna, Austria;
| | - Andreas Spittler
- Department of Surgery and Core Facility Flow Cytometry, Medical University of Vienna, 1090 Vienna, Austria;
| | - Verena Tretter
- Department of Anesthesia, General Intensive Care and Pain Medicine, Medical University of Vienna, 1090 Vienna, Austria (B.H.); (K.K.)
| |
Collapse
|
7
|
Loric S, Denis JA, Desbene C, Sabbah M, Conti M. Extracellular Vesicles in Breast Cancer: From Biology and Function to Clinical Diagnosis and Therapeutic Management. Int J Mol Sci 2023; 24:7208. [PMID: 37108371 PMCID: PMC10139222 DOI: 10.3390/ijms24087208] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/03/2023] [Accepted: 04/09/2023] [Indexed: 04/29/2023] Open
Abstract
Breast cancer (BC) is the first worldwide most frequent cancer in both sexes and the most commonly diagnosed in females. Although BC mortality has been thoroughly declining over the past decades, there are still considerable differences between women diagnosed with early BC and when metastatic BC is diagnosed. BC treatment choice is widely dependent on precise histological and molecular characterization. However, recurrence or distant metastasis still occurs even with the most recent efficient therapies. Thus, a better understanding of the different factors underlying tumor escape is mainly mandatory. Among the leading candidates is the continuous interplay between tumor cells and their microenvironment, where extracellular vesicles play a significant role. Among extracellular vesicles, smaller ones, also called exosomes, can carry biomolecules, such as lipids, proteins, and nucleic acids, and generate signal transmission through an intercellular transfer of their content. This mechanism allows tumor cells to recruit and modify the adjacent and systemic microenvironment to support further invasion and dissemination. By reciprocity, stromal cells can also use exosomes to profoundly modify tumor cell behavior. This review intends to cover the most recent literature on the role of extracellular vesicle production in normal and cancerous breast tissues. Specific attention is paid to the use of extracellular vesicles for early BC diagnosis, follow-up, and prognosis because exosomes are actually under the spotlight of researchers as a high-potential source of liquid biopsies. Extracellular vesicles in BC treatment as new targets for therapy or efficient nanovectors to drive drug delivery are also summarized.
Collapse
Affiliation(s)
- Sylvain Loric
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
| | | | - Cédric Desbene
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
| | - Michèle Sabbah
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
| | - Marc Conti
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
- INTEGRACELL SAS, 91160 Longjumeau, France
| |
Collapse
|
8
|
Zapater A, Barbé F, Sánchez-de-la-Torre M. Micro-RNA in obstructive sleep apnoea: biomarker of cardiovascular outcome? Curr Opin Pulm Med 2022; 28:559-570. [PMID: 36081397 DOI: 10.1097/mcp.0000000000000913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Obstructive sleep apnoea (OSA) is a global health problem with important cardiovascular consequences. Risk assessment tools are essential in OSA to identify patients at increased risk of cardiovascular disease and to achieve a cost-effective clinical management of the disease in the era of precision medicine. The objective is to provide an updated perspective on the role of microRNAs (miRNAs) in OSA as a biomarker of cardiovascular risk. RECENT FINDINGS Specific miRNAs have already been associated with patients with OSA and specific cardiovascular diseases such as hypertension, myocardial infarction or endothelial dysfunction. Numerous studies have addressed the use of miRNAs to identify the cardiovascular risk associated with OSA, both in patients and in animals with in vivo hypoxia models. Thus, these studies identified profiles of differentially expressed miRNAs in patients with OSA. In addition, the in vitro studies suggest that therapies with miRNA inhibitors that could help reduce cardiovascular risk. Therefore, this review highlights the primary approaches of the potential of miRNAs as biomarkers at the prognostic, diagnostic and therapeutic strategy levels. SUMMARY Given the heterogeneity of OSA and its cardiovascular consequences, miRNAs have emerged as powerful biomarkers that can help improve the clinical management of OSA and its cardiovascular risk.
Collapse
Affiliation(s)
- Andrea Zapater
- Precision Medicine in Chronic Diseases, Hospital Universitari Arnau de Vilanova-Santa Maria, Department of Nursing and Physiotherapy, Faculty of Nursing and Physiotherapy, University of Lleida, IRB Lleida, Lleida
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid
| | - Ferran Barbé
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid
- Translation Research in Respiratory Medicine, Hospital Universitari Arnau de Vilanova-Santa Maria, IRB Lleida, Lleida, Spain
| | - Manuel Sánchez-de-la-Torre
- Precision Medicine in Chronic Diseases, Hospital Universitari Arnau de Vilanova-Santa Maria, Department of Nursing and Physiotherapy, Faculty of Nursing and Physiotherapy, University of Lleida, IRB Lleida, Lleida
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Madrid
| |
Collapse
|
9
|
Soccio P, Moriondo G, Lacedonia D, Tondo P, Quarato CMI, Foschino Barbaro MP, Scioscia G. EVs-miRNA: The New Molecular Markers for Chronic Respiratory Diseases. Life (Basel) 2022; 12:1544. [PMID: 36294979 PMCID: PMC9605003 DOI: 10.3390/life12101544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 09/30/2022] [Accepted: 10/02/2022] [Indexed: 11/16/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD), asthma and sleep disorders are chronic respiratory diseases that affect the airways, compromising lung function over time. These diseases affect hundreds of millions of people around the world and their frequency seems to be increasing every year. Extracellular vesicles (EVs) are small-sized vesicles released by every cell in the body. They are present in most body fluids and contain various biomolecules including proteins, lipids, mRNA and non-coding RNA (micro-RNA). The EVs can release their cargo, specifically micro-RNAs (miRNAs), to both neighboring and/or distal cells, playing a fundamental role in cell-cell communication. Recent studies have shown their possible role in the pathogenesis of various chronic respiratory diseases. The expression of miRNAs and, in particular, of miRNAs contained within the extracellular vesicles seems to be a good starting point in order to identify new potential biomarkers of disease, allowing a non-invasive clinical diagnosis. In this review we summarize some studies, present in the literature, about the functions of extracellular vesicles and miRNAs contained in extracellular vesicles in chronic respiratory diseases and we discuss the potential clinical applications of EVs and EVs-miRNAs for their possible use such as future biomarkers.
Collapse
Affiliation(s)
- Piera Soccio
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Giorgia Moriondo
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Donato Lacedonia
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
- Institute of Respiratory Diseases, Policlinico Riuniti of Foggia, 71122 Foggia, Italy
| | - Pasquale Tondo
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Carla Maria Irene Quarato
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
- Institute of Respiratory Diseases, Policlinico Riuniti of Foggia, 71122 Foggia, Italy
| | - Maria Pia Foschino Barbaro
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
- Institute of Respiratory Diseases, Policlinico Riuniti of Foggia, 71122 Foggia, Italy
| | - Giulia Scioscia
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
- Institute of Respiratory Diseases, Policlinico Riuniti of Foggia, 71122 Foggia, Italy
| |
Collapse
|
10
|
Extracellular Vesicles as Drivers of Immunoinflammation in Atherothrombosis. Cells 2022; 11:cells11111845. [PMID: 35681540 PMCID: PMC9180657 DOI: 10.3390/cells11111845] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 02/07/2023] Open
Abstract
Atherosclerotic cardiovascular disease is the leading cause of morbidity and mortality all over the world. Extracellular vesicles (EVs), small lipid-bilayer membrane vesicles released by most cellular types, exert pivotal and multifaceted roles in physiology and disease. Emerging evidence emphasizes the importance of EVs in intercellular communication processes with key effects on cell survival, endothelial homeostasis, inflammation, neoangiogenesis, and thrombosis. This review focuses on EVs as effective signaling molecules able to both derail vascular homeostasis and induce vascular dysfunction, inflammation, plaque progression, and thrombus formation as well as drive anti-inflammation, vascular repair, and atheroprotection. We provide a comprehensive and updated summary of the role of EVs in the development or regression of atherosclerotic lesions, highlighting the link between thrombosis and inflammation. Importantly, we also critically describe their potential clinical use as disease biomarkers or therapeutic agents in atherothrombosis.
Collapse
|
11
|
Wang X, Li Z, Du Y, Xing Y, Guo Y, Zhang Y, Guo R, Gong W, Nie S, Wang X. lncRNA Mirt1: A Critical Regulatory Factor in Chronic Intermittent Hypoxia Exaggerated Post-MI Cardiac Remodeling. Front Genet 2022; 13:818823. [PMID: 35222538 PMCID: PMC8864109 DOI: 10.3389/fgene.2022.818823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Chronic intermittent hypoxia (CIH) is the main feature of obstructive sleep apnea (OSA) and is known to exaggerate cardiac remodeling after myocardial infarction (MI). However, the specific contribution of CIH to overall OSA-induced pathological complications and the transcriptomic mechanisms underlying CIH-exaggerated post-MI remodeling remains unclear. In this study, we used RNA-sequencing to construct the expression profiles of cardiac mRNAs, microRNAs, and long non-coding RNAs (lncRNA) in four groups of C57BL/6J mice (Sham, CIH, MI, MI + CIH) to evaluate how CIH regulates cardiac remodeling after MI. Compared with the other three groups, the MI + CIH group exhibited 345 lncRNAs, 35 microRNAs, and 5,220 differentially expressed mRNAs. Further analysis showed that CIH led to significant changes in Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway of the differentially expressed mRNAs. Co-expression network analysis identified two core lncRNAs (Mirt1 and AC125351.1) and two core microRNAs (miR-466i-5p and miR-574-5p) during the development of CIH-exaggerated post-MI remodeling, and they were verified by quantitative real-time PCR (qRT-PCR). LncRNA-mRNA correlation analysis further showed that lncRNA Mirt1 was positively correlated with Apbb1ip and Lcp2. In addition, microRNA-mRNA correlation analysis showed that microRNA miR-466i-5p was positively correlated with Snai2, Cdc27, and Ngfr. Furthermore, combining with lncRNA-mRNA and miRNA-mRNA networks, 44 RNAs were identified in the competitive endogenous RNA (ceRNA) network. Mirt1 acts as a ceRNA to bind to miR-466i-5p to further regulate the expression levels of the target gene, thereby aggravating cardiac remodeling after MI. In conclusion, our study provides a systematic perspective on the potential functions of mRNAs, microRNAs, and lncRNAs in CIH-exaggerated post-MI cardiac remodeling. Our data suggest that lncRNA Mirt1 may be the most critical regulator of MI aggravated by CIH.
Collapse
Affiliation(s)
- Xinxia Wang
- Department of Respiratory and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Zexuan Li
- Center for Coronary Artery Disease, Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yunhui Du
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yuanyuan Xing
- Comprehensive Ultrasound Department, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yingying Guo
- Center for Coronary Artery Disease, Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yushi Zhang
- Center for Coronary Artery Disease, Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ruifeng Guo
- Center for Coronary Artery Disease, Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Wei Gong
- Center for Coronary Artery Disease, Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Shaoping Nie
- Center for Coronary Artery Disease, Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xiao Wang
- Center for Coronary Artery Disease, Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- *Correspondence: Xiao Wang,
| |
Collapse
|
12
|
Sanz-Rubio D, Khalyfa A, Qiao Z, Ullate J, Marin JM, Kheirandish-Gozal L, Gozal D. Cell-Selective Altered Cargo Properties of Extracellular Vesicles Following In Vitro Exposures to Intermittent Hypoxia. Int J Mol Sci 2021; 22:ijms22115604. [PMID: 34070558 PMCID: PMC8198838 DOI: 10.3390/ijms22115604] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/09/2021] [Accepted: 05/19/2021] [Indexed: 01/09/2023] Open
Abstract
Intermittent hypoxia (IH), a hallmark of obstructive sleep apnea (OSA), is associated with cardiovascular and metabolic dysfunction. However, the mechanisms underlying these morbidities remain poorly delineated. Extracellular vesicles (EVs) mediate intercellular communications, play pivotal roles in a multitude of physiological and pathological processes, and could mediate IH-induced cellular effects. Here, the effects of IH on human primary cells and the release of EVs were examined. Microvascular endothelial cells (HMVEC-d), THP1 monocytes, THP1 macrophages M0, THP1 macrophages M1, THP1 macrophages M2, pre-adipocytes, and differentiated adipocytes (HAd) were exposed to either room air (RA) or IH for 24 h. Secreted EVs were isolated and characterized using transmission electron microscopy, nanoparticle tracking analysis, and Western blotting. The effects of each of the cell-derived EVs on endothelial cell (EC) monolayer barrier integrity, on naïve THP1 macrophage polarity, and on adipocyte insulin sensitivity were also evaluated. IH did not alter EVs cell quantal release, but IH-EVs derived from HMVEC-d (p < 0.01), THP1 M0 (p < 0.01) and HAd (p < 0.05) significantly disrupted HMVEC-d monolayer integrity, particularly after H2O2 pre-conditioning. IH-EVs from HMVEC-d and THP1 M0 elicited M2-polarity changes did not alter insulin sensitivity responses. IH induces cell-selective changes in EVs cargo, which primarily seem to target the emergence of endothelial dysfunction. Thus, changes in EVs cargo from selected cell sources in vivo may play causal roles in some of the adverse outcomes associated with OSA.
Collapse
Affiliation(s)
- David Sanz-Rubio
- Department of Child Health, Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO 65201, USA; (D.S.-R.); (Z.Q.); (J.U.); (L.K.-G.); (D.G.)
- Translational Research Unit, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria de Aragón (IISAragón), 50009 Zaragoza, Spain;
| | - Abdelnaby Khalyfa
- Department of Child Health, Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO 65201, USA; (D.S.-R.); (Z.Q.); (J.U.); (L.K.-G.); (D.G.)
- Correspondence: ; Tel.: +1-573-884-7685
| | - Zhuanhong Qiao
- Department of Child Health, Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO 65201, USA; (D.S.-R.); (Z.Q.); (J.U.); (L.K.-G.); (D.G.)
| | - Jorge Ullate
- Department of Child Health, Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO 65201, USA; (D.S.-R.); (Z.Q.); (J.U.); (L.K.-G.); (D.G.)
| | - José M. Marin
- Translational Research Unit, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria de Aragón (IISAragón), 50009 Zaragoza, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERes), 28029 Madrid, Spain
| | - Leila Kheirandish-Gozal
- Department of Child Health, Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO 65201, USA; (D.S.-R.); (Z.Q.); (J.U.); (L.K.-G.); (D.G.)
| | - David Gozal
- Department of Child Health, Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO 65201, USA; (D.S.-R.); (Z.Q.); (J.U.); (L.K.-G.); (D.G.)
| |
Collapse
|